Sie sind auf Seite 1von 43

CLINICAL MICROBIOLOGY REVIEWS, Apr. 1999, p. 243–285 Vol. 12, No.

2
0893-8512/99/$04.0010
Copyright © 1999, American Society for Microbiology. All Rights Reserved.

Molecular Techniques for Detection, Species Differentiation,


and Phylogenetic Analysis of Microsporidia
CASPAR FRANZEN* AND ANDREAS MÜLLER
Department of Internal Medicine I, University of Cologne, 50924 Cologne, Germany

INTRODUCTION .......................................................................................................................................................244
MORPHOLOGY OF MICROSPORIDIA ...............................................................................................................244
Morphology and Life Cycle ...................................................................................................................................244
Spore.....................................................................................................................................................................244
Merogony..............................................................................................................................................................244
Sporogony.............................................................................................................................................................245

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


TAXONOMY ...............................................................................................................................................................245
GENUS- AND SPECIES-SPECIFIC CHARACTERISTICS .................................................................................247
Enterocytozoon sp. ....................................................................................................................................................247
Encephalitozoon spp.................................................................................................................................................248
Nosema spp. .............................................................................................................................................................249
Vittaforma sp. ...........................................................................................................................................................250
Pleistophora spp. and Trachipleistophora spp.......................................................................................................250
Other Genera...........................................................................................................................................................251
EPIDEMIOLOGY .......................................................................................................................................................251
Prevalence and Geographic Distribution ............................................................................................................251
Sources of Infection and Transmission ...............................................................................................................251
CLINICAL MANIFESTATIONS ..............................................................................................................................254
Gastrointestinal and Biliary Tract Infections.....................................................................................................254
Enterocytozoon bieneusi........................................................................................................................................254
Encephalitozoon spp.............................................................................................................................................255
Other species .......................................................................................................................................................255
Hepatitis, Pancreatitis, and Peritonitis ...............................................................................................................255
Ocular Infections ....................................................................................................................................................255
Encephalitozoon spp.............................................................................................................................................255
Other species .......................................................................................................................................................255
Sinusitis....................................................................................................................................................................256
Pulmonary Infections .............................................................................................................................................256
Urinary Tract Infections ........................................................................................................................................256
Myositis ....................................................................................................................................................................256
Cerebral Infections .................................................................................................................................................256
Encephalitozoon spp.............................................................................................................................................256
Other species .......................................................................................................................................................256
Rare Manifestations ...............................................................................................................................................256
Urethritis..............................................................................................................................................................256
Prostatic abscess .................................................................................................................................................256
Tongue ulcer ........................................................................................................................................................257
Skeletal involvement...........................................................................................................................................257
Cutaneous microsporidiosis ..............................................................................................................................257
Systemic Infections .................................................................................................................................................257
Encephalitozoon spp.............................................................................................................................................257
Other species .......................................................................................................................................................257
THERAPY ....................................................................................................................................................................257
DIAGNOSTIC METHODS........................................................................................................................................258
Transmission Electron Microscopy ......................................................................................................................258
Light Microscopy ....................................................................................................................................................258
Cytologic diagnosis and stool examination .....................................................................................................259
Histologic diagnosis............................................................................................................................................260
Cell Culture .............................................................................................................................................................261
Animal Models ........................................................................................................................................................261

* Corresponding author. Mailing address: Department of Internal


Medicine I, University of Cologne, Joseph-Stelzmann Str. 9, D-50924
Cologne, Germany. Phone: 49-(0)221-478-4433. Fax: 49-(0)221-478-
6456. E-mail: Caspar.Franzen@Uni-Koeln.de.

243
244 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

Antigen-Based Methods .........................................................................................................................................261


Serologic Testing.....................................................................................................................................................262
MOLECULAR METHODS .......................................................................................................................................262
Small- and Large-Subunit rRNA Genes of Microsporidia ...............................................................................262
a- and b-Tubulin Genes of Microsporidia .........................................................................................................263
Other Genes of Microsporidia ..............................................................................................................................264
DNA Isolation Techniques.....................................................................................................................................265
Molecular Techniques for Diagnosis and Species Differentiation ...................................................................265
Primer pairs and hybridization probes for E. bieneusi..................................................................................265
Primer pairs and hybridization probes for E. intestinalis .............................................................................268
Primer pairs for E. hellem and E. cuniculi ......................................................................................................269
General primer pairs and hybridization probes for several microsporidian species ...............................269
Strain differentiation of Encephalitozoon spp. and E. bieneusi......................................................................270
Comparison of molecular techniques with light microscopy........................................................................271
Molecular Techniques for Phylogenetic Analysis...............................................................................................272
Molecular Techniques for Susceptibility Testing ...............................................................................................275

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


FUTURE TRENDS .....................................................................................................................................................275
CONCLUDING REMARKS ......................................................................................................................................275
ACKNOWLEDGMENTS ...........................................................................................................................................276
REFERENCES ............................................................................................................................................................276

INTRODUCTION field of clinical microbiology to incorporate these techniques


(378). In this paper, we review human microsporidial infec-
Microsporidia are obligate intracellular protozoan parasites tions and newly developed molecular techniques for detection,
that infect a broad range of vertebrates and invertebrates. In species differentiation, and phylogenetic analysis of microspo-
1857 these parasites were first recognized as pathogens in ridia with special emphasis on species that infect humans.
silkworms (254), and long before they were described as hu-
man pathogens, they were recognized as a cause of disease in MORPHOLOGY OF MICROSPORIDIA
many nonhuman hosts including insects, mammals, and fish
All microsporidia are obligate intracellular parasites and
(39, 50, 51, 56). Therefore, they are responsible for consider-
have no active stages outside their host cells. They are consid-
able infectious disease problems in industries such as fisheries
ered to be ancient organisms, evolutionarily placed as an early
and silk production (39, 50, 56). The first human case of mi-
branch leading from prokaryotes to eukaryotes (64, 65, 285,
crosporidial infection was reported in 1959 (237), and only 10
362, 363). Microsporidia lack some typical eukaryotic charac-
well-documented human infections with microsporidia were
teristics. The ribosomes (70S), ribosomal subunits (30S and
described until 1985, when a new species, Enterocytozoon bie-
50S), and rRNAs (16S and 23S) are of prokaryotic size, and the
neusi, was found in a human immunodeficiency virus (HIV)-
rRNA has no separate 5.8S rRNA (362). Although mitochon-
infected patient in France (99, 245). Since then, many infec-
dria, peroxisomes, and a classical stacked Golgi apparatus are
tions with microsporidia have been reported from all over the
missing, they are true eukaryotes with a nucleus, an intracyto-
world, and these parasites are now recognized as one of the
plasmatic membrane system, and chromosome separation by
most common pathogens in HIV-infected patients (7, 36–38,
mitotic spindles (362, 363); polyadenylation occurs on mRNA
53, 55, 56, 60, 66, 72, 87, 88, 96, 124, 130, 131, 136, 142–144,
in microsporidia as in every other eukaryotic organism studied
162, 195, 204–208, 223, 225, 241, 246, 247, 253, 263, 265, 311,
to date (381a).
325, 338, 344, 371, 372, 375).
The term “microsporidia” is a nontaxonomic designation
commonly used for organisms belonging to the phylum Micro- Morphology and Life Cycle
spora. This phylum consists of over 100 genera with almost Spore. Microsporidian spores are between 1 and 20 mm long.
1,000 species. So far only six genera (Enterocytozoon, Enceph- Species that infect mammals are usually small, with diameters
alitozoon [including Septata], Pleistophora, Trachipleistophora, of 1 to 3 mm. The spores have a thick wall, composed of three
Vittaforma, and Nosema) with at least 12 different species be- layers: (i) an electron-dense outer layer called the exospore,
longing to these six genera as well as unclassified microsporidia which is proteinaceous; (ii) an electron-lucent inner layer
have been described as pathogens in humans. called the endospore, which is chitinous; and (iii) a plasma
For most patients with infectious diseases, microbiological membrane enclosing the cytoplasm, the nucleus (sometimes
isolation and identification techniques offer the most rapid and two nuclei), a posterior vacuole, the polaroplast membranes,
specific determination of the etiologic agent (378). This is not and the unique extrusion apparatus. The extrusion apparatus
a suitable procedure for microsporidia, which are obligate in- consists of a coiled polar filament and its anchoring disc, which
tracellular parasites requiring cell culture systems for growth. is characteristic of all microsporidia (Fig. 1). The number and
Visualization of organisms in cytologic smears, tissue sections, arrangement of coils of the polar filament vary among genera
or both is commonly used for diagnosis of infections with and species. Under appropriate conditions inside a suitable
microsporidia. However, ultrastructural analysis by transmis- host, the polar filament is discharged through the thin anterior
sion electron microscopy is usually necessary for exact species end of the spore, thereby penetrating a new host cell and
differentiation. This technique may lack sensitivity, and species inoculating the infective sporoplasm into the host cell (Fig. 1
differentiation can be missed. and 2) (41, 43, 50, 58).
During the last 10 years, the detection of infectious disease Merogony. In suitable host cells, the sporoplasms that are
agents has begun to use nucleic acid-based technologies. Di- released from the spores become meronts. Meronts are
agnosis of infection caused by parasitic organisms is the last rounded, irregular, or elongated simple cells with little differ-
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 245

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest

FIG. 1. Diagram of a microsporidian spore and representative life cycle (merogonic and sporogonic stages vary among different genera).

entiated cytoplasm, enclosed by a plasma membrane. Meronts may have isolated or diplokaryon nuclei. Some sporonts divide
may have isolated or diplokaryon nuclei. Inside the host cell, directly into sporoblasts by binary fission, whereas others be-
there is a phase of repeated divisions by binary or multiple come multinucleated plasmodial stages. Sporoblasts are ovoid
fissions called merogony. Nuclear division may occur without bodies that will mature to spores by synthesis of spore or-
cell division, resulting in multinucleated plasmodial forms (41, ganelles (Fig. 1) (41, 43, 50, 58).
43, 50, 58).
Sporogony. Meronts develop into sporonts, which are char- TAXONOMY
acterized by a dense surface coat. This surface coat later de-
velops into the exospore layer of the spore wall. Sporonts The term “microsporidia” is a nontaxonomic designation
multiply by binary or multiple fission and divide into sporo- commonly used for organisms belonging to the phylum Mi-
blasts that will finally develop into mature spores. Sporonts crospora, which is contained within the subkingdom Protozoa
246 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

intestinalis (10, 166), Trachipleistophora hominis (180), and T. antropophtera (356a). Comparing this taxonomic system with phylogenetic trees generated on the basis of DNA sequence data (see Fig. 13), it seems clear
FIG. 3. Taxonomy of microsporidia infecting humans, using the revised taxonomy of Sprague et al. from 1992 (337), modified in light of the new taxonomic classification of Vittaformae corneae (324), Encephalitozoon
FIG. 2. Giemsa stain of Nosema algerae spore with an extruded polar tube
and with the sporoplasm at the end of the tube. Giemsa stain. Magnification,
3640.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


(50, 337). In 1882 Balbiani classified these parasites as a sep-
arate group, “Microsporidies” (12). Before the middle of this
century, since knowledge of this group of organisms was frag-
mentary, classifications of microsporidia were necessarily sim-
ple and artificial. Subsequently, the taxonomy of microsporidia
has been subjected to several modifications. Major published
microsporidian classifications differ considerably in the char-
acteristics used to produce the major divisions within the mi-
crosporidia (337). Larsson (214) considered that many features
traditionally used for taxonomic systems (for example, the dip-
lokaryon, sporophorous vesicle, meiosis) have evolved inde-
pendently in several lineages, and therefore seemed not to be
useful for phylogenetic analysis. In his classification system,
based on differences in ultrastructural morphology, several
characters were subdivided into well-defined categories,
thereby creating a tree representing the phylogeny of the mi-
crosporidia (214). Weiser (376) based his classification only on
the nuclear condition of the spores (one nucleus in Pleistopho-
ridida or two nuclei in Nosematidida), whereas Issi (183) used
the spore morphology and developmental stages. Until re-
cently, the classification system of Sprague, proposed in 1977
and updated in 1982, was the most widely used (335, 336). In
this scheme the microsporidia were divided into two groups,
based on the presence or absence of a membrane surrounding
the parasites: the Pansporoblastina (membrane present) and
the Apansporoblastina (membrane absent). In systems devel-
oped during the last decade, it seems that differences in chro-
mosome cycles constitute the most fundamental basis for dis-
tinguishing taxa at the highest level (52, 337). Based on this
concept, Sprague et al. (337) proposed a comprehensive revi-
sion of the classification system in which differences in the
nuclear state and their implications for the chromosome cycle
were treated as the most fundamental taxonomic characters
(Fig. 3). The microsporidia were separated into the Dihaplo-
phasea, which have a diplokaryon in some phase of their life
that the system of Sprague et al. (337) is flawed.

cycle, and the Haplophasea, which have unpaired nuclei in all


stages of their life cycle. Phylogenetic trees constructed on the
basis of DNA sequence data now show clearly that this and
other classification systems do not reflect the true relationships
among microsporidia and that the classification of microspo-
ridia should be completely revised.
Nucleotide sequence data of the small-subunit (SSU) rRNA
of a microsporidian from insects, Vairimorpha necatrix, sug-
gested that microsporidia are very ancient organisms and that
the evolutionary developments leading to microsporidia
branched very early from those leading to eukaryotes (363).
DNA data for protein-encoding genes supported this thesis
(35, 64, 65, 186, 187, 387), but phylogenetic trees constructed
on the basis of a- and b-tubulin sequences suggested that the
microsporidia are close relatives of fungi, which may be
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 247

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


FIG. 4. Transmission electron micrograph of duodenal epithelium from an HIV-infected patient heavily parasitized with Enterocytozoon bieneusi. Several cells are
infected with merogonic and sporogonic stages, and one cell is infected with darkly staining spores. Magnification, 36,600.

evolved degeneratively from higher forms (125, 126, 221). Re- GENUS- AND SPECIES-SPECIFIC CHARACTERISTICS
cently, genes encoding Hsp70 (a heat shock protein or chap-
eronin) have been identified in the microsporidia Nosema Enterocytozoon sp.
locustae, V. necatrix, Encephalitozoon hellem, and Encephalito-
To date, there is only one member in the genus Enterocyto-
zoon cuniculi, and phylogenetic analyses have shown unequiv- zoon, Enterocytozoon bieneusi. This organism develops in direct
ocally that these genes are most closely related to those en- contact with the host cell cytoplasm. Meronts often have elec-
coding Hsp70 proteins from the mitochondria of other tron-lucent inclusions which are present throughout the life
eukaryotes, suggesting that microsporidia may be evolved de- cycle. Sporonts form electron-dense precursors of the polar
generatively from higher forms (159, 173, 193, 251a, 275a). tube and the anchoring disk, which develop before sporogonial
This possible degenerative evolution is discussed in more detail plasmodia divide into sporoblasts. Multiple sporoblasts are
below. formed by invagination of the plasma membrane of one large
SSU and large-subunit (LSU) rRNA and protein-encoding sporogonial plasmodium. Spores are oval and small, measuring
DNA sequences are now available for several microsporidian only 1.1 to 1.6 by 0.7 to 1.0 mm, with five to seven coils of the
species, including six species infecting humans. The taxonomy polar tubule, arranged in two rows (Fig. 4) (42, 53, 99, 100,
of microsporidia will be amended significantly in the near fu- 318).
ture when these and newly generated nucleotide sequence data E. bieneusi was first detected by Modigliani et al. (245) and
are considered for future classification systems. For example, described in detail by Desportes et al. (99) in 1985 following
molecular analyses have led to the reclassification of Septata examination of a 29-year-old Haitian AIDS patient with
intestinalis into Encephalitozoon intestinalis (10, 166). However, chronic diarrhea who lived in France. A similar case was de-
this reclassification is still controversial on the basis of ultra- scribed in the United States in the same year (119). Since then,
structural data and rules of taxonomy (49). Several phyloge- the number of reported cases has steadily increased in Europe,
netic trees based on DNA sequence data have been suggested North and South America, Africa, and Australia (7, 36, 53, 55,
recently (9–11, 125, 233, 280, 364, 365, 381, 396) and are 56, 60, 72, 83, 87, 96, 103, 124, 127, 130, 131, 136, 142, 244, 246,
discussed below. 263, 325, 389). The parasite usually infects intestinal entero-
248 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


FIG. 5. Transmission electron micrograph of duodenal epithelium from an HIV-infected patient infected with Encephalitozoon cuniculi. Two meronts with single
nuclei, four sporonts with a thickened plasma membrane and highly developed endoplasmatic reticulum, and four spores inside a parasitophorous vacuole are visible.
Magnification, 317,500.

cytes of HIV-infected patients but has been also detected in intranuclear location of the organism, it has been suggested
lamina propria cells of small-bowel biopsy specimens, biliary that in the absence of significant reasons for the suppression of
tree, gallbladder, liver cells, pancreatic duct, and tracheal, the generic name Nucleospora, the original name N. salmonis
bronchial, and nasal epithelia (93, 129, 165, 250, 278, 279, 310, rather than E. salmonis is valid (120).
367, 368).
The second and last member of the family Enterocytozooidae Encephalitozoon spp.
is Nucleospora salmonis. This microsporidium was originally
described by Hedrick et al. in 1991 (169), but shortly thereafter All Encephalitozoon species develop within parasitophorous
Chilmonczyk et al. (67) described it as Enterocytozoon salmo- vacuoles. Meronts divide by binary fission and usually remain
nis. Ultrastructural examinations showed morphological simi- in the vacular membrane. Sporonts develop a thick surface
larities between E. bieneusi and N. salmonis, with Nucleospora coat which becomes the exospore of spores, and the sporonts
exhibiting most of the distinguishing morphological character- divide into sporoblasts which will develop into spores (Fig. 5).
istics of the family Enterocytozoonidae. However, in contrast to Spores measure 2.0 to 2.5 by 1.0 to 1.5 mm, and the polar
E. bieneusi, N. salmonis grows in the nucleus rather than in the tubule has five to seven coils in a single row (Fig. 6) (50, 57, 58,
cytoplasm of cells and parasitizes fish rather than humans (120, 318, 333).
197, 386). Desportes-Livage et al. (101) further described sev- E. cuniculi was the first microsporidium to be recognized as
eral ultrastructural differences in the development of these two a parasite of mammals. First found in rabbits in 1922 (388), this
genera. Based on rRNA sequence data first generated by Bar- microsporidium was named by Levaditi et al. in 1923 (220).
lough et al. (13), rules of taxonomy, and the morphology and Subsequently, it has been detected in many mammalian hosts,
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 249

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


FIG. 6. Transmission electron micrograph of an Encephalitozoon cuniculi spore in nasal discharge from a patient with AIDS and chronic rhinosinusitis. The spore
contains a polar tubule with six coils lying in a single row and is coated with an electron-dense exospore. Magnification, 355,125.

including humans (50, 51). To date, E. cuniculi is the best placed in the genus Encephalitozoon and renamed Encephali-
studied of the microsporidian species, and much of what is tozoon intestinalis (10, 166). This reclassification is still contro-
known about the pathogenesis of microsporidial disease has versial (49), as discussed below. E. intestinalis shows a unique
been derived from studies of this organism. parasite-secreted fibrillar network surrounding the developing
Two pathogenic species of Encephalitozoon which infect hu- parasites, so that the parasitophorous vacuole appears septate
mans, E. cuniculi and E. hellem, are morphologically similar by (Fig. 7) (46, 47, 59, 266).
light and electron microscopy and can be distinguished only by
antigenic, biochemical, or nucleic acid analysis (104, 112). Sev- Nosema spp.
eral cases of Encephalitozoon infection were reported to occur
in patients with and without AIDS prior to 1991. Light and/or Most Nosema species are parasitic in invertebrates (41, 58).
electron microscopic analysis indicated that these infections Their development takes place in direct contact with the host
appeared to be due to E. cuniculi. However, in 1991 Didier et cell cytoplasm, and nuclei are paired throughout the entire life
al. (104) used biochemical and antigenic methods to describe a cycle (41, 58).
new species of Encephalitozoon, E. hellem, which had been Although microsporidia of the genus Nosema are wide-
found in three patients with AIDS. Since all subsequently pub- spread parasites, only a few human infections with Nosema
lished cases of Encephalitozoon infections in humans appeared spp. have been reported. A case of systemic infection occurred
to be caused by E. hellem (113, 162, 178, 211, 304–306, 369), in a 4-month-old thymus-deficient infant (235). At autopsy,
there was some doubt whether E. cuniculi did in fact cause numerous mature and immature microsporidian spores mea-
human infection (293). However, in 1995 De Groote et al. (95) suring 4.0 to 4.5 by 2.0 to 2.5 mm with nuclei in diplokaryon
and Franzen et al. (144) described two homosexual men with arrangement and 10 to 12 coils of the polar tubule were found.
AIDS and disseminated E. cuniculi infection; identification No other developmental stages were documented, but the fea-
was confirmed by an immunofluorescence assay and by DNA tures of the spores supported its assignment to the genus
identification. Recently, E. cuniculi has been detected in sev- Nosema as a new species, Nosema connori (235, 334). A mi-
eral HIV-infected patients (97, 177, 179, 242, 271, 374). crosporidium species infecting the corneal stroma of a 39-year
A third Encephalitozoon species, E. intestinalis, infecting old man from Ohio was named Nosema ocularum (36, 39, 44).
HIV-infected patients, was first described in 1992 by Orenstein Spores were lying freely in direct contact with the host cell
et al. (266, 268) as a microsporidium with ultrastructural sim- cytoplasm and measured 3.0 by 5.0 mm with 9 to 12 coils of the
ilarities with the genus Encephalitozoon. It was later classified polar tubule (36, 44, 45).
as a new genus and species, Septata intestinalis by Cali et al. Another microsporidium infecting muscle cells of a 31-year-
(47) on the basis of ultrastructural differences. Based on rRNA old HIV-infected patient was described by Cali et al. (48).
sequence data, it has been suggested that this organism be Development took place in direct contact with the muscle cell
250 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


FIG. 7. Transmission electron micrograph of duodenal epithelium of an HIV-infected patient infected with Encephalitozoon intestinalis. One meront with two nuclei,
four sporonts with thickened plasma membrane, and four spores are separated by amorphous material which leads to septation of the parasitophorous vacuole.
Magnification, 317,500.

cytoplasm, and the organisms contained one or two diplocary- individually in the host cell cytoplasm. This organism was orig-
otic pairs of nuclei. The spores measured about 2.5 to 2.9 by 1.9 inally assigned to the genus Nosema and named Nosema cor-
to 2.0 mm, with 7 to 10 turns of the polar tubule. These features neum (317), even though the diplokaryotic arrangement of the
are most closely aligned with the genus Nosema, and this or- nuclei was the only character that conformed with the descrip-
ganism is currently named “Nosema-like microsporidian” (48). tion of the genus Nosema. Based on the ultrastructure of de-
Another Nosema-like microsporidium was identified in fecal velopmental stages in liver cells of experimentally infected
material of a patient with AIDS (239). Because all the para- athymic mice (tetrasporoblastic sporogony, band-like sporonts,
sites were located in partially digested striated muscle cells, it all stages surrounded by a cisterna of host endoplasmatic re-
was concluded that this did not represent a true infection ticulum), this organism was later transferred to a new genus
(239). and named Vittaforma corneae (323, 324). The reclassification
on ultrastructural grounds was later supported by SSU rRNA
Vittaforma sp. gene sequence data, which placed Vittaforma distant from
Nosema (9, 10). A case of disseminated V. corneae infection
In 1990, Davis et al. (92) described an otherwise healthy recently occurred in Switzerland (375).
45-year-old man with an 18-month history of unilateral pro-
gressive central keratitis. Microsporidian spores measuring 3.7 Pleistophora spp. and Trachipleistophora spp.
by 1.0 mm were identified in deep corneal stroma and were
isolated in cell cultures (317). The spores contained polar Pleistophora spp. are common parasites of fish, and only a
tubules with six coils and had nuclei in diplokaryotic arrange- few infections have been reported in humans. Three cases of
ments. In cell culture, all the observed stages were detected Pleistophora-like microsporidian infection involving skeletal
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 251

muscles have been described in two HIV-infected patients and was not a microsporidium (79, 341, 377). Many of the 35
in a non-HIV-infected patient (69, 161, 216). The parasites affected patients lived in or had traveled to tropical or sub-
develop within a vesicle, bounded by a thick parasite-formed tropical areas (96, 295, 329, 371). Intestinal E. bieneusi infec-
coat named the sporophorous vesicle. The spores measured 2.0 tion was also reported in 8 of 990 African children who lived in
to 2.8 by 3.0 to 4.0 mm with 10 to 12 coils of the polar tube. an area of low HIV prevalence, but the HIV serostatus of these
The genus Trachipleistophora was established for a micro- children was unknown (34). Encephalitozoon spores were de-
sporidium responsible for a fourth case of myositis, this time in tected in 20 of 255 stool samples from persons with unknown
a patient with AIDS; organisms were found in corneal scrap- HIV serostatus living in two rural highland villages in Mexico
ings, skeletal muscle, and nasal discharge (138). These para- (133). Although microsporidia seem to be common pathogens
sites were cultivated in vitro and in athymic mice (180). Mer- in HIV-infected patients in Africa (34, 124, 195, 351), it is
onts had two to four nuclei and divided by binary fission. In uncertain whether they are more common in tropical and sub-
sporogony, the surface coat became separated from the plasma tropical areas than in Europe or North America. Little is
membrane and formed a sporophorous vesicle. The parasite known about the epidemiology of microsporidia, but the dis-
differed from the genus Pleistophora, because no multinucleate covery of self-limiting infections with E. bieneusi and E. intes-
sporogonial plasmodium was formed at any stage. Thus, this tinalis in immunocompetent persons suggests that microspo-
organism was placed in a new genus and named Trachipleisto- ridia may be common human pathogens (56). The wide

