Sie sind auf Seite 1von 283

Novel Natural Products from Endophytic Fungi

of Egyptian Medicinal Plants - Chemical and


Biological Characterization

Neue Naturstoffe aus endophytischen Pilzen


ägyptischer Arzneipflanzen - chemische und
biologische Charakterisierung

Inaugural-Dissertation
zur
Erlangung des Doktorgrades
der Mathematisch-Naturwissenschaftlichen Fakultät
der Heinrich-Heine-Universität Düsseldorf

vorgelegt von
Amal E. H. A. Hassan
aus Alexandria, Ägypten

Düsseldorf, 2007
Aus dem Institut für Pharmazeutische Biologie und Biotechnologie
der Heinrich-Heine Universität Düsseldorf

Gedruckt mit der Genehmigung der


Mathematisch-Naturwissenschaftlichen Fakultät der
Heinrich-Heine-Universität Düsseldorf

Gedruckt mit der Unterstützung des


Deutschen Akademischen Austauschdienstes (DAAD)

Referent: Prof. Dr. Peter Proksch


Koreferent: Dr. Rainer Ebel, Juniorprofessor

Tag der mündlichen Prüfung: 25.06.2007


Erklärung

Hiermit erkläre ich ehrenwörtlich, dass ich die vorliegende Dissertation mit dem Titel
„Neue Naturstoffe aus endophytischen Pilzen ägyptischer Arzneipflanzen - chemische und
biologische Charakterisierung“ selbst angefertigt habe. Außer den angegebenen Quellen und
Hilfsmitteln wurden keine weiteren verwendet. Diese Dissertation wurde weder in gleicher
noch in abgewandelter Form in einem anderen Prüfungsverfahren vorgelegt. Weiterhin
erkläre ich, dass ich früher weder akademische Grade erworben habe, noch dies versucht
habe.

Düsseldorf, den 10.05.2007

Amal Hassan
Acknowledgement

Acknowledgement

It is a pleasure to find the chance to show my gratitude and all my regards to J. Prof.
Dr. Rainer Ebel for his instructive supervision, his kind help and his continuous support and
encouragement throughout the completion of this work.
I would like to express my cordial thanks and gratitude to Prof. Dr. rer. nat. Peter
Proksch for giving me the opportunity to pursue my doctoral research at the institute, as well
as for his valuable suggestions, his fruitful discussions, his unforgettable support and for the
excellent work facilities at the Institut für Pharmazeutische Biologie und Biotechnologie,
Heinrich-Heine-Universität, Düsseldorf.
My special thanks to Dr. RuAngelie Edrada-Ebel for her constructive advises, NMR
courses, sharing her expertise in NMR data interpretation as well as for her help and support
in good times and bad times.
Many thanks for the friendly cooperation to Prof. Dr. rer. nat. Werner E. G. Müller
and Renate Steffen, Institut für Physiologische Chemie und Pathobiochemie, University of
Mainz, for carrying out the cytotoxicity tests, PD. Dr. Ute Hentschel, Zentrum für
Infektionsforschung, University of Würzburg, for performing the biofilm inhibition test and
Dr. Michael Kubbutat, ProQinase GmbH, Freiburg, for conducting the protein kinase
inhibition assays.
I also appreciate the sincere cooperation of Dr. W. Peters and his coworkers, Institut
für Anorganische und Strukturchemie, Heinrich-Heine-Universität, Düsseldorf, for 500 MHz
NMR measurements, Dr. Victor Wray, Helmholtz Centre for Infection Research,
Braunschweig, and his coworkers for 600 MHz NMR measurements as well as HR-mass
spectrometry experiments, Dr. H. Keck and Dr. P. Tommes, Institut für Anorganische und
Strukturchemie, Heinrich-Heine-Universität, Düsseldorf, for conducting EI- and FAB-mass
spectrometry experiments.
My deep thanks are also to Prof. Dr. Amin El-Sayed Ali, Department of Crops,
Faculty of Agriculture, Alexandria University, and Prof. Dr. Rafiq El-Gharib Mahmoud,
Department of Botany, Faculty of Science, Alexandria University, for the identification of the
plant material, as well as the molecular biology and antimicrobial assay teams at the institute
for the identification of purified fungal strains and performing antimicrobial assays,
respectively.
I would like to thank my past and present colleagues Dr. Moustafa Abdelgawwad, Dr.
Mohamed Ashour, Dr. Ziyad Baker, Dr. Tu N. Duong, Dr. Gero Eck, Dr. Hefni Effendi, Dr.

iv
Acknowledgement

Wafaa Hassan, Dr. Sabrin Ibrahim, Dr. Yoshi B. Murti, Dr. Suwigarn Pedpradab, Dr. Yudi
Rusman, Dr. Bärbel Steffan, Dr. Franka Teuscher, Dr. Carsten Thoms, Dr. Yasman, Mirko
Bayer, Abdessamad Debbab, Arnulf Diesel, Sherif Elsayed, Clécia Freitas-Richard, Ashraf
Hamed, Triana Hertiani, Ine Dewi Inderiani, Julia Jacob, Ehab Moustafa, Edi W. Sri
Mulonyo, Sofia Ortlepp, Annika Putz, Frank Riebe, Anke Suckow-Schnitker, Yao Wang,
Nadine Weber, Sabri Younes, and all the others for the nice multicultural time I spent with
them, for their help and assistance whenever I needed it. Special thanks to Mareike Thiel for
her administrative help whenever needed, as well as Katrin Rohde and Waltraud Schlag for
their kind help in any technical problem encountered during the work.
My great appreciation to DAAD (German Academic Exchange Service) for the
financial support during my stay in Germany, and to my region reference Margret Leopold for
her kind support during my stay.
Finally, I would like to thank my small scientific family, my father, my mother and
my sister, who were always there for me, especially my parents, who supported me and made
it possible for me to set my own goals and to reach them.
Thank you!

v
Zusammenfassung

Zusammenfassung

Endophytische Pilze produzieren Naturstoffe mit einer Vielfalt an chemischen


Strukturen, die für spezifische medizinische oder agrochemische Anwendungen von großem
Interesse sein könnten. Viele dieser Sekundärstoffe weisen biologische Aktivitäten in
pharmakologisch relevanten Assaysystemen auf, die sie zu potentiellen Leitstrukturen für die
Entwicklung neuer Arzneistoffe machen.
Ziel dieser Arbeit war die Isolierung von Sekundärstoffen aus Endophyten
terrestrischer Pflanzen, gefolgt von Strukturaufklärung und Untersuchung ihres
pharmakologischen Potentials. Vier endophytische Pilze, nämlich Alternaria sp.,
Ampelomyces sp., Stemphylium botryosum und Chaetomium sp., gewonnen aus ägyptischen
Arzneipflanzen, wurden als Naturstoffquellen ausgewählt und über einen Zeitraum von drei
bis vier Wochen in Standkulturen in Wickerham-Flüssigmedium sowie in Reis-Festmedium
angezogen. Die aus der folgenden Extraktion erhaltenen Fraktionen wurden zur Isolierung der
Naturstoffe weiteren chromatographischen Trennmethoden unterzogen.
Zur Strukturaufklärung wurden moderne analytische Verfahren wie die
Massenspektrometrie (MS) und die Kernresonanzspektroskopie (NMR) eingesetzt. Zusätzlich
wurden für einige optisch aktive Verbindungen chirale Derivatisierungsreaktionen
angewendet, um deren absolute Konfiguration zu ermitteln. Schließlich wurden die erhaltenen
Substanzen verschiedenen Biotests unterzogen, um ihre antimikrobiellen, antifungalen und
cytotoxischen Eigenschaften sowie die Wirkung als Inhibitoren verschiedener Proteinkinasen
sowie der Biofilmbildung von Staphylococcus epidermidis zu ermitteln.

1. Alternaria sp.
Drei neue Alternariolderivate wurden aus Alternaria sp., isoliert aus Polygonum
senegalense, gewonnen. Des weiteren wurden aus diesem Pilz vier neue Verbindungen,
nämlich Desmethylaltenusin, 4`-Epialtenuene, Alterlacton und Alternariasäure, isoliert. Die
Alternariolderivate sowie einige strukturverwandte Verbindungen wiesen sowohl ausgeprägte
zytotoxische Eigenschaften im Test mit der Zellinie L5178Y (murines T-Zell Lymphom) als
auch inhibitorische Aktivität gegenüber Proteinkinasen auf.

2. Ampelomyces sp.
Ampelomyces sp. ist ein Isolat aus Urospermum picroides. Aus diesem Pilz wurden
sechs neue Verbindungen isoliert, darunter ein neues Pyron, zwei neue Isocoumarine, zwei

vi
Zusammenfassung

neue sulfatierte Anthrachinone und ein neues Hexahydroanthronol. In den Biotests zeigten
Desmethyldiaportinol, Altersolanol A und Methylalaternin zytotoxische Aktivität gegenüber
L5178Y-Zellen. Des weiteren zeigten Altersolanol A und Methylalaternin inhibitorische
Aktivität gegenüber der Biofilmbildung von S. epidermidis.

3. Stemphylium botryosum
Aus Chenopodium album wurde der Pilz Stemphylium botryosum isoliert. Daraus
konnten Curvularinderivate isoliert werden, die ausgeprägte zytotoxische Eigenschaften im
Test mit der Zellinie L5178Y zeigten.

4. Chaetomium sp.
Schließlich wurde der Pilz Chaetomium sp., gewonnen aus Otanthus maritimus,
untersucht. Ein neues Tetrahydrofuranderivat sowie zwei bekannte Cochliodinolderivate und
Orsellinsäure wurden aus Extrakten dieses Pilzes gewonnen. Die Cochliodinolderivate wiesen
inhibitorische Aktivität gegenüber Proteinkinasen auf; weiterhin zeigten Cochliodinol und
Orsellinsäure ausgeprägte zytotoxische Eigenschaften gegenüber der Zellinie L5178Y.

Insgesamt wurden in dieser Arbeit zweiundvierzig Verbindungen isoliert, von denen


vierzehn neue Naturstoffe darstellen. Sowohl die neuen als auch die bekannten Substanzen
wurden in Hinsicht auf bioaktiven Eigenschaften in verschiedenen Biotests untersucht.
Die Extrake der jeweiligen Wirtspflanzen wurden mit Hilfe von LC/MS gezielt auf die
isolierten Naturstoffe aus den endophytischen Pilzen hin untersucht. Keiner der isolierten
Sekundärstoffe des endophytischen Pilzes Chaetomium sp. war in den Fraktionen von O.
maritimus zu detektieren. Dagegen konnten Komponenten der übrigen Pilzextrakte eindeutig
in Fraktionen der jeweiligen Wirtspflanzen P. senegalense, U. picroides and C. album
nachgewiesen werden.

vii
Contents

Table of Contents

1. Introduction 1
1.1. Endophytes 1
1.2. Endophyte-host plant interaction 2
1.3. Microbial biodiversity 3
1.4. Plant selection for isolation of endophytes 4
1.5. The potential of natural products in drug discovery 5
1.6. The potential of fungal natural products in drug discovery 5
1.7. Endophytic fungi as a source of bioactive natural products 7
1.7.1. Secondary metabolites from endophytes as antibiotics 8
1.7.2. Secondary metabolites from endophytes as antimycotic agents 8
1.7.3. Secondary metabolites from endophytes as antiviral agents 9
1.7.4. Secondary metabolites from endophytes as anticancer agents 10
1.7.5. Secondary metabolites from endophytes with further interesting pharmacological 12
activities
1.8. The potential of microbial natural products in agriculture 13
1.9. Aim and scopes of the study 15

2. Materials and Methods 16

2.1. Materials 16
2.1.1. Biological materials 16
2.1.1.1. Plant material 16
2.1.1.2. Pure fungal strains isolated from the collected plants 16
2.1.2. Media 17
2.1.2.1. Composition of malt agar (MA) medium 17
2.1.2.2. Composition of Wickerham medium for liquid cultures 17
2.1.2.3. Composition of rice medium for solid cultures 18
2.1.2.4. Composition of Luria Bertani (LB) medium 18
2.1.2.5. Composition of yeast medium 18
2.1.2.6. Composition of fungal medium for bioassay 18
2.1.2.7. Composition of potato dextrose agar (PDA) medium for bioassay 19
2.1.2.8. Composition of trypticase soy broth (TSB) 19

viii
Contents

2.1.3. Chemicals 19
2.1.3.1. General laboratory chemicals 19
2.1.3.2. Chemicals for culture media 20
2.1.3.3. Chemicals for agarose gel electrophoresis 20
2.1.4. Chromatography 20
2.1.4.1. Stationary phases 20
2.1.4.2. Spray reagents 21
2.1.5. Solvents 21
2.1.5.1. General solvents 21
2.1.5.2. Solvents for HPLC 21
2.1.5.3. Solvents for optical rotation 22
2.1.5.4. Solvents for NMR 22

2.2. Methods 22
2.2.1. Purification of fungal strains 22
2.2.2. Cultivation of pure fungal strains 22
2.2.2.1. Cultivation for short term storage 22
2.2.2.2. Cultivation for screening and isolation of secondary metabolites 23
2.2.3. Extraction of fungal cultures and host plant material 23
2.2.3.1. Extraction of fungal liquid cultures 23
2.2.3.1.1. Total extraction of culture media and mycelia 23
2.2.3.1.2. Separate extraction of culture media and mycelia 24
2.2.3.2. Extraction of solid rice cultures 25
2.2.3.3. Extraction and fractionation of the plant material 26
2.2.3.4. Solvent-solvent extraction 26
2.2.4. Identification of fungal strains and their taxonomy 27
2.2.4.1. Fungal identification 27
2.2.4.2. Taxonomy 28
2.2.5. Isolation and purification of secondary metabolites 33
2.2.5.1. Isolation of the secondary metabolites from Alternaria sp. 33
2.2.5.1.1. Secondary metabolites isolated from liquid cultures of Alternaria sp. 33
2.2.5.1.2. Secondary metabolites isolated from rice cultures of Alternaria sp. 34
2.2.5.2. Isolation of the secondary metabolites from Ampelomyces sp. 35
2.2.5.2.1. Secondary metabolites isolated from liquid cultures of Ampelomyces sp. 35

ix
Contents

2.2.5.2.2. Secondary metabolites isolated from rice cultures of Ampelomyces sp. 37


2.2.5.3. Isolation of the secondary metabolites from Stemphylium botryosum 38
2.2.5.4. Isolation of the secondary metabolites from Chaetomium sp. 39
2.2.5.5. Chromatographic methods 39
2.2.5.5.1. Thin layer chromatography (TLC) 39
2.2.5.5.2. Vacuum liquid chromatography (VLC) 40
2.2.5.5.3. Column chromatography 40
2.2.5.5.4. Flash chromatography 41
2.2.5.5.5. Preparative high pressure liquid chromatography (HPLC) 41
2.2.5.5.6. Semi-preparative high pressure liquid chromatography (HPLC) 42
2.2.5.5.7. Analytical high pressure liquid chromatography (HPLC) 43
2.2.6. Structure elucidation of the isolated secondary metabolites 43
2.2.6.1. Mass spectrometry (MS) 43
2.2.6.1.1. Electrospray ionization mass spectrometry (ESI-MS) 43
2.2.6.1.2. Electron impact mass spectrometry (EI-MS) 44
2.2.6.1.3. Fast atom bombardment mass spectrometry (FAB-MS) 45
2.2.6.1.4. High resolution mass spectrometry (HR-MS) 45
2.2.6.2. Nuclear magnetic resonance spectroscopy (NMR) 45
2.2.6.3. Optical activity 46
2.2.6.4. Determination of absolute stereochemistry by Mosher reaction 46
2.2.7. Testing the biological activity 47
2.2.7.1. Antimicrobial assay 47
2.2.7.1.1. Agar diffusion assay 47
2.2.7.1.2. Inhibition of biofilm formation 49
2.2.7.2. Cytotoxicity test 49
2.2.7.2.1. Microculture tetrazolium (MTT) assay 49
2.2.7.2.2. Protein kinase assay 50
2.2.8. General laboratory equipments 53

3. Results 54

3.1. Compounds isolated from the endophytic fungus Alternaria sp. 54


3.1.1. Alternariol (1, known compound) 56
3.1.2. Alternariol-5-O-sulphate (2, new compound) 58

x
Contents

3.1.3. Alternariol-5-O-methylether (3, known compound) 60


3.1.4. Alternariol-5-O-methylether-4`-O-sulphate (4, new compound) 62
3.1.5. 3`-Hydroxyalternariol-5-O-methylether (5, new compound) 64
3.1.6. Altenusin (6, known compound) 69
3.1.7. Desmethylaltenusin (7, new compound) 71
3.1.8. Alterlactone (8, new compound) 75
3.1.9. Talaroflavone (9, known compound) 79
3.1.10. Alternaric acid (10, new compound) 81
3.1.11. Altenuene and 4`-epialtenuene (11, known, and 12, new compound) 88
3.1.13. 2,5-Dimethyl-7-hydroxychromone (13, known compound) 93
3.1.14. Altertoxin I (14, known compound) 96
3.1.15. Tenuazonic acid (15, known compound) 99
3.1.16. Bioactivity test results for compounds isolated from the endophytic fungus 102
Alternaria sp.

3.2. Compounds isolated from the endophytic fungus Ampelomyces sp. 104
3.2.1. Methyltriacetic lactone (16, known compound) 106
3.2.2. Ampelopyrone (17, new compound) 108
3.2.3. Desmethyldiaportinol (18, new compound) 112
3.2.4. Desmethyldichlorodiaportin (19, new compound) 114
3.2.5. (+)-Citreoisocoumarin (20, known compound) 116
3.2.6. Macrosporin (21, known compound) 120
3.2.7. Macrosporin-7-O-sulphate (22, new compound) 122
3.2.8. 3-O-Methylalaternin (23, known compound) 125
3.2.9. 3-O-Methylalaternin-7-O-sulphate (24, new compound) 127
3.2.10. Altersolanol A (25, known compound) 130
3.2.11. Ampelanol (26, new compound) 132
3.2.12. Alterporriol D (27, known compound) 138
3.2.13. Alterporriol E (28, known compound) 140
3.2.14. Altersolanol J (29, known compound) 144
3.2.15. Bioactivity test results for compounds isolated from the endophytic fungus 147
Ampelomyces sp.

xi
Contents

3.3. Compounds isolated from the endophytic fungus Stemphylium botryosum 149
3.3.1. Tetrahydroaltersolanol B (30, known compound) 151
3.3.2. Stemphyperylenol (31, known compound) 154
3.3.3. Curvularin (32, known compound) 157
3.3.4. Dehydrocurvularin (33, known compound) 159
3.3.5. Bioactivity test results for compounds isolated from the endophytic fungus 163
Stemphylium botryosum

3.4. Compounds isolated from the endophytic fungus Chaetomium sp. 164
3.4.1. Aureonitolic acid (34, new compound) 166
3.4.2. Cochliodinol (35, known compound) 172
3.4.3. Isocochliodinol (36, known compound) 174
3.4.4. Indole-3-carboxylic acid (37, known compound) 178
3.4.6. Cyclo(alanyltryptophane) (38, known compound) 180
3.4.7. Orsellinic acid (39, known compound) 183
3.4.8. Bioactivity test results for compounds isolated from the endophytic fungus 185
Chaetomium sp.

3.5. Tracing of fungal metabilites in the corresponding plant extracts 186


3.5.1. Tracing of Alternaria metabolites in Polygonum senegalense fractions 186
3.5.2. Tracing of Ampelomyces metabolites in Urospermum picroides fractions 189
3.5.3. Tracing of Stemphylium botryosum metabolites in Chenopodium album fractions 190
3.5.4. Tracing of Chaetomium sp. metabolites in Otanthus maritimus fractions 193

4. Discussion 194
4.1. Choice of culture media 194
4.2. Strategies and methodologies for metabolite profiling 195
4.2.1. HPLC/UV 195
4.2.2. HPLC/ESI-MS 196
4.2.3. Dereplication and partial identification of natural products by UV-based 197
techniques
4.3. Isolation of natural products 197
4.4. Compounds isolated from purified fungal strains 197
4.4.1. Compounds isolated from the endophytic fungus Alternaria sp. 197

xii
Contents

4.4.1.1. Alternariol derivatives 198


4.4.1.1.1. Biosynthesis of alternariol derivatives 198
4.4.1.1.2. Biosynthesis of biphenic acids and related compounds 200
4.4.1.1.3. Bioactivity and structure activity relationship of alternariol and biphenic acid 201
derivatives
4.4.1.1.4. Toxicity studies of alternariol derivatives 202
4.4.1.1.5. Occurrence of sulphated metabolites 203
4.4.1.2. Biosynthesis of 2,5-dimethyl-7-hydroxychromone 203
4.4.1.3. Biosynthesis of reduced perylenequinones 204
4.4.1.4. Tautomerism and biosynthesis of tenuazonic acid 205
4.4.1.5. Bioactivity of selected Alternaria metabolites 206
4.4.2. Compounds isolated from the endophytic fungus Ampelomyces sp. 207
4.4.2.1. Anthraquinones and modified anthraquinones 207
4.4.2.1.1. Biosynthesis of anthraquinones and modified anthraquinones 207
4.4.2.1.2. Bioactivity of anthraquinones and modified anthraquinones 208
4.4.2.2. Pyrone and isocoumarin derivatives 211
4.4.2.2.1. Biosynthesis of pyrone and isocoumarin derivatives 211
4.4.2.2.2. Bioactivity of pyrone and isocoumarin derivatives 212
4.4.3. Compounds isolated from the endophytic fungus Stemphylium botryosum 213
4.4.3.1. Biosynthesis of stemphyperylenol 213
4.4.3.2. Biosynthesis of macrocyclic lactones 214
4.4.3.3. Bioactivity of selected Stemphylium metabolites 215
4.4.4. Compounds isolated from the endophytic fungus Chaetomium sp. 215
4.4.4.1. Biosynthesis of tetrahydrofurans 216
4.4.4.2. Biosynthesis of bis-(3-indolyl)-benzoquinones 217
4.4.4.3. Bioactivity of selected Chaetomium metabolites 218
4.5. Detection of fungal metabolites in the host plant fractions 218

5. Conclusion 221
6. References 228
7. List of abbreviations 247
8. Attachments 249
Curriculum 269

xiii
Introduction

1. Introduction

Fungi play pivotal ecological roles in virtually all ecosystems. Saprotrophic fungi are
important in the cycling of nutrients, especially the carbon that is sequestered in wood and
other plant tissues. Pathogenic and parasitic fungi attack effectively all groups of organisms,
including bacteria, plants, other fungi, and animals, including humans. Other fungi function as
mutualistic symbionts, including mycangial associates of insects, mycorrhizae, lichens, and
endophytes. Through these symbioses, fungi have enabled a diversity of other organisms to
exploit novel habitats and resources. Indeed, the establishment of mycorrhizal associations
may be a key factor that enabled plants to make the transition from aquatic to terrestrial
habitats (Lutzoni et al., 2004).

1.1. Endophytes
Mycologists have come to use the term endophyte for fungi that inhabit living, internal
tissues of plants without causing visible disease symptoms. The term refers only to fungi at
the moment of detection without regard for the actual status of the interaction. Endophytic
fungi living asymptomatically within plant tissues have been found in virtually all plant
species (Saikkonen et al., 1998; Bacon and White, 2000). The definition thus includes a wide
range of fungi, from fungal plant pathogens and saprophytes that have extended latency
periods before disease or external signs of infection appear, to obligate mutualists.
Accordingly, the distinction between classical plant fungal pathogens and mutualists is not
clear and interactions between fungi and host plant are often variable (Saikkonen et al., 1998).
It is hypothesized that there are no neutral interactions, but rather that endophyte-host
interactions involve a balance of antagonisms. There is always at least a certain degree of
virulence on the part of the fungus enabling infection, whereas defense of the plant host limits
development of fungal invaders and disease (Schulz and Boyle, 2005). Many endophytes are
closely related to pathogenic fungi, and presumably evolved from them via an extension of
latency periods and a reduction of virulence (White et al., 1993). It is also hypothesized that
endophytes, in contrast to known pathogens, generally have far greater phenotypic plasticity
and thus more options than pathogens including infection, local but also extensive
colonization, latency, virulence, pathogenicity, or saprophytism (Schulz and Boyle, 2005).

1
Introduction

1.2. Endophyte-host plant interaction


The interactions between host plants and endophytes in natural populations and
communities are poorly understood. The endophyte-host plant symbioses represent a broad
continuum of interactions, from pathogenic to mutualistic, even within the lifespan of an
individual microorganism and its host plant (Freeman and Rodriguez, 1993; Saikkonen et al.,
1998; Schulz et al., 2002). Studies showed that endophytes are more likely to be mutualistic
when reproducing vertically (systemic) by growing into seeds, and more antagonistic to the
host when transmitted horizontally (nonsystemic) via spores (Schardl et al., 1991; Saikkonen
et al., 1998). It is possible to imagine that some of these endophytic microbes may have
devised genetic systems allowing for the transfer of information between themselves and the
higher plant and vice versa (Stierle et al., 1993; Strobel, 2002a). Obviously, this would permit
a more rapid and reliable mechanism of the endophyte to deal with environmental conditions
and perhaps allow for more compatibility with the plant host leading to symbiosis (Strobel,
2002a).
Endophytic fungi are thought to interact mutualistically with their host plants mainly
by increasing host resistance to herbivores and have been termed ‘‘acquired plant defenses’’
(Carroll, 1988; Clay, 1988; Schulz et al., 1999; Faeth and Fagan, 2002). Indeed, agronomic
grass species infected with systemic endophytes show striking toxic and noxious effects on
vertebrate and invertebrate herbivores and pathogens, purportedly resulting from production
of multiple alkaloids by endophytes (Siegel and Bush, 1996). Loline alkaloids, saturated 1-
aminopyrrolizidines with an oxygen bridge, were exclusively found in endophyte-infected
grasses, such as Festuca sp. infected with Neotyphodium sp. Recently, it was demonstrated
that N. uncinatum, the common endophyte of F. pratensis, had the full biosynthetic capacity
for some of the most common loline alkaloids (Blankenship et al., 2001). Lolines are potent
broad-spectrum insecticides, acting both as metabolic toxins and feeding deterrents depending
on the specific insect species. Unlike ergot and indole diterpene alkaloids, these loline
derivatives are much less toxic to mammals (Casabuono and Pomilio, 1997). Similarly,
endophytes of woody plants may provide a defensive role for the host plant because they
produce a wide array of mycotoxins and enzymes that can inhibit the growth of microbes and
invertebrate herbivores (Saikkonen et al., 1998; Tan and Zou, 2001).
Endophytes may also increase host fitness and competitive abilities, by increasing
nutrient uptake, germination success, resistance to drought and water stress, resistance to seed
predators, tolerance to heavy metal presence, tolerance to high salinity, and growth rate by
evolving biochemical pathways to produce plant growth hormones. For instance, the growth

2
Introduction

promoting phytohormone indole-3-acetic acid (IAA) was isolated from cultures of the fungal
endophytes Acremonium coenophialum, Aureobasidium pullulans, Epicoccum purpurascens
and Colletotrichum sp. Together with IAA and indole-3-acetonitrile, cytokinins were also
shown to be produced by an endophytic strain of Hypoxylon serpens (Tan and Zou, 2001). An
imaginable role of endophytes is furthermore to initiate the biological degradation of the dead
or dying host plant that begins the critical processes of nutrient recycling (Tan and Zou, 2001;
Strobel, 2002a; Zhang et al., 2006).
In return, plants provide spatial structure, protection from desiccation, nutrients,
photosynthate and, in the case of vertical-transmission, dissemination to the next generation
of hosts (Clay, 1988; Wolock-Madej and Clay, 1991; Knoch et al., 1993; Saikkonen et al.,
1998; Faeth and Fagan, 2002; Rudgers et al., 2004). It is also possible that the plant may
provide compounds critical for the completion of the life cycle of the endophyte or essential
for its growth or self-defense (Metz et al., 2000; Strobel, 2002a). However, in cases in which
herbivores facilitate spore or hyphal dispersal, nonsystemic endophyte interactions with their
host plants should fall near the antagonistic end of the interaction spectrum (Saikkonen et al.,
1998).
Recent studies suggested that plant and endophyte genotypic combinations together
with environmental conditions are an important source of variation in endophyte-plant
interactions (Faeth and Fagan, 2002). It would seem that many factors changing in the host as
related to the season, age, environment and location may influence the biology of the
endophyte (Strobel and Daisy, 2003).

1.3. Microbial biodiversity


Fungi make up one of the major clades of life. It had been estimated that
approximately 1.5 million fungal species are present on earth of which only about 7% have
been described so far (Hawksworth, 1991). Almost all vascular plant species examined to date
were found to harbor endophytes, thus they are presumably ubiquitous in the plant kingdom
(Tan and Zou, 2001). Because numerous new endophytic species may exist in plants, it
follows that endophytic microorganisms are important components of microbial biodiversity
(Clay, 1992). Ultimately, biological diversity implies chemical diversity because of the
constant chemical innovation that exists in ecosystems where the evolutionary race to survive
is the most active (Strobel and Daisy, 2003). Currently, it is hypothesized that ecology has a
major impact on the profiles of natural products in filamentous fungi. Temperature,
precipitation, humidity, length of season and other climatic factors affect the distribution of

3
Introduction

fungi. Moreover, diverse habitats, as tropical forests, the deep sea, sites of extreme
temperature, salinity or pH, often provide a source of novel microorganisms with the potential
for novel metabolic pathways and compounds (Larsen et al., 2005; Ebel, 2006). However,
temperate ecosystems, especially damp temperate regions, such as those of northern Europe,
eastern North America and North Africa are also rich in fungal diversity. They are generally
taken to have the "standard" fungus flora, i.e. the one first and best known (Bisby, 1943).
Even cold regions can be rich in fungal diversity as a number of these species have recently
been investigated and found to produce several bioactive metabolites (Larsen et al., 2005).
One of the most easily genetically transformable fungal species that has been studied
to date is Pestalotiopsis microspora. The fungus was found to be capable of adding telomeric
repeats to foreign DNA, a phenomenon unusual among fungi (Li et al., 1996). This finding
may have important implications in its biology since it explains at least one mechanism by
which new DNA can be captured by this organism and eventually expressed and replicated.
Such a mechanism may explain how the enormous biochemical variation may have arisen in
Pestalotiopsis microspora (Li et al., 1996). It is also a start in understanding how this fungus
adapts itself to the environment of the plant hosts and it suggests that the uptake of plant DNA
into the fungal genome may occur. In addition, the telomeric repeats have sequences very
similar to human telomeres, which points to the possibility that P. microspora could
conceivably serve as a means to construct artificial human chromosomes (Strobel, 2002a).

1.4. Plant selection for isolation of endophytes


Endophytes, by definition, live in close association with living plant tissues. In order
to acquire endophytes, host plant species should be selected that may be of interest because of
their unique biology, age, endemism, ethnobotanical history, or environmental setting. It
seems that endemic plants growing in moist, warm climates or in areas of great biodiversity
are among the first choices for study. It would appear that microbial competition in such an
area would be fierce given the abundance of both water and plants. As such, the number and
diversity of natural products produced by microbes surviving in such an area would be high.
Moreover, plants growing in harsh or extremely moist environments are sometimes prone to
attack by extremely pathogenic fungi and thus special defense mechanisms are necessary for
survival. Such disease defenses may be offered by the endophyte normally associated with the
plant (Strobel, 2002a, 2002b; Strobel and Daisy, 2003).

4
Introduction

1.5. The potential of natural products in drug discovery


Natural products are produced by all organisms but are mostly known from plants,
including algae, and microorganisms including fungi and prokaryotes. Most of these
organisms coexist in ecosystems and interact with each other in various ways in which often
chemistry plays a major role. It has been proposed that most secondary metabolites serve the
producing organisms by improving their survival fitness (Williams et al., 1989). On the
contrary to primary metabolites that are common in all living cells and are involved in the
formation of biomass and generation of energy, secondary metabolites are often only
produced by one or few species. Many are biologically active, and some of them have been
used by man for thousands of years as traditional medicines and as natural poisons (Larsen et
al., 2005).
From a pharmaceutical point of view, there is a growing need for new antibiotics,
chemotherapeutic agents, and agrochemicals that are highly effective, possess low toxicity,
and have a minor environmental impact. In fact, around 60% of the new drugs registered
during the period 1981-2002 by the FDA as anticancer, antimigraine and anti-hypertensive
agents were either natural products or based on them (Newman et al., 2003). Moreover, a
significant number of the top 35 worldwide selling drugs in the years 2000-2003 were natural
product-derived compounds (Butler, 2004). Natural products have been the traditional
pathfinder compounds, offering an untold diversity of chemical structures unparalleled by
even the largest combinatorial databases. In addition, natural products often serve as lead
structures whose activity can be enhanced by manipulation through combinatorial and
synthetic chemistry (Strobel and Daisy, 2003). Since there are still many unexplored
resources in nature, the potential for finding new organisms and thereby new metabolic
pathways is also enormous.

1.6. The potential of fungal natural products in drug discovery


It was not until Alexander Fleming discovered penicillin G from Penicillium notatum
almost 80 years ago (1928) that fungal microorganisms suddenly became a hunting ground for
novel drug leads (Strobel and Daisy, 2003; Larsen et al., 2005). Hence many pharmaceutical
companies were motivated to start sampling and screening large collections of fungal strains
especially for antibiotics (Butler, 2004). Microorganisms represented a promising rich source
of novel natural product leads having the advantage of feasible production of large quantities
with reasonable cost, by large scale cultivation and fermentation of the source organisms.
About 20 years later several other antibacterial agents such as cephalosporin C had been

5
Introduction

discovered (Newton and Abraham, 1955). Furthermore, griseofulvin was one of the first
antifungal natural products found in filamentous fungi (Grove et al., 1952). Recently,
echinocandin B and pneumocandin B, isolated from Aspergillus rugulovalvus and Glarea
lozoyensis, respectively, were the lead compounds and templates for the semisynthetic
antifungal drugs anidulafungin (Eraxis®) and caspofungin (Cancidas®) (Butler, 2004). In
addition, by the promising new screening strategy for antibiotics, aiming at inhibition of
biofilm formation by Gram-negative bacteria, the quorum sensing inhibitory activity of two
well known fungal mycotoxins, patulin and penicillic acid, isolated from Aspergillus and
Penicillium sp., was described (Rasmussen et al., 2005).
Furthermore, a new era in immunopharmacology began with the discovery of
cyclosporine, isolated from Tolypocladium inflatum, in 1971. It was the first
immunosuppressive drug that allowed selective immunoregulation of T cells without
excessive toxicity and was used as immunosuppressant during organ transplantations (Borel
and Kis, 1991; Butler, 2004). It is now widely exploited in organ and tissue transplant
surgery, to prevent rejection following bone marrow, kidney, liver and heart transplants. It has
revolutionized organ transplant surgery, substantially increasing survival rates in transplant
patients (Dewick, 2006). Another strongly immunosuppressive fungal metabolite that is used
for organ transplantations and for treatment of autoimmune diseases is mycophenolic acid
(Cellcept®, Myfortic®) (Bentley, 2000). This compound was produced by Penicillium,
Aspergillus, Byssochlamys and Septoria species (Larsen et al., 2005).
Another group of fungal derived drugs are the antilipidemic statin compounds. Statins
are the most potent cholesterol-lowering agents available. They are either fermentation-
derived for instance mevastatin and lovastatin (Mevacor®), from Penicillium citrinum and
Aspergillus terreus, respectively, or synthetic analogue compounds such as the major selling
synthetic statins (lipitor®, crestor® and livalo®). Statins lower cholesterol by reversible
competitive inhibition of the rate-limiting enzyme HMG-CoA reductase in the mevalonate
pathway of cholesterol biosynthesis, thus reducing total and low-density lipoprotein
cholesterol levels. As high blood cholesterol levels contribute to the incidence of coronary
heart disease, statins are of potential value in treating high-risk coronary patient (Butler, 2004;
Dewick, 2006). Two lipid-regulating drugs of this class, atorvastatin (lipitor®) and simvastatin
(Zocor®), feature prominently in the top ten drugs by cost reflecting the widespread
implementation of clinical guidelines and recommendations relating to coronary heart disease.
Traditionally, microorganisms were isolated from soil samples and explored for
pharmacologically active natural products which might prove to be suitable for specific

6
Introduction

medicinal or agrochemical applications. Extremely unusual and valuable organic substances


were sometimes produced by these organisms. Nowadays, investigations of soil fungi showed
a reduced hit-rate of novel compounds. Thus, in the search for new sources of therapeutic
agents, marine microorganisms and endophytic fungi associated with plants were found to be
a vast untapped reservoir of metabolic diversity producing a wide array of new biologically
active secondary metabolites.

H O
H H O
H HOOC N S
N
S

O N NH2 O N O O

O O
O
O
COOH COOH O
O
Cl
Penicillin G Cephalosporin C
Griseofulvin
HO
O O

O NH O HO N
H
N N
N N N

HO N O O O O
H O
O O O

HO
N OH H
N N
NH N N
H
N
H
O HO O
O O

HO HN O
N H OH O
N Cyclosporine O
O O HO
HO O

OH O

O
HOOC
Echinocandin B
O

Lovastatin
Mycophenolic acid

Figure 1.1: Fungal natural products as drugs or drug lead compounds.

1.7. Endophytic fungi as a source of bioactive natural products


There is growing evidence that bioactive substances produced by microbial
endophytes may not only be involved in the host-endophyte relationship, but may also
ultimately have applicability in medicine, agriculture and industry (Strobel, 2002a).
Additionally, it is of great relevance in this context that the number of secondary metabolites
produced by fungal endophytes is larger than that of any other endophytic microorganism
class (Zhang et al., 2006). Indeed, endophytic fungi are a very promising source of novel
biologically active compounds, and have proven to yield a considerable hit-rate of novel
compounds when screening larger strain numbers for biological activities (Schulz et al.,
2002). This may be the case because endophytes may have developed close biological

7
Introduction

associations with and inside their hosts, leading to the production of a high number and
diversity of classes of biological derived molecules with a range of biological activities. In
fact, a recent comprehensive study has indicated that 51% of biologically active substances
isolated from endophytic fungi were previously unknown (Stierle et al., 1999; Strobel, 2002b;
Weber et al., 2004; Shen et al., 2006). In the following part examples including novel
bioactive secondary metabolites from endophytic fungi are listed according to their
indications. So far, only a small percentage of these metabolites have been carried forward as
natural product drugs, nevertheless they represent interesting structures which indicate the
great chemical diversity and pharmaceutical potential of endophytic fungi as sources for novel
drug lead compounds.

1.7.1. Secondary metabolites from endophytes as antibiotics


Even though more than 30 000 diseases are clinically described today less than one-
third of these can be treated symptomatically and even a fewer can be cured. The increasing
occurrence of multiresistant pathogenic strains has limited the effect of traditional
antimicrobial treatment. Hence, there is an urgent need for new therapeutic agents with
infectious disease control (Strobel and Daisy, 2003; Larsen et al., 2005).
Guanacastepenes, exemplified by guanacastepene A, represent highly diverse
diterpenoids produced by an unidentified endophytic fungus isolated from Daphnopsis
americana tree. They exhibited pronounced antibiotic activity against drug-resistant strains of
Staphylococcus aureus and Enterococcus faecium (Brady et al., 2001). Chaetoglobosin A
and rhizotonic acid, from endophytic Chaetomium globosum, in Maytenus hookeri, and
Rhizoctonia sp., in Cynodon dactylon, respectively, were reported to be active against the
gastric ulcer involved bacterium Helicobacter pylori (Tikoo et al., 2000; Ma et al., 2004).
Moreover, altersetin purified from an endophytic Alternaria sp. displayed potent activity
against pathogenic Gram-positive bacteria (Hellwig et al., 2002).

1.7.2. Secondary metabolites from endophytes as antimycotic agents


Fungal infections are becoming an increasingly difficult problem as a result of the
AIDS epidemic and the increased numbers of patients with organ transplants whose immune
systems are weakened. Thus, new antimycotics are needed to combat these problems (Strobel,
2002a). A unique peptide antimycotic, termed cryptocandin A, was isolated and
characterized from Cryptosporiopsis quercina, endophytic in Tripterigeum wilfordii, a
medicinal plant belonging to the family Celastraceae that is native to Eurasia (Strobel et al.,

8
Introduction

1999). It is currently being considered by several companies for use against a number of fungi
causing diseases of skin and nails (Strobel, 2002a). Other fungal metabolites with promising
antifungal activity are ambuic acid, described recently from several isolates of P. microspora
found in many of the world’s rainforests (Li et al., 2001), as well as jesterone and
hydroxyjesterone from Pestalotiopsis jesteri, a newly described species of Pestalotiopsis (Li
and Strobel, 2001). Furthermore, a new pentaketide antifungal agent, CR377, was isolated
from the culture broth of an endophytic Fusarium sp., from the plant Selaginella pallescens
collected in Costa Rica, and showed potent activity against Candida albicans in agar diffusion
assays performed on fungal lawns (Brady and Clardy, 2000).

1.7.3. Secondary metabolites from endophytes as antiviral agents


The emergence of resistance and multi-resistance against available drugs, the side
effects and high cost of current therapies as well as the HIV/AIDS epidemic and AIDS-
associated opportunistic infections, such as cytomegalovirus and polyomavirus, made the
development of novel antiviral drugs a central priority.
Cytonic acids A and B were reported as human cytomegalovirus protease inhibitors
from the culture of the endophytic fungus Cytonaema sp. isolated from Quercus sp. (Guo et
al., 2000). In addition, the novel quinone-related metabolites, xanthoviridicatins E and F,
produced by an endophytic Penicillium chrysogenum colonizing an unidentified plant,
inhibited the cleavage reaction of HIV-1 integrase (Singh et al., 2003).

9
Introduction

OH
O
O OH O COOH OH
H HO
OH

O
NH
O
O
O
OH H
O O
O O

O NH

O
H Rhizotonic acid
NH H

Guanacastepene A Altersetin

OH Chaetoglobosin A O
HO
O

O
COOH HO
HO OH
O O O
H
H HO
N OH
H2N N H
H
O N
OH
OH
Jesterone
N O O

HO O O Ambuic acid O
OH NH R2
HO OH
HN R
R1 OH OH O
OH NH

O O
(CH2)14CH3 O O
O
COOH OH O
Cryptocandin A O O
O
R=OCH3, CH3
OH
OH
HO OH Xanthoviridicatins E and F
Cytonic acid A R1=Et, R2=H
Cytonic acid B R1=H, R2=Et

Cytonic acids A and B 23

Figure 1.2: Fungal natural products with antimicrobial activity.

1.7.4. Secondary metabolites from endophytes as anticancer agents


The discovery of the paclitaxel (taxol®) producing endophytic fungus Taxomyces
andreanae from Taxus brevifolia (Strobel et al., 1993; Stierle and Strobel, 1995) evoked the
interest in endophytes as potential new sources for therapeutic agents. This early work set the
stage for a more comprehensive examination of the ability of other Taxus species and other
plants to yield endophytes producing taxol. Taxol is the world’s first billion dollar anticancer
drug and is used to treat a number of other human tissue proliferating diseases as well
(Strobel, 2002a). The mode of action of paclitaxel is to preclude tubulin molecules from
depolymerizing during the processes of cell division (Schiff and Horowitz, 1980). In fact,
tubulin molecules in taxol-sensitive plant pathogenic fungi were found to be affected in the
same manner as human cancer cells, which indicated that taxol, in nature, may provide a
defensive role for the yew tree (Taxus sp.) from which it originates (Young et al., 1992).
Similarly, paclitaxel has been reported to induce a reversible polymerization of plant tubulin
into microtubules, albeit weakly when compared to that of mammalian tubulin (Morejohn and

10
Introduction

Fosket, 1984; Bokros et al., 1993). Further examination of the endophytes of T. wallichiana
yielded Pestalotiopsis microspora which was found to produce taxol as well.
By the finding that many other endophytic fungi such as P. microspora (Strobel et al.,
1996) and Periconia sp. (Li et al., 1998), residing in plants other than Taxus species were also
producing taxol, it appeared that fungi more commonly produce taxol than higher plants, and
the distribution of those fungi is worldwide and not confined to endophytes of yews. Thus, it
may be that taxol had its origin in certain fungi and ultimately, if there is lateral gene transfer,
it may have been in the direction of the microbe to the higher plant. Unfortunately, taxol
production upon fermentation by all endophytes investigated so far is only in the range of
submicrograms to micrograms per liter. Considerable efforts are being made to determine the
feasibility of producing taxol by fermentation, in much the same way as penicillin, which
would effectively reduce its market price (Strobel, 2002a; Strobel and Daisy, 2003).
Moreover, the cytotoxic plant alkaloid, camptothecin, originally described from
Camptotheca acuminate and Nothapodytes foetida, and undergoing clinical trials since 1992
as anticancer drug, was identified in cultures of Entrophospora infrequens endophytic in
Nothapodytes foetida (Amna et al., 2006). Another anticancer drug, which has been given in
chemotherapy treatment for some types of cancer including leukemia, lymphoma, breast and
lung cancer for many years, is the indole derivative vincristine. This drug, available under the
trade names Oncovin®, Vincasar®, and Vincrex®, was originally obtained from
Catharanthus roseus. Very recently, a Chinese group reported preliminary evidence that
vincristine might be produced by Fusarium oxysporum endophytic in the same plant (Zhang
et al., 2006).
On the other hand, endophytic fungi were found to produce interesting bioactive
metabolites not related to the natural products produced by their host plants. For example,
chaetomellic acids A and B, isolated from the culture of an endophytic Chaetomella acutisea,
were found to be specific inhibitors of farnesyl-protein transferase (Lingham et al., 1993; Ishii
et al., 2000). Inhibitors of this enzyme prevent posttranslational modification of Ras proteins,
which serve as central connectors between signals generated at the plasma membrane and
nuclear effectors, thus disrupting the Ras signaling pathway as well as Ras-dependent
proliferative activity in cancerous and precancerous lesions (Kelloff et al., 1997). A similar
activity was observed for the new metabolites preussomerin N1, palmarumycin CP4a, and
palmarumycin CP5 produced by an endophytic Coniothyrium sp. (Tan and Zou, 2001).
Moreover, microcarpalide, a microfilament disrupting agent with weak cytotoxicity to

11
Introduction

mammalian cells, was characterized from fermentation broths of an unidentified endophytic


fungus (Ratnayake et al., 2001).
A further example is the relatively large group of alkaloids known as cytochalasins.
Many of these compounds, possessing antitumor and antibiotic activities, were found in
endophytic fungi, but because of their cellular toxicity they have not been developed into
pharmaceuticals (Wagenaar et al., 2000). Chaetoglobosins are fungal metabolites belonging to
the family of cytochalasins. Some chaetoglobosins have been isolated recently from
endophytic Chaetomium globosum and were shown to exhibit cytotoxic activities against the
human nasopharyngeal epidermoid tumour KB cell line (Vesely et al., 1995; Zhang et al.,
2006).

1.7.5. Secondary metabolites from endophytes with further interesting pharmacological


activities
As mentioned above, immunosuppressive drugs are used today to prevent allograft
rejection in transplant patients, and in the future they could be used to treat autoimmune
diseases such a rheumatoid arthritis and insulin-dependent diabetes (Strobel and Daisy, 2003).
Interestingly, compounds showing immunosuppressive activity were also obtained from
endophytic fungi, for example subglutinols A and B, which are noncytotoxic diterpene
pyrones produced by Fusarium subglutinans, an endophyte of Triptergium wilfordii. In the
mixed lymphocyte reaction assay the subglutinols were roughly as potent as cyclosporine
(Lee et al., 1995b).
L-783,281, is a quinine produced by the plant associated fungus Pseudomassaria sp.
This compound was found to lower blood glucose level in diabetic mice. Thus, the compound
mimics the action of the polypeptide hormone insulin, and unlike insulin, it was not destroyed
by enzymes in the digestive tract and may be given orally (Chem. Eng. News, 2000).
Pestacin and isopestacin, were separated from Pestalotiopsis microspora associated
with Terminalia morobensis. The compounds were able to scavenge superoxide and hydroxyl
free radicals in solution. The antioxidant activity of pestacin is at least one order of magnitude
higher than that of trolox, a vitamin E derivative (Harper et al., 2003). Two cerebrosides with
xanthine oxidase inhibitory activity were identified from an endophytic Fusarium sp. (Shu et
al., 2004). Aurasperone A, from Aspergillus niger, an endophytic fungus obtained from
Cynodon dactylon, is also a xanthine oxidase inhibitor (Song et al., 2004).

12
Introduction

O O OH OH H
O
N
H
NH O O

N
N H
H H
O H O O
HO O O
O O
OH O OH
H
O O O
O
N
Paclitaxel H
O
Vincristine H O O
O OH

OH
Subglutinol A
HO
OH OH
H O OH
OH
OH H

O O O
O
OH

Pestacin Chaetomellic acid A


Microcarpalide

Figure 1.3: Fungal natural products with anticancer, immunosuppressive and antioxidant activities.

1.8. The potential of microbial natural products in agriculture


As the world becomes wary of ecological damage provoked by extensive use of
synthetic insecticides, natural product research continues for the discovery of powerful,
selective, and safe alternatives (Strobel and Daisy, 2003). Many synthetic agricultural agents
have been and currently are being targeted for removal from the market, because of profound
harmful effects on human health and environment. Thus, perhaps endophytic fungi could
serve as a reservoir of untapped biologically based compounds that may present alternative
ways to control farm pests and pathogens (Demain, 2000; Strobel, 2002a). One interesting
finding consisted in the discovery of peramine, which was toxic to insects without any
harmful impact on mammals. This secondary metabolite was characterized in cultures of
Neotyphodium coenophialum, N. lolli, Epichloë festucae and E. typhina associated with tall
fescue, ryegrass and other grasses (Dew et al., 1990). Nodulisporic acids were isolated from
a Nodulisporium sp. endophytic in Bontia daphnoides. They were found to exhibit potent
insecticidal properties against the larvae of the blowfly (Demain, 2000). Another endophytic
fungus, Muscodor vitigenus isolated from Paullina paullinioides, was found to yield
naphthalene as its major product. Heptelidic acid and hydroheptelidic acid, from Phyllosticta
sp. an endophytic fungus of Abies balsamea, have been shown to be toxic to spruce bud worm
(Choristoneura fumiferana) larvae (Calhoun et al., 1992).
Furthermore, several fungal metabolites were inhibitory to the growth of selected crop
phytopathogenic fungi. One example is the unique tetramic acid, known as cryptocin, which
was produced by Cryptosporiopsis quercina endophytic in the medicinal plant Tripterigeum

13
Introduction

wilfordii. It showed potent activity against Pyricularia oryzae, causal agent of rice blast, one
of the most important plant diseases on earth, and is currently being examined as a natural
chemical control agent for rice blast (Li et al., 2000). Some of the first reported
sesquiterpenes produced by fungal endophytes were chokols A-G. They were isolated from
an endophytic Epichloë typhina, from Phleum pretense, and were found to be fungitoxic to
the leaf spot disease pathogen Cladosporium phlei (Koshino et al., 1989).

H O
O H
O O O
N

N
N H COOH
NH H
H
OH COOH
HO

N NH2
H O

O Heptelidic acid
Peramine Nodulisporic acid A N
H OH

O
OH

H
O

HO

Chokol A H
Cryptocin

Figure 1.4: Fungal natural products with agricultural potential.

14
Introduction

1.9. Aim and scopes of the study


Being poorly investigated, endophytes are obviously a rich and reliable source of
bioactive and chemically novel compounds with huge medicinal and agricultural potential.
The aim of this study was the purification of endophytic fungal strains from Egyptian
medicinal plants, the isolation, characterization and structure elucidation of biologically
active secondary metabolites from the extracts of these endophytic fungal strains, and the
preliminary evaluation of their pharmaceutical potential. Four endophytic fungi, Alternaria
sp., Ampelomyces sp., Stemphylium botryosum and Chaetomium sp., were subjected as
biological sources of the study.
In order to isolate the secondary metabolites, the fungi were grown in static liquid
Wickerham medium as well as solid rice medium at room temperature. The cultures were
allowed to grow for 3-4 weeks, followed by harvesting and subsequent extraction with
organic solvents. The obtained raw extracts were then fractionated and separated using
various chromatographic techniques and their fractions were analysed by HPLC-DAD for
their purity and ESI-LC/MS for their molecular weight and fragmentation patterns. The pure
compounds were submitted to state-of-the-art one- and two-dimensional NMR techniques for
structure elucidation. In addition, selected compounds were derivatized in order to determine
their absolute stereochemistry.
Furthermore, fractions and pure compounds were subjected to selected bioassays to
determine their pharmaceutical potential. Thus, antimicrobial activity was studied using the
agar diffusion assay as well as the biofilm test, whereas cytotoxicity was studied in vitro using
mouse lymphoma (L5178Y) cell line. Moreover, fractions and pure compounds were also
tested for their protein kinase inhibitory activity. The latter three assays were conducted in
cooperation with Prof. U. Hentschel, Würzburg, Prof. W. E. G. Müller, Mainz, and
ProQinase, Freiburg, respectively.
Finally, extracts were prepared from the corresponding host plants and fractionated,
and the obtained fractions were analyzed by HPLC and LC/MS for the presence of the
identified fungal metabolites. The samples were then reanalyzed parallel to the pure
substances and retention times as well as MS/MS spectra were compared.

15
Materials and Methods

2. Materials and Methods

2.1. Materials

2.1.1. Biological materials

2.1.1.1. Plant material


Plant samples were collected from different areas in Alexandria, Egypt. Voucher
specimens were identified by Prof. Dr. Amin El-Sayed Ali, Department of Crops, Faculty of
Agriculture, Alexandria University, and Prof. Dr. Rafiq El-Gharib Mahmoud, Department of
Botany, Faculty of Science, Alexandria University. Small stem, leaf and flower pieces were
cut from the plants and placed in plastic bags after any excess moisture was removed. Every
attempt was made to store the materials at 4° C until isolation procedures could be instituted.

2.1.1.2. Pure fungal strains isolated from the collected plants


Table 2.1 shows a list of the endophytic fungal strains isolated from different organs of the
collected plant samples and their corresponding botanical sources.

Table 2.1: Pure fungal strains and their botanical sources


Fungal code Plant part Source
I7L1 leaf Chenopodium album
I7L2 (Amaranthaceae)
II2L1 leaf Polygonum senegalense
II2L2 (Polygonaceae)
II2L3
II2L4
II3F1 flower Solanum nigrum
II3F2 (Solanaceae)
II3F3
II3F4
II3F5
II3F6
II3S stem
III3S2 stem Plantago major
III3S3 (Plantaginaceae)
III3S4
III3L1 leaf
III3L2

16
Materials and Methods

Fungal code Plant part Source


IV16L leaf Euphorbia helioscopia
(Euphorbiaceae)
V2L leaf Otanthus maritimus
V2S1 stem (Asteraceae)
V2S2
VI1F1 flower Urospermum picroides
VI1F2 (Asteraceae)
VI1F3
VI1F4
VI1S Stem
VI1L leaf
VI2F1 flower Aegialophila cretica
VI2F2 (Asteraceae)
VI2S1 stem
VI2S2
VI2S3
VI2S4
VI2S5

2.1.2. Media

2.1.2.1. Composition of malt agar (MA) medium


MA medium was used for short term storage of fungal cultures or fresh seeding for
preparation of liquid cultures.
Agar-agar 15.0 g
Malt extract 15.0 g
Distilled water to 1000 mL
pH 7.4 - 7.8 (adjusted with NaOH/HCl)
For the isolation of endophytic fungi from plant tissues chloramphenicol or streptomycin (0.2
or 0.1 g, respectively) were added to the medium to suppress bacterial growth.

2.1.2.2. Composition of Wickerham medium for liquid cultures


Yeast extract 3.0 g
Malt extract 3.0 g
Peptone 5.0 g
Glucose 10.0 g
Distilled water to 1000 mL
pH 7.2 - 7.4 (adjusted with NaOH/HCl)

17
Materials and Methods

2.1.2.3. Composition of rice medium for solid cultures


Rice 100 g
Distilled water 100 mL
Water was added to the rice and kept overnight before autoclaving.

2.1.2.4. Composition of Luria Bertani (LB) medium


This medium was used to conduct antibacterial assays.
Peptone 10.0 g
Yeast extract 5.0 g
NaCl 10.0 g
Distilled water To 1000 mL
pH 7.0 (adjusted with NaOH/HCl)
To prepare the agar plates, 15.0 g agar were added to 1 L broth media.

2.1.2.5. Composition of yeast medium


This medium was used to perform bioassays using Saccharomyces cerevisiae.
Peptone 5.0 g
Yeast extract 3.0 g
Malt extract 3.0 g
Glucose 10.0 g
Distilled water To 1000 mL
To prepare the agar plates, 15.0 g agar were added to 1 L broth media.

2.1.2.6. Composition of fungal medium for bioassay


Mannitose 50.0 g
Saccharose 50.0 g
Succinic acid 5.4 g
Yeast extract 3.0 g
KH2PO4 0.1 g
MgSO4 0.3 g
FeSO4 10.0 mg
ZnSO4 10.0 mg
Distilled water To 1000 mL

18
Materials and Methods

pH 5.4 (adjusted with NaOH/HCl)

2.1.2.7. Composition of potato dextrose agar (PDA) medium for bioassay


Potato infusion (see below) 1000 mL
Dextrose 20.0 g
Agar 15.0 g
Potato infusion: The potatoes (200 g) were first washed and cut into small pieces, then boiled
in 1000 mL distilled water for 1 hour and filtered to get the potato
infusion.

2.1.2.8. Composition of trypticase soy broth (TSB)


Peptone from casein 17.0 g
Peptone from soymeal 3.0 g
Glucose 2.5 g
NaCl 5.0 g
K2HPO4 2.5 g
Distilled water To 1000 mL
pH 7.3 (adjusted with NaOH/HCl)

2.1.3. Chemicals

2.1.3.1. General laboratory chemicals


Anisaldehyde (4-methoxybenzaldehyde) Merck
(-)-2-Butanol Merck
Dimethylsulfoxide Merck
Formaldehyde Merck
L-(+)-Ascorbic acid Merck
Hydrochloric acid Merck
Potassium hydroxide Merck
Pyridine Merck
Concentrated sulphuric acid Merck
Trifloroacetic acid (TFA) Merck
Concentrated ammonia solution Fluka

19
Materials and Methods

Acetic anhydride Merck


Ortho-phosphoric acid 85% (p.a.) Merck
Sodium hydrogen carbonate Sigma
Trifluroacetic acid (TFA) Merck

2.1.3.2. Chemicals for culture media


Agar-agar Galke
Chloramphenicol Sigma
Glucose Caelo
Malt extract Merck
NaCl Merck
Peptone BD
Streptomycin Sigma
Yeast extract Sigma

2.1.3.3. Chemicals for agarose gel electrophoresis


Agarose Serva
TBE-buffer Merck
Ethidium bromide Serva
Standards NEB

2.1.4. Chromatography

2.1.4.1. Stationary phases


Pre-coated TLC plates, Silica Gel 60 F254, layer thickness 0.2 mm Merck
Silica Gel 60, 0.04 - 0.063 mm mesh size Merck
Pre-coated TLC plates , RP-18, F254 S, layer thickness 0.25 mm Merck
RP-18, 0.04 - 0.063 mm mesh size Merck
Sephadex LH 20, 0.25 - 0.1 mm mesh size Merck
Diaion HP20 Supelco

20
Materials and Methods

2.1.4.2. Spray reagents


The reagents were stored in amber-colored bottles and kept refrigerated until use. TLC
was used to monitor the identity of each of the fractions and the qualitative purity of the
isolated compounds. It was also utilized to optimize the solvent system that would be applied
for column chromatography.

Anisaldehyde/H2SO4 Spray Reagent


Methanol 85 mL
Glacial acetic acid 10 mL
Conc. H2SO4 5 mL (added slowly)
Anisaldehyde 0.5 mL

Vanillin/H2SO4 Spray Reagent


Methanol 85 mL
Conc. H2SO4 15 mL (added slowly)
Vanillin 1g

2.1.5. Solvents

2.1.5.1. General solvents


Acetone, acetonitrile, dichloromethane, ethanol, ethyl acetate, n-hexane and methanol
were used. The solvents were purchased from the Institute of Chemistry, University of
Duesseldorf. They were distilled before using and special grades were used for spectroscopic
measurements.

2.1.5.2. Solvents for HPLC


Acetonitrile LiChroSolv HPLC grade (Merck)
Methanol LiChroSolv HPLC grade (Merck)
Nanopure water distilled and heavy metals free water obtained by
passing distilled water through nano- and ion-
exchange filter cells (Barnstead, France)

21
Materials and Methods

2.1.5.3. Solvents for optical rotation


Chloroform Spectral grade (Sigma)
Methanol Spectral grade (Sigma)

Water Spectral grade (Fluka)

2.1.5.4. Solvents for NMR


Acetone-d6 Uvasol, Merck
Chloroform-d Uvasol, Merck
DMF-d7 Uvasol, Merck
DMSO-d6 Uvasol, Merck
Methanol-d4 Uvasol, Merck
Pyridine-d5 Uvasol, Merck

2.2. Methods

2.2.1. Purification of fungal strains


Plant materials were cut into small pieces, washed with sterilized demineralized water,
then thoroughly surface treated with 70% ethanol for 1-2 minutes and ultimately air dried
under a laminar flow hood. This is done in order to eliminate surface contaminating microbes.
With a sterile scalpel, outer tissues were removed from the plant samples and the inner tissues
were carefully dissected under sterile conditions and placed onto malt agar plates containing
antibiotic (see section 2.1.2.1). After 3-4 weeks of incubation at room temperature, hyphal
tips of the fungi were removed and transferred to fresh malt agar medium. Plates are prepared
in duplicates to eliminate the possibility of contamination. Pure strains were isolated by
repeated inoculation (see Figure 2.1).

