Sie sind auf Seite 1von 10

Journal of Ethnopharmacology 155 (2014) 1076–1085

Contents lists available at ScienceDirect

Journal of Ethnopharmacology
journal homepage: www.elsevier.com/locate/jep

Research Paper

Antiproliferative activity and new argininyl bufadienolide esters from


the “cururú” toad Rhinella (Bufo) schneideri
Guillermo Schmeda-Hirschmann a,n, Cristina Quispe a, Cristina Theoduloz b,
Paulo Teixeira de Sousa Jr.c,d, Carlos Parizotto c,d
a
Laboratorio de Química de Productos Naturales, Instituto de Química de Recursos Naturales, Universidad de Talca, Casilla 747, 3460000 Talca, Chile
b
Laboratorio de Cultivo Celular, Facultad de Ciencias de la Salud, Universidad de Talca, Casilla 747, 3460000 Talca, Chile
c
Laboratório de Pesquisa em Química de Produtos Naturais, Departamento de Química, Universidade Federal de Mato Grosso, Av. Fernando C. da Costa s/n,
78060-900 Cuiabá, Mato Grosso, Brazil
d
Instituto Nacional de Ciência e Tecnologia em Áreas Úmidas (INAU), Rua Nove, 305, Boa Esperanca, 78068-410 Cuiabá, Mato Grosso, Brazil

art ic l e i nf o a b s t r a c t

Article history: Ethnopharmacological relevance: Toads known as “cururú” (Rhinella schneideri) have been used in the
Received 22 January 2014 Brazilian Pantanal and Paraguayan Chaco wetlands to treat erysipelas and cancer. The aim of the study
Received in revised form was to assess the antiproliferative effect of the venom obtained from Rhinella schneideri and to identify its
2 June 2014
constituents by spectroscopic and spectrometric methods.
Accepted 6 June 2014
Available online 16 June 2014
Materials and methods: The venom was obtained by gentle pressing the parotid glands of the toads. The
dry crude drug was analyzed by HPLC–MS–MS and chromatographed on Sephadex LH-20 to obtain
Keywords: purified compounds and fractions for spectroscopic analysis. The venom and fractions were evaluated for
Rhinella schneideri antiproliferative activity towards normal human lung fibroblasts (MRC-5) and four human cancer cell
Bufo
lines: gastric epithelial adenocarcinoma (AGS), lung cancer (SK–MES-1), bladder carcinoma (J82) and
Argininyl bufadienolides
promyelocytic leukemia (HL-60).
Antiproliferative activity
Brazilian pantanal Results: From the Rhinella schneideri venom, 29 compounds were isolated and/or identified by spectro-
scopic and spectrometric means. Three known alkaloids and five argininyl diacids were identified in the
complex mixture by HPLC–MS–MS. Nine out of fifteen argininyl diacid derivatives of the bufadienolides
bufalin, marinobufagin and telocinobufagin are reported for the first time and four argininyl diacids are
described for the first time as natural products. The venom and the fractions 9–13 showed a remarkable
antiproliferative effect, with IC50 values in the range 0.019–0.022, 0.035–0.040, 0.028–0.064, 0.042–0.056
and 0.044–0.052 mg/mL for MRC-5, AGS, SK-MES-1, J82 and HL-60 cell lines, respectively. Under the same
experimental conditions, IC50 values of the reference compound etoposide were 2.296, 0.277, 1.295, 1.884
and 1.059 mg/mL towards MRC-5, AGS, SK-MES-1, J82 and HL-60 cells, respectively.
Conclusions: The venom showed a strong antiproliferative effect towards human cancer cells and
presented a high chemical diversity in its constituents, supporting its use as anticancer agent. These
findings encourage further work on the chemistry and bioactivity of South American toad venoms.
& 2014 Elsevier Ireland Ltd. All rights reserved.

1. Introduction powerful hallucinogen Weil, Davis, 1994. The structure of several


groups of alkaloids discovered in South American amphibians has
Frogs have played a relevant role in South American cultures been revised and show a remarkable chemical diversity (Daly,
since prehistoric times. They were represented in material goods 1998; Daly et al., 2008). In the Guarani tradition of Paraguay and
such as earthen ware, pottery, stone sculpture and gold portrayals the Brazilian Pantanal, “cururú”, the large frog belonging to genus
(Wassen, 1934a) as well as in myths (Wassen, 1934b) all over the Rhinella, is linked to firemyths and to the obtention of fire by the
South American continent. Dendrobatid frogs were used to poison Guarani culture1.
arrows and darts in the Amazon basin (Bisset, 1989) and the large The large toad Rhinella schneideri (Werner, 1894) (synonims:
toad Bufo alvarius (now Rhinella alvarius) was described as a Bufo paracnemis A. Lutz, 1925 and Bufo schneideri Werner, 1894) is

n 1
Corresponding author. Tel.: þ 56 71 2200288. (http://www.virtualberks.com/spanishasasecondlanguage/ElPrimerfuego_
E-mail address: schmeda@utalca.cl (G. Schmeda-Hirschmann). esen.html).

http://dx.doi.org/10.1016/j.jep.2014.06.025
0378-8741/& 2014 Elsevier Ireland Ltd. All rights reserved.
G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085 1077

common in the Paraguayan Chaco wetlands and in the Brazilian associated with geographic and environmental changes that could
Pantanal2 . The large frogs are poisonous when bitten by dogs or explain the current distribution of the different Rhinella species. The
when the venom from the parotid glands came accidentally into formation of the hydrological basins of the Amazon, Paraguay and
contact with the mouth of children. In spite of their abundance, Parana rivers in the late Tertiary and Quaternary, could have played a
frogs were not eaten but sometimes used in healing practices. relevant evolutionary role in the genus speciation. The environmental
Alves, Alves (2011) reported the use of Rhinella schneideri (Werner, conditions of the Chaco and Pantanal wetlands, associated with the
1894) (“cururu” toad), to heal wounds, erysipelas, acne, cancer, to Paraguay River basin, are very different from those of the Brazilian
treat dental caries and to induce abortion in Brazil. The close cerrado, suggesting the occurrence of distinctive bioactive compounds
related species Rhinella jimii (Stevaux, 2002) is used to treat in the toad venoms. The aim of the present work was to verify the
erysipelas in northeastern Brazil (Alves et al., 2012). In the decade traditional indication of use of the Pantanal toad as an antiproliferative
of 1960, a decoction of toad to fight cancer began to be known in agent as well as to identify the toad venom components.
Paraguay. This use seems to be a more recent practice in spite of
some claims that it came from the Guayaki Indians from Eastern
Paraguay. The “cururu” toad is rubbed in varicous wounds and skin
2. Materials and methods
lesions in Paraguayan rural medicine. According to the information
received, the toad is used both to treat cancer, as an aqueous
2.1. Collection of the toad venom
decoction, taken orally or locally applied to treat erysipelas. For the
use against cancer, the toad is killed, skinned and the skin is placed
The toad venom was collected by gentle pressing the parotid
in boiling water (about one liter). The decoction is bottled and
glands from free living individuals at SESC-Pantanal, Mato Grosso,
stored in a cold place. The usual dose was a glass of about 100–
Brazil, on April 26, 2013. The toad population surveyed gathers at
150 ml of the decoction per day.
the river harbor to feed on insects attracted by the town lights.
The toad venom has been used as a crude drug in Oriental
About 30 individuals were captured and released after collecting
medicine and traditional Chinese medicine for a very long time. It
the venom. The secretion was suspended/dissolved in MeOH for
is recommended mainly as an anti-inflammatory (Ma et al., 2007)
preservation until work-up at the laboratory. The toad was
and anticancer (Meng et al., 2009) agent. According to The State
identified as Rhinella (Bufo) schneideri by Prof. Domingos de Jesus
Pharmacopoeia Commission of People´s Republic of China (2005),
Rodrigues, Universidade Federal de Mato Grosso, Núcleo de
the crude drug and its aqueous extract (cinobufacini) are used as
Estudos da Biodiversidade da Amazônia Matogrossense (NEBAM),
cardiotonic, antineoplastic, local anesthetic and antimicrobial
Instituto de Ciências Naturais, Humanas e Sociais, Cep 78557-267
agent. In spite of an increasing interest in the toad venoms as
Sinop, MT, Brazil.
potential sources of bioactive agents, less is known on the use of
frog secretions in South American traditional medicine.
Sciani et al. (2013) described the antiproliferative activity of 2.2. Isolation
crude secretions from Brazilian frogs, obtained by manual compres-
sion of the venom glands. Seven species of Rhinella (formerly Bufo) The freeze-dried solid (1.2 g) was treated with 3  20 mL of
and one Phyllomedusa species were investigated. The venom of DCM:MeOH 1:1 under sonication (5 min each time), filtered and
Rhinella schneiderii showed an IC50 value of 48 mg/mL towards the taken to dryness under reduced pressure to afford 256 mg of a
breast cancer cell MCF-7. However, no reference compound for yellowish oil. The solid remaining after DCM:MeOH extraction was
comparison was included in this study. The extracts of the Brazilian sonicated with CHCl3 (30 mL, 10 min) and MeOH (30 mL, 10 min)
frogs were investigated for the occurrence of already described to obtain additional fractions (97 mg of CHCl3 and 101 mg MeOH-
molecules in the Chinese crude drug ChanSu by HPLC-MS. The solubles). After TLC comparison, the three combined extracts
alkaloids dehydrobufotenin and serotonin were found in all the (453 mg) were pooled for chromatography. Some 440 mg of the
secretions and were more abundant than the steroids (Sciani et al., combined extracts sample was loaded into a Sephadex LH-20
2013). Anjolette et al. (2011) reported two active subfractions from column (column length 40 cm, i.d. 3 cm, Sephadex load: 20 cm)
Rhinella schneiderii venom with effects on the complement system. equilibrated with DCM:MeOH 1:1 v/v to afford 25 fractions. After
In a congress abstract, Baldo et al. (2012a) describe the effect of TLC comparison, the fractions were pooled as follows: 1–3
different fractions from the Rhinella schneideri venom against (160 mg), 4 and 5 (46 mg), 6–11 (35 mg), 12 and 13 (23 mg), 14–
seizures induced by the convulsant agents PTZ and NMDA, suggest- 21 (10 mg), 22–25 (8 mg).
ing a neuroprotective effect of the toad venom molecules. In the
same congress, Baldo et al. (2012b) report the cytotoxicity of the
Rhinella schneideri poison towards murine melanoma cells, HL-60, 2.3. HPLC analysis
HepG2, PC-12 and PBMC cells. The venom showed different effects
on the cells, but IC50 values were not reported. The HPLC system used for DAD analysis of extracts was a
In Brazilian Rhinella species, telocinobufagin, hellebregenin and Shimadzu equipment (Shimadzu Corporation, Kyoto, Japan) con-
bufalin were identified as known cytotoxic/antitumoral compounds sisting of a LC-20AT pump, a SPD-M20A UV diode array detector,
(Sciani et al., 2013). The main bufadienolides marinobufagin, bufalin, CTO-20AC column oven and a LabSolution software. A MultoHigh
telocinobufagin, hellebrigenin, 20 S,21 R-epoxymarinobufagin and 100 RP 18-5m (250  4.6 mm) column (CS-Chromatographie Ser-
β-sitosterol were reported from Rhinella schneideri captured around vice GmbH- Germany) maintained at 25 1C was used. Approxi-
Brasilia in the cerrado phytogeographic region (Cunha-Filho et al., mately 5 mg of the extract obtained as explained above was
2010). The compounds and some semisynthetic derivatives were dissolved in 1 mL MeOH, filtered through 0.45 mm PTFE filter
assessed for cytotoxicity using several cell lines. (Waters) and submitted to HPLC-DAD analysis. The compounds
The distribution range of Rhinella schneideri and the phylogenetic were monitored at 295 nm. The HPLC analysis was performed
relationship of South American Rhinella species were reported by using a linear gradient solvent system consisting of 1% formic acid
Maciel et al. (2010). The results of the study suggest diversification (A) and acetonitrile (B) as follows: from 92% to 46% A over 45 min.
The flow rate was 1 mL/min. The volume injected was 20 mL. The
HPLC conditions were similar to those reported by Gao et al.
2
(http://www.iucnredlist.org/details/54755/0). (2010) for a comparison of venoms from different Bufo species.
1078 G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085