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


phora hominis (180). geographical distribution and the high prevalence among HIV-
Recently, two cases of infection with a Pleistophora-like mi- infected patients suggest that microsporidia may be natural
crosporidian, which also seems to be a species of Trachipleis- parasites of humans, causing disease only in immunosup-
tophora, have been reported (271, 390). Sporogony distin- pressed hosts (56). Recently, microsporidia have been emerg-
guishes this parasite from T. hominis since two different types ing as opportunistic pathogens in organ transplant recipients
of sporophorous vesicles and spores are formed (390), and the being treated with immunosuppressive drugs (194, 284, 296).
parasite has recently been classified as a new species T. More than 1,000 cases of microsporidiosis have been docu-
antropophtera (356a). mented, the majority with E. bieneusi, in HIV-infected patients
One of the Pleistophora-like microsporidia involving skeletal (14, 30, 53, 72, 87, 99, 103, 109, 110, 118, 136, 142, 207, 244,
muscles (69), which was described before T. hominis was de- 246, 263, 278, 279, 326, 344). Between 2 and 50% of HIV-
scribed as a new species, resembles T. hominis, whereas other infected patients with severe immunodeficiency and CD4 cell
Pleistophora-like microsporidia (216) may be different (180, counts below 100/ml and otherwise unexplained diarrhea are
181). infected, depending on the study group and method of diag-
nosis (72, 130, 131, 136, 142, 207, 212, 246, 263). When patients
Other Genera do not suffer from diarrhea, E. bieneusi is only rarely reported
(127, 282, 283). Rabeneck et al. (282, 283) observed no signif-
The collective group Microsporidium is an assemblage of icant difference in the occurrence of microsporidiosis in pa-
identifiable species for which the generic positions are uncer- tients with (18 of 55 [33%]) and without (13 of 51 [25%])
tain because details of their life cycle are missing (50). chronic diarrhea. However, these findings were not duplicated
Microsporidium ceylonensis was identified in a corneal ulcer by other investigators, and support for a pathogenic role for
of an 11-year-old Tamil boy from Sri Lanka. The spores mea- microsporidia is based on its identification, often as the sole
sured 1.5 by 3.5 mm, and no meronts or sporonts were seen (6, pathogen, in several hundred patients worldwide. It seems
50). Microsporidium africanum was detected in corneal stroma likely that, as with other parasites, a relationship exists between
of a 26-year-old woman from Botswana suffering from a per- the intensity of infection and clinical illness. Because intestinal
forated corneal ulcer (50, 277). Spores with 15 to 16 turns of microsporidiosis may be a common infection in humans that
the polar tubule measured 4.5 by 1.5 mm, and no developmen- can exist latently (148, 350, 353), microsporidia are most likely
tal stages of the parasite were seen. to cause disease if the immune status of a host is such that the
Many other genera in several invertebrate phyla and in all infection cannot be controlled. However, quantitation of E.
five classes of vertebrates have been described (39, 50, 58). The bieneusi spores in stool specimens is not correlated with inten-
number of named and unnamed species, now approaching sity of diarrhea (71).
1,000 and belonging to nearly 100 genera, certainly represents Infections with other microsporidian species have been re-
only a small fraction of the total diversity. Examination of new ported less frequently, but more than 100 cases of human
hosts will continue to increase the number of microsporidian infections with Encephalitozoon spp. have been documented
genera and species (58). (68, 95, 102, 113, 121, 122, 136, 143, 144, 195, 215, 238, 243,
247, 266, 291, 297–299, 304–306, 328, 369, 373, 374). Most of
EPIDEMIOLOGY these cases were due to E. intestinalis or E. hellem (68, 113, 121,
122, 136, 143, 195, 238, 243, 247, 266, 291, 297, 298, 304–306,
Prevalence and Geographic Distribution 328, 369, 373), but recently E. cuniculi was detected in several
HIV-infected patients as well (1, 95, 144, 177, 179, 242, 374).
Human infections with microsporidia have been reported Human infections with other species (N. connori, N. ocula-
from all over the world, and the majority of cases have involved rum, V. corneae, Pleistophora spp., T. hominis, T. antropophtera,
HIV-infected patients (1, 14, 36, 37, 53, 56, 68, 72, 83, 87, 88, M. ceylonensis, and M. africanum) have occurred only in a few
92, 95, 99, 102, 103, 109, 110, 113, 118, 121, 122, 136, 142–144, patients so far (6, 44, 48, 50, 69, 92, 138, 161, 216, 235, 277, 375)
177, 179, 195, 204–208, 215, 238, 243, 244, 246, 247, 263, 266, and these infections may represent only random opportunistic
277–279, 291, 297–299, 304–306, 315, 316, 326, 328, 339, 344, events.
367–370, 389–391). Among persons without HIV infection,
only 35 cases of microsporidiosis have been documented (Ta- Sources of Infection and Transmission
bles 1 and 2) (40, 371). Several early reports of suspected cases
could not be confirmed because the original material had been Routes of transmission and sources of human microsporidial
lost or reexamination showed that the responsible organism infections have been difficult to ascertain. Based on the distri-
252
FRANZEN AND MÜLLER
TABLE 1. Case reports of microsporidiosis in patients not infected with HIV with normal or unknown immune status
Microsporidial speciesa Location Age (yr)b Sexc Clinical history Immune status Diagnostic method Yr of study Reference(s)
d e
E. cuniculi Japan 9 M Seizure disorder NK Cytology of CSF and urine 1959 237
E. intestinalis Swizerland 36 M Diarrhea (self-limited) Normal Examination of stool specimens 1995 139
E. intestinalis France (travel to tropical country) NK M Diarrhea Normal Examination of stool specimens 1998 286
E. intestinalis France (travel to tropical country) NK M Diarrhea Normal Examination of stool specimens 1998 286
E. bieneusi Germany (travel to Egypt and Jordan) 26 M Diarrhea (self-limited) Normal Examination of stool specimens 1994 295
E. bieneusi France (returning from tropical country) NK M Diarrhea NK, Crohn’s disease Examination of stool specimens 1994 89
E. bieneusi France (returning from tropical country) NK M Diarrhea NK, cardiac-valve graft Examination of stool specimens 1994 96
E. bieneusi France (travel to tropical country) NK M Diarrhea NK Examination of stool specimens 1994 96
E. bieneusi France (travel to tropical country) NK M Diarrhea NK Examination of stool specimens 1994 96
E. bieneusi France (returning from Africa) NK F Diarrhea NK Examination of stool specimens 1994 96
E. bieneusi France NK M Diarrhea NK Examination of stool specimens 1994 96
E. bieneusi Germany (travel to Turkey) 3 F Diarrhea (self-limited) Normal Examination of stool specimens 1995 329
E. bieneusi Swizerland 26 F Diarrhea (self-limited) Normal Examination of stool specimens 1995 139
E. bieneusi Zambia 7 mo M No symptoms Normal Examination of stool specimens 1997 167
E. bieneusi Spain 24 F Diarrhea (chronic) Normal Examination of stool and 1998 154a
duodenal biopsy specimens
M. ceylonensis Sri Lanka 11 M Keratitis NK Histologic examination 1973 6
M. africanum Botswana 26 F Corneal ulceration NK Histologic examination 1981 277
N. ocularum USA (Ohio) 39 M Keratitis Normal Histologic examination 1991 36, 44
V. corneae USA (South Carolina) (travel to 45 M Keratitis/iritis Normal Histologic examination 1990 92
Caribbean and Central America)
Not determined France 44 F Fever, myalgia Normal Examination of stool specimens 1995 249
Not determined France 72 M No symptoms Liver cirrhosis after Examination of stool specimens 1995 249
hepatitis B
Not determined France 63 M No symptoms Chronic alcoholism Examination of stool specimens 1995 249
Not determined France 68 M No symptoms Chronic alcoholism Examination of stool specimens 1995 249
Not determined France 64 M No symptoms Chronic alcoholism Examination of stool specimens 1995 249
Not determined France (travel to tropical country) NK M Diarrhea Normal Examination of stool specimens 1998 286
Not determined France (travel to tropical country) NK M Diarrhea Normal Examination of stool specimens 1998 286
a
Classification is based only on morphology.
b
Age in years unless otherwise indicated.
c
M, male; F, female.
d
NK, not known.
e
CSF, cerebrospinal fluid.

CLIN. MICROBIOL. REV.


Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 253

TABLE 3. Animal hosts of human microsporidia

Microsporidial speciesa
c
b
a

Not determined

Not determined
N. connori
Pleistophora sp.

E. bieneusi

E. bieneusi
E. bieneusi

E. bieneusi
E. cuniculi
M, male; F, female.
Age in years unless otherwise indicated.
Classification is based only on morphology.
Species Nonhuman host Reference(s)

E. bieneusi Pigs, dogs, macaque monkeys 234


E. intestinalis Dogs, donkeys, pigs, cows, goats 32a
E. hellem Birds (parrots) 28
E. cuniculi Several mammals 50, 97, 98, 111,
118, 181,
236a, 365
France

India
USA (Washington D.C.)
USA (Florida)

Spain

France
USA (Boston)

USA (Boston)
Sweden (born in Colombia)
Strain I Rabbits, mice
Strain II Mice, blue foxes
Strain III Domestic dogs, rodents, goats,
sheep, swine, horses, foxes,

Location
cats
N. connori None known
N. ocularum None known

TABLE 2. Case reports of microsporidiosis in patients not infected with HIV but with immunodeficiency
Nosema-like sp. None known
V. corneae None known

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


Pleistophora spp. Fish 50
T. hominis None known
T. antropophtera None known
Age (yr)b

M. africanum None known


52

27

20

66

48
42

48
4 mo

M. ceylonensis None known


Sexc
M

F
M
M

M
M

F
M

bution of lesions, oral, respiratory, and ocular routes of infec-


tion are possible and are supported by evidence obtained from
Diarrhea (chronic),

Pneumonia
Disseminated infection
Myositis

Diarrhea (chronic)

Diarrhea
Diarrhea (chronic)

Diarrhea (self-limited)
Seizure disorder
malabsorption

experimentally infected rabbits (80, 153, 313), mice (106, 156,


Clinical history

209, 300, 342), and monkeys (106, 343). There is considerable


serologic evidence that humans without clinical signs of infec-
tion have been exposed to microsporidia (174–176, 327, 353).
Whether these persons are chronically or actively infected is
unknown.
Microsporidia are released into the environment via stool,
urine, and respiratory secretions. Persons or animals infected
with microsporidia are possible sources of infection. Experi-
Immunosuppression after

Immunosuppression after
Thymic aplasia
T-cell immunodeficiency

Lymphocytopenia and

Immunosuppression after
Decreased CD4 cell count

Immunosuppression after
Low CD4/CD8 cell ratio

mental Encephalitozoon infections of several animals by the


heart-lung transplantation

transplantation
bone marrow

hypogammaglobulinemia

heart-lung transplantation

liver transplantation

oral, tracheal, and rectal routes have been reported (80, 153,
209, 313). Person-to-person transmission of microsporidia may
Immune status

be significant. In a case-control study, intestinal microsporidi-


osis was associated with male homosexuality, thereby suggest-
ing sexual routes of transmission (181a). Person-to-person
transmission was suspected in an HIV-seronegative partner of
an HIV-infected man with intestinal microsporidiosis due to E.
intestinalis (139). Another male patient with microsporidial
urethritis had a sexual partner with diarrhea due to intestinal
microsporidiosis (27).
Examination of stool specimens

Histologic examination (autopsy)


Histologic examination (autopsy)
Histologic examination

Examination of stool and

Examination of stool specimens

Examination of stool specimens


Examination of stool specimens

Cytology of urine, serum antibody

Whether microsporidiosis in humans is a zoonosis is un-


(gastrointestinal biopsy)
and histologic examination

duodenal biopsy specimens

known, and no direct proof of transmission from animals to


humans has been documented, with the exception of one case
Diagnostic method

where a 10-year-old girl seroconverted after close contact with


a dog infected with E. cuniculi (239a). Animal reservoirs of
microsporidia infecting humans have been confirmed recently
(Table 3). E. cuniculi is commonly found in several mammals
(50, 51), and Encephalitozoon spp. have occasionally been
found in lovebirds (50, 196). E. hellem was recently detected in
birds (parrots) (28), E. intestinalis has been found in different
mammalian animals (donkey, dog, pig, cow, and goat) in Mex-
ico (32a), and Enterocytozoon bieneusi has been found in stool
samples of pigs and dogs in Switzerland (98) and in simian
Yr of study

immunodeficiency virus-infected macaques (234). E. hellem in-


1995

1994

1998
1973
1985

1996

1995
1996

1984

fection of birds, E. bieneusi infections of pigs, dogs, and mon-


keys, and E. intestinalis infection of different mammals were
confirmed by molecular techniques with rRNA data (32a, 98,
Reference

234). Molecular analysis of different human, rabbit, dog,


249

194

154a
235
216

284

296
365a

21

mouse, and blue fox E. cuniculi isolates showed that all isolates
from humans were of the same subtype as isolates from dogs
254 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

TABLE 4. Clinical manifestations of human disease is unknown. Results from recent studies involving mo-
microsporidial infections lecular techniques seemed to indicate the presence of E. intes-
Reference(s)
tinalis, E. bieneusi and V. corneae in raw sewage, tertiary efflu-
Species Clinical manifestation ents, surface water, and groundwater in France and the United
(first reports)
States (123a, 332a); there is one report of a presumably water-
E. bieneusi Enteritis 99, 119, 245 borne outbreak in Lyon (France) during summer 1995 (78a).
Cholangitis, cholecystitis 240, 278 Risk factors for intestinal microsporidiosis also suggest water
Pneumonia, bronchitis 367
Sinusitis, rhinitis 129, 165
as the source of infection (133, 181a). In a case-control study,
the only two factors associated with intestinal microsporidiosis
E. intestinalis Enteritis 266 were swimming in a pool and male homosexuality, both sug-
Cholangitis, cholecystitis 268, 385 gesting that the mode of transmission is fecal-oral (181a).
Nephritis, urinary tract infection 47, 268 However, no seasonal variation in the prevalence of micros-
Sinusitis, rhinitis 121, 147 poridiosis in HIV-infected patients was seen over 4 years in
Bronchitis 121, 150 southern California, suggesting a constant presence of micro-
Keratoconjunctivitis 226 sporidia in the environment rather than a seasonal association
Disseminated infection 47, 268 with recreational water use or seasonal contamination of the

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


E. hellem Keratoconjunctivitis 104, 306
water supply (75a).
Sinusitis, rhinitis 178, 211
Pneumonia, bronchiolitis 304, 305
Nephritis, urinary tract infection 304 CLINICAL MANIFESTATIONS
Prostatic abscess 308
Disseminated infection 304 Microsporidiosis is truly an emerging infectious disease with
a rapidly broadening clinical spectrum of diseases. The spec-
E. cuniculi Hepatitis, peritonitisa 340, 392 trum of diseases includes gastrointestinal, pulmonary, nasal,
Encephalitis 237, 242, ocular, muscular, cerebral, and systemic infections. Microspo-
374 ridiosis should be considered in the differential diagnosis of
Intestinal infection 144 HIV-related symptomatic disease of virtually all organ systems
Urinary tract infection 95, 144 (Table 4) (271).
Keratoconjunctivitis 95, 144
Sinusitis, rhinitis 95, 144
Disseminated infection 95, 144 Gastrointestinal and Biliary Tract Infections
T. hominis Myositis 138, 180 Enterocytozoon bieneusi. Intestinal infections with microspo-
Keratoconjunctivitis 138, 180 ridia have been found mainly in HIV-infected patients, and
Sinusitis, rhinitis 138, 180 most infections have been due to E. bieneusi. It is most com-
T. antropophtera Encephalitis 271, 390
mon in patients with severe immunodeficiency and a CD4 cell
Myositis 271, 390 count below 100/ml (60, 127, 130, 131, 142, 246, 332). The
Disseminated infection 271, 390 parasites cause a severe, nonbloody, nonmucoid diarrhea with
up to 10 or even more bowel movements per day, slowly pro-
Pleistophora spp. Myositis 216, 229, gressive weight loss, and malabsorption of fat, D-xylose, and
230 vitamin B12 (60, 78, 127, 130, 131, 204, 205, 212). Intestinal
infection is associated with lactase deficiency and a reduced
V. corneae Keratitis 92, 317, 324 activity of alkaline phosphatase and a-glucosidase at the basal
part of the vilus and with reduced villus height and a vilus
N. ocularum Keratoconjunctivitis 36, 44
surface reduction (301). Diarrhea appears gradually and may
N. connori Disseminated infection 235, 334 continue for months. Patients are often reluctant to eat and
may complain of nausea (60, 78, 204, 205 212). Some patients
Nosema-like sp. Myositis 239 have intermittent diarrhea, but only a few excrete microspo-
ridial spores without having diarrhea (282, 283, 326, 330). In
M. africanum Corneal ulcer 50, 277 groups of patients with chronic diarrhea who were negative for
other enteric pathogens, the prevalence of E. bieneusi was
M. ceylonensis Corneal ulcer 6, 50 between 7 and 50% (127, 130, 131, 207, 246, 263). Some pa-
a
Species not confirmed because classification was based only on ultrastructure tients have coinfections with other pathogens (30, 171, 370);
morphology. cryptosporidia are the most common pathogens coinfecting
patients with intestinal microsporidiosis (157, 171, 370).
E. bieneusi infection of the biliary tract, with or without
cholecystitis, is responsible for some of AIDS-related cholan-
and rabbits (97, 98, 108, 111, 181, 236). This fact supports the giopathies which are not explained by Cryptosporidium spp. or
hypothesis that human infections with E. cuniculi may be a cytomegalovirus infection (33, 201, 240, 278, 279).
zoonosis (97, 98, 111, 181, 236). Dissemination of E. bieneusi is very uncommon, but it has
Arthropods are the most common hosts of microsporidia, been detected in duodenal lamina propria cells (310), in bron-
and experimental infections of mice by a mosquito microspo- choalveolar lavage fluid and transbronchial biopsy specimens
ridium (Nosema algerae) have been accomplished (342, 345). (93, 367), and in nasal sinuses of HIV-infected patients (129,
Whether insect microsporidia might infect humans is un- 165, 184).
known. Intestinal infections with E. bieneusi in non-HIV-infected
Several microsporidia have been found in surface water sam- patients have been reported in only 15 patients in Germany,
ples (8), but whether human microsporidiosis is a waterborne Switzerland, France, Spain, Zambia, and the United States (96,
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 255

154a, 167, 249, 284, 295, 296, 329): one patient had Crohn’s microsporidia consistent in ultrastructure with E. cuniculi were
disease (96), an African woman had a cardiac-valve graft (96), discovered within areas of mixed nongranulomatous inflamma-
one patient had a congenital disorder of the lymphatic system tion in sections of the omentum magnum (392). The reports of
(154a), another had a low CD4 cell count of unknown origin these two cases were published before E. hellem was described
(365a), and two patients were immunosuppressed due to treat- as a new species. In both instances, diagnosis was made only on
ment associated with liver and heart-lung transplantation (284, an ultrastructural basis, so that the exact species identification
296). The other nine patients were otherwise healthy (Tables 1 is uncertain.
and 2). Although patients usually presented with self-limiting A second case of fulminant hepatic failure caused by micro-
diarrhea, some patients were treated with albendazole or met- sporidial infection with an Encephalitozoon sp. was reported in
ronidazole (284, 296). a 43-year-old homosexual man with AIDS (322). He suffered
Encephalitozoon spp. Similar to Enterocytozoon bieneusi, E. from microsporidial diarrhea 2 months prior to development
intestinalis causes an enteritis with diarrhea, weight loss, and of fulminant hepatitis. The patient died before albendazole
malabsorption (68, 70, 78, 121, 136, 143, 207, 212, 247, 266). became available. The autopsy revealed disseminated micros-
Besides intestinal infections, these parasites may infect the poridial infection involving the liver, gallbladder wall, and a
biliary tract and gallbladder, resulting in cholangitis and cho- mediastinal lymph node.
lecystitis (385). Disseminated infections occur regularly and Both E. bieneusi and E. intestinalis have been detected in

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


involve heavy infections of the urinary tract including the kid- nonparenchymal liver cells of several HIV-infected patients,
neys (68, 121, 143, 150, 247, 268). Left untreated, small bowel but the patients did not show any signs of hepatitis (14, 268,
infection with E. intestinalis can lead to perforation and peri- 278, 279).
tonitis (331). E. cuniculi only occasionally infects the gastroin- Disseminated Trachipleistophora antropophtera infection in-
testinal tract, and its pathogenicity in humans is unknown. volving several organ systems including the liver and the pan-
Franzen et al. (144) described an AIDS patient with a widely creas was reported in an 8-year-old HIV-infected girl with
disseminated E. cuniculi infection including the gastrointesti- seizures and cerebral lesions. This patient died after empirical
nal tract but with no accompanying gastrointestinal symptoms. antitoxoplasma therapy (271, 390).
Weber et al. (374) described a second patient with dissemi-
nated infection due to E. cuniculi who had no gastrointestinal
symptoms but who had microsporidian spores in the stool Ocular Infections
samples.
Among persons not infected with HIV, only three cases of Beside gastrointestinal infection, ocular microsporidiosis is
intestinal infection with an Encephalitozoon spp. have been the most common manifestation of microsporidiosis in humans
reported (139). A 36-year-old HIV-seronegative homosexual (225).
man was asked to provide stool for examination after E. intes- Encephalitozoon spp. In HIV-infected patients, keratocon-
tinalis was demonstrated in stool samples of his HIV-infected junctivitis may be caused by all three Encephalitozoon spp. (E.
partner. E. intestinalis was detected in two of seven stool sam- hellem, E. cuniculi, and E. intestinalis) (44, 45, 102, 104, 105,
ples from the non-HIV-infected man and again 4 months later, 113, 144, 152, 211, 224–226, 238, 243, 291, 306, 319, 369). Most
together with Isospora belli, when he became mildly symptom- patients present with bilateral conjunctival inflammation and
atic after a trip to Brazil (139). Two other patients were trav- also exhibit bilateral punctate epithelial keratopathy, leading
elers presenting with chronic diarrhea, and microsporidian to decreased visual acuity. The keratoconjunctivitis is often
spores were detected in their stools (286). Molecular identifi- asymptomatic or moderate but can be severe; it rarely leads to
cation of microsporidian species as E. intestinalis was based on corneal ulcers (225).
PCR amplification of an SSU rRNA sequence. Albendazole Other species. Keratitis with corneal stroma infection was
treatment led to the elimination of spores in the stool, but the described in an otherwise healthy 45-year-old man from South
clinical signs persisted. Carolina who developed decreased vision in his left eye during
Other species. A Nosema-like microsporidium has been an 18-month history of unilateral progressive central keratitis
identified in fecal material of a patient with AIDS (239). The (92). There was no history of prior trauma. Corneal biopsy
parasites were located in partially digested striated muscle revealed microsporidia invading deep into the corneal stroma.
cells, suggesting that infected animal musculature had been This organism was successfully propagated in vitro and was
ingested. It was concluded that this represents an incidental named Nosema corneum (317). On the basis of ultrastructural
finding rather than a true infection (239). data, it is now in a new genus and has been renamed Vittaforma
corneae (324).
Hepatitis, Pancreatitis, and Peritonitis A microsporidium was found to be responsible for the symp-
toms in a 39-year old man from Ohio who developed blurred
Hepatitis caused by an Encephalitozoon spp. that was clas- vision and irritation in his left eye. His visual symptoms per-
sified as E. cuniculi on an ultrastructural basis was reported in sisted despite the discovery and surgical removal of a foreign
a 35-year-old HIV-infected patient from southern Florida with body (36, 39). A subsequent biopsy of the persistent corneal
a CD4 cell count of 48/ml (340). He presented with fatigue, ulcer revealed organisms with typical microsporidian ultra-
diarrhea, and weight loss. He subsequently developed fever structure; the species was named Nosema ocularum (44).
and died of hepatocellular necrosis. Autopsy confirmed the Trachipleistophora hominis was found in the corneal scrap-
diagnosis of microsporidian hepatitis. ings of an HIV-infected patient with disseminated infection
Peritonitis due to E. cuniculi was described in a 45-year-old who suffered from myositis and keratoconjunctivitis (138).
HIV-infected man with a CD4 cell count of 57/ml (392). The In 1973 and 1981, two cases with corneal involvement were
patient presented with a 13-kg weight loss over the course of a documented in a 11-year-old Tamil boy from Sri Lanka with a
year and was treated with trimethoprim-sulfamethoxazole be- corneal ulcer and a 26-year-old woman from Botswana suffer-
cause of Pneumocystis carinii pneumonia. After the end of ing from a perforated corneal ulcer (6, 277). Both otherwise
therapy, he developed renal failure and a tumorlike mass was healthy patients did not have HIV infection. The genera could
recognized in the abdomen. He died, and at limited autopsy not be determined, and the organisms were named Microspo-
256 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

ridium ceylonensis and Microsporidium africanum, respectively myositis with fever, myalgia, and progressive weakness. Mi-
(50). crosporidian spores were detected in a muscle biopsy speci-
men.
Sinusitis In an Australian patient who presented with a severe, pro-
gressive myositis associated with fever and weight loss, Pleis-
Sinusitis is a common manifestation of human microsporidi- tophora-like microsporidia were demonstrated in corneal
osis. All three Encephalitozoon spp. have caused rhinosinusitis scrapings, skeletal muscle, and nasal discharge (138). The or-
in several HIV-infected patients (129, 144, 147, 211, 250, 274, ganisms were cultivated in vitro as well as in athymic mice.
292). E. bieneusi and T. hominis have also been detected in Since these parasites differed from Pleistophora, the new genus
sinus biopsy specimens from HIV-infected patients (129, 138, and species Trachipleistophora hominis was established (180).
165, 184); the patients suffered from severe rhinitis, and nasal A Nosema-like microsporidium was detected by Cali et al. in
polyps were often present. a biopsy specimen from the left quadriceps of a 31-year-old
patient with AIDS and myositis (48).
Pulmonary Infections
Pulmonary infections with microsporidia have been reported Cerebral Infections
less frequently than other manifestations (150, 213, 287, 297– Encephalitozoon spp. Two cases of disseminated Encephali-