2.2.2. Cultivation of pure fungal strains

2.2.2.1. Cultivation for short term storage


Fungi were grown on malt agar medium under room temperature for several days.
When fungal hyphae almost cover the surface of the MA plate, cultures were stored at 4º C
for a maximum period of 6 months, and then re-inoculated onto fresh MA media.

22
Materials and Methods

2.2.2.2. Cultivation for screening and isolation of secondary metabolites


Mass growth of pure fungi for screening as well as isolation and identification of
secondary metabolites was carried out by transferring fresh fungal cultures into Erlenmeyer
flasks (1L each) containing 300 mL of Wickerham medium for liquid cultures or 100 g rice
for solid cultures. The cultures were then incubated at room temperature (no shaking) for 21
and 30 days, respectively. Large scale cultivation was carried out using 30 and 10 1L
Erlenmeyer flasks for liquid and solid rice cultures, respectively.

Figure 2.1: Isolation, purification and cultivation of fungal strains

2.2.3. Extraction of fungal cultures and host plant material

2.2.3.1. Extraction of fungal liquid cultures

2.2.3.1.1. Total extraction of culture media and mycelia


250 mL EtOAc were added to each Erlenmeyer flask containing 300 mL culture
medium and left overnight to stop cell growth. Culture media and mycelia were then extracted
in the Ultraturrax for 10 min for cell destruction, followed by vacuum filtration using
Buchner. The mycelium residue was discarded while culture filtrates were collected and

23
Materials and Methods

extracted with EtOAc and n-BuOH till exhaustion. The combined EtOAc phases were washed
with distilled water and then taken to dryness. The dry residue was then partitioned between
n-hexane and 90% MeOH. The extraction scheme is described in Figure 2.2.

+ EtOAc
Cell suspension
Ultra Turrax
filter

Filtrate

EtOAc

Water phase EtOAc

n-BuOH evaporate

Water phase n-BuOH Residue

90% MeOH
+ n-Hexane

90% MeOH n-Hexane

Figure 2.2: Total extraction of culture media and mycelia

2.2.3.1.2. Separate extraction of culture media and mycelia


Fungal mycelia were seperated from culture media and left in MeOH overnight. Using
Ultraturrax cells were destructed and extracted for 10 min, followed by filtration and repeated
extraction till exhaustion. The culture media were extracted in the same manner as described
above in 2.2.3.1.2 to obtain the EtOAc extract. The extraction scheme is described in Figure
2.3.

24
Materials and Methods

Mycelium Medium

+ MeOH EtOAc
Ultra Turrax

Cell suspension
Water phase EtOAc ext.

filter
n-BuOH evaporate

MeOH ext.
Water phase n-BuOH ext. Residue
evaporate
90% MeOH
+ n-Hexane
Residue
90% MeOH fr. n-Hexane fr.
90% MeOH
+ n-Hexane

90% MeOH fr. n-Hexane fr.

Figure 2.3: Separate extraction of culture media and mycelia

2.2.3.2. Extraction of solid rice cultures


250 mL EtOAc were added to the cultures and left overnight. Culture media were then
cut in pieces to allow complete extraction and left for 3–5 days. Then filtration was done
followed by repeated extraction with EtOAc and MeOH till exhaustion. The combined EtOAc
phases were washed with distilled water and then taken to dryness. The dry residues obtained
from EtOAc and MeOH extracts were partitioned between n-hexane and 90% MeOH. The
extraction scheme is described in Figure 2.4.

25
Materials and Methods

1.
90%
+ EtOAc EtOAc evaporate MeOH fr.
ext. n-Hexane
filter + 90% MeOH
+ n-Hexane fr.
2.
90%
+ MeOH MeOH evaporate MeOH fr.
ext.
filter + 90% MeOH n-Hexane
+ n-Hexane fr.

Figure 2.4: Extraction of solid rice media

2.2.3.3. Extraction and fractionation of the plant material


The plant samples were frozen at - 80° C. The freeze dried samples were extracted
with 90% MeOH overnight with shaking and the resulting extracts were dried. The dried
residues were subjected to partitionation between n-hexane and 90% MeOH. The 90% MeOH
soluble fractions was fractionated over Diaion HP-20 using H2O:MeOH and MeOH:acetone
gradient elution and the obtained fractions were analysed by HPLC and LC/MS.

2.2.3.4. Solvent-solvent extraction


Solvent-solvent extraction is a widely employed technique to separate organic compounds
from a mixture. It involves the separation of compounds into two immiscible solvents. Since
the technique is based upon an unequal distribution of solutes between two solvents with
different polarities, the solutes will be more soluble in one solvent compared to the other. The
distribution of a component A between two phases can be expressed by the distribution
coefficient (K):
[A]top phase
K=
[A]lower phase
where, [A] is the concentration of solute A.
The following general principles should be considered in choosing the solvents:
- the solvents involved in the extraction must be immiscible
- the solvents must not react with the components that will be separated
- the solvents should be easily removed by evaporation after the process
In this study, solvent-solvent extraction was the first step in the separation process. It was
meant to “clean” the ethyl acetate extract from salts and other undesirable polar constituents

26
Materials and Methods

by water-ethyl acetate extraction. Subsequently, the methanol-n-hexane extraction was


applied to remove fatty acids and other undesirable non polar components.

2.2.4. Identification of fungal strains and their taxonomy

2.2.4.1. Fungal identification


Fungal strains were identified using a molecular biological protocol by DNA
amplification and sequencing of the internal transcribed spacer (ITS) region. This was carried
out by Ine Dewi Indriani and Arnulf Diesel at the Institut für Pharmazeutische Biologie und
Biotechnologie, Heinrich-Heine-Universität Düsseldorf.

DNA isolation
Fungal DNA isolation and purification was performed using DNeasy® Plant Mini Kit
(QIAgen). The lyophilized fungal mycelia were pulverized and disrupted with the help of
glass beads. Then cell lysis was carried out by addition of lysis Buffer AP-1 and RNAse-A
solution followed by incubation of the mixture at 65º C. The remaining detergent, protein and
polysaccharide were precipitated by addition of Buffer AP-2 to the lysate. The lysate was then
applied to the Qiashredder™ Mini Spin Column and centrifuged to remove the cell debris and
other remaining precipitates. The lysate was then transferred to a new tube.
An adequate volume of ethanolic Buffer AP3/E was added to the lysate and the
mixture was then applied to DNeasy Mini Spin Column. After centrifugation, the filtrate was
discarded. The column was washed by addition of ethanolic Buffer AW followed by
centrifugation. Another portion of Buffer AW was added to the column and centrifuged at
maximum speed to dry the membrane in the column from residual ethanol.
Fungal DNA, which is incorporated into the membrane, was eluted by addition of
Buffer AE directly to the membrane in the DNeasy column. The column was then incubated
at room temperature for 5 minutes and then centrifuged to collect the filtrate, which was the
fungal DNA dissolved in Buffer AE.

DNA amplification
The isolated DNA was then amplified by Polymerase Chain Reaction (PCR). The PCR
was carried out using HotStarTaq Master Mix Kit (QIAgen). The Master Mix contains
HotStarTaq®DNA Polymerase, PCR buffer (with MgCl2) and dNTPs.

27
Materials and Methods

ITS 1 (with base sequences TCCGTAGGTGAACCTGCGG) and ITS 4 (with base


sequences TCCTCCGCTTATTGATATGC) (Invitrogen), as primers, were mixed with
HotstarTaq Master Mix Kit and DNA template. Thus, each PCR reaction mixture contained 5-
10 ng of genomic DNA, 1 µM each of the primers ITS 1 and ITS 4, and 1 U of Hot start Taq-
Polymerase (Invitrogen) in a total volume of 50 µL. The mixture was then applied to the
thermal cycler (BioRad) using the programmed PCR cycle as outlined below:
- Initial activation step in 95º C for 15 minutes to activate HotStarTaq®DNA Polymerase
- Cycling steps which were repeated 35 times:
Denaturing: 1 minute at 95º C, annealing: 1 minute at 56º C, extension: 1
minute at 72º C
- Final extension for 10 minutes in 72º C

Purification of PCR products and DNA sequencing


The PCR product was purified using 2% Agarose-Gel-Electrophoresis at 75 V for 60
minutes in TBE buffer. The agarose gel was then stained using 1% ethidium bromide. A 500
bp stained DNA fragment was then excised from the agarose gel. The next step of PCR
product purification was performed using Perfectprep® Gel Cleanup Kit (Eppendorf). The
binding buffer was mixed to the PCR product and incubated at 50º C for 10 minutes in an
eppendorf thermomixer at 1000 rpm. The mixture was mixed with a volume of isopropanol
and then centrifuged. The filtrate was discarded and the column was washed with wash buffer
twice followed by centrifugation.
Amplified fungal DNA (PCR product), which was incorporated into the column, was
eluted by addition of elution buffer or molecular biology grade water to the centre of the
column. The column was then centrifuged to collect the filtrate, which was the fungal DNA
dissolved in elution buffer. The amplified fungal DNA was then submitted for sequencing by
a commercial service and the base sequence was compared with publicly available databases
such as GenBank with the help of Blast-Algorithmus.

2.2.4.2. Taxonomy

Alternaria sp.
The fungus Alternaria sp. was isolated from fresh leaves of wildly growing
Polygonum senegalense (Polygonaceae) (see Figure 2.5). The plant was collected in April
2004 from Alexandria, Egypt.

28
Materials and Methods

Taxonomy
Phylum Ascomycota;
Subphylum Pezizomycotina;
Class Dothideomycetes;
Order Pleosporales;
Family Pleosporaceae;
Genus Alternaria;
Species Alternaria sp.

A B

C D

Figure 2.5: Alternaria sp. (A: Polygonum senegalense. B: Pure strain on malt agar plate. C: Liquid
culture in Wickerham medium. D: Rice culture).

29
Materials and Methods

Ampelomyces sp.
The fungus Ampelomyces sp. was isolated from fresh flowers of wildly growing
Urospermum picroides (Asteraceae) (see Figure 2.6). The plant was collected in April 2004
from Alexandria, Egypt.

Taxonomy
Phylum Ascomycota;
Subphylum Pezizomycotina;
Class Dothideomycetes;
Order Pleosporales;
Family Leptosphaeriaceae;
Genus Ampelomyces;
Species Ampelomyces sp.

A B

C D

Figure 2.6: Ampelomyces sp. (A: Urospermum picroides. B: Pure strain on malt agar plate. C: Liquid
culture in Wickerham medium. D: Rice culture).

30
Materials and Methods

Stemphylium botryosum
The fungus Stemphylium botryosum was isolated from fresh leaves of wildly growing
Chenopodium album (Amaranthaceae) (see Figure 2.7). The plant was collected in April 2004
from Alexandria, Egypt.

Taxonomy
Phylum Ascomycota;
Subphylum Pezizomycotina;
Class Dothideomycetes;
Order Pleosporales;
Family Pleosporaceae;
Genus Stemphylium;
Species Stemphylium botryosum.

A
B

C D

Figure 2.7: Stemphylium botryosum. (A: Chenopodium album. B: Pure strain on malt agar plate. C:
Liquid culture in Wickerham medium. D: Rice culture).

31
Materials and Methods

Chaetomium sp.
The fungus Chaetomium sp. was isolated from fresh stems of wildly growing Otanthus
maritimus (Asteraceae) (see Figure 2.8). The plant was collected in April 2004 from
Alexandria, Egypt.

Taxonomy
Phylum Ascomycota;
Subphylum Pezizomycotina;
Class Sordariomycetes;
Order Sordariales;
Family Chaetomiaceae;
Genus Chaetomium;
Species Chaetomium sp.

A
B

C D

Figure 2.8: Chaetomium sp. (A: Otanthus maritimus. B: Pure strain on malt agar plate. C: Liquid
culture in Wickerham medium. D: Rice culture).

32
Materials and Methods

2.2.5. Isolation and purification of secondary metabolites

2.2.5.1. Isolation of the secondary metabolites from Alternaria sp.

2.2.5.1.1. Secondary metabolites isolated from liquid cultures of Alternaria sp.

n-BuOH
extract
Sephadex
100% MeOH

Tenuazonic Fraction Fraction Fraction


acid 3 5 7
27.5 mg

Fraction Alternariol Alternariol Adenosine


2 monomethyl- sulphate 3.0 mg
ether sulphate 60.9 mg
5.9 mg

EtOAc
extract

Solvent fractionation

90% MeOH n-Hexane


extract extract
Sephadex
100% MeOH

Tenuazonic Fraction Fraction Fraction Fraction


acid 3 5 7 9
101.2 mg

Fraction Altenusin Alternariol Alternariol


2 24.6 mg mono- 189.2 mg
methylether
Semipreparative HPLC Semipreparative HPLC 14.1 mg

2,5-Dimethyl-7- Altertoxin I
hydroxychromone 3.5 mg
4.6 mg

33
Materials and Methods

2.2.5.1.2. Secondary metabolites isolated from rice cultures of Alternaria sp.

EtOAc
extract
Solvent fractionation

90% MeOH n-Hexane


fraction fraction
VLC
DCM:MeOH

Fraction Fraction Fraction Altenusin Fraction


1 3 5 353.0 9

Alternariol Fraction Fraction Fraction Fraction


mono- 4 6 8 10
methylether Sephadex
221.8 mg Preparative HPLC Preparative HPLC Preparative HPLC 100% MeOH
Talaroflavone Altenuene Desmethyl- Fraction
5.0 mg and 4`-epi- altenusin 9.1
altenuene 5.6 mg
18.8 mg
Alterlactone Alteric acid
8.4 mg Altenusin 16.9 mg
Alternariol 8.1 mg
6.0 mg Fraction
Alternariol
8.9 mg 9.3

Hydroxy-
alternariol
mono-
methylether
5.8 mg

Alternariol
mono-
methylether
3.1 mg

34
Materials and Methods

2.2.5.2. Isolation of the secondary metabolites from Ampelomyces sp.

2.2.5.2.1. Secondary metabolites isolated from liquid cultures of Ampelomyces sp.

MeOH
extract

Solvent fractionation

n-Hexane DCM EtOAc Aquous


fraction fraction fraction fraction
Silica
Sephadex
Hexane:EtOAc Sephadex 100% MeOH
100% MeOH
Ergosterol
23.5 mg Fraction
1
Fraction Fraction Fraction
1 2 3
Methylalaternin
sulphate
Silica
1.0 mg
EtOAc:MeOH:H2O
Cerebroside C
23.4 mg Macrosporin
sulphate
1.5 mg

Fraction
4

35
Materials and Methods

EtOAc
extract
VLC
n-Hexane:EtOAc:MeOH

Fraction Fraction Fraction Fraction


1 3 5 7

Fraction Fraction Fraction


2 4 6

Sephadex
50% MeOH:DCM Preparative HPLC

Methyltri- Fraction
acetic lactone 4.3
4.2 mg

Ampelopyrone
Semipreparative
2.1 mg
HPLC
Fraction Macrosporin
2.1 6.2 mg

Citreoiso-
Methyl- Fraction coumarin
alaternin 2.4 0.5 mg
0.5 mg

36
Materials and Methods

2.2.5.2.2. Secondary metabolites isolated from rice cultures of Ampelomyces sp.

EtOAc
extract
Solvent fractionation

90% MeOH n-Hexane


fraction fraction
VLC
Hexane:EtOAc:DCM:MeOH

Fraction Fraction Fraction Fraction Fraction


1 3 5 7 9

Macrosporin Methyl- Fraction Fraction Fraction


15.4 mg alaternin 6 8 10
3.0 mg Sephadex
100% MeOH
Fraction Ampelanol
Fraction Fraction Fraction Fraction 8.4 mg
9.2
6.1 6.3 6.5 6.7
Fraction Fraction Fraction
Fraction Fraction Fraction 9.1 9.3
6.4 9.5
6.2 6.6 Preparative
Semi- Sephadex
100% MeOH HPLC
preparative
HPLC
Alterporriol D Alterporriol E
Desmethyl- 10.5 mg 12.3 mg
diaportinol Semi-
Preparative 1.5 mg preparative
HPLC HPLC Fraction Fraction Fraction
8.1 8.3 8.5
Altersolanol A Desmethyl-
15.7 mg dichloro- Fraction Fraction
diaportin 8.2 8.4
1.0 mg Preparativ Semipreparative
e HPLC HPLC

Ampelanol Altersolanol J Methyl- Macrosporin


10.2 mg 1.8 mg alaternin sulphate
sulphate 5.6 mg
1.5 mg

37
Materials and Methods

2.2.5.3. Isolation of the secondary metabolites from Stemphylium botryosum

Secondary metabolites isolated from rice cultures of Stemphylium botryosum

EtOAc
extract

Solvent fractionation

90% MeOH n-Hexane


fraction fraction
VLC
DCM:MeOH

Macrosporin Fraction Fraction Altersolanol A


3.7 mg 3 5 3.3 mg

Fraction Fraction Fraction Fraction


2 4 6 8

Preparative Preparative Preparative HPLC


HPLC HPLC
Tetrahydro-
Curvularin altersolanol B
1.9 mg 5.6 mg

Stemphyperylenol Dehydro-
3.9 mg curvularin
15.5 mg

38
Materials and Methods

2.2.5.4. Isolation of the secondary metabolites from Chaetomium sp.

Secondary metabolites isolated from liquid cultures of Chaetomium sp.

EtOAc
extract

Solvent fractionation

90% MeOH n-Hexane


fraction fraction
Sephadex
100% MeOH

Fraction Fraction Fraction Fraction Fraction


1 3 5 7 9

Fraction Orsellinic Indol- Fraction


2 acid carboxylic 8
Semipreparative 49.6 mg acid
Preparative
HPLC 20.1 mg HPLC

Cycloalanyl- Aureonitolic Cochliodinol Isocochliodinol


tryptophan acid 55.7 mg 8.8 mg
5.4 mg 1.7 mg

2.2.5.5. Chromatographic methods

2.2.5.5.1. Thin layer chromatography (TLC)


Chromatography refers to any separation method in which the components are
distributed between stationary phase and mobile phase. The separation occurs because sample
components have different affinities for the stationary and mobile phases and therefore move
at different rates along the TLC plates and the column. TLC was performed on pre-coated
TLC plates with silica gel 60 F254 (layer thickness 0.2 mm, E. Merck, Darmstadt, Germany)
with the following eluents:

39
Materials and Methods

For polar compounds EtOAc:MeOH:H2O (30:5:4, 30:6:5 and 30:7:6)


For semi-polar compounds DCM:MeOH (95:5, 90:10, 85:15, 80:20 and 70:30)
DCM:MeOH:EtOAc (90:10:5 and 80:20:10)
For non-polar compounds n-Hexane:EtOAc (95:5, 90:10, 85:15, 80:20 and 70:30)
n-Hexane:MeOH (95:5 and 90:10)
TLC on reversed phase RP18 F254 (layer thickness 0.25 mm, Merck, Darmstadt,
Germany) was used for polar substances and using the different solvent systems of
MeOH:H2O (90:10, 80:20, 70:30 and 60:40). The band separation on TLC was detected under
UV lamp at 254 and 366 nm, followed by spraying the TLC plates with anisaldehyde/H2SO4
or vaniline/H2SO4 reagent and subsequent heating at 110 °C.

2.2.5.5.2. Vacuum liquid chromatography (VLC)


Vacuum liquid chromatography is a useful method as an initial isolation procedure for
large amounts of sample. The apparatus consists of a 500 cm sintered glass filter funnel with
an inner diameter of 12 cm. Silica gel 60 was packed to a hard cake at a height of 5-10 cm
under applied vacuum. The sample used was adsorbed onto a small amount of silica gel using
volatile solvents. The resulting sample mixture was then packed onto the top of the column.
Using step gradient elution with non-polar solvent (e.g. n-Hexane or DCM) and increasing
amounts of polar solvent (e.g. EtOAc or MeOH) successive fractions were collected. The
flow was produced by vacuum and the column was allowed to run dry after each fraction
collected.

2.2.5.5.3. Column chromatography


Fractions derived from VLC were subjected to repeated separation through column
chromatography using appropriate stationary and mobile phase solvent systems previously
determined by TLC. The following separation systems were used:
I. Normal phase chromatography using a polar stationary phase, typically silica gel, in
conjunction with a non-polar mobile phase (e.g. n-Hexane, DCM) with gradually
increasing amounts of a polar solvent (e.g. EtOAc or MeOH). Thus hydrophobic
compounds elute more quickly than do hydrophilic compounds.
II. Reversed phase (RP) chromatography using a non polar stationary phase and a polar
mobile phase (e.g. H2O, MeOH). The stationary phase consists of silica packed with n-
alkyl chains covalently bound. For instance, C-8 signifies an octanyl chain and C-18 an
octadecyl ligand in the matrix. The more hydrophobic the matrix on each ligand, the

40
Materials and Methods

greater the tendency of the column to retain hydrophobic moieties. Thus hydrophilic
compounds elute more quickly than do hydrophobic compounds. Elution was performed
using H2O with gradually increasing amounts of MeOH.
III. Size exclusion chromatography involves separations based on molecular size of
compounds being analyzed. The stationary phase consists of porous beads (Sephadex
LH-20). The larger compounds will be excluded from the interior of the bead and thus
will elute first. The smaller compounds will be allowed to enter the beads and elute
according to their ability to exit from the small sized pores they were internalized
through. Elution was performed using MeOH or MeOH:DCM (1:1).
IV. Ion exclusion chromatography uses ion exchange resin beds (Diaion HP-20) that act as a
charged solid separation medium. The components of the processed sample have
different electrical affinities to this medium and are, as a result, differently retained by
the resins due to these different affinities. Therefore, by elution, these components can
be recovered separately at the outlet of the resins bed. Elution was performed using H2O
with gradually increasing amounts of MeOH and acetone.

2.2.5.5.4. Flash chromatography


Flash chromatography is a preparative column chromatography based on optimized
prepacked columns and an air pressure driven eluent at a high flow rate. It is a simple and
quick technique widely used to separate a variety of organic compounds. Normally, the
columns are dry Silica Gel 60 GF254 pre-packed, of 18 cm height, vertically clamped and
assembled in the system. The column is filled and saturated with the desired mobile phase just
prior to sample loading. Samples are dissolved in a small volume of the initial solvent used
and the resulting mixture was then packed onto the top of the column using special syringe.
The mobile phase (isocratic or gradient elution) is then pumped through the column with the
help of air pressure resulting in sample separation. This technique is considered as a low to
medium pressure technique and is applied to samples from few milligrams to some gram of
sample.

2.2.5.5.5. Preparative high pressure liquid chromatography (HPLC)


This process was used for isolation and purification of compounds from fractions
previously separated using column chromatographic separation. The most appropriate solvent
systems were determined before running the HPLC separation. The mobile phase
combination was MeOH or acetonitrile and nanopure H2O with or without 0.01 % TFA or 0.1

41
Materials and Methods

% formic acid, pumped in gradient or isocratic manner depending on the compounds retention
time. Each injection consisted of 20-80 mg of the fraction dissolved in 400 mL of the solvent
system. The solvent system was pumped through the column at a rate of 20 mL/min. The
eluted peaks were detected by the online UV detector and collected separately in Erlenmeyer
flasks.
Preparative HPLC system specifications are described as follows:
Pump Varian, PrepStar 218
Detector Varian, ProStar 320 UV-Vis detector
HPLC Program Varian Star (V. 6)

Column Varian Dynamax (250 × 4.6 mm, ID and 250 × 21.4


mm, ID), pre-packed with Microsorb 60-8 C18, with
integrated pre-column.

2.2.5.5.6. Semi-preparative high pressure liquid chromatography (HPLC)


This process was used for purification of compounds from fractions previously
separated using column chromatographic separation. The most appropriate solvent system
was determined before running the HPLC separation. The mobile phase combination was
MeOH and nanopure H2O with or without 0.01 % TFA or 0.1 % formic acid, pumped in
gradient or isocratic manner depending on the compounds retention time. Each injection
consisted of 1-3 mg of the fraction dissolved in 1 mL of the solvent system. The solvent
system was pumped through the column at a rate of 5 mL/min. The eluted peaks were
detected by the online UV detector and collected separately in Erlenmeyer flasks. The
separation column (125 × 4 mm, ID) was pre-filled with Eurospher C18 (Knauer, Berlin,
Germany).
Semi-preparative HPLC system specifications are described as follows:
Pump Merck Hitachi L-7100
Detector Merck Hitachi UV detector L-7400
Column Knauer (300 × 8 mm, ID), prepacked with Eurosphere
100-10 C18, with integrated pre-column.

42
Materials and Methods

2.2.5.5.7. Analytical high pressure liquid chromatography (HPLC)


Analytical HPLC was used to identify the distribution of peaks either from extracts or
fractions, as well as to evaluate the purity of isolated compounds. The solvent gradient used
started with MeOH:nanopure H2O (10:90), adjusted to pH 2 with phosphoric acid, and
reached to 100 % MeOH in 35 minutes. The autosampler injected 20 µL sample. All peaks
were detected by UV-VIS photodiode array detector. In some cases, special programs were
used. HPLC instrument consists of the pump, the detector, the injector, the separation column
and the reservoir of mobile phase. The separation column (125 × 2 mm, ID) was pre-filled
with Eurospher-100 C18 (5 µm), with integrated pre-column (Knauer, Berlin, Germany).
LC/UV system specifications are described as follows:
Pump Dionex P580A LPG
Detector Dionex Photodiode Array Detector UVD 340S
Column thermostat STH 585
Autosampler ASI-100T
HPLC Program Chromeleon (V. 6.3)
Column Knauer (125 × 4 mm, ID), pre-packed with Eurosphere
100-5 C18, with integrated pre-column.

2.2.6. Structure elucidation of the isolated secondary metabolites

2.2.6.1. Mass spectrometry (MS)


Mass spectrometers use the difference in mass-to-charge ratio (m/z) of ionized
molecules to separate them from each other. Mass spectrometry is therefore useful for
quantification of atoms or molecules and also for determination of chemical and structural
information of molecules. A mass spectrometer consists of an ion source, ion detector and
mass-selective analyzer. The output of mass spectrometers shows a plot of relative intensity
vs. the mass-to-charge ratio (m/z).

2.2.6.1.1. Electrospray ionization mass spectrometry (ESI-MS)


A mass spectrometer is an analytical instrument used to determine the molecular
weight of a compound. Basically, mass spectrometers are divided into three parts; ionization
source, analyzer and detector, which should be maintained under high vacuum conditions in
order to maintain the ions travel through the instrument without any hindrance from air
molecules. Once a sample was injected into the ionization source, the molecules are ionized.

43
Materials and Methods

The ions were then passed and extracted into the analyzer. In the analyzer, the ions were
separated according to their mass (m) to charge (z) ratio (m/z). Once the separated ions flow
into the detector, the signals are transmitted to the data system where the mass spectrum is
recorded.

Liquid chromatography mass spectrometry (LC/MS)


High pressure liquid chromatography is a powerful method for the separation of
complex mixtures, especially when many of the components may have similar polarities. If a
mass spectrum of each component can be recorded as it elutes from the LC column, quick
characterization of the components is greatly facilitated. Usually, ESI-MS is interfaced with
LC to make an effective on-line LC/MS. HPLC/ESI-MS was carried out using a Finnigan
LCQ-DECA mass spectrometer connected to a UV detector. The samples were dissolved in
water/MeOH mixtures and injected to HPLC/ESI-MS set-up. For standard MS/MS
measurements, a solvent gradient that started with acetonitrile:nanopure H2O (10:90),
adjusted with 0.1 % HCOOH, and reached to 100 % acetonitrile in 35 minutes was used.
LC/UV/MS system specifications are described as follows:
HPLC system Agilent 1100 series (pump, detector and autosampler)
Finnigan LC Q-DECA
MS spectrometer Knauer, (250 × 2 mm, ID), prepacked with Eurosphere 100-5
Column C18, with integrated pre-column.

2.2.6.1.2. Electron impact mass spectrometry (EI-MS)


Analysis involves vaporizing a compound in an evacuated chamber and then
bombarding it with electrons having 25.80 eV (2.4-7.6 MJ/mol) of energy. The high energy
electron stream not only ionizes an organic molecule (requiring about 7-10 eV) but also
causes extensive fragmentation (the strongest single bonds in organic molecules have
strengths of about 4 eV). The advantage is that fragmentation is extensive, giving rise to a
pattern of fragment ions which can help to characterize the compound. The disadvantage is
the frequent absence of a molecular ion.
Low resolution EI-MS was measured on a Finnigan MAT 8430 mass spectrometer.
Measurements were done by Dr. Peter Tommes, Institut für Anorganische and
Strukturchemie, Heinrich-Heine Universität, Düsseldorf.

44
Materials and Methods

2.2.6.1.3. Fast atom bombardment mass spectrometry (FAB-MS)


This was the first widely accepted method that employs energy sudden ionization.
FAB is useful for compounds, especially polar molecules, unresponsive to either EI or CI
mass spectrometry. It enables both non-volatile and high molecular weight compounds to be
analyzed. In this technique, a sample is dissolved or dispersed in a polar and relatively non-
volatile liquid matrix, introduced into the source on a copper probe tip. Then, this matrix is
bombarded with a beam of atoms of about 8 Kev. It uses a beam of neutral gas (Ar or Xe
atoms) and both positive and negative ion FAB spectra can be obtained.
Low resolution FAB-MS was measured on a Finnigan MAT 8430 mass spectrometer.
Measurements were done by Dr. Peter Tommes, Institut für Anorganische and
Strukturchemie, Heinrich-Heine Universität, Düsseldorf.

2.2.6.1.4. High resolution mass spectrometry (HR-MS)


High resolution is achieved by passing the ion beam through an electrostatic analyzer
before it enters the magnetic sector. In such a double focusing mass spectrometer, ion masses
can be measured with an accuracy of about 1 ppm. With measurement of this accuracy, the
atomic composition of the molecular ions can be determined.
HRESI-MS was measured on a Micromass Qtof 2 mass spectrometer at Helmholtz
Centre for Infection Research, Braunschweig. The time-of-flight analyzer separates ions
according to their mass-to-charge ratios (m/z) by measuring the time it takes for ions to travel
through a field free region known as the flight.

2.2.6.2. Nuclear magnetic resonance spectroscopy (NMR)


Nuclear magnetic resonance is a phenomenon which occurs when the nuclei of certain
atoms are immersed in a static magnetic field and exposed to a second oscillating magnetic
field. Some nuclei experience this phenomenon, and others do not, dependent upon whether
they possess a property called spin. It is used to study physical, chemical, and biological
properties of matter. As a consequence, NMR spectroscopy finds applications in several areas
of science. NMR spectroscopy is routinely used by chemists to study chemical structure using
simple one dimensional technique. Two dimensional techniques are used to determine the
structure of more complicated molecules.
NMR spectra were recorded at 300º K on a Bruker ARX-500 by Dr. Peter Tommes,
Institut für Anorganische und Strukturchemie, Heinrich-Heine Universität, Düsseldorf. Some
measurements were also performed at the Helmholtz Centre for Infection Research,

45
Materials and Methods

Braunschweig, by Dr. Victor Wray using an AVANCE DMX-600 NMR spectrometer. All 1D
and 2D spectra were obtained using the standard Bruker software. The samples were
dissolved in different solvents, the choice of which was dependent on the solubility of the
samples. Residual solvent signals were used as internal standards (reference signal). The
observed chemical shift (δ) values were given in ppm and the coupling constants (J) in Hz.

2.2.6.3. Optical activity


Optically active compounds contain at least one chiral centre. Optical activity is a
microscopic property of a collection of these molecules that arises from the way they interact
with light. Optical rotation was determined on a Perkin-Elmer-241 MC polarimeter. The
substance was stored in a 0.5 mL cuvette with 0.1 dm length. The angle of rotation was
measured at the wavelength of 546 and 579 nm of a mercury vapour lamp at room
temperature (25º C). The specific optical rotation was calculated using the expression:
[α]579 × 3.199
T
[α]D =
[α]579
4.199 -
[α]546

T
With [α]D = the specific rotation at the wavelength of the sodium D-line, 589 nm, at certain
temperature T.
[α]579 and [α]546 = the optical rotation at wavelengths 579 and 546 nm, respectively,
calculated using the formula:
100 α
[α]λ =
l×c
Where α = the measured angle of rotation in degrees,
l = the length in dm of the polarimeter tube,
c = the concentration of the substance expressed in g/100 mL.

2.2.6.4. Determination of absolute stereochemistry by Mosher reaction


The reaction was performed according to a modified Mosher ester procedure described
by Su et al. (Ohtani et al., 1991; Su et al., 2002).

Reaction with (R)-(-)-α-(trifluoromethyl) phenylacetyl chloride


The compounds (1 mg of each) were transferred into NMR tubes and were dried under
vacuum. Deuterated pyridine (0.5 mL) and (R)-MTPA chloride were added into the NMR

46
Materials and Methods

tube immediately under a N2 gas stream. The reagent was added in the ratio of 0.14 mM
reagent to 0.10 mM of the compound (Dale and Mosher, 1973). The NMR tubes were shaken
carefully to mix the samples and MTPA chloride evenly. The reaction NMR tubes were
permitted to stand at room temperature and monitored by 1H-NMR until the reaction was
found to be complete. 1H-1H COSY was measured to confirm the assignment of the signals.

Reaction with (S)-MTPA chloride


Another portion of each compound (1 mg) was transferred into NMR tube. The
reaction was performed in the same manner as described before to yield the (S)-MTPA ester.

2.2.7. Testing the biological activity


Finding biologically important compounds from endophytic fungi is only achieved if,
and when, assay systems have been devised that will allow for successful biologically guided
fractionation of the culture extracts.

2.2.7.1. Antimicrobial assay

2.2.7.1.1. Agar diffusion assay


This method was used to detect the capability of a substance to inhibit the growth of
microorganisms by measuring the diameter of inhibition zone around a tested compound on
agar plate. The agar diffusion assay was performed according to the Bauer-Kirby-Test (Bauer
et al., 1966).

Microorganisms
Crude extracts and isolated pure compounds were tested for activity against the
following standard strains: Gram-positive bacteria Bacillus subtilis,
Gram-negative bacteria Escherichia coli,
yeast Saccharomyces cerevisiae,
and the fungi Cladosporium cucumerinum and C. herbarum.

Culture preparation
Prior to testing, bacterial liquid cultures were prepared by subculture of a few colonies
(3 to 10) of the organism to be tested in 4 mL semi-liquid medium (containing only 0.01%
agar) followed by incubation to allow growth of organisms. The liquid cultures were then

47
Materials and Methods

seperately mixed in the Ultraturrax to produce suspensions of moderate cloudiness. The


suspension was diluted with sterile saline solution to a density visually equivalent to that of a
BaSO4 standards, prepared by adding 0.5 mL of 1% BaCl2 to 99.5 mL of 1% H2SO4 (0.36 N).
The prepared broth was inoculated onto agar plates using an Eppendorf pipette and
homogenously dispersed by means of a sterile spatula.
For the preparation of fungal cultures mycelia of C. cucumerinum and C. herbarum
(after growing the fungi for about one month) were put into a fresh fungal medium (see
section 2.1.1.6) and destroyed using Ultraturax. The cell debris (extracted mycellium) was
removed by vacuum filtration and the filtrate (medium containing fungal spores) was then
used for the next steps of the assay.

Anti-bacterial Assay
For the assay performed using E. coli and B. subtilis. A 100 – 200 µl of bacterial liquid
culture, in an exponential growth phase, was spread on to the surface of Luria Bertani (LB)
agar plate (see section 2.1.2.4). Immediately, 10-20 µl of tested samples were loaded onto the
disc paper (5 mm diameter, Oxoid Ltd.), to give final disc loading concentrations of 250-500
µg for crude extracts as well as 50-100 µg for pure compounds. The impregnated discs were
placed onto the surface of LB plates previously seeded with the selected test organisms, along
with discs containing solvent blanks. The culture was then incubated at 37º C for 24-48 hrs
(depending on the microbial culture being used). Antimicrobial activity was recorded as the
clear zone of inhibition surrounding the disc (diameter measured in mm) and compared to
Penicillin G, streptomycin and Gentamycin as positive controls.
The same technique was also applied to the assay using S. cerevisiae in a yeast
medium (see section 2.1.2.5) and incubation at 27º C for 24 hrs.

Anti-fungal Assay
A 100 ml of fungal spore suspension was spread out onto the surface of potato
dextrose agar medium (PDA, see section 2.1.2.7). Immediately, 10-20 µl of tested samples
were loaded onto the disc paper (5 mm diameter, Oxoid Ltd.), to give final disc loading
concentrations of 250-500 µg for crude extracts as well as 50-100 µg for pure compounds.
The impregnated discs were transferred onto the surface of the PDA medium. The fungal
cultures were then incubated at room temperature for several days and growth inhibition was
measured around the discs. The result was then compared to positive control (nystatin).

48
Materials and Methods

2.2.7.1.2. Inhibition of biofilm formation


Biofilm inhibition assays were carried out by PD. Dr. U. Hentschel, Zentrum für
Infektionsforschung, Würzburg.
The biofilm formation was determined by a simple adhesion assay in polystyrene
microtiter plates. For this purpose, Staphylococcus epidermidis cultures were diluted with
fresh TSB medium (see section 2.1.2.8) in the ratio of 1:100 (1980 µl medium + 20 µl
culture). 200 µl of the prepared suspension were pipetted into each well of a 96-well tissue
culture plate (8 time application per strain) and incubated at 37°C for 18 hrs. S. epidermidis
RP62A (wild type) was used as positive control, and S. carnosus TM 300 as negative control.
Samples to be tested were added to growing or already formed biofilms. After incubation, the
wells were carefully emptied and the plate washed three times with PBS-buffer (phosphate
buffered saline), and any remaining biofilm was heat-fixed on a hotplate at ca 60° C and
stained with crystal violet dye for 5 min, and excess dye was washed off with water. After
drying, the optical density of the adhering biofilm was determined by ELISA-Reader at 490
nm. Values lower than 0.120 were considered negative, strains with values between 0.120 and
0.240 were considered as weak adherents and results higher than 0.240 as strong adherents.
The limit of 0.120 corresponds to the three-way average value of the negative control.

2.2.7.2. Cytotoxicity test

2.2.7.2.1. Microculture tetrazolium (MTT) assay


Cytotoxicity tests were carried out by Prof. Dr. W. E. G. Müller, Institut für
Physiologische Chemie und Pathobiochemie, University of Mainz, Mainz. The cytotoxicity
was tested against L5178Y mouse lymphoma cells using the microculture tetrazolium (MTT)
assay, and compared to that of untreated controls (Carmichael, DeGraff, Gazdar, Minna, and
Mitchell, 1987).

Cell cultures
L5178Y mouse lymphoma cells were grown in Eagle’s minimal essential medium
supplement with 10% horse serum in roller tube culture. The medium contained 100 units/mL
penicillin and 100 µg/mL streptomycin. The cells were maintained in a humified atmosphere
at 37° C with 5% CO2.

49
Materials and Methods

MTT colorimetric assay


Of the test samples, stock solutions in ethanol 96% (v/v) were prepared. Exponentially
growing cells were harvested, counted and diluted appropriately. Of the cell suspension, 50
µL containing 3750 cells were pipetted into 96-well microtiter plates. Subsequently, 50 µL of
a solution of the test samples containing the appropriate concentration was added to each well.
The concentration range was 3 and 10 µg/mL. The small amount of ethanol present in the
wells did not affect the experiments. The test plates were incubated at 37° C with 5% CO2 for
72 h. A solution of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was
prepared at 5 mg/mL in phosphate buffered saline (PBS; 1.5 mM KH2PO4, 6.5 mM Na2HPO4,
137 mM NaCl, 2.7 mM KCl; pH 7.4) and from this solution, 20 µL was pipetted into each
well. The yellow MTT penetrates the healthy living cells and in the presence of mitochondrial
dehydrogenases, MTT is transformed to its blue formazan complex. After an incubation
period of 3 h 45 min at 37° C in a humidified incubator with 5% CO2, the medium was
centrifuged (15 min, 20 °C, 210 x g) with 200 µL DMSO, the cells were lysed to liberate the
formed formazan product. After thorough mixing, the absorbance was measured at 520 nm
using a scanning microtiter-well spectrophotometer. The colour intensity is correlated with
the number of healthy living cells. Cell survival was calculated using the formula:
absorbance of treated cells – absorbance of culture medium
Survival % = 100 x
absorbance of untreated cells – absorbance of culture medium

All experiments were carried out in triplicates and repeated three times. As controls, media
with 0.1% EGMME/DMSO were included in the experiments.

2.2.7.2.2. Protein kinase assay


Protein kinase assays were carried out by Dr. Michael Kubbutat (ProQinase GmbH,
Freiburg, Germany).
Protein kinase enzymes are integral components of numerous signal transduction
pathways involved in the regulation of cell growth, differentiation, and response to changes in
the extracellular environtment. Consequently, kinases are major targets for potentially
developing novel drugs to treat diseases such as cancer and various inflammatory disorders.
The inhibitory potency of the samples was determined using 24 protein kinases (see
Table 2.2). The IC50 profile of compounds/fractions showing an inhibitory potency of ≥ 40%
with at least one of the 24 kinases at an assay concentration of 1 × 10-06 g/mL was determined.
IC50 values were measured by testing 10 concentrations of each sample in singlicate (n=1).

50
Materials and Methods

Sample preparation
The compounds/fractions were provided as 1 × 10-03 g/mL stock solutions in 100%
DMSO (1000 or 500 µL) in micronic boxes. The boxes stored at -20o C. Prior to the assays,
100 µL of the stock solutions was transferred into separate microtiter plates. Subsequently,
they were subjected to serial, semi-logarithmic dilution using 100% DMSO as a solvent
resulting in 10 different concentrations. 100% DMSO was used as control. Subsequently, 7 ×
5 µL of each concentration were aliquoted and diluted with 45 µL H2O only a few minutes
before the transfer into the assay plate to minimize precipitation. The plates were shaken
thoroughly and then used for the transfer of 5 µL compound solution into the assay plates.

Recombinant protein kinases


All protein kinases were expressed in Sf9 insect cells as human recombinant GST-
fusion proteins or His-tagged proteins by means of the baculovirus expression system.
Kinases were purified by affinity chromatography using either GSH-agarose (Sigma) or Ni-
NTH-agarose (Qiagen). Purity was checked by SDS-PAGE/silver staining and the identity of
each kinase was verified by western blot analysis with kinase specific antibodies or by mass
spectrometry.

Protein kinase assay


A proprietary protein kinase assay (33PanQinase® Activity Assay) was used for
measuring the kinase activity of the protein kinases. All kinase assays were performed in 96-
well FlashPlatesTM from Perkin Elmer/NEN (Boston, MA, USA) in a 50 µL reaction volume.
The reaction mixture was pipetted in the following order: 20 µL assay buffer, 5 µL ATP
solution in H2O, 5 µL test compound in 10% DMSO and 10 µL substrate/10 µL enzyme
solution (premixed). The assay for all enzymes contained 60 mM HEPES-NaOH (pH 7.5), 3
mM MgCl2, 3mM MnCl2, 3 µM Na-orthovanadate, 1.2 mM DTT, 50 µg/mL PEG20000, 1 µM
[γ-33P]-ATP. The reaction mixtures were incubated at 30° C for 80 minutes and stopped with
50 µL 2% (v/v) H3PO4. The plates were aspirated and washed two times with 200 µL of 0.9%
33
(w/v) NaCl or 200 µL H2O. Incorporation of Pi was determined with a microplate
scintillation counter (Microbeta Trilux, Wallac). All assays were performed with a
BeckmanCoulter/Sagian robotic system.

51
Materials and Methods

Table 2.2: List of protein kinases and their substrates


Family Kinase Substrate Oncologically Disease
relevant
mechanism
Serine/threonine AKT1/PKB GCS3(14-27) Apoptosis Gastric cancer
kinases alpha (Staal, 1987)
ARK5 Autophos. Apoptosis Colorectal cancer
(Kusakai et al.,
2004)
Aurora A tetra(LRRWSLG) Proliferation Pancreatic cancers
(Li et al., 2003)
Aurora B tetra(LRRWSLG) Proliferation Breast cancer
(Keen and Taylor,
2004)
CDK2/Cyclin Histone H1 Proliferation Pancreatic cancer
A (Iseki et al., 1998)
CDK4/Cyclin Rb-CTF Proliferation Breast cancer
D1 (Yu et al., 2006)
CK2-alpha1 p53-CTM Proliferation Rhabdomyosarcoma
(Izeradjene et al.,
2004)
COT Autophos. Proliferation Breast cancer
(Sourvinos, 1999)
PLK-1 Casein Proliferation Prostate cancer
(Weichert et al.,
2004)
B-RAF-VE MEK1-KM Proliferation Thyroid cancer
(Ouyang et al.,
2006)
SAK Auotphos. Proliferation Colorectal cancer
(Macmillan et al.,
2001)
Receptor EGFR Poly(Glu,Tyr)4:1 Proliferation Glioblastoma
tyrosine multiforme
kinase (National Cancer
Institute, 2005)
EPHB4 Poly(Glu,Tyr)4:1 Angiogenesis Prostate cancer
(Xia et al., 2005)
ERBB2 Poly(Glu,Tyr)4:1 Proliferation Gastric carcinomas
(Lee et al., 2005)
FLT3 Poly Proliferation Leukemia
(Ala,Glu,Lys,tyr)6:2:4:1 (Menezes et al.,
2005)
IGF1-R Poly(Glu,Tyr)4:1 Apoptosis Breast cancer
(Zhang and Yee,
2000)
INS-R Poly “counter Ovarian cancer
(Ala,Glu,Lys,tyr)6:2:4:1 kinase” (Kalli et al., 2002)
MET Poly Metastasis Lung cancer
(Ala,Glu,Lys,tyr)6:2:4:1 (Qiao, 2002)

52
Materials and Methods

Family Kinase Substrate Oncologically Disease


relevant
mechanism
PDGFR-beta Poly Proliferation Prostate cancer
(Ala,Glu,Lys,tyr)6:2:4:1 (Hofer et al., 2004)
TIE-2 Poly(Glu,Tyr)4:1 Angiogenesis Rheumatoid
arthritis
(DeBusk et al.,
2003)
VEGF-R2 Poly(Glu,Tyr)4:1 Angiogenesis Pancreatic cancers
(Li et al., 2003)
VEGF-R3 Poly(Glu,Tyr)4:1 Angiogenesis Breast cancer
(Garces et al.,
2006)
Soluble FAK Poly(Glu,Tyr)4:1 Metastasis Breast cancer
tyrosine (Schmitz et al.,
kinase 2005)
SRC Poly(Glu,Tyr)4:1 Metastasis Colon cancer
(Dehm et al., 2001)

2.2.8. General laboratory equipments


Autoclave Varioklav, H&P
Balances Mettler 200, Mettler AT 250,
Mettler PE 1600, Sartorious MC1 AC210S
Centrifuge Biofuge pico, Heraeus
Cleanbench HERAsafe, Heraeus
Digital pH meter 420Aplus, Orion
Drying Ovens Kelvitron t, Heraeus
Fraction collector Cygnet, ISCO
Freeze dryer Lyovac GT2, Steris
- 80 °C Freezer Forma Scientific, 86-Freezer
Hot plate Camag
Magnetic stirrer Combi Mag, IKA
Rotary evaporator Vacuubrand, IKA
Sonicator Sonorex RK 102, Bandelin
Syringes Hamilton
Ultra Turrax T18 basic, IKA
UV Lamp Camag (254 and 366)
Vacuum centrifuge SpeedVac SPD 111V, Savant

53
Results

3. Results

3.1. Compounds isolated from the endophytic fungus Alternaria sp.


This endophytic fungal strain of the genus Alternaria was isolated from leaves of
Polygonum senegalense growing in Egypt. The pure fungal strain was cultivated on liquid
Wickerham medium and rice solid medium. Interestingly, chemical screening studies
indicated a clear difference between Alternaria extracts obtained from liquid Wickerham
medium and rice cultures. Comparison of the HPLC chromatograms of the EtOAc extracts of
both cultures showed a different chemical pattern. While the extract of liquid cultures showed
alternariol (1) and tenuazonic acid (14) as main components, altenusin (6) was the major
substance detected in the rice culture extract, with no traces of tenuazonic acid (see Figure
3.1A-B). The yield of EtOAc dried extract from rice cultures was much higher than that from
liquid cultures with a ratio of 11:1, respectively. Moreover, extracts obtained from liquid and
solid cultures were subjected to some preliminary biological screening assays, i.e.
antibacterial, antifungal, cytotoxicity and protein kinase assays. Interestingly, extracts
obtained from rice cultures showed higher cytotoxic and antifungal activity compared to those
of liquid cultures, while the latter had higher antibacterial activity (see Table 3.1).
In this part of the investigation results on the natural products produced by Alternaria
sp. when grown in liquid medium and on solid rice medium are presented.

54
Results

AH040119 #2 R-EtOAc UV_VIS_1


180
mAU WVL:235 nm

A 15
3 - 23,135
4 - 27,391

1
100
3

50 6 - 31,143

7 - 35,291
5 - 30,871 9 - 48,036
1 - 20,215
2 - 22,249 8 - 47,359

min
-20
0,0 10,0 20,0 30,0 40,0 50,0 60,0

AH060225 #13 Rrice UV_VIS_1


200
mAU WVL:235 nm
5 - 22,271

B 6
150

1
100 6 - 26,586
3 9 - 36,585
4 - 21,371
8 - 34,417
50 3 - 19,483 7 - 30,273

2 - 15,548 10 - 47,446
1 - 14,901

min
-20
0,0 10,0 20,0 30,0 40,0 50,0 60,0

Figure 3.1A-B: EtOAc extracts of Alternaria sp. cultures. A: HPLC chromatogram of


EtOAc extract of liquid cultures (Wickerham medium). B: HPLC
chromatogram of EtOAc extract of rice cultures. 1: Alternariol. 3:
Alternariol monomethyl ether. 6: Altenusin. 15: Tenuazonic acid.

Table 3.1: Biological screening test results for Alternaria liquid and rice extracts

Extracts tested L5178Y growth in % Protein kinase activity Antimicrobial activity


(Conc. 10 µg/mL) (Conc. 1 µg/mL) IZ [mm], 0.5 mg
BS SC CH
Alternaria liquid n-BuOH 99.8 Active 7 6 0
Alternaria liquid EtOAc 52.8 13 0 0
Alternaria rice EtOAc 0.1 Active 0 0 9
BS: B. subtilis, SC: S. cerevisiae, CH: C. herbarum.

55
Results

3.1.1. Alternariol (1, known compound)

Alternariol
Synonym(s) 3,7,9-Trihydroxy-1-methyl-6H-dibenzo[b,d]pyran-6-one
Sample code RE8, RR3.3, RR5.2
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 204.1 mg
Physical Description reddish white needles
Molecular Formula C14H10O5
Molecular Weight 258 g/mol
Retention time HPLC 27.3 min (standard gradient)

O 7
OH

2` 2
3` 3
1` 1
HO 4` 4
5` 6` 6 5

OH

700 AH040209 #9
RE8 UV_VIS_2
mAU WVL:254 nm

[M+H]+
1 - 27,247

500

375

250

[2M+Na]+
125

-100 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
[M-H]-
%
255.9
213.9

[2M-H]-

-10,0 nm
200 250 300 350 400 450 500 550 595

56
Results

Alternariol (1) was isolated from the EtOAc extracts of liquid and rice cultures of
Alternaria sp. as reddish white needles (204.1 mg). It showed UV absorbances at λmax
(MeOH) 206.1, 255.8, 299.8 and 339.7 nm. Positive and negative ESI-MS showed molecular
ion peaks at m/z 259.2 [M+H]+ (base peak) and m/z 257.4 [M-H]- (base peak), respectively,
indicating a molecular weight of 258 g/mol. The 1H and 13
C NMR spectra (Table 3.2a and
3.3) indicated the presence of an aromatic methyl group at δH 2.71 and δC 25.8 as well as four
aromatic protons. The coupling constants observed for the aromatic ring protons indicated the
presence of two aromatic rings, each bearing a pair of meta-coupled protons at δH 7.20 (J=2.0
Hz), 6.32 (J=2.0 Hz), 6.65 (J=2.5 Hz) and 6.55 (J=2.5 Hz) assigned for H-4, H-6, H-3` and
H-5`, respectively. The signal at δC 166.8 in the 13C NMR spectrum indicated the presence of
13
a conjugated lactone (C-7). The C NMR spectrum revealing the presence of 15 carbon
atoms as well as 1H NMR and mass spectra supported a molecular formula of C15H12O5.
Further confirmation was achieved by interpretation of the HMBC spectrum (see Table 3.2a
and Figure 3.2) showing correlations of the meta-coupled protons, H-4 to C-2, C-3 and C-6 as
well as H-6 to C-2, C-4 and C-5, thereby establishing the structure of one aromatic ring. The
correlations of the aromatic methyl group to C-(1`-6`) and those observed for H-3` and H-5`
confirmed the structure of the second aromatic ring. Furthermore, correlations of the methyl
group to C-6 and of H-6 to C-1` proved the point of attachment of both rings. UV, 1H, 13
C
NMR and mass spectral data were found to be identical to published data for alternariol
(Stinson et al., 1986), previously reported from several Alternaria species (Freeman, 1966;
Coombe et al., 1970; Bradburn et al., 1994).

57
Results

3.1.2. Alternariol-5-O-sulphate (2, new compound)

Alternariol-5-O-sulphate
Synonym(s) 3,7,9-Trihydroxy-1-methyl-6H-dibenzo[b,d]pyran-6-one-9-sulphate
Sample code RB6
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 60.9 mg
Physical Description reddish white needles
Molecular Formula C14H10O8S
Molecular Weight 338 g/mol
Retention time HPLC 22.8 min (standard gradient)

O 7
OH

2` 2
3` 3
1` 1
HO 4` 4
5` 6` 6 5

OSO3H

AH040202 #3 RB6 UV_VIS_3


200
mAU WVL:280 nm
1 - 22,795 [M+H-SO3]+
150

100 [M+H]+

50

min
-20
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
249.6
214.5

[M-HSO3]-
345.5

-10,0 nm
200 250 300 350 400 450 500 550 595

58
Results

Compound 2 was isolated from the n-BuOH extract of liquid cultures of Alternaria sp.
in the form of reddish white needles (60.9 mg). It displayed UV absorbances at λmax (MeOH)
214.6, 249.7, 287.6 and 345.1 nm, showing high similarity to UV spectra typical for
alternariol derivatives. The HRESI-MS exhibited a strong peak at m/z 339.0170 [M+H]+
indicating a molecular formula of C14H10O8S (calculated 339.0174, ∆ 0.0004). The 1H NMR
spectrum (see Table 3.2a) showed signals for a methyl group at δ 2.75 and two pairs of meta-
coupled aromatic protons at δH 7.83 (J=2.0 Hz), 6.86 (J=2.0 Hz), 6.67 (J=2.0 Hz) and 6.57
(J=2.0 Hz) corresponding to H-6, H-4, H-5` and H-3`, respectively. The dibenzo-α-pyrone
structure was confirmed by the NOE effects observed both on H-6 and H-5` upon irradiation
of the 6`-methyl group. Interpretation of the HMBC spectrum (see Table 3.2a and Figure 3.2)
showed that the correlations observed for 6`-CH3 were identical to those observed for 1.
Moreover, correlations of the meta-coupled protons, H-4 to C-2, C-3, C-5 and C-6 as well as
H-6 to C-2, C-4 and C-5, were similar in both compounds indicating similar structures.
However, comparison of 1H and 13C NMR data (Table 3.2a and 3.3) with those measured for
alternariol (1) showed good congruence except for the downfield shifts observed for H-4 and
H-6, as well as the upfield shift of C-5, of 6.7 ppm, and downfield shifts of C-4 and C-6, of
4.1 and 2.6 ppm, respectively, indicating the presence of a sulphate substitution at C-5
(Ragan, 1978). This deduction was confirmed by the fragment formed by loss of 80 mass
units in the mass spectrum of 2 and the hypsochromic shift in the UV spectrum of 2 compared
to that of 1, which is attributed to the electron withdrawing effect of the sulphate group
(Plasencia and Mirocha, 1991). The compound was thus identified as the new natural product
alternariol-5-O-sulphate.

59
Results

3.1.3. Alternariol-5-O-methyl ether (3, known compound)

Alternariol-5-O-methyl ether
Synonym(s) 3,7-Dihydroxy-9-methoxy-1-methyl-6H-dibenzo[b,d]pyran-6-one,
Djalonensone
Sample code RE6, RR2, RR3.5
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 239.0 mg
Physical Description reddish white needles
Molecular Formula C15H12O5
Molecular Weight 272 g/mol
Retention time HPLC 31.0 min (standard gradient)

O 7
OH

2` 2
3` 3
1` 1
HO 4` 4
5` 6` 6 5

400 AH040209 #7 RE6 UV_VIS_2


mAU WVL:254 nm
[M+H]+
1 - Djalonensone - 30,966

300

200

[2M+Na]+
100

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
[M-H]-
%
256.8
216.0

[2M-H]-

-10,0 nm
200 250 300 350 400 450 500 550 595

60
Results

Alternariol-5-O-methyl ether (3) was isolated from the EtOAc extracts of liquid and
rice cultures of Alternaria sp. in the form of reddish white needles (239.0 mg). It exhibited
UV absorbances at λmax (MeOH) 216.2, 256.9, 287.0 and 338.8 nm, having the typical pattern
of alternariol derivatives. Positive and negative ESI-MS showed molecular ion peaks at m/z
273.2 [M+H]+ (base peak) and m/z 271.3 [M-H]- (base peak), respectively, indicating a
molecular weight of 272 g/mol with an increase of 14 mass units compared to alternariol (1)
and thus supporting a molecular formula of C15H12O5. The 1H and 13
C NMR spectra (see
Table 3.2b and 3.3) showed a methoxy group at δH 3.91 and δC 55.9, an aromatic methyl
group at δH 2.76 and δC 25.3 as well as four aromatic protons at δH 7.28 (J=1.8 Hz), 6.70
(J=2.5 Hz), 6.61 (J=2.5 Hz) and 6.54 (J=1.8 Hz) assigned for H-6, H-5`, H-3` and H-4,
respectively. Interpretation of the HMBC spectrum (see Table 3.2b and Figure 3.3) showing
correlations similar to those observed for 1 indicated a close structure resemblance. The
structure was confirmed by comparison of UV, 1H, 13
C NMR and mass spectral data with
published data for alternariol-5-O-methyl ether (Onocha et al., 1995), also known as
djalonensone, previously reported from several Alternaria species (Freeman, 1966; Coombe
et al., 1970; Bradburn et al., 1994) as well as from Anthocleista djalonensis (Onocha et al.,
1995).

61
Results

3.1.4. Alternariol-5-O-methyl ether-4`-O-sulphate (4, new compound)

Alternariol-5-O-methyl ether-4`-O-sulphate
Synonym(s) 3,7-Dihydroxy-9-methoxy-1-methyl-6H-dibenzo[b,d]pyran-6-one-
3-sulphate
Sample code RB4
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 5.9 mg
Physical Description reddish white needles
Molecular Formula C15H12O8S
Molecular Weight 352 g/mol
Retention time HPLC 25.5 min (standard gradient)

O 7
OH

2` 2
3` 3
1` 1
HO3SO 4` 4
5` 6` 6 5

700 AH040202 #2
RB4 UV_VIS_2
mAU WVL:254 nm
1 - 25,514
[M+H-SO3]+
500

375

250 [M+H]+

125 2 - 27,291

min
-100
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
253.7

[M-HSO3]-
202.6

336.7

-10,0 nm
200 250 300 350 400 450 500 550 595

62
Results

Compound 4 was isolated from the n-BuOH extract of liquid cultures of Alternaria sp.
as reddish white needles (5.9 mg). The UV spectrum showed λmax (MeOH) at 202.5, 253.5,
285.3 and 336.8 nm. The HRESI-MS showed [M+H]+ at m/z 353.0320 (calculated 353.0331,
∆ 0.0011), indicating a molecular formula of C15H12O8S and an increase of 14 mass units
more than 2. The compound showed physical and UV spectral features similar to those of 1, 2
and 3. The 1H NMR spectrum (see Table 3.2b) showed signals for a methoxy group at δH
3.84, an aromatic methyl group at δH 2.74 and two pairs of meta-coupled aromatic protons at
δH 7.27 (J=2.5 Hz), 7.14 (J=2.5 Hz), 7.08 (J=2.5 Hz) and 6.52 (J=2.5 Hz) corresponding to H-
6, H-3`, H-5` and H-4, respectively. Comparison of the 1H and 13
C NMR spectra of 4 (see
Table 3.2b and 3.3) with those of 2 and 3 suggested a close relationship between their
structures. In addition, interpretation of the HMBC spectrum (see Table 3.2b and Figure 3.3)
showed that correlations of the meta-coupled protons H-4 and H-6 were identical to those
observed for 3. Moreover, correlations of the meta-coupled protons, H-3` to C-2`, C-4` and C-
5` as well as H-5` to C-1`, C-3`, C-4` and 6`-CH3, were similar in both compounds indicating
similar structures. However, in spite of close resemblance of 1H NMR spectral features of 4
and 3 exceptions were the downfield shifts observed for H-3` and H-5` along with the upfield
shift of C-4`, by 5.1 ppm, and downfield shifts of C-3` and C-5`, by 6.4 and 4.6 ppm,
respectively, in a similar pattern as in 2 indicating a sulphate group to be attached at C-4`
(Ragan, 1978). Presence of the fragment formed by loss of 80 mass units in the mass
spectrum and the hypsochromic shift in the UV spectrum of 4 confirmed the structure
(Plasencia and Mirocha, 1991). Thus, compound 4 was identified as the new natural product
alternariol-5-O-methyl ether-4`-O-sulphate.