2.4. QTOF-analysis diluted with complete medium, and incubated for 72 h in a


humidified incubator with 5% CO2 in air at 37 1C. Untreated cells
ESI–MS–MS analyses were conducted in a Micromass (Man- were used as 100% viability controls. Control cells were grown
chester, UK) Q-TOF micro instrument. The samples were directly with complete medium and 1% DMSO. At the end of the incubation
infused at a flow rate of 10.0 mL/min using a syringe pump the MTT reduction assay was performed as described previously
(Harvard Apparatus, Holliston, United States of America). ESI mass (Rodriguez, Haun, 1999). The final concentration of MTT was 1 mg/
and tandem mass spectra were acquired in the positive ion mode. mL. Etoposide (E1383), vinblastin (V1377) and camptothecin
The following operating conditions were used: 3.0 kV capillary (C9911) from Sigma-Aldrich (St. Louis, MO, USA) were used as
voltage, 40 V cone voltage and desolvation gas temperature of reference substances. Each experiment was carried out three times
100 1C. Tandem ESI–MS–MS spectra were collected by causing in quadruplicate. The results were transformed to percentage of
collision-induced dissociation (CID) of the mass-selected proto- controls and the IC50 value was obtained adjusting the dose-
nated molecules using argon as the buffer gas and collision response curve to a sigmoidal model.
energies from 5 to 45 eV. Mass-selection was performed by
Quadrupole 1 using a unitary m/z window, and collisions were
performed in the rf-only quadrupole collision cell, followed by 3. Results and discussion
Time of Flight (TOF) mass analysis. ESI–MS were acquired over an
m/z range of 80–1000 amu. The chemical composition and antiproliferative activity of the
parotid glands venom from the “cururu” toad Rhinella schneideri
2.5. HPLC–MS–MS analysis was investigated. The crude venom extract was permeated into a
Sephadex LH-20 column and the constituents of the CC fractions
Mass spectra were recorded using an Agilent 1100 (USA) liquid were analyzed by TLC, 1HNMR and QTOF-MS. While the TLC was
chromatography system connected through a split to an Esquire helpful in the identification of main known constituents, NMR
4000 Ion Trap LC/MS system (Bruker Daltoniks, Germany) or a spectra allowed identification of the main unknown metabolites
Merck-Hitachi 6200 Intelligent Pump and L 4000 UV detector and indicated the occurrence of minor compounds, related to the
coupled to a EBE trisector VG Autospec Micromass spectrometer main products. The main compounds isolated and fully character-
(Micromass-Water Autospec, U.K.) operating at 70 eV. Full scan ized by spectroscopic and spectrometric means were used as
mass spectra were measured between m/z 150 and 2000 u in reference both for the analysis of the different fractions by
positive ion mode. For the Ion Trap LC/MS system, nitrogen was QTOF-MS as well as for HPLC–MS–MS. The HPLC chromatogram
used as nebulizer gas at 27.5 psi, 350 1C and at a flow rate of 8 L/min. of the venom is presented in Fig. 1. The structures of the
The mass spectrometric conditions for positive and negative ion compounds isolated and/or tentatively identified in the “cururu”
mode were: electrospray needle, 4000 V; end plate offset, 500 V; toad venom are shown in Figs. 2 and 3.
skimmer 1, 56.0 V; skimmer 2, 6.0 V; capillary exit offset, 84.6 V;
capillary exit, 140.6 V. Collision induced dissociation (CID) spectra 3.1. NMR and QTOF–MS analysis
were obtained with a fragmentation amplitude of 1.00 V (MS/MS)
using helium as the collision gas. QTOF–MS analysis was performed to get an insight into the
identity of the minor compounds in the fraction pools 1–3, 4 and
2.6. NMR analysis 5, 6–11, 12 and 13, 14–21 and 22–25. The general MS pattern of the
bufadienolides shows the loss of the terpene moiety leading to the
The NMR spectra were recorded on a Bruker Avance 400 argininyl diacid, allowing the identification of the steroid and the
(Bruker, Rheinstetten, Germany) spectrometer at 400 MHz for 1H diacid linked to the basic aminoacid and the steroid (Hu et al.,
and 100 MHz for 13C in CDCl3 or MeOH-d4. Chemical shifts are 2011; Gao et al., 2010; Lenaerts et al., 2013). The fragmentation
given in ppm with residual chloroform or methanol as the internal pattern in MS allows a clear differentiation of the bufadienolides
standard. based on bufalin, marinobufagin or telocinobufagin, characterized
by the neutral loss of [M-H2O] of 368, 382 and 384 amu for bufalin,
2.7. Antiproliferative assay marinobufagin and telocinobufagin, respectively.
The 1H NMR spectrum of fractions 1–3 presented typical
The MTT reduction assay was used to determine the viability of signals for the α-pyrone ring of bufadienolides, with a main
MRC-5 normal human lung fibroblasts (ATCC, CCL-171), AGS compound 14 showing a dd at δ 7.92 (1H, J ¼9.6 and 2.4 Hz), a d
human gastric adenocarcinoma cells (ATCC, CRL-1739), SK-MES-1 at δ 7.48 (1H, J ¼1.6 Hz) and a d at δ 6.30 (1H, J ¼9.6 Hz) and a
human lung cancer cells (ATCC, HTB-58), J82 human bladder minor constituent 7 with a dd at δ 8.01 (1H, J¼ 9.6 and 2.4 Hz), a d
carcinoma cells (ATCC, HTB-1), and HL-60 human promyelocytic at δ 7.46 (1H, J¼ 1.6 Hz) and a d at δ 6.31 (1H, J¼ 9.6 Hz).
leukemia (ATCC, CCL-240) from the American Type Culture Collec- Furthermore, the br s at δ 5.04, dd at δ 4.30 (1H, J ¼6.8, 5.2 Hz),
tion (Manassas, VA, USA). MRC-5, J82, and SK-MES-1 cells were the s at δ 3.46, the t at δ 2.26 and 2.15 and the multiplets at 1.40–
grown in Eagle's Minimum Essential Medium, containing 2 mM 1.68 and 1.16–1.38, pointed out to argininyl diacid derivatives of
L-glutamine, 1 mM sodium pyruvate and 1.5 g/L NaHCO3. AGS cells bufadienolides. The methyl singlets at δ 1.01 and 0.81 for the main
were grown in Ham's F-12 medium supplemented with 2 mM L- and δ 0.98 and 0.74 for the minor constituent, can be assigned to
glutamine and 1.5 g/L NaHCO3. HL-60 cells were grown in suspen- H-19 and H-18 of the terpene moiety, respectively.
sion in RPMI 1640 medium containing 1 mM sodium pyruvate and QTOF-MS analysis of the fraction 1–3 showed a main com-
2 g/L NaHCO3. All media were additionally supplemented with 10% pound with an ion at m/z 713.6476 [Mþ H] þ which in MS/MS
heat inactivated fetal bovine serum, 100 IU/mL penicillin G, and shows loss of water (695.6282) leading to a base peak at 331.2944.
10 mg/mL streptomycin. Cells were sub-cultured once a week and The fragment agrees with a molecular formula of C14H26N4O5
the medium was changed every two days. For the assay, cells were accounting for a C-8 dicarboxylic acid linked to arginine. The
plated in 96-well plates (100 mL/well) at a density of 50,000 cells/ additional fragment at m/z 175.1832 [C6H14N4O2 þH] þ confirms
mL. One day after seeding, the cells were treated with the medium the identity of the aminoacid. The neutral loss of 382 amu [M–
containing the compounds at concentrations ranging from 0 to H2O] agrees with the molecular formula C24H32O5 (400 amu) for
100 mg/mL, first dissolved in DMSO (final concentration of 1%), marinobufagin. The compound was identified as 3-(N-suberoyl
G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085 1079