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


299, 304, 305, 328, 369). Infection of the lower respiratory tract tozoon infection with cerebral involvement were reported in a
may be asymptomatic or associated with bronchiolitis; it is 9-year-old Japanese boy and in a 2-year-old Columbian boy.
rarely associated with pneumonia or progressive respiratory Both patients suffered from cerebral symptoms such as head-
failure in HIV-infected patients (150, 287, 297–299, 304, 305, ache, vomiting, spastic convulsions, and convulsive seizures.
369). All three Encephalitozoon spp. have been detected in Encephalitozoon-like organisms were found in urine from both
bronchial epithelial cells of HIV-infected patients with dissem- patients and in cerebrospinal fluid from one patient. The exact
inated Encephalitozoon infection, whereas pulmonary involve- species differentiation of these two parasites is uncertain (21,
ment with E. bieneusi has been reported only in two patients 237) (see “Systemic infections” below).
(93, 367). Cerebral microsporidiosis due to E. cuniculi was recently
Pulmonary microsporidial infection was also found in a 27- described by Weber et al. (374) in a 29-year-old HIV-infected
year-old woman from India with chronic myeloid leukemia man with a CD4 cell count of 0 cells/ml. The patient was
undergoing allogenic bone marrow transplantation (194). The hospitalized because of headache, visual and cognitive impair-
patient died of a fungal infection, and the diagnosis of pulmo- ment, nausea, and vomiting. Magnetic resonance imaging
nary microsporidiosis was reached only postmortem. Ultra- scans showed right maxillary sinusitis and multiple small, con-
structural examinations confirmed the organism to be a mi- trast-enhanced lesions in the hippocampal, mesencephalitic,
crosporidium, but taxonomic classification could not be done and intracortical regions. Examination of cerebrospinal fluid
because the organism could not be identified as any of the showed microsporidial spores, which were also detected in
known pathogenic species of microsporidia (194). sputum, urine, and stool specimens. The microsporidium was
cultivated in vitro and was classified as E. cuniculi by Western
Urinary Tract Infections blot analysis, ribotyping, and sequencing of the rRNA inter-
genic spacer region (374). A similar case was reported by
Infections of the urinary tract are a common finding in Mertens et al. (242) in a 25-year-old HIV-infected woman.
HIV-infected patients with disseminated Encephalitozoon in- Microsporidian spores, classified as E. cuniculi by immunohis-
fections. The clinical presentation and consequences of the tochemistry and PCR, were detected in the brain, heart, kid-
presence of microsporidia in the urinary system can vary re- neys, urinary bladder, spleen, lymph nodes, adrenals, and tra-
markably; patients may be asymptomatic with or without mi- chea at autopsy.
crohematuria, they may have cystitis and intestinal nephritis Other species. Cerebral involvement with Trachipleistophora
with dysuria and gross hematuria, or they may experience pro- antropophtera was reported in two AIDS patients, a 33-year-
gressive renal failure (1, 121, 144, 150, 242, 268, 304, 305, 369). old man and an 8-year-old girl, with seizures and cerebral
lesions, who died after empirical anti-toxoplasma therapy (20,
Myositis 271, 390). At autopsy, a pansporoblastic microsporidium was
seen in several organ systems including the brain (271, 390).
Myositis caused by Pleistophora-like microsporidia has been
described in four immunocompromised patients. Ledford et al.
(216) reported a 20-year-old HIV-seronegative man who had a Rare Manifestations
severe immunodeficiency of unknown origin (CD4 cells, 66/ml) Urethritis. Two cases of urethritis associated with microspo-
with progressive generalized muscle weakness and contrac- ridia were found in patients with AIDS who suffered from
tures for 7 months, fever, generalized lymphadenopathy, and urethritis, sinusitis, and diarrhea (27, 77). Encephalitozoon-like
an 18-kg weight loss. Pleistophora-like microsporidian spores spores were detected in a smear of expressed urethral pus as
were seen in muscle biopsy specimens from the quadriceps and well as in stool samples, nasal discharge, sputum, and urine of
deltoid (216). Four years after his initial clinical presentation, one patient (77) and in stool samples of the second patient
the patient was still immunodeficient but remained seronega- (27). Both patients were treated with albendazole, and the
tive for HIV (229, 230). symptoms disappeared.
Chupp et al. (69) reported a 33-year-old Haitian man with Prostatic abscess. A prostatic abscess due to E. hellem was
AIDS who was admitted to the hospital with fever, cough, and found in an AIDS patient with disseminated E. hellem infec-
diffuse myalgias and weakness (69, 281). A Pleistophora-like tion (308). The prostate was of normal size with a 1.5- by
microsporidium was detected in muscle cells in a biopsy spec- 1.8-cm central periurethral abscess containing necrotic pros-
imen from the right quadriceps. A similar case was reported by tatic tissue. Tissue Gram stain revealed gram-positive micro-
Grau et al. (161) in a 35-year old HIV-infected Spanish man sporidian spores, which were identified as E. hellem by an
who originated from The Gambia. The patient suffered from indirect fluorescence assay.
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 257

Tongue ulcer. A shallow 1-cm ulceration on the dorsum of newly recognized pansporoblastic microsporidium, T. antropo-
the tongue was observed in an HIV-infected patient with se- phtera caused disseminated infection involving the brain, heart,
vere immunodeficiency (15 CD4 cells/ml) and disseminated kidneys, pancreas, thyroid, parathyroid, liver, bone marrow,
infection due to E. cuniculi (95). Spores were identified in lymph nodes, and spleen in an HIV-infected 8-year-old child
several samples and in soft tissue beneath the tongue ulcer. (271, 390). T. antropophtera infection of the brain was also seen
The microsporidian was identified as E. cuniculi by immuno- in a 33-year-old HIV-infected male in whom autopsy was lim-
fluorescent staining, in vitro cultivation, and molecular analysis ited to the brain (20, 271, 390).
of the SSU rRNA gene by PCR. The patient was treated with
albendazole, and the symptoms resolved within 2 weeks (95). THERAPY
Skeletal involvement. Only two cases of skeletal involvement
with microsporidia have been found in patients with AIDS Successful treatment of microsporidiosis in immunodeficient
(19). Both patients suffered from disseminated microsporidial patients is limited. Several in vitro culture systems and animal
infections. In one patient the mandible and associated soft models have been used to identify potential antimicrobial
tissues were involved. Species identification was not done. agents for treatment of microsporidiosis. Different drugs con-
Cutaneous microsporidiosis. One case of nodular cutaneous trol the levels of microsporidial infection in invertebrate hosts;
microsporidiosis that resolved with oral clindamycin therapy these include fumagillin, an antibiotic produced by Aspergillus

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


was found in an HIV-infected patient (200a). Underlying os- fumigatus, and itraconazole for control of Nosema apis in
teomyelitis that also resolved after therapy was not proven to honey bees and other microsporidia in weevils (54). However,
be caused by the microsporidia. Species differentiation by PCR in vitro investigations with Nosema bombycis showed no effect
techniques was not successful. of itraconazole and metronidazole on the number of cells in-
fected or on the spore harvest (54). On the other hand, al-
Systemic Infections bendazole had marked effects on these parameters, and several
ultrastructural changes in the parasites were noted (54, 163).
Encephalitozoon spp. The first case of documented human Other in vitro models used to evaluate drug efficacy included
microsporidial infection was a case of disseminated Encepha- E. cuniculi, E. hellem, and E. intestinalis (16, 117, 141, 168, 218,
litozoon infection in a 9-year-old Japanese boy who suffered 219, 380). These studies showed that albendazole, fumagillin,
from recurrent fever, headache, vomiting, and spastic convul- 5-fluorouracil, sparfloxacin, oxibendazole, and propamidine is-
sions reported in 1959. Encephalitozoon-like organisms were ethionate inhibited E. cuniculi growth in cell cultures. Chloro-
found in cerebrospinal fluid and urine. He was treated with quine, pefloxacin, azithromycin, rifabutin, and thiabendazole
sulfisoxazole and penicillin and recovered (237). were partially effective at high concentrations. Arprinocid,
A similar case occurred in 1984 in a 2-year-old Columbian metronidazole, minocycline, doxycycline, itraconazole, and di-
boy who lived in Sweden. He had convulsive seizures, and fluoromethylornithine were not evaluable, since the concentra-
gram-positive organisms consistent with an Encephalitozoon tions that inhibited microsporidia were also toxic for the cells
sp. were found in urine. Anti-E. cuniculi antibodies (immuno- in the cell culture. Pyrimethamine, piritrexim, sulfonamides,
globulin G [IgG] and IgM) were detected in serum samples paronomycin, roxithromycin, atovaquone, flucytosine, toltra-
(21). zuril, ronidazole, and ganciclovir were ineffective (16). Spore
Disseminated infections with all three Encephalitozoon spp. germination of E. hellem and E. intestinalis was inhibited by
are now increasingly recognized in severely immunosuppressed nifedipine, metronidazole, and nitric oxide donors (168), and
HIV-infected patients, usually in those with CD4 cell counts E. hellem spore germination was also inhibited by cytochalasin
below 100/ml (95, 121, 144, 150, 162, 215, 247, 268, 297, 304, D, demecolcine, and itraconazole (218). TNP-470, a semisyn-
305, 328, 369). The spectrum of disease has expanded to in- thetic analogue of fumagillin, was highly effective against all
clude keratoconjunctivitis, bronchiolitis and pneumonia, sinus- three Encephalitozoon spp. and V. corneae in cell cultures (82,
itis, nephritis, urethritis, cystitis, prostatitis, hepatitis, peritoni- 111a). A fluorescent probe, designated calcein, and confocal
tis, gastroenteritis, and cholangitis, but there are clear microscopy have been used to demonstrate drug-induced ef-
differences in the typical distribution pattern for each micro- fects in Encephalitozoon-infected green monkey kidney cells,
sporidian species: E. hellem parasitizes mainly the keratocon- and both albendazole and fumagillin caused different types of
junctiva, urinary tract, nasal sinuses, and bronchial system; on parasite changes (219). In vivo efficacy of albendazole, fum-
the other hand, E. intestinalis appears to be confined mainly to agillin, and TNP-470 against E. cuniculi has been demon-
the gastrointestinal and biliary tract with dissemination to the strated in experimentally infected SCID mice, athymic mice,
kidneys, eyes, nasal sinuses, and sometimes the respiratory and rabbits (82, 210, 314). Unfortunately, long-term in vitro
tract; finally, E. cuniculi causes widely disseminated infections cultivation of Enterocytozoon bieneusi has not been feasible so
involving nearly all organ systems, but the clinical manifesta- far; therefore, a direct assay of the effects of agents on this
tions vary substantially, ranging from no symptoms to severe parasite is not yet practicable.
disease (144, 150, 162, 215, 247, 268, 304, 305, 328, 369). Based on these in vitro studies, several drugs have been used
Other species. In 1973, Nosema infection and Pneumocystis to treat microsporidial infections in humans. Until recently,
carinii pneumonia were diagnosed at autopsy in a 4-month-old blinded, placebo-controlled comparative trials were lacking.
athymic male infant (235). Shortly after birth, the child devel- Therefore, most clinical experience in the therapy of human
oped diarrhea, vomiting, fever, dyspnea, weight loss, and me- microsporidiosis consists of only anecdotal observations. Sev-
chanical ileus. Laparatomy and several antibiotics failed to eral case reports and small case series have shown that al-
alter the clinical course, and at autopsy sporoblasts with ma- bendazole was highly effective for treatment of Encephalito-
ture and immature spores of a Nosema sp. were seen in almost zoon infection in HIV-infected patients and led to impressive
all tissues examined except the spleen (235). The parasite was clinical improvement and eradication of the parasites (1, 77,
named Nosema connori (334). 95, 109, 121, 143, 144, 150, 162, 185, 211, 215, 247, 248a, 269,
T. hominis was reported as the cause of myositis in a 34- 328, 373). However, since some patients relapsed after therapy,
year-old HIV-infected man; parasites were recognized in cor- maintenance therapy may be necessary for these patients
neal scrapings, skeletal muscle, and nasal discharge (138). The (248a, 373). Symptomatic improvement with reduction of clin-
258 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

ical findings was also achieved with topical fumagillin (113, well to albendazole therapy, exact species differentiation of
158, 314) in several HIV-infected patients with microsporidial microsporidia infecting humans is absolutely necessary.
keratoconjunctivitis due to Encephalitozoon species. Resolu-
tion of E. hellem infection of the corneal epithelium of an DIAGNOSTIC METHODS
AIDS patient with itraconazole was also reported (391), but
this finding could not be duplicated (113). Itraconazole seems Diagnosis of human microsporidiosis is dependent on the
to be ineffective in preventing Enterocytozoon bieneusi infec- identification of spores in clinical samples, e.g., stool speci-
tion in HIV-infected patients (2). Keratoconjunctivitis due to mens, duodenal or bile juice, urine, conjunctival smears, bron-
an Encephalitozoon species in another AIDS patient re- choalveolar lavage fluid, sputum, nasal discharge, or biopsy
sponded to topical dibromopropamidine isethionate (238). tissues. The detection of spores in clinical samples, however, is
Infections due to E. bieneusi are much more difficult to treat, a laborious, challenging, and time-consuming task because the
and currently there is no acceptable treatment. Pneumocystis tiny organisms can easily be missed. Originally, definitive di-
carinii prophylaxis with co-trimoxazole seems to have no influ- agnosis of microsporidiosis required transmission electron mi-
ence on the prevalence of intestinal microsporidiosis in HIV- croscopy, but during the last few years new staining methods,
infected patients, suggesting that this drug may be ineffective suitable for light microscopy, have been developed. Microspo-
(3). Improvement or disappearance of diarrhea caused by E. ridia have now been found in virtually every tissue and body
fluid in humans.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


bieneusi has been reported after treatment with metronidazole,
Although the diagnosis and identification of microsporidia
but repeated biopsies showed that microsporidia persisted
by light microscopy have greatly improved during the last few
(127, 128), and other investigators did not observe any re-
years, species differentiation is usually impossible by these
sponse after treatment with metronidazole (114, 136). The
techniques. Immunofluorescent staining techniques have been
efficacy of albendazole for E. bieneusi infections is controver-
developed for species differentiation of microsporidia, but an-
sial (257). Diarrhea may improve in 50 to 60% of patients (29,
tibodies used in these procedures are available only at research
103, 114), but persistence of organisms was seen in posttreat- laboratories so far. Similarly, cell culture systems can be used
ment biopsy specimens despite several ultrastructural changes for in vitro cultivation of microsporidia, but this is not a suit-
in the parasite (29, 31, 114). Other trials with albendazole able technique for routine use because it is laborious and
showed much lower response rates (142, 172, 246). Double- time-consuming.
blind placebo-controlled trials with albendazole are in A variety of serological tests has been developed to detect
progress. Despite a remarkable clinical response with atova- antibodies to microsporidia, but the sensitivity and specificity
quone in symptomatic AIDS patients with intestinal E. bieneusi of these tests are unknown. Also, these tests are not suitable
infection, there was no apparent decrease in the parasite bur- methods to diagnose infections in immunosuppressed persons.
den in either stools or biopsy specimens (5). A double-blind Recent success in nucleotide sequencing of various micros-
placebo-controlled study with atovaquone is also under way. poridia has now led to the application of new molecular tech-
Similarly, azithromycin treatment showed only partial effect on niques for the diagnosis of human microsporidiosis.
diarrhea in E. bieneusi-infected patients, and microsporidial
infection persisted on repeat biopsy and stool examinations
Transmission Electron Microscopy
(172). Improvement of diarrhea with clearance of microspo-
ridian shedding in stool was observed in three E. bieneusi- Originally, definitive diagnosis of microsporidiosis required
infected patients treated with furazolidone (116). Purified fum- ultrastructural examination of biopsy tissues, body fluid spec-
agillin was also able to clear E. bieneusi infection from stool as imens (urine, nasal discharge, sinus aspirates, sputum, bron-
well as intestinal biopsy specimens in three patients, but the choalveolar lavage fluid, duodenal or bile juice, cerebrospinal
drug is toxic and causes thrombocytopenia in nearly all patients fluid), or stool samples by transmission electron microscopy,
(248). These observations must be confirmed by treatment of because of the small size of the organisms and their poor and
more patients with these two drugs. As mentioned above, variable staining characteristics (83, 88, 241, 346). Visualiza-
TNP-470, a synthetic analogue of fumagillin that is less toxic, is tion of the unique ultrastructure of the spores with their char-
as effective as fumagillin against several microsporidian species acteristic coiled polar tube is diagnostic. Microsporidia can be
in vitro and in athymic mice and holds promise as a new identified to the genus or even species level based on the
antimicrosporidial compound (111a). fine-structure features of the spores and proliferative forms,
Of note, elevated tumor necrosis factor alpha levels in stool method of division, and nature of the host cell-parasite inter-
samples have been demonstrated in patients with microspo- face. In tissue, all stages of the life cycle can often be observed,
ridial diarrhea. Therefore, the anti-tumor necrosis factor alpha whereas in body fluids or stool samples only spores are visible.
agent thalidomide has been used to treat diarrhea due to E. The ultrastructural characteristics of microsporidian species
bieneusi. Some patients responded to this therapy, but again found in humans are summarized in Table 5.
this observation must be confirmed in controlled trials (320, Detection of microsporidia by transmission electron micros-
321). Symptomatic improvement has been achieved with oct- copy is highly specific, but the technique may lack sensitivity,
especially when performed on body fluids and stool samples.
reotide, but this drug has no effect on the parasites (325).
However, large studies to evaluate the sensitivity of transmis-
Combination antiretroviral therapy that includes a protease
sion electron microscopy are lacking (62, 73). Likewise, sample
inhibitor can restore immunity to microsporidia. The use of
preparation and examination are laborious and time-consum-
potent antiretroviral therapy in patients with advanced HIV
ing (62, 73).
infection can improve symptoms due to microsporidiosis and in
some cases leads to disappearance of the parasites (61, 75b,
140, 160a). However, the rapid time to release in patients with Light Microscopy
declining CD4 lymphocyte counts suggest that the microspo- Histologic examination of biopsy specimens or cytologic ex-
ridial infections are not eradicated. amination of body fluids by light microscopy allows diagnosis
Since no effective therapy is available for E. bieneusi infec- of microsporidial infection, but genus or species differentiation
tion whereas infections with Encephalitozoon spp. respond very is uncertain (76, 227). The size of the spores and the distribu-
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 259

Special features

Vacuole

Nucleus

Arrangement of polar tube

No. of coils of polar tubule

Size of spores (mm)


Characteristic
Electron-lucent

No vacuole, in direct

Unikaryotic

Two rows

5–7

1.1–1.6 by 0.7–1.0
sporonts
beginning in the
polar tubule
development of the
inclusion,

cell cytoplasma
contact with the host

E. bieneusi

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


TABLE 5. Morphological characteristics of microsporidia infecting humans
Septated parasitophorous

Unikaryotic

One row

5–7

2.0–2.5 by 1.0–1.5
intestinalis
vacuole in E.

Encephalitozoon spp.

FIG. 8. Encephalitozoon cuniculi spores in conjunctival swab from an HIV-


infected patient with disseminated infection. Modified chromotrope-based stain.
Magnification, 3870.

tion pattern of the infection may be of limited use, but exact


species differentiation is impossible.
Cytologic diagnosis and stool examination. Microsporidian
Diplosporoblastic

No vacuole, in direct

Diplokaryotic

One row

7–12

2.5–5.0 by 2.0–2.5
sporogony

cell cytoplasma
contact with the host

spores have been detected in several body fluids and stool


samples (264, 347). The outer layer of the spore wall (exo-
Nosema spp.

spore) is proteinaceous, and the inner layer (endospore) is


chitinous (26, 50) so that Gram, Giemsa, and trichrome stains,
as well as fluorescent dyes, have been advocated for staining
microsporidian spores (74). Gram and Giemsa stains are not
suitable for cytologic diagnosis because they do not differen-
tiate between microsporidia and other elements present in
body fluids or stool specimens that can be confused with mi-
Tetrasporoblastic

No vacuole, in direct

Diplokaryotic

One row

5–7

3.05–4.55 by 0.77–1.27

crosporidian spores (75). Microscopic examination of body


reticulum
endoplasmatic
cisterna of host
surrounded by a
sporonts, all stages are
sporogony, band-like

cell cytoplasma
contact with the host

fluids to diagnose microsporidia did not become routine until


special chromotrope-based and fluorescent stains were devel-
V. corneae

oped. Nowadays these chromotrope- and/or fluorochrome-


based stains are used in several modifications.
The chromotrope-based stain developed by Weber et al.
(366) has markedly improved spore detection in stool samples
and body fluids. In this technique, involving a modification of
the trichrome stain with a concentration of chromotrope 2R
that is 10 times higher than that used in the trichrome stain,
Multinucleate

Sporophorous vesicle

Unikaryotic

Two rows

9–12

3.2–3.4 by 2.8

microsporidian spores stain characteristic pinkish red. Usually


plasmodia
sporogonial

Pleistophora spp.

the spores have a characteristic appearance when examined


under high-power magnification (31,000). The spore wall
stains intense red, and some spores show a distinct beltlike
stripe that grids the spores diagonally or equatorially (Fig. 8)
(366). This staining technique is lengthy, and spores are diffi-
cult to detect if only a few are present in the sample (94).
Several modifications (changes in temperature and staining
time [203], decrease in the phosphotungstic acid level, and
No multinucleate

Unikaryotic
Sporophorous vesicle

One or two rows

11

4.0 by 2.4
Trachipleistophora spp.

substitution of a color-fast counterstain [294]) of the chromo-


antropophtera
development of T.
are formed during
vesicles and spores
sporophorous
different types of
plasmodia, two
sporogonial

trope-based stain have been suggested for speeding the process


and for better contrast between the spores and the back-
ground. The improved hot Gram-chromotrope technique pro-
vides some real advantages in staining microsporidia for light
microscopy. In this procedure, samples are stained in solutions
of crystal violet and iodine used in Gram’s stain and then in a
260 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

FIG. 10. Paraffin-embedded duodenal biopsy specimen from a patient with

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


AIDS with intestinal Enterocytozoon bieneusi infection. The microsporidial
spores are easily visualized within the enterocytes. Fluorescence microscopy after
Uvitex 2B stain. Magnification, 31,000.