63
Results

3.1.5. 3`-Hydroxyalternariol-5-O-methyl ether (5, new compound)

3`-Hydroxyalternariol-5-O-methyl ether
Synonym(s) 3,7,8-Trihydroxy-9-methoxy-1-methyl-6H-dibenzo[b,d]pyran-6-one
Sample code RR3.4
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 5.8 mg
Physical Description violet needles
Molecular Formula C15H12O6
Molecular Weight 288 g/mol
Retention time HPLC 28.3 min (standard gradient)

HO O 7
OH

2` 2
3` 3
1` 1
HO 4` 4
5` 6` 6 5

AH060616 #10 1-4 UV_VIS_1


700
mAU WVL:235 nm
1 - 28,331
[M+H]+

400

[2M+Na]+

200

2 - Weichmacher (Phthalat) - 37,127

3 - 44,096
4 - 47,784

min
-100
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
260.1
236.0
203.4

-10,0 nm
200 250 300 350 400 450 500 550595

64
Results

Compound 5 was isolated from the EtOAc extract of rice cultures of Alternaria sp.. It
was isolated as violet needles (5.8 mg). The UV spectrum showed absorbances at λmax
(MeOH) 203.4, 236.0, 260.1 and 340.0 nm, showing high similarity to UV spectra typical for
alternariol derivatives 1, 2, 3 and 4. The HRESI-MS showed [M+H]+ at m/z 289.0720
(calculated 289.0712, ∆ 0.0008), establishing the composition C15H12O6 and indicating an
increase of 16 mass units in the molecular weight compared to 3. UV and NMR spectra of 5
had close similarity to those of 3. The 1H NMR spectrum (see Table 3.2b) resembled that of 3
except for the absence of one meta-coupled pair of aromatic protons and the presence of an
aromatic proton singlet at δH 6.82 assigned for H-5`. In addition, the 1H and 13C NMR spectra
(Table 3.2b and 3.3) showed a methoxy group at δH 3.99 and δC 56.3, an aromatic methyl
group at δH 2.74 and δC 24.9 and a pair of aromatic protons at δH 7.30 (J=1.8 Hz) and 6.63
(J=1.8 Hz) assigned to H-6 and H-4, respectively. Interpretation of the HMBC spectrum (see
Table 3.2b and Figure 3.4) showed that correlations of the meta-coupled protons H-4 and H-6
were identical to those observed for 3. On the other hand, H-5` correlated to C-1`, C-3`, C-4`
and 6`-CH3 with an upfield shift observed for C-4`and a downfield shift for C-3` compared to
the corresponding chemical shifts in compounds 1-4. These findings suggested the presence
of an additional hydroxy substitution on the aromatic ring which was placed at C-3`. This was
13
further supported by the upfield shifts of C-2` and C-6` in the C NMR spectrum. Thus
compound 5 was identified as the new natural product 3`-hydroxyalternariol-5-O-methyl
ether.

65
Results

R1 O 7
OH

2` 2
3` 3
1` 1
R2O 4` 4
5` 6` 6 5

OR3

Nr. Compound R1 R2 R3
1 Alternariol H H H
2 Alternariol-5-O-sulphate H H SO3H
3 Alternariol-5-O-methyl ether H H CH3
4 Alternariol-5-O-methyl ether-4`-O-sulphate H SO3H CH3
5 3`-Hydroxyalternariol-5-O-methyl ether OH H CH3

Table 3.2a: 1H NMR and HMBC data of compounds 1-2 at 500 MHz

Nr. 1 2
δH (MeOD) HMBC δH (MeOD) HMBC
4 6.32,d (2.0) 2,3,6 6.86,d (2.0) 2,3,5,6
6 7.20,d (2.0) 2,4,5,1` 7.83,d (2.0) 2,4,5
3` 6.55,d (2.5) 1`,2`,4`,5` 6.57,d (2.0) 1`,2`,4`,5`
5` 6.65,d (2.5) CH3, 1`,3`,4` 6.67,d (2.0) CH3, 1`,3`,4`
CH3 2.71,s 1`,2`,3`,5`,6`,6 2.75,s 1`,2`,3`,5`,6`,6

Table 3.2b: 1H NMR and HMBC data of compounds 3-5 at 500 MHz

Nr. 3 3a 4 5
δH (MeOD) HMBC δH (CDCl3) δH (MeOD) HMBC δH (DMF-d7) HMBC
4 6.54,d (1.8) 2,3,5,6 6.3,d (2.0) 6.52,d (2.5) 2,3,6 6.63, d (1.8) 2,3,5,6
6 7.28,d (1.8) 2,4,5,1` 7.1,d (2.0) 7.27,d (2.5) 2,4,5,1` 7.30, d (1.8) 2,4,5,1`
3` 6.61,d (2.5) 1`,2`,4`,5` 6.5,d (2.0) 7.14,d (2.5) 2`,4`,5`
5` 6.70,d (2.5) CH3, 1`,3`,4` 6.5,d (2.0) 7.08,d (2.5) CH3, 1`,3`,4` 6.82, s CH3, 1`,3`,4`
CH3 2.76,s 1`,5`,6`,6 2.62,s 2.74,s 1`,5`,6`,6 2.74, s 1`,2`,3`,5`,6`,6
OCH3 3.91,s 5 3.79,s 3.84,s 5 3.99, s 5
3-OH 12.03, br s
a) Onocha et al., 1995.

66
Results

Table 3.3: 13C NMR data of compounds 1-5 at 125 MHz

Nr. 1 1a 2 3 4 5
δC δC δC δC δC δC
(MeOD) (DMSO-d6) (MeOD)b (MeOD)b (MeOD) (DMF-d7)
1 139.8 138.1 138.8 139.5
2 99.1 97.4 101.6 99.6 100.5 99.2
3 166.1 164.1 165.8 165.8 165.4
4 101.9 100.9 106.0 99.7 100.9 99.7
5 166.8 165.5 160.1 167.7 168.1 167.3
6 105.4 104.4 108.0 104.5 105.9 104.2
7 166.8 164.7 165.8
1` 110.9 109.0 109.7 110.5 115.1 110.1
2` 154.4 152.6 153.2 154.3 153.6 142.3
3` 102.7 101.6 101.3 102.4 108.8 132.4
4` 159.8 158.4 159.0 159.6 154.5 148.2
5` 118.5 117.6 117.6 118.3 122.9 117.7
6` 140.0 138.3 139.0 139.3 139.5 127.3
CH3 25.8 25.3 24.6 25.3 25.7 24.9
OCH3 55.9 56.3 56.3
a) Stinson et al., 1986.
b) Derived from HMBC spectrum.

4
6 5` 3` 4 3`
6 5`
(ppm)

2
4 100

3` H6/C4
6 H4/C6 C4
1` H6/C4 H4/C6 C6
5` 120

6` 140

2`
4`
C5
160

H6/C5 H4/C5 H6/C5 H4/C5


5

(ppm) 7.40 7.20 7.00 6.80 6.60 6.40 6.20

Figure 3.2: HMBC spectra of compounds 1 and 2.

67
Results

6 5` 3` 4 6 3` 5`
4

(ppm) (ppm)

100 100

C3` H5`/C3`
H5`/C3`
C3`

C5` 120

H3`/C5`
120

C5`

H3`/C5`

140
140

H3`/C4`
C4`
H5`/C4`
H5`/C4`
C4` 160

H3`/C4` 160

(ppm) 7.40 7.20 7.00 6.80 6.60 6.40 (ppm) 7.60 7.40 7.20 7.00 6.80 6.60 6.40

Figure 3.3: HMBC spectra of compounds 3 and 4.

OCH3 CH3

6 5` 4
(ppm)

2
4 100

6
1`

5` CH3/C5`
120

6` CH3/C6`
3`
H5`/C3` CH3/C3`
1 140
2` CH3/C2`
4` H5`/C4`

3 160

7 5
(ppm) 7.2 6.4 5.6 4.8 4.0 3.2 2.4

Figure 3.4: HMBC spectra of compound 5.

68
Results

3.1.6. Altenusin (6, known compound)

Altenusin
Synonym(s) 3,4',5'-Trihydroxy-5-methoxy-2'-methyl[1,1'-biphenyl]-2-carboxylic
acid
Sample code RE4, RR7, RR8.2
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 377.6 mg
Physical Description reddish white prisms
Molecular Formula C15H14O6
Molecular Weight 290 g/mol
Retention time HPLC 23.0 min (standard gradient)
O

HO HO 7 OH

2` 2
3` 3
1` 1
HO 4` 4
5` 6` 6 5

600 AH040209 #6 RE4 UV_VIS_1


mAU WVL:235 nm
1 - 22,971 [M+H]+

375

250

125

2 - 26,611 3 - 47,607

-100 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
215.4

[2M-H]-
256.5

[M-H-CO2]-

-10,0 nm
200 250 300 350 400 450 500 550 595

69
Results

Altenusin (6) was isolated from the EtOAc extracts of liquid and rice cultures of
Alternaria sp. in the form of reddish white prisms (377.6 mg). It exhibited UV absorbances at
λmax (MeOH) 202.6, 214.6, 256.9 and 293.0 nm. Positive and negative ESI-MS showed
molecular ion peaks at m/z 290.9 [M+H]+ (base peak) and m/z 289.2 [M-H]- (base peak),
respectively, indicating a molecular weight of 290 g/mol. The 1H NMR spectrum (see Table
3.4) showed two aromatic singlets at δH 6.57 and 6.47 assigned to H-5` and H-2`,
respectively, as well as meta-related hydrogens at δH 6.42 and 6.16 (each doublet, J=2.5 Hz)
corresponding to H-4 and H-6, respectively. A methoxy signal was seen at δH 3.79 and an
13
aromatic methyl at δH 1.89. The signal at δC 174.3 in the C NMR spectrum indicated the
presence of a carboxy group (C-7). This was further confirmed by the presence of a fragment
at m/z 245.3 [M-CO2-H]- in the negative ESI-MS. The 13
C NMR spectrum (see Table 3.4)
revealing the presence of 15 carbon atoms together with 1H NMR and mass spectra supported
a molecular formula of C15H14O6. The structure was established on basis of the HMBC data
(see Table 3.4 and Figure 3.5) showing correlations of the meta-coupled protons, H-4 to C-2,
C-3, C-5 and C-6 as well as H-6 to C-2 and C-4, thereby establishing the structure of one
aromatic ring. The methoxy group was found to correlate to C-5 thereby indicating its
location there. Furthermore, from the correlations of the aromatic methyl group to C-1`, C-5`
and C-6`, H-5` to C-1`, C-3`, C-4` and 6`-CH3 as well as those observed for H-2` to C-3`, C-
4` and C-6` the structure of the second aromatic ring was deduced. The correlation of H-6 to
C-1` and that of H-2` to C-1 established the C1-C1` bond attaching both rings. The obtained
UV, 1H, 13C NMR and mass spectral data were found to be identical with published data for
altenusin (Nakanishi et al., 1995), previously reported from Alternaria (Coombe et al., 1970)
and Penicillium species (Nakanishi et al., 1995).

70
Results

3.1.7. Desmethylaltenusin (7, new compound)

Desmethylaltenusin
Synonym(s) 3,5,4',5'-Tetrahydroxy-2'-methyl[1,1'-biphenyl]-2-carboxylic acid
Sample code RR8.1
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 5.6 mg
Physical Description viscous reddish oil
Molecular Formula C14H12O6
Molecular Weight 276 g/mol
Retention time HPLC 17.6 min (standard gradient)

HO HO 7 OH

2` 2
3` 3
1` 1
HO 4` 4
5` 6` 6 5

OH

AH060625 #2 RR8-1 UV_VIS_2


700
mAU WVL:254 nm

1 - 17,578
(+)-ESI-MS: no ionization
500

375

250

125

2 - 19,536 3 - 37,149

min
-100
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
[M-H
223.

-CO2]-
5
255.
4

-10,0 nm
200 250 300 350 400 450 500 550 595

71
Results

Compound 7 was isolated from the EtOAc extract of rice cultures of Alternaria sp. as
viscous reddish oil (5.6 mg). It displayed UV absorbances at λmax (MeOH) 202.6, 223.5, 255.4
and 293.0 nm. The HR-MS exhibited a strong peak at m/z 299.0520 [M+Na]+ indicating a
molecular formula of C14H12O6 (calculated 299.0531, ∆ 0.0011) as well as a loss of 14 mass
units compared to altenusin (6). Both compounds showed similar UV spectra. Comparing the
1 13
H and C NMR spectra of 7 with those of 6 (see Table 3.4) suggested a close relationship
between both structures. The main difference between 7 and 6 was the absence of the
methoxy group signal of 6 and the upfield shifts observed for C-4 and C-6, by 1.8 and 3.2
ppm, respectively, as well as for their corresponding meta-related hydrogens at δH 6.25 and
6.03 (each doublet, J=2.2 Hz), respectively. Two aromatic singlets at δH 6.55 and 6.46
assigned to H-5` and H-2`, respectively, were also detected in the 1H NMR spectrum, as well
as an aromatic methyl signal at δH 1.90. The structure was further confirmed by interpretation
of the HMBC spectrum (see Table 3.4 and Figure 3.6) which showed that all observed
correlations were corresponding to those observed in the spectrum of 6 except for the lacking
methoxy group. In addition, the characteristic fragment at m/z 231.6 [M-CO2-H]- observed in
the negative ESI-MS confirmed the presence of a carboxylic group substituent as in case of
altenusin. Thus the compound was confirmed to be desmethylaltenusin, representing a new
natural product.

72
Results

HO HO 1 OH

2` 2
3` 3
1` 7
HO 4` 4
5` 6` 6 5

OR

Nr. Compound R
6 Altenusin CH3
7 Desmethylaltenusin H

Table 3.4: 1H,13C NMR and HMBC data of compounds 6 and 7 at 500 (1H) and 125 MHz (13C)

Nr. 6 6a 7
δH HMBC δC δH δC δH HMBC δC
(MeOD) (MeOD) (DMSO-d6) (DMSO-d6) (MeOD) (MeOD)
1 148.0 145.0 148.3
2 107.0 108.8 106.5
3 164.9 161.6 163.0
4 6.42, d (2.5) 2,3,5,6 100.5 6.43, d (2.7) 99.6 6.25, d (2.2) 2,3,5,6 102.3
5 165.7 162.0 165.7
6 6.15, d (2.5) 2,4,1` 111.4 6.10, d (2.7) 108.9 6.03, d (2.2) 2,4,1` 112.1
7 174.3 171.6
1` 135.3 132.4 135.8
2` 6.47, s 3`,4`,6`,1 116.6 6.42, s 115.9 6.46, s 3`,4`,6`,1 116.6
3` 143.2 142.1 143.2
4` 144.9 143.9 144.7
5` 6.57, s CH3, 1`,3`,4` 117.3 6.54, s 116.6 6.55, s CH3, 1`,3`,4` 117.2
6` 127.3 124.9 127.3
CH3 1.89, s 1`,5`,6` 19.2 1.86, s 18.8 1.90, s 1`,5`,6`,3` 19.3
OCH3 3.79, s 5 55.9 3.76, s 55.3
a) Nakanishi et al., 1995.

73
Results

OCH3 CH3
2`
6
5` 4
(ppm)
H6/C4
4 100

2 H4/C2 H6/C2
6
H4/C6
2` CH3/C5`
5` 120

H2`/C6` CH3/C6`
6`
H5`/C1` H6/C1` CH3/C1`
1`
H5`/C3`,C4` 140
3`
4` H2`/C3`,C4`
1 H2`/C1

160

3 H4/C3 OCH3/C5
5 H4/C5
(ppm) 6.4 5.6 4.8 4.0 3.2 2.4 1.6

Figure 3.5: HMBC spectra of compound 6.

CH3
5` 2` 4 6
4 H6/C4
(ppm)

2 H4/C2
H6/C2
H4/C6 110.0
6
2` CH3/C5`
5` 120.0

6` H2`/C6` CH3/C6`
130.0

H5`/C1` H6/C1` CH3/C1`


1`
H5`/C3`,C4` 140.0
3`
4` H2`/C3`,C4`
1
H2`/C1 150.0

160.0
3 H4/C3
5 H4/C5
(ppm) 6.4 5.6 4.8 4.0 3.2 2.4

Figure 3.6: HMBC spectra of compound 7.

74
Results

3.1.8. Alterlactone (8, new compound)

Alterlactone
Sample code RR3.2
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 8.4 mg
Physical Description white flakes
Molecular Formula C15H12O6
Molecular Weight 288 g/mol
Retention time HPLC 22.0 min (standard gradient)

OH O

8 O 6
7a 7
9
5

O 10 11a 11b 4a
11
4
1
3
2
OH
HO

AH060803 #2 RR4c-4 UV_VIS_3


300
mAU WVL:280 nm
1 - 22,033

[2M+Na]+
200
[M+H]+

100

2 - 26,959 3 - 37,114

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
206.0
221.1

254.4

[M-H-CO2]-

-10,0 nm
200 250 300 350 400 450 500 550 595

75
Results

Compound 8 was isolated from the EtOAc extract of rice cultures of Alternaria sp. in
form of white flakes (8.4 mg). The UV spectrum showed absorbances at λmax (MeOH) 206.0,
221.1 and 254.4 nm. The HRESI-MS gave a [M+H]+ at m/z 289.0700 (calculated 289.0712, ∆
13
0.0012), indicating the molecular formula to be C14H14O6. Its C NMR spectrum (see Table
3.5) displayed one methoxy group, twelve aromatic carbons, a carbonyl function and an
oxygenated methylene group corresponding to a pair of doublets at δH 4.80 and 4.85 (J=11.1
Hz) in the 1H NMR spectrum. This could be accounted for by assuming that the methylene
protons are situated on a ring and their non-equivalence must result from a steric factor of the
biphenyl system. Confirmation was achieved by an HMBC 3J correlation of the methylene
protons with the carbonyl carbon (C-7) suggesting that the lactone ring was not formed
through connection of the carbonyl carbon to the phenolic hydroxy group as in the previously
discussed alternariol derivatives (1-5), but instead the carbonyl carbon was linked through an
ester to the hydroxymethyl group to construct an additional seven-membered lactone ring. The
meta-coupled hydrogens, observed in the 1H NMR spectrum at δH 6.45 and 6.50 (each
doublet, J=2.2 Hz), were assigned to H-9 and H-11, respectively. Both protons showed
ROESY correlations (see Table 3.5 and Figure 3.8) to the methoxy group indicating its
location at C-10. In addition, the correlations observed for H-9 and H-11 to C-7a in the
HMBC spectrum (see Table 3.5 and Figure 3.7), as well as the chelated nature of 8-OH
deduced from its appearance at δH 10.21, confirmed the attachment of the aromatic ring to the
lactone ring at C-7a. Furthermore, the hydroxy substituents were placed in ortho-position at
C-2 and C-3 on basis of the chemical shift values of the carbon atoms appearing at δC 146.6
and 145.9, respectively. The neighboring position of H-1 and H-4, observed at δH 7.03 and
6.90 in the 1H NMR spectrum, respectively, was deduced from their HMBC correlations to C-
3 and C-2, respectively. Furthermore, H-4 showed both ROESY and HMBC correlations to
the methylene group as well as HMBC correlations to C-4a and C-11b. This together with the
ROESY correlation observed between H-1 and H-11 as well as the HMBC correlations of H-
11 to C-11b and those of H-1 to C-4a, C-11a and C-11b indicated the attachment of the
aromatic rings through the C11a-C11b bond. The structure was further confirmed by
comparing NMR data of 8 to those reported for ulocladol (8a) (Höller et al., 1999) and
graphislactone D (8b) (Tanahashi et al., 1997). Both compounds differ from 8 in having an
additional hydroxy function at C-1 as well as methoxy groups at C-3 (both) and C-8
(graphislactone D). Thus the compound was identified as the new natural product to which we
propose the name alterlactone. It is worth mentioning that this is only the third report for
isolation of this carbon skeleton in nature.

76
Results

OR3 O

8 O 6
7a 7
9
5

O 10 11a 11b 4a
11
4
R1 1 3
2
OR2
HO

Nr. Compound R1 R2 R3
8 Alterlactone H H H
8a Ulocladol OH CH3 H
8b Graphislactone D OH CH3 CH3

Table 3.5: 1H,13C NMR, ROESY and HMBC data of compound 8 at 500 (1H) and 125 MHz (13C)

Nr. 8 8ba
δH ROESY HMBC δC δH δC (DMSO-
(DMSO-d6) (DMSO-d6) (DMSO-d6) d6)
1 7.03, s 11 3,4a,11a,11b 115.5 143.8
2 146.6 135.7
3 145.9 147.6
4 6.90, s 5 2,3,5,11b,4a 115.5 6.78, s 103.9
4a 140.0 135.1
5 4.80, d (11.3) 4 4,7,11b 67.8 4.74, dd (12.0) 68.3
4.85, d (11.0) 4.78, dd (12.0)
7 168.7 166.1
7a 109.5 113.2
8 159.9 158.6
9 6.45, d (2.2) OCH3 7a,8,10,11 100.8 6.66, d (2.0) 97.7
10 162.2 160.6
11 6.50, d (2.2) OCH3,1 7a,9,10,11b 105.0 6.89, d (2.0) 106.8
11a 126.6 127.3
11b 129.8 118.2
1-OH 9.10, brsb)
b
2-OH 9.37, brs 8.98, brsb)
3-OH 9.47, brsb
3-OCH3 3.84, s 56.0
8-OH 10.21, brs
8-OCH3 3.81, s 55.3
10-OCH3 3.81, s 9,11 10 55.4 3.82, s 55.8
a) Tanahashi et al., 1997.
b) Assignements may be interchanged.

77
Results

14
9 11 5
(ppm)

5
H4/C5

80

H11/C9
9 100

11 H9/C11
7a H9/C7a H11/C7a H5/C4
H5/C4
1, 4
120

11a H1/C11a H5/C11b


11b H1/C11b H4/C11b
H11/C11b

4a H1/C4a H4/C4a
140

3
2 H1/C3 H4/C3
H4/C2

8 H9/C8 160

10 H9/C10 H11/C10
H5/C7
7
(ppm) 7.2 6.8 6.4 6.0 5.6 5.2 4.8

Figure 3.7: HMBC spectrum of compound 8.

1 4 9 11 1 4 9 11

H1/H11
11
OCH3
9
4
1
H4/CH2
5 5

Figure 3.8: ROESY spectrum of compound 8.

78
Results

3.1.9. Talaroflavone (9, known compound)

Talaroflavone
Sample code RR3.1
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 5.0 mg
Physical Description white flakes
Molecular Formula C14H12O6
Molecular Weight 276 g/mol
20
Optical Rotation [α]D + 179 (c 0.7, MeOH)
Retention time HPLC 15.9 min (standard gradient)
OH
O

1 7
2 6
O
5
1`
3 O
2` (R) (S) 4
OH
5`
4`
3`

AH060616 #4 1-1c UV_VIS_2


350
mAU WVL:254 nm

1 - 15,944 (+)-ESI-MS: no ionization

200

100

2 - 17,034 3 - 37,115

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
219.5

[M-H]-
260.2

295.1

-10,0
nm
200 250 300 350 400 450 500 550 595

79
Results

Talaroflavone (9) was isolated from the EtOAc extract of rice cultures of Alternaria
sp. in the form of white flakes (5.0 mg). It displayed UV absorbances at λmax (MeOH) 219.5,
260.2 and 295.1 nm. The negative ESI-MS showed a pseudo-molecular ion peak at m/z 275.2
[M-H]- (base peak) indicating a molecular weight of 276 g/mol. 1H NMR and mass spectra
supported a molecular formula of C14H12O6. The 1H NMR spectrum exhibited meta-coupled
protons at δH 6.43 (broad singlet) and 6.04 (doublet, J=1.2 Hz) assigned to H-4 and H-6,
respectively, as well as a methoxy signal at δH 3.79. These 1H NMR data (see Table 3.6)
resembled those of the aromatic portion of metabolites 6 and 7. Furthermore, a carbinolic
13
hydrogen at δH 4.73 and a C NMR signal at δC 79.0 were consistent with the secondary
13
alcohol group at C-5`. Signals in the C NMR (see Table 3.7) spectrum at δC 201.0 (C-4`),
172.0 (C-2`), 130.5 (C-3`) and 13.0 (2`-CH3), and 1H NMR resonances at δH 1.85 (2`-CH3)
and 6.34 (H-3`) were consistent with a 3-methylcyclopentenone. This substructure was further
confirmed by interpretation of the HMBC spectrum (see Table 3.6) showing correlations of
2`-CH3 to C-1`, C-2` and C-3`, H-3` to C-1`, C-4` and C-5` as well as H-5` to C-1`and C-4`.
The degrees of unsaturation calculated from the molecular formula were found to be 9
[(2C+2-H)/2], where the aromatic ring and the cyclopentenone account for 4 and 3 degrees of
unsaturation, respectively. A third ring, joining the aromatic and enone portions, incorporated
the quaternary spiro carbon at δC 94.0 (C-1`) and the ester carbonyl group (C-1), thus
accounting for the remaining two degrees of unsaturation inspired by the molecular formula.
This was also supported by the correlation observed for H-4 to C-1` in the HMBC spectrum.
In order to determine the relative configuration of this compound, a NOESY
experiment was performed. The 2`-CH3 group was found to correlate to H-4 and H-3`,
additionally, H-5` correlated to H-4 and H-3` (see Table 3.6 and Figure 3.9). Furthermore, for
the determination of the absolute configuration of the chiral centre at C-5` the modified
Mosher procedure was applied. The observed shift difference between the (S)-MTPA ester
and its (R)-MTPA ester epimer allowed for assigning of the chiral centre at C-5` to have S-
configuration (see Table 3.6a). Both experiments permitted the assignment of the
stereochemistry of talaroflavone to be as shown in 9a, which is reported for the first time.
The structure was confirmed by comparison of UV, 1H, 13
C NMR and mass spectral
data with published data for talaroflavone previously isolated from the soil fungus
Talaromyces flavus for the first time (Ayer and Racok, 1990a).

80
Results

3.1.10. Alternaric acid (10, new compound)

Alternaric acid
Synonym(s) 2-hydroxy-6-(4-hydroxy-2-methyl-5-oxocyclopent-1-enyl)-4-
methoxybenzoic acid
Sample code RR9.2
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 16.9 mg
Physical Description viscous yellow oil
Molecular Formula C14H14O6
Molecular Weight 278 g/mol
20
Optical Rotation [α]D + 75 (c 1.0, MeOH)
Retention time HPLC 19.7 min (standard gradient)
O

HO 1 OH

2` 2
7
3`
1`
3 6
(R)
4` 5` 5
HO 4
O O

300 AH060602 #2 RR11-1 UV_VIS_2


mAU WVL:254 nm

1 - 19,685

[M+H]+
200
[2M+Na]+
[M+Na]+
100

2 - 37,098

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
217.9

[M-H-CO2]-
301.7

-10,0 nm
200 250 300 350 400 450 500 550 595

81
Results

Compound 10 was isolated from the EtOAc extract of rice cultures of Alternaria sp. as
viscous yellow oil (16.9 mg). It exhibited UV absorbances at λmax (MeOH) 217.9, 260 (sh)
and 301.7 nm. The HRESI-MS showed the pseudo-molecular ion [M+Na]+ at m/z 301.0700
(calculated 301.0688, ∆ 0.0012), providing the molecular formula C14H14O6. The 1H NMR
spectrum (see Table 3.6) showed meta-coupled hydrogens at δH 6.40 and 6.07 (each doublet,
J=2.5 Hz) corresponding to H-6 and H-4, respectively, and a methoxy group at δH 3.78, thus
resembling 1H NMR data observed for the aromatic portion of metabolites 6, 7 and 9 (Tables
3.4 and 3.6). Similar to talaroflavone (9), a secondary alcohol group was detected, as a
carbinolic hydrogen at δH 4.38 and a 13C NMR signal at δC 73.2, which was found to be part
of a saturated spin system by coupling to two methylene hydrogens at δH 2.49 (brd, J=17.0
Hz) and 3.00 (dd, J=6.9, 17.0 Hz). Evidence for this substructure was found in the COSY
spectrum (see Table 3.6 and Figure 3.10). A singlet at δH 1.99 (3H) in the 1H NMR with a
13
corresponding C NMR signal at δC 17.8 indicated the presence of a vinyl methyl group. It
was located adjacent to the methylene protons on basis of the long range COSY correlations
observed for 2`-CH3 to 3`-CH2 as well as similar correlations appearing in the ROESY
spectrum (see Table 3.6, Figure 3.10 and 3.11). The ROESY spectrum showed also strong
correlations between the 2`-CH3 group and H-4, as well as between the methoxy signal and
both H-4 and H-6. Furthermore, interpretation of the HMBC spectrum (see Table 3.6 and
Figure 3.12) showed correlations of 2`-CH3 to C-1`, C-2` and C-3` as well as H-3` to C-5`
confirming the five-membered ring substructure. This was also supported by the correlation
observed for H-4 to C-1` in the HMBC spectrum. In addition, the HMBC correlation observed
for H-4 to C-1` established the C1`-C3 bond, attaching both rings.
In order to determine the absolute configuration of the metabolite the modified
Mosher procedure was applied. The observed shift difference between the (S)-MTPA ester
and its (R)-MTPA ester epimer allowed for the assignment of the chiral centre at C-4` to have
R-configuration as shown in 10 (see Table 3.6b).
The compound was identified as the new natural product to which we propose the
name alternaric acid.

82
Results

OH O
O
1 7 HO 1 OH
2 6
2` 2
O 7
5 3`
1` 1`
3 3 6
O (R)
2` (R) (S) 4
OH 4` 5` 5
5` HO 4
4`
3` O O

9 Talaroflavone 10 Alteric acid

OH
O

(R) O
OH
(S)

9a

83
Results

Table 3.6: 1H NMR, NOESY, COSY, ROESY and HMBC data of compounds 9 and 10 at 500 MHz

Nr. 9 9 a) 10
δH δH NOESY HMBC δH δH COSY ROESY HMBC
(MeOD) (DMSO-d6) (MeOD) (MeOD)
1
2
3
4 6.43, brs 6.43, s 2,5,6, 1` 6.44, d (2.0) 6.07, d (2.5) 6 OCH3,CH3 2,5,6,1`
5
6 6.04, d (1.2) 5.90, s 2,4,5,7 6.05, d (2.0) 6.40, d (2.5) OCH3,4 OCH3 2,4,5,7
7
1`
2`
3` 6.34, s 6.38, s 1`,4`,5` 6.34, q (1.5) A:3.00, dd CH3,3`A,4` CH3 1`,2`,4`,5`
(6.9, 17.3)
B:2.49, brd CH3,3`B,4` CH3 2`,4`
(17.0)
4` 4.38, dd CH3,3`A,3`B
(2.8, 6.3)
5` 4.73, s 4.61, d (6.1) 3`,4 1`,4` 4.74, s
2`-CH3 1.85, s 1.74, s 3`,4 1`,2`,3` 1.86, d (1.5) 1.99, s 3`A,3`B,4` 1`,2`,3`,5`
5-OCH3 3.79, s 3.73, s 4,6 5 3.78, s 6 5
5`-OH 6.08, d (6.1)
7-OH 11.00, brs
a) Ayer and Racok, 1990a.

Table 3.6a: Chemical shift difference between the (2`S)-MTPA and (2`R)-MTPA ester of 9

Nr. Chemical shift (δH, in C5D5N, at 500 MHz) ∆


9 (S)-MTPA ester (R)-MTPA ester δS-δR
3` 6.3617 1.7912 1.7279 0.0633
CH3 1.7953 6.4461 6.4603 - 0.0142

Table 3.6b: Chemical shift difference between the (2`S)-MTPA and (2`R)-MTPA ester of 10

Nr. Chemical shift (δH, in C5D5N, at 500 MHz) ∆


10 (S)-MTPA ester (R)-MTPA ester δS-δR
3`A 3.0503 3.3192 3.3166 0.0026
3`B 2.8074 2.8994 2.7070 0.1924
CH3 1.9447 1.9352 1.9320 0.0032

84
Results

Table 3.7: 13C NMR data of compounds 9 and 10 at 125 MHz

Nr. 9 9a 10
d
δC (DMSO-d6) δC (MeOD) δC (MeOD) δC (MeOD)
1 166.6 170.2b) 164.3
2 104.9 106.2 106.5 109.9
3 149.8 150.8 137.3
4 99.2 100.5 103.5c 110.2
5 165.6 168.2 168.5b 164.3
6 102.0 102.5 101.0c 101.4
7 158.2 159.1 159.9 166.0
1` 91.8 94.0 94.2 142.5
2` 168.7 171.8 171.4b 165.2
3` 130.4 130.7 131.4 41.8
4` 200.1 201.5 202.0 73.2
5` 78.2 79.3 79.7 209.0
CH3 13.0 13.9 13.4 17.8
OCH3 55.7 56.1 56.5 55.8
a) Ayer and Racok, 1990a.
b), c) Assignements may be interchanged.
d) Derived from HMBC spectrum.

3` 4 OCH3 CH3
6
5`-OH
5`

H3`/CH3 H4/CH3
CH3

H6/OCH3 H4/OCH3
OCH3
H3`/H5` H4/H5`
5`

5`-OH
4
3`
6

Figure 3.9: NOESY spectrum of compound 9.

85
Results

5-OCH3
CH
2`-CH
3 3

4` 3`A 3`B
(ppm)

2`CH3 CH3/H3`A CH3/H3`B

H3`B/H4` 2.4
3`B

3`A

H3`A/H4` H3`B/H3`A 3.2

5OCH3

4.0

4`

(ppm) 4.4 4.0 3.6 3.2 2.8 2.4 2.0

Figure 3.10: COSY spectrum of compound 10.

6 4 5-OCH3 2`-CH3
3`B
4`
3`A

H3`B/2`CH3

2`-CH3 H4/2`CH3 H3`A/2`CH3

3`B

3`A

5-OCH3 H6/5OCH3

4` H4/5OCH3

4
6

Figure 3.11: ROESY spectrum of compound 10.

86
Results

5- OCH3
2`-CH3

3`A 3`B
(ppm)
3` CH3/C3`
5 OCH3
4` H3`B/C4`
H3`A/C4` 80

6
4
2 120

3
H3`A/C1`
1` CH3/C1`

1,5 OCH3/C5 160


CH3/C2`
2` 7 H3`A/C4` H3`B/C4`

200

5` H3`A/C5` CH3/C5`
(ppm) 4.0 3.6 3.2 2.8 2.4 2.0

Figure 3.12: HMBC spectrum of compound 10.

87
Results

3.1.11. Altenuene and 4`-epialtenuene (11, known, and 12, new compound)

Altenuene and 4`-epialtenuene


Synonym(s) 2,3,4,4a-Tetrahydro-2,3,7-trihydroxy-9-methoxy-4a-methyl-6H-
dibenzo[b,d]pyran-6-one
Sample code RR5.1
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 18.8 mg
Physical Description Viscous yellow oil
Molecular Formula C15H16O6
Molecular Weight 292 g/mol
20
Optical Rotation [α]D racemic
Retention time HPLC 21.8 (4`-epialtenuene), 22.5 min (altenuene) (standard gradient)

O
O
O OH
O OH
7
3` 2
2` 3
1` H
HO 4` 4
1 HO
H
5`
6` 6 5 H O
H O OH
OH

Altenuene 4`-Epialtenuene
AH060803 #3 RR5b-2 UV_VIS_3
180
m AU WVL:280 nm

2 - 22,504

1 - 21,837
100

50

3 - 37,058

m in
-20
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
241.4

4`-Epialtenuene
[M+H]+
280.6

318.3

-10,0
nm
200 250 300 350 400 450 500 550 595

88
Results

70,0
%
Altenuene [M-H]-
243.6

281.7

322.5

-10,0 nm
200 250 300 350 400 450 500 550 595

Compounds 11 and 12 were obtained as an inseperable mixture from the EtOAc


extract of rice cultures of Alternaria sp. in the form of a viscous yellow oil (18.8 mg). The UV
spectrum of compound 11 showed λmax (MeOH) at 243.6, 281.7 and 322.5 nm. Compound 12,
on the other hand, showed UV absorbances at λmax (MeOH) 241.4, 280.6 and 318.3 nm. Their
HRESI-MS showed [M+H]+ at m/z 293.1020 (calculated 293.1025, ∆ 0.0005), indicating the
molecular formula to be C15H16O6. The major compound in the fraction was identified as
altenuene (11) by comparison of UV, 1H, 13
C NMR and mass spectral data with published
data (Bradburn et al., 1994). The 1H NMR spectrum showed a pair of meta-coupled protons
(δH 6.64, H-6, and δH 6.45, H-4) and a methoxy group at δH 3.86, thus reminiscent of the
aromatic portion of metabolites 6-9 (see Tables 3.4, 3.5, 3.6 and 3.8). In addition, the
diastereotopic methylene protons CH2-3 appeared as the AB part of an ABX-type system with
δH 1.96 (dd, J=14.5 and 9.1 Hz) and δH 2.40 (dd, J=14.5 and 3.7 Hz), consistent with axially
and equatorially situated protons, respectively, and with a near antiperiplanar relationship
between H-3`ax and H-4`ax (δH 3.77) in a half-chair conformation (11a). Moreover, a
coupling constant of 2.8 Hz between H-6` (δH 6.21) and H-5` (δH 4.06) was in agreement with
a pseudoequatorial orientation of the C-5` hydroxy group (Bradburn et al., 1994). These data
were in accordance with reported X-ray crystallographic data for altenuene (McPhail et al.,
1973) as well as an in depth analysis of its stereochemistry by Bradburn et al. (Bradburn et
al., 1994). The structure was corroborated by interpretation of the HMBC spectrum showing
correlations of CH3-2` to C1`-C4`, H-4` to C-2` and C-6`, H-5` to C-1` and C-3` as well as H-
6` to C-2`, C-4` and C-1 (see Tables 3.8 and Figure 3.13). Moreover, further evidence was
detected in the ROESY spectrum showing correlations between the 2`-methyl group, the CH2-
3` methylene protons (δH 2.40 and 1.96) and H-5` (δH 4.06) (see Tables 3.8 and Figure 3.14).
Thus, the structure was confirmed to be that of the known compound altenuene, previously
isolated from Alternaria species (McPhail et al., 1973; Bradburn et al., 1994).

89
Results

In contrast, the 1H NMR spectrum of 12 showed similar 1H NMR data for the aromatic
portion of the compound, but significantly different resonances and coupling patterns for the
CH2-3` methylene protons at δH 2.15 (br t, J=12.3 Hz) and δH 2.25 (dd, J=11.9 and 3.7 Hz),
which could be explained with this methylene group situated adjacent to an equatorial 4`-
hydroxy group. This, taken together with strong ROESY correlations between the 2`-methyl
group, one of the methylene protons (δH 2.25), H-4` (δH 3.73), and H-5` (δH 4.20) (see Figure
3.14), indicated the adoption of the alternative half-chair conformation and the placement of
the 4`-hydroxy group in the equatorial position. Thus, the relative stereochemistry shown in
12a was assigned to 12, which identifies the compound as the previously unreported 4`-
epialtenuene. All remaining NMR spectral data were in accord with this conclusion (see
Tables 3.8 and 3.9, and Figure 3.13). Assignment of all signals belonging to 12 was easily
possible because of the lower amount of 12 present in the mixture with 11 (1:2). It was not
possible to determine the absolute stereochemistry as the compounds were obtained as
inseparable mixture. Moreover, the compounds were found to be optically inactive, probably
due to their racemic nature as reported in literature (McPhail et al., 1973).

O OH
7
3` 2
2` 3

R1 4` 1`
4
1
R2
5`
6` 6 5
H O
OH

Nr. Compound R1 R2
11 Altenuene OH H
12 4`-Epialtenuene H OH

H H

3` 4`
H H OH H
H 2` 1` H
5`
2` 1` 6` 6` 5`
O O OH
OH OH H
4` 3`

H H

11a 12a

90
Results

Table 3.8: 1H NMR and HMBC data of compounds 11 and 12 at 500 MHz

Nr. 11 11a 12
δH COSY ROESY HMBC δH (CDCl3- δH COSY ROESY HMBC
(MeOD) DMSO-d6) (MeOD)
1
2
3
4 6.45, d (2.2) 6 2,3,5,6 6.41, d (2.3) 6.46, d (2.2) 6 2,3,5,6
5
6 6.64, d (2.2) 4 6` 4,5,1` 6.53, d (2.3) 6.63, d (2.2) 4 6` 4,5,1`
7


3´α 2.40, dd 3`β,4`α 2`CH3,5´β 2`CH3,1`,2`,4`,5` 2.54, dd 2.15, brt (12.3) 3`β,4`α 5´β 2`CH3,2`,4`,5`
(14.5, 3.7) (14.9, 4.2)
3`β 1.96, dd 3`α,4`α 2`CH3,5´β 2`CH3,1`,2`,4`,5` 1.84, dd 2.25, dd 3`α,4`α 2`CH3 2`CH3,1`,2`,4`,5`
(14.5, 9.1) (14.9, 11.0) (11.9, 3.7)
4´α 3.77, ddd 3`α,β,5`β 6` 2`,5`,6` 3.77, m
(9.1, 5.6, 3.7)
4´β 3.73, ddd 3`α,β,5`β 2`CH3,6`
(12.3, 8.2, 3.7)
5´β 4.06, dd 4`α,6` 2`CH3,3`α,β 1`,3`,4`,6` 4.11, d (4.6) 4.20, dd 4`α,6` 2`CH3,3`α 1`,4`,6`
(5.6, 2.8) (8.2, 2.5)
6´ 6.21, d (2.8) 3`α,β 4´α,6 2`,4`,1 6.41, d (1.7) 6.17, d (2.5) 3`α 4´β,6 2`,4`,1
2`-CH3 1.49, s 3`α,β,5´β 1`,2`,3`,4` 1.49, s 1.54, s 3`β,4´β,5´β 1`,2`,3`
5-OCH3 3.86,s 5 3.86,s 3.85, s 5
a) Bradburn et al., 1994.

Table 3.9: 13C NMR data of compounds 11 and 12 at 125 MHz

Nr. 11 11a 12
δC (MeOD) δC (CDCl3-DMSO-d6) δC (MeOD)
1 140.7 139.1 139.0
2 101.5 101.2
3 165.2 165.7 165.4
4 101.7 100.4 102.0
5 167.8 168.7b 167.9
6 103.7 102.4 103.7
7 170.3 165.7b 169.6
1` 134.7 130.9c 134.2
2` 82.4 81.0 83.5
3` 40.8 40.2 44.5
4` 70.6 69.6 72.0
5` 72.2 72.4 74.2
6` 131.4 132.5c 130.4
2`-CH3 28.0 27.8 26.6
5-OCH3 56.3 55.5 56.3
a) Bradburn et al., 1994.
b), c) Assignements may be interchanged.

91
Results

2`- CH3

2`- CH3*
3`β*
3`α*
5`β 4`α 4`β* 3`α 3`β
5`β*
CH23`*/C3`* (ppm)

H5`β/C3` CH23`/C3` CH3/C3`


40
CH3*/C3`*

H5`β/C4` H3`α/C4` CH23`*/C4`* H3`β/C4` CH3/C4`


H4`α/C5`
H3`β/C5`
H5`β*/C4`* H3`α/C5`
H4`α/C2` CH23`*/C5`* CH3/C2` 80
CH23`*/C2`* CH3*/C2`*
H3`α/C2` H3`β/C2`

120
H5`β/C6`
H5`β*/C6`* H4`α/C6` H3`β*/C2`* CH3*/C1`*
H5`β*/C1`* CH3/C1`
H5`β/C1`
CH23`/C1`

160
OCH3/C5

(ppm) 4.0 3.2 2.4 1.6

Figure 3.13: HMBC spectrum of compounds 11 and 12(*) (obtained as inseperable mixture).

2`-CH3*
3`α* 2`-CH3
5`β
4`α 3`α 3`β

5`β* 4`β* 3`β*

H5`β/2`-CH3
2`-CH3
H5`β*/2`-CH3*
2`-CH3* H4`β*/2`-CH3*
3`β H5`β/H3`β
3`α*
3`β* H5`β*/H3`β*
3`α

4`α 4`β*

5`β
5`β*

Figure 3.14: ROESY spectrum of compounds 11 and 12(*) (obtained as inseperable mixture).

92
Results

3.1.13. 2,5-Dimethyl-7-hydroxychromone (13, known compound)

2,5-Dimethyl-7-hydroxychromone
Synonym(s) 7-Hydroxy-2,5-dimethyl-4H-1-benzopyran-4-one
Sample code RE3.1
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 4.6 mg
Physical Description Viscous yellow oil
Molecular Formula C11H10O3
Molecular Weight 190 g/mol
Retention time HPLC 22.7 min (standard gradient)

5
4a 4
6 3

2
7 8a
HO O
8 1

AH050427b #4 RE3-1 UV_VIS_2


350
mAU WVL:254 nm
2 - 22,741
[M+H]+

200

100

1 - 19,666
3 - 49,828

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
222.2
212.0

249.5

-10,0 nm
200 250 300 350 400 450 500 550 595

93
Results

2,5-Dimethyl-7-hydroxychromone (13) was isolated from the EtOAc extract of liquid


cultures of Alternaria sp. as viscous yellow oil (4.6 mg). It displayed UV absorbances at λmax
(MeOH) 212.0, 222.2, 243.5, 249.5 and 291.0 nm indicating a chromone skeleton. Positive
and negative ESI-MS showed molecular ion peaks at m/z 191.2 [M+H]+ (base peak) and m/z
189.3 [M-H]- (base peak), respectively, indicating a molecular weight of 190 g/mol. The 1H
NMR (see Table 3.10) indicated the presence of two aromatic methyl groups at δH 2.25 and
2.63, a vinyl proton at δH 5.96, and a pair of aromatic meta-coupled protons at δH 6.59 and
6.62 (each d, J=2.2 Hz). The upfield chemical shift of the vinyl proton appearing at δH 5.96
indicated it to reside at C-3 and thus in the α-position of an α,β-unsaturated carbonyl
substructure (Kashiwada et al., 1984; Kimura et al., 1992). The location of the methyl group
at C-5 was confirmed by the HMBC correlations of its protons to C-5, C-6 and C-4a (see
Table 3.10), while both H-6 and H-8 correlated to C-7 and C-4a, thus establishing the
substitution pattern of the aromatic ring. The structure was further confirmed by comparison
1 13
of UV, H, C NMR and mass spectra with published data for 2,5-dimethyl-7-
hydroxychromone (Kashiwada et al., 1984). This is the first report of this natural product
from an endophytic fungal source. However, it was previously isolated from the soil fungus
Talaromyces flavus (Ayer and Racok, 1990a) and its positional isomer, altechromone A, was
obtained from Alternaria sp. (Kimura et al., 1992).

94
Results

10
O

5
4a 4
6 3

2
7 8a
HO O 9
8 1

13 2,5-Dimethyl-7-hydroxychromone

Table 3.10: 1H, 13C NMR and HMBC data of compound 13 at 500 (1H) and 125 MHz (13C)

Nr. 13 13a
δH (DMSO-d6) HMBC δC (DMSO-d6) δH (DMSO-d6) δC (DMSO-d6)
2 163.8 163.9
3 5.96, s 2,4a,9 110.7 5.96, s 116.4
4 178.2 178.4
4a 114.2 114.1
5 141.4 141.5
6 6.59, d (2.2) 4a,7,8,10 116.4 6.60, s 110.5
7 160.9 160.6b
8 6.62, d (2.2) 4a,6,7,8a 100.5 6.60, s 100.4
8a 159.1 159.1b
2-CH3 2.25, s 2,3 19.3 2.28, s 19.2
5-CH3 2.63, s 4a,5,6 22.4 2.66, s 22.3
7-OH 10.58, brs
a) Kashiwada, Nonaka and Nishioka, 1984.
b) Assignements may be interchanged.

95
Results

3.1.14. Altertoxin I (14, known compound)

Altertoxin I
Synonym(s) Hexahydro-1,4,9,12a-tetrahydroxyperylene-3,10-quinone
1,2,7,8,12b-pentahydro-l,4,6b,l0-tetrahydroxyperylene-3,9-dione
Sample code RE5.1
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 3.5 mg
Physical Description Reddish brown powder
Molecular Formula C20H16O6
Molecular Weight 352 g/mol
26
Optical Rotation [α]D + 380 (c 0.2, acetone)
Retention time HPLC 24.5 min (standard gradient)

OH O

4
5 3a 3 2

6 3b 1
6a 12b
OH
6b H OH
9b 12a
7 12
8 10 11
9a
9

OH O

450 AH050624 #3 RE5-1 UV_VIS_2


mAU WVL:254 nm

1 - 24,485

[M-2H2O]+
300

200

100

2 - 25,728

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
257.9
215.0

284.3

-10,0 nm
200 250 300 350 400 450 500 550 595

96
Results

Altertoxin I (14) was isolated from the EtOAc extract of liquid cultures of Alternaria
sp. as reddish brown powder (3.5 mg). It displayed UV absorbances at λmax (MeOH) 215.0,
257.9, 284.3 and 358.0 nm. Positive and negative ESI-MS showed molecular ion peaks at m/z
317.3 [M-2H2O]+ (base peak) and m/z 351.2 [M-H]- (base peak), respectively, indicating a
13
molecular weight of 352 g/mol. The C NMR spectrum (see Table 3.11) contained signals
due to twenty carbon atoms, which together with 1H NMR and mass spectra indicated a
molecular formula of C20H16O6. The 1H resonances were assigned to two AX spin systems of
ortho-coupled aromatic protons, one appearing at δH 8.05 and 7.03 (H-6 and H-5,
respectively), and the other one at δH 7.99 and 6.93 (H-7 and H-8, respectively), and to two
chelated phenolic hydroxy groups (δH 12.72 and 12.31, 4- and 9-OH, respectively). In
addition, C(1)H2-C(2)H2 and C(11)H2-C(12)HOH-C(12a)H fragments were observed and
13
assembled on basis of the COSY spectrum (see Table 3.11). The aromatic C resonances
were attributed to two tetra-substituted aromatic rings and two carbonyl carbon atoms, while
the remaining signals were assigned to three methylene and two methine sp3 hybridized
carbons, one of which is oxygen-bearing. The chelated phenolic hydroxy groups were located
at C-4 and C-9 on basis of the observed HMBC correlations of H-5, H-6 and 4-OH to C-4 as
well as those of H-7, H-8 and 9-OH to C-9 (see Table 3.11). Consequently, the carbonyl
groups were located at C-3 and C-10. Furthermore, HMBC correlations were observed for 4-
OH to C-3a, H-5 to C-3a and C-6a, H-6 to C-4 and C-3b, CH2-1 to C-3, C-3b and C-12b
besides CH2-2 to C-3 and C-12b, thereby establishing the structure of the northern part of the
molecule. The southern part exhibited similar HMBC correlations for the aromatic portion, in
addition to the correlations observed for H-8 to C-9a, CH2-11 to C-9a and C-10 as well as H-
12a to C-6b and C-9a. The attachment of both parts of the molecule was deduced from the
HMBC correlations of H-6 to C-6b, H-7 to C-6a along with those of H-12a to C-1 and C-3b
suggesting the structure of altertoxin I (14) with the phenyl rings being conjugated.
The relative stereochemistry was derived from detailed examination of the proton
spectrum (see Table 3.11). The large value of J11ax-12 (11.1 Hz) and J12-12a (8.1) indicated that
H-12 is trans to H-11ax and H-12a, and that all of these hydrogens are axially positioned.
Accordingly, the hydroxy group 12-OH was placed in an equatorial position. The structure
was further confirmed by comparing UV, 1H, 13
C NMR, mass spectral data and [α]D value
with published data for altertoxin I, previously reported from several Alternaria species
(Okuno et al., 1983; Stack et al., 1986; Hradil et al., 1989).

97
Results

OH O

4
5 3a 3 2

6 3b 1
6a 12b
OH
6b H OH
9b 12a
7 12
8 11
9a 10
9

OH O

14 Altertoxin I

Table 3.11: 1H, 13C NMR, COSY and HMBC data of compound 14 at 500 (1H) and 125 MHz (13C)

Nr. 14 14a
δH (DMSO-d6) COSY HMBC δC (DMSO-d6) δH (DMSO-d6) δC (DMSO-d6)
1ax 2.29, dt (14.0, 4.1) 1eq,2ax,2eq 34.7 2.30, dt (13.0, 3.0) 34.8
1eq 2.96, m 1ax,2ax,2eq 2,3,3b,12a,12b 3.0, m
2ax 3.07, ddd (17.3, 14.0, 4.5) 1ax,1eq,2eq 1,3 33.4 3.0, m 33.5
2eq 2.57, ddd (17.3, 4.1, 2.5) 1ax,1eq,2ax 3,12b 2.59, dt (15.0, 3.0)
3 206.0 206.0
3a 113.7 113.8
3b 140.6 138.4
4 160.9 161.0
5 7.03, d (8.8) 6 3a,3b,4,6a 117.8 7.1, d (8.8) 117.8
6 8.05, d (8.8) 5 3a,3b,4,6b 132.8 8.1, d (8.8) 132.9
6a 123.4 123.5
6b 124.7 124.8
7 7.99, d (8.8) 8 6a,9,9a,9b 132.5 8.0, d (8.8) 132.5
8 6.93, d (8.8) 7,12a 6b,9,9a,9b 115.5 6.9, d (8.8) 115.5
9 160.4 160.4
9a 116.5 116.5
9b 138.3 140.7
10 204.1 204.2
11ax 2.97, dd (15.4, 11.1) 11eq,12 9a,10,12,12a 47.4 3.0, m 47.5
11eq 2.85, dd (15.4, 4.4) 11ax,12 9a,10,12,12a 2.86, m
12 4.52, m 11ax,11eq,12a,12-OH 64.6 4.5, m 64.7
12-OH 5.37, brs 12 51.3 2.86, m 51.4
12a 2.94, d (8.1) 8,12 1,3b,6b,7,9,9a,9b,11,12,12b 67.9 68.0
12b
12b-OH 5.27, s 3b,12a
4-OH 12.72, s 3a,4,5,6 12.4
9-OH 12.31, s 7,8,9,9a 12.7
a) Stack et al., 1986.

98
Results

3.1.15. Tenuazonic acid (15, known compound)

Tenuazonic acid
Synonym(s) 3-Acetyl-1,5-dihydro-4-hydroxy-5-(1-methylpropyl)-2H-pyrrol-2-
one
Sample code RB1, RE1
Biological source Alternaria sp. (from Polygonum senegalense)
Sample amount 128.7 mg
Physical Description Viscous brown oil
Molecular Formula C10H15O3N
Molecular Weight 197 g/mol
20
Optical Rotation [α]D - 134 (c 0.2, CHCl3)
Retention time HPLC 22.3 min (standard gradient)

HO 7
6
4 3
9
5 2
10 8
1 O
N
H H

11

900 AH050427b #3
RE1 UV_VIS_2
mAU WVL:254 nm
[M+H]+
2 - 22,310

600

400

200

1 - 19,328

-100 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
276.8
218.9

563.2

-10,0 nm
200 250 300 350 400 450 500 550 595

99
Results

Tenuazonic acid (15) was obtained as yellowish brown viscous oil (101.2 mg). It
displayed UV absorbances at λmax (MeOH) 218.9 and 276.8 nm. Positive and negative ESI-
MS showed molecular ion peaks at m/z 198.1 [M+H]+ (base peak) and m/z 196.3 [M-H]- (base
peak), respectively, indicating a molecular weight of 197 g/mol and thus the presence of an
odd number of N-atoms in the structure. The 13C NMR spectrum revealing 10 carbon atoms,
together with 1H NMR and mass spectral data supported a molecular formula of C10H15O3N.
The 1H and 13
C NMR spectra (see Table 3.12) indicated three methyl groups, more
specifically a methyl ketone group at δH 2.33 (s) and δC 19.8, a methyl group at δH 0.80 (t,
J=7.2 Hz) and δC 11.7, which based on the multiplicity was assigned as adjacent to a
methylene group, and a methyl group at δH 0.90 (d, J=6.9 Hz) and δC 15.4 located besides a
methine group. In the COSY spectrum the latter signals correlated forming one common spin
system of a 2-butyl side chain. Using a substructure and molecular weight based search the
structure of tenuazonic acid was found to match the obtained data. The structure was further
confirmed by interpretation of the HMBC spectrum (see Table 3.12) in addition to
comparison of UV, 1H, 13
C NMR, mass spectral data and [α]D value with published data for
tenuazonic acid (Nolte et al., 1980). This compound was previously isolated from several
Alternaria (Rosett et al., 1957; Meronuck et al., 1972) and Aspergillus species (Kaczka et al.,
1964) as well as from Piricularia oryzae (Umetsu et al., 1972) and Phoma sorghina (Steyn
and Rabie, 1976).

100
Results

HO 7
6
4 3
9
5 2
10 8
1 O
N
H H

11

15 Tenuazonic acid

Table 3.12: 1H, 13C NMR and HMBC data of compound 15 at 500 (1H) and 125 MHz (13C)

Nr. 15 15a
b
δH (CDCl3) δH (DMSO-d6) COSY HMBC δC (DMSO-d6) δH (CDCl3) δC (CDCl3)
2 174.6 175.6
3 102.2 102.5
4 195.6 195.5
5 3.77, d (3.4) 3.77, d (2.8) 8 2,4,8,9,11 65.9 3.75 67.4
6 184.7 184.0
7 2.43, s 2.33, s 3,6 19.8 2.43 19.4
8 1.94, m 1.78, m 5,9A,9B,11 5,10 36.4 1.92 37.0
9A 1.35, m 1.24, m 8,9B,10 5,8,10,11 23.2 1.30 23.6
9B 1.23, m 1.09, m 8,9A,10 5,8,10,11
10 0.87, t (7.2) 0.80, t (7.2) 9A,9B 8,9 11.7 0.89 11.7
11 0.99, d (6.9) 0.90, d (6.9) 8 5,8,9 15.4 1.00 15.8
NH 7.06, s 8.74, brs 6.22
OH 11.10,brs
a) Nolte, Steyn, and Wessels, 1980.
b) Derived from HMBC spectrum.

101
Results

3.1.16. Bioactivity test results for compounds isolated from the endophytic fungus
Alternaria sp.
The isolated compounds were subjected to bioassays aimed to determine their
cytotoxicity and their protein kinase inhibitory profiles. The results are shown in Tables 3.13
and 3.14.

Table 3.13: Cytotoxicity assay results for the compounds isolated from Alternaria sp. liquid and rice
extracts

Nr. Compound tested L5178Y growth in %* EC50* EC50


(Conc. 10 µg/mL) (µg/mL) (µmol/L)
1 Alternariol 3.7 1.7 6.6
2 Alternariol-5-O-sulphate 0.9 4.5 13.3
3 Alternariol-5-O-methyl 1.8 7.8 28.7
ether
4 Alternariol-5-O-methyl 89.0
ether-4`-O-sulphate
5 3`-Hydroxyalternariol-5- 47.1
O-methyl ether
6 Altenusin 1.2 6.8 23.4
7 Desmethylaltenusin 0.8 6.2 22.5
8 Talaroflavone 92.9
9 Alternaric acid 101.2
10 Alterlactone 11.8
11 and Altenuene and 94.0
12 4`-epialtenuene
13 Altertoxin I 77.3
14 2,5-Dimethyl-7- 95.0
hydroxychromone
15 Tenuazonic acid 57.9
* Data provided by Prof. W. E. G. Müller, Mainz.

All alternariol derivatives as well as alterlactone proved to be highly active against


L5178Y cell line except for 3`-hydroxyalternariol-5-O-methyl ether and alternariol-5-O-
methyl ether-4`-O-sulphate which showed moderate and very weak cytotoxic activity,
respectively. Altenusin and its desmethyl derivative were also highly active in the assay.
Tenuazonic acid was moderately active as well, while altertoxin I showed weak activity.

102
Results

Table 3.14: Protein kinase assay results for the compounds isolated from Alternaria sp. liquid and
rice extracts

Activity on various protein kinases based on IC50 [g/mL]*

CDK4/CycD1

PDGFR-beta
CDK2/CycA
Compound tested

CK2-alpha1

B-RAF-VE
VEGF-R2
VEGF-R3

Aurora-A
Aurora-B
(Conc. 1µg/mL)

IGF1-R
ERBB2
EPHB4
EGF-R

INS-R
ARK5
AKT1

PAK4
PDK1

PLK1
FLT3

MET

TIE2
FAK

SAK

COT
SRC
Ellagic acid A A MA H A A A MA A A 0 0 MA MA A A A A H A MA
Alternariol M0 MMMMA M0 A A A 0 0 MMMA MMA MA MMM
Alternariol-5-O- MMMMMMA M0 A A A 0 0 A MMA MMMMA MMA
sulphate
Alternariol-5-O-methyl M M M M M M M A M M M M 0 0 MM0 MM0 MMMMMM
ether
Alternariol-5-O-methyl 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
ether-4`-O- sulphate
3`-Hydroxyalternariol- M M M M A M A A M A M M 0 0 MM0 A MMMMA MMM
5-O-methyl ether
Altenusin MM0 0 MMM0 0 MMM0 0 0 M 0 M M M M M M M M M
Desmethylaltenusin MM0 0 MMMM0 MMM0 0 0 M 0 M M M M 0 M M M M
Alterlactone M0 0 0 MMMM0 MMM0 0 0 0 0 M M M 0 0 M 0 0 M
Talaroflavone 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Alteric acid 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Altenuene and 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 M 0 0 0 0 0 0 0 0
4`-epialtenuene
Altertoxin I MM0 MMMMM0 MMA 0 0 0 M0 A MMM0 MMMM
2,5-Dimethyl-7- 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
hydroxychromone
Tenuazonic acid 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
H: highly active, A: active, M: moderately active, 0: not active
* Data provided by ProQinase, Freiburg.