argininyl) marinobufagin, namely marinobufotoxin (compound spectra. The compounds A, B and C have not been previously
14). The spectral data are in agreement with marinobufotoxin, described as toad venom constituents.
reported from Bufo ictericus and Bufo marinus by Linde-Tempel Fractions 4 and 5, showed a 1H NMR spectrum close to that of
(1970) and synthesized by Pettit, Kamano (1974). A second con- fractions 1–3, bearing characteristic signals for argininyl esters of
stituent (compound 7) with m/z 699.5363 [Mþ H] þ shows a neutral bufadienolides. The fraction was analyzed by QTOF-MS and then
loss of 368 amu pointing out to a terpene of 386 amu, differing from treated with diazomethane in dietyl ether to afford the methyl
compound 14 in one oxygen function and an unsaturation equiva- ester derivatives of the natural products. In the 1H NMR spectrum
lent, in agreement with bufalin. The base peak at 331 indicates the of the methylated fraction, the OMe singlet at δ 3.37 supports the
presence of a suberoyl arginine moiety. The compound 7 was presence of the free COOH function in the aminoacid moiety while
identified as 3-(N-suberoyl argininyl)-3,14-dihydroxybufa-20,22- the whole spectrum of the bufadienolide remains unaltered.
dienolide (bufalitoxin), previously reported from Bufo vulgaris for- The fractions 6–11 were further purified by preparative TLC
mosus by Shimada et al. (1975, 1976, 1977). The compounds 12, 13, (EtOAc:2-propanol:conc NH4OH 9:7:4 v/v/v) to afford compounds
16, A, B and C were identified on the basis of the MS/MS spectra. 10 (5 mg), 24 (2 mg) and 26 (1 mg). The 1H NMR spectrum of
The compound 13 with [MþH] þ 699 differs from compound 14 in fractions 12 and 13 showed a main compound (10), presenting the
one CH2 unit, showing a fragmentation pattern in agreement for 3- structural features characteristic of a bufadienolide. The dd at δ
(N-pimeloyl argininyl) marinobufagin, previously reported from 7.91 (1H, J ¼9.6 and 2.4 Hz), a d at δ 7.46 (1H, J ¼1.6 Hz) and a d at
Bufo marinus (Dictionary of Natural Products on DVD, 2013). δ 6.28 (1H, J ¼9.6 Hz) clearly indicates an α-pyrone ring while an
The QTOF-MS spectrum of compound 12 with a [MþH] þ ion at m at δ 4.14 (1H) and an s at δ 3.61 (1H) were assigned to H-3 and
m/z 685.5116 shows the loss of water and a neutral fragment of 382 H-15, respectively. Two methyl singlets at δ 0.99 and 0.80 are in
from the [MþH] þ peak, leading to the base peak at m/z 303.2126 agreement with H-19 and H-18, respectively. The QTOF-MS of 10
and a fragment at m/z 175, compatible with arginine. The base peak presented a [MþH] þ peak at 401.3618, whose fragmentation
of compound 12 (303 amu) differs from that of compounds 7 and 14 pattern is in full agreement with that of marinobufagin. Therefore,
(331 amu) by 28 amu, and can be explained by a C6 dicarboxylic 10 was identified as 14,15-epoxy-3,5-dihydroxybufa-20,22-dieno-
acid unit acting as a linker between the terpene and aminoacid part lide (marinobufagin). Marinobufagin was previously reported from
of the molecule. The fragmentation pattern of the bufadienolide is skin secretions of Bufo rubescens (Cunha Filho et al., 2005). A
in agreement with that of marinobufagin. Therefore, the compound second constituent, with [MþH] þ at m/z 205.2051 was identified
12 was assigned as 3-(N-adipoyl argininyl) marinobufagin. The by its 1H NMR spectrum as well as QTOF-MS as the alkaloid
compound 16 with [MþH] þ at 741.3877 shows a base peak at bufotenin (24) (C12H16N2O: 204.12626). The MS–MS is in accor-
359.3, differing from the base peak at 331.3 observed for com- dance with the proposed structure (Maciel et al., 2003). Suberoyl
pounds 7 and 14 in 28 amu. Therefore, the diacid linked to the arginine was identified by QTOF analysis of the fraction, as a minor
arginine should be C10 and the neutral loss fragment requires a constituent. After preparative TLC of fractions 12 and 13 (silica gel,
molecular formula of C24H32O5 (400 amu) for the terpene moiety, in EtOAc:2-propanol:conc NH4OH 9:7:4 v/v/v), five zones were
agreement with marinobufagin. The compound 16 differs from scraped off from the plate and analyzed for alkaloids by QTOF-
compound 14 in two CH2 units in the diacid and was assigned as MS. The fraction 12-13-2 showed the alkaloid bufotenin 24 and a
3-(N-sebacyl argininyl) marinobufagin. Compounds 12 and 16 are second constituent (26) with [M þH] þ at m/z 203.1096, in agree-
described for the first time as natural products. ment with dehydrobufotenin (C12H14ON2: 202.1106). Fraction 12-
The QTOF-MS of compounds 7, 14, A, B and C shows loss of 13-4 yielded a mixture of compounds 24 and 26, while fraction 12-
water from the [Mþ H] þ peak and neutral loss of a fragment 13-5 contained nearly pure compound 26. The MS spectra are in
leading to the base peak at m/z 331 and the arginine fragment at agreement with those reported by Maciel et al. (2003).
m/z 175. The diacid in these compounds is a C8 (suberoyl) unit Fractions 14–21 afforded compound 10 while the pooled frac-
acting as a linker between the basic aminoacid and the bufadie- tions 22–25 contained the alkaloids 24–26 and compounds 3 and 4.
nolide. Related compounds were reported from Bufo bufo gargar- The QTOF-MS of compounds 3 and 4 shows [MþH] þ at 331.2058
izans Cantor by Shimada et al. (1985). and 345.2282 amu, respectively and fragmentation leading to
The compound A, with a molecular weight of [Mþ H] þ arginine, pointing out to diacid derivatives of arginine. Compounds
729.5521 (calc for C38H56N4O10: 728.3996) showed a neutral loss 3 and 4 were assigned to suberoyl arginine and azelayl arginine,
of 398 amu [terpene – H2O] in agreement with a steroid moiety respectively. While compound 3 was previously reported from Bufo
with the molecular formula C24H32O6 (416 amu) leading to a base species by Gao et al. (2010), compound 4 was not described as a
peak at [M þH] þ of 331 (suberoyl arginine). The ion at m/z 345, natural product but as part of more complex bufadienolides.
shows the loss of four water molecules from the steroid moiety
[Mþ H  4H2O] þ , suggesting three hydroxy and one epoxy
3.2. HPLC–MS–MS analysis
functions in the bufadienolide. Compound A differs from 14 in
one additional OH group in the steroid moiety and was tentatively
The composition of the toad venom was investigated by
assigned to 3-(N-suberoyl argininyl)-hydroxmarinobufagin. For
positive and negative mode ionization HPLC–MS–MS. The chro-
compound B, the [MþH] þ at 731.5519 shows the neutral loss of
matograms in the positive and negative ion mode are shown in
400 amu, in agreement with a molecular formula C24H34O6
Fig. 1. The results are summarized in Table 1 (A and B). The
(418 amu) for the terpene moiety, and a base peak at 331,
structures of the compounds 1–26 are shown in Figs. 2 and 3.
characteristic for suberoyl arginine. Compound B was identified
as 3-(N-suberoyl argininyl)-hydroxytelocinobufagin. Compound B
might be originated from opening of the 14,15-epoxy ring of A. The 3.2.1. Argininyl diacid derivatives
bufadienolide C with a [Mþ H] þ at 741.3877 presents a base peak The HPLC–MS–MS chromatograms of the toad venom showed
at 331.2043, in agreement with suberoyl arginine and a neutral the occurrence of compound 3 in the positive ion mode and that of
loss of 414 amu [steroid-H2O], in agreement with dihydroxymar- compounds 1–5 in the negative ion mode. All the compounds 1–5
inobufagin C24H32O7 (432 amu). The compound was tentatively showed the loss of the diacid leading to the arginine moiety and
assigned as 3-(N-suberoyl argininyl)-dihydroxymarinobufagin. were assigned as adipyl (C6 diacid), pimeloyl (C7 diacid), suberoyl
However, the exact placement of the additional hydroxy function (C8 diacid), azelayl (C9 diacid) and sebacyl (C10 diacid) arginine,
of compounds A, B and C cannot be given based on the MS–MS respectively. Suberoyl arginine was previously described from Bufo
1080 G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085