Whether concentration techniques are useful for detection of


microsporidian spores in stool specimens remains controver-
FIG. 9. Encephalitozoon intestinalis spores derived from in vitro culture. Flu- sial. Some authors reported that concentration techniques such
orescence microscopy after Uvitex 2B stain. Magnification, 31,000. as the formalin-ethyl acetate concentration procedure or dif-
ferent flotation methods led to a substantial loss of microspo-
ridial spores and thus to false-negative results (62, 366). In
contrast, others found that concentration techniques such as
modified chromotrope solution heated to 50 to 55°C. With this the water-ether sedimentation or centrifugation of KOH-
stain, microsporidian spores are stained dark violet against a treated stool samples increased the yield of microsporidian
pale green background and the total staining time is shortened spores (62, 349). Therefore, one of these concentration tech-
to 5 min (251). niques should be used for stool samples.
The epifluorescence of microsporidian spores stained with Histologic diagnosis. Because of the small size of the spores,
optical brighteners was a second major breakthrough for de- reliable visualization of microsporidia by light microscopy de-
tecting spores in stool samples and body fluids (74, 75, 228, pends on a distinct contrast between spores and other cellular
348, 349). Of these fluorescent stains, Uvitex 2B, Calcofluor contents. Routine hematoxylin-and-eosin stained parasites in
white, and Fungifluor (a formulation of Calcofluor white in tissue are easily overlooked even by experienced pathologists
10% KOH) are the stains of first choice, although Calcofluor (241). Tissue Gram stains such as Brown-Brenn or Brown-
white produces somewhat greater background staining (74). Hopps seem to be very useful for reliable identification of
The staining procedure is easy and quick, but examination microsporidia in paraffin-embedded tissue sections (206, 267).
requires a fluorescent microscope with a 350- to 380-nm exci- Microsporidian spores are Gram variable, but with these stains
tation filter and a high-magnification objective lens (magnifi- they stain dark blue or red against a faint brown-yellow back-
cation, 31,000). Fluorochromes bind to the chitin of the en- ground (206). Fluorescent staining techniques with optical
dospore, and when excited under UV light, the bound dye brighteners are easy and quick to perform on tissues, and the
fluoresces brightly in the visible spectrum (74). Spores are sensitivity of these stains seems to be very high (Fig. 10) (74,
identified by their size, shape, and fluorescent staining prop- 146). A major advantage of these stains is that they can be
erties (Fig. 9) (74, 348). Several modifications of the fluores-
combined with other staining techniques (74). Silver stains
cent stain with Uvitex 2B, originally described by van Gool et
such as the Warthin-Starry stain (136, 137) or a modified chro-
al. (348), were introduced to use on touch preparations (74,
motrope-based trichrome stain (160) are preferred by some,
356), brush cytologic specimens (270), and smears of several
body fluids (74, 107) and for staining of paraffin-embedded but both techniques are time-consuming and interpretation of
material (74, 146). Uvitex 2B was used a number of years ago sections may be difficult.
for detecting nonhuman microsporidia in tissue sections (355). Giemsa, chromotrope 2R, or fluorochrome staining of touch
Although fluorescent stains seem to be more sensitive than preparations of intestinal tissue and of endoscopic brush cyto-
chromotrope-based trichrome stains, they may lead to some logic specimens is useful in the diagnosis of intestinal micros-
false-positive results due to the similarity in staining of small poridiosis, but these techniques require fresh material (15, 74,
yeast cells (74, 75, 94, 228). In addition, other studies have not 270, 288, 309).
shown superior sensitivity of the fluorescent stains over the Semithin sections of resin-embedded biopsy material,
chromotrope-based stains (107, 182). Most authors conclude stained with a variety of different stains (hematoxylin and eo-
that the two techniques should be used simultaneously to en- sin, Giemsa, toluidine blue, methylene blue-azure, and basic
hance performance and to provide greater accuracy, especially fuchsin), are useful methods for visualization of spores and
for patients with light infections (94, 107, 182). tissue stages of microsporidia. However, resin embedding of
Because the number of microsporidia in clinical samples can biopsy specimens is not routinely used in most laboratories
be very small, centrifugation of body fluids may be necessary. (Fig. 11) (263, 267, 273, 309).
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 261

Animal Models

Animal models provide a basis for studying immune re-


sponses and for evaluating diagnostic methods, vaccine candi-
dates, therapeutic strategies, and routes of transmission (106).
Furthermore, they are essential for producing poly- and mono-
clonal antibodies (307, 309, 359, 360). Several animal models
have been established to study microsporidial infections (50,
80, 106, 153, 156, 209, 236, 313, 343, 384). Most of these models
used E. cuniculi as pathogen. This organism had long been
recognized as an important cause of latent infections in labo-
ratory rodents, sometimes complicating the interpretation of
experimental results obtained with these animals (50, 51, 80).
BALB/c and C57Bl/6 athymic mice have been used as animal
models and have been infected intraperitoneally with E. cunic-
uli, E. hellem, or V. corneae (106, 156). SCID mice have also

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


been infected by oral inoculation of E. cuniculi spores. This
animal model was used to study the in vivo efficacy of albenda-
zole against E. cuniculi (209). Experimental E. cuniculi infec-
tions in immunocompetent mice produced only chronic asymp-
tomatic infection. Successful transmission of E. cuniculi to
rabbits by administration of spores orally and rectally has been
reported (80, 153). Simian immunodeficiency virus-infected
rhesus macaque monkeys have been also infected with E. cu-
FIG. 11. Resin-embedded semithin (1-mm) section of duodenal mucosa from
a patient with AIDS and intestinal Enterocytozoon bieneusi infection. Epithelial niculi, E. hellem, and V. corneae per os (106).
cells contain spores of Enterocytozoon bieneusi. Toluidine blue stain. Magnifica- Animal models for E. bieneusi infection are difficult to es-
tion, 3800. tablish. Attempts to infect immunosuppressed gnotobiotic pig-
lets, gamma interferon knockout mice, and SCID mice treated
with anti-gamma interferon monoclonal antibodies with E. bie-
Cell Culture neusi have been unsuccessful. Recently, experimental oral
transmission of E. bieneusi to simian immunodeficiency virus-
The in vitro cultivation of several microsporidian species infected rhesus monkeys has been reported (343).
that infect humans has been of enormous benefit, both for our
understanding of the biologic aspects of the host cell-parasite
relationship and for the development of immunologic reagents Antigen-Based Methods
for diagnosis and species differentiation. In vitro cultures have
been also used to assess the effects of antimicrobial agents on Microsporidium-specific antibodies in immunofluorescence
several microsporidian species including E. cuniculi, E. hellem, tests have been used for the diagnosis and species differentia-
and E. intestinalis (16, 54, 117, 141, 168, 218, 219). In vitro tion of microsporidia. Poly- and monoclonal antibodies were
cultures combined with ultrastructural, biochemical, antigen, also used for Western blot analysis of several microsporidian
or molecular analyses have been used to confirm infections species (4, 84, 85, 132, 198–200, 256, 259, 261, 304–309, 358–
with existing species of microsporidia (95, 177–179, 358–360), 360, 379, 397).
as well as to define new species (104, 180, 317). However, their Immunofluorescent antibody tests involving polyclonal anti-
use in routine clinical diagnosis is not practical because they sera against E. hellem, E. cuniculi, and E. intestinalis produced
are time-consuming and laborious and only specialized labo- in mice or rabbits showed that several species of microsporidia
ratories maintain cell cultures with microsporidia. demonstrated immunological cross-reactivity (4, 259, 359, 360,
Microsporidia have been successfully cultivated in a number 397). This cross-reactivity of the polyclonal antisera against
of mammalian cell lines including monkey and rabbit kidney Encephalitozoon spp. was used for the detection of several
cells (Vero and RK13), human fetal lung fibroblasts (MRC-5), microsporidian species including E. bieneusi in various clinical
MDCK cells, and several other cell lines (50, 102, 177–179, samples by using different immunofluorescence tests (4, 259,
275, 350, 354). Species that have been cultivated in vitro from 359, 360, 397). However, the cross-reactivity of the antisera
a variety of human specimens include E. hellem (102, 178, 359), limit their use as diagnostic tools because species differentia-
E. cuniculi (95, 177, 179, 312), E. intestinalis (122, 151, 350, tion is not possible with these reagents.
360), V. corneae (317), and T. hominis (180). Attempts to Several monoclonal antibodies which recognized E. hellem
culture Enterocytozoon bieneusi from small intestinal biopsy (4, 359), E. cuniculi, or E. intestinalis (17) were developed.
specimens laden with merogonic stages and spores by using a Most of these monoclonal antibodies are specific to spore
range of cell lines and pretreatments have had limited success antigens (4, 227a, 359), whereas other researchers used polar
(54); to date, E. bieneusi has been propagated only in short tube protein-reactive monoclonal antibodies in combination
term cultures (6 months) (361). In the culture systems (human with monoclonal antibodies that recognize the surfaces of
lung fibroblasts and Vero monkey kidney cells) used, E. bie- spores (17). Some of these monoclonal antibodies are species
neusi seems to exert a greater cytotoxic effect than has been specific, whereas others react against spore walls or the polar
observed with cell cultures of Encephalitozoon spp. The inabil- tubes of several microsporidian species (227a).
ity to grow E. bieneusi in a continuous-culture system may Mono- and polyclonal antibodies are useful tools for species
reflect a need of this organism for some specific nutritional differentiation of microsporidia in different clinical samples
requirements that are not provided by the cell cultures used so (307), but antibody-staining techniques may be less sensitive
far (361). than other techniques. Didier et al. (107) compared a chromo-
262 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

able. Whether antigens of Encephalitozoon spp. cross-react


with those of E. bieneusi has not been determined exactly so far
(259, 302, 379, 397).
However, serologic methods are not useful as diagnostic
tools for microsporidiosis because at least half of the serum
samples from persons without a history of microsporidial in-
fection may have positive titers and immunosuppressed per-
sons may have a poor response to antigen challenge.

MOLECULAR METHODS
Molecular studies of microsporidia are in their infancy. A
limited number of genes have been located, and only a few
DNA sequences have been reported in a few microsporidian
species so far.
FIG. 12. Indirect immunofluorescent staining of Encephalitozoon cuniculi
Compared with other eukaryotes, microsporidia have ex-

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


spores in nasal discharge of an HIV-infected patient with disseminated infection,
using polyclonal anti-Encephalitozoon cuniculi antiserum. Magnification, 3400. tremely small genomes, often in the size range of bacterial
genomes. Pulsed-field gel electrophoresis studies of the karyo-
types of several microsporidian species showed that the hap-
loid genome usually ranges from only 5.3 to 19.5 Mb, which
trope-based stain, a fluorescent stain containing Calcofluor represents the largest microsporidian genome measured to
white, and a fluorescent polyclonal antibody stain. The fluo- date in Glugea atherinae (23, 24, 252). However, the haploid
rescent polyclonal antibody stain was the least sensitive genome of E. cuniculi was estimated to be only 2.9 Mb, which
method for detecting microsporidial spores in stool samples, is smaller than all the other microsporidian genomes. This size
urine, and duodenal fluid (107). Therefore, antibodies should is about half that of the previously smallest known microspo-
be used for species differentiation in samples only when the ridian genome, 5.3 Mb in Nosema locustae (23). Eleven chro-
initial diagnosis of microsporidiosis by using fluorescent stains mosomal DNA bands obtained by pulsed-field gel electro-
with optical brighteners and/or chromotrope-based stains has phoresis with DNA isolated from E. cuniculi spores ranged
been made (Fig. 12) (107). E. bieneusi-specific antibodies have only from 217 to 315 kb. However, the chromosome number of
not been developed so far. E. cuniculi was larger than that of a Vairimorpha sp. and
Nosema costelytrae, both of which have only eight chromo-
somes (231). The very small genome in E. cuniculi may be
Serologic Testing
related to the early divergence of microsporidia, but it may be
A variety of serological tests (carbon immunoassay, indirect also a derived characteristic, perhaps related to the highly
immunofluorescence test, enzyme-linked immunosorbent as- adapted parasitic lifestyle of this organism.
say, counterimmunoelectrophoresis, and Western blotting) Chromosomal localization of genes in microsporidia were
have been developed to detect IgG and IgM antibodies to done for only a few genes in a limited number of species. Eight
microsporidia, especially to E. cuniculi (18, 21, 22, 174–176, genes in E. cuniculi have been localized so far. Each of the 11
327, 352, 353, 379, 383). Some of these tests are commonly chromosomes of E. cuniculi contains rDNA, which strongly
used to detect antibodies in several animal species (18, 32, 170, contrasts with the location of rDNA within a single but rather
175). Of these assays, the indirect immunofluorescence test large chromosome (760 kb) in Nosema bombycis, an insect
and enzyme-linked immunosorbent assay are probably the microsporidium with a genome of 15.3 Mb (25). Further anal-
most useful because they are easy to perform, but the sensi- ysis of eluted DNA from each E. cuniculi chromosome by
tivity and specificity of all these tests are unknown (174–176). restriction enzyme digestion and rDNA hybridization showed
Antibodies to E. cuniculi and E. intestinalis have been found in identical patterns, suggesting the presence of a single rDNA
humans with and without HIV infection (174, 176, 327, 352, unit copy per chromosome (25). However, most probes were
353), but it is uncertain whether these represent true infection, assigned to single chromosomes, indicating a prevalence of
cross-reactivity with other species, or nonspecific reaction. low-copy-number nucleotide sequences within the very small
Serologic surveys for antibodies to E. cuniculi have sug- genome of E. cuniculi. Both b-tubulin and aminopeptidase
gested a possible link between exposure to a tropical environ- genes were located on two different chromosomes (the b-tu-
ment and infection with microsporidia. In patients with malaria bulin gene on chromosomes II and III, and the aminopeptidase
and schistosomiasis, the microsporidial seropositivity rate was gene on chromosomes I and VIII), whereas thymidylate syn-
4.7 and 9.1%, respectively (176). A study of homosexual men in thase, dihydrofolate reductase, and serine hydroxymethyl
Sweden reported that 10 of 30 persons (33%) were seroposi- transferase genes were located only on chromosome I, cdc2-
tive for antibodies to E. cuniculi; all the seropositive patients like and ERCC6-like genes were located only on chromosome
had at some time visited a tropical area (22, 176). Explanations VIII (25), and an Hsp70 gene was located only on chromosome
for this apparent relationship remain speculative, and clinico- XI (275a).
pathologic correlations have not been reported for any of these
serologic surveys. Of interest, a high seroprevalence of anti-
Small- and Large-Subunit rRNA Genes of Microsporidia
bodies against E. intestinalis were observed among Dutch
blood donors (24 of 300 [8%]), pregnant French women (13 of Although microsporidia are true eucaryotic organisms with a
276 [5%]), and patients with various infectious and noninfec- nucleus, an intracytoplasmatic membrane system, and chromo-
tious diseases (6 of 150 [4%]) (353). some separation by mitotic spindles, their rRNA genes show
For E. bieneusi, which has not been propagated in long-term features related to prokaryotic sequences (86, 362). They are
cell cultures so far, specific serologic assays remain unavail- composed of a 16S SSU rRNA and a 23S LSU rRNA sepa-
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 263

rated by an intergenic nontranscribed spacer (363). In contrast and there are significant differences between the SSU rRNA
to true eukaryotic organisms, microsporidia lack a separate sequence determined by Zhu et al. and that obtained by others
5.8S rRNA, which is believed to be a universal eukaryotic (10, 164, 359, 364) (accession no. L07255, L17072, L39107,
characteristic, and also a second intergenic spacer (363–365). X98469, X98470, and X98467) which includes also bases in
As in the prokaryotes, microsporidia have LSU rRNA whose conserved regions of the rRNA gene. Again, some dissimilar-
59 region corresponds to the 5.8S rRNA of eukaryotes (363– ities may be caused by polymorphisms, but dissimilarities in
365). The SSU and LSU rRNA genes are shorter than typical conserved regions indicate that the sequence reported by Zhu
eukaryotic SSU and LSU rRNA genes and lack several uni- et al. contained several sequencing errors.
versal sequences (158a, 275b, 363–365). The complete E. hellem SSU rRNA sequence data were first
The first sequence data of SSU-rRNA of a microsporidium, described by Visvesvara et al. (359). They used a primer pair
V. necatrix, were reported by Vossbrinck et al. in 1987 (363) based on nucleotides 1 to 21 (Micro-F [the nucleotide se-
(accession no. M24612). Subsequently, PCR amplification with quence of this primer is identical to that of primer V1]) and
primers complementary to conserved sequences of the V. ne- 1244 to 1218 (Micro-R) of the V. necatrix SSU rRNA gene to
catrix SSU rRNA gene has been used to amplify and sequence amplify, clone, and sequence the SSU rRNA gene of E. hellem,
DNA fragments of the SSU rRNA gene from several micro- E. cuniculi, and E. intestinalis (359, 360). The obtained se-
sporidia that infect humans (164–166, 359, 364, 365, 393–396). quences are deposited in the GenBank database under acces-

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


Several nucleotide sequences of SSU and LSU rRNA genes sion no. L19070, L17072, and U09929.
including the intergenic spacer regions of different microspo- SSU rRNA sequences of V. corneae (accession no. L39112
ridian species, including six species infecting humans (E. bie- and U11046) and T. hominis (accession no. AJ002605) are
neusi, E. cuniculi, E. hellem, E. intestinalis, T. hominis, and V. available via the GenBank database as well.
corneae), have been published and are accessible via the Gen- Vossbrinck et al. (364) used highly conserved primers 530f in
Bank and EMBL databases (October 1998) (Table 6). the SSU rRNA and 580r and 228r in the LSU rRNA to amplify
The rRNA sequences of E. bieneusi were first reported by the 39 end of the SSU rRNA gene, the intergenic spacer, and
Zhu et al. and deposited in the GenBank database under the 59 end of the LSU-rRNA gene of E. hellem (accession no.
accession no. L07123 and L20290 (395). Gene amplification L13331) and E. cuniculi (accession no. L13332). The same
was performed with DNA extracted from human intestinal primers were used by Zhu et al. (396) to amplify the same
biopsy specimens from HIV-infected patients with electron region of SSU and LSU rRNA genes including the intergenic
microscopy-confirmed E. bieneusi infection. One primer pair spacer of E. bieneusi (accession no. L20290) and E. intestinalis
was in a conserved region of the SSU rRNA (V1 and 1492), (accession no. L20292) (396). The 39 end of the SSU-rRNA
and a second was in a conserved region of the SSU rRNA gene, the intergenic spacer, and the 59 end of the LSU-rRNA
(530f) and LSU rRNA (580r). Primer V1 is based on the gene of several microsporidian species found in humans have
sequence of the SSU rRNA gene of V. necatrix at the 59 end, now been sequenced by many researchers and are available via
and primer 1492 is based on rRNA sequences highly conserved the GenBank or EMBL database (Table 6).
across many organisms at the 39 end. An amplification product To date, complete LSU rRNA sequences have been deter-
of about 1,300 bp was obtained, cloned, and sequenced (395). mined only for E. cuniculi (accession no. AJ005581) and N.
Resequencing of the SSU and LSU rRNA regions of different apis (accession no. U97150). The sizes were estimated to be
E. bieneusi isolates by other researchers (89, 165, 289, 361) about 2,487 and 2,480 bp, respectively, which is in the size
(accession no. L16868, and AF024657) showed a 2.3 to 2.7% range of prokaryotic 23S rRNA; this shortening is essentially
dissimilarity even in conserved regions with respect to the due to truncation of divergent domains, with most of them
sequences reported by Zhu et al. Some of these differences being removed (158a, 275b). Most variable stems of the con-
may be caused by polymorphisms, but many are probably due served core are also deleted, reducing the LSU rRNA to only
to unresolved sequencing artifacts in the sequence reported by the structural features preserved in all living cells (275b).
Zhu et al. (395).
E. intestinalis SSU rRNA sequences were reported by sev-
eral researchers (10, 166, 360, 394, 396). Zhu et al. used primer a- and b-Tubulin Genes of Microsporidia
pair V1 and 1492 to amplify a 1,300-bp DNA fragment from
intestinal biopsy specimens with electron microscopy-con- The tubulin gene family consists of three distinct but highly
firmed E. intestinalis infection (394, 396). The DNA fragment conserved subfamilies, a-, b-, and g-tubulin. a- and b-tubulins
was cloned and sequenced, and the obtained sequence was are the most abundant in eukaryotic cells and have been stud-
deposited in the GenBank database under accession no. ied most extensively. Microtubules are a characteristic feature
L19567 (394, 396). Resequencing of the SSU rRNA region of of all eukaryotic organisms, since they are major components
different E. intestinalis isolates by other investigators (10, 166, of the cytoskeleton and the mitotic spindle. Microtubules form
360) (accession no. U09929, L39113, L16866, and L16867) by polymerization of tubulin, a dimer of a- and b-subunits,
showed several dissimilarities to the sequence reported by Zhu each approximately 440 amino acids long (125, 189). The func-
et al. Although some may be caused by polymorphisms, dis- tion of g-tubulin is less clear, although it is known to be im-
similarities are also reported in conserved regions of the SSU- portant in microtubule-organizing centers and has been impli-
rRNA, thus indicating that the E. intestinalis sequence of Zhu cated in several other processes. About 100 b-tubulin
et al. may contain several sequencing errors. sequences as well as many a-tubulin sequences from a wide
The E. cuniculi SSU rRNA sequence was also first reported range of eukaryotic cells have been reported. In contrast to
by Zhu et al. and was deposited in the GenBank database rRNA sequences and many other proteins, deletions or inser-
under accession no. Z19563 (393). They amplified the SSU- tions are rare in tubulin genes, so that there are no problems
rRNA gene with primer pair V1 and 1492 from rabbit kidney in alignment. Phylogenetic analysis with a- and b-tubulin se-
cells (RK-13) infected with E. cuniculi in vitro. A 1,200-bp quences is consistent in several respects with rRNA-based phy-
DNA fragment was amplified, cloned, and sequenced (393). logenetics but has generated different results for some amito-
Several further SSU and LSU rRNA sequences of different E. chondrial protozoa including microsporidia (125).
cuniculi strains have been deposited in the GenBank database, b-Tubulin sequences of E. cuniculi (accession no. L31807)
264 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

TABLE 6. Human microsporidian genes in GenBanka


Accession no. Gene Organism Reference(s)

X98470 SSU rRNA E. cuniculi Donovan 181


X98469 SSU rRNA E. cuniculi Stewart (host, Canis familiaris) 181
X98467 SSU rRNA E. cuniculi D. Owen (host, Mus musculus) 181
L39107 SSU rRNA E. cuniculi 10
L17072 SSU rRNA E. cuniculi 359
L13295 SSU rRNA E. cuniculi partial cds 364
Z19563 SSU rRNA E. cuniculi 393
L07255 SSU rRNA E. cuniculi 164
AJ005581 SSU rRNA E. cuniculi 275b
L13393 SSU rRNA E. hellem partial cds 364
L39108 SSU rRNA E. hellem 10
L19070 SSU rRNA E. hellem 359
AF039229, AF039230 SSU rRNA E. hellem (host, Electus roratus) partial cds 281a
U39297 SSU rRNA E. intestinalis partial cds 148
U09929 SSU rRNA E. intestinalis 360

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


L39113 SSU rRNA E. intestinalis 10
L19567 SSU rRNA E. intestinalis 394
L16867 SSU rRNA Encephalitozoon sp. 166a
L16866 SSU rRNA Encephalitozoon sp. 164
L16868 SSU rRNA E. bieneusi 165
L07123 SSU rRNA E. bieneusi 395
AF023245 SSU rRNA E. bieneusi (host, Macaca mulatta) 234a
U61180 SSU rRNA E. bieneusi (host, pig) 98
AF024657 SSU rRNA E. bieneusi 89
AJ002605 SSU rRNA T. hominis 66a
L39112 SSU rRNA V. cornea 10
U11046 SSU rRNA V. cornea 276a
X98466 39-end SSU, ITS,b 59-end LSU E. cuniculi Donovan 181
X98468 39-end SSU, ITS, 59-end LSU E. cuniculi D. Owen (host, Mus musculus) 181
L29560 39-end SSU, ITS, 59-end LSU E. cuniculi 189
L13332 39-end SSU, ITS, 59-end LSU E. cuniculi 364
AJ005581 39-end SSU, ITS, 59-end LSU E. cuniculi 275b
AF069064 39-end SSU, ITS, 59-end LSU E. hellem 151a
L13331 39-end SSU, ITS, 59-end LSU E. hellem 364
L29556 39-end SSU, ITS, 59-end LSU E. hellem 189
AF069064 39-end SSU, ITS, 59-end LSU E. hellem 151a
L20292 39-end SSU, ITS, 59-end LSU E. intestinalis 396
Y11611 39-end SSU, ITS, 59-end LSU E. intestinalis 301a
L20290 39-end SSU, ITS, 59-end LSU E. bieneusi 396
U61180 39-end SSU, ITS, 59-end LSU E. bieneusi (host, pig) 98
AF023245 39-end SSU, ITS, 59-end LSU E. bieneusi (host, Macaca mulatta) 234a
AF059610 39-end SSU, ITS, 59-end LSU E. bieneusi (host, Canis familiaris) 236a
AJ005581 LSU rRNA E. cuniculi 275b
U66908 a-Tubulin E. hellem 192
L47271 b-Tubulin E. hellem 221
L31808 b-Tubulin E. hellem partial cds 125
L31807 b-Tubulin E. cuniculi partial cds 125
L47274 b-Tubulin E. intestinalis partial cds 221
AJ005666 Polar tube protein E. cuniculi 95a
AF044915 Polar tube protein 55 precursor E. hellem 200
AF054829 Ribosomal protein L27a gene E. cuniculi 25a
AJ005644 Dihydrofolate reductase E. cuniculi 124a
AJ005644 Serine hydroxymethyltransferase E. cuniculi 124a
AJ005644 Aminopeptidase E. cuniculi 124a
AJ005644 Thymidylate synthase E. cuniculi 124a
AF031701 Actin (ACT1) E. hellem 126a
a
For a more complete list including microsporidian species not infecting humans, see reference 381a.
b
ITS, internal transcribed spacer.

and E. intestinalis (accession no. L47274) and a- and b-tubulin U66907) and Spraguea lophii (U66906) have been determined
sequences of E. hellem (accession no. U66908, L47271, and and are available via the GenBank or EMBL databases (192).
L31808) were amplified with primers corresponding to con-
served sequences (125, 126, 188, 189, 192, 221). Southern blots Other Genes of Microsporidia
indicate that E. cuniculi, E. hellem, and E. intestinalis possess a
single b-tubulin gene copy and that the three b-tubulin se- Besides rRNA and a- and b-tubulin genes, only those en-
quences are very similar to each other (125, 188, 189). coding isoleucyl-tRNA synthase (accession no. L37097) and
a-Tubulin sequences of Nosema locustae (accession no. glutamyl-tRNA synthetase in N. locustae (AF005490 and
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 265