Results of protein kinase assay showed a similar activity profile as the cytotoxicity
assay results. All alternariol derivatives as well as alterlactone were active on various protein
kinases, while alternariol-5-O-methyl ether-4`-O-sulphate proved to be an exception with
being inactive for all tested protein kinases. As a reference ellagic acid was used, since it
represents a highly active substance with structural similarity to alternariol. Furthermore,
altenusin, its desmethyl derivative and altertoxin I were also active in the assay.

103
Results

3.2. Compounds isolated from the endophytic fungus Ampelomyces sp.


This endophytic fungal strain of the genus Ampelomyces was isolated from flowers of
Urospermum picroides growing in Egypt. The pure fungal strain was cultivated on liquid
Wickerham medium and solid rice medium. Chemical screening indicated a clear difference
between Ampelomyces extracts obtained from liquid Wickerham medium and rice cultures.
HPLC chromatograms of the EtOAc extract of the fungus grown on solid rice medium
showed altersolanol A (25) and ampelanol (26) as main components. When grown on liquid
medium, the major substance detected in the extract was macrosporin (21) with no traces of
25 or 26 (see Figure 3.15A-B). Similar to Alternaria extracts, the yield of rice cultures was
much higher than that of liquid cultures with a weight ratio of 18:1 of dried extracts,
respectively. Antibacterial, antifungal, cytotoxicity and protein kinase assay results showed
that extracts obtained from rice cultures were much more active in the preliminary biological
screening tests compared to the liquid culture extracts (see Table 3.15).
In this part of the investigation results on the natural products produced by
Ampelomyces sp. when grown in liquid medium and on solid rice medium are presented.

104
Results

200 AH041006 #6 VI1F1IIbEt UV_VIS_1


mAU WVL:235 nm
11 - 32,861

A 21
150
6 - 19,653

100
7 - 20,069
3 - 15,858 10 - 24,343
9 - 22,578 12 - 33,450
8 - 20,772
50 5 - 18,283
4 - 16,426
2 - 15,188
1 - 10,688

-20 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

350 AH060811B #3 ARE UV_VIS_1


mAU WVL:235 nm

B
2 - 16,559
1 - 16,159

26
25
200

12 - 37,169

100
4 - 18,322
11 - 33,863
9 - 31,186
6 7- 20,298
3 - 17,961
- 20,993 8 - 27,74110 - 33,289
5 - 19,777

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

Figure 3.15A-B: EtOAc extracts of Ampelomyces sp. cultures. A: HPLC


chromatogram of EtOAc extract of liquid cultures (Wickerham
medium). B: HPLC chromatogram of EtOAc extract of rice
cultures. 21: Macrosporin. 25: Altersolanol A. 26: Ampelanol.

Table 3.15: Biological screening test results for Ampelomyces liquid and rice extracts

Extracts tested L5178Y growth in % Protein kinase activity Antimicrobial activity


(Conc. 10 µg/mL) (Conc. 1 µg/mL) IZ [mm], 0.5 mg
BS SC CH
Ampelomyces liquid n-BuOH 98.3 0 0 0
Ampelomyces liquid EtOAc 108.2 0 0 0
Ampelomyces liquid MeOH 65.9 0 0 0
Ampelomyces rice EtOAc 0.3 Active 9 0 0
BS: B. subtilis, SC: S. cerevisiae, CH: C. herbarum.

105
Results

3.2.1. Methyltriacetic lactone (16, known compound)

Methyltriacetic lactone
Synonym(s) 4-Hydroxy-3,6-dimethyl-2H-pyran-2-one
Sample code AE4.2
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 4.2 mg
Physical Description viscous yellow oil
Molecular Formula C7H8O3
Molecular Weight 140 g/mol
Retention time HPLC 13.3 min (standard gradient)

OH

4
5 3

2
6 O O

AH060604 #5 AE4-2a UV_VIS_3


300
mAU WVL:280 nm
2 - 13,291
[M+H]+

200

100

4 - MA-Medium H - 37,105
1 - 11,0543 - 15,944
5 - 52,056

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
202.4

[M-H]-
287.9

560.8

-10,0 nm
200 250 300 350 400 450 500 550 595

106
Results

Methyltriacetic lactone (16) was isolated from the EtOAc extract of liquid cultures of
Ampelomyces sp. as viscous yellow oil (4.2 mg). It displayed UV absorbances at λmax (MeOH)
202.4 and 287.9 nm. Positive and negative ESI-MS showed molecular ion peaks at m/z 141.0
[M+H]+ (base peak) and m/z 139.1 [M-H]-, respectively, indicating a molecular weight of 140
g/mol. The 1H and 13
C NMR spectra (Table 3.16) indicated the presence of two aromatic
methyl groups at δH 2.08 and δC 19.2 (6-CH3) as well as δH 1.69 and δC 8.5 (3-CH3), an
aromatic proton singlet at δH 5.88 assigned to H-5 as well as the corresponding tertiary
13
aromatic carbon at δC 101.0. Furthermore, the C NMR spectrum showed a quaternary
carbon at δC 95.0 corresponding to C-3, and two oxygenated quaternary carbons at δC 167.3
and 158.6, corresponding to C-4 and C-6, respectively. The signal at δC 165.3 indicated the
13
presence of a conjugated lactone (C-2). The C NMR spectrum revealing the presence of 7
carbon atoms as well as 1H NMR and mass spectra supported a molecular formula of C7H8O3.
The attachment of the methyl groups at C-3 and C-6 was confirmed by the observed HMBC
correlation of 3-CH3 to C-2, C-3 and C-4, and of 6-CH3 to C-5 and C-6 (see Table 3.16 and
Figure 3.16). The structure was confirmed by comparison of UV, 1H, 13
C NMR and mass
spectral data with published data for methyltriacetic lactone (Fehr et al., 1999), previously
isolated from Penicillium species (Acker et al., 1966; Savard et al., 1994).

107
Results

3.2.2. Ampelopyrone (17, new compound)

Ampelopyrone
Synonym(s) 6-(2-hydroxypropyl)-3-methyl-2-oxo-2H-pyran-4-yl acetate
Sample code AE4.4
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 2.1 mg
Physical Description viscous yellow oil
Molecular Formula C11H13O5
Molecular Weight 226 g/mol
20
Optical Rotation [α]D + 67 (c 0.2, MeOH)
Retention time HPLC 18.2 min (standard gradient)

10 O
11

4
5 3

2
7 6 O O

(R) 8
9
HO

400 AH060604 #10 AE4-4a UV_VIS_3


mAU WVL:280 nm
1 - 18,150

300

200
[M+H]+
[M+Na]+
100
2 - 21,284 3 - 37,109

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
203.2

288.4

560.4

-10,0 nm
200 250 300 350 400 450 500 550 595

108
Results

Compound 17 was isolated from the EtOAc extract of liquid cultures of Ampelomyces
sp. in the form of viscous yellow oil (2.1 mg). It showed UV absorbances at λmax (MeOH)
203.2 and 288.4 nm, showing high similarity to the UV spectrum of methyltriacetic lactone
(16). The HRESI-MS exhibited a strong peak at m/z 227.0910 [M+H]+ indicating a molecular
formula of C11H14O5 (calculated 227.0919, ∆ 0.0009). The 1H and 13
C NMR spectra (Table
3.16) indicated the presence of three methyl groups, i.e. an aromatic methyl group located at
the α-position of the carbonyl group of the conjugated lactone at δH 1.71 and δC 8.4 (3-CH3),
an acetoxy methyl group at δH 1.94 and δC 20.8, and a methyl group at δH 1.19 (d, J=6.3 Hz)
and δC 19.4 (CH3-9). The latter was found to be part of a saturated spin system by coupling to
13
a carbinolic hydrogen at δH 5.02 and a C NMR signal at δC 67.6, indicating a secondary
alcohol group, which in turn was adjacent to a methylene group (δH 2.65 and δC 38.5).
Evidence for this substructure was found in the COSY spectrum. The attachment of the side
chain to C-6 was established by the HMBC correlation of CH2-7 to C-5 (Table 3.16 and
Figure 3.17). Similar to methyltriacetic lactone, an aromatic proton singlet at δH 5.97 assigned
to H-5 as well as the corresponding tertiary aromatic carbon at δC 101.2 were detected in the
1
H and 13C NMR spectra. The 13C NMR spectrum showed also a quaternary carbon at δC 96.7
corresponding to C-3, and two oxygenated quaternary carbons at δC 165.5 and 158.5,
corresponding to C-4 and C-6, respectively. The signal at δC 164.7 indicated the presence of a
conjugated lactone (C-2). The α-pyrone found in 17 was confirmed by the HMBC correlation
of 3-CH3 to C-2, C-3 and C-4 and of H-5 to C-3 (Table 3.16 and Figure 3.17).
In order to determine the absolute configuration of the metabolite we applied the
modified Mosher procedure in an NMR tube. The observed shift differences between the (S)-
MTPA ester and its (R)-MTPA ester epimer led to the assignment of the chiral centre at C-8
of ampelopyrone as shown in 17 (see Table 3.16a).
Thus, 17 was identified as a new natural product for which we suggest the name
ampelopyrone.

109
Results

OH 10 O
11
4 4
5 3 3
5

2 2
6 7 6
O O O O

(R) 8
9
HO

16 Methyltriacetic lactone 17 Ampelopyrone

Table 3.16: 1H,13C NMR and HMBC data of compounds 16 and 17 at 500 (1H) and 125 MHz (13C)

Nr. 16 16a 17
δH HMBC δC δH δC δH COSY HMBC δC
(MeOD) (MeOD) (DMSO-d6) (DMSO-d6) (DMSO-d6) (DMSO-d6)
2 165.3 164.8 164.7
3 95.0 96.2 96.7
4 167.3 165.0 165.5
5 5.88,s 6-CH3,3,4,6 101.0 5.98, s 99.6 5.97, s 7 3,6,7 101.2
6 158.6 159.2 158.5
7 2.65, m 5,8 5,6,8 38.5
8 5.02, m 7,9 67.6
9 1.19, d (6.3) 8 7,8 19.4
10 169.6
11 1.94, s 10 20.8
3-CH3 1.69, s 2,3,4 8.5 1.74, s 8.2 1.71, s 2,3,4 8.4
6-CH3 2.08, s 5,6 19.2 2.14 19.1
8-OH 4.02, brs
a) Fehr et al., 1999.

Table 3.16a: Chemical shift difference between the (2`S)-MTPA and (2`R)-MTPA ester of 17

Nr. Chemical shift (δH, in C5D5N, at 500 MHz) ∆


17 (S)-MTPA ester (R)-MTPA ester δS-δR
5 6.1884 6.4853 6.4909 - 0.0056
7 2.6693 2.7084 2.7138 - 0.0054
9 1.1900 1.1914 1.1908 0.0006
11 2.2442 1.9920 1.9895 0.0025
3-CH3 1.9138 1.8155 1.8313 - 0.0158

110
Results

6-CH3 3-CH3

(ppm)
3-CH3

6-CH3 H5/6-CH3

40

80

3 H5/C3 3-CH3/C3
5
6-CH3/C5

120

6 H5/C6 6-CH3/C6 160


2 H5/C4 3-CH3/C2
4 3-CH3/C5
(ppm) 5.6 4.8 4.0 3.2 2.4 1.6

Figure 3.16: HMBC spectrum of compound 16.

11
3-CH3
9
5
8 8-OH 7
(ppm)
3-CH3
9
11
7
H5/C7 40
CH3-9/C7

CH2-7/C8
8
CH3-9/C8
80

H5/C3
3
5 CH2-7/C5 3-CH3/C3

120

6 H5/C6 CH2-7/C6 160


2,4
10 3-CH3/C2,C4
CH3-11/C10
(ppm) 5.6 4.8 4.0 3.2 2.4 1.6 0.8

Figure 3.17: HMBC spectrum of compound 17.

111
Results

3.2.3. Desmethyldiaportinol (18, new compound)

Desmethyldiaportinol
Synonym(s) 6,8-Dihydroxy-3-(2-hydroxypropyl)-isocoumarin
Sample code AR6.3.1
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 1.5 mg
Physical Description viscous yellowish oil
Molecular Formula C12H12O6
Molecular Weight 252 g/mol
20
Optical Rotation [α]D + 51 (c 0.5, MeOH)
Retention time HPLC 18.3 min (standard gradient)

HO
5 4 9
6 4a 3 10 11 OH

7 O OH
8a 1
8

OH O

160 AH060601 #3 AR16/17-3 UV_VIS_2


mAU WVL:254 nm

+
[2M+Na]+
1 - 18,287 [M+Na]

100

2 - 37,104

50

min
-20
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
244.0

277.3

326.1

-10,0 nm
200 250 300 350 400 450 500 550 595

112
Results

Compound 18 was isolated from the EtOAc extract of rice cultures of Ampelomyces
sp. as a viscous yellow oil (1.5 mg). It showed UV absorbances at λmax (MeOH) 244.0, 277.3
and 326.1 nm characteristic of isocoumarin derivatives (Larsen and Breinholt, 1999). The
HRESI-MS exhibited a prominent peak at m/z 275.0540 [M+Na]+ indicating a molecular
formula of C12H12O6 (calculated 275.0532, ∆ 0.0008). The 1H NMR spectrum (see Table
3.17) displayed characteristic signals attributable to protons H-4, H-5 and H-7, appearing at
δH 6.43 (s), 6.42 (d, J=2.2 Hz) and 6.37 (d, J=2.2 Hz), respectively, in a 3,6,8-trisubstituted
isocoumarin ring system. A downfield one-proton singlet signal (δH 11.14) indicated the
presence of a strongly hydrogen-bonded phenolic proton at C-8. 1H NMR, COSY and NOE
spectra (see Table 3.17) confirmed the substitution pattern and demonstrated the presence of a
CH2CHCH2 fragment consisting of two methylene protons detected at δH 2.76 (dd, J=14.5,
3.7 Hz, H-9A) and 2.52 (dd, J=14.5, 8.8 Hz, H-9B), a carbinolic hydrogen at δH 4.02,
indicating a secondary alcohol group, and a hydroxymethyl group at δH 3.55 (d, J=5.3 Hz)
(see Figure 3.18). The attachment of the side chain at C-3 was further confirmed by the NOE
correlation of H-4 to CH2-9 (Table 3.17). Comparison of UV, 1H, 13C NMR and mass spectral
data with literature data indicated the similarity of 18 to the known diaportinol (18a), in which
the hydroxy group at C-6 is methoxylated (Larsen and Breinholt, 1999). The relative
stereochemistry was derived from the obtained [α]D value found to have identical sign as that
measured for similar structures (Larsen and Breinholt, 1999). Thus, 18 was identified as a
new natural product, and was given the name desmethyldiaportinol.

113
Results

3.2.4. Desmethyldichlorodiaportin (19, new compound)

Desmethyldichlorodiaportin
Synonym(s) 6,8-Dihydroxy-3-(2-hydroxy-3,3-dichloropropyl)-isocoumarin
Sample code AR6.4.1
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 1.0 mg
Physical Description viscous yellowish oil
Molecular Formula C12H10O5Cl2
Molecular Weight 304 g/mol
20
Optical Rotation [α]D + 19 (c 0.3, MeOH)
Retention time HPLC 24.6 min (standard gradient)

Cl

HO
5 4 9
6 4a 3 10 11 Cl

7 O OH
8a 1
8

OH O

180 AH060601 #4 AR18-2 UV_VIS_2


mAU WVL:254 nm

1 - 24,641
150

100

2 - 37,106
[M+H]+

50

-20 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
244.2

277.5

326.9

-10,0 nm
200 250 300 350 400 450 500 550 595

114
Results

Compound 19 was isolated from the EtOAc extract of rice cultures of Ampelomyces
sp. in the form of a viscous yellow oil (1.0 mg). It displayed UV absorbances at λmax (MeOH)
244.2, 277.5 and 326.9 nm characteristic of isocoumarin derivatives and very similar to those
reported for desmethyldiaportinol (18). The HRESI-MS exhibited a prominent peak at m/z
326.9800 [M+Na]+ indicating a molecular formula of C12H10O5Cl2 (calculated 326.9802, ∆
0.0002). The negative ESI-MS showed the presence of molecular ion peaks at m/z 303.5,
305.4 and 307.4 [M-H]-, with the distinctive isotope pattern caused by two chloro atoms in the
molecule, indicating the dichlorosubstitution in this compound. The 1H NMR spectrum (see
Table 3.17) showed the characteristic signals assigned to protons H-4, H-5 and H-7 observed
at δH 6.50 (s), 6.42 (d, J=2.0 Hz) and 6.38 (d, J=2.0 Hz), respectively, indicating a 3,6,8-
trisubstituted isocoumarin ring system, as in the case of desmethyldiaportinol. 1H NMR,
COSY and NOE spectra (see Table 3.17) confirmed the substitution pattern and demonstrated
the presence of an analogous CH2CH(OH)CH as described above, with two methylene
protons detected at δH 2.98 (dd, J=14.5, 3.4 Hz, H-9A) and 2.76 (dd, J=14.5, 9.2 Hz, H-9B),
an oxygenated carbinolic hydrogen at δH 4.39, and a methine group at δH 6.20 (d, J=3.1 Hz)
1
(see Figure 3.18). Thus, the H NMR data were highly similar to those of
desmethyldiaportinol (18), except for the marked downfield shift of H-11 which indicated
dichlorosubstitution at this position (Table 3.17). The attachment of the side chain at C-3 was
confirmed by the NOE correlation of H-4 to CH2-9 (Table 3.17). Thus, 19 was identified as
the 10-deoxy-10,10-dichloro congener of 18 and represents a new natural product for which
we suggest the name desmethyldichlorodiaportin.

115
Results

3.2.5. (+)-Citreoisocoumarin (20, known compound)

(+)-Citreoisocoumarin
Synonym(s) 3-(2-Hydroxypentyl-4-ketone)-6,8-dihydroxyisocoumarin
Sample code AE4.3.1
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 0.5 mg
Physical Description viscous yellowish oil
Molecular Formula C14H14O6
Molecular Weight 278 g/mol
20
Optical Rotation [α]D + 15 (c 0.1, MeOH)
Retention time HPLC 20.2 min (standard gradient)

HO
5 4 9
10 12 13
6 4a 3 11

7 O OH O
8a 1
8

OH O

400 AH060727 #2
AE4-5 UV_VIS_2
mAU WVL:254 nm

1 - 20,166 [2M+H]+
300

[M+H]+
200

[M+Na]+

100

2 - 37,082

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
244.5

277.5

326.3

-10,0 nm
200 250 300 350 400 450 500 550 595

116
Results

(+)-Citreoisocoumarin (20) was isolated from the EtOAc extract of liquid cultures of
Ampelomyces sp. as viscous yellow oil (0.5 mg). It showed UV absorbances at λmax (MeOH)
244.5, 277.5 and 326.3 nm characteristic of isocoumarin derivatives as described for 18 and
19. Positive and negative ESI-MS showed molecular ion peaks at m/z 279.1 [M+H]+ and m/z
277.5 [M-H]- (base peak), respectively, indicating a molecular weight of 278 g/mol. The 1H
NMR spectrum (see Table 3.18) showed the characteristic signals corresponding to protons
H-4, H-5 and H-7 observed at δH 6.39, 6.36 (d, J=2.0 Hz) and 6.3 (d, J=2.0 Hz), respectively,
indicating a 3,6,8-trisubstituted isocoumarin ring system, similar to previously discussed
isocoumarin derivatives 18 and 19 (see Table 3.17). It showed two methylene groups, with
the protons resonating at δH 2.71 (dd, J=16.1, 4.4 Hz, H-11A), 2.65 (dd, J=16.1, 7.8 Hz, H-
11B), 2.66 (dd, J=14.3, 4.4 Hz, H-9A) and 2.57 (dd, J=14.3, 8.2 Hz, H-9B), both of which
coupled to an oxygenated methine group at δH 4.44, indicating a secondary alcohol group.
Additionally, a singlet at δH 2.13 (3H) was assigned to a methyl group adjacent to a keto
function. Thus, 20 was identified as the known (+)citreoisocoumarin, and its UV, 1H NMR,
mass spectral data and [α]D value were in agreement with published data (Lai et al., 1991;
Watanabe et al., 1998). This compound was originally isolated from Aspergillus (Watanabe et
al., 1998, 1999) and Penicillium species (Lai et al., 1991).

117
Results

R3

R1O
5 4 9
6 4a 3 10 11 R2

7 O OH
8a 1
8

OH O

Nr. Compound R1 R2 R3
18 Desmethyldiaportinol H OH H
18a Diaportinol CH3 OH H
19 Desmethyldichlorodiaportin H Cl Cl
19a Dichlorodiaportin CH3 Cl Cl
20 Citreoisocoumarin H COCH3 H

Table 3.17: 1H NMR, COSY and NOE data of compounds 18 and 19 at 500 MHz

Nr. 18 18aa 19 19aa


δH δH COSY NOE δH δH δH COSY NOE δH
(MeOD) (Acetone- (Acetone- (MeOD) (Acetone- (Acetone-
d6) d 6) d6) d6)
4 6.38, s 6.43, s 9A 5,9A,9B 6.46, s 6.41, s 6.50, s 9A 5,9B,9A 6.57, s
5 6.29, s 6.42, d (2.2) 7 4 6.53, s 6.30, s 6.42, d (2.0) 7 4 6.53, d (2.2)
7 6.29, s 6.37, d (2.2) 5 6.53, s 6.30, s 6.38, d (2.0) 5 6.47, d (2.2)
9A 2.74, dd 2.76, dd 4,9B,10 2.79, dd 2.96, dd 2.98, dd 4,9B,10 3.02, dd
(14.6, 4.1) (14.5, 3.7) (14.7, 3.4) (14.8, 3.1) (14.5, 3.4) (14.7, 3.3)
9B 2.52, dd 2.52, dd 9A,10 2.56, dd 2.72, dd 2.76, dd 9A,10 2.80, dd
(14.6, 8.8) (14.5, 8.8) (14.7, 8.8) (14.8, 9.4) (14.5, 9.2) (14.7, 9.2)
10 3.99, m 4.02, m 9A,9B,11 4.04, m 4.29, ddd 4.39, ddd 9A,9B,11 4.41, m
(9.4, 3.4, 3.1) (9.2,3.4, 3.1)
11 3.54, d (5.3) 3.55, d (5.3) 10 3.58, d (5.4) 6.02, d (3.4) 6.20, d (3.1) 10 10 6.20, d (3.3)

OCH3 3.91, s 3.90, s


6-OH
8-OH 11.14, brs 11.16, s 11.08, s
10-OH 5.25, brs
a) Larsen and Breinholt, 1999.

118
Results

Table 3.18: 1H NMR data of compound 20 at 500 MHz

Nr. (+)-20 (-)-20a (+)-20b


δH (MeOD) δH (Acetone-d6) δH (MeOD) δH (Acetone-d6)
4 6.33, s 6.39, s 6.33, s 6.53, s
5 6.24, d (2.2) 6.36, d (2.0) 6.39, d (2.2) 6.49, d (2.1)
7 6.23, d (2.2) 6.33, d (2.0) 6.32, d (2.2) 6.45, d (2.1)
9 A 2.66, dd (14.5, 5.0) A 2.66, dd (14.3, 4.4) 2.67, d (6.3) A 2.83, dd (14.5, 4.8)
B 2.58, dd (14.5, 7.5) B 2.57, dd (14.3, 8.2) B 2.72, dd (14.5, 8.1)
10 4.43, m 4.44, m 4.47, m 4.55, m
11 2.68, d (5.9) A 2.71, dd (16.1, 4.4) 2.71, d (6.3) A 2.86, dd (16.3, 4.7)
B 2.65, dd (16.1, 7.8) B 2.80, dd (16.3, 7.8)
13 2.17, s 2.13, s 2.23, s 2.23, s
6-OH 9.70, brs
8-OH 11.22, brs
a) Lai et al., 1991.
b) Watanabe et al., 1998.

5 11 5,7
4
7 9A
10 11
9B 4
10 9A 9B
(ppm)
H9B/ H9A (ppm)
9B
9B H9B/ H9A
9A H9B/H10 H9B/ H10
H9A/H10 9A
H9A/H4 H9A/ H4 H9A/ H10
3.2
3.2

11

4.0
10 4.0
10
H11/H10
H10/ H11

4.8
4.8

5.6 5.6

11
5 5,7
7
6.4 6.4
4 4

(ppm) 5.6 4.8 4.0 3.2 2.4 (ppm) 5.6 4.8 4.0 3.2

Figure 3.18: COSY spectra of compounds 18 and 19.

119
Results

3.2.6. Macrosporin (21, known compound)

Macrosporin
Synonym(s) 1,7-Dihydroxy-3-methoxy-6-methylanthraquinone
Sample code AE2.3, AR2
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 21.6 mg
Physical Description yellow crystals
Molecular Formula C16H12O5
Molecular Weight 284 g/mol
Retention time HPLC 33.2 min (standard gradient)

4 5
O
3 4a 10 10a 6

2 7
9a 9 8a OH
1 8
OH O

1.000 AH060322 #5 AmpHxD1/1 UV_VIS_3


mAU WVL:280 nm
1 - 33,222

800
[M+H]+
600

400

200
2 - 37,083

-100 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
284.4
225.4

380.2

-10,0 nm
200 250 300 350 400 450 500 550 595

120
Results

Macrosporin (21) was isolated from the EtOAc extract of liquid and rice cultures of
Ampelomyces sp. as yellow crystals (21.6 mg). It exhibited UV absorbances at λmax (MeOH)
225.4, 284.4 and 380.2 nm suggesting an anthraquinone as the basic structure. Positive and
negative ESI-MS showed molecular ion peaks at m/z 285.3 [M+H]+ (base peak) and m/z 283.6
[M-H]- (base peak), respectively, indicating a molecular weight of 284 g/mol. 1H and 13
C
NMR spectra (see Table 3.19) indicated the presence of an aromatic methyl group at δH 2.34
13
and δC 16.4, and a methoxy singlet at δH 4.00 and δC 56.7. The C NMR spectrum showed
resonances at δC 187.6 and 181.2, indicative of a quinone, and three aromatic carbons
connected to oxygens at δC 167.0, 165.8 and 162.5. The 1H NMR spectrum revealed the
presence of four aromatic protons, two of which were doublets occurring at δH 7.19 and 6.80
with a coupling constant of 2.5 Hz, indicating their meta disposition in the ring system,
corresponding to H-4 and H-2, respectively, while the remaining two aromatic proton singlets
at δH 7.95 and 7.67, were assigned to the para-coupled protons H-5 and H-8, respectively, of
the other aromatic ring. The 13C NMR spectrum displayed 16 carbon atoms, and together with
1
H NMR and mass spectral data the molecular formula of C16H12O5 was derived. The
compound was thus identified as the known macrosporin, which was confirmed by
interpretation of HMBC spectra (see Table 3.19 and Figure 3.19) and comparison of UV, 1H,
13
C NMR and mass spectral data with published data (Suemitsu et al., 1984, 1989).
Macrosporin was previously reported from several Alternaria species (Stoessl et al., 1983;
Lazarovits et al., 1988; Suemitsu et al., 1989) as well as from Phomopsis juniperovora
(Wheeler and Wheeler, 1975), Dactylaria lutea (Becker et al., 1978), Dichotomophthora
lutea (Hosoe et al., 1990) and Pleospora sp. (Ge et al., 2005).

121
Results

3.2.7. Macrosporin-7-O-sulphate (22, new compound)

Macrosporin-7-O-sulphate
Synonym(s) 1-Hydroxy-3-methoxy-6-methylanthraquinone-7-sulphate
Sample code AME3, AR8.4.2
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 7.1 mg
Physical Description yellow crystals
Molecular Formula C16H12O8S
Molecular Weight 364 g/mol
Retention time HPLC 26.8 min (standard gradient)

4 5
O
3 4a 10 10a 6

7
2 9a 9 8a OSO3H
1 8
OH O

AH060326 #3 AmpMeEt65 UV_VIS_3


100
mAU WVL:280 nm
1 - 26,831 [M+H]+
80

2 - 36,963

60

40
[M+H-SO3]+

20

min
-10
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-HSO3]-
267.3
278.2
203.1

[M-H]-

-10,0 nm
200 250 300 350 400 450 500 550 595

122
Results

Compound 22 was isolated from the MeOH extract of liquid cultures and EtOAc
extract of rice cultures of Ampelomyces sp. in the form of yellow crystals (7.1 mg). Its UV
spectrum showed λmax (MeOH) at 203.1, 267.3, 278.3 and 420.0 nm. The HRESI-MS
exhibited a prominent peak at m/z 408.9970 [M+2Na]+ indicating a molecular formula of
C16H12O8S (calculated 408.9969, ∆ 0.0001). Comparison of 1H, 13
C NMR and HMBC data
(Table 3.19 and Figure 3.19) with those measured for macrosporin (21) showed good
accordance except for the downfield shifts observed for H-8, as well as the upfield shift of C-
7, by 6.5 ppm, and downfield shifts of C-6 and C-8, by 5.0 and 7.2 ppm, respectively,
indicating the presence of a sulphate substitution at C-7 (Ragan, 1978). This assumption was
corroborated by the fragment formed through loss of 80 mass units in the mass spectra of 22
and the hypsochromic shift in the UV spectrum of 22 compared to that of 21, which is
attributed to the electron withdrawing effect of the sulphate group (Plasencia and Mirocha,
1991). The compound was thus identified as the new natural product macrosporin-7-O-
sulphate.

4 5
O
3 4a 10 10a 6

7
2 9a 9 8a OR
1 8
OH O

Nr. Compound R
21 Macrosporin H
22 Macrosporin sulphate SO3H

123
Results

Table 3.19: 1H, 13C NMR and HMBC data of compounds 21 and 22 at 500 (1H) and 125 MHz (13C)

Nr. 21 21a,b 22
δH HMBC δC δH δC δH HMBC δC
(DMF-d7) (DMF-d7) (THF-d8)a (THF-d8)b (MeOD) (MeOD)
1 165.8 166.4 166.0
2 6.80, d (2.5) 1,3,4,9a 106.0 6.72, d (2.5) 106.1 6.74, d (2.5) 1,3,4,9a 107.0
3 167.0 167.4 167.0
4 7.19, d (2.5) 2,9a,10 107.8 7.30, d (2.5) 108.0 7.27, d (2.5) 2,3,9a,10 108.0
4a 136.0 136.5
5 7.95, s CH3,7,8a,9,10 130.8 8.00, s 131.1 8.06, s CH3,7,8a,9,10 131.0
6 133.0 133.0 138.0
7 162.5 162.3 156.0
8 7.67, s 6,7,8a,9,10,10a 111.8 7.54, s 111.6 8.36, s 6,7,8a,9,10,10a 119.0
8a 134.1 134.6 134.0
9 187.6 187.9 187.0
9a 111.1 111.5 111.0
10 181.2 181.2 182.0
10a 126.0 126.9 130.0
CH3 2.34, s 5,6,7,8 16.4 2.33, s 16.4 2.46, s 5,6,7,8 17.0
OCH3 4.00, s 3 56.7 3.93, s 56.4 3.92, s 3 56.0
a) Suemitsu et al., 1984.
b) Suemitsu et al., 1989.

OCH3 OCH3
CH3
CH3

8
5 4 2 8 5 4 2

(ppm) (ppm)
2
4
9a
8
CH3/C8
C8 120
120
CH3/C8
10a
5
6 H8/C6
8a
4a CH3/C6 C6
140
140
H8/C6 CH3/C6

C7
H5/C7
7 H8/C7
H8/C7 160
CH3/C7 160

1 H5/C7 CH3/C7
3

180 180
10

(ppm) 8.00 (ppm) 8.00 7.00 6.00 5.00 4.00 3.00


7.00 6.00 5.00 4.00 3.00 2.00

Figure 3.18: HMBC spectra of compounds 21 and 22.

124
Results

3.2.8. 3-O-Methylalaternin (23, known compound)

3-O-Methylalaternin
Synonym(s) 1,2,8-Trihydroxy-6-methoxy-3-methylanthraquinone
Sample code AE2.2, AR4
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 3.5 mg
Physical Description orange crystals
Molecular Formula C16H12O6
Molecular Weight 300 g/mol
Retention time HPLC 34.0 min (standard gradient)

4 5
O
3 4a 10 10a 6

2 7
9a 9 8a
OH
1 8

OH O OH

AH060904 #6 ARE-14 UV_VIS_3


400
mAU WVL:280 nm
1 - 33,985
[M+H]+
300

200

100

2 - Weichmacher (Phthalat) - 37,348

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
281.5
229.8
207.3

-10,0 nm
200 250 300 350 400 450 500 550 595

125
Results

3-O-Methylalaternin (23) was isolated from the EtOAc extract of liquid and rice
cultures of Ampelomyces sp. as orange crystals (3.5 mg). It showed UV absorbances at λmax
(MeOH) 207.3, 229.8, 281.5 and 435.0 nm suggesting an anthraquinone as the basic structure
and showing high similarity to UV spectra of macrosporin (21). Positive and negative ESI-
MS showed molecular ion peaks at m/z 301.6 [M+H]+ (base peak) and m/z 299.8 [M-H]- (base
peak), respectively, indicating a molecular weight of 300 g/mol, and further indicating an
increase of 16 mass units compared to 21 which established the composition C16H12O6. UV
and NMR spectra of 23 had close similarity to those of 21. The 1H NMR spectrum (see Table
3.20) resembled that of 21 except for the absence of the para-coupled pair of aromatic protons
and the presence of an aromatic proton singlet at δH 7.62 assigned to H-5. The meta-coupled
pair appeared as doublets occurring at δH 7.24 and 6.84 with a coupling constant of 2.0 Hz,
13
corresponding to H-4 and H-2, respectively. The C resonances at δC 187.6 and 180.1
indicated a quinone structure, and both 1H NMR and 13C NMR data (see Table 3.20) indicated
the presence of an aromatic methyl group at δH 2.34 and δC 16.4, and a methoxy singlet at δH
4.01 and δC 56.7. Proton signals were assigned to their corresponding carbons by HMBC (see
Table 3.20 and Figure 3.19). The structure was confirmed by comparison of UV, 1H , 13
C
NMR and mass spectral data with published data for alaternin (23a) (Lee et al., 1998). 3-O-
Methylalaternin was previously reported from Alternaria species (Stoessl, 1969b; Stoessl et
al., 1983).

126
Results

3.2.9. 3-O-Methylalaternin-7-O-sulphate (24, new compound)

3-O-Methylalaternin-7-O-sulphate
Synonym(s) 1,8-Dihydroxy-6-methoxy-3-methylanthraquinone-2-sulphate
Sample code AME3, AR8.4.1
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 2.5 mg
Physical Description orange crystals
Molecular Formula C16H12O9S
Molecular Weight 380 g/mol
Retention time HPLC 26.5 min (standard gradient)

4 5
O
3 4a 10 10a 6

2 7
9a 9 8a
OSO3H
1 8

OH O OH

AH060326 #6 AmpMeEt68 UV_VIS_3


80,0
mAU WVL:280 nm
1 - Hydroxyemodin - 26,501
2 - MA-Medium H - 37,118 [M+H]+
60,0

40,0
[M+H-SO3]+

20,0

min
-10,0
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-HSO3]-
226.3

273.1

438.4

[M-H]-

-10,0 nm
200 250 300 350 400 450 500 550 595

127
Results

Compound 24 was isolated from the MeOH extract of liquid cultures as well as EtOAc
extract of rice cultures of Ampelomyces sp. in the form of orange crystals (2.5 mg). It had UV
absorbances at λmax (MeOH) 226.3, 273.1 and 438.4 nm. The HRESI-MS exhibited a
prominent peak at m/z 424.9910 [M+2Na]+ , consistent with a molecular formula of
C16H12O9S (calculated 424.9919, ∆ 0.0009). The 1H NMR spectrum (see Table 3.20) showed
signals for a methyl group at δH 2.52, a methoxy group at δH 3.94, a pair of meta-coupled
aromatic protons at δH 7.31 and 6.77 (J=2.5 Hz), and an aromatic singlet at δH 7.65,
corresponding to H-4, H-2 and H-5, respectively. The aromatic methyl group showed HMBC
correlations to carbons at δC 146.0, 143.5 and 122.5, assigned to C-7, C-6, and C-5,
respectively. Similar to macrosporin sulphate (22), comparison of the chemical shifts
observed for C-7 and C-6 to those recorded for the respective carbon atoms in methylalaternin
(23) (see Table 3.20 and Figure 3.19) clearly revealed a prominent upfield shift for C-7 and a
downfield shift of C-6, thus indicating the presence of a sulphate substitution at C-7 (Ragan,
1978). Presence of the sulphate substituent was confirmed by the fragment formed by loss of
80 mass units in the mass spectrum of 24 and the hypsochromic shift in the UV spectrum of
24 compared to that of 23, which is attributed to the electron withdrawing effect of the
sulphate group (Plasencia and Mirocha, 1991). The compound was thus identified as 3-O-
methylalaternin-7-O-sulphate. This is the first example of the isolation of 24 as a natural
product.

4 5
R1O 10a
3 4a 10 6

2 7
9a 9 8a
OR2
1 8

OH O OH

Nr. Compound R1 R2
23 Methylalaternin CH3 H
23a Alaternin H H
24 Methylalaternin sulphate CH3 SO3H

128
Results

Table 3.20: 1H, 13C NMR and HMBC data of compounds 23 and 24 at 500 (1H) and 125 MHz (13C)

Nr. 23 23aa 24
δH HMBC δC δH δC δH (MeOD) HMBC δC
(DMF-d7) (DMF-d7) (DMSO-d6) (DMSO-d6) (MeOD)b
1 165.7 164.4
2 6.84, d (2.0) 1,3,4,9a 106.1 6.72, d (2.5) 107.2 6.77, d (2.5)
3 167.5 165.6 167.0
4 7.24, d (2.0) 2,9a,10 107.9 7.30, d (2.5) 108.5 7.31, d (2.5)
4a 136.9 131.3
5 7.62, s CH3,7,8a,10 123.3 7.47, s 122.9 7.65, s 122.5
6 132.0 135.6 143.5
7 152.5 150.1 146.0
8 149.2
8a 114.9 113.9
9 187.6 190.1
9a 110.0 109.0
10 180.1 179.9
10a 124.0 122.9
CH3 2.34, s 5,6,7 16.4 16.2 2.52, s 5,6,7
OCH3 4.01, s 3 56.7 3.94, s 3
a) Lee et al., 1998.
b) Derived from HMBC spectrum.

OCH3 CH3

OCH3 CH3

(ppm)

120

C5 C5
CH3/C5
CH3/C5 128

C6 CH3/C6
136

C6
CH3/C6
144
C7
CH3/C7
152
CH3/C7
C7
160

OCH3/C3 OCH3/C3 168

(ppm) 3.6 3.2 2.8

Figure 3.19: HMBC spectra of compounds 23 and 24.

129
Results

3.2.10. Altersolanol A (25, known compound)

Altersolanol A
Synonym(s) 7-Methoxy-2-methyl-1β,2α,3α,4β,5-pentahydroxy-1,3,4-
trihydroanthraquinone
Sample code AR6.2.1
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 15.7 mg
Physical Description orange yellow crystals
Molecular Formula C16H16O8
Molecular Weight 336 g/mol
22
Optical Rotation [α]D - 149 (c 1.2 mg/mL, EtOH)
Retention time HPLC 16.2 min (standard gradient)

O OH

8
O
7 8a 9 9a 1
2 OH

6
10a 10 4a 4 3
OH
5

OH O OH

AH061120b #3 AR-A UV_VIS_2


300
mAU WVL:254 nm
1 - 16,201
[M+H]+

200

100

2 - 37,478

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
220.5

[M-H]-
269.5

434.5

-10,0 nm
200 250 300 350 400 450 500 550 595

130
Results

Altersolanol A (25) was isolated from the EtOAc extract of rice cultures of
Ampelomyces sp. as orange yellow crystals (15.7 mg). It showed UV absorbances at λmax
(MeOH) 220.5, 269.5 and 434.5 nm suggesting a quinone as the basic structure. Positive and
negative ESI-MS showed molecular ion peaks at m/z 337.3 [M+H]+ (base peak) and m/z 335.3
[M-H]- (base peak), respectively, indicating a molecular weight of 336 g/mol, while
inspection of the NMR data suggested a molecular formula of C16H16O8. The 1H NMR
spectrum (see Table 3.21) displayed signals for four alcoholic hydroxyl groups, three doublets
at δH 5.67, 5.03 and 4.88 (J=5.9, 5.3, and 6.3 Hz, respectively) and one singlet at δH 4.46,
assigned for 1-OH, 4-OH, and 3-OH, respectively. In addition, the spectrum contained a
broad singlet (chelated phenol) at δH 12.11 (5-OH). These signals were not observed in the 1H
NMR spectrum measured in MeOD, confirming their facile exchange. Furthermore, a singlet
was detected at δH 1.23 corresponding to an aliphatic methyl group (2-CH3), together with
three carbinolic protons at δH 4.47 (H-4), 4.31 (H-1) and 3.63 (H-3). The pair of meta-coupled
aromatic protons at δH 7.00 (H-8) and 6.81 (H-6) and the aromatic methoxy group (δH 3.89)
were comparable to the respective signals of metabolites 21-24 (see Tables 3.19 and 3.20).
The nature of the non-aromatic carbocycle was evident from the COSY spectrum (see Table
3.21), establishing the planar structure of 25 as identical to altersolanol A (25a) (Stoessl,
1969a). Moreover, correlations of 2-CH3 with H-3, 1-OH, and 4-OH in the ROESY spectrum
indicated that also the relative stereochemistry corresponded to that of altersolanol A (25a),
which was also in agreement with the observed coupling constants. This assignment was
further corroborated by the very similar experimental UV, 1H, 13
C NMR, mass spectral data
and [α]D value obtained for 25a in comparison to published data for altersolanol A (Yagi et
al., 1993; Okamura et al., 1993, 1996). Altersolanol A was previously isolated from several
Alternaria species (Stoessl et al., 1983; Lazarovits et al., 1988; Yagi et al., 1993; Okamura et
al., 1993, 1996).

131
Results

3.2.11. Ampelanol (26, new compound)

Ampelanol
Synonym(s) 7-Methoxy-2-methyl-1β,2α,3α,4β,5,9α-hexahydroxy-1,3,4,4a,9,9a-
hexahydroanthracen-10-one
Sample code AR8.2.1, AR9.4
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 18.6 mg
Physical Description white crystals
Molecular Formula C16H20O8
Molecular Weight 340 g/mol
20
Optical Rotation [α]D - 64 (c 0.5 mg/mL, MeOH)
Retention time HPLC 16.4 min (standard gradient)

OH OH
H
O
8
7 8a 9 1
9a 2 OH
4a
6 10a 10 4 3
OH
5
H
OH O OH

AH060605 #3 ARE15-1b UV_VIS_3


900
mAU WVL:280 nm
[M+H]+
1 - 16,409

600 [2M+Na]+

400

200

2 - 37,104

min
-100
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
283.4

[M-H]-
218.1
231.6

[M+HCOO]-
[2M+HCOO]-

-10,0 nm
200 250 300 350 400 450 500 550 595

132
Results

Compound 26 was isolated from the EtOAc extract of rice cultures of Ampelomyces
sp. in the form of white crystals (18.6 mg). It had UV absorbances at λmax (MeOH) 218.1,
231.6, 283.4 and 318.0 nm. The HRESI-MS exhibited a prominent peak at m/z 341.1230
[M+H]+ indicating a molecular formula of C16H20O8 (calculated 341.1236, ∆ 0.0006) as well
as an increase of four mass units compared to altersolanol A (25). The 1H NMR spectrum (see
Table 3.21) contained five exchangeable alcoholic hydroxyl groups, two doublets at δH 5.49
and 5.06, a broad singlet at δH 4.41 and two singlets at δH 4.46 and 4.21, assigned to 9-OH, 1-
OH, 3-OH, 4-OH, and 2-OH, respectively. In addition, a singlet for a chelated phenol
appeared at δH 12.57 which was likewise exchangeable and was attributed to 5-OH. A singlet
corresponding to an aliphatic methyl group was detected at δH 1.20 (2-CH3), while four
carbinolic protons resonated at δH 4.67 (H-9), 3.82 (H-4), 3.76 (H-1), and 3.36 (H-3). Similar
to compounds 21-25, the meta-coupled H-8 and H-6 appeared at δH 6.72 and 6.35, while the
aromatic methoxy group was detected at δH 3.83. In the COSY spectrum, the less shielded
aryl proton H-8 exhibited a long range correlation to a peri-proton (H-9), which in turn
coupled to both the hydroxy signal at δH 5.49 (9-OH) and the ring junction proton at δH 2.30
(H-9a). These results indicated that the quinone carbonyl at C-9 in 25 had been reduced to a
hydroxy group at the respective position in 26, while the double bond between C-9a and C-4a
in 25 likewise was reduced in 26, also in accord with the increase in the molecular weight of 4
amu compared to altersolanol A and the absence of color for this compound. The complete
aliphatic spin system comprising H-9, H-9a, H-1, H-4a, H-4, and H-3, together with the
corresponding hydroxy functions, was clearly discernible in the COSY spectrum (see Figure
3.20). Furthermore, in the HMBC spectrum (see Figure 3.22) the correlations attributed to the
two protons at the ring junction, i.e. H-9a (to C-4a and C-9) and H-4a (to C-4, C-9, C-9a, and
C-10), as well as the correlation of H-8 to C-9 fully supported the assignment of the planar
structure as depicted.
The relative stereochemistry was deduced from the coupling constants in the 1H NMR
spectrum as well as correlations in the ROESY spectrum (see Table 3.21). The large values of
J3-4 (9.4 Hz), J4-4a (9.4 Hz), J4a-9a (13.2 Hz) and J9-9a (10.5 Hz) could only be explained by a
series of mutual diaxial relationships and thus proved that all of these hydrogens were axially
positioned, while correspondingly, the 2.2 Hz coupling between H-9a and H-1 indicated an
equatorial position for the latter. Correlations of H-9 to H-1 and H-4a, 2-CH3 to both H-1 and
H-3, as well as H-4a to H-3 and 4-OH in the ROESY spectrum, indicated their position at the
β-face of the molecule. On the other hand, correlations of H4 to 2-OH and 3-OH, 9-OH to
both H-1 and H-9a, and H-9a to 2-OH indicated their α-orientation (see Table 3.21). These

133
Results

data indicated the adoption of chair conformation for the aliphatic carbocycle and allowed to
deducing the relative stereochemistry as shown in 26a. The structure was further confirmed
by comparing NMR data of 26 to those reported for altersolanol A (25) (Yagi et al., 1993;
Okamura et al., 1993, 1996) and tetrahydroaltersolanol B (Stoessl and Stothers, 1983) which
differ from 26 in lacking the 1- and 4-OH groups. Thus, 26 was identified as a new natural
product for which we propose the name ampelanol.

O OH OH OH
H
8 8
O O
7 8a 9 9a 1 7 8a 9 1
2 OH 9a 2 OH

6 4a
10a 10 6
4a 4 3
OH 10a 10 4 3
OH
5 5
H
OH O OH OH O OH

25 Altersolanol A 26 Ampelanol

HO O

O O HH
Me
OH H OH
H
HO OH H
O OH
HO
H OH
H
OH
O
HO
OH H
H

25a 26a

134
Results

Table 3.21: 1H NMR, COSY, ROESY and HMBC data of compounds 25 and 26 at 500 MHz

Nr. 25 25a 26
δH (DMSO- ROESY HMBC δH (DMSO- δH (DMSO- COSY ROESY HMBC
d6) d6) d6)
1 4.31, d (3.7) CH3 CH3,2,3,4a,9,9a 4.38, d (4.0) 3.76, br s 9a,1OH CH3,9,9a,9OH
1-OH 5.67, d (5.9) CH3,3 1,2,9a 5.30, s 5.06, d (4.7) 1 CH3,9
2-OH 4.46, s 1,3 4.48, br s 4.21, s CH3 CH3,4,9a 1
3 3.63, dd CH3,1OH, CH3,1,4 3.64, m (7.0) 3.36, d (9.4) 4,3OH CH3,4a 4
(5.6, 6.3) 4OH
3-OH 4.88, d (6.3) 2,3,4 5.00, d (7.0) 4.41, br s 3 4
4 4.47, m 3,4a,9a 4.54, m (7.0) 3.82, m 3,4a,4OH 2OH,3OH 3,10
4-OH 5.03, d (5.3) CH3,3 5.71, d (7.0) 4.46, s 4 4a 3
4a 2.63, dd (13.2, 9.4) 4 3,9,4OH 4,9,9a,10
5-OH 12.11, br s 12.15, s 12.57, s 6 5,6,10a
6 6.81, br s OCH3 5,7,8,10a 6.72, d (2.0) 6.35, d (1.8) 8 OCH3,5OH 5,7,8,10a
8 7.00, br s OCH3 6,7,9,10a 6.93, d (2.0) 6.72, d (1.8) 6,9 OCH3,9,9OH 6,7,9,10a
9 4.67, dd (10.5, 6.2) 8,9a,9OH 1,4a,8,1OH
9-OH 5.49, d (6.2) 9 1,8,9a
9a 2.30, ddd (13.2, 10.5, 1,4a,9 1,2OH,9OH 4a,9
2.2)
CH3 1.23, s 1,3,1OH,4OH 1,2,3 1.24, s 1.20, s 2OH 1,3,1OH,2OH 1,2,3
OCH3 3.89, s 6,8 7 3.90, s 3.83, s 6,8 7
a) Yagi et al., 1993.

Table 3.22: 13C NMR data of compounds 25 and 26 at 125 MHz

Nr. 25 25a 26
δC (DMSO-d6) δC (DMSO-d6) δC (DMSO-d6)
1 68.5 68.3 71.2
2 72.9 72.7 72.9
3 73.8 73.6 74.0
4 68.5 68.3 70.7
4a 144.5 144.2 48.1
5 163.2 162.9 164.2
6 105.9 105.6 99.0
7 165.4 165.1 166.0
8 106.6 106.4 104.6
8a 133.3 132.9 152.0
9 183.7 183.3 66.4
9a 142.1 141.8 44.5
10 188.5 188.1 206.1
10a 109.5 109.2 109.1
CH3 22.3 22.2 23.8
OCH3 56.2 56.1 55.6
a) Yagi et al., 1993.

135
Results

OCH3

6 4-OH
8 3-OH 4 3
1 4a 9a
9-OH 1-OH 9 2-OH

(ppm)
9a H9a/H1
H9a/H9 2.4

4a
H9a/H4a

H3/H4 3.2
H3/3-OH
3 H4/4-OH
OCH3 H1/1-OH
1
4
H4a/H4 4.0
2-OH
3-OH
4-OH
H9/9-OH
9
4.8
1-OH

9-OH
5.6

H6/H8
6
6.4

8
H9/H8

(ppm) 6.4 5.6 4.8 4.0 3.2 2.4

Figure 3.20: COSY spectrum of compound 26.

CH3
4-OH 1 4a
3
2-OH
4 9a
1-OH
9-OH H9 3-OH

1-OH/CH3 H1/CH3
CH3 CH3

H9a 9a

H1/H4a
H4a 4a
H9/H4a

H9/H3

H3 3
9-OH/H1 H9a/H1,4
H1 1
1,4
2-OH/H1,4
H4 4

Figure 3.21A: ROESY spectrum of compound 26.

136
Results

6
8

1-OH9
9-OH

9
1-OH/H9
1-OH

9-OH

6
9-OH/H8 H9/H8
8

Figure 3.21A: ROESY spectrum of compound 26.

OCH3 CH3

6
8 4-OH 2-OH
1 4a 9a
9-OH 1-OH 9 3-OH 4 3

(ppm)
9a
H4a/C9a
4a H9a/C4a
OCH3

9 H8/C9 H4a/C9 H9a/C9


4 2-OH/C1 H3/C4 CH3/C1
1
2 H4a/C4 CH3/C2
3
4-OH/C3 H4/C3 CH3/C3 80

H8/C6
6
8 H6/C8
10a
H8/C10a H6/C10a
120

8a

5 H6/C5 OCH3/C7 160


7 H6/C7
H8/C7

200
H4/C10 H4a/C10
10
(ppm)
7.00 6.00 5.00 4.00 3.00 2.00 1.00

Figure 3.22: HMBC spectrum of compound 26.

137
Results

3.2.12. Alterporriol D (27, known compound)

Alterporriol D
Synonym(s) 7,7`-Dimethoxy-2,2`-dimethyl-1β,2α,3α,4β,5,1`α,2`β,3`β,4`α,5`-
decahydroxy-1,1`,3,3`,4,4`-hexahydro-8,8`-bianthraquinone
Sample code AR9.2.1
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 10.5 mg
Physical Description red crystals
Molecular Formula C32H30O16
Molecular Weight 670 g/mol
20
Optical Rotation [α]D - 795 (c 0.01, EtOH)
Retention time HPLC 16.0 min (standard gradient)
OH O OH

5` OH
10`a 10` 4`a 4` 3`
6`
OH
7`
8`a 9` 9`a 1` 2`
O
8`
O OH

O OH (atropisomer of 28)
O 8
8a 9 9a 1 2
7 OH

6 10a 10 4a 4 3 OH
5

OH O OH

500 AH061029b #6 AD1 UV_VIS_2


mAU WVL:254 nm
1 - 16,002

400 [M+H]+

300

200

100

2 - Weichmacher (Phthalat) - 37,036

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
227.0

[M-H]-
276.0

459.8

-10,0 nm
200 250 300 350 400 450 500 550 595

138
Results

Alterporriol D (27) was isolated from the EtOAc extract of rice cultures of
Ampelomyces sp. as red crystals (10.5 mg). It showed UV absorbances at λmax (MeOH) 227.0,
276.0 and 459.8 nm, thereby almost superimposable and closely resembling those of
altersolanol A (25). Positive and negative ESI-MS showed molecular ion peaks at m/z 671.0
[M+H]+ (base peak) and m/z 669.5 [M-H]- (base peak), respectively, indicating a molecular
weight of 670 g/mol. The 1H NMR spectral data of alterporriol D and altersolanol A
resembled each other in many aspects except for the absence of the meta-coupled pair of
aromatic protons found for the latter, which was replaced by a single aromatic proton singlet
at δH 6.90 in the spectrum of 27, assigned for H-6,6` (see Table 3.21 and 3.23). Likewise, the
13
C NMR spectra of both compounds showed high similarity with the exception of the
downfield shift of the signal corresponding to C-8 and the slight upfield shift of C-8a in the
spectrum of 27 compared to that of 25 (see Table 3.22 and 3.24). Similar NMR and UV
spectra as well as the molecular weight being twice that of altersolanol A (336 g/mol) with the
loss of two hydrogens suggested that 27 was a known compound, representing a symmetrical
dimer of 25, formed through phenolic oxidation at C-8. This assignment was corroborated by
interpretation of COSY, ROESY and HMBC spectra which were reminiscent to a high degree
to those acquired for altersolanol A (see Table 3.21 and 3.23, and Figure 3.23). Further
confirmation was achieved by comparison of UV, 1H, 13C NMR, mass spectral data and [α]D
with published data for alterporriol D, previously reported from Alternaria solani and A. porri
(Lazarovits et al., 1988; Suemitsu et al., 1989).

139
Results

3.2.13. Alterporriol E (28, known compound)

Alterporriol E
Synonym(s) 7,7`-Dimethoxy-2,2`-dimethyl-1β,2α,3α,4β,5,1`α,2`β,3`β,4`α,5`-
decahydroxy-1,1`,3,3`,4,4`-hexahydro-8,8`-bianthraquinone
Sample code AR9.2.2
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 12.3 mg
Physical Description red crystals
Molecular Formula C32H30O16
Molecular Weight 670 g/mol
20
Optical Rotation [α]D - 688 (c 0.01, EtOH)
Retention time HPLC 17.7 min (standard gradient)

OH O OH

5` OH
10`a 10` 4`a 4` 3`
6`
OH
7`
8`a 9` 9`a 1` 2`
O
8`
O OH

O OH
(atropisomer of 27)
O 8
8a 9 9a 1 2
7 OH

6 10a 10 4a 4 3 OH
5

OH O OH

300 AH061029b #8 AD2 UV_VIS_2


mAU WVL:254 nm
1 - 17,719

[M+H]+
200

100

2 - Weichmacher (Phthalat) - 37,044

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
226.1

[M-H]-
275.8

-10,0 nm
200 250 300 350 400 450 500 550 595

140
Results

Alterporriol E (28) was isolated from the EtOAc extract of rice cultures of
Ampelomyces sp. as red crystals (12.3 mg). It had UV absorbances at λmax (MeOH) 226.1,
275.8 and 460.0 nm, almost superimposable to those of altersolanol A (25) and alterporriol D
(27). Positive and negative ESI-MS showed molecular ion peaks at m/z 671.0 [M+H]+ (base
peak) and m/z 669.3 [M-H]- (base peak), respectively, indicating a molecular weight of 670
g/mol identical to that of 27. Moreover, the fragment patterns in the mass spectra of both
compounds were similar to each other. The 1H NMR spectral data of alterporriol E were
almost identical to those of alterporriol D (27) (see Table 3.23). Moreover, by comparing the
13
C NMR spectra of both compounds, the agreement between them was within 1 ppm (see
Table 3.24). Likewise, COSY, ROESY and HMBC spectra showed basically the same set of
correlations as observed for alterporriol D (see Table 3.23 and Figure 3.23). This striking
similarity ruled out the possibility of 28 being a diastereomer of 27, and could neatly be
explained with both compounds representing atropisomers. Due to a complete set of ortho
substituents around the C8-C8` bond, rotation about this single bond is restricted and thus
lead to the existence of two separate atropisomers which do not interconvert at room
temperature. However, since the compounds do contain additional stereogenic centres with
identical configuration, both atropisomers do not behave like mirror images of each other, and
can thus be isolated using achiral chromatographic separation techniques. Further
confirmation of the structure was achieved by comparison of UV, 1H, 13
C NMR, mass
spectral data and [α]D with published data for alterporriol E, previously reported from
Alternaria solani and A. porri (Lazarovits et al., 1988; Suemitsu et al., 1989). Interestingly,
both publications also reported the simultaneous occurrence of both atropisomers alterporriol
D and alterporriol E in the respective fungal strains.

141
Results

OH O OH

5` OH
10`a
6` 10` 4`a 4` 3`
OH
7`
8`a 9` 9`a 1` 2`
O
8`
O OH

O OH

O 8
8a 9 9a 1 2
7 OH

6 10a 10 4a 4 3 OH
5

OH O OH

27 Alterporriol D
28 Alterporriol E (atropisomer of 27)

Table 3.23: 1H NMR, COSY, ROESY and HMBC data of compounds 27 and 28 at 500 MHz

Nr. 27 27a 28 28a


δH COSY ROESY HMBC δH δH COSY ROESY HMBC δH
(DMSO-d6) (THF-d8) (DMSO-d6) (THF-d8)
1,1` 4.08, d (6.7) 1OH CH3,2,3,4a, 4.26 4.05, d (6.6) 1OH CH3,2,3,4a, 4.26
9,9a 9,9a
1,1`OH 5.65, d (6.7) 1 1,2,9a 5.65, d (6.6) 1 1,2,9a
2,2`OH 4.39, s CH3 1,2,3 4.42, s
3,3` 3.57, d (6.7) 4,3OH 1,4 4.32, d (6.5) 3.54, d (6.6) 4,3OH 1,4 4.35, d (6.4)
3,3`OH 4.83, br s 3 4.86, br s 3
4,4` 4.45, dd 3,4OH 3,4a,9a 4.74, d (6.5) 4.45, dd 3,4OH 4.75, d (6.4)
(6.7, 4.4) (6.6, 4.7)
4,4`OH 5.02, d (4.4) 4 3 5.07, d (4.7) 4
5,5`OH 12.92, br s 13.07, br s
6,6` 6.90, s OCH3 5,7,8,10,10a 6.77 6.92, s OCH3 5,7,8,10a 6.77
2,2`CH3 1.13, s 2OH 1,2,3 1.33 1.12, s 1,2,3 1.36
7,7`OCH3 3.66, s 6 7 3.71 3.69, s 6 7 3.67
a) Suemitsu et al., 1989.

142
Results

Table 3.24: 13C NMR data of compounds 27 and 28 at 125 MHz

Nr. 27 27a 28 28a


δC (DMSO-d6) δC (THF-d8) δC (DMSO-d6) δC (THF-d8)
1,1` 68.1 68.2 68.1 68.3
2,2` 72.8 72.8 72.9 72.9
3,3` 73.8 73.7 73.7 73.8
4,4` 68.3 68.4 68.4 68.3
4a,4`a 143.4 144.5 143.3 143.5
5,5` 163.5 163.5 164.0 163.9
6,6` 104.2 104.1 103.7 103.9
7,7` 163.6 163.6 164.6 164.7
8,8` 121.4 121.4 122.4 122.5
8a,8`a 129.8 129.8 128.8 128.9
9,9` 184.1 184.0 183.8 183.8
9a,9`a 142.7 142.7 142.8 142.8
10,10` 188.8 188.8 188.7 188.8
10a,10`a 109.3 109.3 109.2 109.3
2,2`-CH3 22.2 22.2 22.2 22.2
7,7`-OCH3 56.7 56.7 56.8 56.8
a) Suemitsu et al., 1989.