Fig. 1. HPLC chromatogram of the Rhinella schneideri venom extract. UV chromatogram at 295 nm and total ionic current (TIC). A. Positive ion mode. B. Negative ion mode.
Compounds identification: 1: Adipyl arginine; 2: Pimeloyl arginine; 3: Suberoyl arginine; 4: Azelayl arginine; 5: Sebacyl arginine; 6: Bufalin; 11: 3-(N-glutaryl argininyl)-
marinobufagin; 7: 3-(N-suberoyl argininyl)-bufalin (Bufalitoxin); 8: 3-(N-azelayl argininyl)-bufalin; 9: 3-(N-sebacyl argininyl)-bufalin; 12: 3-(N-adipoyl argininyl)
marinobufagin; 13: 3-(N-pimeloyl argininyl) marinobufagin; 14: 3-(N-suberoyl argininyl) marinobufagin (Marinobufotoxin); 15: 3-(N-azelayl argininyl marinobufagin; 16:
3-(N-sebacyl argininyl) marinobufagin; 17: 3-(N-undecadienoyl argininyl) marinobufagin; 18: 3-(N-dodecadienoyl argininyl) marinobufagin; 19: Telocinobufagin; 20: 3-(N-
pimeloyl argininyl) telocinobufagin; 21: 3-(N-suberoyl argininyl) telocinobufagin (Telocinobufatoxin); 23a: 3-(N-suberoyl argininyl) hellebrigenin; 22: 3-(N-sebacyl
argininyl) telocinobufagin; 24. Bufotenin; 25: Bufotenidin.

venom (Gao et al., 2010). Compounds 1, 2, 4 and 5 were not argininyl) marinobufagin (16), 3-(N-undecadienoyl argininyl) marino-
previously reported as natural products but as esters of dicar- bufagin (17) and 3-(N-dodecadienoyl argininyl) marinobufagin (18).
boxylic acid arginine conjugates of bufadienolides.
3.2.4. Telocinobufagin argininyl diacids
The genine telocinobufagin was isolated from Rhinella jimi by
3.2.2. Bufalin argininyl diacids Tempone et al. (2008). In the positive ion-MS, following the loss of
The compounds 7–9, detected in the positive-ion MS, showed a 384 amu corresponding to [M þH H2O] þ , further fragmentation
common loss of 369 amu corresponding to [Mþ H H2O] þ and of compounds 20–22 lead to the arginine ion, allowing to
similar fragmentation patterns, leading to the m/z ion at 175 amu, differentiate the compounds by the number of CH2 units of the
corresponding to arginine. Compounds 7–9 differ in the number of diacid linker. Compounds 20–22 present a diacid with 5, 6 or 8
CH2 groups in the diacid and were identified as follows: 3-(N- CH2 units, allowing the assignation of the compounds as pimeloyl,
suberoyl argininyl)-bufalin (bufalitoxin) (7), isolated from Bufo suberoyl, and sebacyl argininyl derivatives of telocinobufagin. The
vulgaris formosus venom (Dictionary of Natural Products on DVD compound 21, 3-(N-suberoyl argininyl)-telocinobufagin (Telocino-
2013, Shimada et al., 1977) and 3-(N-sebacyl argininyl)-bufalin (9), bufotoxin) was previously reported from Venenum Bufonis (Hu et
found in Venenum Bufonis by Hu et al. (2011). The compound 8, al., 2011) and from Bufo marinus (Dictionary of Natural Products on
described here for the first time as a natural product, shows azelaic DVD, 2013). Compounds 20 and 22 are reported here for the first
acid as the diester and was identified as 3-(N-azelayl argininyl)- time as natural products.
bufalin based on the MS fragmentation pattern.

3.2.5. Hellebrigenin argininyl diacids


Hellebrigenin is a genine occurring in several toad venoms (Gao
3.2.3. Marinobufagin argininyl diacids
et al., 2008). In our sample, one of the minor constituents
A second group of argininyl diacids include the compounds
(compound 23a) was tentatively identified as 3-(N-suberoyl argi-
11–18, all of them derivatives of marinobufagin 10, isolated from
ninyl) hellebrigenin. The structure follows from the [M-H]– ion at
Rhinella schneideri venom. The QTOF-MS analysis of marinobufagin
727.6 and the loss of 398 amu, leading to the argininyl suberoyl
was reported by Cunha Filho et al. (2005) and our data are in
moiety at m/z 329.1 amu.
agreement with the literature. The compounds 11–18 were detected
in the positive ion-HPLC-MS, following the common loss of 383 amu
corresponding to [MþH H2O] þ . The compounds showed a similar 3.2.6. Alkaloids
fragmentation pattern, leading to the arginine m/z ion at 175 amu, and The alkaloids 24 and 25 were detected in the positive ion mode
differing in the diacid moiety. While compounds 13 and 14 were analysis of the venom, in agreement with the NMR studies and
already described as 3-(N-pimeloyl argininyl) marinobufagin (13) from QTOF-MS analysis of the fractions.
Bufo marinus (Dictionary of Natural Products on DVD, 2013) and 3-(N-
suberoyl argininyl) marinobufagin (Marinobufotoxin) (14) from Bufo 3.3. Compounds
marinus and Bufo ictericus (Dictionary of Natural Products on DVD,
2013), compounds 12, 15–18 are reported here for the first time. The 3.3.1. Argininyl diacids
compounds were identified as 3-(N-adipoyl argininyl) marinobufagin Compound 1: Adipyl arginine. Molecular formula: C12H22N4O5;
̄
(12), 3-(N-azelayl argininyl) marinobufagin (15), 3-(N-sebacyl HPLC–MS–MS [M  H]: 300.9; 300.9 (100), 240.8 (12).
G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085 1081

Table 1
Tentative identification of Rhinella schneideri toad venom constituents by HPLC–MS–MS in the positive (A) and negative ion mode (B).

(A) Positive ion mode.

Rt (min) [MþH] þ MS/MS Tentative identification

7.4 204.8 159.6, 57.4 Bufotenin 24


7.5 218.8 159.6 Bufotenidin 25
9.4 660.8 331.2 Suberoyl arginine 3 [2 M þH] þ
12.0 387.4 351.2 Bufalin 6
21.9 671.7 653.4, 289.1 3-(N-glutaryl argininyl)-marinobufagin 11
22.0 715.7 697.6, 331.3, 278.1 3-(N-suberoyl argininyl) telocinobufagin (Telocinobufatoxin) 21
22.8 701.5 683.6, 317.2 3-(N-pimeloyl argininyl) telocinobufagin 20
23.0 685.6 667.5, 303.2, 250.8 3-(N-adipoyl argininyl) marinobufagin 12
23.9 699.6 681.5, 317.2, 331.3 3-(N-pimeloyl argininyl) marinobufagin 13
24.9 713.7 695.6, 331.3, 278.0 3-(N-suberoyl argininyl) marinobufagin (Marinobufotoxin) 14
25.8 727.7 709.5, 345.3, 343.3 3-(N-azelayl argininyl) marinobufagin 15
27.4 741.7 723.6, 359.3, 278.1 3-(N-sebacyl argininyl) marinobufagin 16
27.5 699.7 681.6, 331.3 3-(N-suberoyl argininyl)-bufalin (Bufalitoxin) 7
27.9 403.2 349.2, 367.2, 350.2, 271.0 Telocinobufagin 19
28.8 755.7 737.6, 373.4 3-(N-undecadienoyl argininyl) marinobufagin 17
28.9 713.7 695.6, 345.4 3-(N-azelayl argininyl)-bufalin 8
30.1 769.7 751.7, 387.4, 306.3 3-(N-dodecadienoyl argininyl) marinobufagin 18
30.2 727.8 709.7, 359.4, 279.0 3-(N-sebacyl argininyl)-bufalin 9