AF005489) (35), homologues of U2 spliceosomal RNA in V. same incubator) or to contamination of the culture with a
necatrix (115) and of U2 and U6 spliceosomal RNA in N. plasmid containing the cloned E. bieneusi SSU rRNA gene
locustae (AF053588 and AF053589) (133a), reverse transcrip- (81). The V1-EB450 primer pair reliably amplifies E. bieneusi
tase (AF019229), chitin synthase (AF019228), and DNA-di- DNA from gastrointestinal biopsy specimens from patients
rected RNA polymerase II (AF019227) in S. lophii (172a), with light and electron microscopy-confirmed E. bieneusi in-
translation elongation factors EF-1a (D32139) and EF-2 fection (81, 145, 149, 381, 395) and from stool specimens from
(D79220) in Glugea plecoglossi (186, 187), a chaperone protein patients with light microscopy-confirmed intestinal microspo-
Hsp70 in N. locustae (U97520), V. necatrix (AF008215), E. ridiosis (262, 339a). Further evaluation of this primer pair gave
hellem, and E. cuniculi (159, 173, 275a), actin (ACT1) in E. negative results with one bile sample that was confirmed to
hellem (AF031701) and N. locustae (AF031702), a polar tube contain E. bieneusi by electron microscopy and with E. bieneusi
protein in E. cuniculi (AJ005666) (95a) and E. hellem derived from a short-term culture (89). Resequencing of the
(AF044915) (200), a ribosomal protein (AF054829), dihydro- SSU rRNA region of different E. bieneusi isolates by other
folate reductase, serine hydroxymethyltransferase, aminopep- researchers showed a 2.3 to 2.7% dissimilarity to the sequence
tidase, and thymidylate synthase (AJ005644) in E. cuniculi reported by Zhu et al. (395), resulting in a base mismatch of
(25a, 124a) have been partially characterized in microsporidia. primer EB450 (position 9 of the EB450 primer has G in place
of C) (89). The upstream primer V1 is not specific for E.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


bieneusi but is directed against a conserved sequence for many
DNA Isolation Techniques microsporidian species (381). The base mismatch of primer
EB450 combined with this lack of specificity may lead to neg-
Microsporidian DNA can be easily extracted from tissue
ative results with some samples. An internal 30-mer oligonu-
samples or in vitro cultures by routine procedures such as
cleotide, EB150, complementary to a region of the amplicon
proteinase K digestion followed by phenol-chloroform extrac-
produced by V1 and EB450 has been used as a probe for
tion and ethanol precipitation or by DNA purification methods
Southern blot hybridization analysis of the PCR products (81,
with commercial kits such as Magic Minipreps (Promega
145, 149, 381, 395) by isotopic and nonisotopic procedures
Corp.) or QIAmp tissue kits (Qiagen) (81, 91, 145, 148, 149,
(145, 149, 395). Probe EB150 hybridized with E. hellem DNA
236).
when amplified by V1 and EB450 (149, 395); again, it is likely
DNA isolation from microsporidian spores is more difficult,
that this was due either to contamination of the original E.
and harsh conditions must be used to destroy the spore wall.
hellem culture with spores of E. bieneusi or to contamination of
Mechanical disruption of the spores with glass beads in com-
the culture with a plasmid containing the cloned E. bieneusi
bination with proteinase K digestion is commonly used (109,
SSU rRNA gene (81).
110, 134). Some authors recommend additional incubation
The primer pair V1-EB450 and a second pair, V1-Mic3
with enzymes that resolve chitin (lyticase or chitinase) (134) or
(Mic3 is located at positions 445 to 427 of the SSU rRNA gene
boiling the samples to release DNA from spores (260, 262).
of E. bieneusi), have been used to confirm E. bieneusi infection
If stool samples are used, treatment of stool with 0.5%
in simian immunodeficiency virus-infected rhesus monkeys.
sodium hypochlorite, 10% formalin, or 1 M KOH is recom-
The specificity of the PCR products was confirmed by DNA
mended to remove PCR inhibitors (134, 190, 222), whereas
sequencing and restriction fragment length polymorphism
others have reported that inhibition can be nullified by simple
analysis with MnlI and DdeI restriction endonucleases (343).
dilution of the samples (262).
The primer pair V1-Mic3 has also been used to amplify a
446-bp fragment from the E. bieneusi SSU rRNA gene of
Molecular Techniques for Diagnosis and HIV-infected patients with intestinal microsporidiosis due to
Species Differentiation E. bieneusi (63).
The primer pair EBIEF1 and EBIER1 described by Da Silva
Several PCR-based methods have been published to amplify et al. (89) amplified cloned E. bieneusi SSU rRNA sequences
different regions of the SSU and LSU rRNA gene as well as and DNA from bile fluid, a duodenal aspirate from an AIDS
the intergenic spacer region for diagnosis and species differ- patient with electron microscopy-confirmed E. bieneusi infec-
entiation of microsporidia infecting humans (135) and animals tion, and a short-term culture of an E. bieneusi isolate. Speci-
(232, 233) (Tables 7 and 8). ficity was tested with human DNA as well as with cloned
Primer pairs and hybridization probes for E. bieneusi. PCR microsporidian SSU rRNA coding regions of 13 species of
diagnosis of E. bieneusi was first reported by Zhu et al. (395). microsporidia, and positive amplification was shown only with
Primer pair V1 and EB450 amplified cloned E. bieneusi SSU the E. bieneusi cloned template. PCR products generated with
rRNA sequences and DNA from E. bieneusi-infected tissues this primer pair were analyzed only by ethidium bromide-
but also amplified E. hellem DNA from cell cultures (81, 145, stained gel analysis, and the identity of the PCR products was
149, 381, 395). Specificity was tested with DNA prepared from not confirmed by Southern blot hybridization, DNA sequenc-
Toxoplasma gondii, Trypanosoma cruzi, Escherichia coli, Sac- ing, or restriction enzyme digestion (89). Estimating the size of
charomyces cerevisiae, E. intestinalis, V. necatrix, E. cuniculi, the PCR-generated fragment by gel electrophoresis alone is
Glugea stephani, N. locustae, N. bombycis, Pleistophora, and E. usually not sufficient to confirm the specificity of a PCR. Every
hellem and with gastrointestinal biopsy specimens infected with primer that is nonspecifically elongated at its 39 end because of
E. cuniculi, E. intestinalis, Cryptosporidium spp., Giardia lam- unwanted annealing to partially complementary sequences not
blia, Mycobacterium avium, and Candida albicans (81, 145, 381, only is lost for specific priming but also enhances further non-
395). Only weak amplification of DNA prepared from E. hel- specific amplifications. This may lead to false-positive results,
lem was seen (395). In other studies, these primers did not which should be ruled out by the confirmation techniques
amplify DNA from E. hellem (81). Based on the SSU rRNA mentioned above.
sequence of E. hellem, amplification should not occur. It is An E. bieneusi-specific primer pair and hybridization probe
likely that the amplification observed by Zhu et al. (395) was were designed by Schuitema et al. (303) and further evaluated
due either to contamination of the original E. hellem culture by David et al. (91) and Liguory et al. (222). The primer pair
with spores of E. bieneusi (which were being cultivated in the and probe were used for species differentiation for samples
266 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

TABLE 7. Primer pairs and hybridization probes used for diagnosis and species differentiation of human microsporidia
Primer pair sequence
Primer designation Hybridization probe

59-CACCAGGTTGATTCTGCCTGAC-39 V1 59-TGTTGCGGTAATTTGGTCTCTGTGTGTAAA-39
59-ACTCAGGTGTTATACTCACGTC-39 EB450

59-GCCTGACGTAGATGCTAGTC-39 59-GTTCAATAGCGATGAGTTTGCTAATGTT-39
59-ATGGTTCTCCAACTGAAACC-39

59-GAAACTTGTCCACTCCTTACG-39 EBIEF1
59-CCATGCACCACTCCTGCCATT-39 EBIER1

59-CACCAGGTTGATTCTGCCTGAC-39 V1
59-CAGCATCCACCATAGACAC-39 Mic3

59-TCAGTTTTGGGTGTGGTATCGG-39 Eb.gc
59-GCTACCCATACACACATCATTC-39 Eb.gt

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


59-CACCAGGTTGATTCTGCCTGAC-39 V1 59-TGTTGATGAACCTTGTGG-39
59-CTCGCTCCTTTACACTCGAA-39 SI500 59-TGGTAGTTTAGGGTAATGGCCTAACTAGGCG-39

59-GGGGGCTAGGAGTGTTTTTG-39 59-TTTGGGGGATTATGTCCTGATGTGGAT-39
59-CAGCAGGCTCCCTCGCCATC-39

59-TTTCGAGTGTAAAGGAGTCGA-39 SINTF1
59-CCGTCCTCGTTCTCCTGC-39 SINTR

59-CACCAGGTTGATTCTGCCTGAC-39 V1
59-CCTGCCCGCTTCAGAACC-39 Sep1

59-TGAGAAGTAAGATGTTTAGCA-39 EHEL-F
59-GTAAAAACACTCTCACACTCA-39 EHEL-R

59-ATGAGAAGTGATGTGTGTGTGCG-39 ECUN-F
59-TGCCATGCACTCACAGGCATC-39 ECUN-R

59-CACCAGGTTGATTCTGCCTGAC-39 PMP1 (V1)


59-CCTCTCCGGAACCAAACCTG-39 PMP2

59-CACCAGGTTGATTCTGCC-39 C1
59-GTGACGGGCGGTGTGTAC-39 C2

59-TGAATG(G/T)GTCCCTGT-39 MSP-1 Nested PCR (first PCR)


59-GGAATTCACACCGCCCGTC(A/G)(C/T)TAT-39 MSP-3
59-GTTCATTCGCACTACT-39 MSP-2B Nested PCR (second PCR)
59-CCAAGCTTATGCTTAAGTCCAGGGAG-39 MSP-4B

59-TGAATG(G/T)GTCCCTGT-39 MSP-1 Nested PCR (first PCR)


59-GGAATTCACACCGCCCGTC(A/G)(C/T)TAT-39 MSP-3
59-TCACTCGCCGCTACT-39 MSP-2A Nested PCR (second PCR)
59-CCAAGCTTATGCTTAAGT(C/T)(A/C)AA(A/G)GGGT-39 MSP-4A

59-CCAGGUTGATUCTGCCUGACG-39 Mic3U Nested PCR (first PCR)


59-TUACCGCGCCUGCUGGCAC-39 Mic421U

59-AAGGAGCCTGAGAGATGGCT-39 Mic266 Nested PCR (second PCR)


59-CAATTGCTTCACCCTAAGGTC-39 Eb379

59-AAGGAGCCTGAGAGATGGCT-39 Mic266 Nested PCR (second PCR)


59-CACCCCTTTGCACTCGCACAC-39 Ec378

59-AAGGAGCCTGAGAGATGGCT-39 Mic266 Nested PCR (second PCR)


59-TGCCCTCCAGTAAATCACAAC-39 Eh410

59-AAGGAGCCTGAGAGATGGCT-39 Mic266 Nested PCR (second PCR)


59-CCTCCAATCAATCTCGACTC-39 Ei395

59-TGCAGTTAAAATGTCCGTAGT-39 Int530f 59-TAGCGGCTGACGAAGCTGC-39


59-TTTCACTCGCCGCTACTCAG-39 Int580r 59-CGGGCAGGAGAACGAGGACGG-39
59-CGGGCAGGAGAACGAGGACGG-39
a
ISR, intergenic spacer region.
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 267

TABLE 7—Continued
Probe
Species Target gene Amplicon size (bp) Reference(s)
designation

EB150 E. bieneusi SSU rRNA 348 145, 149, 260, 262, 381, 395

E. bieneusi SSU rRNA 1,265 91, 222, 303

E. bieneusi SSU rRNA 607 89

E. bieneusi SSU rRNA 443 343

E. bieneusi SSU rRNA, ISRa, LSU rRNA 210 357

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


SI60 E. intestinalis SSU rRNA 370 81, 148, 149, 260, 262

E. intestinalis SSU rRNA 930 91, 222, 303

E. intestinalis SSU rRNA 520 90

E. intestinalis SSU rRNA 874 343

E. hellem SSU rRNA 547 359, 360

E. cuniculi SSU rRNA 549 359, 360

E. bieneusi, E. cuniculi, E. intestinalis, E. hellem SSU rRNA 250, 268, 270, 279 134

E. bieneusi, E. cuniculi, E. intestinalis, E. hellem SSU rRNA 1,200 286

E. bieneusi SSU rRNA, ISR, LSU-rRNA 508 190, 191

E. cuniculi, E. intestinalis, E. hellem SSU rRNA, ISR, LSU rRNA 289–305 190, 191

E. bieneusi, E. cuniculi, E. intestinalis, E. hellem SSU rRNA 410, 419, 421, 433 202

E. bieneusi SSU rRNA 132

E. cuniculi SSU rRNA 113 202

E. hellem SSU rRNA 134 202

E. intestinalis SSU rRNA 128 202

E. cuniculi SSU rRNA, ISR, LSU rRNA ca. 1,000 109


E. hellem
E. intestinalis
268 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

TABLE 8. Primer pairs used for phylogenetic analysis of human microsporidia


Approximate
Primer
Primer pair Species amplified Target gene amplicon size Reference(s)
designation
(bp)

59-CACCAGGTTGATTCTGCCTGAC-39 V1 E. bieneusi, E. intestinalis, SSU rRNA 1,200 363, 381, 395,


59-GGTTACCTTGTTACGACTT-39 1492 E. cuniculi, E. hellem, 396
other species

59-CACCAGGTTGATTCTGCC-39 18f E. bieneusi, E. intestinalis, SSU rRNA 1,300 10


59-TTATGATCCTGCTAATGGTTC-39 1537r E. cuniculi, E. hellem,
other species

59-CACCAGGTTGATTCTGCCTGA-39 Micro-F E. intestinalis, E. cuniculi, SSU rRNA 1,300 359, 360


59-TTATGATCCTGCTAATGGTTCTCCAAC-39 Micro-R E. hellem, other
species

59-CACCAGGTTGATTCTGCCTGA-39 Micro-F E. bieneusi SSU rRNA 1,200 361

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


59-CCAACTGAAACCTTGTTACGACTT-39 1492N4

59-CACCAGGTTGATTCTGCCTGACG-39 E. bieneusi, E. intestinalis, SSU rRNA 1,300 303


59-TTATGATCCTGCTAATGGTTCTCC-39 E. cuniculi, E. hellem,
other species

59-GTGCCAGC(C/A)GCCGCGG-39 530f E. bieneusi, E. intestinalis, SSU rRNA, 1,550, 1,350, 108, 364, 381,
59-GGTCCGTGTTTCAAGACGG-39 580r E. cuniculi, E. hellem, ISRa, 1,350, 1,350, 396
V. cornae, other LSU rRNA 1,300b
species

59-TGCAGTTAAAATGTCCGTAGT-39 int580f E. intestinalis, E. cuniculi, SSU rRNA, 1,000, 1,000, 108, 303
59-TTTCACTCGCCGCTACTCAG-39 int580r E. hellem, other ISR, 1,000b
species LSU rRNA
a
ISR, intergenic spacer region.
b
Sizes given for listed species respectively.

that were found to be PCR positive by using another primer priming reaction and used for in situ hybridization of paraffin-
pair, which amplifies different microsporidian species (see be- embedded jejunal biopsy specimens from patients with intes-
low) (91), and for diagnosis of intestinal infection with E. tinal E. bieneusi infection (63). The same technique was used
bieneusi by using DNA from stool samples (222). Examination with necropsy-derived tissue samples from simian immunode-
of 31 stool specimens from 26 patients with intestinal E. bie- ficiency virus-infected monkeys after experimental E. bieneusi
neusi infection, 6 stool specimens from 3 patients with E. in- infection (343). Histologic examination of tissue sections failed
testinalis infection, and 61 stool specimens from 45 patients to identify microsporidial infection, but in situ hybridization
without intestinal microsporidiosis showed a sensitivity and with the E. bieneusi probe revealed characteristic supranuclear
specificity of 100% (222). When this primer pair was used with staining of scattered villous-tip epithelial cells within the jeju-
target DNA from 25 gastrointestinal biopsy specimens from nal mucosa, which was also confirmed by electron microscopy
patients with confirmed E. bieneusi infection, 23 samples were (343).
found to be positive; no amplification was seen with DNA from Primer pairs and hybridization probes for E. intestinalis. A
three biopsy specimens from patients with confirmed E. intes- primer pair, V1 and SI500, described by Weiss et al. (381)
tinalis infection (91). amplified cloned E. intestinalis SSU rRNA sequences as well as
The unique rRNA intergenic spacer sequence of E. bieneusi DNA from E. intestinalis-infected tissues, body fluids, stool
was used to design a primer pair, Eb.gc and Eb.gt, which samples, and cell cultures (81, 148–150, 260, 262, 381). These
amplified a 210-bp DNA fragment (357). This primer pair was primers did not amplify cloned E. bieneusi SSU rRNA se-
used to amplify DNA from gastrointestinal biopsy specimens, quences or DNA from E. bieneusi- or E. cuniculi-infected tis-
stool samples, and duodenal aspirates of patients with E. bie- sue samples (81, 148). The specificity of this primer pair was
neusi infection but not from material infected with E. intesti- further tested with DNA prepared from a variety of microspo-
nalis, Isospora belli, Cryptosporidium spp., or G. lamblia. The ridian species, S. cerevisiae, and E. coli, and no amplification
identity of the amplified PCR products was not confirmed by was observed (81). To confirm the identity of the PCR prod-
Southern blot hybridization, DNA sequencing, or restriction ucts, an internal 18-mer oligonucleotide (SI60), that was used
enzyme digestion. Again, false-positive results which may oc- by Weiss et al. as upstream primer in combination with the
cur during unwanted annealing of primers to partially comple- reverse primer SI500 (381), was used for isotopic and noniso-
mentary sequences should be ruled out by the confirmation topic Southern blot hybridization by others (81, 148–150). An-
techniques mentioned above. other internal 30-mer oligonucleotide was used as a hybridiza-
To date, in situ hybridization has only occasionally been tion probe by others (262). Although the primer pair V1-SI500
used to diagnose infections with microsporidia. A 446-bp PCR- reliably amplified DNA products of the correct size from tis-
product, amplified with the E. bieneusi-specific primer pair sues from patients with electron microscopy-confirmed E. in-
V1-Mic3, was labeled with digoxigenin–11-dUTP by a random- testinalis infection, Franzen et al. (148) detected a PCR prod-
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 269

uct in biopsy specimens from patients with E. bieneusi infection Primer pairs for E. hellem and E. cuniculi. Two primer pairs
and in a biopsy specimen from a patient with E. cuniculi in- specific for either E. cuniculi (positions 344 to 364 and 872 to
fection. The PCR products hybridized with the E. intestinalis- 892 of the E. cuniculi SSU rRNA sequence [ECUN-F and
specific probe (SI60), and partial sequencing of the amplified ECUN-R]) or E. hellem (positions 358 to 378 and 884 to 904 of
DNA fragments showed high homology (96 to 100%) to pub- the E. hellem SSU rRNA sequence [EHEL-F and EHEL-R])
lished E. intestinalis sequences. This confirmed that the ampli- were designed for species-specific amplification of DNA frag-
fied PCR products were derived from the SSU rRNA gene of ments by Visvesvara et al. (359, 360). These two primer pairs
E. intestinalis and provided genetic evidence for latent E. in- were used for species differentiation of cultured organisms and
testinalis infection in these patients (148). These results were organisms present in various clinical samples (95, 297, 298, 359,
reinforced in another study which used primer pair V1-SI500 360).
in combination with the hybridization probe SI60 (149). A total Two primer pairs, originally designed for genus- and species-
of 46 HIV-infected patients with diarrhea were studied, and specific detection of Echinococcus multilocularis, surprisingly
PCR gave positive results for E. intestinalis in 10 patients and also amplified DNA of the SSU rRNA gene of E. cuniculi (154,
indicated five cases of double infection with E. intestinalis and 255). The PCR products amplified with these two primer pairs
E. bieneusi. Histologic examination by light microscopy showed were sequenced, and it was shown that the DNA of E. cuniculi
microsporidian spores in all cases, but light microscopy was carried segments that are characteristic for the genus Echino-

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


unable to distinguish between species in almost all cases; un- coccus and the species Echinococcus multilocularis. Perhaps E.
fortunately, electron microscopy results were available only for cuniculi infected host animals of E. multilocularis or E. mul-
3 of the 10 patients (149). Primer pair V1-SI500 in combina- tilocularis larvae, so that the DNA of E. cuniculi may unknow-
tion with the hybridization probe SI60 was also used to detect ingly have been cloned and sequenced in a former analysis, or
E. intestinalis DNA in blood from an HIV-infected patient with some genome sequences may be shared between E. cuniculi
disseminated infection (150). and E. multilocularis (154, 255).
Primer pair SINTF1 and SINTR, described by Da Silva et al. General primer pairs and hybridization probes for several
(90), amplified cloned E. intestinalis SSU rRNA sequences and microsporidian species. Primer pair PMP1 (the nucleotide se-
DNA from a duodenal-jejunal segment from a patient with quence of this primer is identical to that of primer V1) and
AIDS and suspected intestinal microsporidiosis and from eight PMP2 amplified four microsporidian pathogens that infect hu-
different cultured samples of E. intestinalis. Specificity was mans (E. bieneusi, E. hellem, E. cuniculi, and E. intestinalis)
tested with cloned microsporidian SSU rRNA coding regions (134). This primer pair was useful for the detection of micro-
of E. cuniculi, E. hellem, E. intestinalis, E. bieneusi, and V. sporidian DNA from cultured organisms and stool samples as
well as from several other clinical samples including gastroin-
corneae; positive amplification was shown only with the E.
testinal biopsy specimens (123, 134). These primers also am-
intestinalis SSU rRNA as the template. This primer pair was
plified V. corneae from culture (134). To confirm the identity of
also used for species differentiation of microsporidia in stool
PCR amplicons, restriction enzyme digestion of amplified PCR
samples from several animals (32a). PCR products generated
products with PstI and HaeIII was used. Since E. bieneusi does
by this primer pair were analyzed only by ethidium bromide-
not have a PstI restriction site in the amplified region but PstI
stained gel analysis, and the identity of the PCR products was
cuts the Encephalitozoon amplicons into two fragments, E.
not confirmed by Southern blot hybridization, DNA sequenc- bieneusi could be easily differentiated from Encephalitozoon
ing, or restriction enzyme digestion. Again, false-positive re- spp. However, E. intestinalis could not be differentiated from E.
sults cannot be ruled out without confirmation as mentioned cuniculi (134), thus limiting the use of this primer pair for
above. species differentiation. The same primer pair, PMP1-PMP2,
An E. intestinalis-specific primer pair and hybridization was used by Dowd et al. (123), and the detection sensitivity was
probe were designed by Schuitema et al. (303) and were fur- two spores in vitro culture-derived spores. Restriction frag-
ther evaluated by David et al. (91) and Liguory et al. (222). The ment length polymorphism analysis after digestion of the PCR
primer pair and probe were used for species differentiation of product with PstI was used to differentiate E. bieneusi from
samples found to be PCR positive with a primer pair that Encephalitozoon spp., whereas species differentiation of En-
amplified different microsporidian species in a previous PCR cephalitozoon spp. was done by DNA sequencing (123). The
(see below) (91) and for diagnosis of intestinal infection with same authors used this primer pair for the detection of E.
E. intestinalis by using DNA from stool samples (222). Exam- intestinalis, E. bieneusi, and V. corneae in environmental water
ination of 6 stool specimens from 3 patients with E. intestinalis samples as well (123a).
infection, 31 stool specimens from 26 patients with intestinal E. A similar approach was chosen by Raynaud et al. (286), who
bieneusi infection, and 61 stool specimens from 45 patients used the primer pair C1 (partial sequence of primer V1) and
without intestinal microsporidiosis showed a sensitivity and C2 to amplify a conserved region of the SSU rRNA gene of
specificity of 100%, but the number of stool samples from four microsporidia found in humans (E. bieneusi, E. hellem, E.
patients with E. intestinalis infection was very small in this study cuniculi, and E. intestinalis). To confirm the identity of the
(222). When this primer pair was used with target DNA from amplified 1,200-bp PCR products, restriction enzyme digestion
three gastrointestinal biopsy specimens from patients with con- with HindIII and HinfI was used. The amplified PCR products
firmed E. intestinalis infection, all samples were found to be displays one HindIII restriction site in E. bieneusi and none in
PCR-positive and no amplification was seen with DNA from 25 any Encephalitozoon spp. On the other hand, Encephalitozoon
biopsy specimens from patients with confirmed E. bieneusi spp. could be differentiated on the basis of the number of HinfI
infection. Again, the number of biopsy samples from patients restriction sites: one for E. cuniculi, two for E. hellem, and
with E. intestinalis infection was very small in this study (91). three for E. intestinalis. The technique was used to confirm E.
Another E. intestinalis-specific primer pair, V1 and Sep1, has intestinalis infection diagnosed by light microscopy in two non-
been used to exclude E. intestinalis infection in patients with E. HIV-infected travelers (286).
bieneusi-infected stool samples (63) and in simian immunode- Another primer pair that amplified DNA from four known
ficiency virus-infected rhesus monkeys with experimental E. human microsporidian species was designed by Schuitema et
bieneusi infection (343). al. (303) and further evaluated by David et al. (91). For species
270 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

differentiation, digoxigenin-labeled E. bieneusi- and E. intesti- lished sequences of E. bieneusi, and the DNA sequence of the
nalis-specific probes were used in a nonisotopic hybridization amplified intergenic spacer region (accession no. U61180)
assay. Two species-specific primer sets for E. bieneusi and E. showed 97% identity to the corresponding sequence of E.
intestinalis were used in a second PCR (91, 222, 303). About 10 bieneusi (191).
copies of recombinant plasmids carrying amplified SSU rRNA Another nested PCR assay which detects four microsporid-
genes from E. bieneusi and E. intestinalis were detected by ian species that infect humans (E. bieneusi, E. hellem, E. cu-
using the primer pairs and the hybridization assay. Of 28 in- niculi, and E. intestinalis) was developed by Kock et al. (202). A
testinal biopsy specimens from patients with electron micros- PCR with upstream primer Mic3U and downstream primer
copy-confirmed infection due to E. bieneusi or E. intestinalis, 26 Mic421U was used to amplify 410- to 433-bp DNA fragments
were considered PCR positive, and species differentiation was of the SSU rRNA gene of all the above mentioned microspo-
correctly done in all cases. No false-positive results were seen ridia. A nested PCR with upstream primer Mic266 and down-
in 23 intestinal biopsy specimens from patients without intes- stream primers Eb379, Ec378, Eh410, and Ei395 was used to
tinal microsporidiosis (91). amplify species-specific, 113- to 134-bp DNA fragments. The
A set of pan-Encephalitozoon primers, int530f and int580r, assay was tested with culture-derived spores of E. cuniculi and
selected from sequences conserved between E. cuniculi and E. E. hellem and with stool samples spiked with culture-derived
hellem, was first described by Schuitema et al. (303) and was spores. Stool samples from HIV-infected patients with elec-