1-OH 2-OH 1 1-OH 1


4-OH 4 3 4-OH 3-OH 4 2-OH 3
3-OH

1-OH/H3
1-OH/H3 3
3
4-OH/H3
4-OH/H3

2-OH/H1
2-OH/H1 1
1

2-OH 2-OH
4 4 3-OH/H4
3-OH/H4
3-OH 3-OH
4-OH 4-OH

1-OH
1-OH

Figure 3.23: ROESY spectra of compounds 27 and 28.

143
Results

3.2.14. Altersolanol J (29, known compound)

Altersolanol J
Synonym(s) 7-Methoxy-2-methyl-2α,3α,5,10β-tetrahydroxy-1,3,4,4a,9a,10-
hexahydroanthracen-9-one
Sample code AR8.2.2
Biological source Ampelomyces sp. (from Urospermum picroides)
Sample amount 1.8 mg
Physical Description yellowish white powder
Molecular Formula C16H20O6
Molecular Weight 308 g/mol
22
Optical Rotation [α]D - 46 (c 0.7, MeOH)
Retention time HPLC 19.6 min (standard gradient)

O
H
O
8
7 8a 9 1
9a 2 OH
4a
6 10a 10 4 3
OH
5
H
OH OH

AH061220 #4 AR15-3 UV_VIS_4


160
mAU WVL:340 nm

1 - 19,570

100

[M+H]+

50

-20 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
220.8

268.9

328.8

-10,0 nm
200 250 300 350 400 450 500 550 595

144
Results

Compound 29 was isolated from the EtOAc extract of rice cultures of Ampelomyces
sp. in the form of yellowish white powder (1.8 mg). It showed UV absorbances at λmax
(MeOH) 218.5, 268.9 and 328.7 nm. Positive and negative ESI-MS showed molecular ion
peaks at m/z 308.9 [M+H]+ and m/z 307.3 [M-H]- (base peak), respectively, indicating a
molecular weight of 308 g/mol and a decrease of 32 amu compared to ampelanol (26),
formally corresponding to the loss of two oxygen atoms. The 1H NMR spectrum (see Table
3.25) showed two alcoholic hydroxyl groups at δH 4.37 (3-OH) and 3.92 (2-OHs), while a
singlet corresponding to the aliphatic methyl group 2-CH3 was detected at δH 1.16.
Additionally, the spectrum contained two carbinolic protons at δH 4.76 (H-10) and 3.15 (H-3),
a set of meta-coupled aromatic protons at δH 6.80 (H-8) and 6.57 (H-6), as well as an aromatic
methoxy group (δH 3.71). The latter features were already familiar from the spectra of 25 and
26 (see Tables 3.21). However, absence of the long range correlation of H-8 to a peri-proton,
observed in the COSY spectrum of 26, together with the singlet at δH 9.91 corresponding to a
non-chelated aromatic hydroxyl group (5-OH) indicated that the quinone carbonyl present at
C-10 in 25, had been reduced to a hydroxyl group. As in the case of ampelanol (26), this
explained the absence of color for this compound. The complete aliphatic spin system in 29
was assembled in a straight forward manner on the basis of COSY data (see Table 3.25).
The relative stereochemistry was deduced from the coupling constants in the 1H NMR
spectrum (see Table 3.25). The large values of J4a-9a (13.2 Hz) and J4a-10 (9.8 Hz) indicated the
diaxial orientation of H-4a to both H-9a and H-10. A 12.1 Hz value for J3-4ax indicated an
axial position for also for H-3. The structure was further confirmed by comparing UV, 1H
NMR, mass spectral data and [α]D value with published data for altersolanol J, previously
reported from an undetermined fungicolous hyphomycete resembling Cladosporium (Höller
et al., 2002).

145
Results

O
H
O
8
7 8a 9 1
9a 2 OH
4a
6 10a 10 4 3
OH
5
H
OH OH

29 Altersolanol J

Table 3.25: 1H-NMR and COSY data of compound 29 at 500 MHz

Nr. 29 29a
δH (DMSO-d6) COSY δH (DMSO-d6)
1ax 1.26, dd (13.8, 11.9) 1eq,9a 1.27, dd (14.0, 12.0)
1eq 2.06, dd (13.8 ,3.7) 1ax,9a 2.07, dd (14.0, 3.8)
2-OH 3.92, br s 3.93, s
3 masked by water peak 4ax,4eq,3-OH 3.16, ddd (12.0, 6.6, 4.5)
3-OH 4.37, brs 3 4.38, d (6.6)
4ax 1.56, br ddd (12.1, 12.1, 12.1) 3,4eq,4a 1.57, br ddd (12.0, 12.0, 12.0)
4eq 2.09, ddd (3.7, 4.4, 11.7) 3,4ax,4a 2.10, m
4a 1.82, dddd (13.2, 12.1, 9.8, 4.4) 4ax,4eq,9a,10 1.84, m
5-OH 9.91, s 10.1, s
6 6.57, d (2.2) 8 6.59, d (2.5)
8 6.80, d (2.2) 6 6.83, d (2.5)
9a masked by solvent peak 1ax,1eq,4a 2.51, ddd (13.0, 12.0, 3.8)
10 4.76, d (9.8) 4a,10-OH 4.78, dd (9.6, 6.6)
10-OH 6.38, d (6.6)
CH3 1.16, s 1.17, s
OCH3 3.71, s 3.73, s
a) Höller, Gloer and Wicklow, 2002.

146
Results

3.2.15. Bioactivity test results for compounds isolated from the endophytic fungus
Ampelomyces sp.
The isolated compounds were subjected to cytotoxicity and protein kinase bioassays.
Some of the isolated pure compounds were also subjected to Staphylococcus epidermidis
biofilm inhibition assays. The results are shown in Tables 3.26 and 3.27.

Table 3.26: Cytotoxicity and biofilm inhibition test results for the compounds isolated from
Ampelomyces sp. liquid and rice extracts

Nr. Compound tested L5178Y growth EC50a EC50 Biofilm MICb


a b
in % (µg/mL) (µmol/L) inhibition in % (µg/mL)
(Conc. 10 µg/mL) (Conc. 50 µg/mL)
16 Methyltriacetic lactone 100.5
17 Ampelopyrone 102.6
18 Desmethyldiaportinol - 0.4 7.30 1.6
19 Desmethyl- 41.4
dichlorodiaportin
20 Citreoisocoumarin 99.5
21 Macrosporin 54.5 na > 50.0
22 Macrosporin sulphate 74.8 na > 50.0
23 Methylalaternin 4.9 1.25 4.2 100 12.5
24 Methylalaternin sulphate 93.6 na > 50.0
25 Altersolanol A 0.0 0.21 0.6 50 > 50.0
26 Ampelanol 69.1 na > 50.0
27 Alterporriol D 64.8 na > 50.0
28 Alterporriol E 92.1 na > 50.0
29 Altersolanol J 35.7 na > 50.0
na: not active
a) Data provided by Prof. W. E. G. Müller, Mainz.
b) Data provided by Dr. U. Hentschel, Würzburg.

The isocoumarins, desmethyldiaportinol and desmethyldichlorodiaportin, showed high


and moderate activity against L5178Y cell line, respectively. Furthermore, the anthraquinone
metabolites, macrosporin and 3-O-methylalaternin, proved to be moderately and highly
active, respectively, while no activity was detected for their sulphated derivatives.
Altersolanol A was very active in this bioassay compared to ampelanol, altersolanol J and
alterporriol D, which showed moderate activity. The latter was more active than its
atropisomer, alterporriol E, which displayed almost no activity against the cancer cell line
used in the assay. On the other hand, 3-O-methylalaternin inhibited biofilm formation of S.
epidermidis by 100%, while altersolanol A was moderately active in the assay showing 50%
inhibition of biofilm formation.

147
Results

Table 3.27: Protein kinase assay results for the compounds isolated from Ampelomyces sp. liquid and
rice extracts

Activity on various protein kinases based on IC50 [g/mL]*

CDK4/CycD1

PDGFR-beta
CDK2/CycA
Compound tested

CK2-alpha1

B-RAF-VE
VEGF-R2
VEGF-R3

Aurora-A
Aurora-B
(Conc. 1µg/mL)

IGF1-R
ERBB2
EPHB4
EGF-R

INS-R
ARK5
AKT1

PAK4
PDK1

PLK1
FLT3

MET

TIE2
FAK

SAK

COT
SRC
Methyltriacetic 0 0 0 0 0 0 0 0 0 0 M0 0 0 0 0 0 0 0 0 0 M0 0 0 0
lactone
Ampelopyrone 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 A 0 0 0 M0 0 0 0
Desmethyldiaportinol 0 0 0 M 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Desmethyl- 0 0 0 0 0 M0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 M0 0 0 0
dichlorodiaportin
Citreoisocoumarin 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Macrosporin M 0 M 0 0 0 M 0 0 0 M A 0 0 0 0 0 A M M 0 0 M M M M
Macrosporin sulphate 0 0 M 0 0 0 0 0 0 0 0 M 0 0 0 0 0 M 0 0 0 M 0 0 0 0
Methylalaternin M 0 0 0 M M M M 0 0 M M 0 0 0 M 0 M M M 0 0 M M M 0
Methylalaternin 0 0 M 0 0 0 0 M 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
sulphate
Altersolanol A M 0 0 M M M M M M 0 M A 0 0 0 A 0 M 0 0 0 M M M 0 0
Ampelanol 0 M 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Alterporriol D 0 0 0 M 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Alterporriol E 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 M 0 0 0 0 0
Altersolanol J 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
S: strongly active, A: active, M: moderately active, 0: not active
* Data provided by ProQinase, Freiburg.

Results of protein kinase assay showed a similar activity profile as the cytotoxicity
assay results. Only altersolanol A and 3-O-methylalaternin were active on various protein
kinases, macrosporin was also active on some protein kinases, while the remaining
compounds were either active on very few protein kinases or completely inactive for all tested
protein kinases.

148
Results

3.3. Compounds isolated from the endophytic fungus Stemphylium botryosum


The endophytic fungus Stemphylium botryosum was isolated from leaves of
Chenopodium album growing in Egypt. The pure fungal strain was cultivated on liquid
Wickerham medium and on rice solid medium. Interestingly, chemical screening studies
indicated a clear difference between Stemphylium botryosum extracts obtained from liquid
(Wickerham) and rice cultures. Comparison of the HPLC chromatograms of the EtOAc
extracts of both cultures showed that extracts of liquid cultures had a very complex chemical
pattern compared to those obtained from rice cultures. HPLC chromatograms of the EtOAc
extract of the fungus grown on solid rice medium showed dehydrocurvularin (33) and
macrosporin (21) as main components. When grown on liquid medium the major substance
detected in the extract was stemphyperylenol (31) (see Figure 3.25A-B). Moreover, the yield
of rice cultures was higher than that of liquid cultures with a ratio of 4:1 of dried extract,
respectively. Antibacterial, antifungal, cytotoxicity and protein kinase assay results showed
that extracts obtained from rice cultures were much more active in the preliminary biological
screening tests compared to the liquid culture extracts (see Table 3.28). Due to the complex
chemical pattern, low yield and low activity of extract obtained from liquid cultures, rice
culture extracts were chosen for further investigation.
In this part results of investigation of the natural products produced by Stemphylium
botryosum when grown on solid rice medium are presented.

149
Results

250 AH041006 #2 I7LIIIaiiEt UV_VIS_2


mAU WVL:254 nm
6 - 25,123
200 31

150
8 -10
27,416
- 28,628
12 - 32,865
7 - 26,030
100
2 - 19,484
9 - 27,970
4 - 21,223 11 - 30,750
1 - 16,032
3 - 20,360
50
5 - 22,007

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

AH060608 #6 SREt UV_VIS_2


220
mAU WVL:254 nm
7 - 21,950

33 12 - 32,853

150
1 - 1,197
21

100
10 - 24,983

3 - 15,970
50 4 - 19,398 11 - 27,700
8 - 23,521 13 - 37,109
9 - 23,900
56- -20,265
20,823
2 - 2,290

-20 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

Figure 3.25A-B: EtOAc extracts of Stemphylium botryosum cultures. A: HPLC


chromatogram of EtOAc extract of liquid cultures (Wickerham
medium). B: HPLC chromatogram of EtOAc extract of rice cultures.
21: Macrosporin. 31: Stemphyperylenol. 33: Dehydrocurvularin.

Table 3.28: Biological screening test results for Stemphylium botryosum liquid and rice extracts

Extracts tested L5178Y growth in Protein kinase Antimicrobial


% activity activity
(Conc. 10 µg/mL) (Conc. 1 µg/mL) IZ [mm], 0.5 mg
BS SC CH
Stemphylium liquid n-BuOH 96.5 0 0 0
Stemphylium liquid EtOAc 66.8 0 0 0
Stemphylium rice EtOAc - 2.9 Active 12 0 0
BS: B. subtilis, SC: S. cerevisiae, CH: C. herbarum.

150
Results

3.3.1. Tetrahydroaltersolanol B (30, known compound)

Tetrahydroaltersolanol B
Synonym(s) 7-Methoxy-2-methyl-2α,3α,5,9α-tetrahydroxy-1,3,4,4a,9,9a-
hexahydroanthracen-10-one
Sample code SR5.1
Biological source Stemphylium botryosum (from Urospermum picroides)
Sample amount 5.6 mg
Physical Description yellowish white powder
Molecular Formula C16H20O6
Molecular Weight 308 g/mol
25
Optical Rotation [α]D - 27.0º (c 0.18, MeOH)
Retention time HPLC 20.5 min (standard gradient)

OH
H
O
8
7 8a 9 1
9a 2 OH
4a
6
10a 10 4 3
OH
5
H
OH O

AH060811b #2 SR-5 UV_VIS_3


300
mAU WVL:280 nm
2 - 20,456
[M+H]+
200

[2M+Na]+
100
1 - 19,613 3 - 33,287

4 - 49,673

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
216.3

281.7
231.0

-10,0 nm
200 250 300 350 400 450 500 550 595

151
Results

Tetrahydroaltersolanol B (30) was isolated from the EtOAc extract of rice cultures of
Stemphylium botryosum as a yellowish white powder (5.6 mg). It showed UV absorbances at
λmax (MeOH) 216.1, 231.0, 281.7 and 320.0 nm, with a similar pattern to the UV spectrum
recorded for ampelanol (26). Positive and negative ESI-MS showed molecular ion peaks at
m/z 309.1 [M+H]+ (base peak) and m/z 307.6 [M-H]- (base peak), respectively, indicating a
molecular weight of 308 g/mol, identical to that of altersolanol J (29). The 1H NMR spectrum
resembled that of 29 (see Tables 3.25 and 3.29), showing three exchangeable alcoholic
hydroxyl groups at δH 5.61, 4.43, and 3.79 assigned to 9-OH, 3-OH, and 2-OH, respectively.
The singlet corresponding to the aliphatic methyl group (2-CH3) was detected at δH 1.15.
Additionally, two carbinolic protons appearing at δH 4.26 (d, J=10.8 Hz) and 3.28 (dd, J=12.2,
4.2 Hz) were assigned to H-9 and H-3, respectively. The 1H NMR spectrum also showed
meta-coupled aromatic protons at δH 6.68 (H-8) and 6.34 (H-6) as well as an aromatic
methoxy group (δH 3.80). These were features already familiar from the spectra of 25 and 26
(see Table 3.21). In addition, a chelated phenolic OH was found to resonate at δH 12.89 (5-
OH). Importantly, the less shielded of the two aryl protons H-8 exhibited a second splitting of
1.2 Hz, indicating a long range coupling with the peri-proton H-9, which was confirmed by
the corresponding correlation observed in the COSY spectrum. Accordingly, similar to
ampelanol (26), the quinone carbonyl corresponding to that at C-9 in 25 had been reduced to a
hydroxy group, with the consequent loss of color for this compound. As for 26, H-9 coupled
to the proton situated at the junction of the trans-decaline-like ring system (H-9a), which in
turn coupled to a diastereotopic methylene group (CH2-1) as well as to the other ring junction
proton (H-4a). The further members of the aliphatic spin system, i.e. CH2-4 and H-3, were
clearly discernible in the COSY spectrum (see Table 3.29).
The relative stereochemistry of 30 with the exception of C-2 could be resolved by
analysis of the coupling constants of signals in the high field region (see Table 3.29). The
large values of J4a-9a (11.6 Hz) and J9a-9 (10.8 Hz) indicated their respective diaxial
relationship. The value for J3-4ax (12.2 Hz) furthermore indicated an axial position for H-3. In
addition, spectroscopical properties discovered during this study proved virtually identical to
UV, 1H NMR, mass spectral data, and the [α]D value published for tetrahydroaltersolanol B
(Stoessl and Stothers, 1983), thus confirming the identity of 30 with this compound.
Tetrahydroaltersolanol B had previously been reported from culture filtrates of Alternaria
solani (Stoessl and Stothers, 1983).

152
Results

OH
H
O
8
7 8a 9 1
9a 2 OH
4a
6
10a 10 4 3
OH
5
H
OH O

30 Tetrahydroaltersolanol B

Table 3.29: 1H NMR and COSY data of compound 30 at 500 MHz

Nr. 30 30a
δH (DMSO-d6) COSY δH (DMSO-d6)
1ax 1.19, dd (13.2, 12.1) 1eq,9a 1.20, dd (13.0, 12.0)
1eq 2.15, dd (13.2 ,3.4) 1ax,9a 2.16, dd (13.0, 3.3)
2-OH 3.79, brs 3.80, s
3 3.28, dd (12.2, 4.2) 4ax,4eq,3-OH 3.30, ddd (10.7, 6.5, 4.6)
3-OH 4.43, brs 3 4.45, d (6.5)
4ax 1.45, q (12.2) 3,4eq,4a 1.46, dt (12.0, 12.0)
4eq 2.11, ddd (12.2, 4.2, 3.7) 3,4ax,4a 2.13, m
4a 2.44, m 4ax,4eq,9a 2.46, dt (12.0, 3.7)
5-OH 12.89, s 12.91, s
6 6.34, d (2.3) 8 6.35, d (2.5)
8 6.68, dd (2.3, 1.2) 6,9 6.70, dd (2.5, 1.1)
9 4.26, br d (10.8) 8,9a,9-OH 4.29, m
9-OH 5.61, br s 9 5.63, d (7.4)
9a 1.94, dddd (12.1, 11.6, 10.8, 3.4) 1ax,1eq,4a,9 1.96, m
CH3 1.15, s 1.16, s
OCH3 3.80, s 3.82, s
a) Stoessl and Stothers, 1983.

153
Results

3.3.2. Stemphyperylenol (31, known compound)

Stemphyperylenol
Synonym(s) Hexahydro-1,4,7,10-tetrahydroxyperylene-3,9-quinone
Sample code SR2.1
Biological source Stemphylium botryosum (from Urospermum picroides)
Sample amount 3.9 mg
Physical Description Reddish brown powder
Molecular Formula C20H16O6
Molecular Weight 352 g/mol
23
Optical Rotation [α]D + 411º (c 0.2, MeOH)
Retention time HPLC 25.4 min (standard gradient)
OH O

4
5 3a 3 2

6 3b 1
6a 12b OH
H H
HO 12a
6b 9b
12
7
8 11
9 9a
10
O OH

60,0 AH060803 #6 SR-8 UV_VIS_2


mAU WVL:254 nm

1 - 25,372

37,5

2 - 37,110
25,0

12,5

3 - 49,761
0,0

min
-20,0
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
217.4

261.3

[M-H]-
342.0

-10,0
nm
200 250 300 350 400 450 500 550 595

154
Results

Stemphyperylenol (31) was isolated from the EtOAc extract of rice cultures of
Stemphylium botryosum in the form of a reddish brown powder (3.9 mg). The UV spectrum
showed λmax (MeOH) at 217.4, 261.3 and 342.0 nm. Negative ESI-MS showed a molecular
ion peak at m/z 351.3 [M-H]- (base peak) indicating a molecular weight of 352 g/mol,
identical to that of altertoxin I (14). The 1H and 13
C NMR spectra (see Table 3.30), which
contained signals due to only eight protons and ten carbon atoms, indicated that the molecule
was a symmetrical C10 dimer. The 1H resonances were assigned to an AX spin system of
ortho-coupled aromatic protons appearing at δH 8.14 and 6.81 (H-6/H-12 and H-5/H-11,
respectively), to a chelated phenolic hydroxyl group (δH 12.09, 4-OH/10-OH), and to a CH2-
CHOH-CH fragment (CH2-2, H-1 and H-12b/CH2-8, H-7 and H-6b, respectively). The
respective substructures were also assembled on basis of the COSY spectrum (see Table
13
3.30). The C resonances were attributed to a tetra-substituted aromatic ring and a carbonyl
carbon atom, the remaining signals assigned to one methylene and two methine sp3 carbons,
one of which was oxygen-bearing. From the HMBC spectrum, it was possible to place the
chelated phenolic hydroxyl group at C-4, since this carbon showed HMBC correlations to H-
5, H-6 and 4-OH. Consequently, the carbonyl group was located at C-3. Furthermore, CH2-
2/8 displayed correlations with C-3/9, thereby establishing the C2-C3/C8-C9 bonds. The
correlations of H-6b/12b to C-6a/12a and of H-6/12 to C-6b/12b indicated the connection of
the two halves of the molecule to give the planar structure of 31. This was further confirmed
by the long-range coupling between the aromatic (H-6/12) and benzylic (H-6b/12b) protons
observed in the COSY spectrum.
The relative stereochemistry was derived from detailed examination of the coupling
constants (see Table 3.30). The large values of J1/7-2ax/8ax (11.9 Hz) and J1/7-12b/6b (8.8 Hz)
indicated the diaxial (= trans) position of H-1/7 with regard to both H-12b/6b and H-2ax/8ax.
This suggested that the cyclohexanone rings preferentially adopted a half-chair conformation
with the hydroxy groups in an equatorial position and H-6b/H-12b in an axial position. The
obtained data were in excellent agreement with UV, 1H, 13
C NMR, mass spectral data and
[α]D value published for stemphyperylenol (Arnone and Nasini, 1986), confirming that 31 and
the latter were identical. Stemphyperylenol had previously been described from Alternaria
cassiae (Hradil et al., 1989) and Stemphylium botryosum var. Lactucum (Arnone and Nasini,
1986).

155
Results

OH O

4
5 3a 3 2

6 3b 1
6a 12b OH
H H
HO 12a
9b
6b 12
7
8 11
9 9a
10
O OH

31 Stemphyperylenol

Table 3.30: 1H-, 13C-NMR, COSY and HMBC data of compound 31 at 500 (1H) nd 125 MHz (13C)

Nr. 31 31a
δH COSY HMBC δC δH δC
(DMSO-d6) (DMSO-d6) (Acetone-d6) (Acetone-d6)
1,7 4.59, m 2/8ax,2/8eq,6/12b,1/7-OH 66.5 4.76 68.32
1-,7-OH 5.75, brs 1/7 4.97
2ax,8ax 3.14, dd (15.4, 11.9) 1/7,2/8eq 1/7,3/9,6/12b 46.8 3.17 47.84
2eq,8eq 2.91, dd (15.4, 4.4) 1/7,2/8ax 1/7,3/9,6/12b 3.07
3,9 203.5 204.01
3a,9a 114.8 115.92
3b,9b 143.0 143.73
4,10 159.1 161.03
5,11 6.85, d (8.8) 6/12 3/9a,4/10,6/12a 114.4 6.81 115.55
6,12 8.01, d (8.8) 5/11,6/12b 3/9b,4/10,6/12b 134.6 8.14 135.58
6a,12a 129.9 130.90
6b,12b 3.71, d (8.8) 1/7,6/12 1/7,3/9b,6/12a 44.6 3.75 46.05
4-,10-OH 12.02, s 3/9a,4/10,5/11 12.09
a) Arnone and Nasini, 1986.

156
Results

3.3.3. Curvularin (32, known compound)

Curvularin
Synonym(s) 11,13-dihydroxy-4-methyl-4,5,6,7,8,9-hexahydro-1H-
benzo[d][1]oxacyclododecine-2,10-dione
Sample code SR3.1
Biological source Stemphylium botryosum (from Urospermum picroides)
Sample amount 1.9 mg
Physical Description yellow powder
Molecular Formula C16H20O5
Molecular Weight 292 g/mol
20
Optical Rotation [α]D - 28.5º (c 0.4, EtOH)
Retention time HPLC 23.9 min (standard gradient)

O O
15
1
4 14
HO 2
5 3
13
10
6 9 12
8
7 11
OH O

AH060803 #5 SR-7 UV_VIS_1


300
mAU WVL:235 nm
1 - 23,859
[M+H]+
[2M+Na]+
200

2 - 36,984

100

3 - 47,703

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
202.5
222.0

271.9

-10,0 nm
200 250 300 350 400 450 500 550 595

157
Results

Curvularin (32) was isolated from the EtOAc extract of rice cultures of Stemphylium
botryosum in the form of a yellow powder (1.9 mg). It showed UV absorbances at λmax
(MeOH) 202.5, 222.0, 271.9 and 298.9 nm, reminiscent to those of ampelanol (26) and
tetrahydroaltersolanol B (30). Positive and negative ESI-MS displayed molecular ion peaks at
m/z 293.2 [M+H]+ (base peak) and m/z 291.5 [M-H]- (base peak), respectively, indicating a
molecular weight of 292 g/mol. The 1H NMR spectrum (see Table 3.31) showed a pair of
meta-coupled aromatic protons at δH 6.24 (H-6) and 6.21 (H-4), respectively. In addition, the
spectrum contained a diastereotopic methylene function resonating at δH 3.85 and 3.61 (CH2-
2) as well as an extended aliphatic spin system which could only be completely assembled
with help of the COSY spectrum due to a significant degree of overlapping (see Table 3.31).
It consisted of an aliphatic methyl group (δH 1.12, 15-CH3) adjacent to an oxygenated methine
group (δH 4.92, H-15), which was further connected to a chain of five methylene groups. Four
of them appeared as a complex set of multiplets resonating between 1.26 and 1.74 ppm (CH2-
11-CH2-14), while the downfield chemical shift of the last one (δH 3.20 and 2.73, CH2-10)
indicated its position adjacent to a carbonyl function, which was conjugated to the aromatic
ring on basis of the occurrence of the characteristic ion with m/z 205 (C11H9O4) in the mass
spectrum, the structure of which is presumably 32a (Munro et al., 1967). Furthermore, the
COSY spectrum also revealed a long range coupling between H-4 and CH2-2. In the HMBC
spectrum, CH2-2 displayed correlations to C-3, C-4, C-8, and the ester carbonyl at δC 173.0
(C-1), the oxygen atom of which had to be connected to H-15. Overall, the substructures
established so far suggested that 32 was identical to the known curvularin, a macrocyclic
lactone rather widespread in fungi, which had previously been reported from Curvularia sp.
(Birch et al., 1959), Alternaria sp. (Robeson and Strobel, 1981, 1985) and Penicillium sp. (Lai
et al., 1989). This assumption was confirmed by comparison of spectral properties of 32 with
data reported in the literature (Munro et al., 1967; Ghisalberti et al., 1993). Based on the
almost identical value obtained for the [α]D it was possible to deduce the absolute
configuration at C-15 to be S (Ghisalberti et al., 1993).

158
Results

3.3.4. Dehydrocurvularin (33, known compound)

Dehydrocurvularin
Synonym(s) 11,13-dihydroxy-4-methyl-4,5,6,7-tetrahydro-1H-
benzo[d][1]oxacyclododecine-2,10-dione
Sample code SR5.2
Biological source Stemphylium botryosum (from Urospermum picroides)
Sample amount 15.5 mg
Physical Description brown powder
Molecular Formula C16H18O5
Molecular Weight 290 g/mol
22
Optical Rotation [α]D - 79.8º (c 3.0, EtOH)
Retention time HPLC 22.4 min (standard gradient)

O O
15
1
4 14
HO 2
5 3
13
10
6 9 12
8
7 11
OH O

AH060810 #2 SR-6 UV_VIS_1


400
mAU WVL:235 nm
2 - 22,362

300

200 [M+H]
+
3 - 37,187

100
1 - 22,035

4 - 47,510

min
-50
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
204.1
228.4

287.6

-10,0 nm
200 250 300 350 400 450 500 550 595

159
Results

Dehydrocurvularin (33) was isolated from the EtOAc extract of rice cultures of
Stemphylium botryosum in the form of brown powder (15.5 mg). Its UV spectrum showed
λmax (MeOH) at 204.1, 228.4 and 287.6 nm. Positive and negative ESI-MS showed molecular
ion peaks at m/z 291.2 [M+H]+ and m/z 289.7 [M-H]- (base peak), respectively, indicating a
molecular weight of 290 g/mol, and thus 2 mass units less than that of curvularin (32). The 1H
NMR spectrum (see Table 3.31) suggested a close relationship to 32, except for the
appearance of an ABX2 system, with two olefinic protons forming the AB part (δH 6.49, H-10,
and δH 6.57, H-11), and a coupling constant of 15.4 Hz consistent with a trans-configuration
at the double bond. These olefinic protons were found to be part of an extended spin system
detected in the COSY spectrum, consisting of the adjacent CH2-12 (δH 2.38, 2.29), and the
remaining signals for CH2-13, CH2-14, H-15, and 15-CH3 which showed close similarity to
the respective signals observed for curvularin 32. As in the case of curvularin, presence of the
characteristic ion with m/z 203 (C11H7O4, 33a) in the EI-mass spectrum indicated that the
carbonyl function at C-9 was conjugated to the aromatic ring (Munro et al., 1967). Thus, 33
was identified as the known 10-dehydro congener of curvularin. This was corroborated by
very similar spectral characteristics both in the HMBC spectrum (see Table 3.31) and the ESI-
MS as well as by comparison with published data for dehydrocurvularin (Munro et al., 1967;
Lai et al., 1989) and curvularin (Ghisalberti et al., 1993). Again, the [α]D value obtained in
this study suggested the S-configuration at C-15 (Munro et al., 1967). Dehydrocurvularin was
previously reported from Curvularia sp. (Munro et al., 1967), Penicillium sp. (Lai et al.,
1989), Alternaria sp. (Arai et al., 1989; Robeson and Strobel, 1981, 1985) and Stemphylium
radicinum (Grove, 1971).

160
Results

O O O O
15 15
1 1
4 14 4 14
HO 2 HO 2
5 3 5 3
13 13
10 10
6 9 12 6 9 12
8 8
7 11 7 11
OH O OH O

32 Curvularin 33 Dehydrocurvularin

O+ O+

C C

HO HO

OH O OH O

32a 33a
C11H9O4 C11H7O4
m/z 205 m/z 203

161
Results

Table 3.31: 1H-NMR, COSY and HMBC data of compounds 32 and 33 at 500 MHz

Nr. 32 32a 33
δH COSY HMBC δH δH COSY HMBC
(MeOD) (Acetone-d6) (MeOD)
1
2A 3.85, d (15.7) 2B,4 1,3,4,8 3.77, d (15.7) 3.72, d (16.7) 2B,4 1,3,4,8
2B 3.61, d (15.7) 2A,4 1,3,4,8 3.69, d (15.7) 3.44, d (16.7) 2A,4 1,3,4,8
3
4 6.21, d (2.2) 2A/B,6 2,6,8 6.33, d (2.3) 6.28, d (2.0) 2A/B,6 2,5,6,8
5
6 6.24, d (2.2) 4 4,5,8 6.37, d (2.3) 6.24, d (2.0) 4 4,5,8
7
8
9
10 A 3.20, ddd 10B,11A/B A 3.10, ddd 6.49, d (15.4) 11 8,9,11,12
(15.2, 8.8, 2.8) (15.4, 8.7, 3.0)
B 2.73, ddd 10A,11A/B B 2.75, ddd
(15.2, 9.7, 2.8) (15.4, 9.7, 2.9)
11 A 1.74, m 10A/B,11B,12A/B A 1.73, m 6.57, ddd 10,12A/B 9,10,12,13
(15.4, 8.2, 5.6)
B 1.57, m 10A/B,11A,12A/B B 1.52, m
12A 1.39, m 11A/B,12B,13A/B 1.41, m 2.38, m 11,12B,13A/B 10,11,13,14
12B 1.26, m 11A/B,12A,13A/B 1.25, m 2.29, m 11,12A,13A/B 10,11,13,14
13A 1.47, m 12A/B,13B,14A/B 1.45, m 1.96, m 12A/B,13B,14A/B 11,12,14,15
13B 1.30, m 12A/B,13A,14A/B 1.28, m 1.56, m 12A/B,13A,14A/B 12,15
14A 1.59, m 13A/B,14B,15 1.58, m 1.85, m 13A/B,14B,15 CH3,12,13
14B 1.43, m 13A/B,14A,15 1.43, m 1.58, m 13A/B,14A,15 12,13
15 4.92, m CH3,14A/B 4.90, m 4.79, m CH3,14A/B 1,13,14
CH3 1.12, d (6.3) 15 14,15 1.10, d (6.4) 1.18, d (6.6) 15 14,15
a) Ghisalberti et al., 1993.

Table 3.32: 13C-NMR data of compounds 32 and 33 at 125 MHz

Nr. 32 32a 33
δC (MeOD)b δC (MeOD) δC (MeOD)
1 173.0 172.8 173.0
2 40.6 40.5 42.4
3 137.5 137.2 137.0
4 112.6 112.2 112.2
5 161.5 161.2 162.0
6 102.9 102.7 102.8
7 159.5 162.2
8 121.0 120.9 117.8
9 209.7 200.0
10 44.6 133.3
11 23.8 154.3
12 27.7 34.2
13 24.9 25.4
14 33.0 32.9 35.1
15 73.9 73.7 74.2
CH3 20.5 20.4 20.3
a) Ghisalberti et al., 1993.
b) Derived from HMBC spectrum.

162
Results

3.3.5. Bioactivity test results for compounds isolated from the endophytic fungus
Stemphylium botryosum
The isolated compounds were subjected to bioassays aimed to determine their
cytotoxicity and their protein kinase inhibitory profiles. The results are shown in Tables 3.33
and 3.34.

Table 3.33: Cytotoxicity test results for the compounds isolated from Stemphylium
botryosum rice extracts

Nr. Compound tested L5178Y growth in %* EC50* EC50


(Conc. 10 µg/mL) (µg/mL) (µmol/L)
30 Tetrahydro- 38.9
altersolanol B
31 Stemphyperylenol 31.6
32 Curvularin 6.6 4.7 16.0
33 Dehydrocurvularin - 3.8 0.43 1.4
* Data provided by Prof. W. E. G. Müller, Mainz.

Curvularin and dehydrocurvularin showed high activity against the L5178Y cell line,
whereas tetrahydroaltersolanol B and stemphyperylenol were moderately active.

Table 3.34: Protein kinase assay results for the compounds isolated from Stemphylium botryosum
rice extract

Activity on various protein kinases based on IC50 [g/mL]*


CDK4/CycD1

PDGFR-beta
CDK2/CycA

Compound tested
CK2-alpha1

B-RAF-VE
VEGF-R2
VEGF-R3

Aurora-A
Aurora-B

(Conc. 1µg/mL)
IGF1-R
ERBB2
EPHB4
EGF-R

INS-R
ARK5
AKT1

PAK4
PDK1

PLK1
FLT3

MET

TIE2
FAK

SAK

COT
SRC

Tetrahydro- 0 0 0 M0 0 0 0 0 0 MM0 0 0 0 0 M0 0 0 A 0 0 0 0
altersolanol B
Stemphyperylenol 0 0 0 0 0 0 0 0 0 0 M0 0 0 0 0 0 M0 0 0 0 0 0 0 0
Curvularin 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Dehydrocurvularin 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
S: strongly active, A: active, M: moderately active, 0: not active
* Data provided by ProQinase, Freiburg.

The results of the protein kinase assay showed a different activity profile than the
cytotoxicity assay results. Tetrahydroaltersolanol B was active against a few protein kinases
whereas stemphyperylenol inhibited only two of the tested enzymes. On the other hand,
curvularin and dehydrocurvularin were inactive for all tested protein kinases in spite of their
pronounced activity in the cytotoxicity assay.

163
Results

3.4. Compounds isolated from the endophytic fungus Chaetomium sp.


This undescribed endophytic fungal strain of the genus Chaetomium was isolated from
fresh stems of Otanthus maritimus growing in Egypt. The pure fungal strain was cultivated on
liquid Wickerham medium and rice solid medium. Preliminary biological and chemical
screening studies indicated slight differences between Chaetomium extracts obtained from
liquid and rice cultures. Comparison of the HPLC chromatograms of the EtOAc extracts of
both cultures showed that both extracts had cochliodinol (35) as their main component. While
this was the only peak observed in extracts obtained from rice cultures, liquid culture extracts
showed additional peaks for orsellinic acid (39) and aureonitolic acid (34) (see Figure 3.26A-
B). Similar to observations made with other fungal strains throughout this thesis, the yield of
rice cultures was higher than that of liquid cultures with a ratio of 2:1 of dried extract,
respectively. Furthermore, extracts obtained from rice cultures were slightly more active in
preliminary biological screening tests compared to the liquid culture extracts (see Table 3.35).
In this part results of investigating the natural products produced by Chaetomium sp.
when grown in liquid medium or on solid rice medium are presented.

164
Results

350 AH040709 #9 V2SIEtOAc UV_VIS_1


mAU WVL:235 nm
4 - Monocerin - 34,357
A
35

200 3 - Fettsäure - 26,660

34
39 1 - PAI 1/1 - 18,638
100
2 - 20,524 5 - meta-Chloro-para-hydroxy-phenyl-essigsäureamid - 47,526

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

500 AH060614 #3 ChEtR UV_VIS_1


mAU WVL:235 nm
1 - 33,356

400 B
35

300

200

4 - 37,109
100
3 - 35,469
2 - 34,133 5 - 47,811

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

Figure 3.26A-B: EtOAc extracts of Chaetomium sp. cultures. A: HPLC chromatogram


of EtOAc extract of liquid cultures (Wickerham medium). B: HPLC
chromatogram of EtOAc extract of rice cultures. 34: Aureonitolic acid.
35: Cochliodinol. 39: Orsellinic acid.

Table 3.35: Biological screening test results for Chaetomium liquid and rice extracts

Extracts tested L5178Y growth in Protein kinase Antimicrobial


% activity activity
(Conc. 10 µg/mL) (Conc. 1 µg/mL) IZ [mm], 0.5 mg
BS SC CH
Chaetomium liquid n-BuOH 99.5 0 0 0
Chaetomium liquid EtOAc 72.4 Active 0 0 0
Chaetomium rice EtOAc 52.6 Active 10 0 0
BS: B. subtilis, SC: S. cerevisiae, CH: C. herbarum.

165
Results

3.4.1. Aureonitolic acid (34, new compound)

Aureonitolic acid
Synonym(s) (2E,4E)-5-((3S,4R,5S)-5-((E)-buta-1,3-dienyl)-4-hydroxy-
tetrahydrofuran-3-yl)penta-2,4-dienoic acid
Sample code ChE2.2
Biological source Chaetomium sp. (from Otanthus maritimus)
Sample amount 1.7 mg
Physical Description viscous colourless oil
Molecular Formula C13H16O4
Molecular Weight 236 g/mol
20
Optical Rotation [α]D - 5.0 (c 0.5, MeOH)
Retention time HPLC 19.1 min (standard gradient)

12
13 O 10
8
2 4 6 11
9
HOOC 7
3 5
OH

AH050106 #2 V276-85/1 UV_VIS_3


700
mAU WVL:280 nm
2 - 19,149 (+)-ESI-MS: no ionization

400

3 - 19,404

200

1 - 1,091 4 - 50,147

min
-100
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-HCOO]-
263.1
225.2

[M-H]-
[2M-H]-
566.3

-10,0 nm
200 250 300 350 400 450 500 550 595

166
Results

Aureonitolic acid (34) was isolated from the EtOAc extracts of liquid cultures of
Chaetomium sp. as viscous colourless oil (1.7 mg). It showed UV absorbances at λmax
(MeOH) 225.2 and 263.1 nm. The HRESI-MS exhibited a strong peak at m/z 259.0940
[M+Na]+ indicating a molecular formula of C13H16O4 (calculated 259.0946, ∆ 0.0006). The
1
H NMR and COSY spectra (see Table 3.36 and Figure 3.27) showed two major spin systems,
which on the basis of the observed coupling constants were shown to consist of two
conjugated double bonds each (C-2 through C-5 as well as C-9 through C-12, respectively).
Both were connected to a central 3-hydroxytetrahydrofuran moiety at its positions 4 and 2,
respectively. The coupling constants observed for the terminal methylene protons H-12A and
H-12B indicated mutual geminal as well as vicinal couplings to H-11 as in aureonitol (34a,
13
see below). The C NMR spectrum (see Table 3.37) showed two oxymethine carbons at δC
82.3 and 86.2, as well as one oxymethylene carbon at δC 71.7 assigned to C-7, C-8 and C-13,
respectively, which would correspond to positions 3, 2 and 5 of the central 3-
hydroxytetrahydrofuran ring. Signals for H-7 and H-13B overlapped at δH 3.80, H-8 and H-
13A at δH 4.05 in the 1H NMR spectrum (see Table 3.36). The carbon framework of 34 which
was basically already evident from the COSY spectrum was confirmed by inspection of the
HMBC spectrum (see Figure 3.28). Key correlations include H-2 to C-1, C-3 and C-4, H-5 to
C-3, C-4, C-6, C-7 and C-13, H-6 to C-4, C-5, C-7 and C-13, H-7 to C-5, C-6, C-8 and C-9,
H-8 to C-7 and C-10, H-9 to C-8 and C-11 as well as those of CH2-13 to C-5, C-7 and C-8.
In order to determine the relative configuration of the compound, 1D NOE difference
spectra were acquired. Irradiation of H-3, H-6 and H-9 gave enhancements as listed in Table
3.36. Most importantly, H-6 exhibited a pronounced NOE with H-8, and correspondingly, H-9
with H-7. These results together with the coupling constants observed in the 1H NMR
spectrum are in agreement with a syn configuration of the two carbon chains and a trans
configuration of the hydroxy group at the ether ring, as well as an all trans configuration of
the double bonds in the side chains. All spectroscopical data obtained for 34 were in
agreement with the corresponding signals reported for aureonitol (Abraham and Arfmann,
1992; Bohlmann and Ziesche, 1979; Seto et al., 1979), except for the fact that the terminal
methyl group in one of the side chains of the latter was replaced by a carboxylic acid group,
13
as indicated by the signal at δC 174.5 in the C NMR spectrum and the presence of a
fragment at m/z 190.8 [M-CO2-H]- in the negative mode ESI-MS. Thus, 34 was identified as a
new natural product for which we propose the name aureonitolic acid. Aureonitol was
previously reported from Chaetomium species (Abraham and Arfmann, 1992; Seto et al.,
1979) as well as from Helichrysum aureo-nitens (Bohlmann and Ziesche, 1979).

167
Results

12
13 O 10
8
2 4 6 11
9
R 7
3 5
OH

Nr. Compound R
34 Aureonitolic acid COOH
34a Aureonitol CH3

Table 3.36: 1H NMR, COSY, NOE and HMBC data of compound 34 at 500 MHz

Nr. 34 34aa
δH (MeOD) COSY NOE HMBC δH (CDCl3)
1 1.74, dd (6.5, 1.5)
2 5.89, d (15.1) 3 1,3,4 5.65, dq (14.5, 6.5)
3 7.00, dd (14.8, 10.7) 2,4 5 1,4,5 6.00, ddq (14.5, 10.0, 1.5)
4 6.30, dd (15.3, 10.7) 3,5 2,3,6 6.12, dd (15.0, 10.0)
5 5.89, dd (15.3, 8.5) 4,6 3,4,6,7,13 5.40, dd (15.0, 9.0)
6 2.82, q (7.8) 5,7,13A,13B 4,8,13A>>5,7 4,5,7,13 2.84, dddd (9.0, 8.5, 7.5)
7 3.80, dd (6.8) 6,8 5,6,8,9 3.72, dd (7.5)
8 4.05, dd (6.8) 7,9 7,10 4.12, dd (7.5)
9 5.70, dd (14.8, 6.9) 8,10 7,11 8,11 5.68, dd (14.5, 7.5)
10 6.28, m 9 8,12 6.32, m (14.5, 10.0)
11 6.29, m 12A,12B 12 6.32, m (17.0, 10.0)
12A 5.21, d (16.1, 1.7) 11,12B 10 5.22, d (17.0)
12B 5.08, d (9.4, 1.7) 11,12A 10,11 5.10, d (10.0)
13A 4.05, dd (8.4) 6,13B 5,7,8 4.10, dd (8.5)
13B 3.80, dd (8.4) 6,13A 5,7,8 3.70, dd (8.5)
a) Abraham and Arfmann, 1992.

Table 3.37: 13C NMR data of compound 34 at 125 MHz

Nr. 34 34aa
δC (MeOD) δC (CDCl3)
1 174.5 18.0
2 128.0 128.3
3 139.0 130.9
4 132.5 133.1
5 138.1 129.3
6 52.4 51.6
7 82.3 81.7
8 86.2 84.8
9 133.1 133.2
10 134.1 131.0
11 137.7 136.2
12 118.0 118.1
13 71.7 71.1
a) Abraham and Arfmann, 1992.

168
Results

2,5 12B 8,13A 7,13B


9 12A 6
(ppm)

H6/H7,13B
6 H6/H13B

3.2

7,13B H7/H8
H13B/H13A
H8/H9 4.0

8,13A

4.8

12B
12A

5.6
9
H5/H6
2,5

(ppm) 5.6 4.8 4.0 3.2

Figure 3.27A: Expansion of the COSY spectrum of compound 34.

3 11,10,4 2,5 9 12A 12B


(ppm)

12B H12A/H12B
12A 5.2

5.6

9 H9/H10
H2/H3
2,5
6.0

11,10,4
H4/H5 6.4

H11/H12B
H11/H12A

6.8

3
H3/H4
(ppm) 6.8 6.4 6.0 5.6 5.2 4.8

Figure 3.27B: Expansion of the COSY spectrum of compound 34.

169
Results

8,13A
2,5 12B 7,13B
3 4,10,11 12A 6
9

H4/C6
C6
H5/C6 H7/C6

H5/C13 H6/C13 C13

H5/C7 H8,13A/C7
H13B/C7
C7
H10/C8 H6/C7
C8
H9/C8 H13A/C8 H7,13B/C8

Figure 3.28A: Expansion of the HMBC spectrum of compound 34.

170
Results

2,5 8,13A
3 12B 7,13B
4,10,11 9 12A 6

H4/C2
H2,5/C4 H7/C9
H3/C4 CH28/C10 H6/C4
H9/C11 H8/C10
H3/C5 H12A/C11
H6/C5
H4/C3 H2,5/C3
H7,13B/C5
H13A/C5

H2/C1

H3/C1

Figure 3.28B: Expansion of the HMBC spectrum of compound 34.

171
Results

3.4.2. Cochliodinol (35, known compound)

Cochliodinol
Synonym(s) 2,5-Dihydroxy-3,6-bis[5-(3-methyl-2-butenyl)-1H-indol-3-yl]-2,5-
cyclohexadiene-1,4-dione
Sample code ChE8.1, ChR3
Biological source Chaetomium sp. (from Otanthus maritimus)
Sample amount 455.7 mg
Physical Description purple crystals
Molecular Formula C32H30N2O4
Molecular Weight 506 g/mol
Retention time HPLC 33.0 min (standard gradient)

1
HN 2 O
7a
7 3 OH
6` 1`
3a 2`
6
4
5 5` 4` 3`
HO
9 8
O
NH
10
11
12

500 AH040906 #4 V2Et227-233 UV_VIS_2


mAU WVL:254 nm
1 - 33,012
[M+H]+
400

300

200

100

2 - 51,612

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
224.0

279.8

472.0

-10,0 nm
200 250 300 350 400 450 500 550 595

172
Results

Cochliodinol (35) was isolated from the EtOAc extracts of liquid and rice cultures of
Chaetomium sp. as purple crystals (455.7 mg). The UV spectrum showed λmax (MeOH) at
224.0, 279.8 and 472.0 nm, indicative of an indole chromophore. Positive and negative ESI-
MS showed molecular ion peaks at m/z 507.3 [M+H]+ (base peak) and m/z 505.7 [M-H]- (base
peak), respectively, indicating a molecular weight of 506 g/mol. Together with inspection of
the NMR spectra, a molecular formula of C32H30N2O4 was derived. However, the 1H NMR
spectrum only gave signals in a ratio of 4:1:2:6 for aromatic, olefinic, methylene and methyl
protons, respectively. All resonances therefore had to originate from pairs of chemically
equivalent groups due to symmetry in the molecule. Furthermore, aromatic proton and carbon
resonances had chemical shifts and multiplicities consistent with the presence of either a 3,5-
or a 3,6-disubstituted indole residue (see Table 3.38 and 3.39), since the aromatic multiplet
signals clearly indicated a 1,2,4-trisubstituted phenyl substructure. The presence of a 2-
methyl-but-2-enyl (= isoprenyl) substituent was established by a metastable ion observed in
the mass spectrum indicating the loss of a neutral fragment of 69 mass units. In addition, the
1
H NMR spectrum indicated that the olefinic proton (H-9) at δH 5.37 was allylically coupled
to the methyl protons at δH 1.74 and 1.72, and vicinally coupled to the methylene group (CH2-
8) at δH 3.40. From the COSY spectrum, the attachment of the isoprenyl side chain to the
aromatic side of the indole substructure (either at C-5 or C-6) was evident by long range
correlations of CH2-8 to both H-4 and H-6. Accordingly, correlations of H-2 to both C-3a and
C-7a indicated that a further substituent had to reside at C-3 in the heteroaromatic ring of the
indole moiety. Most diagnostic for unambiguously deducing the position of the isoprenyl side
chain proved a correlation of H-4 to C-3 in the HMBC spectrum, the corresponding proton
signal was the one exhibiting the meta-coupling, thus representing the proton immediately
adjacent to the substituent. Based on consideration of symmetry as outlined above, two of
these 3,5-disubstituted indole systems were present in 35. However, in the 13C NMR spectrum
only one signal (δC 111.9, C-3`/C-6`) was observed belonging to the central unit connecting
these two substructures. The molecular weight indicated this to be a symmetrical 2,5-
dihydroxy-1,4-quinone ring, thus suggesting that 35 was identical to the known cochliodinol.
According to the literature, the non-appearance of the signal for C-1`, C-2`, C-4` and C-5`
(due to the rapid interconversion of two equivalent tautomeric forms only one instead of two
signals is to be expected) seems to be a typical feature of this substructure. The identification
of 35 as 2,5-dihydroxy-3,6-bis[5-(3-methyl-2-butenyl)-3-indolyl]-benzoquinone was
1 13
confirmed by comparison of UV, H, C NMR and mass spectral data with published data for
cochliodinol (Jerram et al., 1975; Sekita, 1983). Cochliodinol was previously reported from

173
Results

several Chaetomium species (Jerram et al., 1975; Sekita et al., 1981; Sekita, 1983; Brewer et
al., 1984).

3.4.3. Isocochliodinol (36, known compound)

Isocochliodinol
Synonym(s) 2,5-Dihydroxy-3,6-bis[6-(3-methyl-2-butenyl)-1H-indol-3-yl]-2,5-
cyclohexadiene-1,4-dione
Sample code ChE8.2, ChR5
Biological source Chaetomium sp. (from Otanthus maritimus)
Sample amount 8.8 mg
Physical Description purple crystals
Molecular Formula C32H30N2O4
Molecular Weight 506 g/mol
Retention time HPLC 33.8 min (standard gradient)
1
HN 2 O
7a
7 3 OH
6` 1`
3a 2`
8
6
12 4
5 5` 4` 3`
9 HO
10
O
11 NH

500 AH041014 #7 V2SIIbEt64-76 UV_VIS_2


mAU WVL:254 nm

1 - 33,821
[M+H]+
400

300

200

100

-50 min
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
226.3

282.7

474.2

-10,0 nm
200 250 300 350 400 450 500 550 595

174
Results

Isocochliodinol (36) was isolated from the EtOAc extracts of liquid and rice cultures
of Chaetomium sp. as purple crystals (8.8 mg). Its UV spectrum showed λmax (MeOH) at
226.3, 282.7 and 474.2 nm, with very high similarity to that of 35. Positive and negative ESI-
MS showed molecular ion peaks at m/z 507.3 [M+H]+ (base peak) and m/z 505.7 [M-H]- (base
peak), respectively, indicating a molecular weight of 506 g/mol and thus identical to that of
cochliodinol (35). The 1H NMR spectrum disclosed identical spin systems as described above
for 35, with the mass spectrum also supporting the presence of a 2-methyl-but-2-enyl group.
Thus, 36 had to represent a symmetrical isomer of cochliodinol (35), leaving a positional
isomer with the prenyl groups attached to C-6 as the most probable alternative. As in the case
of cochliodinol, the COSY spectrum (see Table 3.38) confirmed that the isoprenyl group was
attached to the aromatic side of the indole substructure since CH2-8 exhibited long range
correlations to both H-7 and H-5. In addition, in the HMBC spectrum (see Table 3.38) a
correlation of H-4 to C-3 was detected. However, in the case of 36, this proton signal was the
one exhibiting the ortho-coupling, proving that the side chain was situated meta with regard to
H-4 and thus resided at C-6. Based on these observations, 36 was identified as the known
isocochliodinol which was confirmed by comparison of its UV, 1H, 13
C NMR and mass
spectral data with published data (Sekita, 1983). The compound was previously obtained from
several Chaetomium spp. (Sekita et al., 1981; Sekita, 1983; Brewer et al., 1984).

175
Results

1
HN 2 O
7a
7 3 OH
6` 1`
3a 2`
6
4
5 5` 4` 3`
HO
9 8
O
NH
10
11
12

35 Cochliodinol

1
HN 2 O
7a
7 3 OH
6` 1`
3a 2`
8
6
12 4
5 5` 4` 3`
9 HO
10
O
11 NH

36 Isocochliodinol

176
Results

Table 3.38: 1H NMR, COSY and HMBC data of compounds 35 and 36 at 500 MHz

Nr. 35 35a 36 36a


δH COSY HMBC δH δH COSY HMBC δH
(MeOD) (THF-d8) (DMSO-d6) (THF-d8)
NH 10.30, br s 11.21,s 2 2,3,3a,7a 10.14, br s
2 7.50, s 3,3a,7a 7.51, d (2.4) 7.43, d (2.5) NH 3,3a,7a 7.39, d (2.4)
3
4 7.32, d (0.9) 6,8 3,6,7a,8 7.39, br s 7.32, d (8.2) 5 3,6,7a 7.38, d (8.3)
5 6.81, br d 4,7,8 3a,7,8 6.73, dd
(8.2) (8.3, 1.4)
6 6.94, dd 4,7,8 4,7a,8 6.90, dd
(8.5, 1.5) (8.3, 1.4)
7 7.29, d (8.2) 6 3a,5 7.19, d (8.3) 7.16, br s 5,8 3a,5,8 7.01, d (1.4)
8 3.40, d (7.2) 4,6,9,11,12 4,5,6,9,10 3.40, d (6.8) 3.42, d (7.2) 5,7,9,11,12 5,6,7,9,10, 3.32, d (7.3)
9 5.37, tq 8,11,12 8,11,12 5.36, tqq 5.35, br t (7.2) 8,11,12 8,11,12 5.29, tqq
(7.2, 1.5) (6.8, 1.4, 1.4) (7.3, 1.5, 1.5)
10
11 1.74, br s 8,9 9,10,12 1.72, br s 1.71, br s 8,9 9,10,12 1.639, d (1.5)
12 1.72, d (0.95) 8,9 9,10,11 1.72, br s 1.71, br s 8,9 9,10,11 1.636, d (1.5)
OH 9.72, s 9.56, s
a) Sekita, 1983.

Table 3.39: 13C NMR data of compounds 35 and 36 at 125 MHz

Nr. 35 35a 36
δC (MeOD)b δC (THF-d8) δC (DMSO-d6)b
1`,4` 168.5
2`,5` 168.5
3`,6` 111.9 112.1
2 128.4 128.0
3 106.6 105.5 105.8
3a 128.3 132.7 126.0
4 121.8 122.8
5 133.5 127.9 121.1
6 123.0 122.0 135.8
7 111.7 111.5 111.6
7a 135.5 135.8 135.2
8 35.6 35.6 35.0
9 126.1 126.2 126.0
10 131.0 131.0 131.2
11 17.9 17.9 18.0
12 25.9 25.9 26.0
a) Sekita, 1983.
b) Derived from HMBC spectra.

177
Results

3.4.4. Indole-3-carboxylic acid (37, known compound)

Indole-3-carboxylic acid
Synonym(s) 1H-indole-3-carboxylic acid
Sample code ChE6
Biological source Chaetomium sp. (from Otanthus maritimus)
Sample amount 20.1 mg
Physical Description brown needles
Molecular Formula C9H7NO2
Molecular Weight 161 g/mol
Retention time HPLC 18.4 min (standard gradient)

O
OH

4
3
5
2
6
N1
H
7

AH060203 #12 156-181Et UV_VIS_3


160
mA WVL:280 nm
2 - 18,408
U

100

[M+H]+
50

3 - 36,675

1 - 15,877

mi
-20 n
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
211.0
227.1

281.2

-10,0 nm
200 250 300 350 400 450 500 550 595

178
Results

Indole-3-carboxylic acid (37) was isolated from the EtOAc extracts of liquid cultures
of Chaetomium sp. as brown needles (20.1 mg). It had UV absorbances at λmax (MeOH) 211.0,
227.1 and 281.2 nm, indicative of an indole chromophore. Positive and negative ESI-MS
showed molecular ion peaks at m/z 162.0 [M+H]+ and m/z 160.2 [M-H]- (base peak),
respectively, indicating a molecular weight of 161 g/mol and suggesting the molecular
formula C9H7NO2. The presence of a fragment at m/z 116.0 [M-CO2-H]- in the negative ESI-
MS indicated that the compound contained a carboxylic acid function. The 1H NMR spectrum
(see Table 3.40) showed 7 proton resonances that included an aromatic ABCD spin system at
δH 8.06 (ddd, J=6.9, 2.2, 0.6 Hz, H-4), 7.42 (ddd, J=6.9, 1.2, 0.6 Hz, H-7), 7.18 (dt, J=6.9, 2.2
Hz, H-6), 7.15 (dt, J=6.9, 1.2 Hz, H-5), as well as a proton resonance at δH 7.93 (s, H-2). The
obtained UV, 1H NMR and mass spectral data were identical to those reported for indole-3-
carboxylic acid (Aldrich, 1992; Hiort, 2002).

O
OH

4
3
5
2
6
N1
H
7

37 Indole-3-carboxylic acid

Table 3.40: 1H NMR data of compound 37 at 500 MHz

Nr. 37 37a
δH (MeOD) δH (DMSO-d6)
1 11.43, s
2 7.93, s 7.70, s
4 8.06, ddd (6.9, 2.2, 0.6) 8.14, d (7.6)
5 7.15, dt (6.9, 1.2) 7.02, dt (6.9, 1.3)
6 7.18, dt (6.9, 2.2) 7.05, dt (6.9, 1.3)
7 7.42, ddd (6.9, 1.2, 0.6) 7.35, d (7.6)
a) Hiort, 2002.

179
Results

3.4.6. Cyclo(alanyltryptophane) (38, known compound)

Cyclo(alanyltryptophane)
Synonym(s) 3-(1H-Indol-3-ylmethyl)-6-methyl-2,5-diketopiperazine
Sample code Ch2.1
Biological source Chaetomium sp. (from Otanthus maritimus)
Sample amount 5.4 mg
Physical Description white crystals
Molecular Formula C14H15N3O2
Molecular Weight 257 g/mol
Retention time HPLC 13.9 min (standard gradient)

O
H
4` N
5`
3` 6`
8
2`
4 N 1`
3a 3 H
5 O

2
6 1
7a N
7 H

700 AH060411 #2 Et25-40/3 UV_VIS_2


mAU WVL:254 nm

2 - 13,944 [2M+H]+
500

375

250
[M+H]+

125
3 - 37,050
1 - 13,028

min
-100
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
%
[M-H]-
225.9

[2M-HCOO]-
[M-HCOO]-
279.6
287.5

-10,0 nm
200 250 300 350 400 450 500 550 595

180
Results

Cyclo(alanyltryptophane) (38) was isolated from the EtOAc extracts of liquid cultures
of Chaetomium sp. as white crystals (5.4 mg). It showed UV absorbances at λmax (MeOH)
225.9, 279.6 and 287.5 nm, indicative of an indole chromophore. Positive and negative ESI-
MS showed molecular ion peaks at m/z 258.0 [M+H]+ and m/z 256.4 [M-H]- (base peak),
respectively, revealing a molecular weight of 257 g/mol and suggesting the molecular formula
C14H15N3O2. The 1H NMR spectrum showed the characteristic 3-substituted indole signals, as
also observed in the spectrum of 37 (see Tables 3.40 and 3.41), namely the aromatic ABCD
spin system appearing at δH 7.55 (br d, J=7.7 Hz, H-4), 7.29 (br d, J=7.7 Hz, H-7), 7.01 (br t,
J=7.7 Hz, H-6), 6.92 (br t, J=7.7 Hz, H-5), as well as two proton singlets at δH 10.90 and 7.02,
assigned to NH-1 and H-2, respectively. Additionally, the protons of an aliphatic methylene
group (CH2-8) were observed at δH 3.22 (J=14.3, 3.9 Hz) and 2.99 (J=14.3, 4.5 Hz). In the
COSY spectrum (see Table 3.41), this methylene group was found to be part of a saturated
spin system coupling to a signal at δH 4.09 (br s, H-3`) and a signal at δH 8.02 (br s, NH-2`).
Interpretation of the HMBC spectrum (see Table 3.41) showed that CH2-8 correlated to C-3,
C-3a as well as C-3`, confirming the 3-substituted indole structure. Moreover, both CH2-8 and
H-3` showed HMBC correlations to an oxygenated carbon signal at δC 166.5 (C-4`), thereby
establishing the C3`-C4` bond and the tryptophane substructure. Furthermore, the COSY
spectrum showed another saturated spin system composed of an aliphatic methyl group
detected at δH 0.38 (d, J=6.8 Hz), a broad quartet at δH 3.57 (J=6.8 Hz, H-6`) and a signal at
δH 7.90 (brs, NH-5`). All the latter proton signals showed HMBC correlations to an
oxygenated carbon atom appearing at δC 167.8 (C-1`), thus confirming an alanine
substructure. The HMBC correlation between NH-5` and C-3` as well as the pronounced
upfield shift of the methyl group indicated that both amino acids were arranged into a cyclic
dipeptide or diketopiperazine. The obtained UV, 1H, 13
C NMR and mass spectral data were
identical to published data for cyclo(alanyltryptophane) (Hiort, 2002) previously isolated from
Aspergillus sp. (Marchelli et al., 1975). Since all possible stereoisomers of this simple
metabolite have already been found in nature, and moreover its origin as a true natural
product, rather than an artifact resulting for example from the amino acid-containing culture
medium and formed upon autoclaving, is debatable, no efforts were undertaken to elucidate
the absolute stereochemistry of 38.