(B) Negative ion mode

̄
Rt (min) [M  H] MS/MS Tentative identification

̄
5.2 603.0 300.9, 240.8 [2M  H] Adipyl arginine 1
̄
6.8 631.0 315.0 [2M  H] Pimeloyl arginine 2
̄
9.2 659.0 329.1 [2M  H] Suberoyl arginine 3
10.3 357.2 312.0, 172.5 Sebacyl arginine 5
12.9 343.2 301.0, 172.6 Azelayl arginine 4
22.5 683.7 641.5, 301.0, 240.8 3-(N-adipoyl argininyl) marinobufagin 12
22.8 699.5 657.4, 315.0, 655.3, 272.8 3-(N-pimeloyl argininyl) telocinobufagin 20
23.4 727.6 685.5, 329.1, 268.9 3-(N-suberoyl argininyl) hellebrigenin 23a
23.6 697.6 655.4, 315.1 3-(N-pimeloyl argininyl) marinobufagin 13
23.8 713.5 671.4, 329.1, 287.0, 268.9 3-(N-suberoyl argininyl) telocinobufagin (Telocinobufatoxin) 21
24.5 711.5 669.4, 329.2, 287.1, 270.1 3-(N-suberoyl argininyl) marinobufagin (Marinobufotoxin) 14
25.8 725.4 683.4, 343.1, 684.3 3-((N-azelayl argininyl) marinobufagin) 15
26.3 741.5 357.1, 699.5, 340.2, 315.1, 297.1 3-(N-sebacyl argininyl) telocinobufagin 22
27.5 739.5 697.6, 357.2, 315.1, 297.0 3-(N-sebacyl argininyl) marinobufagin 16
27.5 697.6 655.6, 329.1 3-(N-suberoyl argininyl)-bufalin (Bufalitoxin) 7

Compound 2: Pimeloyl arginine. Molecular formula: Compound 8: 3-(N-azelayl argininyl)-bufalin. Molecular for-
̄
C13H24N4O5; HPLC–MS–MS [M  H]: 315.0. mula: C39H60N4O8; HPLC–MS–MS [Mþ H] þ : 713.7; 695.6 (100),
Compound 3: Suberoyl arginine. Molecular formula: 345.4 (11).
C14H26N4O5; QTOF–MS–MS [MþH] þ : 331.2058; 331 (19), 272 Compound 9: 3-(N-sebacyl argininyl)-bufalin Molecular for-
̄
(26), 250 (100), 175 (68), 158 (71); HPLC–MS–MS [M  H]: 329.1. mula: C40H62N4O8; HPLC–MS–MS [Mþ H] þ : 727.8; 709.7 (100),
Compound 4: Azelayl arginine. Molecular formula: C15H28N4O5; 359.4 (15), 279.0 (4).
̄
HPLC–MS–MS [M  H]: 343.2; 301.0 (100), 172.6 (28). QTOF–MS–
þ
MS [Mþ H] : 345.2282.
Compound 5: Sebacyl arginine. Molecular formula: C16H30N4O5;
̄
HPLC–MS–MS [M  H]: 357.2; 312.0 (100), 172.5 (11). 3.3.3. Marinobufagin argininyl diacid derivatives
Compound 10: 14,15-epoxy-3,5-dihydroxybufa-20,22-dienolide
(marinobufagin); Molecular formula: C24H32O5; QTOF–MS–MS
(20 eV) [M þH] þ : 401.3618; 401 (100), 383 (12), 365 (40), 347
3.3.2. Bufalin argininyl diacid derivatives (30), 339 (22), 319 (12), 269 (17), 253 (20), 159 (23). Marinobufagin
Compound 6: 3,14-dihydroxybufa-20,22-dienolide (Bufalin). acetate (compound 10a: QTOF–MS–MS: 907.7390 [2 MþNa] þ ;
Molecular formula: C24H34O4; HPLC–MS–MS [Mþ H] þ : 387.4; 465.3669 [M þNa] þ ; 443.3846 [MþH] þ .
351.2 (100). Compound 11: 3-(N-glutaryl argininyl)-marinobufagin. Molecu-
Compound 7: 3-(N-suberoyl argininyl)-bufalin (Bufalitoxin). lar formula: C36H54N4O8; HPLC–MS–MS [M þH] þ : 671.7; 653.4
Molecular formula: C38H58N4O8; QTOF–MS–MS (40 eV) [MþH] þ : (60), 289.1 (100).
699.5363 (22), 681.5162 (22), 331.2511 (100), 317.2320 (65), Compound 12: 3-(N-adipoyl argininyl) marinobufagin. Molecu-
250.1973 (56), 175.1503 (52). HPLC–MS–MS [MþH] þ : 699.7; lar formula: C36H52N4O9; exact mass: 684.3734; QTOF–MS–MS
681.6 (100), 331.3 (31); HPLC–MS–MS [M  H]-: 697.5; 655.6 [MþH] þ : (40 eV): 685.5116 (19), 667.5118 (13), 655.5584 (13),
(100), 329.1 (31). 1H NMR: δ 8.01 (dd, J ¼9.6 and 2.4 Hz, H-22), δ 331.2511 (19), 303.2126 (100), 250.1591 (21), 175.1567 (16); HPLC–
7.46 (d, J ¼1.6 Hz, H-21), δ 6.31 (d, J ¼9.6 Hz, H-23), δ 5.25 m, δ MS–MS [Mþ H] þ : 685.6; 667.5 (67), 303.2 (100), 250.8 (5); HPLC–
5.04 (br s, NH), δ 4.30 (dd, J¼ 6.8, 5.2 Hz, H-3), δ 2.61 (d, J ¼10.4, MS–MS [M  H]-: 683.7; 641.5 (100), 301.0 (99), 240.8 (34).
H-17), δ 2.26 (t, J ¼7.2), δ 2.15 (t, J ¼7.2), δ 1.40-1.68 (m), δ 1.16-1.38 Compound 13: 3-(N-pimeloyl argininyl) marinobufagin. Mole-
(m), 0.98 s (H-19), 0.74 s (H-18). cular formula: C37H54N4O9; QTOF–MS–MS [M þH] þ : 699.5363;
1082 G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085

HPLC–MS–MS [MþH] þ : 699.6; 681.5 (86), 317.2 (100), 331.3 (26);


̄
HPLC–MS–MS [M  H]: 697.6; 655.4 (44), 315.1 (100).
Compound 14: 3-(N-suberoyl argininyl) marinobufagin (Mari-
nobufotoxin). Molecular formula: C38H56N4O9; QTOF–MS–MS
(40 eV) [MþH] þ : 713.4387 (100), 695.4443 (4), 331.2131 (56),
278.1512 (10), 175.1186 (5); HPLC–MS–MS [MþH] þ : 713.7; 695.6
(75), 331.3 (100), 278.0 (12); HPLC–MS–MS [M  H]-: 711.5; 669.4
(45), 329.2 (1 0 0), 287.1 (24), 270.1 (9). 1H NMR: δ 7.92 (dd, J ¼10.0
and 2.8 Hz, H-22), δ 7.48 (d, J ¼2.0 Hz, H-21), δ 6.30 (d, J¼ 10.0 Hz,
H-23), δ 5.16 (br s, NH), δ 4.30 (dd, J ¼6.8, 5.2 Hz, H-3), δ 3.64 (s,
H-15), δ 2.61 (d, J¼ 10.4, H-17), Diacid –CH2- δ 2.41 m, 2.34-2.36
(brt, J ¼7.2), δ 2.27 (t, J ¼7., H.4), δ 1.40-1.68 (m), δ 1.16-1.38 (m),
Arginine δ 3.20-3.25 m; 1.01 s (H-19), 0.81 s (H-18).
Compound 15: 3-(N-azelayl argininyl) marinobufagin. Molecu-
lar formula: C39H58N4O9; HPLC–MS–MS [M þH] þ : 727.7; 709.5
̄
(40), 345.3 (100), 343.3 (25); HPLC–MS–MS [M  H]: 725.4 683.4
(55), 343.1 (100), 684.3 (51).
Compound 16: 3-(N-sebacyl argininyl) marinobufagin. Molecu-
lar formula: C40H60N4O9; QTOF MS–MS (40 eV) [Mþ H] þ : 741.3877
(55), 723.4843 (16), 359.2509 (100), 331.1955 (32), 175.1378 (33).
HPLC–MS–MS [Mþ H] þ : 741.7; 723.6 (54), 359.3 (100), 278.1 (11).
̄
HPLC–MS–MS [M  H]: 739.4; 697.6 (90), 357.2 (100), 315.1 (15),
297.0 (30).
Compound 17: 3-(N-undecadienoyl argininyl) marinobufagin.
Molecular formula: C41H62N4O9; HPLC–MS–MS [MþH] þ : 755.7;
737.6 (77), 373.4 (100).
Compound 18: 3-(N-dodecadienoyl argininyl) marinobufagin.
Molecular formula: C42H64N4O9; HPLC–MS–MS [Mþ H] þ : 769.7;
Fig. 2. Compounds identified in Rhinella (Bufo) schneideri toad venom. 751.7 (54), 387.4 (100), 306.3 (11).