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


further used by Didier et al. (108–111). This primer pair am- tron microscopy-confirmed E. bieneusi (n 5 8), Encephalito-
plified a product of approximately 1,000 bp that includes a zoon spp. (n 5 2), and E. intestinalis (n 5 1) infection and
large portion of the SSU rRNA gene, the intergenic spacer, intestinal biopsy specimens from three patients with electron
and a small portion of the LSU rRNA gene of E. hellem, E. microscopy-confirmed E. bieneusi (n 5 2) and E. intestinalis
cuniculi, and E. intestinalis, but did not amplify DNA from (n 5 1) infection were also examined (202). Without exception,
culture-derived V. corneae or intestinal biopsy-derived E. bie- the PCR assay verified electron microscopy-confirmed infec-
neusi as the target (108). Species differentiation was done by tions in stool samples and biopsy specimens of all patients as
Southern blot hybridization with a nonradioactive, chemilumi- well as in spiked stool samples and tissue cultures (202).
nescent 39-oligolabeling and detection system involving spe- Strain differentiation of Encephalitozoon spp. and E. bie-
cies-specific hybridization probes specific for E. cuniculi, E. neusi. Molecular techniques have confirmed the existence of
hellem, and E. intestinalis (109). The specificity of the three different strains of E. cuniculi. Didier et al. (108) first demon-
hybridization probes was determined by using culture-derived strated differences in the intergenic spacer region of eight E.
E. cuniculi, E. hellem, E. intestinalis, and V. corneae isolates cuniculi isolates from mice, rabbits, and dogs. Two primer
(109), but the sensitivity was not determined, and the clinical pairs, int530f-int580r and 530f-580r, were used to amplify the
utility is limited because the primer pair did not amplify E. 39 end of the SSU rRNA gene, the intergenic spacer, and the
bieneusi DNA. 59 end of the LSU rRNA gene of E. cuniculi (108). Using a
A nested PCR assay which detects four microsporidian spe- double-stranded DNA heteroduplex mobility assay and restric-
cies that infect humans (E. bieneusi, E. hellem, E. cuniculi, and tion fragment length polymorphism after digestion with the
E. intestinalis) and an additional four species not known to be restriction endonuclease FokI, they found differences in the
pathogenic to humans (V. necatrix, V. lymantriae, Ameson isolates from mice, rabbits, and dogs. After DNA sequencing
[Nosema] michaelis, and Ichthyosporidium giganteum) was de- of the intergenic spacer region, the isolates were shown to
scribed by Katzwinkel-Wladarsch et al. (190, 191). Stool sam- differ by a small repetitive sequence of 59-GTTT-39. This se-
ples spiked with E. cuniculi spores and stool samples, nasal quence was repeated twice in two isolates from mice, three
discharge, urine, sputum, and cerebrospinal fluid from patients times in three isolates from rabbits and in one isolate from a
with light microscopy-confirmed E. bieneusi, E. hellem, E. cu- mouse, and four times in the two isolates from domestic dogs.
niculi, or E. intestinalis infection were assayed. Two upstream These differences were used to characterize a rabbit strain
primers, MSP-1 and MSP-3, complementary to the 39 region of (strain I), a mouse strain (strain II), and a dog strain (strain
the SSU rRNA gene of the microsporidian species mentioned III). The three strains also differed antigenically by sodium
above and four downstream primers, MSP-2B and MSP-4B for dodecyl sulfate-polyacrylamide gel electrophoresis and West-
E. bieneusi and MSP-2A and MSP-4A for the other mentioned ern blotting.
microsporidian species, complementary to the 59 region of the Hollister et al. (181) also studied the intergenic spacer re-
LSU rRNA gene were used (190, 191). E. bieneusi was distin- gion of three isolates of E. cuniculi: a putative mouse isolate
guished from Encephalitozoon spp. because the PCR products (strain D. Owen [accession no. X98467 and X98468]), a dog
differed in size, and E. cuniculi could be distinguished from E. isolate (strain Stewart [accession no. X98469]), and a human
intestinalis by restriction endonuclease digestion with MnlI, isolate (strain Donovan [accession no. X98466 and X98470])
which resulted in distinct restriction fragment patterns. The established in vitro from the urine of a patient with AIDS (1,
results were confirmed by DNA sequence analysis, and the 177, 179). In the intergenic spacer region, four tetranucleotide
generated sequences were homologous to previously published repeats 59-GTTT-39 were found for the dog and human iso-
DNA sequences. The limit of detection varied between 3 and lates and three were found for the mouse isolate (181). An-
100 spores per 0.1 g of stool (190). Comparison of this PCR other human E. cuniculi isolate from a patient with AIDS (95)
assay with light microscopy on 34 specimens from 31 HIV- also corresponded to dog strain III, with four tetranucleotide
infected patients produced identical results in 82% of speci- repeats (111, 189).
mens (28 of 34) (191). Of the discrepant results, four samples Deplazes et al. (97) used primers that corresponded to po-
were microscopy negative and PCR positive and two were sitions 1 to 19 (which are the first 19 bases of primer V1) and
microscopy positive and PCR negative. Species differentiation 1277 to 1299 of the SSU rRNA gene sequence of E. cuniculi to
was 100% accurate. These primer pairs were also used for amplify SSU rRNA gene fragments from organisms cultured
amplification of E. bieneusi DNA sequences from fecal samples from six HIV-infected patients infected with E. cuniculi, E.
of pigs with suspected E. bieneusi infection confirmed by light hellem, and E. intestinalis and from nine rabbits infected with
microscopy (191). The intergenic spacer region of the ampli- E. cuniculi. PCR products were cleaved with restriction en-
fied PCR products was sequenced and compared with pub- zymes (MboI and HpaII) and analyzed on ethidium bromide-
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 271

TABLE 9. Number of 59-GTTT-39 repeats in the intergenic spacer polymorphisms but also by several sequencing errors. Liguory
region of different E. cuniculi isolates from different hosts and et al. (222a) identified four genetically unrelated lineages
geographical areas among 78 different isolates of E. bieneusi from 65 patients with
No. of No. of Geographic intestinal microsporidiosis. Strain differentiation was done by
Source Reference(s) restriction fragment length polymorphism with NlaIII and
repeats isolates area
Fnu4HI after amplification of the intergenic spacer region with
2 Blue fox 4 Norway 236 the primer pair Eb.gc and Eb.gt. Type I strains were found in
Mouse 2 Czech Republic, UK 108
the majority of samples (78%), whereas type II, III, and IV
3 Rabbit 11 Swizerland 236 strains were found only in 12, 5, and 5% of the samples,
Rabbit 3 USA 108 respectively. The same strains were found in sequential stool
Human 6 Swizerland 97, 98 specimens from the same patients. DNA sequences were de-
termined only for strain I, which showed 100% homology to
4 Human 3 Mexico, USA 97, 98, 111, the E. bieneusi sequence of Zhu et al. (accession no. L20290)
189 (396), and for strain II, which showed seven mismatches with
Dog 2 USA 108 this sequence (97% identity) (222a). rRNA sequences of E.
bieneusi isolates from humans, a pig (accession no. U61180),

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


and rhesus macaques (accession no. AF023245) exhibit addi-
tional variability over the intergenic spacer region and the 59
stained gels. Restriction fragment length polymorphisms of all end of the LSU rRNA gene (62a). The rRNA sequence of the
E. cuniculi isolates from humans did not differ from the rabbit pig belongs to the type IV lineage, which has been found in
isolates (97). Further analysis of the intergenic spacer region of only 5% of humans (222a).
these six E. cuniculi isolates from humans and of different E. When rRNA genes are used to discriminate among strains,
cuniculi isolates of animal origin was done by Mathis et al. several problems must be considered. The presence of differ-
(236). Five E. cuniculi isolates from Swiss patients were found ent genotypes for individual isolates does not automatically
to be homologous to isolates from rabbits, whereas the sixth warrant their consideration as different strains, and different
isolate, from a patient from Mexico, corresponded to dog copies of a gene within one genome may not be identical (289).
strain III (236). It is not clear whether different prevalences of strains in hu-
In the light of these data (summarized in Table 9), dogs or mans reflect different sources of strains that lead to human
rabbits might be the source of human infection. However, it is infection or are related to the pathogenicity of strains in par-
difficult to understand how host-related differences can be ticular types of hosts. However, comparisons of different
maintained in microsporidial isolates when strains can be strains from humans, animals, and environmental sources will
transmitted among different types of hosts, at least experimen- provide a better understanding of the epidemiology of micro-
tally. Too few isolates of E. cuniculi have been obtained and sporidial infection.
analyzed so far to assess host specificity or to determine if Comparison of molecular techniques with light microscopy.
humans are at risk of infection by exposure to dogs or rabbits. In-house validations of PCR protocols for the detection of
More data on strains from many different hosts including hu- microsporidia, especially by those who developed them, are
mans are needed, before the epidemiology of E. cuniculi is generally satisfactory to excellent. However, until recently
clarified. none of these techniques had been validated in a blinded,
E. hellem has been found once in a bird, and E. intestinalis externally controlled fashion. In a multicenter evaluation of
has recently been detected in several mammals. It remains the detection of microsporidia in stool specimens by light mi-
unclear if these animals are relevant reservoirs of the parasites. croscopy and PCR, quality parameters were determined with
Double-stranded DNA heteroduplex mobility assays, restric- identical sets of 50 stool samples for six laboratories using light
tion fragment length polymorphisms after digestion with the microscopy and six laboratories using their own PCR protocol
restriction endonucleases, and DNA sequencing of the inter- (290). A total of 14 clinical samples contained E. bieneusi
genic spacer between the SSU rRNA and the LSU rRNA of spores, and 14 samples were spiked with spores of E. hellem, E.
different E. hellem and E. intestinalis isolates from different cuniculi, and E. intestinalis at different concentrations ranging
geographical regions (Europe and America) showed no differ- from 102 to 106 spores/g; 22 samples were negative. The aver-
ences among these isolates (109, 110, 364). However, Schnitt- age overall sensitivity was 67% (36 to 96%) for laboratories
ger et al. recently reported genotypic differences of the SSU using PCR and 54% (25 to 71%) for laboratories using light
and LSU rRNA coding regions in E. intestinalis (GenBank microscopy, and the specifities were 98 and 95%, respectively.
accession no. Y11611). Again, too few isolates of E. hellem and A uniform detection limit of between 104 and 106 spores/g of
E. intestinalis have been analyzed so far to determine if there stool was apparent for light microscopy, while PCR detected
are different strains of these species. Additional isolates must concentrations as low as 102 spores/g. However, the greatest
be analyzed to help define the epidemiology of these micro- differences were found between individual laboratories rather
sporidian species. than between the two techniques used (290). These apparent
Three distinct genotypes of E. bieneusi have been identified differences in quality between laboratories suggest that indi-
by Rinder et al. (289) by amplification of a 484-bp DNA frag- vidual laboratories first should improve their established tech-
ment between primers MSP-3 and MSP-4B from 12 stool sam- nique as much as possible and not give up one method in favor
ples of eight HIV-infected patients (289). Nine polymorphic of the other. Nevertheless, it will certainly be helpful to have a
sites were found in the 243-bp intergenic spacer region of the second technique available when confirmation of the first is
rRNA gene. The genotypes were stable in samples taken dur- desired. Since the threshold of detection for light microscopy
ing 11 weeks of infection. The intergenic spacer rRNA E. seems to be between 104 and 106 spores/g of stool, epidemio-
bieneusi sequence reported by Zhu et al. (accession no. logical studies involving only light microscopy may give prev-
L20290) (396) was not considered to be a fourth genotype by alence data which did not reflect the true prevalence of mi-
the authors. This sequence is only 86.8% identical to those of crosporidia. Application of PCR to stool samples offers an
the other three genotypes; this may be caused not only by approach that can be adapted for processing large numbers of
272 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

specimens to define the true prevalence of intestinal micros- rRNA sequences for phylogenetic construction of microspo-
poridiosis in human populations and to determine whether ridia pathogenic to humans were also first used by Vossbrinck
microsporidia are present in the general population (272). et al. (364). They used a highly conserved primer pair, 530f in
Because all the participating laboratories in this trial re- the SSU rRNA and 580r in the LSU rRNA, to amplify a DNA
mained anonymous, no recommendations about DNA isola- fragment of about 1,350 bp that contains the 39 end of the SSU
tion techniques, primer pairs, or PCR methods can be given. rRNA, the intergenic spacer, and the 59 end of the LSU rRNA
Nevertheless, testing all clinical samples with separate species- of E. hellem, E. cuniculi, and V. corneae obtained from tissue
specific primer pairs is a time-consuming task. An efficient culture (364). Restriction digests of the amplified DNA frag-
approach for PCR detection of microsporidia in clinical sam- ments with SauIIIA, EcoRI, DraI, and HinfI showed clear
ples would involve using general microsporidian primer pairs differences among the three species. Three regions of the am-
that amplify several species. The species of microsporidia de- plified DNA fragment were sequenced with primer 228r (in the
tected could then be determined by PCR with species-specific LSU rRNA) in addition to primers 530f and 580r. The se-
primer pairs. Primers V1 and PMP2 amplify microsporidian quence difference between E. hellem and E. cuniculi was
DNA of different microsporidian species. Primer V1 is used by 23.1%, while the sequence difference between the two Enceph-
several researchers, and primer PMP2 has also been evaluated alitozoon spp. and other studied families (Vairimorpha and
by different groups (123, 134, 149). Other published general Ichtyosporidium) ranged between 54.2 and 56.2% (364). These

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


microsporidian primer pairs (190, 202, 286, 303) have not been data indicate that E. hellem and E. cuniculi are different or-
evaluated by others so far. The primer pairs V1-EB450 and ganisms which are closely related.
V1-SI500, although constructed by using SSU rRNA se- The same primers (530f, 580r, and 228r) were used by Zhu
quences which appear to contain several sequencing errors, are et al. (396) for phylogenetic analysis of E. bieneusi, E. intesti-
best evaluated for diagnosis of microsporidiosis due to E. bie- nalis, and A. michaelis. E. bieneusi and E. intestinalis DNA was
neusi and E. intestinalis. Several researchers have published extracted from intestinal biopsy specimens from patients with
studies with these primer pairs, usually with excellent results electron microscopy-confirmed microsporidial infection;
(81, 145, 148–150, 260, 262, 339a, 381, 395). spores of A. michaelis were obtained from the muscle of blue
crabs (Callinectus sapidus). Following amplification and se-
Molecular Techniques for Phylogenetic Analysis quencing of PCR product, sequences were aligned with those
of several other microsporidian species including E. hellem and
Molecular techniques are rapidly becoming an important E. cuniculi. By using three sequenced regions, Zhu et al. cal-
and integral part of biosystematic studies. Traditional classifi- culated that there was a 16.7% sequence difference between E.
cations of microsporidia have been done on the basis of mor- hellem and E. cuniculi and that there were 21.9 and 26.4%
phological characteristics, but the four major published classi- sequence differences between E. intestinalis and E. cuniculi and
fications differ significantly, and the taxonomy of microsporidia between E. intestinalis and E. hellem, respectively (396). These
is still controversial (183, 214, 335–337, 376). Microsporidia are differences calculated by Zhu et al. (396) differed from those
prime candidates for phylogenetic analysis based on DNA se- reported by Vossbrinck et al. (364), perhaps due to differences
quence data because of the relatively small number of useful in alignment or to the version of PAUP (phylogenetic analysis
morphological characters. Advances in DNA sequencing tech- using parsimony) used. Based on their data, Zhu et al. (396)
nology have resulted in a great increase in the number of useful stated that E. cuniculi, E. hellem, and E. intestinalis are closely
characters available for phylogenetic analysis. Moreover, DNA related, most likely at the family level, with E. cuniculi and E.
amplification by PCR has made it possible to obtain sequence hellem belonging to the same genus and E. intestinalis belong-
data from organisms which cannot be cultured and for which ing to the Encephalitozoonidae family but as a separate genus.
material cannot be obtained in sufficient quantities for cloning Much larger differences were found between the other micro-
and sequencing. sporidia. Since E. bieneusi differed from E. intestinalis by
16S rRNA genes are found in all prokaryotic organisms and 45.7%, from E. cuniculi by 43.2%, and from E. hellem by
accumulate mutations at a low, constant rate over time. Hence, 42.2%, it is clear that E. bieneusi is placed in a separate family
they may be used as “molecular clocks” (258, 387). Highly (396).
variable portions of the SSU-rRNA genes provide unique Baker et al. presented a phylogeny of various species of
signatures for any organism and useful information about microsporidia based on sequence data of the whole SSU rRNA
possible relationships (387). Several groups have been using gene (10). DNA sequences either were obtained by PCR with
molecular approaches to develop taxonomic systems for mi- primer pair 18f (partial sequence of primer V1) and 1537r,
crosporidia based on rRNA genes and also on protein-coding complementary to conserved regions of the SSU rRNA gene at
DNA sequences. This analysis has yielded insights into new the 39 and 59 ends, followed by sequencing with several internal
relationships among microsporidia, which are being continu- primers or were obtained from the GenBank database. Results
ously integrated into a revised taxonomy. For example, it has of parsimony analysis with PAUP suggested that the 16 studied
been shown that Nosema trichoplusiae is a synonym for Nosema microsporidian species could be divided into four distinct
bombycis based on the sequence of the SSU rRNA coding groups referred to as the Ichtyosporidium group, the Enceph-
region (276). Therefore, new molecular techniques have led alitozoon group, the Endoreticulatus group, and the Vairimor-
not only to the discovery of new organisms but also to the pha/Nosema group (Fig. 13). Delineation of Vairimorpha/
elimination of an old organism. Nosema as a group was additionally supported by LSU rRNA
The first microsporidian DNA sequence data were reported data obtained from several other species of Nosema and Vairi-
by Vossbrinck et al. (363). The SSU rRNA gene of the insect morpha (9). From the branching pattern in Fig. 13 and the
microsporidian V. necatrix was sequenced, and a phylogenetic sequence difference between V. corneae (formerly N. corneum)
tree was then constructed. The root of the eukaryotic tree lay and N. bombycis (27.9%) it seems clear, that V. corneae is
between the V. necatrix rRNA sequence and those of all other unrelated to the other Nosema spp. Baker et al. (9, 10) placed
eukaryotic lineages for which sequences were available, show- V. corneae in the Endoreticularis group because the parasite is
ing that the V. necatrix sequence is consistent with a very early most closely related to Endoreticulatus schubergi (7.3% dis-
branch in the eukaryotic line of descent (363). tance). This supports the reclassification of N. corneum to a
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 273

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


FIG. 13. Tree representing the phylogenetic relationship of several microsporidian species as determined by SSU rRNA sequence analysis. This was the most
pasimonious tree found by using the branch-and-bound option of PAUP. Reprinted from reference 101 with permission of the publisher.

new genus as V. corneae which was done based on the ultra- amplified with primers complementary to conserved regions at
structure of developmental stages in the livers of experimen- the 39 and 59 end of the SSU rRNA gene (164, 166). Restric-
tally infected atymic mice (the diplokaryotic arrangement of tion enzyme digestion of the amplified DNA with HphI and
the nuclei was the only character that conformed to the de- BsiHKAI produced different restriction fragment length poly-
scription of the genus Nosema) (324). Baker et al. (10) placed morphism patterns for all the species examined. The degree of
in the Encephalitozoon group the genera which Cali et al. sequence homology was established by a pairwise comparison
placed in the family Encephalitozoonidae (47). By using parsi- with the Bestfit program. SSU rRNA gene sequences of E.
mony (branch-and-bound option of PAUP), E. intestinalis was cuniculi and E. hellem showed a lower degree of homology
most closely related to E. cuniculi (Fig. 13), and by using (89.7%) than did those of E. cuniculi and E. intestinalis (90.7%)
distance methods (unweighted pair group method with arith- or E. hellem and E. intestinalis (91.4%). The E. intestinalis
metic mean [UPGMA]), E. hellem was most closely related to sequence was only approximately 70% identical to those of
E. intestinalis (mean distance values, E. intestinalis/E. hellem 5 microsporidia of other genera such as E. bieneusi or V. necatrix.
0.043, E. intestinalis/E. cuniculi 5 0.059, E. hellem/E. cuniculi 5 Although sequence dissimilarity values are not suitable meth-
0.066), but in no case was E. hellem most closely related to E. ods for phylogenetic analysis, the authors concluded that E.
cuniculi (10). Because of the close relationship between the cuniculi, E. hellem, and E. intestinalis belong to the same genus
three Encephalitozoon spp., Baker et al. proposed that E. in- and proposed the reclassification of Septata intestinalis to En-
testinalis, originally described as Septata intestinalis by Cali et cephalitozoon intestinalis (166).
al. (47), be designated Encephalitozoon intestinalis and that the The data reported by Hartskeerl et al. (166) and Baker et al.
morphological definition of the genus be changed to reflect this (10) differed significantly from those of Zhu et al. (396). Harts-
designation (10). keerl et al. and Baker et al. placed E. intestinalis within the
Similar conclusions were drawn by Hartskeerl et al. on the genus Encephalitozoon, while in the phylogeny of Zhu et al., E.
basis of SSU rRNA sequence data (166). The SSU rRNA gene intestinalis was placed in a genus of its own (as originally
of E. intestinalis, E. cuniculi, E. hellem, and E. bieneusi was proposed by Cali et al. [47]). There are several possible expla-
274 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

nations for this. First, Zhu et al. used sequence data of the 39 branching pattern comparable to the phylogeny of Baker et al.
and 59 ends of the SSU and LSU rRNA genes and the inter- (9, 10) for the species used in both analysis.
genic spacer for their phylogenetic analysis, whereas Harts- The phylogenetic relationships of microsporidia were also
keerl et al. and Baker et al. used the sequence of the whole analysed by riboprinting (280). The SSU rRNA and the inter-
SSU rRNA gene. However, Zhu et al. stated that their results genic spacer region of different microsporidia from fish, in-
were unchanged when the whole SSU rRNA gene sequence sects, and shrimp were amplified and digested with restriction
was used. Second, the alignment methods differed. Phyloge- enzymes. The generated riboprints were analyzed, and phylo-
netic analysis depends on comparison of homologous charac- genetic trees were constructed by using the MIX and DOL-
ters, and only regions in which exact alignment is possible LOP programs of the PHYLIP package, in which each frag-
should be used. Third, and this may be the most important ment is considered a different character. While sequencing
point, the E. cuniculi, E. hellem, and E. intestinalis sequences gives more detailed information on the phylogenetic relation-
obtained by Zhu et al. (393–396) differed significantly even in ship of different species and genera, riboprinting is a faster and
conserved regions of the rDNA from those reported by Baker cheaper way of determining the identity and phylogeny of
et al. (10) and by Hartskeerl et al. (164, 166). microsporidia. This approach will work at the genus level, but
Cali et al. (49) did not agree with the reclassification of DNA sequencing should be used to define the relationships of
Septata intestinalis into Encephalitozoon intestinalis. The sporo- different species.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


gony of this species is tetrasporous (47). On the other hand, a- and b-tubulin sequences obtained by amplification with
there are no supporting published data for tetrasporous devel- primer pairs directed against conserved sequences from differ-
opment of E. cuniculi, the type organism from which the genus ent microsporidian species (E. hellem, E. intestinalis, E. cunic-
was established. Until E. cuniculi is demonstrated to be tetra- uli, N. locustae, and Spraguea lophii) have been used by several
sporous, the genus Encephalitozoon cannot be modified to in- authors to construct phylogenetic trees (125, 126, 188, 221).
clude this feature, thus excluding Encephalitozoon (Septata) The b-tubulin sequences from E. hellem, E. intestinalis, and E.
intestinalis. They also noted that the morphological presence of cuniculi were closely related, but that of E. intestinalis was
secretions from E. intestinalis and the lack of such secretions in more closely related to that of E. cuniculi than was that of E.
E. cuniculi was not addressed in the reclassification of E. in- hellem (221), reinforcing the reclassification of E. intestinalis.
testinalis by Hartskeerl et al. (166) and Baker et al. (10). In The generated a- and b-tubulin trees were nearly identical in
other microsporidia, this feature has been given taxonomic topology, but in several important aspects these trees are in-
value. For example, some genera in the family Pleistophoridae consistent with phylogenetic trees constructed on the basis of
were created on the basis of variations in secretion (49). Based SSU rRNA sequence data. The few SSU rRNA-based molec-
ular trees for which microsporidian data are available suggest
on these data, Cali et al. excluded E. intestinalis from the genus
that microsporidia are one of the most ancient eucaryotic lin-
Encephalitozoon until E. cuniculi is shown to be tetrasporous
eages (10, 11, 155, 217, 363), but the a- and b-tubulin trees
(49).
suggest that the microsporidia are close relatives of fungi,
Whereas molecular phylogenies suggest that the family En-
which invites speculation that microsporidia evolved degenera-
cephalitozoonidae should contain one genus, morphologic dif-
tively from higher forms (126, 192, 221).
ferences between E. hellem, E. cuniculi, and E. intestinalis sug-
Phylogenetic analysis of the complete nucleotide sequence
gest that three genera should be used. In cell culture, E. hellem of the genes putatively encoding translation elongation factors
displays variable development (49, 102); some parasites occur 1a and 2 from the fish microsporidium Glugea plecoglossi sug-
within a parasitophorous vacuole, while others develop in di- gests that G. plecoglossi represents the earliest branch of the
rect contact with the host cell cytoplasm. This behavior has eukaryotes among the analyzed eukaryotic species (186, 187).
never been observed for E. cuniculi. Such differences have This reinforces the hypothesis that microsporidia are extremely
been used historically for genus-level classification in micro- ancient eukaryotes that diverged before the occurrence of mi-
sporidia. Consequently, Cali et al. (49) proposed the removal tochondrial symbiosis rather than evolving by degeneration
of E. hellem from the genus Encephalitozoon and its reclassi- from higher forms.
fication in a genus of its own, creating three genera in the One of the original reasons behind the proposal that micro-
family Encephalitozoonidae. This would satisfy both morpho- sporidia are ancient eukaryotes is their lack of mitochondria
logical and molecular criteria (49). (64). The presence of mitochondrion-derived genes reveals
Docker et al. (120) compared sequences of the SSU rRNA that the ancestor of a lineage contained a mitochondrion, even
gene, the intergenic spacer region, and the LSU rRNA gene of though an organelle has not been identified in the lineage
N. salmonis with that of E. bieneusi amplified with primer pair (193). Recently, genes encoding a chaperone protein, Hsp70
530f and 580r; the two sequences differed in composition by (70-kDa heat shock protein), have been identified in the mi-
19.8%. This genetic divergence between the two species was crosporidia N. locustae, V. necatrix, E. hellem, and E. cuniculi
sufficiently large to place the two species into two different (159, 173, 275a). Phylogenetic analyses have shown close rela-
genera in the family Enterocytozoonidae (120, 197), as origi- tionships to Hsp70 proteins from mitochondria of other eu-
nally proposed by Hedrick et al. (169). This placement is sup- karyotes. In addition to the detection of genes encoding Hsp70
ported by morphological data. Although N. salmonis exhibits proteins in microsporidia, tRNA synthetase genes consistent
most of the distinguishing morphological characteristics of the with the presence of mitochondria have been found in micro-
family Enterocytozoonidae, the distinctively different hosts and sporidia (381a). The simplest interpretation of these data is
sites of development support the placement of Nucleospora that microsporidia have lost mitochondria while retaining ge-
and Enterocytozoon into separate genera within the family En- netic evidence of their past presence. Interestingly, these mi-
terocytozoonidae (101, 120, 197). crosporidian Hsp70 sequences appear as a sister group of fungi
Another SSU rRNA-based phylogeny of microsporidia has in Hsp70 phylogenies, supporting the list of reasons to believe
been published by Malone and McIvor (233). Although they that microsporidia are closely related to fungi, as a highly
included several other microsporidian species in their analysis specialized group that adapted extensively to an exclusively
than were analyzed by Baker et al. (9, 10) the calculated phy- intracellular parasitic mode of life by degenerative evolution
logenetic tree, using the neighbor-joining method, showed a (193, 251a).
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 275