181
Results

O
H
4` N
5`
3` 6`
8 H
2`
4 N 1`
3 H
5 3a O

2
6 1
7a N
7 H

38 Cyclo(alanyltryptophane)

Table 3.41: 1H, 13C NMR, COSY and HMBC data of compound 38 at 500 (1H) and 125 MHz (13C)

Nr. 38 38a
δH COSY HMBC δC δH δC
(DMSO-d6) (DMSO-d6)b (DMSO-d6) (DMSO-d6)
1 10.90, s 2,4 2,3,3a,7a 10.88, s
2 7.02, d (0.6) 1,8A,8B 3,3a,7a 124.5 7.05, m 124.7
3 108.1 108.3
3a 127.5 127.7
4 7.55, br d (7.7) 1,5 3,6,7a 118.6 7.55, d (7.6) 118.9
5 6.92, br t (7.7) 4,6 3a,7 118.0 6.90, m 118.2
6 7.01, br t (7.7) 5,7 4,7a 120.4 7.05, m 121.1
7 7.29, br d (7.7) 6 3a,5 110.9 7.29, d (8.1) 110.7
7a 135.5 135.9
8 A 3.22, dd (14.3, 3.9) 2,8B,3` 3,3a,3` 3.23, dd (14.5, 4.1) 28.7
B 2.99, dd (14.3, 4.5) 2,8A,3` 2,3,3a,3`,4` 3.00, dd (14.5, 4.4)
1` 167.8 168.0
2` 8.02, brs 3` 4`,6` 7.99, d (1.3)
3` 4.09, brs 8A,8B,2`,6` 4` 55.4 4.09, brs 55.3
4` 166.5 166.7
5` 7.90, brs 6` 1`,3` 7.89, d (1.3)
6` 3.57, brq (6.8) CH3,3`,5` CH3,1` 49.5 3.58, brq (6.9) 49.3
CH3 0.38, d (6.8) 6` 1`,6` 19.0 0.41, d (6.9) 18.8
a) Hiort, 2002.
b) Derived from HMBC spectrum.

182
Results

3.4.7. Orsellinic acid (39, known compound)

Orsellinic acid
Synonym(s) 2,4-Dihydroxy-6-methylbenzoic acid
Sample code ChE4
Biological source Chaetomium sp. (from Otanthus maritimus)
Sample amount 49.6 mg
Physical Description brown needles
Molecular Formula C8H8O4
Molecular Weight 168 g/mol
Retention time HPLC 18.0 min (standard gradient)

HO O

1 OH
6
2

5 3
4

OH

AH040902 #11 V2Et106-120 UV_VIS_3


700
mAU WVL:280 nm

1 - 18,005

400
[M+H]+

200

2 - 18,986 3 - 27,167
4 - 51,532

min
-100
0,0 10,0 20,0 30,0 40,0 50,0 60,0

70,0
% [M-H]-
221.3

260.7

296.6

-10,0 nm
200 250 300 350 400 450 500 550 595

183
Results

Orsellinic acid (39) was isolated from the EtOAc extracts of liquid cultures of
Chaetomium sp. as brown needles (49.6 mg). It displayed UV absorbances at λmax (MeOH)
221.3, 260.7 and 296.6 nm. Positive and negative ESI-MS showed molecular ion peaks at m/z
169.0 [M+H]+ (base peak) and m/z 167.4 [M-H]- (base peak), respectively, revealing a
molecular weight of 168 g/mol. 1H and 13
C NMR spectra (see Table 3.42) indicated the
presence of an aromatic methyl group at δH 2.38 and δC 23.4 as well as a pair of meta-coupled
protons at δH 6.16 and 6.11 (each d, J=2.2 Hz) assigned to H-3 and H-5, respectively.
13
Additionally, the C NMR spectrum (see Table 3.42) contained 6 aromatic carbon signals,
two of which were hydroxylated, and a signal at δC 173.2 indicative of an aromatic carboxylic
acid function. This was further confirmed by a fragment at m/z 123.0 [M-CO2-H]- in the
13
negative ESI-MS. The C, 1H NMR and mass spectra supported a molecular formula of
C8H8O4. The obtained UV, 1H NMR and mass spectral data were found to be almost identical
to published data for orsellinic acid (Evans and Staunton, 1988). Orsellinic acid was
previously isolated from lichens (Maass, 1975) as well as cultures of Penicillium sp. and other
microorganisms (Birkinshaw and Gowlland, 1962).

HO O

1 OH
6
2

5 3
4

OH

39 Orsellinic acid

Table 3.42: 1H and 13C NMR data of compound 39 at 500 (1H) and 125 MHz (13C)

Nr. 39 39a
δH (DMSO-d6) δC (DMSO-d6) δH (MeOD)
1 104.8
2 161.9
3 6.11, d (2.2 Hz) 100.4 6.20, br s
4 164.4
5 6.16, d (2.2 Hz) 110.9 6.20, br s
6 142.8
CH3 2.38, s 23.4 2.54, s
COOH 173.2
2-OH 12.63, br s
4-OH 10.13, br s
a) Evans and Staunton, 1988.

184
Results

3.4.8. Bioactivity test results for compounds isolated from the endophytic fungus
Chaetomium sp.
The isolated compounds were subjected to bioassays aimed to determine their
cytotoxicity and their protein kinase inhibitory profiles. The results are shown in tables 3.43
and 3.44.

Table 3.43: Cytotoxicity test results for the compounds isolated from Chaetomium sp. rice
extracts

Nr. Compound tested L5178Y growth in %* EC50* EC50


(Conc. 10 µg/mL) (µg/mL) (µmol/L)
34 Aureonitolic acid 79.6
35 Cochliodinol - 2.3 7.0 14.0
36 Isocochliodinol 75.8
37 Indol-3-carboxylic acid 93.7
38 Cyclo(alanyl- 104.7
tryptophane)
39 Orsellinic acid 1.0 2.7 16.0
* Data provided by Prof. W. E. G. Müller, Mainz.

Orsellinic acid and cochliodinol proved to be highly active against the L5178Y cancer
cell line, whereas a weak activity was observed for isocochliodinol and aureonitolic acid.

Table 3.44: Protein kinase assay results for the compounds isolated from Chaetomium sp. liquid
extract

Activity toward selected protein kinases*


CDK4/CycD1

PDGFR-beta
CDK2/CycA

Compound tested
CK2-alpha1

B-RAF-VE
VEGF-R2
VEGF-R3

Aurora-A
Aurora-B

(Conc. 1µg/mL)
IGF1-R
ERBB2
EPHB4
EGF-R

INS-R
ARK5
AKT1

PAK4
PDK1

PLK1
FLT3

MET

TIE2
FAK

SAK

COT
SRC

Aureonitolic acid 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Cochliodinol A M M 0 A A A M 0 A M A 0 0 M M 0 A A A M M A A A A
Isocochliodinol A M M 0 A A A M 0 0 M M 0 0 M M 0 M M M M M M M M M
Orsellinic acid 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
S: strongly active, A: active, M: moderately active, 0: not active
* Data provided by ProQinase, Freiburg.

The results of the protein kinase assay showed a different activity profile in
comparison to the cytotoxicity assay. Cochliodinol and isocochliodinol were active on various
protein kinases, while aureonitolic acid and orsellinic acid were inactive for all tested protein
kinases, in spite of the high activity of orsellinic acid in the cytotoxicity assay.

185
Results

3.5. Tracing of fungal metabilites in the corresponding plant extracts


With the different compounds isolated from endophytic fungal strains in the course of
this thesis at hand, extracts of their respective host plants were screened to detect the presence
of these metabolites. To this aim, crude plant extracts were prepared and divided into
subfractions by liquid-liquid partitioning and subsequent fractionation over Diaion HP-20
using H2O:MeOH and MeOH:acetone gradient elution. Each of these fractions were then
analyzed by LC-MS, with specifically preparing mass chromatograms obtained at the
respective base or other characteristic intense peaks for the individual isolated fungal
metabolites. If matching mass spectra were suspected, co-elution studies with the
corresponding metabolite and the respective plant fraction were carried out to compare the
corresponding retention times.

3.5.1. Tracing of Alternaria metabolites in Polygonum senegalense fractions


All of the compounds isolated from Alternaria sp. were detected in the crude extract
of this fungus. Most remarkably, alternariol, alternariol-5-O-methyl ether and altenusin were
also traced in subfractions of the Polygonum senegalense extract (see Table 3.45 and Figures
3.29-31), the host plant from which the fungal strain was originally obtained.

Table 3.45: Compounds detected in P. senegalense fractions

Compound Fraction* Polarity of eluting solvent


Altenusin 2 50% MeOH:H2O
Alternariol 3 75% MeOH:H2O
Alternariol monomethyl ether 4 100% MeOH
* The 90% MeOH fraction was fractionated over Diaion HP-20 using H2O:MeOH and MeOH:acetone
gradient elution.

186
Results

A: Mass chromatogram and full MS of altenusin in P. senegalense fraction 2


RT: 5.00 - 45.00 Hassan527 #591 RT: 18.69 AV: 1 NL: 1.81E6
18.58 N L: 1.83E6 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 273.3
Bas e Peak m /z=
100

[M-H]-
95 272.5-273.5 F: - c

90
m/z 273.3 ES I Full m s [
100.00-1000.00]
95

MS Has s an527 90
85
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60

Relative Abundance
55
55
50
50
45 45
40 40
35 19.18 258.4
35
30 30
25 19.68 25
20 20

15 15 274.3

10 22.33 10
317.3 547.0
22.75
5 25.79 28.63 5 186.3 230.3
16.92 31.55 33.98 35.40 37.79 174.3
6.62 13.65 16.63 40.37 43.06 318.4 371.5 453.1 600.3 659.4 688.4 781.8 831.5 865.5 939.3 965.1
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

B: Mass chromatogram and full MS of pure altenusin


RT: 5.00 - 45.00 Hassan528 #590 RT: 18.34 AV: 1 NL: 2.97E6
18.34 N L: 2.97E6 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 273.4
Bas e Peak m /z=
100
272.5-273.5 F: - c

[M-H]-
95

90 m/z 273.4 ES I Full m s [


100.00-1000.00]
MS Has s an528
95

90
85
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60

Relative Abundance
55
55
50
50
45 45
40 40
35 35
30 258.5
30
25 25
20 20
274.3
15 15

10 10
547.1
5 19.22 5 186.4
17.63 21.15 24.19 27.26 29.26 174.4 230.4 571.5 600.4 644.0 701.1
12.49 15.33 33.85 35.65 36.91 44.38 336.1 371.3 445.4 497.8 790.0 819.0 883.0 933.8 961.7
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

Figure 3.29: MS detection of altenusin (A: in P. senegalense fraction 2, B: pure compound).

A: Mass chromatogram and full MS of alternariol in P. senegalense fraction 3


RT: 5.00 - 45.00 Hassan529 #552 RT: 17.86 AV: 1 NL: 2.43E6
17.86 NL: T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 2.43E6 257.3
100
95 m/z 257.3 Bas e Peak F: -
c E SI Full m s [ 95
90

85
100.00-
1000.00] MS
Has s an529
90 [M-H]-
85
80
80
75
32.98 75
70
70
65
65
60
Relative Abundance

60
Relative Abundance

55
55
50
50
45 45
40 40
35 35
30 30
31.00
25 25
20 20

15 15
258.3
28.98
10 10 515.0
22.87 213.5
5 12.78 20.38 28.81 5
16.78 25.78 34.86 37.11 551.0
5.69 10.13 11.12 41.65 43.39 147.2 211.3 293.0 339.0 513.6 569.2 638.2 704.9 766.9 834.7 857.0 903.2 959.2
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

187
Results

B: Mass chromatogram and full MS of pure alternariol


RT: 5.00 - 45.00 Hassan530 #582 RT: 17.90 AV: 1 NL: 6.90E5
17.99 N L: 7.24E5 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 257.3
17.81 Bas e Peak m /z=
100
95
m/z 257.3 256.5-257.5 F: - c
ES I Full m s [ 95

[M-H]-
90 100.00-1000.00]
MS Has s an530 90
85
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60

Relative Abundance
55
55
50
50
45 45
40 40
35 35
30 30
25 25
20 20
258.3
15 15

10 10
213.3
5 5
13.40 15.36 18.99 25.44 147.0 268.8 342.0 533.9 637.9 698.1 731.8 798.3 827.2 891.0 986.7
31.47 32.45 34.26 393.0 930.3
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

Figure 3.30: MS detection of alternariol (A: in P. senegalense fraction 3, B: pure compound).

A: Mass chromatogram and full MS of alternariol-5-O-methyl ether in P. senegalense fraction


4
RT: 5.00 - 45.00 Hassan532 #750 RT: 22.65 AV: 1 NL: 5.74E5
22.65 N L: 5.74E5 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 271.3
Bas e Peak m /z=
100
95

90
m/z 271.3 270.5-271.5 F: - c
ES I Full m s [
100.00-1000.00]
95 [M-H]-
MS Has s an532 90
85
85
80
80
75
75
70
70
65
65
60 256.3
Relative Abundance

60
Relative Abundance

55
55
50
50
45 45
40 40
35 35
30 30
25 9.75 25
20 12.09 20

15 15 255.3
272.3
10 10
228.3
5 5 185.4
15.15 16.55 18.59 601.3 623.8
6.51 9.31 25.17 26.61 29.64 183.1 338.9 382.8 406.2 454.8 541.0 750.8 780.4 859.6 952.3 967.4
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

B: Mass chromatogram and full MS of pure alternariol-5-O-methyl ether


RT: 5.00 - 45.00 Hassan531 #705 RT: 22.67 AV: 1 NL: 2.79E6
22.67 N L: T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 3.22E6 271.3
100
m /z=

[M-H]-
95

90 m/z 271.3 263.4-278.0 F: -


c ESI Full m s [
100.00-1000.00]
95

90
85 MS Has s an531
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60
Relative Abundance

55
55
50 256.5
50
45 45
40 40
35 35
30 30
25 25
20 20
272.3
15 15 255.6
10 10 228.5

5 5 543.1
7.73 10.62 12.33 13.57 17.66 20.47 25.63 28.98 34.63 35.12 38.05 40.33 42.02 183.5 213.4 325.5 356.2 414.6 479.6 578.9 694.6 717.9 778.8 814.5 870.0 952.9 992.4
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

Figure 3.31: MS detection of alternariol-5-O-methyl ether (A: in P. senegalense fraction 4, B: pure


compound).

188
Results

3.5.2. Tracing of Ampelomyces metabolites in Urospermum picroides fractions


All of the compounds isolated from Ampelomyces sp. were detected in the crude
extract of this fungus. Most remarkably, macrosporin and 3-O-methylalaternin were also
traced in subfraction 5 (eluted by 100% acetone) of the Urospermum picroides extract (see
Figures 3.32-33), the host plant from which the fungal strain was originally obtained.

A: Mass chromatogram and full MS of macrosporin in U. picroides fraction 5


RT: 5.00 - 45.00 Hassan499 #853 RT: 25.35 AV: 1 NL: 1.94E6
25.26 NL: T: - c ESI sid=25.00 Full ms [ 100.00-1000.00]
100 2.27E6 283.5
95 m/z 283.5 Bas e Peak F: -
100

[M-H]-
c E SI Full m s [ 95
90 100.00-
1000.00] MS 90
85 Has s an499
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60
Relative Abundance

55 268.7
55
50
50
43.05
45 45
40 40
26.66
35 35
30 35.01
30
17.39
25 25
20.68 31.10
20 40.83 20
15.11 21.61 36.50
13.72
15 15
39.02
27.88 34.00 240.6
10 19.98 10
23.17
5 10.34 5 212.6
169.6 319.6 379.6
6.86 9.90 12.37 397.6 481.1 525.9 611.7 642.5 683.5 708.6 793.7 877.5 903.1 925.7 990.5
0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

B: Mass chromatogram and full MS of pure macrosporin


RT: 5.00 - 45.00 Hassan508 #847 RT: 25.00 AV: 1 NL: 4.05E6
25.00 N L: T: - c ESI sid=25.00 Full ms [ 100.00-1000.00]
100 4.89E6 283.5
100
95

90
m/z 283.5 m /z=
273.2-292.7 F: -
c ESI Full m s [
95 [M-H]-
100.00-1000.00] 90
85 MS Has s an508
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60
Relative Abundance

268.7
55
55
50
50
45 45
40 40
35 35
30 30
25 25
20 20
16.20
15 15 240.6
14.33
10 10

5 5 211.7
26.11 28.43 29.62 465.5 567.0
7.17 10.46 13.95 16.73 21.94 34.97 38.89 40.04 43.34 141.3 183.4 285.5 323.5 415.5 487.6 602.9 656.5 732.3 790.6 863.3 936.5 965.8
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

Figure 3.32: MS detection of macrosporin (A: in U. picroides fraction 5, B: pure compound).

189
Results

A: Mass chromatogram and full MS of 3-O-methylalaternin in U. picroides fraction 5


RT: 5.00 - 45.00 Has s an512 #742 RT: 23.39 AV: 1 NL: 8.42E4
23.39 NL: 3.96E4 T: - c ESI s id=25.00 Full m s [ 100.00-1000.00]
100 347.4
Bas e Peak m/z=
100
95 298.5-299.5 F: - c

90 m/z 299.4 ESI Full ms [


100.00-1000.00]
MS Hass an512
95

90
85
85
80
80
75

70
75

70
[M-H]-
65
65
60
Relative Abundance

60

Relative Abundance
55 16.04 55
50
50
299.4
45 45
40 40
35 35
30 30
25 21.37
25
14.31
13.85 42.27
20 20
21.10
13.33 39.28 284.1
15 15
17.59
18.76 984.3
10 10.28 11.64 37.19 10 311.4 363.2 431.2 774.7 806.2
596.1 620.4 645.5 681.4 901.7 920.1
5 5 147.1 482.9 546.8 834.3

0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Time (min) m /z

B: Mass chromatogram and full MS of pure 3-O-methylalaternin


RT: 5.00 - 45.00 Has s an514 #705 RT: 23.06 AV: 1 NL: 3.28E5
23.06 NL: T: - c ESI s id=25.00 Full m s [ 100.00-1000.00]
100 3.28E5 299.2
100
95 m/z=

90 m/z 299.2 298.5-299.5 F: -


c ESI Full ms [
100.00-1000.00]
95

90
[M-H]-
85 MS Hass an514
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60
Relative Abundance

55
55
50
50
45 45
40 40
35 35
30 30
25 25 284.2
20 256.3
20
23.99
15 15
21.11 655.1
10 10 300.3 653.2
5 24.19 5 621.1 675.5
11.44 16.86 28.57 30.46 227.3 357.0 389.3 940.2 992.6
6.83 35.83 468.8 708.2
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Time (min) m /z

Figure 3.33: MS detection of 3-O-methylalaternin (A: in U. picroides fraction 5, B: pure compound).

3.5.3. Tracing of Stemphylium botryosum metabolites in Chenopodium album fractions


All of the compounds isolated from Stemphylium botryosum were detected in the
crude extract of this fungus. Most remarkably, tetrahydroaltersolanol B, curvularin,
stemphyperylenol and macrosporin were also traced in subfractions of the Chenopodium
album extract (see Table 3.46 and Figures 3.34-37), the host plant from which the fungal
strain was originally obtained.

Table 3.46: Compounds detected in C. album fractions

Compound Fraction* Polarity of eluting solvent


Tetrahydroaltersolanol B 2 50% MeOH:H2O
Curvularin 4 100% MeOH
Stemphyperylenol 4 100% MeOH
Macrosporin 5 100% Acetone
* The 90% MeOH fraction was fractionated over Diaion HP-20 using H2O:MeOH and MeOH:acetone
gradient elution.

190
Results

A: Mass chromatogram and full MS of tetrahydroaltersolanol B in C. album fraction 2


RT: 5.00 - 45.00 Hassan543 #351 RT: 11.06 AV: 1 NL: 4.34E4
11.06 N L: 4.34E4 T: - c ESI sid=25.00 Full ms [ 100.00-1000.00]
100 307.3
Bas e Peak m /z=
100
306.5-307.5 F: - c
95

90
m/z 307.3 ES I Full m s [
100.00-1000.00]
95
[M-H]-
MS Has s an543 90
85 11.36
85
80
80
75
75
70
70
65
10.91 65
60
Relative Abundance

60

Relative Abundance
55
55
50
50
45 45 569.0
40 40
289.3
35 35
30 30
25 25 983.9
367.3
11.97
5.81 6.96 33.02
20 29.09 20
40.73 135.2
9.83 13.57 16.01 21.92 32.75 42.53
15 18.83 23.87 15 847.1 912.7
285.7 379.1 629.0 830.3
16.39 24.88 34.05 927.5
39.89 338.3 437.6 694.2 748.0
10 10 161.0 262.4 593.0 875.8
544.5
9.20 19.56 690.9
5 5

0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

B: Mass chromatogram and full MS of pure tetrahydroaltersolanol B


RT: 5.00 - 45.00 Hassan544 #338 RT: 10.53 AV: 1 NL: 2.20E5
10.53 N L: 2.20E5 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 307.4
Bas e Peak m /z=
100
95

90
m/z 307.4 306.5-307.5 F: - c
ES I Full m s [
100.00-1000.00]
95 [M-H]-
MS Has s an544 90
85
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60

Relative Abundance
55
55
50
50
45 45
40 40
35 35
30 30
25 25
20 20

15 15 709.5
725.4
10 10
308.5 615.3 638.3
14.82 564.4 885.2 917.1
5 6.82 15.89 32.07 5 478.3 507.2 690.2 793.5 828.5 996.8
13.52 20.34 21.81 25.40 329.3 412.3
42.98 255.4
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

Figure 3.34: MS detection of tetrahydroaltersolanol B (A: in C. album fraction 2, B: pure compound).

A: Mass chromatogram and full MS of curvularin in C. album fraction 4


RT: 5.00 - 45.00 Hassan547 #539 RT: 16.72 AV: 1 NL: 2.39E4
16.65 N L: 3.27E4 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 291.4
Bas e Peak m /z=
100

[M-H]-
95 290.5-291.5 F: - c

90
m/z 291.4 ES I Full m s [
100.00-1000.00]
95

MS Has s an547 90
85
85
80
80
75
75
70
70
65 21.69
65
60
Relative Abundance

60
Relative Abundance

55
55
50
50
45 45
40 40
22.38
35 137.2
35
30 30 609.4
697.3 747.2
22.65 523.8 933.3
25 553.3 792.7
25 415.2 967.5
329.7 666.0 927.4
20 6.73 27.29 20 721.5
40.65 42.57 445.1 519.9
25.46 978.1
15 27.83 15 632.2 814.5
11.70 37.04
10 29.27 10
32.97
5 5

0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

191
Results

B: Mass chromatogram and full MS of pure curvularin


RT: 5.00 - 45.00 Hassan545 #520 RT: 16.77 AV: 1 NL: 1.03E6
16.77 N L: 1.03E6 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 291.4
Bas e Peak m /z=
100
290.5-291.5 F: - c

[M-H]-
95

90
m/z 291.4 ES I Full m s [
100.00-1000.00]
MS Has s an545
95

90
85
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60

Relative Abundance
55
55
50
50
45 45
40 40
35 35
30 30
25 25
20 20
292.4
15 15

10 10

5 15.63 22.05 23.23 5


18.17 25.24 27.78 30.74 33.06 161.1 190.3 247.3 305.3
9.31 11.06 13.77 36.47 39.77 376.3 441.4 503.3 583.0 605.3 619.7 733.9 797.7 848.6 885.6 962.9
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

Figure 3.35: MS detection of curvularin (A: in C. album fraction 4, B: pure compound).

A: Mass chromatogram and full MS of stemphyperylenol in C. album fraction 4


RT: 5.00 - 45.00 Hassan547 #501 RT: 15.53 AV: 1 NL: 3.15E5
15.53 N L: 1.44E5 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 333.4
Bas e Peak m /z=
100
95 m/z 351.3 350.5-351.5 F: - c
ES I Full m s [ 95
90 100.00-1000.00]
MS Has s an547 90
85
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60
Relative Abundance

55
55
50

45
50

45
351.3 [M-H]-
40 40
35 35
30 30
25 25
20 261.4
20
303.4
15 15
7.64
10 10
7.77 233.5 739.0
21.07 28.56 33.81 38.35 387.3
5 7.51 13.10 24.81 5 197.0 515.0 565.2 804.4 916.2 994.7
19.15 21.80 28.76 403.2 486.3 651.2 703.5 733.8 852.9
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

B: Mass chromatogram and full MS of pure stemphyperylenol


RT: 5.00 - 45.00 Hassan546 #496 RT: 15.73 AV: 1 NL: 5.10E5
15.73 N L: 2.68E5 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 333.4
Bas e Peak m /z=
100
95

90
15.55
m/z 351.2 350.5-351.5 F: - c
ES I Full m s [
100.00-1000.00]
95

MS Has s an546 90
85
85
80
80
75
75
70
70
65
65
60
Relative Abundance

60
Relative Abundance

55

50
55 351.2 [M-H]-
50
45 45
40 40
35 35
30 30
25 25
20 20 261.4
15 15
303.5
10 10 387.1

5 16.31 20.71 22.10 28.18 29.24 35.76 5


12.88 14.38 39.96 43.27 160.8 248.2
388.0 459.2
512.8 529.2 644.5 740.8 765.9 807.1 863.7 926.6 940.9
32.45
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

Figure 3.36: MS detection of stemphyperylenol (A: in C. album fraction 4, B: pure compound).

192
Results

A: Mass chromatogram and full MS of macrosporin in C. album fraction 5


RT: 5.00 - 45.00 Has san549 #777 RT: 24.57 AV: 1 NL: 7.80E5
24.57 N L: 7.80E5 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 283.4
Bas e Peak m /z=
100
282.5-283.5 F: - c

[M-H]-
95

90
m/z 283.4 ES I Full m s [
100.00-1000.00]
95

MS Has s an549 90
85
85
80
80
75
75 268.4
70
70
65
65
60
Relative Abundance

60

Relative Abundance
55
55
50
50
45 45
40 40
35 35
30 30
25 25
20 20 240.5
15 15 284.4
10 10 211.5
10.69
5 5 169.3
293.4 465.5 505.7
6.09 9.51 12.67 16.78 20.96 23.47 27.23 29.27 35.86 37.57 39.05 44.74 373.0 411.3 593.4 613.3 659.1 681.4 775.6 847.7 871.5 926.6 985.5
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

B: Mass chromatogram and full MS of pure macrosporin


RT: 5.00 - 45.00 Has san548 #772 RT: 24.59 AV: 1 NL: 6.18E5
24.59 N L: 6.18E5 T: - c ESI s id=25.00 Full ms [ 100.00-1000.00]
100 283.3
Bas e Peak m /z=
100
95 m/z 283.3 282.5-283.5 F: - c
ES I Full m s [ 95
90

85
100.00-1000.00]
MS Has s an548 90 [M-H]-
85
80
80
75 268.3
75
70
70
65
65
60
Relative Abundance

60

Relative Abundance
55
55
50
50
45 45
40 40
35 35
30 30
25 25
240.3
20 20

15 15 284.3
211.3
10 10
25.21 197.2
5 25.89 5 169.3
11.31 14.49 18.73 23.66 30.05 31.97 35.69 37.11 40.79 43.30 285.5 420.4 438.2 469.3 541.2 627.1 689.6 727.3 798.5
840.4 876.4 944.9 964.5
0 0
5 10 15 20 25 30 35 40 45 100 200 300 400 500 600 700 800 900 1000
Tim e (m in) m/z

Figure 3.37: MS detection of macrosporin (A: in C. album fraction 5, B: pure compound).

3.5.4. Tracing of Chaetomium sp. metabolites in Otanthus maritimus fractions


In a manner completely analogous to the procedures described above for Polygonum
senegalense, Urospermum picroides and Chenopodium album, similar investigations were
also carried out with extracts of Otanthus maritimus, from which the endophytic fungus
Chaetomium sp. was isolated. For these studies, intense MS peaks for cochliodinol,
isocochliodinol, orsellinic acid and aureonitolic acid were used. However, it was not possible
to detect any of these secondary metabolites in any of the host-pant-derived fractions, even
though all of them were detectable in the crude extract of the fungus.

193
Discussion

4. Discussion

4.1. Choice of culture media


The physiology of secondary metabolism has often been neglected and still few of the
regulatory features involved in the biosynthesis of natural products have been elucidated. One
of the factors having great impact on growth and production of secondary metabolites from
microorganisms is medium composition and culture conditions (Bills, 1995). Thus it may be
necessary to use several media and growth conditions when strains are to be investigated for
their full metabolic potential in order to generate conditions that will allow the expression of
as broad a range of secondary metabolites as possible for a given strain to increase the chance
to generate novel drug candidates (Larsen et al., 2005). Furthermore, some natural products
are only produced under certain environmental conditions and if all trace metals, phosphate
and other medium factors are present in certain ranges of concentrations (Knight et al., 2003).
Thus, optimal media for good metabolite production can change for different genera being
investigated (Larsen et al., 2005).
Different and relatively easy to control conditions to investigate in a discovery
programme include growing cultures at both solid and liquid conditions, incubation at two or
more temperatures, incubation at two or more shaker speeds, incubation for at least two
different time periods, media with at least two different pH levels, choosing carbon and
nitrogen sources at different concentrations, high- or low phosphate content, adding trace
minerals etc. (Knight et al., 2003).
Some authors strongly argue in favour of using solid substrate fermentations in studies
of fungal metabolites since fungi, unlike other microorganisms, typically grow in nature on
solid substrates such as wood, roots and leaves of plants (Nielsen et al., 2004). On the other
hand, some believe and argue that all metabolites can be expressed in liquid culture by
varying carbohydrate composition, nitrogen source, oxygen tension, pH, redox potential,
water activity, as the right conditions will produce intracellular conditions that will trigger
production of a certain metabolite. It was found that metabolites associated to spore or
sclerotia formation are often produced under solid conditions, whereas the production of other
metabolites is enhanced under liquid conditions (Larsen et al., 2005, Nielsen et al., 2004).
Of course, in a screening effort aimed at the discovery of novel natural products which
includes a variety of different strains, one has to focus on a limited set of parameters, since
otherwise the sheer number of extracts generated will easily be overwhelming. Thus, in this
study chosen fungal strains were cultured in liquid (Wickerham) medium as well as on solid

194
Discussion

rice medium. Bioactivity and chemical profiles of the obtained extracts from both cultures
were compared and subjected to further investigation. HPLC chromatograms of the EtOAc
extracts of liquid and rice cultures showed different chemical patterns for all the fungal strains
investigated in this study. Moreover, EtOAc extracts of liquid and rice cultures showed
different antimicrobial and cytotoxic activities in preliminary biological screening tests which
was in accordance with the different chemical picture. It was also observed as a general trait
that the yield of dry extract obtained from rice cultures was higher than that of liquid cultures
with varying rations in all investigated fungal strains. However, it cannot be excluded that this
finding, at least to a certain degree, was due to the fact that more polar material, e.g. sugars or
amino acids, were extracted from the culture medium in the case of the solid rice cultures
compared to the liquid medium

4.2. Strategies and methodologies for metabolite profiling


In order for natural product chemistry to continue to be competitive with purely
synthetic based discovery methods, natural product research needs to continually improve the
efficiency of the selection, screening, dereplication, isolation and structure elucidation
processes (Butler, 2004). Hence, talented microbial strains can be selected to be included in
screening programs, which together with the use of spectroscopic methods in combination
with chemoinformatics can be used as part of an effective dereplication strategy (Larsen et al.,
2005). In fact the chemical diversity and the resources of natural products are immense and
nowhere near fully exploited. Fungi are known to produce species specific profiles of natural
products which can be used as efficient tools to select some representative strains for
biological testing. Thus, extracts for chemical fractionation were selected based on the
biological activity and chemical profiles of the crude extract. Metabolite profiling is not an
easy task to perform since natural products display a very high structural diversity.
Consequently a single analytical technique does not exist, which is capable of profiling all
secondary metabolites in the biological source (Wolfender et al., 2005). However, advanced
analytical and spectroscopic methods, like the hyphenated techniques coupled to HPLC, can
give a good idea about the different substructures and/or functional groups of the structure.

4.2.1. HPLC/UV
With the advancement of HPLC as well as much more stable and better columns for
high resolution separation, combined with fast UV diode array detectors it has become easy to
acquire the UV spectrum of practically every single component from an extract, provided a

195
Discussion

suitable chromophore. Consequently the UV spectrum has turned into one of the most readily
accessible pieces of information related to structure of natural products which increased the
interest in exploiting its usefulness (Cannall, 1998).
In the present study, a lot of chemical compounds that share similar chromophoric
functions were examined by the hyphenated technique HPLC/UV-photodiode array detection
(LC/UVDAD) which showed very often this also translated into similar UV spectra, even
though there were significant differences in terms of additional non-chromophoric functions
or molecular weights (e.g. alternariol derivatives (1-5), altenusin and desmethyl altenusin (6-
7), altenuene and 4`-epialtenuene (11-12), isocoumarin derivatives (18-20), ampelanol (26)
and tetrahydroaltersolanol B (30) and the cochliodinol derivatives (35-36)).

4.2.2. HPLC/ESI-MS
With the arrival of electrospray ionization mass spectrometry (ESI-MS) and the
associated techniques about 25 years ago the scientific community obtained a highly versatile
tool for studies of natural products. ESI-MS has the advantage of being a soft and sensitive
ionization technique which can be optimized to produce mainly protonated or sodiated ions
(assuming positive ESI) from a very broad range of natural products (Smedsgaard and
Frisvad, 1996). Moreover, modern LC-MS system allow to switch between positive and
negative ionization very quickly, i.e. in the order of 1 second per spectrum, thus allowing to
obtain complimentary information to securely identify the molecular weight of an analyte, or
to obtain molecular weight information for compounds not ionizing upon positive, but only
upon negative ionization. Hence, ESI-MS represents a rapid method to differentiate and
estimate the presence of secondary metabolites in microbial extracts.
Furthermore, this method is also helpful in establishing the relation between closely
related natural products. In the context of the present study, this proved extremely valuable in
detecting the characteristic loss of 80 mass units from the new sulphated alternariol
derivatives (2 and 4) and anthraquinone derivatives (22 and 24) indicating the presence of a
sulphate group. It is difficult to come to a definite conclusion, but there is some degree of
probability that at least some members of this series of sulphated compounds had simply
overlooked in previous studies.

196
Discussion

4.2.3. Dereplication and partial identification of natural products by UV-based


techniques
Apart from exact structural formulae structural databases usually also contain physical
and chemical data including UV maxima and minima characteristic of the included
compounds. Moreover, modern HPLC-DAD systems allow to generate a library of UV
spectra in a rather straightforward manner which in turn is extremely valuable for
dereplication of compounds previously isolated. Many natural products such as polyketides
and alkaloids derived from aromatic amino acids have characteristic UV spectra due to their
polyunsaturated nature. In addition many such natural products often have one or more
carbonyl groups as part of ketone, carboxylic acid, ester or amide functional groups. Thus, the
UV based library established at the Institute of Pharmaceutical Biology at Düsseldorf in the
last 10 years was extensively used for dereplication purposes to investigate isolated fungal
strains for production of natural products but also as an approach to discover possible novel
bioactive metabolites with structural features similar to that of already known bioactive
compounds.

4.3. Isolation of natural products


Chromatographic techniques were used to isolate and purify the chemically most
interesting substances. Ideally the structurally unusual or novel compounds are also
responsible for the activity of the extract. The approach proved indeed efficient with respect
to isolating numerous new compounds, many of which probably being responsible for most of
the biological activity observed for the crude extract (e.g. alternariol and altenusin derivatives
in Alternaria extracts, altersolanol A in Ampelomyces rice culture extract, curvularin
derivatives in Stemphylium rice culture extract as well as cochliodinol and orsellinic acid in
Chaetomium liquid culture extract).

4.4. Compounds isolated from purified fungal strains

4.4.1. Compounds isolated from the endophytic fungus Alternaria sp.


Alternaria sp. was isolated from leaves of Polygonum senegalense growing wild in
Egypt. External application of an extract of the fresh leaves of this plant is reported in folk
medicine to be highly effective in treating skin troubles. Species of Polygonum are known in
traditional medicine for their diuretic, cholagic, antihemorragic and antiseptic actions

197
Discussion

(Smolarz, 2002). In addition, it was found that crude aqueous methanol extract of P.
senegalense exhibited molluscicidal activity (Dossaji and Kubo, 1980).
Alternaria species have a widespread distribution. Many species are plant pathogens,
which cause both pre- and post-harvest decay. A number of metabolites of polyketide origin,
including many α-dibenzopyrones such as alternariol and alternariol monomethyl ether have
been isolated from species of Alternaria (Stinson et al., 1986, Onocha et al., 1995). Many of
these metabolites were reported to be toxic to mammalian systems, nevertheless, our interest
in the metabolites produced by Alternaria was stimulated by the recent report of the
estrogenic potential of alternariol in cultured mammalian cells (Lehmann et al., 2006).
Chemical investigation of the ethyl acetate extract of the fungus grown in liquid culture
lead to the isolation of new sulphated derivatives of alternariol and its monomethyl ether (2
and 4) as well as the known compounds alternariol (1), alternariol-5-O-methyl ether (3),
altenusin (6), 2,5-dimethyl-7-hydroxychromone (13), altertoxin I (14), tenuazonic acid (15)
and adenosine. When grown on solid rice medium the fungus yielded four new compounds,
identified as 3`-hydroxyalternariol-5-O-methyl ether (5), desmethylaltenusin (7), alterlactone
(8) and alternaric acid (10) in addition to the known compounds 1, 3 and 6, talaroflavone (9)
and altenuene (11). Furthermore, we isolated a new altenuene isomer that was given the trivial
name 4`-epialtenuene (12).

4.4.1.1. Alternariol derivatives

4.4.1.1.1. Biosynthesis of alternariol derivatives


The biosynthesis of alternariol had been reported to involve assembling a heptaketide
chain, folded as shown in Figure 4.1, aromatizing, cyclizing, and release of alternariol in a
single step. The enzyme responsible for the production of alternariol is a fungal polyketide
synthetase. The central theme of polyketide biosynthesis is that iterative decarboxylative
Claisen condensations of malonyl thiolesters result in a growing carbon chain (Liu et al.,
1998). The folding of the polyketide chain was established by labeling studies, feeding 13C2-
labelled acetate to the appropriate organism and establishing the location of labeled intact C2
units in the final product by 13C NMR spectrometry. Whilst the precise sequence of reactions
involved is not known, the essential features would include two aldol condensations followed
by enolization in both rings to give a biphenyl, and lactonization would then lead to
alternariol. The oxygenation pattern in alternariol shows alternate oxygens on both aromatic
rings, and an acetate origin is readily presumed, even though some oxygens have been

198
Discussion

consumed in ring formation processes. The lone methyl ‘start of chain’ usually obvious in
acetate-derived compounds is also detected, though the carboxyl ‘end of chain’ reacted with a
convenient hydroxyl function, which may have arisen through enolization, to form the lactone
function. The formation of alternariol monomethyl ether from alternariol is known to occur
catalyzed by a O-methyltransferase in a transmethylation reaction involving S-adenosyl
methionine (Stinson et al., 1986, Dewick, 2006).
An alternative mechanism for alternariol biosynthesis has been also suggested, which
would proceed through the rearrangement of a norlichexanthone intermediate, in analogy to a
well-documented step in aflatoxin biosynthesis. In this case the polyketide chain is assembled
on the surface of the enzyme in a configuration that facilitated the formation of
norlichexanthone (see Figure 4.1). Then oxidative cleavage of the aromatic phloroglucinol
ring occurs, followed by limited rotation of the aryl fragment and ring closure to produce the
coupling pattern observed in alternariol (Stinson et al., 1986).

O
O OH
[O]

HO

[O] [SAM]
OH
Rotation OH OH
HO Ring closure
O COOH COOH
HO
Desmethylaltenusin (7)
O HO
Altenusin (6)
Alterlactone (8) OH
OH
[O]
SCoA - H2O
* OH
O
O

O
Acetyl-CoA *
2 x Aldol Enolization
COOH * * * - H2O
O O O O OH
SCoA
6x *
* * * COOH COOH
O O O SCoA O O HO OH
O
Malonyl-CoA Heptaketide chain
Lactone
O OH OH
formation

* Ox
* OH
*
* * * *
O O
*
SCoA HO * *
O SCoA O SCoA
OH

* * *
* * O Rotation
O * * O * * O OH
Ring closure
* * * *
O
* * * * *
HO HO HO O O
Heptaketide chain Norlichexanthone Alternariol (1)

Figure 4.1: Postulated biosynthesis of alternariol derivatives (Stinson et al., 1986, Dewick, 2006).

199
Discussion

4.4.1.1.2. Biosynthesis of biphenic acids and related compounds


The formation of altenusin (6) and the previously unreported desmethylaltenusin (7)
may be envisaged as proceeding by cyclization of a heptaketide precursor to give the biphenyl
derivative shown in Figure 4.1 (similar to alternariol biosynthesis). An oxidation and
reduction sequence, and methylation of the phenolic hydroxyl, in case of 6, affords the
biphenyl metabolites 6 and 7. The previously unreported alterlactone (8) is most probably
biosynthetized through the same pathway from altenusin (6) by oxidation of the aromatic
methyl group, rotation and closure of the lactone ring at a different site compared to
alternariol derivatives (1-5), namely at the aromatic hydroxymethyl group.
The heptaketide biphenyl intermediate in the biosynthesis of biphenic acids 6 and 7 is
also believed to be the precursor to the structurally and biosynthetically interesting spirocyclic
metabolite, talaroflavone (9), previously isolated from Talaromyces flavus (Ayer and Racok,
13
1990a), as well as the new natural product, alteric acid (10). Feeding C-labelled sodium
acetate to T. flavus resulted in the incorporation of label at six carbons of 9. A biosynthetic
pathway that accounts for this is illustrated in Figure 4.2 (Ayer and Racok, 1990b).

OH OH

* *
HOOC * H+ HOOC *

O
* * * * *
CH313COONa * O * O

* * * *
HO OH HO O
[O]
-
OH

H2O
[O]
OH OH
O OH
*
HOOC *
* * *
HOOC *
O
Cyclize * Cyclize
* * *
* * O [O] * O - *CO2 * O
* OH OH

* * O
* [O], - H2O O -
HO
O [H] *COOH
Talaroflavone (9)
O OH

HO

HO
O
Alteric acid (10)

Figure 4.2: Postulated biosynthesis of talaroflavone (Ayer and Racok, 1990b).

200
Discussion

4.4.1.1.3. Bioactivity and structure activity relationship of alternariol and biphenic acid
derivatives
Alternariol (1) was found to have fairly powerful activity against some Gram-positive
and Gram-negative bacteria. Furthermore, the compound had no phytotoxic effect when
sprayed on to young carrot seedlings and applied to their roots (Raistrick et al., 1953,
Freeman, 1966).
Recently, alternariol (1) was reported to show estrogenic potential in cultured
mammalian cells. Furthermore, it inhibited cell proliferation by interference with the cell
cycle (Lehmann et al., 2006). The results of our cytotoxicity test of the isolated alternariol
derivatives from the endophytic fungus Alternaria sp. toward L5178Y (mouse lymphoma)
cell line (see Table 3.13), strongly suggest that the free hydroxyl group at C-4`, which is
present in all strongly active alternariol derivatives (1-3), plays an important role in the
cytotoxic activity. Upon substitution of the 4`-OH by a sulphate group in alternariol
monomethyl ether sulphate (4) activity was greatly decreased. Moreover, presence of a
hydroxyl substituent at C-3` reduced the activity of hydroxyalternariol monomethyl ether (5)
to only moderate. Substitution of the 5-OH by a sulphate group in alternariol sulphate (2) or a
methyl group in alternariol monomethyl ether (3) had no effect on the activity of the
compounds (see Figure 4.3). On the other hand, altenusin (6) and desmethylaltenusin (7)
showed strong activities as well. Substitution of the 5-OH by a methyl group in
desmethylaltenusin (7) had no effect on the activity of the compound. Moreover, alterlactone
(8) was also active in the cytotoxicity assays. Interestingly, these results were in accordance
with the results of protein kinase inhibition assay except for hydroxyalternariol monomethyl
ether (5) which showed a protein kinase inhibitory activity comparable to that of alternariol
(1) (see Table 3.14). Both altenusin (6) and desmethylaltenusin (7) were active against several
protein kinases tested in the assay, which is in accordance with the recent report of altenusin
as potent protein kinase inhibitor (Oyama et al., 2004).

201
Discussion

O OH
3` 3

4` 1` 1
HO

OH

Alternariol (1)

Substitution at 4`-OH greatly Substitution at 5-OH does


decreases bioactivity by not affect bioactivity as in
∼100% as in O

O OH
O OH substitution at C-3`
O OH decreases cytotoxic activity by
∼50% with no effect on protein HO

HO3SO
kinase inhibition as in
O OSO3H

O
HO O OH Alternariol-5-O-sulphate (2)
O
Alternariol-5-O-methyl HO
O OH
ether sulphate (4)
O
HO

3`-Hydroxyalternariol-5-
O-methyl ether (5) O
Alternariol-5-O-
methyl ether (3)

Figure 4.3: Structure-activity relationship for alternariol derivatives.

4.4.1.1.4. Toxicity studies of alternariol derivatives


The chicken embryo assay is often used as a convenient assay for toxicity of
mycotoxins as well as other compounds. Investigation of the effects in the chicken embryo
assay for alternariol (1), alternariol monomethyl ether (3) and altenuene (11) showed that
these compounds exhibited no toxic effect at doses up to 1.000, 500, and 100 µg per egg,
respectively, when administered to 7-day-old chicken embryos by yolk sac injection. In
addition to the lack of mortality, these metabolites exerted no teratogenic effect in the
developing embryo (Griffin and Chu, 1983). In another study, one day-old chicks were fed a
standard diet supplemented with purified 3 at levels of up to 100 mg/kg of feed for 4 weeks
exhibiting no mortality or significant loss in performance, indicating a general lack of toxicity
of 3 in poultry systems (Griffin and Chu, 1983). Similarly, alternariol (1) and alternariol
monomethyl ether (3) were shown to be nonmutagenic to Salmonella strains in the Ames
Salmonella typhimurium assay (Davis and Stack, 1994). Thus, the finding of other
investigators that alternariol monomethyl ether (3) was weakly mutagenic could have resulted
from the presence of a small amount of one of the highly mutagenic altertoxins in the sample
of alternariol monomethyl ether originally tested (Davis and Stack, 1994).

202
Discussion

4.4.1.1.5. Occurrence of sulphated metabolites


In the plant kingdom, sulphated products, mainly flavonoids, have been isolated from
more than 250 species, including dicotyledons and monocotelydons (Barron et al., 1988).
Sulphated phlorotannins were also detected in marine algae (Glombitza and Knöss, 1992). In
fungi, however, few studies of sulphate conjugates appear in the literature. Choline sulphate is
produced by Aspergillus nidulans presumably as sulphur reserve (Hussey et al., 1965). The
sulphate conjugation of aromatic hydrocarbons in liquid fermentation by Cunninghamella
elegans was also reported (Cerniglia et al., 1982). Furthermore, zearalenone sulphate was
detected in four species of Fusarium as well as a sterol sulphate in corn cultures of F.
graminearum (Vesonder et al., 1990, Plasencia and Mirocha, 1991). However, the mechanism
of sulphate conjugation in Fusarium sp. is unknown (Plasencia and Mirocha, 1991). In this
study sulphated derivatives of alternariol and its monomethyl ether (2 and 4, respectively)
were isolated from the n-BuOH extract of Alternaria liquid culture. In addition, the sulphated
anthraquinones, macrosporin sulphate (22) and methylalaternin sulphate (24), were isolated
from MeOH and EtOAc extracts of Ampelomyces liquid and rice cultures, respectively (see
4.4.2.).

4.4.1.2. Biosynthesis of 2,5-dimethyl-7-hydroxychromone


2,5-Dimethyl-7-hydroxychromone (13) has been isolated from the roots of Polygonum
cuspidatum, a plant used in Chinese and Japanese traditional medicine (Kimura et al., 1983),
from the aerial parts of Hypericum perforatum (Yin et al., 2004), and from a Japanese
commercial rhubarb sample (Kashiwada et al., 1984). The first report for its isolation from a
fungal source, Talaromyces flavus, was in 1990 (Ayer and Racok, 1990a). The biosynthesis of
13 did not seem to follow the pattern of most chromones that arise from a pentaketide
precursor but probably originates from a hexaketide precursor as illustrated in Figure 4.4
(Ayer and Racok, 1990a).

203
Discussion

O O O SCoA HO SCoA
OH O

O O O

O O

Hydrolysis
- CO2

HO O HO
OH O

Cyclization
- H2O

O O

2,5-Dimethyl-7-
hydroxychromone (13)

Figure 4.4: Postulated biosynthesis of 2,5-Dimethyl-7-hydroxychromone (Ayer and Racok,


1990a).

4.4.1.3. Biosynthesis of reduced perylenequinones


Altertoxin I (14) is an example of reduced perylenequinones so far identified in fungi
of the morphologically closely related genera Alternaria and Stemphylium. The biosynthesis
of these compounds occurs most probably via oxidative coupling of two molecules of a
tetralone derivative, which in turn is synthesized from a pentaketide derivative (Okuno et al.,
1983) by so-called head-to-head coupling, followed by reduction and hydroxylation in
different positions (Arnone et al., 1986). The proposed biosynthetic pathway was confirmed
by an incorporation experiment of 13C-labelled sodium acetate and may be depicted as shown
in Figure 4.5 (Okuno et al., 1983).

OH O OH O

O O * * *

* * * OH
O O * *
*CH3COO- OH
H
* * OH OH
O OH
* *
* * *

OH O OH O
Altertoxin I (14)

Figure 4.5: Proposed biosynthetic pathway of reduced perylenequinones (Okuno et al., 1983).

204
Discussion

4.4.1.4. Tautomerism and biosynthesis of tenuazonic acid


The tautomeric behavior of 3-acylpyrrolidine-2,4-diones (e.g. tenuazonic acid (15))
involves two sets of rapidly interchanging internal tautomers (a↔b) and (c↔d), where each
set arises through proton transfer along the intramolecular hydrogen bond, together with two
pairs of slowly interconverting external tautomers (ab↔cd), arising from the rotation of the
acyl side chain (see Figure 4.6). It was found that the internal tautomerization occurs too
rapidly to be detected on the time scale of an NMR experiment, the external tautomerism,
however, occurs at a rate which can be measured on the NMR time scale. In non polar
solvents (e.g. CDCl3) the interconversion between the external enolic tautomers (ab↔cd) was
found to be a comparatively slow process, while interconversion between the pairs of internal
tautomers (a↔b, c↔d) was found to be fast. Thus, the two sets of resonances observed in the
NMR spectra are attributed to the external tautomers (ab) and (cd). In polar solvents (e.g.
CD3OD) the two external pairs were found to interconvert at a much faster rate and, therefore,
the NMR signals of the external tautomers coalesce (Nolte, 1980; Royles, 1995).

O
O

HN
HN
fast
O
O R
R
O H
O
H
slow b
a
O

O
HN

O
O H
H
O
slow
O
O HN
HN

fast
R
R
O
O
d
c

Figure 4.6: Tautomerism of 3-acylpyrrolidine-2,4-diones (Nolte, 1980).

205
Discussion

From biosynthetic studies on tenuazonic acid (15) established by feeding experiments


using 14C-labelled acetate it was concluded that the biosynthesis occurs via cyclization of N-
acetoacetyl-L-isoleucine to produce tenuazonic acid as shown in Figure 4.7 (Royles, 1995).

COOH

COOH
NH
O
NH
2 x MeC*OOH *
NH2
O

* O
O
OH
N-acetoacetyl-L-isoleucine Tenuazonic acid (15)

Figure 4.7: Postulated biosynthesis of tenuazonic acid (Royles, 1995).

4.4.1.5. Bioactivity of selected Alternaria metabolites


Altertoxin I (14) had been previously isolated from a number of Alternaria spp.
(Okuno et al., 1983, Stack et al., 1986, Hradil et al., 1989). The compound showed a high
level of phytotoxicity toward corn (B73), as measured by the size of necrotic lesions 72 h
after application of 1 µg of compound, with strong selectivity as well (Hradil et al., 1989).
Tenuazonic acid (15) was originally isolated from the culture filtrate of Alternaria
tenuis and subsequently other species have also been found to produce it. The compound
exhibited a low level of antibacterial activity and showed an inhibitory effect on several
viruses including poliovirus MEF-1, ECHO-9, parainfluenza-3, vaccinia, and herpes simplex
(HF) (Miller et al., 1963). It also showed the ability to inhibit human adenocarcenoma
growing in the embryonated egg with a proposed mode of action involving the inhibition of
amino acid incorporation into microsomal proteins. However, the compound has been of
limited value due to its extreme toxicity (Royles, 1995).
In this study both compounds 14 and 15 were tested for cytotoxicity toward L5178Y
cell line where they showed weak and moderate activities, respectively. In the protein kinase
inhibition assay, however, 14 showed to be active on various protein kinases, while 15
showed no activity.

206
Discussion

4.4.2. Compounds isolated from the endophytic fungus Ampelomyces sp.


The Ampelomyces sp. strain investigated was isolated from flowers of Urospermum
picroides growing wild in Egypt. U. picroides is typical for the traditional Mediterranean diet
and its extract shows anti-inflammatory activities (Strzelecka, et al., 2005).
Historically, pycnidial fungi belonging to the genus Ampelomyces were among the
first mycoparasites to be studied in detail and were also the first fungi used as biocontrol
agents of plant parasitic fungi (Yarwood, 1932; Sundheim and Krekling, 1982). The
interactions between host plants, powdery mildew fungi and Ampelomyces mycoparasites are
one of the most evident cases of tritrophic relationships in nature, in which organisms on three
different trophic levels integrate functionally through host-parasite interactions (Kiss et al.,
2004). While it seems likely that fungal metabolites are involved in many reported
interspecies interactions, Ampelomyces mycoparasites attracted our attention because they
have rarely been studied chemically.
Extracts of the fungus grown in liquid culture afforded a new pyrone, ampelopyrone
(17), two new sulphated derivatives of macrosporin (22) and methylalaternin (24) together
with the known compounds methyltriacetic lactone (16), citreoisocoumarin (20), macrosporin
(21), methylalaternin (23), ergosterol and cerebroside C. From extracts of the fungus grown
on solid rice medium we obtained two new isocoumarins, desmethyldiaportinol (18) and
desmethyldichlorodiaportin (19) and a new hexahydroanthronol, ampelanol (26), as well as
compounds 21-24, altersolanol A (25), the atropisomers, alterporriols D and E (27 and 28,
respectively), and altersolanol J (29).

4.4.2.1. Anthraquinones and modified anthraquinones

4.4.2.1.1. Biosynthesis of anthraquinones and modified anthraquinones


Fungi are known to form anthraquinones by linear head-to-tail combination of acetate
and malonate, namely, octaketide chains, catalyzed by a fungal polyketide synthase, followed
by the loss of carboxylic acid carbon from the terminal unit at C-3, but the detailed sequence
of condensation, dehydration and hydroxylation steps is not well known (see Figure 4.8). The
periphery of the carbon skeleton is constructed by folding the octaketide chain, and then the
ring at the centre of the fold is formed first, followed in turn by the next two rings (Ohnishi et
al., 1991, Dewick, 2006). The validity of the octaketide pathway was confirmed by
spectroscopic studies on the biosynthesis of macrosporin (21) utilizing single and double
labeled acetates (Stoessl et al., 1983, Suemitsu et al., 1989, Ohnishi et al., 1992).

207
Discussion

Altersolanol A (25) is clearly closely related to its anthraquinone co-metabolites 21-24


since they have the same meta-substituted aromatic ring and a C-methyl group in their
biogenetic 2-position. It is indeed possible that the anthraquinones are formed from
altersolanol derivatives by dehydration brought about enzymatically or by acid catalysis
within the mold tissue. However, the reverse may also be true, i.e. that 25 is derived from
anthraquinone precursors. At any rate, it is very probable that altersolanol A (25) and
macrosporin (21) share a common biogenetic origin (Stoessl, 1969b).

[SAM]
O

SCoA [O]
OH
O
O OH O
Acetyl-CoA Oxidation Macrosporin (21)
O O O Enolization
O OH
COOH COOH
- CO2
O
SCoA O O O OH
7x
Octaketide chain
O
OH

Malonyl-CoA
OH O OH

Altersolanol A (25)
OH O OH

OH

OH Oxidative coupling
Dimerization at C-8
O

O OH

O OH

OH

OH

OH O OH

Alterporriols D and E (27 and 28)

Figure 4.8: Postulated biosynthesis of anthraquinones and modified anthraquinones (Stoessl


et al., 1983, Suemitsu et al., 1989, Ohnishi et al., 1991, 1992).

4.4.2.1.2. Bioactivity of anthraquinones and modified anthraquinones


Altersolanol A (25) inhibited the growth of Gram-positive bacteria and Pseudomonas
aeruginosa IFO 3080 when tested by the broth dilution method (Yagi et al., 1993). It was
found that the compound acts as an electron acceptor in the bacterial membrane to inhibit
bacterial growth (Haraguchi et al., 1992). Moreover, 25 was found to be highly phytotoxic

208
Discussion

when injected into tomato leaves. However, using this assay, the two altersolanol A dimers,
27 and 28, were only very weakly phytotoxic (Lazarovits et al., 1988).
The anthraquinones and modified anthraquinones isolated from the endophytic fungus
Ampelomyces sp. in this study were tested for cytotoxicity toward L5178Y (mouse
lymphoma) cell line (see Table 3.26). The obtained results showed that the
tetrahydroanthraquinone, altersolanol A (25), was the most active compound. The anthranol
derivatives, ampelanol (26), altersolanol J (29) and tetrahydroaltersolanol B (30) showed
moderate to weak activities, suggesting that the para-quinone moiety is of great importance
for the cytotoxic activity. Furthermore, the monomer 25 showed much higher activity than its
dimers 27 and 28. Methylalaternin (23) was the most active under the anthraquinone
derivatives, while macrosporin (21) only showed moderate activity. Sulphate substitution at 7-
OH in 22 and 24 reduced the activity indicating the possible contribution of this hydroxy
group to the activity (see Figure 4.9). Furthermore, results of protein kinase inhibition assay
showed a similar pattern of activity, with altersolanol A (25) being the most active compound
inhibiting various protein kinases in the protein kinase inhibition assay, while methylalaternin
(23) and macrosporin (21) were less active (see Table 3.27).

209
Discussion

Increased cytotoxic activity


O O O O

O O O O

OH OH OSO3H OSO3H

OH O OH OH O OH O OH O OH

Methylalaternin (23) Macrosporin (21) Macrosporin sulphate (22) Methylalaternin sulphate (24)

Decreased cytotoxic activity

O OH

OH

OH

OH O OH

Altersolanol A (25)

Dimerization decreses Reduction of quinone carbonyl


bioactivity as in decreses bioactivity as in
OH O OH OH OH
H
OH O

OH
OH

O OH
H
O OH OH O OH

O OH Ampelanol (26)
O
O H
OH O

OH
OH
OH
OH O OH H
OH OH
Alterporriols D and E
(27 and 28) Altersolanol J (29)

Figure 4.9: Structure-activity relationship of anthraquinones and modified anthraquinones.

One very promising new approach for developing novel antibiotics is based on the fact
that bacterial colonization and pathogenesis seemingly depends on the ability of the bacteria
to communicate and thereby coordinate the behavior of the entire population (Hall-Stoodley
et al., 2004). Population activity such as biofilm formation is coordinated by simple
communication systems which in many Gram-negative bacteria is based on homoserine
lactone (HSL) signals, which have been described in numerous pathogens (Costerton et al.,
1999). HSL systems are referred to as quorum sensing (QS) systems, i.e. they express target
genes in relation to the quorum size (or density) of the population and in most known cases
control expression of virulence factors. A screening strategy aiming at inhibition of QS is
therefore not targeting bacterial growth but instead blocking the coordination of bacterial
population activity. This means that a quorum sensing inhibiting (QSI) drug is not generating

210
Discussion

a selective pressure on the bacteria, and it is therefore unlikely that bacteria will develop
resistance towards a given QSI compound (Larsen et al., 2005). Upon testing the
anthraquinones and modified anthraquinones for inhibition of biofilm formation of
Staphylococcus epidermidis, methylalaternin (23) showed very high activity with a MIC of
12.5 µg/mL and complete inhibition of biofilm formation, whereas altersolanol A (25) having
MIC of >50 µg/mL inhibited biofilm formation by 50%.