O 3.3.4. Telocinobufagin argininyl diacid derivatives


O Compound 19: Telocinobufagin. HPLC–MS–MS [Mþ H] þ : 403.2;
349.2 (100), 367.2 (42), 350.2 (25), 271.0 (7).
Bufalin argininyl diacids Compound 20: 3-(N-pimeloyl argininyl) telocinobufagin. Mole-
̄
cular formula: C37H56N4O9; HPLC–MS–MS [M  H]: 699.5; 657.4
n Diacid
OH (97), 315.0 (100), 655.3 (44), 272.8 (55). HPLC–MS–MS [M þH] þ :
O 701.5; 683.6 (95), 317.2 (100).
H 7 6 suberic acid
n(H2C) O 8 7 azelaic acid
Compound 21: 3-(N-suberoyl argininyl) telocinobufagin (Telo-
HN NH2 cinobufatoxin). Molecular formula: C38H58N4O9; HPLC–MS–MS
HN O 9 8 sebacic acid ̄
HN [M H]: 713.5; 671.4 (54), 329.1 (100), 287.0 (21), 268.9 (22);
COOH
HPLC–MS–MS [MþH] þ : 715.7; 697.6 (96), 331.3 (100), 278.1 (19).
O Marinobufagin argininyl Compound 22: 3-(N-sebacyl argininyl) telocinobufagin. Mole-
̄
O diacids cular formula: C40H62N4O9; HPLC–MS–MS [M H]: 741.5; 357.1
(100), 699.5 (20), 340.2 (6), 315.1 (27), 297.1 (9).
n Diacid
11 3 glutaric acid
12 4 adipic acid
3.3.5. Hellebrigenin argininyl diacid derivatives
O
13 5 pimelic acid Compound 23a: 3-(N-suberoyl argininyl) hellebrigenin. Mole-
O ̄
OH
14 6 suberic acid cular formula: C39H60N4O9; HPLC–MS–MS [M  H]: 727.6; 685.5
n(H2C) O
(100), 329.1 (31), 268.9 (13).
15 7 azelaic acid
HN NH2
HN O 16 8 sebacic acid
HN 17 9 undecadienoic acid
COOH
18 10 dodecadienoic acid 3.3.6. Alkaloids
Compound 24. Bufotenin. HPLC–MS–MS [M þH] þ : 204.8; 159.6
(100), 57.4 (44); QTOF–MS–MS: [M þH] þ : 205.1263; 205 (100),
O
160 (100).
O
Compound 25: Bufotenidin (BTD). HPLC–MS–MS [Mþ H] þ : 219
Telocinobufagin argininyl
(100), 160 (66).
diacids
Compound 26: Dehydrobufotenin. QTOF–MS–MS: [M þH] þ :
203.1227; 203 (55), 188 (100), 173 (6).
OH
O n Diacid
OH
20 5 pimelic acid
n(H2C) O 3.3.7. Polihydroxy argininyl diacid derivatives
21 6 suberic acid
HN NH2 Compound A. 3-(N-suberoyl argininyl)-hydroxymarinobufagin.
HN O 22 8 sebacic acid
HN Molecular formula: C38H56N4O10, QTOF–MS–MS (40 eV) [M þH] þ :
COOH
729.4277 (51), 711.4509 (18), 345.2379 (33), 331.2043 (100),
Fig. 3. Argininyl diacid derivatives from Rhinella schneideri bufadienolides. 278.1674 (16), 175.1122 (8).
G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085 1083

Compound B. 3-(N-suberoyl argininyl)-hydroxytelocinobufagin. In the Chinese crude drug Ch’an Su, the HPLC pattern of the
Molecular formula: C38H58N4O10, QTOF–MS–MS (45 eV) [MþH] þ : venom shows three differential zones, characterized by the elution of
731.4405 (91), 713.4128 (10), 331.2131 (100), 278.1674 (20), suberoyl arginine in the first third of the chromatogram, followed by
175.1186 (13). the bufadienolide genines with different oxidation patterns and the
Compound C. 3-(N-suberoyl argininyl)-dihydroxymarinobufa- diacid esters of bufadienolides at the end of the chromatographic
gin. Molecular formula: C38H56N4O11, QTOF–MS–MS (45 eV): run. In the Pantanal toad, the simple alkaloids and argininyl diesters
745.4355 (44), 727.4439 (14), 379.2471 [MþH  3H2O] (5), elutes first, followed by the argininyl diacid derivatives of the
331.2043 (100), 278.1674 (17), 250.1729 (9), 175.1314 (11). bufadienolides bufalin, marinobufagin and telocinobufagin. Liquid
chromatography/electrospray ionization tandem mass spectrometry
3.4. Bufadienolides and alkaloids in Rhinella schneideri venom is an efficient method for the analysis of constituents in the Chinese
crude drug Venenum Bufonis (Hu et al., 2011) as well as to compare
From the parotid gland venom of the Brazilian Pantanal toad the composition of different Bufo (Rhinella) venoms (Gao et al., 2010)
Rhinella schneideri, 29 compounds were identified or tentatively or for dosage methods of marinobufagenin in biofluids (Lenaerts et
identified by a combination of spectroscopic and spectrometric al., 2013). In the Chinese (Chan Su) and Japanese (Sen So) crude toad
means. The venom showed as main compounds the bufadieno- venom, large variation in the oxidation pattern of the bufadienoides
lides 7, marinobufagin 10 and marinobufotoxin 14 but also several was observed. In the Chinese crude drug (Gao et al., 2010), sulfate,
minor compounds differing in the homologous diacid side chain acetate and C8 diacids (suberoyl) were the main esters of the
linking arginine and the bufadienolide as well as in the number of bufadienolides. The composition of the Brazilian Rhinella schneideri
hydroxy groups in the steroidal moiety. Kamano et al. (1968) venom show differences with that of the Chinese crude drug Ch'an
described marinobufagin 10 from the crude drug Ch'an Su. Su in the identity of the minor components as well as in the
Compound 14 was previously reported by Linde-Tempel (1970) oxidation patterns of the main compounds.
and Shimada et al. (1976). Compounds 10 and 14 were synthesized Looking for novel bufadienolides with relevant biological
by Pettit, Kamano (1974). A comprehensive 1H and 13C NMR activities, Ye et al. (2004, 2005) used the fungi Mucor spinosus
chemical shift assignment for bufadienolides from the Chinese and Mucor polymorphosporus to modify bufalin and resibufogenin,
drug Ch'an Su was reported (Kamano et al., 2001). Shimada et al. respectively. The microbial oxidation products were assessed for
(1977) pointed out to the possible occurrence of other homologous cytotoxicity and the authors suggest that 3-OH glycosylation or
diacid side chain linking arginine and marinobufagin in the living hydroxylation at C-1β or C-12β might be relevant to afford more
animals. In a careful study of the crude drug, Shimada et al. (1977) polar bufadienolides with higher cytotoxicity.
reported the occurrence of several diacid conjugates of arginine, The antimicrobial effect of the bufadienolides telocinobufagin
including the 3-suberoyl arginine ester of bufogenin and the and marinobufagin was published by Cunha Filho et al. (2005). The
3-pimeloyl, 3-adipoyl and 3-succinoyl arginine esters of bufalin. compounds were characterized in the parotid secretion from Bufo
The related compound bufalin 3-succinoylarginine ester was rubescens (Rhinella rubescens) after purification by RP-HPLC and
isolated from the skin of the Japanese toad Bufo vulgaris formosus QTOF and NMR analysis. Both compounds presented antimicrobial
Boulenger (Shimada et al., 1975). In a study undertaken with the effect with MIC of 128 mg/mL against Staphylococcus aureus ATCC
dried body of the Korean toad Bufo bufo gargarizans Cantor, 25213 and MICs of 64 and 16 mg/mL towards Escherichia coli ATCC
Shimada et al. (1985) described 3-succinyl, 3-adipoyl, 3-pimeloyl 25922, respectively. The MICs for the reference compounds amox-
and 3-suberoyl arginine esters of cinobufagin. Compounds 3, 6 and icillin, iminipenem and trimethoprim against Escherichia coli were
10 were reported as constituents of the Chinese crude drug o22, 14 and 40 mg/mL, respectively. Trimetoprim, used as refer-
Chansu (Gao et al., 2010). The alkaloids bufotenin (24), bufotenidin ence against Staphylococcus aureus presented an MIC o160 mg/mL.
(25) and dehydrobufotenin (26) were previously identified in the Both compounds show an interesting effect against Escherichia coli.
parotid glands secretions of Bufo (Rhinella) schneideri and several Furthermore, bufadienolides from Rhinella jimi were assessed for
Brazilian Rhinella (Bufo) species by Maciel et al. (2003). trypanocidal effect against Trypanosoma cruzi trypomastigotes and
Three additional suberoyl argininyl esters of hydroxymarino- Leishmania chagasi promastigotes (Tempone et al., 2008). The
bufagin, hydroxytelocinobufagin and dihydroxytelocinobufagin, compounds were isolated from the toad venom by HPLC and were
namely compounds A, B and C, were tentatively identified based identified by mass spectrometry and NMR spectroscopy analysis.
on QTOF–MS–MS analysis. The exact position of the hydroxy group The bufadienolides telocinobufagin and hellebrigenin did not
of the three minor compounds, remains to be established. induced hemolysis up to 200 mg/mL and reduced growth of