The concept that microsporidia are ancient eukaryotes was (chromosome I) of E. cuniculi is already available, and system-
also supported by their apparent lack of spliceosomal introns. atic sequencing of this chromosome will be completed in the
The fact that the microsporidia N. locustae and V. necatrix near future (25, 124a). Complete mapping and gene sequenc-
possess core spliceosomal components (U2 and U6 small nu- ing of the whole genome of E. cuniculi are in progress (124a),
clear RNAs) (115, 133a) is consistent with the recent diver- and this will be useful in advancing our knowledge of physio-
gence of the microsporidia. It has been suggested that micro- logically important genes and control regions of coding and
sporidia lost most of their spliceosomal introns during the noncoding sequences in microsporidia.
radical genome size reduction accompanying the adaptation to rRNA data have suggested that microsporidia are one of the
an intracellular parasitic lifestyle (133a). most ancient eukaryotic lineages. On the other hand, the re-
Phylogenetic analysis of 19 eukaryotic and archeal LSU latedness of microsporidia to fungi and animals, as suggested
rRNA sequences, including the LSU rRNA sequence of E. by the a- and b-tubulin and mitochondrion-type Hsp70 se-
cuniculi, produced different trees depending on the maximum- quences, invites speculation that microsporidia evolved degen-
likelihood method used. FastDNAml produced a tree in which eratively from higher forms. Examination of protein-coding
three fast-evolving lineages including microsporidia are pre- sequences from microsporidia will be of interest to answer this
dicted to have diverged early from other eukaryotic lineages, question and is under way.
whereas the BASEML method placed the Encephalitozoon The taxonomy of microsporidia has undergone several

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


sequence in a relatively late-emerging position with a very high changes during the last few years and will continue to change
long terminal branch, indicating a high rate of evolution significantly in the near future when new DNA-based data are
(275b). incorporated into new classification systems. As a first step,
Why do different microsporidian genes suggest contradic- molecular data suggest that the family Encephalitozoonidae
tory phylogenies? The fact that a phylogeny can be obtained should contain only one instead of two genera, but morpho-
for a group of organisms does not guarantee that it really logical data and rules of taxonomy demand three genera. How-
reflects the evolutionary history (381a). Much of our evolu- ever, there is no longer justification for two genera within the
tionary thinking is currently based on the phylogeny of SSU- family Encephalitozoonidae.
rRNA, which may be biased by the long-branch attraction Different strains of E. cuniculi and E. bieneusi have already
artefact. Like any other single-gene phylogeny, it may be mis- been identified, and more isolates from different hosts and
leading in some aspects but correct in others. The phylogenies environmental samples are currently being examined to sub-
presented above clearly point to the dangers of placing too stantiate the belief that human microsporidiosis is a zoonotic
much confidence in the phylogny of a single gene, and more or waterborne disease. With respect to other species, no dif-
DNA sequence data of protein-encoding sequences from mi- ferent strains have been identified so far, but examination of
crosporidia will be needed to answer this question. different isolates from different geographic areas and hosts by
molecular techniques is under way.
Molecular Techniques for Susceptibility Testing Animal hosts have been identified for E. cuniculi and, most
recently, for E. hellem, E. intestinalis, and E. bieneusi. Further
Several antibacterial protein synthesis inhibitors have anti- examination of different hosts and environmental samples by
parasitic activity, in particular the aminoglycoside paromomy- the highly sensitive molecular techniques is under way. These
cin. The paromomycin binding site includes several nucleotides studies may lead to the identification of further animal reser-
at the 39 end of the SSU rRNA, and mutational analysis has voirs for microsporidia which infect humans. The highly sen-
specifically implicated the C1409 z G1491 base pair (E. coli num- sitive molecular techniques may be also helpful for identifying
bering) at the base of the 47-479 hairpin (189). SSU rRNA asymptomatic carriers of microsporidia.
from all analyzed microsporidia lack this base pair, predicting At present only limited data have been published comparing
that they would be paromomycin resistant (189). molecular techniques for diagnosis of microsporidia with tra-
Several drugs, notably colchicine, vinca alkaloides, and ben- ditional methods of determining the sensitivity and specificity
zimidazoles, are effectors of microtubule polymerization. of these new techniques. Nevertheless, these studies indicate
Among these agents, the benzimidazoles are unique in being that the sensitivity and specificity of molecular techniques can
selectively toxic to certain lower eukaryotes. The b-tubulin be very high. One international, blinded multicenter study
subunit has been identified as the primary target of benzimid- comparing traditional light-microscopic techniques with PCR
azole. Benzimidazoles such as albendazole act by disrupting has been published recently (290), but further studies are
microtubules through b-tubulin binding, and six different needed.
amino acid residues have been implicated in benzimidazole
susceptibility by mutational analysis of fungi (125, 188). Muta-
CONCLUDING REMARKS
tions in His-6, Phe-167, Glu-198, Phe-200, and Arg-241 confer
resistance to the derivate benomyl, while a mutation in Ala-165 Microsporidia are an important cause of disease in HIV-
confers thiabendazole resistance (221). Both E. hellem and E. infected patients and are now increasingly also recognized as
cuniculi b-tubulins include His-6, Phe-167, Glu-198, Phe-200, pathogens in non-HIV-infected patients with or without im-
and Arg-241, predicting that these microsporidia would be munosuppression. Therefore, methods for the diagnosis and
susceptible to benzimidazoles (125, 188). In addition, E. hellem species differentiation of microsporidia should be available in
and E. intestinalis b-tubulins include Val-268, and mutational every laboratory performing parasitological examination of
and comparative sequence analyses with the yeast S. cervisiae clinical specimens.
suggest that Val-268 also plays a role in benzimidazole suscep- Although molecular techniques, especially PCR, seem to be
tibility (126). very sensitive and specific and are useful for the diagnosis and
species differentiation of microsporidia, large comparative
FUTURE TRENDS blinded studies determining sensitivity and specificity are lack-
ing. Therefore, screening of clinical samples for microsporidia
The genomes of microsporidia, especially E. cuniculi, are by these techniques is not recommended at the moment. For
very small. A genomic library of the smallest chromosome the detection of microsporidia in clinical samples, chemofluo-
276 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

rescent stains or chromotrope-based stains should be used as associated with microsporidia infection of the common bile duct mucosa in
the methods of first choice. Nevertheless, accurate taxonomic a patient with HIV infection. Ann. Intern. Med. 117:401–402.
15. Beauvais, B., C. Sarfati, J. M. Molina, A. Lesourd, M. Lariviere, and F.
classification of microsporidia is essential because the different Derouin. 1993. Comparative evaluation of five diagnostic methods for dem-
genera and species exhibit different biological and epidemio- onstrating microsporidia in stool and intestinal biopsy specimens. Ann.
logical characteristics, which influence approaches to preven- Trop. Med. Parasitol. 87:99–102.
tion and treatment. Application of PCR methods offers an 16. Beauvais, B., C. Sarfati, S. Challier, and F. Derouin. 1994. In vitro model
to assess effect of antimicrobial agents on Encephalitozoon cuniculi. Anti-
approach that can be adapted for processing large numbers of microb. Agents Chemother. 38:2440–2448.
specimens to define the epidemiological extent of microspo- 17. Beckers, P. J. A., G. J. M. M. Derks, T. van Gool, F. R. J. Rietveld, and
ridian infection in human populations (272). Therefore, iden- R. W. Sauerwein. 1996. Encepahlitozoon intestinalis-specific monoclonal
tification of microsporidia by light microscopy should be fol- antibodies for laboratory diagnosis of microsporidiosis. J. Clin. Microbiol.
34:282–285.
lowed by confirmation and specifies differentiation with 18. Beckwith, C., N. Peterson, J. J. Liu, and J. A. Shadduck. 1988. Dot enzyme-
molecular techniques. linked immunosorbent assay (dot ELISA) for antibodies to Encephalito-
As is true for many other new and emerging pathogens, we zoon cuniculi. Lab. Anim. Sci. 38:573–576.
have just scratched the surface of a complex and evolving 19. Belcher, J. W., S. A. Guttenberg, and B. M. Schmookler. 1997. Microspo-
ridiosis of the mandible in a patient with acquired immunodeficiency syn-
relation between the phylum Microspora and humans (382). drome. J. Oral Maxillofac. Surg. 55:424–426.
Application of molecular techniques to diagnosis, species dif- 20. Berg, J., L. E. Diaz, and B. S. Bender. 1996. Microsporidia in humans. Ann.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


ferentiation, and phylogentic analysis of microsporidia will Intern. Med. 125:522–523.
lead to an enormously increased knowledge of these organisms 21. Bergquist, N. R., G. Stintzing, L. Smedman, T. Waller, and T. Andersson.
1984. Diagnosis of encephalitozoonosis in man by serological tests. Br.
in the near future. Med. J. 288:902.
22. Bergquist, R., L. Morfeldt Mansson, P. O. Pehrson, B. Petrini, and J.
Wasserman. 1984. Antibody against Encephalitozoon cuniculi in Swedish
ACKNOWLEDGMENTS homosexual men. Scand. J. Infect. Dis. 16:389–391.
23. Biderre, C., M. Pagès, G. Méténier, E. U. Canning, and C. P. Vivarès. 1995.
Evidence for the smallest nuclear genome (2.9 Mb) in the microsporidium
We acknowledge and appreciate the assistance of Britta Franzen
Encephalitozoon cuniculi. Mol. Biochem. Parasitol. 74:229–231.
during preparation of the manuscript. 24. Biderre, C., M. Pagès, G. Méténier, D. David, J. Bata, G. Prensier, and
Portions of this work were supported by the Köln Fortune program/ C. P. Vivarès. 1994. On small genomes in eukaryotic organisms: molecular
Faculty of Medicine, University of Cologne, Cologne, Germany. karyotypes of two microsporidian species (Protozoa) parasites of verte-
brates. C. R. Acad. Sci. 317:399–404.
25. Biderre, C., F. Duffieux, E. Peyretaillade, P. Glaser, P. Peyret, A. Danchin,
REFERENCES M. Pagès, G. Méténier, and C. P. Vivarès. 1997. Mapping of repetitive and
nonrepetitive DNA probes to chromosomes of the microsporidian Enceph-
1. Aarons, E. J., D. Woodrow, W. S. Hollister, E. U. Canning, N. Francis, and alitozoon cuniculi. Gene 191:39–45.
B. G. Gazzard. 1994. Reversible renal failure caused by a microsporidian 25a.Biderre, C., G. Méténier, and C. P. Vivarès. 1998. A small spliceosomal-
infection. AIDS 8:1119–1121. type intron occurs in a ribosomal protein gene of the microsporidian En-
2. Albrecht, H., H. J. Stellbrinck, and I. Sobottka. 1995. Failure of itracon- cephalitozoon cuniculi. Mol. Biochem. Parasitol. 94:283–286.
azole to prevent Enterocytozoon bieneusi infection. Genitourin. Med. 71: 26. Bigliardi, E., M. G. Selmi, P. Lupetti, S. Corona, S. Gatti, M. Scaglia, and
325–326. L. Sacchi. 1996. Microsporidian spore wall: ultrastructural findings on En-
3. Albrecht, H., I. Sobottka, H. J. Stellbrinck, and H. Greten. 1995. Does cephalitozoon hellem exospore. J. Eukaryot. Microbiol. 43:181–186.
choice of Pneumocystis carinii prophylaxis influence the prevalence of En- 27. Birthistle, K., P. Moore, and P. Hay. 1996. Microsporidia: a new sexually
terocytozoon bieneusi microsporidiosis in AIDS patients? AIDS 9:302–304. transmissible cause of urethritis. Genitourin. Med. 72:445.
4. Aldras, A. M., J. M. Orenstein, D. P. Kotler, J. A. Shadduck, and E. S. 28. Black, S. S., L. A. Steinohrt, D. C. Bertucci, L. B. Rogers, and E. S. Didier.
Didier. 1994. Detection of microsporidia by indirect immunofluorescence 1997. Encephalitozoon hellem in budgerigars (Melopsittacus undulatus). Vet.
antibody test using polyclonal and monoclonal antibodies. J. Clin. Micro- Pathol. 34:189–198.
biol. 32:608–612. 29. Blanshard, C., D. S. Ellis, D. G. Tovey, S. Dowell, and B. G. Gazzard. 1992.
5. Anwar-Bruni, D. M., S. E. Hogan, D. A. Schwartz, C. Mel Wilcox, R. T. Treatment of intestinal microsporidiosis with albendazole in patients with
Bryan, and J. L. Lennox. 1996. Atovaquone is effective treatment for AIDS. AIDS 6:311–313.
symptoms of gastrointestinal microsporidiosis in HIV-1-infected patients. 30. Blanshard, C., W. S. Hollister, C. S. Peacock, D. G. Tovey, D. S. Ellis, E. U.
AIDS 10:619–623. Canning, and B. G. Gazzard. 1992. Simultaneous infection with two types
6. Ashton, N., and P. A. Wirasinha. 1973. Encephalitozoonosis (nosematosis) of intestinal microsporidia in a patient with AIDS. Gut 33:418–420.
of the cornea. Br. J. Ophthalmol. 57:669–674. 31. Blanshard, C., D. S. Ellis, S. P. Dowell, G. Tovey, and B. G. Gazzard. 1993.
7. Asmuth, D. M., P. C. DeGirolami, M. Federman, C. R. Ezratty, D. K. Electron microscopic changes in Enterocytozoon bieneusi following treat-
Pleskow, G. Desai, and C. A. Wanke. 1994. Clinical features of microspo- ment with albendazole. J. Clin. Pathol. 46:898–902.
ridiosis in patients with AIDS. Clin. Infect. Dis. 18:819–825. 32. Boot, R., F. van Knapen, B. C. Kruijt, and H. C. Walvoort. 1988. Serological
8. Avery, S. W., and A. H. Undeen. 1987. The isolation of microsporidia and evidence for Encephalitozoon cuniculi infection (nosemiasis) in gnotobiotic
other pathogens from concentrated ditch water. J. Am. Mosq. Control guineapigs. Lab. Anim. 22:337–342.
Assoc. 3:54–58. 32a.Bornay-Llinares, F. J., A. J. da Silva, H. Moura, D. A. Schwartz, G. S.
9. Baker, M. D., C. R. Vossbrinck, J. V. Maddox, and A. H. Undeen. 1994. Visvesvara, N. J. Pieniazek, A. Cruz-López, P. Hernández-Jaúregui, J.
Phylogenetic relationship among Vairimorpha and Nosema species (micros- Guerrero, and F. J. Enriques. 1998. Immunologic, microscopic, and mo-
poridia) based on ribosomal RNA sequence data. J. Invertebr. Pathol. lecular evidence of Encephalitozoon intestinalis (Septata intestinalis) infec-
64:100–106. tion in mammals other than humans. J. Infect. Dis. 178:820–826.
10. Baker, M. D., C. R. Vossbrinck, E. S. Didier, J. V. Maddox, and J. A. 33. Bouche, H., C. Housset, J. L. Dumont, F. Carnot, Y. Menu, B. Aveline, J.
Shadduck. 1995. Small subunit ribosomal DNA phylogeny of various mi- Belghiti, B. Boboc, S. Erlinger, P. Berthelot, and S. Pol. 1993. AIDS-related
crosporidia with emphasis on AIDS related forms. J. Eukaryot. Microbiol. cholangitis: diagnostic features and course in 15 patients. J. Hepatol. 17:
42:564–570. 34–39.
11. Baker, M. D., C. R. Vossbrink, J. J. Becnel, and J. V. Maddox. 1997. 34. Bretagne, S., F. Foulet, W. Alkassoum, J. Fleury Feith, and M. Develoux.
Phylogenetic position of Amblyospora Hazard & Oldacre (Microspora: Am- 1993. Prévalence des spores d’Enterocytozoon bieneusi dans les selles de
blyosporidae) based on small subunit rRNA data and its implication for the patients sidéens et d’enfants Africains non infectés par le VIH. Bull. Soc.
evolution of the microsporidia. J. Eukaryot. Microbiol. 44:220–225. Pathol. Exot. 86:351–357.
12. Balbiani, G. 1882. Sur les microsporidies ou psorospermies des articules. 35. Brown, J. R., and W. F. Doolittle. 1995. Root of the universal tree of life
C. R. Hebd. Seances Acad. Sci. Paris 95:1168–1171. based on ancient aminoacyl-tRNA synthetase gene duplications. Proc. Natl.
13. Barlough, J. E., T. S. McDowell, A. Milani, L. Bigornia, S. B. Slemenda, Acad. Sci. USA 92:2441–2445.
N. J. Pieniazek, and R. P. Hedrick. 1995. Nested polymerase chain reaction 36. Bryan, R. T., A. Cali, R. L. Owen, and H. C. Spencer. 1991. Microsporidia:
for detection of Enterocytozoon salmonis genomic DNA in chinook salmon opportunistic pathogens in patients with AIDS. Prog. Clin. Parasitol. 2:1–
Oncorynchus tshawytscha. Dis. Aquat. Org. 23:17–23. 26.
14. Beaugerie, L., M. F. Teilhac, A. M. Deluol, J. Fritsch, P. M. Girard, W. 37. Bryan, R. T., and R. Weber. 1993. Microsporidia. Emerging pathogens in
Rozenbaum, Y. Le Quintrec, and F. P. Chatelet. 1992. Cholangiopathy immunodeficient persons. Arch. Pathol. Lab. Med. 117:1243–1245.
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 277

38. Bryan, R. T. 1995. Microsporidiosis as an AIDS-related opportunistic in- stools and in intestinal biopsy specimens from infected patients. Clin. Di-
fection. Clin. Infect. Dis. 21(Suppl. 1):S62–S65. agn. Lab. Immunol. 4:405–408.
39. Bryan, R. T. 1995. Microsporidia, p. 2513–2524. In G. L. Mandell, J. E. 64. Cavalier-Smith, T. 1987. Eukaryotes with no mitochondria. Nature 326:
Bennett, and R. Dolin (ed.) Principles and practice of infectious diseases, 332–333.
4th ed. Churchill Livingstone, Inc., New York, N.Y. 65. Cavalier-Smith, T. 1991. Archamoebae: the ancestral eukaryotes? Biosys-
40. Bryan, R. T., R. Weber, and D. A. Schwartz. 1997. Microsporidiosis in tems 25:25–38.
patients who are not infected with human immunodeficiency virus. Clin. 66. Centers for Disease Control. 1990. Microsporidian keratoconjunctivitis in
Infect. Dis. 24:534–535. patients with AIDS. Morbid. Mortal. Weekly Rep. 39:188–189.
41. Cali, A., and R. L. Owen. 1988. Microsporidiosis, p. 928–949. In A. Balows, 66a.Cherey, S. A. Unpublished data.
W. Hausler, Jr., and E. H. Lennette (ed.), The laboratory diagnosis of 67. Chilmonczyk, S., W. T. Cox, and R. P. Hedrick. 1991. Enterocytozoon
infectious diseases: principles and practice, vol. 1. Springer-Verlag, New salmonis n. sp.: an intranuclear microsporidium from salmonid fish. J.
York, N.Y. Protozool. 38:264–269.
42. Cali, A., and R. L. Owen. 1990. Intracellular development of Enterocyto- 68. Chu, P., and A. B. West. 1996. Encephalitozoon (Septata) intestinalis. Cyto-
zoon, a unique microsporidian found in the intestine of AIDS patients. J. logic, histologic, and electron microscopic features of a systemic intestinal
Protozool. 37:145–155. pathogen. Am. J. Clin. Pathol. 106:606–614.
43. Cali, A. 1991. General microsporidian features and recent findings on AIDS 69. Chupp, G. L., J. Alroy, L. S. Adelman, J. C. Breen, and P. R. Skolnik. 1993.
isolates. J. Protozool. 38:625–630. Myositis due to Pleistophora (Microsporidia) in a patient with AIDS. Clin.
44. Cali, A., D. M. Meisler, C. Y. Lowder, R. Lembach, L. Ayers, P. M. Infect. Dis. 16:15–21.
Takvorian, I. Rutherford, D. L. Longworth, J. McMahon, and R. T. Bryan. 70. Clark, S., S. Morgan, and J. Williams. 1995. Chronic diarrhoea associated
1991. Corneal microsporidioses: characterization and identification. J. Pro- with Septata intestinalis. Postgrad. Med. J. 71:764.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