4.4.2.2. Pyrone and isocoumarin derivatives

4.4.2.2.1. Biosynthesis of pyrone and isocoumarin derivatives


From a biogenetic point of view, β-polyketo carboxylic acids are expected to convert
into both the corresponding pyrones and phenolic compounds according to the mode of
enzymatic cyclization as shown in Figure 4.10 (Lai et al., 1991).

OH

OH O

R OH

O [SAM]
COOH
Or
R O O
R O O
HO R

O SCoA
O

Methyltriacetic lactone (16),


ampelopyrone (17) and OH O
related pyrone compounds

Desmethyldiaportinol (18),
R: Polyene or poly-β-keto chain desmethyldichlorodiaportin (19)
and citreoisocoumarin (20)

Figure 4.10: Biosynthetic pathways of pyrones and isocoumarins (Lai et al., 1991).

The isocoumarin derivatives 18-20 are typical heptaketide compounds with oxygen
atoms located at alternate carbons. The carbonyl carbons in the side chain (R) might be
reduced by fungal reductases to hydroxyl groups (Watanabe et al., 1998).
On the other hand, recently naturally occurring organohalogen compounds have
assumed an important role in the field of natural products. The number of natural

211
Discussion

organohalogen compounds has multiplied about 250 times in the past 40 years. Fungi and
lichens are known to be a bountiful source of such metabolites, and the earliest examples of
natural chlorine-containing metabolites include the fungal metabolites griseofulvin and
chloramphenicol. The mechanism for the formation of organohalogen compounds was
reported to initially involve the oxidation of halide by a peroxidase enzyme and hydrogen
peroxide (Gribble, 1998). A more recent study presented evidence that NADH-dependent
halogenases rather than haloperoxidases are the enzymes that actually do the chlorination
(Hohaus et al., 1997).

4.4.2.2.2. Bioactivity of pyrone and isocoumarin derivatives


Pyrone and isocoumarin derivatives were subjected to cytotoxicity testing toward
L5178Y (mouse lymphoma) cell line (see Table 3.26). Desmethyldiaportinol (18) was the
most active of the compounds, whereas desmethyldichlorodiaportin (19) and
citreoisocoumarin (20) were found to be moderately active and inactive, respectively. This
indicated that presence of bulky substituents on the side chain attached to the isocoumarin
structure may result in reduction and loss of activity (see Figure 4.11). On the other hand, the
pyrone compounds 16 and 17 were found to be inactive in the test (see Figure 12).
Furthermore, all compounds were inactive when tested for protein kinase inhibition, which
suggested that desmethydiaportinol cytotoxic acitivity did not involve interaction with protein
kinases.

Increased cytotoxic activity


Cl

HO HO HO
OH Cl

O OH O OH O OH O

OH O OH O OH O

Desmethyldiaportinol (18) Desmethyldichlorodiaportin (19) Citreoisocoumarin (20)

Decreased cytotoxic activity

Figure 4.11: Structure-activity relationship of isocoumarin derivatives.

212
Discussion

OH O

O O O O

Mehyltriacetic
lactone (16) HO
Ampelopyrone (17)

Figure 4.12: Structures of inactive pyrones.

4.4.3. Compounds isolated from the endophytic fungus Stemphylium botryosum


Stemphylium botryosum was isolated from Chenopodium album, a plant growing wild
in Egypt. C. album is reported in folk medicine to possess anthelmintic properties and the
seed oil is effective against many forms of intestinal parasites. The plant was also used in the
past as oral contraceptive (Laszlo and Henshaw, 1954). It is also used in the Indian Himalayan
Region for treating liver diseases (Samant and Pant, 2006).
Stemphylium botryosum is a mould which causes leaf spot of lettuce, a disease of
economic importance in many countries. Both saprotrophic and pathogenic forms of
Stemphylium occur on a wide range of plants. Many species of Stemphylium are economically
important pathogens of agricultural crops. Usually, the toxicity of moulds is related to the
production of one or more phytotoxins, which is the case in Stemphylium species that are
reported to produce a wide array of toxins (Arnone and Nasini, 1986, Camara et al., 2002).
Chemical investigation of the EtOAc extract of Stemphylium botryosum, grown on
solid rice medium, yielded altersolanol A (25), tetrahydroaltersolanol B (30),
stemphyperylenol (31), as well as the macrocyclic lactones, curvularin (32) and
dehydrocurvularin (33), in addition to macrosporin (21).

4.4.3.1. Biosynthesis of stemphyperylenol


The biosynthesis of stemphyperylenol (31) occurs as described above in the context of
other reduced perylenequinones (see 4.4.1.3.). However, it is remarkable that, whereas all the
compounds so far found appear to derive from so-called head-to-head coupling,
stemphyperylenol (see Figure 4.13) seems to be an unusual example of a head-to-tail coupling
of pentaketide-derived moieties (Arnone and Nasini, 1986).

213
Discussion

OH O

OH
H H
HO

O OH

Figure 4.13: Structure of stemphyperylenol (31).

4.4.3.2. Biosynthesis of macrocyclic lactones


The fungal metabolite curvularin (32) is a macrolide octaketide produced by some
Curvularia (Coombe et al., 1968), Alternaria (Robeson and Strobel, 1981) and Penicillium
(Lai et al., 1989) species. Interestingly, curvularin-type metabolites had never been previously
described for the chemically well-investigated Stemphylium botryosum. Curvularin (32) and
dehydrocurvularin (33) are lactones containing a twelve membered ring. The acyl-resorcinol
fragment is probably formed by cyclization of an intermediate containing a carbon chain
formed by serial head-to-tail linkage of acetate units typical of polyketide biogenesis (Liu et
13 14
al., 1998). Mould incorporation of labeled C- or C-acetate units into 32 or 33 indicated
that the C16 metabolites are derived from eight acetate units, very probably through linear
precursors (Birch et al., 1959, Arai et al., 1989). More recent studies (see Figure 4.14) also
showed that diketides I and its reduced congener II are the precursors in the biosynthetic
pathway, and additionally, that the double bond of the tetraketide intermediate III is essential
for incorporation. This strongly supports the hypothesis that the unsaturated tetraketide III
represents the final oxidation state achieved on the polyketide synthase prior to addition of the
next C2 unit. Since the subsequent penta-, hexa-, hepta-, and octaketide intermediates require
no reduction, this suggested that dehydrocurvularin (33) is the initial polyketide synthetase
product (Liu et al., 1998). Curvularin (32) could be obtained by reduction of
dehydrocurvularin (33) (Arai et al., 1989). The proposed biosynthetic pathway is illustrated in
Figure 4.14.

214
Discussion

HO
SCoA

O HO
O
Acetyl-CoA [H]
Head-to-tail linkage SCoA SCoA + 2 C2
CoAS
COOH [H], - H2O

7x SCoA O Diketide O Diketide Tetraketide


O

+ 4 C2
O
Malonyl-CoA

OH
O O O HO
O O

HO O
HO
[H]
Enolization
-H2O O

OH O O O
OH O

Curvularin (32) Dehydrocurvularin (33)

Figure 4.14: Biosynthesis of macrocyclic lactones (Birch et al., 1959, Liu et al., 1998).

4.4.3.3. Bioactivity of selected Stemphylium metabolites


Stemphyperylenol (31) was reported to show antibacterial activity in vitro against
Bacillus subtilis, B. cereus and E. coli (Arnone and Nasini, 1986). In the present study, this
compound showed moderate activity when tested for cytotoxicity toward L5178Y cell line.
Curvularin (32) and dehydrocurvularin (33) exhibited antifungal and antibacterial
activity as well as non-specific phytotoxicity (Robeson and Strobel, 1981). More
interestingly, curvularin (32) showed remarkable cytotoxic activity towards sea urchin
embryogenesis, blocking cell division at concentrations of 2.5 µg/mL by specifically
disordering microtubule centers and inducing barrel-shaped spindles (Kobayashi et al., 1988).
The results obtained by testing 32 and 33 for cytotoxic activity toward L5178Y (mouse
lymphoma) cell line (see Table 3.33) were in accordance with the data reported in literature.
Dehydrocurvularin (33) was found to be highly active with an EC50 value of 0.43 µg/mL, and
curvularin (32) was also very active having an EC50 value of 4.7 µg/mL. However, both
compounds were inactive in the protein kinase inhibition assay, which also strongly points to
a mechanism of cytotoxic activity not involving interactions with protein kinases.

4.4.4. Compounds isolated from the endophytic fungus Chaetomium sp.


Chaetomium sp. was isolated from fresh stems of Otanthus maritimus growing wild on
the sandy Mediterranean coast in Egypt. The genus Otanthus, found mainly in the
Mediterranean region, belongs to family Asteraceae and is represented by a single species.

215
Discussion

Otanthus maritimus has been reported in the past to exhibit a significant array of biological
and pharmacological activities including the treatment of dysentery and inflammation of the
urinary bladder (Muselli et al., 2007). Dry specimens of O. maritimus have been traditionally
used as decoration and at the same time as a means of repelling flying insects from household
areas (Christodoulopoulou et al., 2005). Moreover, it was reported to be used by the Bedouins
for treating asthmatic bronchitis (Jakupovic et al.,1988).
The genus Chaetomium is a member of the subphylum Ascomycotina, family
Chaetomiaceae. Members of this family are cellulolytic and occur naturally on paper and
cotton fabrics (Alexopoulous et al., 1996). Chaetomium species are reported to be widespread
in soil and plant debris, where they are important agents of cellulose degradation (Abbott et
al., 1995, Carlile et al., 2001). As pathogens of crop plants, timber and ornamental trees, they
received comprehensive attention with regard to the production of mycotoxins (Alexopoulous
et al., 1996).
The EtOAc extract of Chaetomium sp. liquid cultures afforded the previously
unreported aureonitolic acid (34), as well as the known compounds cochliodinol (35),
isocochliodinol (36), indole-3-carboxylic acid (37), cyclo(alanyltryptophane) (38) and
orsellinic acid (39).

4.4.4.1. Biosynthesis of tetrahydrofurans


The previously unreported tetrahydrofuran aureonitolic acid (34) is structurally very
similar to the fungal metabolite aureonitol (34a), isolated for the first time in 1967 from the
culture broth of Chaetomium coarctatum (Abraham and Arfmann, 1992). Thus, both
compounds are presumably biosynthesized as illustrated in Figure 4.15. The epoxide
intermediate a, comprising seven acetate units, is rearranged to the aldehyde b which is then
reduced to the alcohol c. C is then epoxidized again to the intermediate d which is
subsequently opened intramolecularly by the hydroxyl group to 34a (Seto et al., 1979,
Abraham and Arfmann, 1992). This biosynthetic pathway was established by feeding
experiments using 13C-labelled precursors (Seto et al., 1979).

216
Discussion

H
O
COOH
*CH3 COOH * * * * *
* *
H a
Rearrangement
OH O

H [H] H

* *
c b
Epoxide
formation
OH
H O H
H H

d O
H [O] H OH Aureonitol (34a)

HO

OH
Aureonitolic acid (34)
O

Figure 4.15: Proposed biosynthesis of aureonitol and aureonitolic acid (Seto et al., 1979,
Abraham and Arfmann, 1992).

4.4.4.2. Biosynthesis of bis-(3-indolyl)-benzoquinones


The purple pigment cochliodinol (35) and related compounds were found to be
common metabolic products of the genus Chaetomium (Sekita et al., 1981). As suggested by
the chemical structure (see Figure 4.16), cochliodinol (35) and isocochliodinol (36) are
presumed to be biosynthesized from tryptophane and and isopentenyl unit derived from
mevalonic acid. To confirm the participation of tryptophane and mevalonate, administration
13 14
experiments were attempted using C- and C-precursors. Very surprisingly, the results
indicated that tryptophane was also incorporated into the oxygenated carbon atoms of the
benzoquinone ring of cochliodinol (Yamamoto et al., 1976; Taylor and Walter, 1978).

HN HN
O O

OH OH

HO HO

O O
NH NH

Cochliodinol (35) Isocochliodinol (36)

Figure 4.16: Structures of bis-(3-indolyl)-benzoquinones.

217
Discussion

4.4.4.3. Bioactivity of selected Chaetomium metabolites


Cochliodinol (35) and related compounds are produced by several Chaetomium
species. It was found that these quinonoid metabolites inhibit the growth and metabolism of a
range of bacterial genera (Brewer et al., 1984). In this study cochliodinol (35) and
isocochliodinol (36) were tested for cytotoxic activity against L5178Y (mouse lymphoma)
cell line (see Table 3.43). Interestingly, 35 was found to be very active with an EC50 value of
7.0 µg/mL, while 36 was only weakly active inhibiting L5178Y growth to 75.8 % at a
concentration of 10.0 µg/mL. Thus, it may be concluded that cytotoxic activity was affected
by the position of prenyl substituents at the indole rings.
Furthermore, results of brine shrimp lethality test showed that chain elongation
(increase in lipophilicity) caused a rise in the cytotoxic activity of orsellinates. In addition, the
reduction of activity upon substitution at 4-OH suggested that the hydroxy group at the C4
position causes effect in the cytotoxic activity of these compounds (Gomes et al., 2006). In
our study orsellinic acid (36) was found to be very active in the cytotoxic test against L5178Y
(mouse lymphoma) cell line (see Table 3.43). It inhibited L5178Y growth to only 1.0 % at a
concentration of 10.0 µg/mL, with an EC50 value of 2.7 µg/mL. On the other hand, orsellinic
acid was inactive in the protein kinase inhibition assay, indicating that the mechanism of
cytotoxic activity was most probably not due to interaction with protein kinases.

4.5. Detection of fungal metabolites in the host plant fractions


The fungal metabolite alternariol monomethyl ether (3) was isolated for the first time
from a plant source, Anthocleista djalonensis, in 1995 (Onocha et al., 1995). This plant is of
West African origin and is used in traditional medicine for the treatment of various diseases
(Onocha et al., 1995). Furthermore, 2,5-dimethyl-7-hydroxychromone (13) has been isolated
from Polygonum cuspidatum (Kimura et al., 1983), Hypericum perforatum (Yin et al., 2004)
and Rhei rhizoma (Kashiwada, Nonaka and Nishioka, 1984). Aureonitol (34a), a fungal
metabolite isolated from Chaetomium species, was isolated from an extract of Helichrysum
aureo-nitens (Bohlmann and Ziesche, 1979). These reports of isolation of fungal metabolites
from higher plants by circumstantial evidence evoked our interest to see if the metabolites we
isolated from the fungal species, investigated in this study, were detectable in fractions of the
corresponding host plants.
The fungal metabolites were traced in the host plant subfractions using LC/MS, an
analytical technique that provides high sensitivity and specificity even for very complex
extracts or fractions. Moreover, to increase the specificity of the method co-elution studies

218
Discussion

with the corresponding pure metabolites and the respective plant fractions were carried out
and spectra were evaluated for matching of retention times, presence of the molecular ion of
the target compounds, patterns of MS and MS/MS spectra of the pure substances and the
substances detected in the host plant fractions. Compared with LC/UV the LC/MS method
was found to be approximately twenty-five times more sensitive, with the lower limit of
quantification twenty-five times lower than that of LC/UV (10 ng/mL) for equivalent sample
volumes (Baldrey et al., 2002). Another difference between the two methods was that the
LC/UV method needs a cleaner extract as it is less specific and any endogenous compounds
with a similar retention time and similar maximum of absorption would interfere (Baldrey et
al., 2002). This illustrated the fact that LC/MS is in many aspects superior to LC/UV, and
thus, for the current study where sensitivity is an issue and limited amounts of substances or
complex fractions were to be investigated, would be the method of choice.
As a result, LC/MS analysis showed the presence of the Alternaria metabolites
alternariol (1), alternariol monomethyl ether (3) and altenusin (6) in the fractions of the host
plant, Polygonum senegalense. The Ampelomyces metabolites macrosporin (21) and
methylalaternin (23) were also detected in the fractions of Urospermum picroides. Similarly,
the Stemphylium metabolites macrosporin (21), tetrahydroaltersolanol B (30),
stemphyperylenol (31) and curvularin (32) were detected in Chenopodium album.
Interestingly, the substances detected were found in both liquid and rice cultures of the
endophytic fungus, while the substances obtained from only one of both cultures were not
detected in the host plant fractions. These results suggest the possible production of such
metabolites by the endophytic fungus under its normal physiological conditions of growth
within the tissues of the healthy plants, implying their possible contribution to the mutualistic
interaction between the endophyte and its host plant and proving the contribution of the
fungal endophyte to the chemical composition of the host plant. In the case of secondary
metabolites of the endophytic fungus Chaetomium sp., it was not possible to detect any of the
isolated secondary metabolites in any of the fractions of Otanthus maritimus, even though all
of them were detectable in the crude extract of the fungus. Thus, it could be concluded that
the fungal metabolites were either not produced in planta or present in very minute quantities
beyond the limit of detection of the very sensitive technique applied.
This evidence for presence of the fungal metabolites in the corresponding host plant
was quite surprising and encourages a quantification study which would be of great
significance. It is worth mentioning that, apart from a few studies more or less by chance
reporting the isolation of typical fungal metabolites from plant sources, the presence of

219
Discussion

secondary metabolites of endophytic fungi in the same host plants from which they had
originally been isolated has rarely been investigated. The reason could also be the use of less
sensitive methods of detection (e.g. LC/UV) resulting in lack of positive evidence. Recently,
it was demonstrated that Neotyphodium uncinatum, the common endophyte of Festuca
pratensis, had the full biosynthetic capacity for some of the most common loline alkaloids,
which were formerly exclusively found in endophyte-infected grasses. The identity of the
alkaloids was confirmed by GC/MS and 13C-NMR spectroscopic analysis (Blankenship et al.,
2001). Intensive studies of grass-endophyte associations, however, showed that endophytes in
the grasses produce physiologically active alkaloids in the tissues of their host, which cause
toxicosis to grazing livestock, increase resistance to invertebrate herbivores and pathogenic
microorganisms, and may inhibit germination and growth of other grasses. Experiments
demonstrated that plant growth and seed production can be increased by infection as well.
Ecologically, certain loline analog alkaloids have been demonstrated to contribute to the
allelopathic properties of host grasses (Clay, 1988; Joost, 1995; Siegel and Bush, 1997; Tan
and Zou, 2001). Moreover, endophyte-infected grasses usually possess an increased tolerance
to drought and aluminium toxicity (Malinowski and Belesky, 2000).
Thus, the present study has proved, for the first time, that the postulated, and hitherto
only for grass-endophyte associations proven hypothesis, that endophyte infection enhances
host plant fitness and competitiveness in stressful environments by producing functional
metabolites, could be also the case in other plant-endophyte associations, supported by the
unequivocal detection of fungal metabolites in three out of four investigated host plants
indicating that this could actually be a general case. It may also be hypothesized that fungal
metabolites reported previously from other plants presumably also originate from endophytic
fungi colonizing these plants. This finding supplies an important contribution to the question
of the ecological function of secondary metabolites produced by endophytic fungi, which
could lead to a better understanding of this interesting group of organisms as well as help in
the specific search for new bioactive substances with pharmaceutical potential to assist in
solving not only human, but also animal and plant health problems.

220
Conclusion

5. Conclusion

Plant endophytic fungi produce natural products with a large diversity of chemical
structures which might prove to be suitable for specific medicinal or agrochemical
applications. Most of these secondary metabolites show biological activities in
pharmaceutically relevant bioassay systems and thus represent potential lead structures which
could be optimized to yield effective therapeutic and bioactive agents.
The aim of this work was the isolation of secondary metabolites from endophytic
fungi, followed by structure elucidation and examination of their pharmocological potential.
Four endophytic fungal strains (Alternaria sp., Ampelomyces sp., Stemphylium botryosum and
Chaetomium sp.), obtained from Egyptian medicinal plants, were selected as biological
sources. The fungi were grown in liquid Wickerham medium as well as in solid rice medium
for a period of three to four weeks. The extracts obtained were then subjected to different
chromatographic separation techniques in order to isolate the secondary metabolites.
Structure elucidation of secondary metabolites was performed using state-of-the-art
analytical techniques, including mass spectrometry (MS) and nuclear magnetic resonance
(NMR) experiments. In addition, in the case of selected optically active natural products,
chiral derivatisation methods were applied in order to determine their absolute configuration.
Finally, the isolated compounds were subjected to various bioassays to examine their anti-
microbial, antifungal and cytotoxic activities as well as inhibitory profiles towards selected
protein kinases and their potential suppression of Staphylococcus epidermidis biofilm
formation.

1. Alternaria sp.
Three new alternariol derivatives were obtained from Alternaria sp., isolated from
Polygonum senegalense. Moreover, four new compounds including desmethylaltenusin, 4`-
epialtenuene, alterlactone and alternaric acid were isolated. The alternariol derivatives and
some of the structurally related compounds showed high cytotoxic activity when tested
against L5178Y mouse lymphoma cell line as well as pronounced protein kinase inhibitory
activity

2. Ampelomyces sp.
Ampelomyces sp. was isolated from Urospermum picroides. Six new compounds were
obtained from this fungal strain, including a new pyrone, two new isocoumarines, two new

221
Conclusion

sulphated anthraquinones and a new hexahydroanthronol. Desmethyldiaportinol, altersolanol


A and methylalaternin showed cytotoxic activity against L5178Y cells. Moreover,
altersolanol A and methylalaternin inhibited S. epidermidis biofilm formation.

3. Stemphylium botryosum
The fungal strain Stemphylium botryosum was isolated from Chenopodium album.
From this fungus curvularin derivatives, showing high cytotoxic activity against L5178Y,
were isolated.

4. Chaetomium sp.
Finally, the fungal strain Chaetomium sp., isolated from Otanthus maritimus, was
investigated. A new tetrahydrofuran derivative as well as known cochliodinol derivatives and
orsellinic acid were obtained from extracts of this fungus. The cochliodinol derivatives
inhibited various protein kinases tested in the bioassay. Furthermore, cochliodinol and
orsellinic acid showed high cytotoxic activity when tested against L5178Y lymphoma cell
line.

A total of forty-two compounds were isolated in this study, fourteen of which were
identified as new natural products. Both known and new compounds were tested for their
biological activities using different bioassay systems.
Furthermore, the fungal metabolites were traced in the corresponding host plant
subfractions using LC/MS. None of secondary metabolites of the endophytic fungus
Chaetomium sp. was found in any of the fractions of O. maritimus. On the other hand, major
compounds of the remaining fungal extracts could unequivocally be detected in fractions of
the respective host plants P. senegalense, U. picroides and C. album.

222
Conclusion

Table 5.1: Summary of the isolated compounds


Compound name Structure Source Comment
O
Alternariol Alternaria sp. Known
O OH

HO

OH
O
Alternariol-5-O-sulphate Alternaria sp. New
O OH

HO

OSO3H
O
Alternariol-5-O-methyl ether Alternaria sp. Known
O OH

HO

OCH3
O
Alternariol-5-O-methylether-4`-O- Alternaria sp. New
O OH
sulphate

HO3SO

OCH3
O
3`-Hydroxyalternariol-5-O- Alternaria sp. New
HO O OH
methylether

HO

O
O
Altenusin Alternaria sp. Known
HO HO OH

HO

O
O
Desmethylaltenusin Alternaria sp. New
HO HO OH

HO

OH
OH O
Alterlactone Alternaria sp. New
O

OH
HO

223
Conclusion

Compound name Structure Source Comment


OH
Talaroflavone O
Alternaria sp. Known
O

O
(R) (S)
OH

O
O
Alternaric acid Alternaria sp. New
HO OH

(R)
HO
O O

O
Altenuene Alternaria sp. Known
O OH

HO
H

H O
OH
O
4`-Epialtenuene New
O OH

H
HO

H O
OH

2,5-Dimethyl-7-hydroxychromone HO O Alternaria sp. Known

O
OH O
Altertoxin I Alternaria sp. Known

OH
H OH

OH O
O
Tenuazonic acid Alternaria sp. Known
HO

O
N
H

OH
Methyltriacetic lactone Ampelomyces Known
sp.

O O

224
Conclusion

Compound name Structure Source Comment


O
Ampelopyrone Ampelomyces New
sp.
O

O O

(R)
HO

Desmethyldiaportinol HO Ampelomyces New


OH
sp.
O OH

OH O
Cl
Desmethyldichlorodiaportin Ampelomyces New
HO
Cl
sp.
O OH

OH O
HO
Citreoisocoumarin Ampelomyces Known
O OH O sp.
OH O
O OH
Macrosporin Ampelomyces Known
HO
sp. and
Stemphylium
O
botryosum
O
O OH
Macrosporin sulphate Ampelomyces New
HO3SO
sp.

O
OH O OH
3-O-Methylalaternin Ampelomyces Known
HO
sp.

O
OH O OSO3H
3-O-Methylalaternin sulphate Ampelomyces New
HO
sp.

O
O OH
Altersolanol A Ampelomyces Known
O
sp. and
OH
Stemphylium
OH botryosum
OH O OH
OH OH
Ampelanol H Ampelomyces New
O
sp.
OH

OH
H
OH O OH

225
Conclusion

Compound name Structure Source Comment


OH O OH

Alterporriol D OH
Ampelomyces Known
OH sp.
O

O OH

O OH

OH

OH

OH O OH
OH O OH

Alterporriol E (atropsiomer of OH
Ampelomyces Known
alterporriol D) OH sp.
O

O OH

O OH

OH

OH

OH O OH
O
Altersolanol J H Ampelomyces Known
O
sp.
OH

OH
H
OH OH
OH
Tetrahydroaltersolanol B H
Stemphylium Known
O botryosum
OH

OH
H
OH O
OH O
Stemphyperylenol Stemphylium Known
botryosum
OH
H H
HO

O OH
O O
Curvularin Stemphylium Known
HO
botryosum

OH O
O O
Dehydrocurvularin Stemphylium Known
HO
botryosum

OH O

Aureonitolic acid O
Chaetomium sp. New

HO

OH

Cochliodinol HN
1
2
O
Chaetomium sp. Known
7a
7
OH
3
3a
6

4
5
HO
9 8
O
NH
10
11
12

226
Conclusion

Compound name Structure Source Comment


Isocochliodinol HN 2
Chaetomium sp. Known
1 O
7a
7
OH
3
6 3a

11 8
4
5
9 HO
10

O
12
NH

Indole-3-carboxylic acid COOH Chaetomium sp. Known

N
H
O
Cyclo(alanyltryptophane) H
N
Chaetomium sp. Known

N
H
O

N
H
O
Orsellinic acid Chaetomium sp. Known

OH

OH

227
References

6. References

Abbott, S. P., Sigler, L., McAleer, R., McGough, D. A., Rinaldi, M. G, and Mizell, G. (1995).
Fatal cerebral mycoses caused by the ascomycete Chaetomium strumarium. J. Clin.
Microbiol., 33(10), 2692-2698.
Abraham, W. R., and Arfmann, H. A. (1992). Rearranged tetrahydrofurans from Chaetomium
cochlioides. Phytochemistry, 31(7), 2405-2408.
Acker, T. E., Brenneisen, P. E., and Tanenbaum, S. W. (1966). Isolation, structure, and
radiochemical synthesis of 3,6-dimethyl-4-hydroxy-2-pyrone. J. Am. Chem. Soc., 88(4),
834-837.
Aldrich library of 13C and 1H FT NMR spectra (1992). Sigma-Aldrich, 3, 138A.
Alexopoulous, C. J., Mims, C. W., and Blackwell, M. (1996). Introductory mycology, 4th ed.,
John Wiley & Sons, Inc., New York, US, p. 182, 363.
Amna, T., Puri, S. C., Verma, V., Sharma, J. P., Khajuria, R. K., Musarrat, J., Spiteller, M.,
and Qazi, G. N. (2006). Bioreactor studies on the endophytic fungus Entrophospora
infrequens for the production of an anticancer alkaloid camptothecin. Can. J.
Microbiol., 52(3), 189-196.
Arai, K., Rawlings, B. J., Yoshizawa, Y., and Vederas, J. C. (1989). Biosyntheses of
antibiotic A26771B by Penicillium turbatum and dehydrocurvularin by Alternaria
cinerariae: Comparison of stereochemistry of polyketide and fatty acid enoyl thiol ester
reductases. J. Am. Chem. Soc., 111(9), 3391-3399.
Arnone, A., Nasini, G., Merlini L., and Assante G. (1986). Secondary mould metabolites. Part
16. Stemphyltoxins, new reduced perylenequinone metabolites from Stemphylium
botryosum var. Lactucum. J. Chem. Soc. Perkin Trans. I, 525-530.
Ayer, W. A., and Racok, J. S. (1990a). The metabolites of Talaromyces flavus: Part 1.
Metabolites of the organic extracts. Can. J. Chem., 68(11), 2085-2094.
Ayer, W. A., and Racok, J. S. (1990b). The metabolites of Talaromyces flavus: Part 2.
Biological activity and biosynthetic studies. Can. J. Chem., 68(11), 2095-2101.
Bacon, C. W., and White, J. F. (2000). Microbial endophytes, Marcel Dekker, Inc., New
York, pp. 4-5.
Baldrey, S. F., Brodie, R. R., Morris, G. R., Jenkins, E. H., and Brookes, S. T. (2002).
Comparison of LC-UV and LC-MS-MS for the determination of taxol. Chromatographia,
55(Supp. 1), 187-192.

228
References

Barron, D., Varin, L., Ibrahim, R. K., Harborne, J. B., and Williams, C. A. (1988). Sulphated
flavonoids – an update. Phytochemistry, 27(8), 2375-2395.
Bauer, A. W., Kirby, W. M. M., Sherriss, J. C., and Turck, M. (1966). Antibiotic
susceptibility testing by a standardized single disc method. Amer. J. Clin. Pathol., 45(4),
493-496.
Becker, A. M., Rickards, R. W., Schmalzl, K. J., and Yick, H. C. (1978). Metabolites of
Dactylaria lutea. J. Antibiot., 31(4), 324-329.
Bentley, R. (2000). Mycophenolic acid: A one hundred year odyssey from antibiotic to
immunosuppressant. Chem. Rev., 100(10), 3801-3826.
Bills, G. F. (1995). Analyses of microfungal diversity from a user's perspective. Can. J. Bot.,
73(Suppl.1), 33-41.
Birch, A. J., Musgrave, O. C., Rickards, R. W., and Smith, H. (1959). Studies in relation to
biosynthesis. Part XX. The structure and biosynthesis of curvularin. J. Chem. Soc., 4,
3146-3152.
Birkinshaw, J. H., and Gowlland, A. (1962). Studies in the biochemistry of micro-organisms.
110. Production and biosynthesis of orsellinic acid by Penicillium madriti G. Smith.
Biochem. J., 84(2), 342-347.
Bisby, G. R. (1943). Geographical distribution of fungi. The Botanical Review, 9, 466-482.
Blankenship, J. D., Spiering, M. J., Wilkinson, H. H., Fannin, F. F., Bush, L. P., and Schardl,
C. L. (2001). Production of loline alkaloids by the grass endophyte, Neotyphodium
uncinatum, in defined media. Phytochemistry, 58(3), 395-401.
Bohlmann, F., and Ziesche, J. (1979). Ein ungewöhnliches Tetrahydrofuran-Derivat aus
Helichrysum aureo-nitens. Phytochemistry, 18(4), 664-665.
Bokros, C. L., Hugdahl, J. D., Hanesworth, V. R., Murthy, J. V., and Morejohn, L. C. (1993).
Characterization of the reversible taxol-induced polymerization of plant tubulin into
microtubules. Biochemistry, 32(13), 3437-3447.
Borel, J. F., and Kis, Z. L. (1991). The discovery and development of cyclosporine.
Transplant. Proc., 23, 1867-1874.
Bradburn, N., Coker, R. D., Blunden, G., Turner, C. H., and Crabb, T. A. (1994). 5`-
Epialtenuene and neoaltenuene,dibenzo-α-pyrones from Alternaria lternate cultured on
rice. Phytochemistry, 35(3), 665-669.
Brady, S. F., and Clardy, J. (2000). CR377, a new pentaketide antifungal agent isolated from
an endophytic fungus. J. Nat. Prod., 63(10), 1447-1448.

229
References

Brady, S. F., Bondi, S. M., and Clardy, J. (2001). The guanacastepenes: A highly diverse
family of secondary metabolites produced by an endophytic fungus. J. Am. Chem. Soc.,
123(40), 9900-9901.
Brewer, D., Jen, W. C., Jones, G. A., and Taylor, A. (1984). The antibacterial activity of some
naturally occurring 2,5-dihydroxy-1,4-benzoquinones. Can. J. Microbiol., 30(8), 1068-
1072.
Butler, M. S. (2004). The role of natural product chemistry in drug discovery. J. Nat. Prod.,
67(12), 2141-2153.
Calhoun, L. A., Findlay, J. A., Miller, J. D., and Whitney, N. J. (1992). Metabolites toxic to
spruce budworm from balsam fir needle endophytes. Mycol. Res., 96, 281-286.
Camara, M. P. S., O’Neill, N. R., and Van Berkum, P. (2002). Phylogeny of Stemphylium spp.
based on ITS and glyceraldehyde-3-phosphate dehydrogenase gene sequences.
Mycologia, 94(4), 660-672.
Cannall, R. J. P. (1998). Methods in Biotechnology, Vol. 4: Natural products isolation,
Humana Press Inc., Totowa, NJ, pp.1.
Carlile, M. J., Watkinson, S. C., and Gooday, G. W. (2001). The fungi, 2nd ed., Academic
press, London, UK, p. 53.
Carmichael, J., DeGraff, W. G., Gazdar, A. F., Minna, J. D., and Mitchell, J. B. (1987).
Evaluation of a tetrazolium-based semiautomated colorimetric assay: Assessment of
radiosensitivity. Cancer Res., 47(4), 943-946.
Carroll, G. C. (1988). Fungal endophytes in stems and leaves: From latent pathogen to
mutualistic symbiont. Ecology, 69(1), 2-9.
Casabuono, A. C., and Pomilio, A. B. (1997). Alkaloids from endophyte-infected Festuca
argentina. J. Ethnopharmacol., 57(1), 1-9.
Cerniglia, C. E., Freeman, J. P., and Mitchum, R. K. (1982). Glucuronide and sulfate
conjugation in the fungal metabolism of aromatic hydrocarbons. Appl. Environ.
Microbiol., 43(5), 1070-1075.
Chem. Eng. News (2000). Natural product yields small molecule that mimics insulin. 77, 9-10.
Christodoulopoulou, L., Tsoukatou, M., Tziveleka, L. A., Vagias, C., Prtrakis, P. V., and
Roussis, V. (2005). Piperidinyl amides with insecticidal activity from the maritime plant
Otanthus maritimus. J. Agric. Food Chem., 53(), 1435-1439.
Clay, K. (1988). Fungal endophytes of grasses: A defensive mutualism between plants and
fungi. Ecology, 69(1), 10-16.

230
References

Clay, K. (1993). Fungal endophytes of plants: Biological and chemical diversity. Nat. Toxins,
1(3), 147-149.
Coombe, R. G., Jacobs, J. J., and Watson, T. R. (1968). Constituents of some Curvularia
species. Aust. J. Chem., 21(3), 783-788.
Coombe, R. G., Jacobs, J. J., and Watson, T. R. (1970). Metabolites of some Alternaria
species. The structures of altenusin and dehydroaltenusin. Aust. J. Chem., 23(11), 2343-
2351.
Costerton, J. W., Stewart, P. S., and Greenberg, E. P. (1999). Bacterial biofilms: A common
cause of persistent infections. Science, 284(5418), 1318-1322.
Dale, J. A., and Mosher, H. S. (1973). Nuclear magnetic resonance enantiomer reagents.
Configurational correlations via nuclear magnetic resonance chemical shifts of
diastereomeric mandelate, O-methylmandelate, and α-methoxy-α-
trifluoromethylphenylacetate (MTPA) esters. J. Am. Chem. Soc., 95(2), 512-519.
Davis, V. M., and Stack, M. E. (1994). Evaluation of alternariol and alternariol methyl ether
for mutagenic activity in Salmonella typhimurium. Appl. Environ. Microbiol., 60(10),
3901-3902.
DeBusk, L.M., Chen, Y., Nishishita, T., Chen, J., Thomas, J.W., and Lin, P.C. (2003). Tie2
receptor tyrosine kinase, a major mediator of tumor necrosis factor alpha-induced
angiogenesis in rheumatoid arthritis. Arthritis Rheum., 48(9), 2461-2471.
Dehm, S., Senger M. A., and Bonham, K. (2001). SRC transcriptional activation in a subset of
human colon cancer cell lines. FEBS Letters, 487(3), 367-371.
Demain, A. L. (2000). Microbial natural products: A past with a future. In: Biodiversity: New
leads for pharmaceutical and agrochemical industries. Wrigley, S. K., Hayes, M. A.,
Thomas, R., Chrystal, E. J. T., and Nicholson, N. (ed.), The Royal Society of Chemistry,
Cambridge, UK, p. 3-16.
Dew, R. K., Boissonneault, G. A., Gay, N., Boling, J. A., Cross, R. J., and Cohen, D. A.
(1990). The effect of the endophyte (Acremonium coenophialum) and associated toxin(s)
of tall fescue on serum titer response to immunization and spleen cell flow cytometry
analysis and response to mitogens. Vet. Immunol. Immunopathol., 26(3), 285-295.
Dewick, P. M. (2006). Medicinal natural products. A biosynthetic approach. John Wiley &
Sons Ltd, Baffins Lane, Chichester, England.
Dossaji, S., and Kubo, I. (1980). Quercetin 3-(2``-galloylglucoside), a molluscicidal flavonoid
from Polygonum senegalense. Phytochemistry, 19(3), 482.

231
References

Ebel, R. (2006). Secondary metabolites from marine-derived fungi. In: Frontiers in Marine
Biotechnology, (Proksch, P., Müller, W. E. G., eds.). Horizon Scientific Press, Norwich,
p. 73-143.
Evans, G. E., and Staunton, J. (1988). An investigation of the biosynthesis of citromycetin in
13 14
Penicillium frequentans using C- and C-labelled precursors. J. Chem. Soc. Perkin
Trans. I, 755-761.
Faeth, S. H., and Fagan, W. F. (2002). Fungal endophytes: Common host plant symbionts but
uncommon mutualists. Integr. Comp. Biol., 42(2), 360-368.
Fehr, M. J., Consiglio, G., Scalone, M., and Schmid, R. (1999). Asymmetric hydrogenation of
substituted 2-pyrones. J. Org. Chem., 64(16), 5768-5776.
Freeman, G. G. (1966). Isolation of alternariol and alternariol monomethyl ether from
Alternaria dauci (Kühn) Groves and Skolko. Phytochemistry, 5(4), 719-725.
Freeman, S., and Rodriguez, R. J. (1993). Genetic conversion of a fungal plant pathogen to a
nonpathogenic, endophytic mutualist. Science, 260(5104), 75-78.
Garces, C. A., Kurenova, E. V., Golubovska, V. M., and Cance, W. G. (2006). Vascular
endothelial growth factor receptor-3 and focal adhesion kinase bind and suppress
apoptosis in breast cancer cells. Cancer Research, 66, 1446-1454.
Ghisalberti, E. L., Hockless, D. C. R., Rowland, C. Y., and White, A. H. (1993). Structural
study of curvularin, a cell division inhibitor. Aust. J. Chem., 46(4), 571-575.
Glombitza, K.-W., and Knöss, W. (1992). Sulphated phlorotannins from the brown alga
Pleurophycus gardneri. Phytochemistry, 31(1), 279-281.
Gomes, A. T., Honda, N. K., Roese, F. M., Muzzi, R. M., and Sauer, L. Z. (2006). Cytotoxic
activity of orsellinates. Z. Naturforsch. C: Biosciences, 61(9/10), 653-657.
Gribble, G. W. (1998). Naturally occurring organohalogen compounds. Acc. Chem. Res.,
31(3), 141-152.
Griffin, G. F., and Chu, F. S. (1983). Toxicity of the Alternaria metabolites alternariol,
alternariol methyl ether, altenuene, and tenuazonic acid in the chicken embryo assay.
Appl. Environ. Microbiol., 46(6), 1420-1422.
Grove, J. F. (1971). Metabolic products of Stemphylium radicinum. Part IV. Minor products.
J. Chem. Soc. C, 2261-2263.
Grove, J. F., MacMillan, J., Mulholland, T. P. C., and Rogers, M. A. T. (1952). Griseofulvin.
Part IV. Structure. J. Chem. Soc., 3977-3987.

232
References

Guo, B., Dai, J.-R., Ng, S., Huang, Y., Leong, C., Ong, W., and Karte, B. K. (2000). Cytonic
acids A and B: Novel tridepside inhibitors of hCMV protease from the endophytic fungus
Cytonaema species. J. Nat. Prod., 63(5), 602-604.
Hall-Stoodley, L., Costerton, J. W., Stoodley, P. (2004). Bacterial biofilms: From the natural
environment to infectious diseases. Nature Rev. Microbiology, 2, 95-108.
Haraguchi, H., Abo, T., Hashimoto, K., and Yagi, A. (1992). Action-mode of antimicrobial
altersolanol A in Pseudomonas aeruginosa. Biosci. Biotech. Biochem., 56(8), 1221-1224.
Harper, J. K., Arif, A. M., Ford, E. J., Strobel, G. A., Porco, J. A., Tomer, Jr. D. P., Oneill, K.
L., Heider, E. M., and Grant, D. M. (2003). Pestacin: A 1,3-dihydro isobenzofuran from
Pestalotiopsis microspora possessing antioxidant and antimycotic activities. Tetrahedron,
59(14), 2471-2476.
Hellwig, V., Grothe, T., Mayer-Bartschmid, A., Endermann, R., Geschke, F. U., Henkel, T.,
and Stadler, M. (2002). Altersetin, a new antibiotic from cultures of endophytic
Alternaria spp. Taxonomy, fermentation, isolation, structure elucidation and biological
activities. J. Antibiot., 55(10), 881-892.
Hiort, J. (2002). Neue Naturstoffe aus Schwamm-assoziierten Pilzen des Mittelmeeres.
Isolierung, Strukturaufklärung und biologische Aktivität. Heinrich-Heine-Universität.
Hofer, M. D., Fecko, A., Shen, R., Setlur, S. R., Pienta, K. G., Tomlins, S. A., Chinnaiya, A.
M., and Rubin, M. A. (2004). Expression of the platelet-derived growth factor receptor in
prostate cancer and treatment implications with tyrosine kinase inhibitors. Neoplasia,
6(5), 503-512.
Hohaus, K., Altmann, A., Burd, W., Fischer, I., Hammer, P. E., Hill, D. S., Ligon, J. M., and
van Pée, K.-H. (1997). NADH-abhängige Halogenasen sind wahrscheinlich eher an der
Biosynthese von Halogenmetaboliten beteiligt als Haloperoxidasen. Angew. Chem.,
109(18), 2102-2104.
Höller, U., Gloer, J. B., and Wicklow, D. T. (2002). Biologically active polyketide
metabolites from an undetermined fungicolous hyphomycete resembling Cladosporium.
J. Nat. Prod., 65(6), 876-882.
Höller, U., König, G. M., and Wright, A. D. (1999). A new tyrosine kinase inhibitor from a
marine isolate of Ulocladium botrytis and new metabolites from the marine fungi
Asteromyces cruciatus and Varicosporina ramulosa. Eur. J. Org. Chem., (11), 2949-
2955.

233
References

Horn, W. S., Simmonds, M. S. J., Schwartz, R. E., and Blaney, W. M. (1995).


Phomopsichalasin, a novel antimicrobial agent from an endophytic Phomopsis sp.
Tetrahedron, 51(14), 3969-3978.
Hosoe, T., Nozawa, K., Udagawa, S.-I., Nakajima, S., and Kawai, K.-I. (1990). An
anthraquinone derivative from Dichotomophthora lutea. Phytochemistry, 29(3), 997-999.
Hradil, C. M., Hallock, Y. F., Clardy, J., Kenfield, D. S., and Strobel, G. (1989). Phytotoxins
from Alternaria cassiae. Phytochemistry, 28(1), 73-75.
Hussey, C., Orsi, B. A., Scott, J., and Spencer, B. (1965). Mechanism of choline sulphate
utilization in fungi. Nature, 207(4997), 632-634.
Iseki, H., Ko, T. C., Xue, X. Y., Seapan, A., and Townsend, C. M. Jr. (1998). A novel strategy
for inhibiting growth of human pancreatic cancer cells by blocking cyclin dependent
kinase activity. J. Gastrointest. Surg., 2(1), 36-43.
Ishii, T., Hayashi, K., Hida, T., Yamamoto, Y., and Nozaki, Y. (2000). TAN-1813, a novel
ras-farnesyltransferase inhibitor produced by Phoma sp.
Taxonomy, fermentation, isolation and biological activities in vitro and in vivo. J.
Antibiot., 53(8), 765-778.
Izeradjene, K., Douglas, L., Delaney, A., and Houghton, J. A. (2004). Influence of casein
kinase II in tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in
human rhabdomyosarcoma cells. Clinical Cancer Research, 10, 6650-6660.
Jakupovic, J., Boeker, R., Grenz M., Paredes, L., Bohlmann, F., and Seif El-Din, A. (1988).
Highly oxygenated guaianolides from Otanthus maritimus. Phytochemistry, 27(4), 1135-
1140.
Jerram, W. A., McInnes, A. G., Maass, W. S. G., Smith, D. G., Taylor, A., and Walter, J. A.
(1975). The chemistry of cochliodinol, a metabolite of Chaetomium spp. Can. J. Chem.,
53(5), 727-737.
Joost, R. E. (1995). Acremonium in fescue and ryegrass: Boon or bane? A review. J. Anim.
Sci., 73, 881-888.
Kaczka, E. A., Gitterman, C. O., Dulaney, E. L., Smith, M. C., Hendlin, D., Woodruff, H. B.,
and Folkers, K. (1964). Discovery of inhibitory activity of tenuazonic acid for growth of
human adenocarcinoma-1. Biochem. Biophys. Res. Commun., 14(1), 54-57.
Kalli, K. R., Falowo, O. I., Bale, L. K., Zschunke, M. A., Roche, P. C., and Conover, C. A.
(2002). Functional insulin receptors on human epithelial ovarian carcinoma cells:
Implications for IGF-II mitogenic signalling. Endocrinology, 143(9), 3259-3267.

234
References

Kashiwada, Y., Nonaka, G.-I., and Nishioka, I. (1984). Studies on rhubarb (Rhei hizome).
V. Isolation and characterization of chromone and chromanone derivatives. Chem.
Pharm. Bull., 32(9), 3493-3500.
Keen, N., and Taylor, S. (2004). Aurora-kinase inhibitors as anticancer agents. Nature Rev.
Cancers, 4, 927-936.
Kelloff, G. J., Lubet, R. A., Fay, J. R., Steele, V. E., Boone, C. W., Crowell, J. A., and
Sigman, C. C. (1997). Farnesyl protein transferase inhibitors as potential cancer
chemopreventives. Cancer Epidem. Biom. Prev., 6(4), 267-282.
Kimura, Y., Kozawa, M., Baba, K., and Hata, K. (1983). New constitutents of roots of
Polygonum cuspidatum. Planta Med., 48(7), 164-168.
Kimura, Y., Mizuno, T., Nakajima, H., and Hamasaki, T. (1992). Altechromones A and B,
new plant growth regulators produced by the fungus, Alternaria sp. Biosci. Biotech.
Biochem., 56(10), 1664-1665.
Kiss, L., Russell, J. C., Szentivanyi, O., Xu, X., and Jeffries, P. (2004). Biology and
biocontrol potential of Ampelomyces mycoparasites, natural antagonists of powdery
mildew fungi. Biocontrol Science and Technology, 14(7), 635-651.
Knight, V., Sanglier, J. J., DiTullio, D., Braccili, S., Bonner, P., Waters, J., Hughes, D., and
Zhang, L. (2003). Diversifying microbial natural products for drug discovery. Appl.
Microbiol. Biotechnol., 62(5-6), 446-458.
Knoch, T. R., Faeth, S. H., and Arnott, D. L. (1993). Endophytic fungi alter foraging and
dispersal by desert seed-harvesting ants. Oecologia, 95(4), 470–475.
Kobayashi, A., Hino, T., Yata, S., Itoh, T. J., Sato, H., and Kawazu, K. (1988). Unique
spindle poisons, curvularin and its derivatives, isolated from Penicillium species. Agric.
Biol. Chem., 52(12), 3119-3123.
Kosemura, S. (2003). Meroterpenoids from Penicillium citreo-viride B. IFO 4692 and 6200
hybrid. Tetrahedron, 59(27), 5055-5072.
Koshino, H., Togiya, S., Terada, S.-I., Yoshihara, T., Sakamura, S., Shimanuki, T., Sato, T.,
and Tajimi, A. (1989). New fungitoxic sesquiterpenoids, chokols A-G, from stromata of
Epichloe typhina and the absolute configuration of chokol E. Agric. Biol. Chem., 53(3),
789-796.
Kusakai, G., Suzuki, A., Ogura, T., Miyamoto, S., Ochiai, A., Kaminishi, M., and Esumi, H.
(2004). ARK5 expression in colorectal cancer and its implications for tumor progression.
Am. J. Pathol., 164, 987-995.

235
References

Lai, S., Shizuri, Y., Yamamura, S., Kawai, K., and Furukawa, H. (1991). Three new phenolic
metabolites from Penicillium species. Heterocycles, 32(2), 297-305.
Lai, S., Shizuri, Y., Yamamura, S., Kawai, K., Terada, Y., and Furukawa, H. (1989). Novel
curvularin-type metabolites of a hybrid strain ME 0005 derived from Penicillium citreo-
viride B. IFO 6200 and 4692. Tetrahedron letters, 30(17), 2241-2244.
Larsen, T. O., Smedsgaard, J., Nielsen, K. F., Hansen, M. E., and Frisvad, J. C. (2005).
Phenotypic taxonomy and metabolite profiling in microbial drug discovery. Nat. Prod.
Rep., 22(6), 672-695.
Laszlo, H., and Henshaw, P. S. (1954). Plant materials used by primitive peoples to affect
fertility. Science, 119, 626-630.
Lazarovits, G., Steele, R. W., and Stoessl, A. (1988). Dimers of altersolanol A from
Alternaria solani. Z. Naturforsch. Sect. C, 43c(11-12), 813-817.
Lee, H. J., Choi, J. S., Jung, J. H., and Kang, S. S. (1998). Alaternin glucoside isomer from
Cassia tora. Phytochemistry, 49(5), 1403-1404.
Lee, J. W., Soung, Y. H., Kim, S. Y., Park, W. S., Nam, S. W., Kim, S. H., Lee, J. Y., Yoo, N.
J., and Lee, S. H. (2005). ERBB2 kinase domain mutation in a gastric cancer metastasis.
APMIS, 113, 683-687.
Lee, J., Lobkovsky, E., Pliam, N. B., Strobel, G., and Clardy, J. (1995a). Subglutinols A and
B: Immunosuppressive compounds from the endophytic fungus - Fusarium subglutinans.
J. Org. Chem., 60(22), 7076-7077.
Lehmann, L., Wagner, J., and Metzler, M. (2006). Estrogenic and clastogenic potential of the
mycotoxin alternariol in cultured mammalian cells. Food Chem. Toxicol., 44(3), 398-408.
Li, D., Zhu, J., Firozi, P., Abbruzzesse, J. L., Evans, D. B., Cleary, K., Fries, H., and Sen, S.
(2003). Overexpression of oncogenic STK15/BTAK/Aurora A kinase in human
pancreatic cancer. Clinical Cancer Research, 9, 991-997.
Li, J. Y. and Strobel, G. A. (2001). Jesterone and hydroxyjesterone antioomycete
cyclohexenenone epoxides from the endophytic fungus - Pestalotiopsis jesteri.
Phytochemistry, 57(2), 261-265.
Li, J. Y., Harper, J. K., Grant, D. M., Tombe, B. O., Bashyal, B., Hess, W. M., and Strobel, G.
A. (2001). Ambuic acid, a highly functionalized cyclohexenone with antifungal activity
from Pestalotiopsis spp. and Monochaetia sp. Phytochemistry, 56(5), 463-468.
Li, J. Y., Sidhu, R. S., Ford, E., Hess, W. M., and Strobel, G. A. (1998). The induction of
taxol production in the endophytic fungus – Periconia sp. from Torreya grandifolia. J.
Ind. Microbiol. Biotechnol., 20(5), 259-264.

236
References

Li, J. Y., Strobel, G. A., Harper, J. K., Lobkovsky, E., and Clardy, J. (2000). Cryptocin, a
potent tetramic acid antimycotic from the endophytic fungus Cryptosporiopsis cf.
quercina. Org. Lett., 2(6), 767-770.
Li, J. Y., Strobel, G. A., Sidhu, R., Hess, W. M., and Ford, E. (1996). Endophytic taxol
producing fungi from bald cypress Taxodium distichum. Microbiology, 142(8), 2223-
2226.
Lingham, R. B., Silverman, K. C., Bills, G. F., Cascales, C., Sanchez, M., Jankins, R. G.,
Gartner, S. E., Martin, I., Diez, M. T., Pelaez, F., Mochales, S., Kong, Y.-L., Burg, R. W.,
Meinz, M. S., Huang, L., Nallin-Omstead, M., Mosser, S. D., Schaber, M. D., Omer, C.
A., Pompliano, D. L., Gibbs, J. B., and Singh, S. B. (1993). Chaetomella acutiseta
produces chaetomellic acids A and B which are reversible inhibitors of farnesyl-protien
transferase. Appl. Microbiol. Biotechnol., 40(2-3), 370-374.
Liu, Y., Li, Z., and Vederas, J. C. (1998). Biosynthetic incorporation of advanced precursors
into dehydrocurvularin, a polyketide phytotoxin from Alternaria cinerariae. Tetrahedron,
54(52), 15937-15958.
Lutzoni, F., Kauff, F., Cox, C. J., McLaughlin, D., Celio, G., Dentinger, B., Padamsee, M.,
Hibbett, D., James, T. J., Baloch, E., Grube, M., Reeb, V., Hofstetter, V., Schoch, C.,
Arnold, A. E., Miadlikowska, J., Spatafora, J., Johnson, D., Hambleton, S., Crockett, M.,
Shoemaker, R., Sung, G.-H., Lücking, R., Lumbsch, T., O’Donnell, K., Binder, M.,
Diederich, P., Ertz, D., Gueidan, C., Hansen, K., Harris, R. C., Hosaka, K., Lim, Y.-W.,
Matheny, B., Nishida, H., Pfister, D., Rogers, J., Rossman, A., Schmitt, I., Sipman, H.,
Stone, J., Sugiyama, J., Yahr, R., and Vilgalys, R. (2004). Assembling the fungal tree of
life: Progress, classification, and evolution of subcellular traits. American Journal of
Botany, 91(10), 1446–1480.
Ma, Y. M., Li, Y., Liu, J. Y., Song, Y. C., and Tan, R. X. (2004). Anti-Helicobacter pylori
metabolites from Rhizoctonia sp. Cy064, an endophytic fungus in Cynodon dactylon.
Fitoterapia, 75(5), 451-456.
Maass, W. S. G. (1975). Lichen substances VII. Identification of orsellinate derivatives from
Lobaria linita. The Bryologist, 78(2), 178-182.
Macmillan, J. C., Hudson J. W., Bull, S., Dennis, J. W., and Swallow, C. J. (2001).
Comparative expression of the mitotic regulators SAK and PLK in colorectal cancer.
Annals of Surgical Oncology, 8, 729-740.

237
References

Malinowski, D. P., and Belesky, D. P. (2000). Adaptation of endophyte-infected cool-season


grasses to environmental stresses: Mechanisms of drought and mineral stress tolerance.
Crop sci., 40(4), 923-940.
Marchelli, R., Dossena, A., and Casnati, G. (1975). Biosynthesis of neoechinulin by
Aspergillus amstelodami from cyclo-L-[U-14C]alanyl-L[5,7-3H2]tryptophyl. J. Chem.
Soc., Chem. Commun., 779-780.
McPhail, A., Miller, R. W., Harvan, D., and Pero, R. W. (1973). X-ray crystal structure
revision of the fungal metabolite (+/-)-altenuene. J. Chem. Soc., Chem. Commun., 682.
Menezes, D. E. L., Peng, J., Garrett, E. N., Louie, S. G., Lee, S. H., Wiesmann, M., Tang, Y.,
Shephard, L., Goldbeck, C., Oei, Y., Ye, H., Aukerman, S. L., and Heise, C. (2005).
CHIR-258: A potent inhibitor of FLT3 kinase in experimental tumor xenograft models of
human acute myelogenous leukemia. Clinical Cancer Research, 11, 5281-5291.
Meronuck, R. A., Steele, J. A., Mirocha, C. J., and Christensen, C. M. (1972). Tenuazonic
acid, a toxin produced by Alternaria lternate. Appl. Microbiol., 23(3), 613-617.
Metz, A., Haddad, A., Worapong, J., Long, D., Ford, E., Hess, W. M., and Strobel, G. A.
(2000). Induction of the sexual stage of Pestalotiopsis microspora, a taxol producing
fungus. Microbiology, 146(8), 2079-2089.
Miller, F. A. , Rightsel, W. A. , Sloan, B. J. , Ehrlich, J., French, J. C., Bartz, Q. R., and
Dixon, G. J. (1963). Antiviral activity of tenuazonic acid. Nature, 200(4913), 1338.
Morejohn, L. C., and Fosket, D. E. (1984). Taxol-induced rose microtubule polymerization in
vitro and its inhibition by colchicines. J. Cell Biol., 99(1), 141-147.
Munro, H. D., Musgrave, O. C., and Templeton, R. (1967). Curvularin. Part V. The
compound C16H18O5, αβ-Dehydrocurvularin. J. Chem. Soc. C, 947-948.
Muselli, A., Rossi, P.-G., Desjobert, J.-M., Bernardini, A.-F., Berti, L., Costa, J. (2007).
Chemical composition and antibacterial activity of Otanthus maritimus (L.) Hoffmanns.
and Link essential oils from Corsica. Flav. Frag. J., 22(3), 217-223.
Nakanishi, S., Toki, S., Saitoh, Y., Tsukuda, E., Kawahara, K., Ando, K., and Matsuda, Y.
(1995). Isolation of myosin light chain kinase inhibitors from microorganisms:
Dehydroaltenusin, altenusin, atrovenetinone, and cyclooctasulfur. Biosci. Biotech.
Biochem., 59(7), 1333-1335.
National Cancer Institute. (22 November 2005). Mutations in gliboblastoma in multiforme
predict responsed to target therapies. NCI Cancer Bulletin (online), 2(45).
Newman, D. J., Cragg, G. M., and Hyde, K. D. (2003). Natural products as sources of new
drugs over the period 1981-2002. J. Nat. Prod., 66(7), 1022-1037.

238
References

Newton, G. G. F., and Abraham, E. P. (1955). Cephalosporin C, a new antibiotic containing


sulphur and D-α-aminoadipic acid. Nature, 175(4456), 548.
Nielsen, K. F, Larsen T. O., and Frisvad, J. C. (2004). Lightweight expanded clay aggregates
(LECA), a new up-scaleable matrix for production of microfungal metabolites. J.
Antibiot., 57(1), 29-36.
Nolte, M. J., Steyn, P. S., and Wessels, P. L. (1980). Structural investigation of 3-
acylpyrrolidine-2,4-diones by nuclear magnetic resonance spectroscopy and x-ray
crystallography. J. Chem. Soc. Perkin Trans. I, 1057-1065.
Ohnishi, K., Suemitsu, R., Kubota, M., Matano, H., and Yamada, Y. (1991). Biosynthesis of
alterporriol D and E by Alternaria porri. Phytochemistry, 30(8), 2593-2595.
Ohnishi, K., Tanabe, H., Hayashi, S., and Suemitsu, R. (1992). Biosynthesis of alterporriol A
by Alternaria porri. Biosci. Biotech. Biochem., 56(1), 42-43.
Ohtani, I., Kusumi, T., Kashman, Y., and Kakisawa, H. (1991). High-field FT NMR
application of Mosher’s method. The absolute configurations of marine terpenoids. J. Am.
Chem. Soc., 113(11), 4092-4096.
Okamura, N., Haraguchi, H., Hashimoto, K., and Yagi, A. (1993). Altersolanol-related
antimicrobial compounds from a strain of Alternaria solani. Phytochemistry, 34(4), 1005-
1009.
Okamura, N., Mimura, K., Haraguchi, H., Shingu, K., Miyahara, K., and Yagi, A. (1996).
Altersolanol-related compounds from the culture liquid of Alternaria solani.
Phytochemistry, 42(1), 77-80.
Okuno, T., Natsume, I., Sawai, K., Sawamura, K., Furusaki, A., and Matsumoto, T. (1983).
Structure of antifungal and phytotoxic pigments produced by Alternaria sps. Tetrahedron
Lett., 24(50), 5653-5656.
Okuno, T., Natsume, I., Sawai, K., Sawamura, K., Furusaki, A., and Matsumoto, T. (1983).
Structure of antifungal and phytotoxic pigments produced by Alternaria sps. Tetrahedron
Lett., 24(50), 5653-5656.
Onocha, P. A., Okorie, D. A., Connolly, J. D., and Roycroft, D. S. (1995). Monoterpene diol,
iridoid glucoside and dibenzo-α-pyrone from Anthocleista djalonensis. Phytochemistry,
40(4), 1183-1189.
Ouyang, B., Knauf, J. A., Smith, E. P., Zhang, L., Ramsey, T., Yusuff, N., Batt, D., and
Fagin, J. A. (2006). Inhibitors of Raf kinase activity block growth of thyroid cancer cells
with RET/PTC or BRAF mutations in vitro and in vivo. Clinical Cancer Research, 12,
1785-1793.