Table 2
Antiproliferative activity of Rhinella schneideri crude extract and fractions expressed as IC50 values (mg/mL 7 SEM).

a b c d e
MRC-5 AGS SK-MES-1 J82 HL-60

Crude 0.0417 0.003 0.0767 0.004 0.093 7 0.065 0.086 7 0.004 0.1027 0.005
Fr. 1-3 0.1267 0.008 0.1837 0.007 0.1947 0.009 0.1347 0.008 0.1737 0.011
Fr. 4-5 0.2357 0.016 0.3127 0.018 0.3537 0.021 0.2457 0.017 0.5217 0.026
Fr. 6-8 0.1007 0.005 0.1677 0.110 0.2107 0.015 0.1837 0.011 0.234 7 0.014
Fr. 9-11 0.022 7 0.001 0.0407 0.002 0.0647 0.003 0.056 7 0.004 0.054 7 0.002
Fr. 12-13 0.0197 0.001 0.0357 0.002 0.028 7 0.005 0.0427 0.003 0.0447 0.002
Fr. 14-21 0.026 7 0.001 0.0427 0.002 0.082 7 0.003 0.0777 0.005 0.052 7 0.003
Etoposide 2.296 7 0.115 0.2777 0.011 1.295 7 0.078 1.884 7 0.075 1.059 7 0.002
Camptothecin 0.0026 7 0.0003 0.00777 0.0006 0.0059 7 0.0004 0.00217 0.0002 0.00457 0.0004
Vinblastine 0.0025 7 0.0003 0.0025 7 0.0001 0.00137 0.0001 0.0081 7 0.0006 0.00037 0.00002

a
Normal lung MRC-5 fibroblasts.
b
Human epithelial gastric cells AGS.
c
SK-MES-1 lung cancer cells.
d
J82 bladder carcinoma cells
e
Promyelocytic leukemia HL-60 cells. Each concentration was tested in quadruplicate and repeated three times in separate experiments.
1084 G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085

Leishmania chagasi with IC50 of 61.21 and 126.2 mg/mL, respec- analysis and assessed for cytotoxicity. According to the authors,
tively. The latter activity is not related to the ethnopharmacologi- Rhaebo guttatus yielded only marinobufagin. The antiprolifera-
cal data on the “cururu” frog. tive effect of five naturally occurring bufadienolides from the
parotid gland secretion of Rhinella schneideri (captured near
3.5. Antiproliferative activity Brasilia) was reported by Cunha-Filho et al. (2010). The authors
prepared several semisynthetic derivatives and found that all
The crude venom and the CC fractions of the venom were natural and synthetic compounds presented moderate to strong
assessed for antiproliferative effect using the MTT reduction assay activity towards human HL-60, SF-295, MDA-MB-435 and HCT-8
in five different human cell lines: normal lung fibroblasts (MRC-5), cancer cells.
human epithelial gastric (AGS), lung cancer (SK-MES-1), bladder
carcinoma (J82) and promyelocytic leukemia (HL-60) cells. Etopo-
side, camptothecin and vinblastine were used as reference anti- 4. Conclusions
proliferative compounds. The concentrations of the extract/
fractions inhibiting cell growth by 50% (IC50 values, mg/mL) were Our study demonstrated that argininyl diacid esters of bufa-
obtained adjusting the dose-response curves to a sigmoidal model. dienolides are the main constituents of the Rhinella schneideri
The results are summarized in Table 2. venom and that the most active antiproliferative compounds are
The crude venom showed a strong antiproliferative effect bufadienolides. Marinobufagin was associated with higher activity
towards all the cell lines, with IC50 values ranging from 0.041 to but the presence of alkaloids as minor constituents in the same
0.102 mg/mL. After chromatography, the fractions 1–3, 4 and 5 and fractions might play a role in the effect. The strong antiprolifera-
6–8 presented less effect than the crude venom. The fractions 9– tive activity of Rhinella schneideri crude venom extract and frac-
13, however, showed a remarkable antiproliferative effect, with tions, associated with the identification of constituents, supports
IC50 values in the range 0.019–0.022, 0.035–0.040, 0.028–0.064, the traditional claim of using this toad to treat cancer. However,
0.042–0.056 and 0.044–0.052 mg/mL for MRC-5, AGS, SK-MES-1, further pharmacological studies are required to disclose the
J82 and HL-60 cell lines, respectively. Under the same experi- potential of the “cururu” toad venom in antitumor therapy. The
mental conditions, IC50 values (in mg/mL) of etoposide towards the new compounds detected and identified in the venom pointed out
different cell lines were: 2.296 (MRC-5), 0.277 (AGS), 1.295 (SK- to the relevance of new investigations on South American amphi-
MES-1), 1.884 (J82) and 1.059 (HL-60). Camptothecin and vinblas- bians, using updated techniques and combining spectroscopic and
tine were more active with IC50 values in the range of 0.0021– spectrometric methods with working hypothesis based on tradi-
0.0077 and 0.0003–0.0081 mg/mL, respectively. In spite of relevant tional uses of natural resources.
antiproliferative effect, no selectivity is observed for the toad
venom, fractions or reference compounds. While the less effective
fractions 1–3 and 4 and 5 contained mainly the higher molecular Acknowledgments
weight argininyl diesters of bufadienolides, the most active frac-
tions 9–13 and 14–21 yielded marinobufagin 10 as the main We are grateful to Conselho Nacional de Desenvolvimento Cientí-
compound with alkaloids 24 and 26 as minor constituents. fico e Tecnológico (CNPq-Brazil) and FONDECYT No. 1110054 (Chile)
The aqueous extract of the skin and parotid glands of Bufo bufo for financial support.
gargarizans was shown to inhibit cell proliferation of the liver
carcinoma cell lines HepG2 and Bel-7402 (Qi et al., 2010). The
aqueous extract, as used in traditional medicine, induces apoptosis References
through a disruption of the mitochondrial membrane potential.
However, in this report, the concentration of solubles is not given. Alves, R.R.N., Alves, H.N., 2011. The faunal drugstore: animal-based remedies used
in traditional medicines in Latin America. Journal of Ethnobiology and Ethno-
The total crude drug (20 g) to the final volume after concentration medicine 7 (9), 43.
to 1 ml was referred as 20 g crude drug/mL for the experiments (Qi Alves, R.R.N., Neta, R.O., de, S., Trovao, D.M., de, B., Barbosa, J.E., de, L., Barros, A.T.,
et al., 2010). When expressed as mg crude drug/mL, IC50 values for Dias, T.L.P., 2012. Traditional uses of medicinal animals in the semi-arid región
of Brazil. Journal of Ethnobiology and Ethnomedicine 8 (41), 7.
HepG2 cells at 24, 48 and 72 h were 0.20, 0.06 and 0.02 mg/mL,
Anjolette, F.A.P., Pereira, J.C., Azzolini, A.E.C., Sampaio, S.V., Assis-Pandochi, A.I.,
respectively while for Bel-7402 cells, the values were 0.15, 0.06 Arantes, E.C., Pereira-Crott, L.S., 2011. Complement system modulation by
and 0.02 mg/mL, respectively. No reference compound was used in components from Rhinella schneideri poison. Molecular Immunology 48 (14),
this study. Further work on the crude drug and the main active 1694–1695.
Baldo, M.A., Liberato, J.L., Godoy, L.D., Ferreira dos Santos, W., Arantes, E.C., 2012a.
bufadienolides from the crude drug (Qi et al., 2011) showed a New perspective for therapy against seizures using molecules from Rhinella
relevant apoptosis-inducing effect of cinobufacini with the follow- schneideri toad poison. Toxicon 60 (2), 103–104.
ing IC50 values (in parentheses): human leukemia U937 (1.36 mg/ Baldo, E.C.F., da Silva, C.P., Sampaio, S.V., Arantes, E.C., 2012b. Evaluation of the
cytotoxic activity of Rhinella schneideri toad poison on tumor cells and on
mL, 24 h), human leukemia HL-60 (0.25 mg/mL, 48 h), human healthy mononuclear cells. Toxicon 60, 103.
hepatocellular carcinoma Bel-7402 and HepG2 (80 mg/mL, 48 h). Bisset, N.G., 1989. Arrow and dart poisons. Journal of Ethnopharmacology 25, 1–41.
However, all the structures of bufadienolides shown in the report Cunha Filho, G.A., Schwartz, C.A., Resck, I.S., Murta, M.M., Lemos, S.S., Castro, M.S.,
Kyaw, C., Pires Jr., O.R., Leite, J.R.S., Bloch Jr., C., Schwartz, E.F., 2005. Anti-
of Qi et al. (2011) are wrong. microbial activity of the bufadienolides marinobufagin and telocinobufagin
Giri et al. (2006) reported the cytotoxic, antiproliferative and isolated as major components from skin secretion of the toad Bufo rubescens.
apoptosis-inducing effect of extracts from the Indian toad Bufo Toxicon 45, 777–782.
Cunha-Filho, G.A., Resck, I.S., Cavalcanti, B.C., Pessoa, C.Ó., Moraes, M.O., Ferreira, J.R.
melanostictus using human myeloid leukemia U937 and K562 O., Rodrigues, F.A.R., dos Santos, M.L., 2010. Cytotoxic profile of natural and
cells. The toad skin was extracted with methanol, with 1.3% w/w some modified bufadienolides from toad Rhinella schneideri parotoid gland
yield. The extract induces cell arrest at the G1 phase of the cell secretion. Toxicon 56, 339–348.
Daly, J.W., 1998. The nature and origin of amphibian alkaloids. In: Cordell, G.A. (Ed.),
cycle and showed an IC50 value of 55.0–66.2 mg/mL for U937 and
The Alkaloids: Chemistry and Biology. Academic Press, New York, pp. 141–169.
81.1–92.2 mg/mL for K562 cells, respectively. The antiprolifera- Daly, J.W., Garraffo, H.M., Spande, T.F., Yeh, H.J.C., Peltzer, P.M., Cacivio, P.M., Baldo, J.
tive effect of the Brazilian Rhinella marina and Rhaebo guttatus D., Faivovich, J., 2008. Indolizidine 239Q and quinolizidine 275I. Major alkaloids
venom was reported by Ferreira et al. (2013). From Rhinella in two Argentinian bufonid toads (Melanophryniscus). Toxicon 52, 858–870.
Dictionary of Natural Products on DVD (2013), CRC Press, Boca Raton, FL, USA.
marina, the bufadienolides telocinobufagin, marinobufagin, Ferreira, P.M.P., Lima, D.J.B., Debiasi, B.W., Soares, B.M., Machado, K., da, C., Noronha,
bufalin and resinobufogenin were identified by HPLC-MS J., da, C., Rodrigues, D. de J., Sinhorin, A.P., Pessoa, C., Vieira Jr., G.M., 2013.
G. Schmeda-Hirschmann et al. / Journal of Ethnopharmacology 155 (2014) 1076–1085 1085

Antiproliferative activity of Rhinella marina and Rhaebo guttatus venom extracts Cantor, induces apoptosis through a mitochondria-mediated pathway in
from Southern Amazon. Toxicon 72, 43–51. human hepatocellular carcinoma cells. Journal of Ethnopharmacology 128,
Gao, H., Zehl, M., Leitner, A., Wu, X., Wang, Z., Kopp., B., 2010. Comparison of toad 654–661.
venoms from different Bufo species by HPLC and LC-DAD-MS/MS. Journal of Qi, F., Li, A., Inagaki, Y., Kokudo, N., Tamura, S., Nakata, M., Tang, W., 2011. Antitumor
Ethnopharmacology 131, 368–376. activity of extracts and compounds from the skin of the toad Bufo bufo
Giri, B., Gomes, A., Debnath, A., Saha, A., Biswas, A.K., Dasgupta, S.C., Gomes, A., gargarizans Cantor. International Immunopharmacology 11, 342–349.
2006. Antiproliferative, cytotoxic and apoptogenic activity of Indian toad (Bufo Rodriguez, J.A., Haun, M., 1999. Cytotoxicity of trans-dehydrocrotonin from Croton
melanostictus, Schneider) skin extract on U937 and K562 cells. Toxicon 48, cajucara on V79 cells and rat hepatocytes. Planta Medica 65, 522–526.
388–400. Sciani, J.M., de-Sá-Júnior, P.L., Ferreira, A.K., Pereira, A., Antoniazzi, M.M., Jared, C.,
Hu, Y., Yu, Z., Yang, Z.J., Zhu, G., Fond, W., 2011. Comprehensive chemical analysis of Pimenta, D.C., 2013. Cytotoxic and antiproliferative effects of crude amphibian
Venenum Bufonis by using liquid chromatography/electrospray ionization skin secretions on breast tumor cells. Biomedicine & Preventive Nutrition 3,
tandem mass spectrometry. Journal of Pharmaceutical and Biomedical Analysis 10–18.
56, 210–220. Shimada, K., Ro, J.S., Ohishi, K., Nambara, T., 1985. Isolation and characterization of
Kamano, Y., Nogawa, T., Yamashita, A., Pettit, G.R., 2001. The 1H and 13C NMR cinobufagin 3-glutaroyl-L-arginine ester from Bufo bufo gargarizans Cantor.
chemical shift assignments for thirteen bufadienolides isolated from the Chemical and Pharmaceutical Bulletin 33, 2767–2771.
traditional Chinese drug Ch'an Su. Collection of Czechoslovak Chemical Com- Shimada, K., Fujii, Y., Niizaki, Y., Nambara, T., 1975. Isolation of gamabufotalin 3-
munications 66, 1841–1848. pimeloylarginine ester from the skin of Japanese toad. Tetrahedron Letters 9,
Kamano, Y., Yamamoto, H., Hatayama, K., Tanaka, Y., Shinohara, M., Komatsu, M.,
653–654.
1968. The isolation and structure of new bufadienolide, resibufagin and the
Shimada, K., Sato, Y., Fujii, Y., Nambara, T., 1976. Occurrence of bufalitoxin,
isolation of marinobufagin. Tetrahedron Letters 54, 5669–5672.
cinobufotoxin and their homologs in Japanese toad. Chemical and Pharmaceu-
Lenaerts, C., Demeyer, M., Gerbaux, P., Blankert, B., 2013. Analytical aspects of
tical Bulletin 24, 1118–1120.
marinobufagenin. Clinica Chimica Acta 421, 193–201.
Shimada, K., Fujii, Y., Yamashita, E., Niizaki, Y., Sato, Y., Nambara, T., 1977. Studies on
Linde-Tempel, H.O., 1970. Konstitution der Bufotoxine. Über Krötengifte, 38.
cardiotonic steroids from the skin of Japanese toad. Chemical and Pharmaceu-
Mitteilung. Helvetica Chimica Acta 53, 2188–2196.
tical Bulletin 25, 714–730.
Ma, H.Y., Kou, J.P., Wang, J.R., Yu, B.Y., 2007. Evaluation of the anti-inflammatory and
Tempone, A.G., Pimenta, D.C., Lebrun, I., Sartorelli, P., Taniwaki, N.N., de Andrade Jr.,
analgesic activities of Liu-Shen-Wan and its individual fractions. Journal of
Ethnopharmacology 112, 108–114. H.F., Antoniazzi, M.M., Jared, C., 2008. Antileishmanial and antitrypanosomal
Maciel, N.M., Schwartz, C.A., Pires Júnior, O.R., Sebben, A., Castro, M.S., Sousa, M.V., activity of bufadienolides isolated from the toad Rhinella jimi parotoid macrog-
Fontes, W., Schwartz, E.N.F., 2003. Composition of indolealkylamines of Bufo land secretion. Toxicon 52, 13–21.
rubescens cutaneous secretions compared to six other Brazilian bufonids with The State Pharmacopoeia Commission of People´s Republic of China, 2005.
phylogenetic implications. Comparative Biochemistry and Physiology Part B: Pharmacopoeia of the People´s Republic of China, Beijing. China Chemical
Biochemistry and Molecular Biology 134, 641–649. Industry Press, 265–266.
Maciel, N.M., Collevatti, R.G., Colli, G.R., Schwartz, E.F., 2010. Late Miocene Wassen, H., 1934a. The frog motive among the South American Indians. Anthropos
diversification and phylogenetic relationships of the huge toads in the Rhinella 29, 319–370.
marina (Linnaeus, 1758) species group (Anura: Bufonidae). Molecular Phyloge- Wassen, H., 1934b. The frog in Indian mythology and imaginative world. Anthropos
netics and Evolution 57, 787–797. 29, 613–658.
Meng, Z., Yang, P., Shen, Y., Bei, W., Zhang, Y., Ge, Y., Newman, R.A., Cohen, L., Liu, L., Weil, A.T., Davis, W., 1994. Bufo alvarius: a potent hallucinogen of animal origin.
Thornton, B., Chang, D.Z., Liao, Z., Kurzrock, R., 2009. Pilot study of Huachansu Journal of Ethnopharmacology 41, 1–8.
in patients with hepatocellular carcinoma, nonsmall-cell lung cancer, or Ye, M., Han, J., An, D., Tu, G., Guo, D., 2005. New cytotoxic bufadienolides from the
pancreatic cancer. Cancer 115, 5309–5318. biotransformation of resibufogenin by Mucor polymorphosporus. Tetrahedron
Pettit, G.R., Kamano, Y., 1974. Bufadienolides. 28. Marinobufotoxin. Journal of 61, 8947–8955.
Organic Chemistry 39, 3003–3006. Ye, M., Qu, G., Guo, H., Guo, D., 2004. Novel cytotoxic bufadienolides derived from
Qi, F., Li, A., Zhao, L., Xu, H., Inagaki, Y., Wang, D., Cui, X., Gao, B., Kokudo, N., Nakata, bufalin by microbial hydroxylation and their structure-activity relationships.
M., Tang, W., 2010. Cinobufacini, an aqueous extract from Bufo bufo gargarizans Journal of Steroid Biochemistry & Molecular Biology 91, 87–98.

Das könnte Ihnen auch gefallen