239
References

Oyama, M., Xu, Z., Lee, K.-H., Spitzer, T. D., Kitrinos, P., McDonald, O. B., Jones, R. R.
J., Garvey, E. P. (2004). Fungal metabolites as potent protein kinase inhibitors:
Identification of a novel metabolite and novel activities of known metabolites. Letters in
Drug Design and Discovery, 1(1), 24-29.
Plasencia, J., and Mirocha, C. J. (1991). Isolation and characterization of zearalenone sulfate
produced by Fusarium spp. Appl. Environ. Microbiol., 57(1), 146-150.
Plasencia, J., and Mirocha, C. J. (1991). Isolation and characterization of zearalenone sulfate
produced by Fusarium spp. Phytochemistry, 57(1), 146-150.
Qiao, H., Hung, W., Tremblay, E., Wojcik, J., Gui, J., Ho, J., Klassen, J., Campling, B., and
Elliot, B. (2002). Constitutive activation of met kinase in non-small-cell lung carcinomas
correlates with anchorage-independent cell survival. J. Cell Biochem., 86(4), 665-677.
Ragan, M. A. (1978). Phenol sulfate esters: ultraviolet, infrared, 1H and 13C nuclear magnetic
resonance spectroscopic investigation. Can. J. Chem., 56(20), 2681-2685.
Raistrick, H., Stickings, C. E., and Thomas, R. (1953). Studies in the biochemistry of micro-
organisms. 90. Alternariol and alternariol monomethyl ether, metabolic products of
Alternaria tenuis. Biochem. J., 55(3), 421-433.
Rasmussen, T. B., Skinderso, M. E., Bjarnsholt, T., Phipps, R. K., Christensen, K. B.,
Andersen, J. B., Koch, B., Larsen, T. O., Hentzer, M., Hoiby, N., and Givskov, M.
(2005). Identity and effects of quorum-sensing inhibitors produced by Penicillium
species. Microbiology, 151(5), 1325-1340.
Ratnayake, A. S., Yoshida, W. Y., Mooberry, S. L., and Hemscheidt, T. (2001). The structure
of microcarpalide, a microfilament disrupting agent from an endophytic fungus. Org.
Lett., 3(22), 3479-3481.
Robeson, D. J., and Strobel, G. A. (1981). αβ-Dehydrocurvularin and curvularin from
Alternaria cinerariae. Z. Naturforsch., 36c, 1081-1083.
Robeson, D. J., and Strobel, G. A. (1985). The identification of a major phytotoxic component
from Alternaria macrospora as αβ-Dehydrocurvularin. J. Nat. Prod., 48(1), 139-141.
Rosett, T., Sankhala, R. H., Stickings, C. E., Taylor, M. E. U., and Thomas, R. (1957).
Studies in the biochemistry of micro-organisms. 103. Metabolites of Alternaria tenuis
Auct.: Culture filtrate products. Biochem. J., 67(3), 390-400.
Royles, B. J. L. (1995). Naturally occurring tetramic acids: Structure, isolation, and synthesis.
Chem. Rev., 95(6), 1981-2001.
Rudgers, J. A., Koslow, J. M., and Clay, K. (2004). Endophytic fungi alter relationships
between diversity and ecosystem properties. Ecology Letters, 7(1), 42-51.

240
References

Saikkonen, K., Faeth, S. H., Helander, M., and Sullivan, T. J. (1998). Fungal endophytes: A
continuum of interactions with host plants. Annu. Rev. Ecol. Syst., 29, 319-343.
Samant, S. S., and Pant, S. (2006). Diversity, Distribution Pattern and Conservation Status of
the Plants Used in Liver Diseases/Ailmentsin Indian Himalayan Region. Journal of
Mountain Science, 3(1), 28-47.
Savard, M. E., Miller, J. D., Blais, L. A., Seifert, K. A., and Samson, R. A. (1994). Secondary
metabolites of Penicillium bilaii strain PB-50. Mycopathologia, 127(1), 19-27.
Schardl, C. L., Liu, J.-S., White, J. F., Finkel, R. A., An, Z., and Siegel, M. R. (1991).
Molecular phylogenetic relationships of nonpathogenic grass mycosymbionts and
clavicipitaceous plant pathogens. Plant Syst. Evol., 178(1-2), 27-41.
Schiff, P. B., and Horowitz, S. B. (1980). Taxol stabilizes microtubules in mouse fibroblast
cells. Proc. Natl. Acad. Sci. USA, 77, 1561-1565.
Schmitz, K. J., Grabellus, F., Callies, R., Otterbach, F., Wohlschlaeger, J., Levkau, B.,
Kimmig, R., Schmid, K. W., and Baba, H. A. (2005). High expression of focal adhesion
kinase (p125FAK) in node-negative breast cancer is related to overexpression of HER-
2/neu and activated Akt kinase but does not predict outcome. Breast Cancer Res., 7(2),
R194-R203.
Schulz, B., and Boyle, C. (2005). The endophytic continuum. Mycological research, 109(6),
661-686.
Schulz, B., Boyle, C., Draeger, S., Römmert, A.-K., and Krohn, K. (2002). Endophytic fungi:
a source of novel biologically active secondary metabolites Mycol. Res., 106(9), 996-
1004.
Schulz, B., Boyle, C., Draeger, S., Römmert, A.-K., and Krohn, K. (2002). Endophytic fungi:
a source of novel biologically active secondary metabolites. Mycol. Res., 106(9), 996-
1004.
Schulz, B., Römmert, A.-K., Dammann, U., Aust, H.-J., and Strack, D. (1999). The
endophyte-host interaction: a balanced antagonism? Mycol. Res., 103(10), 1275-1283.
Sekita, S. (1983). Isocochliodinol and neocochliodinol, bis (3-indolyl)-benzoquinones from
Chaetomium spp. Chem. Pharm. Bull., 31(9), 2998-3001.
Sekita, S., Yoshihira, K., Natori, S., Udagawa, S., Muroi, T., Sugiyama, Y., Kurata, H., and
Umeda, M. (1981). Mycotoxin production by Chaetomium spp. and related fungi. Can. J.
Microbiol., 27(8), 766-772.

241
References

13
Seto, H., Saito, M., Uzawa, J., and Yonehara, H. (1979). Utilization of C-13C coupling in
structural and biosynthetic studies. XII1 Biosynthesis of 2-(but-1,3-dienyl)-3-hydro-4-
(penta-1,3-dienyl)-tetrahydrofuran, a metabolite of Chaetomium coarctatum.
Heterocycles, 13(1), 247-253.
Shen, L., Ye, Y.-H., Wang, X.-T., Zhu, H.-L., Xu, C., Song, Y.-C., Li, H., Tan, R.-X. (2006).
Structure and total synthesis of aspernigerin: A novel cytotoxic endophyte metabolite.
Chemistry - A European Journal, 12(16), 4393-4396.
Shu, R. G., Wang, F. W., Yang, Y. M., Liu, Y. X., and Tan, R. X. (2004). Antibacterial and
xanthine oxidase inhibitory cerebrosides from Fusarium sp. IFB-121, and endophytic
fungus in Quercus variabilis. Lipids, 39(7), 667-673.
Siegel, M. R., and Bush, L. P. (1996). Defensive chemicals in grass-fungal endophyte
associations. Recent Adv. Phytochem., 30, 81-118.
Siegel, M. R., and Bush, L. P. (1997). Toxin production in grass/endophyte associations.
Chapt. 10. In: The Mycota, Part V, Plant Relationships, (Carroll, G. C., and Tudzynski,
P., eds.). Springer-Verlag, Berlin.
Singh, S. B., Zink, D. L., Guan, Z., Collado, J., Pelaez, F., Felock, P. J., and Hazuda, D. J.
(2003). Isolation, structure, and HIV-1 integrase inhibitory activity of xanthoviridicatin E
and F, two novel fungal metabolites produced by Penicillium chrysogenum. Helv. Chim.
Acta, 86(10), 3380-3385.
Smedsgaard, J., and Frisvad, J. C. (1996). Using direct electrospray mass spectrometry in
taxonomy and secondary metabolite profiling of crude fungal extracts. J. Microbiol.
Methods, 25(1), 5-17.
Smolarz, H. D. (2002). Flavonoids from Polygonum lapathifolium ssp. Tomentosum. Pharm.
Biol., 40(5), 390-394.
Song, Y. C., Li, H., Ye, Y. H., Shan, C. Y., Yang, Y. M., and Tan, R. X. (2004). Endophytic
naphthopyrone metabolites are co-inhibitors of xanthine oxidase, SW1116 cell and some
microbial growths. FEMS Microbiol. Lett., 241(1), 67-72.
Sourvinos, G., Tsatsanis, C., and Spandidos, D. A. (1999). Overexpression of the Tpl-2/Cot
oncogene in human breast cancer. Oncogene, 18, 4968-4973.
Staal, S. P. (1987). Molecular cloning of the akt oncogene and its human homologues AKT1
and AKT2: Amplification of AKT1 in a primary human gastric adenocarcinoma, Proc.
Natl. Acad. Sci. USA, 84(14), 5034-5037.

242
References

Stack, M. E., Mazzola, E. P., Page, S. W., Pohland, A. E., Highet, R. J., Tempesta, M. S., and
Corley, D. G. (1986). Mutagenic perylenequinone metabolites of Alternaria lternate:
Altertoxins I, II, and III. J. Nat. Prod., 49(5), 866-871.
Staunton, J., and Weissman, K. J. (2001). Polyketide biosynthesis: A millennium review. Nat.
Prod. Rep., 18(4), 380-416.
Steyn, P. S., and Rabie, C. J. (1976). Characterization of magnesium and calcium tenuazonate
from Phoma sorghina. Phytochemistry, 15(12), 1977-1979.
Stierle, A., and Stobel, G. (1995). The search for a taxol-producing microorganism among the
endophytic fungi of the pacific yew, Taxus brevifolia. J. Nat. Prod., 58(9), 1315-1324.
Stierle, A., Stierle, D., and Bugni, T. (1999). Sequoiatones A and B: Novel antitumor
metabolites isolated from a redwood endophyte. J. Org. Chem., 64(15), 5479-5484.
Stierle, A., Strobel, G. A., and Stierle, D. (1993). Taxol and taxane production by Taxomyces
andreanae, an endophytic fungus of Pacific yew. Science, 260(5105), 214-216.
Stinson, E. E., Wise, W. B., Moreau, R. A., Jurewicz, A. J., and Pfeffer, P. E. (1986).
Alternariol: Evidence for biosynthesis via norlichexanthone. Can. J. Chem., 64(8), 1590-
1594.
Stoessl, A. (1969a). Relative stereochemistry of altersolanol A. Can. J. Chem., 47(5), 777-
784.
Stoessl, A. (1969b). Some metabolites of Alternaria solani. Can. J. Chem., 47(5), 767-776.
Stoessl, A., and Stothers, J. B. (1983). Tetrahydroaltersolanol B, a hexahydroanthronol from
Alternaria solani. Can. J. Chem., 61(2), 378-382.
Stoessl, A., Unwin, C. H., and Stothers, J. B. (1983). On the biosynthesis of some polyketide
metabolites in Alternaria solani: 13C and 2Hmr studies. Can. J. Chem., 61(2), 372-377.
Strobel, G. A. (2002a). Microbial gifts from rain forests. Can. J. Plant Pathol., 24(1), 14-20.
Strobel, G. A. (2002b). Rainforest endophytes and bioactive products. Crit. Rev. Biotechnol.,
22(4), 315-333.
Strobel, G. A., Miller, R. V., Miller, C., Condron, M., Teplow, D. B., and Hess, W. M.
(1999). Cryptocandin, a potent antimycotic from the endophytic fungus Cryptosporiopsis
cf. quercina. Microbiology, 145(8), 1919–1926.
Strobel, G. A., Stierle, A., Stierle, D., and Hess, W. M. (1993). Taxomyces andreanae a
proposed new taxon for a bulbilliferous hypomycete associated with Pacific yew.
Mycotaxon, 47, 71-78.

243
References

Strobel, G. A., Yang, X., Sears, J., Kramer, R., Sidhu, R. S., and Hess, W. M. (1993). Taxol
from Pestalotiopsis microspora, an endophytic fungus of Taxus wallichiana.
Microbiology, 142, 435-440.
Strobel, G., and Daisy, B. (2003). Bioprospecting for microbial endophytes and their natural
products. Microbiol. Mol. Biol. Rev., 67(4), 491-502.
Strzelecka, M., Bzowska, M., Koziel, J., Szuba, B., Dubiel, O., Riviera, N. D., Heinrich, M.,
and Bereta, J. (2005). Anti-inflammatory effects of extracts from some traditional
Mediterranean diet plants. J. Physiol. Pharmacol., 56(1), 139-156.
Su, B.-N., Park, E. J., Mbwambo, Z. H., Santarsiero, B. D., Mesecar, A. D., Fong, H. H. S.,
Pezzuto, J. M., and Kinghorn, A. D. (2002). New chemical constituents of Euphorbia
quinquecostata and absolute configuration assignment by a convenient Mosher ester
procedure carried out in NMR tubes. J. Nat. Prod., 65(9), 1278-1282.
Suemitsu, R., Ohnishi, K., Yanagawase, S., Yamamoto, K., and Yamada, Y. (1989).
Biosynthesis of macrosporin by Alternaria porri. Phytochemistry, 28(6), 1621-1622.
Suemitsu, R., Sakurai, Y., Nakachi, K., Miyoshi, I., Kubota, M., and Ohnishi, K. (1989).
Alterporriol D and E, modified bianthraquinones from Alternaria porri (Ellis) Ciferri.
Agric. Biol. Chem., 53(5), 1301-1304.
Suemitsu, R., Sano, T., Yamamoto, M., Arimoto, Y., Morimatsu, F., and Nabeshima, T.
(1984). Structural elucidation of alterporriol B, a novel metabolic pigment produced by
Alternaria porri (Ellis) Ciferri. Agric. Biol. Chem., 48(10), 2611-2613.
Sundheim, L., and Krekling, T. (1982). Host-parasite relationships of the hyperparasite
Ampelomyces quisqualis and its powdery mildew host Sphaerotheca fuliginea. J.
Phytopath., 104, 202-210.
Tan, R. X., and Zou, W. X. (2001). Endophyts: A rich source of functional metabolites. Nat.
Prod. Rep., 18(4), 448-459.
Tanahashi, T., Kuroishi, M., Kuwahara, A., Nagakura, N., and Hamada, N. (1997). Four
phenolics from the cultured lichen mycobiont of Graphis scripta var. pulverulenta. Chem.
Pharm. Bull., 45(7), 1183-1185.
13
Taylor, A., and Walter, J. A. (1978). Determination of low levels of incorporation of C-
labelled precursors. Biosynthesis of the 2,5-dihydroxycyclohexadiene-1,4-dione system
of cochliodinol. Phytochemistry, 17(6), 1045-1048.
Tikoo, A, Shakri, R., Conolly, L., Hirokawa, Y., Shishido, T., Bowers, B., Ye, L., Kohama,
K., Simpson, R. J., and Maruta, H. (2000). Treatment of ras-induced cancers by the F-
actin-bundling drug MKT-077. Cancer J., 6(3), 162-168.

244
References

Umetsu, N., Kuji, J., and Tamari, K. (1972). Investigation on the toxin production by several
blast fungus strains and isolation of tenuazonic acid as a novel toxin. Agr. Biol. Chem.
Jap., 36, 859-866.
Vesely, D., Vesela, D., and Jelinek, R. (1995). Penicillium aurantiogriseum Dierckx produces
chaetoglobosin A toxic to embryonic chickens. Mycopathologia, 132(1), 31-33.
Vesonder, R. F., Burmeister, H. R., and Tjarks, L. W. (1990). 4,4,24-trimethylcholesta-
8,14,24(28)-trien-2alpha,3beta,11alpha,12beta-tetrol 12-acetate, 3-sulphate from
Fusarium graminearum. Phytochemistry, 29(7), 2263-2265.
Wagenaar, M., Corwin, J., Strobel, G. A., and Clardy, J. (2000). Three new chytochalasins
produced by an endophytic fungus in the genus Rhinocladiella. J. Nat. Prod., 63(12),
1692-1695.
Watanabe, A., Fujii, I., Sankawa, U., Mayoorga, M. E., Timberlake, W. E., and Ebizuka, Y.
(1999). Re-identification of Aspergillus nidulans wA gene to code for a polyketide
synthase of naphthopyrone. Tetrahedron Lett., 40(1), 91-94.
Watanabe, A., Ono, Y., Fujii, I., Sankawa, U., Mayoorga, M. E., Timberlake, W. E., and
Ebizuka, Y. (1998). Product identification of polyketide synthase coded by Aspergillus
nidulans wA gene. Tetrahedron Lett., 39(42), 7733-7736.
Weber, D., Sternere, O., Anke, T., Gorzalczancy, S., Martino, V., Acevedo, C. (2004).
Phomol, a new antiinflammatory metabolite from an endophyte of the medicinal plant
Erythrina crista-galli. J. Antibiot., 57(9), 559-563.
Weichert, W., Schmidt, M., Gekeler, V., Denkert, C., Stephan, C., Jung, K., Loening, S.,
Dietel, M., and Kristiansen, G. (2004). Polo-like kinase 1 is overexpressed in prostate
cancer and linked to higher tumor grades. Prostate, 60(3), 240-245.
Wheeler, M. M., and Wheeler, D. M. S. (1975). Anthraquinone pigments from the
phytopathogen Phomopsis juniperovora Hahn. Phytochemistry, 14(1), 288-289.
White, J. F., Morgan-Jones, G., and Morrow, A. C. (1993). Taxonomy, life cycle,
reproduction and detection of Acremonium endophytes. Agric. Ecosyst. Environ., 44(1-4),
13-37.
Williams, D. H., Stone, M. J., Hauck, P. R., and Rahman, S. K. (1989). Why are secondary
metabolites (natural products) biosynthesized? J. Nat. Prod., 52(6), 1189-1208.
Wolfender, J. L., Queiroz, E. F., and Hostettmann, K. (2005). Phytochemistry in the
microgram domain – a LC-NMR perspective. Magn. Reson. Chem., 43(9), 697-709.
Wolock-Madej, C., and Clay, K. (1991). Avian seed preference and weight loss experiment:
The role of fungal-infected fescue seeds. Oecologia, 88(2), 296-302.

245
References

Xia, G., Kumar, S. R., Masood, R., Zu, S., Reddy, R., Krasnaperov, V., Quinn, D. I., Henshal,
S. M., Sutherland, R. L., Pinski, J. K., Daneshmand, S., Buscarini, M., Stein, J. P., Zong,
C., Broek, D., Roy-Burman, P., and Gill, P. S. (2005). EphB4 expression and biological
significance in prostate. Cancer Res., 65(11), 4623-4632.
Yagi, A., Okamura, N., Haraguchi, H., Abo, T., and Hashimoto, K. (1993). Antimicrobial
tetrahydroanthraquinones from a strain of Alternaria solani. Phytochemistry, 33(1), 87-
91.
Yamamoto,Y., Nishimura, K., and Kiriyama, N. (1976). Studies on the metabolic products of
Aspergillus terreus. I. Metabolites of the strain IFO 6123. Chem. Pharm. Bull., 24(8),
1853-1859.
Yarwood, C. E. (1932). Ampelomyces quisqualis on clover mildew. Phytopathology, 22, 31.
Yin, Z.-Q., Wang, Y., Ye, W.-C., Zhao, S.-X. (2004). Chemical constituents of Hypericum
perforatum (St. John’s wort) growing in China. Biochem. Syst. Ecol., 32(5), 521-523.
Young, D. H., Michelotti, E. J., Sivendell, C. S., and Krauss, N. E. (1992). Antifungal
properties of taxol and various analogues. Experientia, 48(9), 882-885.
Yu, Q., Sicinska, E., Geng, Y., Ahnstrom, M., Zagozdzou, A., Kong, Y., Gardner, H.,
Kiyokawa, H., Harris, L. N., Stal, O., and Sicinski, P. (2006). Requirement for CDK4
kinase function in breast cancer. Cancer cell, 9(1), 23-32.
Zhang, H. W., Song, Y. C., and Tan, R. X. (2006). Biology and chemistry of endophytes. Nat.
Prod. Rep., 23(5), 753-771.
Zhang, X., and Yee, D. (2000). Tyrosine kinase signalling in breast cancer: Insulin-like
growth factors and their receptors in breast cancer. Breast Cancer Res., 2(3), 170-175.

246
Abbreviations

7. List of Abbreviations

[α]D specific rotation at the sodium D-line


br broad signal
CDCl3 deuterated chloroform
CHCl3 chloroform
CI chemical ionization
COSY correlation spectroscopy
d doublet
DCM dichloromethane
dd doublet of doublet
DEPT distortionless enhancement by polarization transfer
DMSO dimethyl sulfoxide
DNA Deoxyribonucleic acid
ED effective dose
EI electron impact ionization
ESI electrospray ionization
et al. et altera (and others)
EtOAc ethyl acetate
eV electronvolt
FAB fast atom bombardment
g gram
HMBC heteronuclear multiple bond connectivity
HMQC heteronuclear multiple quantum coherence
H2O water
HPLC high performance liquid chromatography
H3PO4 phosphoric acid
hr hour
HR-MS high resolution mass spectrometry
Hz Herz
IZ inhibition zone
L liter
LC liquid chromatography

247
Abbreviations

LC/MS liquid chromatography-mass spectrometery


m multiplet
M molar
MeOD deuterated methanol
MeOH methanol
mg milligram
MHz mega Herz
min minute
mL milliliter
mm millimeter
MS mass spectrometry
MTT microculture tetrazolium assay
m/z mass per charge
µg microgram
µL microliter
µM micromol
NaCl sodium chloride
ng nanogram
NMR nuclear magnetic resonance
NOE nuclear Overhauser effect
NOESY nuclear Overhauser and exchange spectroscopy
PCR polymerase chain reaction
ppm parts per million
q quartet
ROESY rotating frame overhauser enhancement spectroscopy
RP 18 reversed phase C 18
s singlet
t triplet
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC thin layer chromatography
UV ultra-violet
VLC vacuum liquid chromatography

248
Attachements

8. Attachments

Attachment 1: The 1H NMR spectrum of alternariol (1).

3614.21
3612.00

3337.09
3334.57
3291.06
3288.54

3173.47
3171.58

1957.81

1364.80
CH3

4
6 3`
5` X
Integral

0.8769

0.9130

0.8785

0.8457

0.7420

3.0000
8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5
(ppm)

Attachment 2: The 1H NMR spectrum of alternariol-5-O-sulphate (2).


7.8361
7.8323

6.8628
6.8584

6.6775
6.6731
6.5798
6.5741

2.7560

CH3

4
3`
6 5`
Integral

0.9596

0.9370

0.9744
0.9125

3.0000

7.8 7.6 7.4 7.2 7.0 6.8 6.6 6.4 6.2 6.0 5.8 5.6 5.4 5.2 5.0 4.8 4.6 4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2
(ppm)

249
Attachments

Attachment 3: The 1H NMR spectrum of alternariol-5-O-methylether (3).


3644.79
3642.90

3352.23
3349.70
3307.46
3304.94
3276.56
3274.67

1959.39

1383.08

653.56
649.15
639.69
458.73
453.06
451.16
445.49
440.44
438.55
433.51
430.36
426.57
OCH3
CH3

4
6 3`
5`
Integral

0.8313

0.8943

0.8002

0.8288

3.0000

2.5347

2.8158

1.7776
8.0 7.5 7.0 6.5 6.0 5.5 5.0 4.5 4.0 3.5 3.0 2.5 2.0 1.5 1.0
(ppm)

Attachment 4: The 1H NMR spectrum of alternariol-5-O-methylether-4`-O-sulphate (4).


7.3722
7.3671
7.2335
7.2284
7.1755
7.1704

6.6182
6.6132

3.9304

2.8335

OCH3

CH3

3` 4
6 5`
Integral

1.0035

0.9360
0.9222

0.9215

2.9982

3.0000

7.8 7.6 7.4 7.2 7.0 6.8 6.6 6.4 6.2 6.0 5.8 5.6 5.4 5.2 5.0 4.8 4.6 4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2
(ppm)

250
Integral

8.8
Integral

12.5

8.4
0.7549 6017.57

12.0

8.0
11.5
3-OH

7.6
11.0

7.2
0.9673 7.0658
0.1385

10.5

NH
0.2972 6.8433

6.8
0.0468

10.0

6.4
9.5

6.0
9.0

5.6
8.5

5.2
8.0

4.8
7.5
3655.92

(ppm)
(ppm)
1.0000 3654.03

4.4

251
7.0
0.0642 0.9342 3413.80
3.9462

4.0
0.3416
6 5`

3.9392 1.0403 3316.07


3.7772
4

0.1985

6.5
3.7703 3314.18

5
1.0000

3.6
6.0

3.2
2.4862 5.5
2.4383

Attachment 6: The 1H NMR spectrum of tenuazonic acid (15).


1.9700

2.8
1.9630
1.9567
1.9498
5.0

1.4667 1.9448
1.9372
2.7658

2.4
1.9290
1.3844

7
1.3762
4.5

1.3705
1.3585

2.0
1.9374 1.3491
1.3434
1.3346
4.0

3.0292 1998.58
1.3194
1.2816

1.6
1.2665
1.2507

9A
1.2438
3.5

1.9167

9B
1.2381
2.3469 1.2230

1.2
1.2085
OCH3

4.4038 1.2041
3.0

1.1921

11
1.7095
1.1770
4.5641 1.0036

0.8
0.9898
1365.84
0.8372
Attachment 5: The 1H NMR spectrum of 3`-hydroxyalternariol-5-O-methylether (5).

0.9652

10
2.5

0.9513
0.9362
0.8902

0.4
0.8750
CH3

0.8605
0.7773
0.7641
Attachements
Attachments

Attachment 7: The 1H NMR spectrum of altenusin (6).


6.5779
6.4764
6.4253
6.4203

6.1593
6.1543

3.7986

1.8981
OCH3

CH3

2`
5`
4 6
Integral

0.9660
0.9414
0.9635

0.9933

3.2435

3.0000
6.8 6.6 6.4 6.2 6.0 5.8 5.6 5.4 5.2 5.0 4.8 4.6 4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2 2.0 1.8 1.6 1.4 1.2
(ppm)

Attachment 8: The 1H NMR spectrum of desmethylaltenusin (7).


6.5584
6.4676

6.2570
6.2526

6.0421
6.0377

1.9063

CH3

2`
5`
4 6
Integral

1.0000
1.0038

1.1344

1.0444

2.9281

6.8 6.6 6.4 6.2 6.0 5.8 5.6 5.4 5.2 5.0 4.8 4.6 4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2 2.0 1.8 1.6 1.4 1.2
(ppm)

252
Integral 3325.74 Integral
3323.53
3317.86

10.5
3315.65

6.8
2.8670 3235.58 0.8214 5110.15

6,6*
3233.37

8-OH
0.9522 3229.27

10.0
1.1900 3227.06
3108.84

6.4
2.4880
3106.00

4,4*
2.8138 3088.35

9.5
4738.14
3085.82 1.8888
1.0000 4691.16

6`

6.0
9.0
2,3-OH
1.0000 3516.49

6`*
7.0

5.6
(ppm)

8.5
1.0010 3452.49

5.2
8.0
3253.24
1.0125 3251.03

6.50
(ppm)
2110.71
0.9736 3228.34

7.5
2108.51

4.8
2102.83 3226.13
2100.31
2038.52
2035.68 1.0000 3516.49

7.0
2032.53 1.0010 3452.49

4.4
2030.00
1 4

1.0451 1931.64 3253.24


9
1.0125

5`β*
1929.12 3251.03

6.5
2.8093 1897.59 0.9736

5`β
3228.34

(ppm)
1893.81

(ppm)

4.0
3226.13

253
7.5749
11

1891.92
2.9334 1888.45

6.0
3.1579 1884.67
1882.78
0.9366 1878.99 Integral

3.6
4`α
1875.84

5.5
1871.43 2423.15

4`β*
1867.33 1.7651
(ppm)
Attachment 9: The 1H NMR spectrum of alterlactone (8).

1863.23 2408.33
4.80

1858.82

3.2
1855.03 5.0
2423.15
5

1.7651
2408.33

2.8
4.5

3`α
4.0

2.8883 1211.26

2.4
1207.48
1.0922 1196.76 2.9053 1906.12
0.8704 1192.98
1136.86
3.5

3`β*
1133.08

2.0
2.9318 1124.88

3`α*
1121.10
OCH3

1089.89

3`β
3.0

1077.28
1064.98

CH3*

1.6
3.1587
995.94
8.2377 986.80
Attachment 10: The 1H NMR spectrum of altenuene (11) and 4`-epialtenuene (12*).
2.5

981.44
972.29
771.79

1.2
CH3
745.30
2.0
Attachements
Attachments

Attachment 11: The 1H NMR spectrum of talaroflavone (9).

5504.54

3220.45
3194.29
3044.54
3037.92
2955.63

2313.12
2307.45

1868.29

874.89
OCH3

3220.45

3194.29

3044.54
3037.92

2955.63

2313.12
2307.45
CH3
Integral

Integral

Integral

Integral
1.1212

1.0000

0.8444

0.9795

0.9895
6.45 6.40 6.35 6.10 5.90 4.60
(ppm) (ppm) (ppm) (ppm)

3` 4
6
5`
5`-OH
7-OH
Integral

0.7438

1.1212
1.0000

0.8444
0.9795

0.9895

3.0550

2.7769
12.0 11.6 11.2 10.8 10.4 10.0 9.6 9.2 8.8 8.4 8.0 7.6 7.2 6.8 6.4 6.0 5.6 5.2 4.8 4.4 4.0 3.6 3.2 2.8 2.4 2.0 1.6 1.2 0.8
(ppm)

Attachment 12: The 1H NMR spectrum of alternaric acid (10).


3206.57
3204.05

3040.11
3037.59

2197.41
2193.94
2191.11

1892.55

1513.92
1506.98
1496.58
1489.64

1257.61
1240.59

997.20
988.06

OCH3

CH3

6
4
4`
3`A 3`B
Integral

1.0000

0.9774

0.7141

2.9489

0.9807

1.0584

3.0906

6.8 6.6 6.4 6.2 6.0 5.8 5.6 5.4 5.2 5.0 4.8 4.6 4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2 2.0 1.8 1.6
(ppm)

254
Integral Integral

11.2

12.8
0.8903 6365.22

10.8
4-OH

12.4
0.8735 6161.55
0.8558 5292.06

9-OH

10.4

12.0
7-OH

11.6
10.0

11.2
9.6

10.8
9.2
Integral

10.4
8.8
6.64

10.0
3312.51

8.4

9.6
0.9818 3310.30

6.62

9.2
8.0
(ppm)
6.60
3298.32
1.0000

8.8
7.6
6.58

8.4
4032.57

7.2
4023.75

6
1.0300 4002.62
3993.80

8.0
1.0202

6.56

7
6.8
3312.51
3310.30
0.9818
8

7.6
3298.32
1.0000

(ppm)

(ppm)

255
6

6.4
3523.11

7.2
3514.28
1.0582 3474.24
1.0000 3465.41

6.0
0.8596 2982.43

6.8
8
3

6.4
5.6

Attachment 14: The 1H NMR spectrum of altertoxin I (14).

6.0
5.2

2689.55

5.6
2640.05
2273.09

12-OH
0.8570
4.8

2268.67

12b-OH
0.8915
2261.42

5.2
2253.85
2249.44
4.4

1876.80
1874.59

4.8
1868.29
1555.54
1.0809
1550.81

12
4.0

1541.04

4.4
1536.63
1533.48
1523.70

4.0
3.6

1519.29
1502.58
2.1885 1490.91

X
1486.82

3.6
1476.73
3.2

2ax 1468.53
1436.69
Attachment 13: The 1H NMR spectrum of 2,5-dimethyl-7-hydroxychromone (13).

1432.28
1eq,11ax,12a

3.2
1.6397 1420.93
1416.51
2.8

3.4451
1294.19
5-CH3
1.1099 1276.85 2.8139 1316.26

2.8
1164.93
11eq
2.0944 2eq 1161.15
2.4

1150.74
1146.96 2.7585 1128.36

2.4
0.8992
2-CH3
1ax 1136.56
1132.77
0.7226 1050.49
2.0

X
1027.16

2.0
Attachements
Attachments

Attachment 15: The 1H NMR spectrum of methyltriacetic lactone (16).

5.8801

2.0822

1.6932
3-CH3
6-CH3

5
Integral

0.9865

3.0014

3.0000
6.4 6.2 6.0 5.8 5.6 5.4 5.2 5.0 4.8 4.6 4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2 2.0 1.8 1.6 1.4 1.2
(ppm)

Attachment 16: The 1H NMR spectrum of ampelopyrone (17).


5.9740

5.0619
5.0493
5.0348
5.0222
5.0108
4.9982

4.0262

2.6665
2.6583
2.6444

1.9498

1.7147

1.2041
1.1914

11

3-CH3
2.6665
2.6583

2.6444
5.0619
5.0493
5.0348
5.0222
5.0108
4.9982

9
Integral

2.0355
1.1028

5 5.05
(ppm)
5.00 2.65
(ppm)

7
8
8-OH
Integral

1.0000

1.1028

0.8889

2.0355

3.3954

3.1757

3.2780

6.4 6.2 6.0 5.8 5.6 5.4 5.2 5.0 4.8 4.6 4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2 2.0 1.8 1.6 1.4 1.2
(ppm)

256
Integral
Integral

4
1.0000 6.4165

6.4
6.4
4
1.0000 6.3831
1.9305 6.3005
1.9438 6.2999

5,7

6.2
5,7

6.2
Integral

6.04
6.0251
0.9769 6.0181

6.0
6.0251

6.0
6.0181
0.9769

(ppm)

11

5.8
6.00
5.8

5.6
5.6

5.4
5.4

5.2
5.2

5.0
5.0

4.8
4.8
4.0224

4.6
4.6
4.0117
Integral 4.3080 4.0035

4.32
4.3010
4.00
1.0803 3.9940
4.2947

4.4
(ppm)
4.3080 4.2890 3.9858

4.4
4.3010 4.2821 3.9751
0.9321 4.2947 4.2758

10
0.9321 4.2890

4.2
(ppm)
4.2821 4.0224

4.2

4.28
4.2758 4.0117
4.0035
Integral 3.9940

(ppm)
4.0
(ppm)
3.9858

4.0
1.0803

257
3.9751

10
3.5496
1.9195 3.5389
3.54
(ppm)

3.8
3.8

3.6
3.6
3.5496
1.9195 3.5389
11

3.4
3.4

3.2
3.2
Integral

3.00

3.0
2.9867 2.9867 2.7680
3.0

1.0098

9A
2.9804 2.9804
2.7598
2.76

2.9571
2.9508
1.0887

(ppm)

2.8
1.0098
(ppm)

2.9571
2.8

2.7497 2.7383
9A
Attachment 17: The 1H NMR spectrum of desmethyldiaportinol (18).

0.9502 2.9508 2.7680

2.95
2.7308 1.0887 2.7302 2.7598
2.7201

9B
2.7383

X
2.72

2.7012

2.6
2.7302
2.6

2.5505
1.0400 2.5328
2.5505 2.5215

2.4
2.55

2.5039
Attachment 18: The 1H NMR spectrum of desmethyldichlorodiaportin (19).
2.7497
2.4

2.75
9B

2.5328
2.7308 1.0400
(ppm)

0.9502 2.5215

2.2
2.7201

(ppm)
2.2

2.5039
2.50

2.7012

2.70

2.0
2.0

1.8
1.8

1.6
1.6
Attachements
Integral

3169.06

6.4
1.0000 3123.97

4
3121.77
1.0147 3118.93
Integral 3116.72
0.9040

6.2
Attachments

6.250
3123.97
1.0147

5,7

6.0
3121.77

6.240
(ppm)
3118.93

5.8
0.9040
3116.72

6.230

5.6
5.4
5.2
5.0
4.8
4.6
Integral 2230.83
2223.89
2230.83 2217.90
1.0927 2210.97

4.45
2223.89 2204.98

4.4
2217.90

10
1.0927

(ppm)
2210.97

4.2
2204.98
4.40

(ppm)
4.0

258
3.8
3.6
3.4
3.2
2.70

1345.88

3.0
2.3363
1339.89
1335.48
1.1785 1326.02
2.65

2.8
1320.98
Attachment 19: The 1H NMR spectrum of (+)citreoisocoumarin (20).

(ppm)

2.3363 1345.88
11

1.1785 1304.90 1339.89


2.6

1335.48
2.60

1.1072 1297.33
1.1072 1326.02
1290.40 1320.98
1304.90
1282.83
9B
9A

1297.33
2.4

1290.40
1282.83
2.2

3.1813 1086.74
13

2.0
1.8
1.6
Attachements

Attachment 20: The 1H NMR spectrum of macrosporin (21).

3990.52

3862.84

3613.15
3610.62

3413.90
3411.38

2003.72

1172.06
OCH3
CH3

8
5
4 2
Integral

1.0000

1.0313

0.8876

0.8972

2.9259

2.9009
9.2 8.8 8.4 8.0 7.6 7.2 6.8 6.4 6.0 5.6 5.2 4.8 4.4 4.0 3.6 3.2 2.8 2.4 2.0 1.6
(ppm)

Attachment 21: The 1H NMR spectrum of macrosporin-7-O-sulphate (22).


4181.37

4032.57

3638.80
3636.28

3373.66
3371.14

1964.12

1230.50

OCH3

CH3

8
5 4 2
Integral

1.0000

0.9705

0.9626

0.9700

3.0936

3.0068

9.2 8.8 8.4 8.0 7.6 7.2 6.8 6.4 6.0 5.6 5.2 4.8 4.4 4.0 3.6 3.2 2.8 2.4 2.0
(ppm)

259
Attachments

Attachment 22: The 1H NMR spectrum of 3-O-methylalaternin (23).

3813.02

3626.39
3624.18

3425.88
3423.99

2010.34

1173.00
OCH3

CH3

4
2
5
Integral

1.0000

1.1047

1.0504

3.1079

3.4797
9.2 8.8 8.4 8.0 7.6 7.2 6.8 6.4 6.0 5.6 5.2 4.8 4.4 4.0 3.6 3.2 2.8 2.4 2.0 1.6
(ppm)

Attachment 23: The 1H NMR spectrum of 3-O-methylalaternin-7-O-sulphate (24).


3828.27

3660.55
3658.03

3391.00
3388.48

1972.63

1263.92

OCH3

CH3

5 4 2

X
Integral

0.4245

0.9644

0.9968

1.0000

2.9513

2.8334

9.2 8.8 8.4 8.0 7.6 7.2 6.8 6.4 6.0 5.6 5.2 4.8 4.4 4.0 3.6 3.2 2.8 2.4 2.0
(ppm)

260
Integral Integral

13.0
12.4
1.0000 0.9563 6060.67
6288.29
5-OH
Integral

12.0

12.5
5-OH
2342.44
2336.77

11.6
0.9750

12.0
2332.35

(ppm)
2326.68

4.65
11.2

11.5
10.8
Integral

11.0
3.85
10.4
1915.89
3.8543 1908.01

10.5
3.80
1898.24

10.0

(ppm)
1879.64

10.0
1.0028

9.6

3.75
9.2

9.5
8.8

9.0
2.65
1322.56
1313.11

8.4
0.9979
1309.32

(ppm)

8.5
1299.86

2.60
8.0

8.0
7.6
Integral

7.5
7.2
1165.56
1.0000 3504.82

7.0
1152.95 1.0013 3410.24

(ppm)
6.8
1.0191

8
8 6

(ppm)
(ppm)
0.9835 1141.60 3365.79

261
6.4

6.5
3178.21
0.9790 3176.32

6
6.0

6.0
2843.08
1.0910 2837.09

5.6
2750.08
1-OH

5.5
0.9106 9-OH 2743.14

2532.55 2518.36

5.2
2527.82 2513.00
4-OH

Attachment 25: The 1H NMR spectrum of ampelanol (26).


0.9098 1.0785 2447.11
2342.44

5.0
1-OH 1.0503 2440.81
2336.77

4.8
2332.35
3-OH

9
0.9750 2326.68
Attachment 24: The 1H NMR spectrum of altersolanol A (25).

1.0080 2233.68

4.5
2.2674 2230.84
2208.77
0.8543
4-OH 3-OH 4.4 2-OH
1.1155 2158.33
4 1

0.9442 2-OH 2106.94 2154.55

4.0
4
4.0

3.8543 1915.89 3.2226 1947.10

1
1.0028 1908.01
1898.24 1.1366 1821.31
3

3.6

3.5
1879.64 1815.64
1.4853 1681.33 1809.33
1671.88
3.2

OCH3
OCH3

3.0
3

4a
2.8

0.9979 1322.56
1313.11

2.5
1309.32
1.0191 1299.86
2.4

1165.56
1152.95

2.0
9a
1141.60
2.0

1.5
1.6

2.9015
3.7718 617.00

1.0
1.2

CH3
0.8
CH3

0.5
Attachements
Integral Integral

13.5

13.5
0.9445

13.0
5-OH 6540.82 0.9898 5-OH 6464.84

13.0
12.5
Attachments

12.5
12.0

12.0
11.5

11.5
11.0

11.0
10.5

10.5
10.0

10.0
9.5

9.5
9.0

9.0
8.5

8.5
8.0

8.0
7.5

7.5
7.0

7.0
(ppm)
1.0000 3453.43

6
6
1.0000 3464.78

(ppm)

262
6.5

6.5
6.0

6.0
Integral
Integral

4.5
2831.42 2832.68
1.0067 2824.80 1.0302 2825.74
4.50

2228.63 2235.57

5.5

5.5
2.1558 2222.01
1-OH 1-OH 1.2192 2231.47
2213.18 2225.16

(ppm)
2541.06 2516.15

4.4
(ppm)

0.9030 4-OH 2536.33 0.9818 4-OH 2511.42

5.0

5.0
4.40

0.9614 2197.42
0.8666 2431.35 0.8556 2418.74
2228.63 2235.57

Attachment 27: The 1H NMR spectrum of alterporriol E (28).


3-OH
Attachment 26: The 1H NMR spectrum of alterporriol D (27).

2231.47
3-OH
4

2222.01 1.2192

4.5

4.5
2.1558 2213.18 2225.16
0.9614
4,2-OH 2-OH 2197.42

1.1713 2045.15

1
1

1.1079 2033.17

4.0

4.0
2026.55 2038.53

3.0292 1846.53 3.2322 1834.55


1.2055 1776.23 1.1421 1791.68
3.5

3
3

3.5
1769.61 1784.43
3.0

3.0
OCH3
OCH3

2.5
2.5

2.0
2.0

1.5
1.5
X

3.4475 563.41 3.4160 569.71

1.0
1.0

X X
X

CH3
CH3

0.5
0.5
Integral Integral

7.0

13.0
8
0.9110 6446.87
3403.31
1.0000

6.8
3400.47
5-OH
1085.80
1076.34

12.5
1071.93 3286.97

6.6
0.6496 1056.80 3284.77
1053.01

6
1086.43 1048.28
3.7555

(ppm)

12.0
1082.96

2.10
1041.35

6.4
1073.19 1038.19
1069.41 1028.11
3.3894 1062.79 1024.32
1058.69

6.2

11.5
1018.02

(ppm)
1054.27

2.10
1050.18
1048.60

6.0
1046.39 940.78

11.0
932.58
929.43

5.8
1.85
919.97

10.5
Integral 1.2023

(ppm)

5.6
907.68
990.59

1.80
987.12 898.53

10.0
978.61 894.43

5.4
975.14

(ppm)
1.95
1.9054
967.26
963.79

9.5
5.2
955.60
952.13 800.49

5.0

9.0
788.82
Integral
1.0810
(ppm)

2388.16

4.8

1.50
744.05 0.8496 776.84 2378.38

8.5
1.55

9a
732.07

4.6
2.7110

1.45
(ppm)
719.78 764.86

8.0
0.3475 707.80

4.4

1.40
1.30

3-OH
0.3392 645.38

7.5
4.2
1.8487
(ppm)

0.3765 631.50
628.04

7.0
608.49

4.0

(ppm)
1.25

(ppm)
3346.24
0.4732 3344.98

X X X X
2-OH 619.52

263
0.9567 3344.04

1.20
3.8

6.5
2.4567

(ppm)
595.25 3173.16 3.1346 1859.46

6
1.0000 3170.64

3.6

6.0
582.95
2.9746
OCH3

3.4
0.7567 9-OH 2808.72

5.5
3.2
3

5.0
3.0
Attachment 28: The 1H NMR spectrum of altersolanol J (29).

2.8
2216.65

4.5
0.7786 3-OH 2140.36
1.0221 2129.96

9
1902.97 1085.80
2.6

1.1955 1637.83 1076.34

4.0
1633.41

X
0.4096 1071.93
1626.16 1056.80
2.4

3.5483 1621.75 1053.01


1086.43 1048.28

3.5
1082.96 1041.35
1073.19 1038.19
2.2

3.2309 1069.41
Attachment 29: The 1H NMR spectrum of tetrahydroaltersolanol B (30). 4eq 1028.11

3
1062.79 3.7555 1024.32

OCH3

3.0
1058.69 1eq 1018.02
2.0

1054.27 940.78
1050.18 932.58
1048.60 929.43
1046.39 1.2023 919.97

2.5
1.8

990.59
4a 907.68
987.12 898.53

X
3.3894 978.61 894.43
1.6

975.14 4ax 800.49

2.0
1.0810

4a
1.9054 967.26 788.82
963.79 0.8036

4eq
1eq
776.84
955.60

9a
764.86
1.4

952.13 750.99

1.5
2.7110 744.05
4ax 1ax 740.27
732.07 1.8487 732.07
2.4567
1.2

1ax 719.78 728.29


2.3392
2.9746 707.80 645.38
608.49

1.0
631.50
595.25 628.04
1.0

582.95 619.52

CH3
576.33
CH3

581.69
Attachements
Integral

12.4
0.9287 6012.43

12.0
Attachments

11.6
4-,10-OH

11.2
10.8
10.4
10.0
9.6
9.2
8.8
8.4
4010.19

6,12
1.0566 4001.36

8.0
3971.41

7.6
(ppm)
7.2

264
3433.26
1.0000 3424.43

6.8
5,11

6.4
6.0
1587.07
0.7808 2879.65
3.16

1575.09
1.3012

5.6
1571.62
(ppm)

1559.64
3.12

1-,7-OH

5.2
Attachment 30: The 1H NMR spectrum of stemphyperylenol (31).

4.8
1.0420 2299.25
2.95

1,7

1469.79
4.4

1465.38
1.2144
(ppm)

1454.34
4.0

1449.93
2.90

1864.19
1.0637 1855.36
1439.84
6b,12b

3.6

1587.07
3.2

1.3012 2ax,8ax 1575.09


1571.62
1.2144 2eq,8eq 1559.64
1469.79
X

2.8

1465.38
X

1454.34
1449.93
1439.84
2.4

1360.08
3297.37 Integral

6.8
Integral
3290.12
Integral 3284.44
0.3068 3281.92

11
6.4
3305.57

6.6
3276.25 3125.55

10
0.7911 0.3068 3299.89 3259.22 3123.34

6.60
3297.37

6
0.9981 3243.77 1.0000 3107.58
3290.12 3105.37

6.2
3143.52

6.4
3284.44 0.9636
0.7911

4
4
3141.63
1.0881 3281.92 3123.34

6.55
3276.25 3121.14

(ppm)
0.9614

6.2
6.0

6
3259.22
0.9981

6.50

6.0
5.8
3243.77

5.8
5.6
Integral

5.6
5.4

6.30
3143.52
1.0881 3141.63

5.4
5.2

(ppm)
6.25
3123.34

5.2
0.9614

5.0
3121.14

15

5.0
4.8
2409.27
2402.97
1.6162

4.8
2396.66

4.6

15
2409.27

4.6
1.6162

4.4
2402.97

4.80
(ppm)
2396.66

4.4
4.2

4.2
4.0
1937.95

4.0
2A
1.1760 1922.19
1816.89

3.8
1801.13
1629.94

(ppm)

(ppm)
3.8
2A
1872.69 1628.36

265
1.1238 1.1550 1626.78

3.6
1855.98
1617.64
2B

1614.80

3.6
1732.40 1608.81

3.4
1715.69 1605.98
1.1755 1681.33 1602.19

2B

3.4
1669.98 1599.67

X
1661.15 1.9976 1593.37

3.2
1590.53
1.9976

3.2
1608.81 1381.51
10A

1605.98 1378.99

3.0
1602.19 1371.74
Attachment 31: The 1H NMR spectrum of curvularin (32).

(ppm)

1369.21

3.0
1599.67
1366.38
1363.54

2.8
1593.37 1356.60
1.1851 1590.53

2.8
1353.77
1204.33
2.6
10B

1197.08
1381.51

2.6
1189.51
1378.99
1183.84
1371.74
2.75

1163.66

Attachment 32: The 1H NMR spectrum of dehydrocurvularin (33).


2.4

1160.82 1369.21
1.1851

2.4
1.1747
12A 1153.26 1366.38
(ppm)

1145.38 1363.54 881.50


1.5051 1138.44 876.77
1356.60
2.2

12B 1132.13 1353.77 873.93

2.2
867.00
2.70

1120.47
996.57 798.59
790.70
2.0

989.32
786.61

2.0
979.55
1.2448 977.02 783.45
779.98
13A 969.77
1.4490
1.8

946.76 746.88

1.8
14A 933.83 1.7786 743.41
11A

919.02 734.90
910.19 728.91
1.6

809.93 3.4260 722.92

1.6
2.9092 801.74
11B,14A 711.26
702.75
13B,14B 799.85
791.96 5.6406 696.12
1.4

1.0879
12A,13A,14B 689.82

1.4
787.55
778.72 683.83
768.95 4.6716 657.66
651.04
766.74
12B,13B
1.2

1.2
3.0000 709.37 646.63
704.32 3.6709 638.43
1.3267

X
695.81 633.39
594.92 631.50
1.0

1.0
588.30 626.77
CH3
559.93 564.03
CH3

554.89 557.72
Attachements
Integral

7.6
3651.73
Integral 3647.94
3645.11
3639.12

13.6
3637.86

7.4
3631.87
3504.50 3618.31

13.2
3493.78

7.00
3.6061 3615.16
2.1108 3489.68 3611.69

(ppm)

7.2
3478.33
Attachments

12.8
3504.50
1.0845 6316.98 3493.78
3489.68

7.0
2.1108

3
3478.33

12.4
3170.32
3164.01 3207.20
3163.07 3197.12

6.4
3155.82 3190.50

6.8

2-OH

12.0
3153.61 3186.71
6.1203 3152.66 3180.09

(ppm)
3148.56 3170.32

6.3
3164.01

11.6
3144.78

6.6
3140.37 3163.07
3129.65 3155.82
3153.61

11.2
3152.66

6.4
3148.56
6.1203 2954.68 3144.78
2.2124 2949.95 3140.37

10.8
5.90
2946.48 3129.65
2939.86

6.2
(ppm)
1.8521 2934.82

4,10,11

10.4
2931.35
0.6488 5069.48

6.0
2954.68

10.0
2.2124 2949.95
2946.48
1.8521 2939.86

4-OH

9.6
5.8
2934.82
1.0000 2931.35

2,5
2873.02

9
0.8313
2866.09

9.2
2858.21

5.6
2851.27

8.8
5.4

8.4
0.8778 12A 2616.40

5.2
0.7865 2614.51
2598.43

8.0
0.9015
2547.04
Integral 0.8244 2545.15

(ppm)
5.0
2535.69
12B

(ppm)
3083.31

7.6

266
1.0000 3081.11

7.2
4.8

(ppm)
0.9281 3056.83
3054.63

6.10

6.8
4.6
Integral
2042.30
2034.11

6.4
3083.31 2028.12
4.5454
2025.59
(ppm)

4.4
1.0000 3081.11
3056.83 2022.13
0.9281
3054.63

6.0
5 3
4.2

5.6
8,13A

Attachment 34: The 1H NMR spectrum of orsellinic acid (39).


Integral 2042.30
4.5454
2034.11
1873.64

4.0
2028.12

5.2
Attachment 33: The 1H NMR spectrum of aureonitolic acid (34).

1867.02 2025.59
4.1048 1865.44 2022.13
(ppm)

1860.08
1856.61

4.8
3.8
3.70

4.1048 1873.64
1867.02
1865.44

4.4
1860.08
3.6

1856.61

4.0
7,13B

3.4

3.6
3.2

3.2
1454.33
2.90

1446.45
3.0

2.8
1.9663 1438.57
1454.33
(ppm)

1.9663 1430.69 1446.45


6
2.85

1422.81 1438.57

2.4
2.7696 1192.04
2.8

1430.69
1422.81

2.0
2.6

CH3

1.6
Integral
Integral

7.8
1.6704
7.4844

7.48
7.5588

7.6
2.0000 7.4062

2
1.6704 7.4844

2
7.5

(ppm)
7.4465 7.3281
2.0000 7.4301

4
2.0000
7.4465 2.1888 7.3117

7.4
7.44
7
7.4301 2.3175 7.0658
7.0621

7.2
2.1359 7.1711
7.0482

4 7
2.3393 7.0457

6
Integral 6.8716

7.0
7.0
7.42
6.8698
6.8553
2.1577
6.8527

5
7.18

6.8
2.1888
2.1359 7.1711 7.4062

(ppm)
(ppm)

6.6
7.40

7.16
6.5

6.4
Integral

6.2
6.88
6.8716
6.8698 Integral
2.1577

6.0
(ppm)
6.86
6.8553

6.0
6.8527

7.3281

6.84

5.8
2.3175

(ppm)
7.32
7.3117

5.6
5.40
5.3928 5.3928 5.3929
5.3903 5.3903

5.5

9
2.9811 5.3903

5.4
5.3808
5.3808 5.3783

9
2.9811 5.3783 5.3758 2.2623 5.3815

5.38
5.3758 5.3663

(ppm)
5.3638
Integral 5.3790

5.2
5.3663 5.3664
7.08
5.3638
5.3639

5.36

5.0
4.6895 4.9465
7.0658

5.0
4.9226 7.0621
2.3393

4.8
(ppm)

7.0482
7.0457
7.04

4.6
(ppm)
(ppm)

267
4.4
4.5
Integral

4.2
Integral

3.44

4.0
3.4280
5.3929

4.0
5.3241

(ppm)
5.40

5.3903

3.8
3.42
3.4135 5.3815
2.2623 5.3790
(ppm)

3.6
3.40
5.3664

8
Attachment 35: The 1H NMR spectrum of cochliodinol (35).

5.3241 3.4280
5.36

5.3639

3.4
3.4135
8

2.3569
3.5
3.4501

Attachment 36: The 1H NMR spectrum of isocochliodinol (36).


2.5909 3.4413

3.2
3.4349

3.0
3.0

3.4501

2.8
2.3569 3.4413
2.5909 3.4349
(ppm)

2.6
2.4
2.5

Integral

2.2
6.3663 1.7367

2.0
(ppm)

6.7285 1.7204
2.0

1.8
15.646
1.70

1.7518

1.6
11

6.3663
1.7367
6.7285 1.7204

1.4
12

1.1014 1.2765
1.5

11,12

1.2
Attachements
Integral Integral

8.4

11.4
4035.72
4035.09
4033.51

8.2
0.8253 5453.79 4028.78

10.8
4027.21
1.0000 4026.58

8.0
Attachments

0.9438 3970.46

10.2
Integral

7.8
4035.72
4035.09
4033.51 3716.36

7.6
1.0000 3715.09

9.6
4028.78

(ppm)
4027.21 3709.74
4026.58 3707.84

7
8.04
0.9921 3707.21

7.4

9.0
3601.60

7.2
2.1740 3600.34

7.44
3716.36 3594.66
3715.09 3593.09

8.4
3587.73

7.0
0.9921 3709.74

5,6
3707.84 3585.84

(ppm)
4014.92 3707.21 3580.16

2`
0.9276 3954.39 3578.90
3780.36 3573.23

6.8
0.8959 3772.17 3571.96

7.8
3651.74
3643.54
0.9746 3515.23

5` 4
6.6
3514.60 3600.34
3507.98 3594.66

7.20
1.0000

7
3500.41 3593.09

7.2
3470.14

2
3587.73

6.4
1.9992 3462.58 2.1740 3585.84

(ppm)
0.9909 3455.33 3580.16
3515.23 3578.90
7.15

6
6.2
3514.60 3573.23

6.6
1.9992 3507.98 3571.96

5
7.0
3500.41

6.0

(ppm)
3470.14
0.9909

6.0
3462.58
3455.33

6.9
5.8
0.2152 2876.19

(ppm)
(ppm)

268
5.6

5.4
5.4

4.8
5.2
1796.09

3.60
1789.47

4.2
1.0271
1782.53

5.0
1.0093 2047.67

(ppm)
1775.91

3`
4.8
1796.09

3.6
1.0271 1789.47

6`
1782.53
1775.91

4.6
1.0743 1623.64
1619.54
1609.14

8A

3.0
1.0277
1605.36
4.4
1506.36
1501.95

8B
1492.18
1487.45
4.2

2.4
Attachment 37: The 1H NMR spectrum of indol-3-carboxylic acid (37).

1623.64
1619.54

Attachment 38: The 1H NMR spectrum of cyclo(alanyltryptophane) (38).


1.0743
4.0

1609.14

(ppm)
1605.36

1.8
3.20
3.8

0.5646 610.38

1.2
1506.36
3.6

1.0277 1501.95

3.00
1492.18

(ppm)
1487.45
3.4

0.6
2.9244 198.02
191.08
3.2

0.0
CH3
3.0
Curriculum

Curriculum Vitae

Name : Amal El-Sayed Hassan Abbas Hassan Aly


Day of birth : 9th of June 1975 in Aachen, Germany
Nationality : Egyptian

Course of Education
- Postgraduate studies at the Institut für Pharmazeutische Biologie und Biotechnologie,
Heinrich-Heine-Universität Düsseldorf, Germany, since 2003
- Courses in Mass Spectrometry at the Institut für Pharmazeutische Chemie, Heinrich-Heine-
Universität Düsseldorf, Germany, 2005
- Courses in Nuclear Magnetic Resonance Spectroscopy at the Institut für Pharmazeutische
Chemie, Heinrich-Heine-Universität Düsseldorf, Germany, 2005-2006
- Receiving the M. Sc. degree in Pharmaceutical Sciences (Pharmacognosy) from the
Faculty of Pharmacy, Alexandria University, Egypt, in 2002
- Postgraduate studies at the Department of Pharmacognosy, Faculty of Pharmacy,
Alexandria University, Egypt, 1999-2002
- Second part of M. Sc. degree in Pharmaceutical Sciences (Pharmacognosy), grade
distinction, in 2001
- First part of M. Sc. degree in Pharmaceutical Sciences (Pharmacognosy), grade distinction,
in 2000
- Graduation with B. Sc. in Pharmacy, grade distinction, from the Faculty of Pharmacy,
Alexandria University, Egypt, in 1998
- Faculty of Pharmacy, Alexandria University, Egypt, from 1994-1998
- Graduation with secondary school certificate, in 1993
- Deutsche Schule der Borromäerinnen in Alexandria, Egypt, from 1980-1993

Scientific Experience
- Ph.D. student at the at the Institut für Pharmazeutische Biologie und Biotechnologie,
Heinrich-Heine-Universität Düsseldorf, Germany, since 2003
- Teaching practical courses in pharmaceutical biology for fifth semester students at the
Institut für Pharmazeutische Biologie und Biotechnologie, Heinrich-Heine-Universität
Düsseldorf, Germany, from 2004-2006
- Assistant lecturer at the Department of Pharmacognosy, Faculty of Pharmacy, Alexandria
University, Egypt, from 2002-2003

269
Curriculum

- Supervising and teaching practical courses in phytochemistry for seventh and eighth
semester students at the Department of Pharmacognosy, Faculty of Pharmacy, Alexandria
University, Egypt, from 2002-2003
- Demonstrator at the Department of Pharmacognosy, Faculty of Pharmacy, Alexandria
University, Egypt, from 1999-2002
- Supervising and teaching practical courses in pharmacognosy and phytochemistry for fifth
and sixth semester students at the Department of Pharmacognosy, Faculty of Pharmacy,
Alexandria University, Egypt, from 2000-2002
- Assistant at the Department of Pharmacognosy, Faculty of Pharmacy, Alexandria
University, Egypt, from 1998-1999
- Teaching practical courses in pharmacognosy and phytochemistry for third to sixth
semester students at the Department of Pharmacognosy, Faculty of Pharmacy, Alexandria
University, Egypt, from 1998-1999

Publications
- El-Ghazooly, Maged G., El-Lakany, Abdalla M., Abou-Shoer, Mohamed I., Aly, Amal H..
Chemical constituents of Helichrysum conglobatum growing in Egypt. Natural Product
Sciences (2003), 9(4), 213-219.

Conferences
- Oral and Poster presentation at the International Congress on the Medicinal and Aromatic
Plants, March 2007, Fes, Morocco.
- Poster presentation at the Pharmaceutical Sciences World Congress, April 2007,
Amsterdam, Netherlands.
- Poster presentation at the International Congress and 55th Annual Meeting of the Society
for Medicinal Plant Research, September 2007, Graz, Austria.

270

Das könnte Ihnen auch gefallen