tozool. 38:215S–217S. 71. Clarridge, J. E., S. Karkhanis, L. Rabeneck, B. Marino, and L. W. Foote.
45. Cali, A., D. M. Meisler, I. Rutherford, C. Y. Lowder, J. T. McMahon, D. L. 1996. Quantitative light microscopic detection of Enterocytozoon bieneusi in
Longworth, and R. T. Bryan. 1991. Corneal microsporidiosis in a patient stool specimens: a longitudinal study of human immunodeficiency virus-
with AIDS. Am. J. Trop. Med. Hyg. 44:463–468. infected microsporidiosis patients. J. Clin. Microbiol. 34:520–523.
46. Cali, A., J. M. Orenstein, D. P. Kotler, and R. Owen. 1991. A comparison 72. Cominos, D., D. L. Paterson, N. I. Walker, A. M. Allworth, and R. J. Kemp.
of two microsporidian parasites in enterocytes of AIDS patients with 1994. Relative infrequency of microsporidial infection in HIV infected
chronic diarrhea. J. Protozool. 38:96S–98S. patients in Queensland. Med. J. Aust. 160:452–453.
47. Cali, A., D. P. Kotler, and J. M. Orenstein. 1993. Septata intestinalis n. g., n. 73. Connolly, G. M., D. S. Ellis, J. E. Williams, G. Tovey, and B. G. Gazzard.
sp., an intestinal microsporidian associated with chronic diarrhea and dis- 1991. Use of electron microscopy in examination of faeces and rectal and
semination in AIDS patients. J. Eukaryot. Microbiol. 40:101–112. jejunal biopsy specimens. J. Clin. Pathol. 44:313–316.
48. Cali, A., P. M. Takvorian, S. Lewin, M. Rendel, C. Sian, M. Wittner, and 74. Conteas, C. N., T. Sowerby, O. G. W. Berlin, F. Dahlan, A. Nguyen, R.
L. M. Weiss. 1996. Identification of a new Nosema-like microsporidian Porschen, J. Donovan, M. Lariviere, and J. M. Orenstein. 1996. Fluores-
associated with myositis in an AIDS patient. J. Eukaryot. Microbiol. 43: cence techniques for diagnosing intestinal microsporidiosis in stool, enteric
108S. fluid, and biopsy specimens from acquired immunodeficiency syndrome
49. Cali, A., L. M. Weiss, and P. M. Takvorian. 1996. Microsporidian taxonomy patients with chronic diarrhea. Arch. Pathol. Lab. Med. 120:847–853.
and the status of Septata intestinalis. J. Eukaryot. Microbiol. 43:106S–107S. 75. Conteas, C. N., J. Donovan, O. G. W. Berlin, T. M. Sowerby, and M.
Lariviere. 1997. Comparison of fluorescent and standard light microscopy
50. Canning, E. U., and J. Lom. 1986. The microsporidia of vertebrates. Aca-
for diagnosis of microsporidia in stools of patients with AIDS and chronic
demic Press, Inc., New York, N.Y.
diarrhoea. AIDS 11:386–387.
51. Canning, E. U., and W. S. Hollister. 1987. Microsporidia of mammals—
75a.Conteas, C. N., O. G. Berlin, M. J. Lariviere, S. S. Pandhumas, C. E. Speck,
widespread pathogens or opportunistic curiosities? Parasitol. Today 3:267–
R. Porschen, and T. Nakaya. 1998. Examination of prevalence and seasonal
273.
variation of intestinal microsporidiosis in the stools of persons with chronic
52. Canning, E. U. 1988. Nuclear division and chromosome cycle in microspo-
diarrhea and human immunodeficiency virus infection. Am. J. Trop. Med.
ridia. Biosystems 21:333–340. Hyg. 58:559–561.
53. Canning, E. U., and W. S. Hollister. 1990. Enterocytozoon bieneusi (Micro- 75b.Conteas, C. N., O. G. Berlin, C. E. Speck, S. S. Pandhumas, M. J. Lariviere,
spora): prevalence and pathogenicity in AIDS patients. Trans. R. Soc. Trop. and C. Fu. 1998. Modification of the clinical course of intestinal microspo-
Med. Hyg. 84:181–186. ridiosis in the acquired immunodeficiency syndrome patients by immune
54. Canning, E. U., and W. S. Hollister. 1991. In vitro and in vivo investigations status and anti-human immunodeficiency virus therapy. Am. J. Trop. Med.
of human microsporidia. J. Protozool. 38:631–635. Hyg. 58:555–558.
55. Canning, E. U., and W. S. Hollister. 1992. Human infections with micro- 76. Corcoran, G. D., D. G. Tovey, A. H. Moody, and P. L. Chiodini. 1995.
sporidia. Rev. Med. Microbiol. 3:35–42. Detection and identification of gastrointestinal microsporidia using non-
56. Canning, E. U., and W. S. Hollister. 1992. The importance of microsporidia invasive techniques. J. Clin. Pathol. 48:725–727.
as opportunistic infections in patients with acquired immune deficiency 77. Corcoran, G. D., J. R. Isaacson, C. Daniels, and P. L. Chiodini. 1996.
syndrome. Eur. J. Gastroenterol. Hepatol. 4:422–427. Urethritis associated with disseminated microsporidiosis: clinical response
57. Canning, E. U., A. Curry, C. J. Lacey, and D. Fenwick. 1992. Ultrastructure to albendazole. Clin. Infect. Dis. 22:592–593.
of Encephalitozoon sp. infecting the conjunctival, corneal and nasal epithe- 78. Cornet, M., S. Romand, J. Warszawski, and P. Bourée. 1996. Factors
lia of a patient with AIDS. Eur. J. Protistol. 28:226–237. associated with microsporidial and cryptosporidial diarrhea in HIV infected
58. Canning, E. U. 1993. Microsporidia, p. 299–385. In J. P. Kreier and J. R. patients. Parasite 4:397–401.
Baker (ed.), Parasitic protozoa, 2nd ed., vol. 6. Academic Press, Inc., New 78a.Cotte, L., M. Rabodonirina, C. Raynal, F. Chapuis, M. A. Piens, and C.
York, N.Y. Trepo. 1997. Outbreak of intestinal microsporidiosis in HIV-infected pa-
59. Canning, E. U., A. S. Field, M. C. Hing, and D. J. Marriott. 1994. Further tients in relation with town water distribution system, abstr. I-147. In Pro-
observations on the ultrastructure of Septata intestinalis Cali, Kotler and gram and Abstracts of the 37th Interscience Conference on Antimicrobial
Orenstein 1993. Eur. J. Protistol. 30:414–422. Agents and Chemotherapy. American Society for Microbiology, Washing-
60. Caramello, P., G. Mazzucco, M. Romeo, A. Ullio, G. De Rosa, A. Lucchini, ton, D.C.
B. Forno, T. Brancale, A. Macor, C. Preziosi, and P. Gioannini. 1995. 79. Coulon, G. 1929. Presence d’un nouvel Encephalitozoon (Encephalitozoon
Clinical and microscopical features of small-intestinal microsporidiosis in brumpti n. sp.) dans le liquide cephalo-rachidien dùn sujet atteint de men-
patients with AIDS. Infection 23:362–368. ingite suraigue. Ann. Parasitol. Hum. Comp. 7:449–452.
61. Carr, A., D. Marriott, A. Field, E. Vasak, and D. A. Cooper. 1998. Treat- 80. Cox, J. C., R. C. Hamilton, and H. D. Attwood. 1979. An investigation of the
ment of HIV-1-associated microsporidiosis and cryptosporidiosis with com- route and progression of Encephalitozoon cuniculi infection in adult rabbits.
bination antiretroviral therapy. Lancet 351:256–261. J. Protozool. 26:260–265.
62. Carter, P. L., D. W. MacPherson, and R. A. McKenzie. 1996. Modified 81. Coyle, C. M., M. Wittner, D. P. Kotler, C. Noyer, J. M. Orenstein, H. B.
technique to recover microsporidian spores in sodium acetate-acetic acid- Tanowitz, and L. M. Weiss. 1996. Prevalence of microsporidiosis due to
formalin-fixed fecal samples by light microscopy and correlation with trans- Enterocytozoon bieneusi and Encephalitozoon (Septata) intestinalis amoung
mission electron microscopy. J. Clin. Microbiol. 34:2670–2673. patients with AIDS-related diarrhea: determination by polymerase chain
62a.Carville, A., K. C. Lin, L. Chalifoux, S. Tzipori, A. Lackner, and K. G. reaction to the microsporidian small-subunit rRNA gene. Clin. Infect. Dis.
Mansfield. 1998. Ribosomal RNA sequences of Enterocytozoon bieneusi 23:1002–1006.
isolates from human, pig and rhesus macaque exhibit variability over the 82. Coyle, C., M. Kent, H. B. Tanowitz, M. Wittner, and L. M. Weiss. 1998.
intergenic spacer region and 59 end of the large subunit ribosomal gene. TNP-470 is an effective antimicrosporidial agent. J. Infect. Dis. 177:515–
GenBank accession no. AF023245. 518.
63. Carville, A., K. Mansfield, G. Widmer, A. Lackner, D. Kotler, P. Wiest, T. 83. Croft, S. L., J. Williams, and I. McGowan. 1997. Intestinal microsporidiosis.
Gumbo, S. Sarbah, and S. Tzipori. 1997. Development and application of Semin. Gastroenterol. 8:45–55.
genetic probes for detection of Enterocytozoon bieneusi in formalin-fixed 84. Croppo, G. P., G. J. Leitch, S. Wallace, and G. S. Visvesvara. 1994. Immu-
278 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

nofluorescence and Western blot analysis of microsporidia using anti-En- ridian, Encephalitozoon hellem (n. sp.), from three AIDS patients with
cephalitozoon hellem immunoglobulin G monoclonal antibodies. J. Eu- keratoconjunctivitis. J. Infect. Dis. 163:617–621.
karyot. Microbiol. 41:31S. 105. Didier, E. S., J. A. Shadduck, P. J. Didier, N. Millichamp, and C. R.
85. Croppo, G. P., G. S. Visvesvara, G. J. Leitch, S. Wallace, and M. A. De Vossbrinck. 1991. Studies on ocular microsporidia. J. Protozool. 38:635–
Groote. 1997. Western blot and immunofluorescence analysis of a human 638.
isolate of Encephalitozoon cuniculi established in culture from the urine of 106. Didier, E. S., P. W. Varner, P. J. Didier, A. M. Aldras, N. J. Millichamp, M.
a patient with AIDS. J. Parasitol. 83:66–69. Murphy-Corb, R. Bohm, and J. A. Shadduck. 1994. Experimental micro-
86. Curgy, J. J., J. Vávra, and C. Vivares. 1980. Presence of ribosomal RNAs sporidiosis in immunocompetent and immunodeficient mice and monkeys.
with prokaryotic properties in microsporidia, eukaryotic organisms. Biol. Folia Parasitol. (Prague) 41:1–11.
Cell. 38:49–51. 107. Didier, E. S., J. M. Orenstein, A. Aldras, D. Bertucci, L. B. Rogers, and
87. Curry, A., L. J. McWilliam, N. Y. Haboubi, and B. K. Mandal. 1988. F. A. Janney. 1995. Comparison of three staining methods for detecting
Microsporidiosis in a British patient with AIDS. J. Clin. Pathol. 41:477–478. microsporidia in fluids. J. Clin. Microbiol. 33:3138–3145.
88. Curry, A., and E. U. Canning. 1993. Human microsporidiosis. J. Infect. 108. Didier, E. S., C. R. Vossbrink, M. D. Baker, L. B. Rogers, D. C. Bertucci,
27:229–236. and J. A. Shadduck. 1995. Identification and characterization of three
89. Da Silva, A. J., D. A. Schwartz, G. S. Visvesvara, H. de Moura, S. B. Encephalitozoon cuniculi strains. Parasitology 111:411–421.
Slemenda, and N. J. Pieniazek. 1996. Sensitive PCR diagnosis of infections 109. Didier, E. S., L. B. Rogers, A. D. Brush, S. Wong, V. Traina-Dorge, and D.
by Enterocytozoon bieneusi (microsporidia) using primers based on the Bertucci. 1996. Diagnosis of disseminated microsporidian Encephalitozoon
region coding for small-subunit rRNA. J. Clin. Microbiol. 34:986–987. hellem infection by PCR-Southern analysis and successful treatment with
90. Da Silva, A. J., S. B. Slemenda, G. S. Visvesvara, D. A. Schwartz, C. Mel albendazole and fumagillin. J. Clin. Microbiol. 34:947–952.
Wilcox, S. Wallace, and N. J. Pieniazek. 1997. Detection of Septata intesti- 110. Didier, E. S., L. B. Rogers, J. M. Orenstein, M. D. Baker, C. R. Vossbrinck,

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


nalis (microsporidia) Cali et al. 1993 using polymerase chain reaction prim- T. Van Gool, R. Hartskeerl, R. Soave, and L. M. Beaudet. 1996. Charac-
ers targeting the small subunit ribosomal RNA coding region. Mol. Diagn. terization of Encephalitozoon (Septata) intestinalis isolates cultured from
2:47–52. nasal mucosa and bronchoalveolar lavage fluids of two AIDS patients. J.
91. David, F., A. R. J. Schuitema, C. Sarfati, O. Liguory, R. A. Hartskeerl, F. Eukaryot. Microbiol. 43:34–43.
Derouin, and J. Molina. 1996. Detection and species identification of in- 111. Didier, E. S., G. S. Visvesvara, M. D. Baker, L. B. Rogers, D. C. Bertucci,
testinal microsporidia by polymerase chain reaction in duodenal biopsies M. A. De Groote, and C. R. Vossbrink. 1996. A microsporidian isolated
from human immunodeficiency virus-infected patients. J. Infect. Dis. 174: from an AIDS patient corresponds to Encephalitozoon cuniculi III, origi-
874–877. nally isolated from domestic dogs. J. Clin. Microbiol. 34:2835–2837.
92. Davis, R. M., R. L. Font, M. S. Keisler, and J. A. Shadduck. 1990. Corneal 111a.Didier, E. S. 1997. Effects of albendazole, fumagillin, and TNP-470 on
microsporidiosis. A case report including ultrastructural observations. Oph- microsporidial replication in vitro. Antimicrob. Agents Chemother. 41:
thalmology 97:953–957. 1541–1546.
93. De Aguila, C., R. Lopez-Velez, S. Fenoy, C. Turrientes, J. Cobo, R. Navajas, 112. Didier, P. J., E. S. Didier, J. M. Orenstein, and J. A. Shadduck. 1991. Fine
G. S. Visvesvara, G. P. Croppo, A. J. Da Silva, and N. J. Peniazek. 1997. structure of a new human microsporidian, Encephalitozoon hellem, in cul-
Identification of Enterocytozoon bieneusi spores in respiratory samples from ture. J. Protozool. 38:502–507.
an AIDS patient with a 2-year history of intestinal microsporidiosis. J. Clin. 113. Diesenhouse, M. C., L. A. Wilson, G. F. Corrent, G. S. Visvesvara, H. E.
Microbiol. 35:1862–1866. Grossniklaus, and R. T. Bryan. 1993. Treatment of microsporidial kerato-
94. De Girolami, P. C., C. R. Ezratty, G. Desai, A. McCullough, D. Asmuth, C. conjunctivitis with topical fumagillin. Am. J. Ophthalmol. 115:293–298.
Wanke, and M. Federman. 1995. Diagnosis of intestinal microsporidiosis by 114. Dieterich, D. T., E. A. Lew, D. P. Kotler, M. A. Poles, and J. M. Orenstein.
examination of stool and duodenal aspirate with Weber’s modified 1994. Treatment with albendazole for intestinal disease due to Enterocyto-
trichrome and Uvitex 2B strains. J. Clin. Microbiol. 33:805–810. zoon bieneusi in patients with AIDS. J. Infect. Dis. 169:178–183.
95. De Groote, M. A., G. Visvesvara, M. L. Wilson, N. J. Pieniazek, S. B. 115. DiMaria, P., B. Palic, B. A. Debrunner-Vossbrinck, J. Lapp, and C. R.
Slemenda, A. J. daSilva, G. J. Leitch, R. T. Bryan, and R. Reves. 1995. Vossbrinck. 1996. Characterization of the highly divergent U2 RNA ho-
Polymerase chain reaction and culture confirmation of disseminated En- molog in the microsporidium Vairimorpha necatrix. Nucleic Acids Res.
cephalitozoon cuniculi in a patient with AIDS: successful therapy with al- 24:515–522.
bendazole. J. Infect. Dis. 171:1375–1378. 116. Dionisio, D., G. Sterrantino, M. Meli, M. Trotta, D. Milo, and F. Leoncini.
95a.Delbac, F., P. Peyret, G. Méténier, D. David, A. Danchin, and C. P. Vivarès. 1995. Use of furazolidone for the treatment of microsporidiosis due to
1998. On proteins of the microsporidian invasive apparatus: complete se- Enterocytozoon bieneusi in patients with AIDS. Recenti Prog. Med. 86:394–
quence of a polar tube protein of Encephalitozoon cuniculi. Mol. Microbiol. 397.
29:825–834. 117. Ditrich, O., Z. Kucerová, and B. Koudela. 1994. In vitro sensitivity of
96. Deluol, A. M., J. L. Poirot, F. Heyer, P. Roux, D. Levy, and W. Rozenbaum. Encephalitozoon cuniculi and Encephalitozzoon hellem to albendazole. J.
1994. Intestinal microsporidiosis: about clinical characteristics and labora- Eukaryot. Microbiol. 41:37S.
tory diagnosis. J. Eukaryot. Microbiol. 41:33S. 118. Ditrich, O., J. Lom, I. Dykova, and J. Vávra. 1994. First case of Enterocy-
97. Deplazes, P., A. Mathis, R. Baumgartner, I. Tanner, and R. Weber. 1996. tozoon bieneusi infection in the Czech Republic: comments on the ultra-
Immunologic and molecular characteristics of Encephalitozoon-like micro- structure and teratoid sporogenesis of the parasite. J. Eukaryot. Microbiol.
sporidia isolated from humans and rabbits indicate that Encephalitozoon 41:35S–36S.
cuniculi is a zoonotic parasite. Clin. Infect. Dis. 22:557–559. 119. Dobbins, W. O., III, and W. M. Weinstein. 1985. Electron microscopy of the
98. Deplazes, P., A. Mathis, C. Müller, and R. Weber. 1996. Molecular epide- intestine and rectum in acquired immunodeficiency syndrome. Gastroen-
miology of Encepahlitozoon cuniculi and first detection of Enterocytozoon terology 88:738–749.
bieneusi in faecal samples of pigs. J. Eukaryot. Microbiol. 43:93S. 120. Docker, M. F., M. L. Kent, D. M. L. Hervio, J. S. Khattra, L. M. Weiss, A.
99. Desportes, I., Y. Le Charpentier, A. Galian, F. Bernard, B. Cochand Pri- Cali, and R. H. Devlin. 1997. Ribosomal DNA sequence of Nucleospora
ollet, A. Lavergne, P. Ravisse, and R. Modigliani. 1985. Occurrence of a salmonis Hedrick, Groff and Baxa 1991 (Microspora: Enterocytozoonidae):
new microsporidan: Enterocytozoon bieneusi n.g., n. sp., in the enterocytes implications for phylogeny and nomenclature. J. Eukaryot. Microbiol. 44:
of a human patient with AIDS. J. Protozool. 32:250–254. 55–60.
100. Desportes-Livage, I., I. Hilmarsdottir, C. Romana, S. Tanguy, A. Datry, 121. Dore, G. J., D. J. Marriott, M. C. Hing, J. L. Harkness, and A. S. Field.
and M. Gentilini. 1991. Characteristics of the microsporidian Enterocyto- 1995. Disseminated microsporidiosis due to Septata intestinalis in nine pa-
zoon bieneusi: a consequence of its development within short-living entero- tients infected with the human immunodeficiency virus: response to therapy
cytes. J. Protozool. 38:111S–113S. with albendazole. Clin. Infect. Dis. 21:70–76.
101. Desportes-Livage, I., S. Chilmonczyk, R. Hedrick, C. Ombrouck, D. Monge, 122. Doultree, J. C., A. L. Maerz, N. J. Ryan, R. W. Baird, E. Wright, S. M.
I. Maiga, and M. Gentilini. 1996. Comparative development of two micro- Crowe, and J. A. Marshall. 1995. In vitro growth of the microsporidian
sporidian species: Enterocytozoon bieneusi and Enterocytozoon salmonis, Septata intestinalis from an AIDS patient with disseminated illness. J. Clin.
reported in AIDS patients and salmonid fish, respectively. J. Eukaryot. Microbiol. 33:463–470.
Microbiol. 43:49–60. 123. Dowd, S. E., C. P. Gerba, F. J. Enriquez, and I. L. Pepper. 1998. PCR
102. Desser, S. S., H. Hong, and Y. J. Yang. 1992. Ultrastructure of the devel- amplification and species determination of microsporidia in formalin-fixed
opment of a species of Encephalitozoon cultured from the eye of an AIDS feces after immunomagnetic separation. Appl. Environ. Microbiol. 64:333–
patient. Parasitol. Res. 78:677–683. 336.
103. Di Candilo, F., G. Bassotti, M. Marroni, F. Baldelli, and A. Morelli. 1993. 123a.Dowd, S. E., C. P. Gerba, and I. L. Pepper. 1998. Confirmation of the
Enterocytozoon bieneusi detection in a patient with human immunodefi- human-pathogenic microsporidia Enterocytozoon bieneusi, Encephalitozoon
ciency virus infection and chronic diarrhoea: response to albendazole treat- intestinalis, and Vittaforma corneae in water. Appl. Environ. Microbiol.
ment. Ital. J. Gastroenterol. 25:321–323. 64:3332–3335.
104. Didier, E. S., P. J. Didier, D. N. Friedberg, S. M. Stenson, J. M. Orenstein, 124. Drobniewski, F., P. Kelly, A. Carew, B. Ngwenya, N. Luo, C. Pankhurst, and
R. W. Yee, F. O. Tio, R. M. Davis, C. Vossbrinck, N. Millichamp, and J. A. M. Farthing. 1995. Human microsporidiosis in African AIDS patients with
Shadduck. 1991. Isolation and characterization of a new human microspo- chronic diarrhea. J. Infect. Dis. 171:515–516.
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 279

124a.Duffieux, F., P. Peyret, B. A. Roe, and C. P. Vivarès. 1998. First report on 146. Franzen, C., A. Müller, B. Salzberger, G. Fätkenheuer, S. Eidt, G. Mahrle,
the systematic sequencing of the small genome of Encephalitozoon cuniculi V. Diehl, and M. Schrappe. 1995. Tissue diagnosis of intestinal microspo-
(Microspora, Protozoa): gene organization of a 4.3 kbp region on chromo- ridiosis using a fluorescent stain with Uvitex 2B. J. Clin. Pathol. 48:1009–
some I. Microb. Comp. Genomics 3:1–11. 1010.
125. Edlind, T., G. Visvesvara, J. Li, and S. Katiyar. 1994. Cryptosporidium and 147. Franzen, C., A. Müller, B. Salzberger, G. Fätkenheuer, V. Diehl, and M.
microsporidial beta-tubulin sequences: predictions of benzimidazole sensi- Schrappe. 1996. Chronic rhinosinusitis in patients with AIDS: potential role
tivity and phylogeny. J. Eukaryot. Microbiol. 41:38S. of microsporidia. AIDS 10:687–688.
126. Edlind, T., S. Katiyar, G. Visvesvara, and J. Li. 1996. Evolutionary origins 148. Franzen, C., R. Küppers, A. Müller, B. Salzberger, G. Fätkenheuer, B.
of microsporidia and basis for bezimidazole sensitivity: an update. J. Eu- Vetten, V. Diehl, and M. Schrappe. 1996. Genetic evidence for latent
karyot. Microbiol. 43:109S. Septata intestinalis infection in human immunodeficiency virus-infected pa-
126a.Edlind, T. Unpublished data. tients with intestinal microsporidiosis. J. Infect. Dis. 173:1038–1040.
127. Eeftinck Schattenkerk, J. K. M. T., T. Van Gool, R. J. van Ketel, J. F. 149. Franzen, C., A. Müller, P. Hegener, P. Hartmann, B. Salzberger, B. Fran-
Bartelsman, C. L. Kuiken, W. J. Terpstra, and P. Reiss. 1991. Clinical zen, V. Diehl, and G. Fätkenheuer. 1996. Polymerase chain reaction for
significance of small-intestinal microsporidiosis in HIV-1-infected individ- microsporidian DNA in gastrointestinal biopsy specimens of HIV-infected
uals. Lancet 337:895–898. patients. AIDS 10:F23–F27.
128. Eeftinck Schattenkerk, J. K. M. T., T. Van Gool, R. J. van Ketel, and 150. Franzen, C., A. Müller, P. Hartmann, M. Kochanek, V. Diehl, and G.
J. F. W. M. Bartelsman. 1991. Microsporidiosis in HIV1-infected individ- Fätkenheuer. 1996. Disseminated Encephalitozoon (Septata) intestinalis in-
uals. Lancet 338:323. fection in a patient with AIDS. N. Engl. J. Med. 335:1610–1611.
129. Eeftinck Schattenkerk, J. K. M. T., T. Van Gool, L. S. Schott, M. van den 151. Franzen, C., A. Müller, G. Fätkenheuer, B. Salzberger, G. Mahrle, V. Diehl,
Bergh Weerman, and J. Dankert. 1992. Chronic rhinosinusitis, a new clin- and M. Schrappe. 1996. In vitro Kultur von Encephalitozoon intestinalis

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


ical syndrome in HIV-infected patients with microsporidiosis, p. 13. In isoliert von einem HIV-infizierten Patienten mit disseminierter Microspo-
Workshop on Intestinal Microsporidia in HIV Infection. ridiose. In 17th Tagung der Deutschen Gesellschaft für Parasitologie.
130. Eeftinck Schattenkerk, J. K. M. T., and T. Van Gool. 1992. Clinical and 151a.Franzen, C., A. Müller, P. Hartmann, P. Hegener, M. Schrappe, V. Diehl,
microbiological aspects of microsporidiosis. Trop. Geogr. Med. 44:287. G. Fätkenheuer, and B. Salzberger. 1998. Polymerase chain reaction for
131. Eeftinck Schattenkerk, J. K. M. T. 1992. Clinical aspects of coccidian and diagnosis and species differentiation of microsporidia. Folia Parasitol. (Pra-
microsporidian parasites. Eur. J. Gastroenterol. Hepatol. 4:468–472. gue) 45:140–148.
132. Enriques, F. J., O. Dittrich, J. D. Palting, and K. Smith. 1997. Simple 152. Friedberg, D. N., S. M. Stenson, J. M. Orenstein, P. M. Tierno, and N. C.
diagnosis of Encephalitozoon sp. microsporidial infections by using a pan- Charles. 1990. Microsporidal keratoconjunctivitis in acquired immunode-
specific antiexospore monoclonal antibody. J. Clin. Microbiol. 35:724–729. ficiency syndrome. Arch. Ophthalmol. 108:504–508.
133. Enriques, F. J., D. Taren, A. Cruz-Lopez, M. Muramoto, J. D. Palting, and 153. Fuentealba, I. C., N. T. Mahoney, J. A. Shadduck, J. Harvill, V. Wicher, and
P. Cruz. 1998. Prevalence of intestinal encephalitozoonosis in Mexico. Clin. K. Wicher. 1992. Hepatic lesions in rabbits infected with Encepahlitozoon
Infect. Dis. 26:1227–1229. cuniculi administered per rectum. Vet. Pathol. 29:536–540.
133a.Fast, N. M., A. J. Roger, C. A. Richardson, and W. F. Doolittle. 1998. U2 154. Furuya, K., H. Nagano, and C. Satoh. 1995. Primers designed for amplifi-
and U6 snRNA genes in the microsporidian Nosema locustae: evidence for cation of Echinococcus multilocularis DNA amplify the DNA of Encepha-
a functional spliceosome. Nucleic Acids Res. 26:3202–3207. litozoon-like spores in the polymerase chain reaction. J. Eukaryot. Micro-
134. Fedorko, D. P., N. A. Nelson, and C. P. Cartwright. 1995. Identification of biol. 42:526–528.
microsporidia in stool specimens by using PCR and restriction endonucle- 154a.Gainzarain, J. C., A. Canut, M. Lozano, A. Labora, F. Carreras, S. Fenoy,
ases. J. Clin. Microbiol. 33:1739–1741. R. Navajas, N. J. Pieniazek, A. J. da Silva, and C. del Aguila. 1998. Detec-
135. Fedorko, D. P., and Y. M. Hijazi. 1996. Application of molecular techniques tion of Enterocytozoon bieneusi in two human immunodeficiency virus-
to the diagnosis of microsporidial infections. Emerging Infect. Dis. 2:183– negative patients with chronic diarrhea by polymerase chain reaction in
192. duodenal specimens and review. Clin. Infect. Dis. 27:394–398.
136. Field, A. S., M. C. Hing, S. T. Milliken, and D. J. Marriott. 1993. Micro- 155. Galtier, N., and M. Gouy. 1995. Inferring phylogenies from DNA sequences
sporidia in the small intestine of HIV-infected patients. A new diagnostic of unequal base compositions. Proc. Natl. Acad. Sci. USA 92:11317–11321.
technique and a new species. Med. J. Aust. 158:390–394. 156. Gannon, J. 1980. The course of infection of Encephalitozoon cuniculi in
137. Field, A. S., D. J. Marriott, and M. C. Hing. 1993. The Warthin-Starry stain immunodeficient and immunocompetent mice. Lab. Anim. 14:189–192.
in the diagnosis of small intestinal microsporidiosis in HIV-infected pa- 157. Garcia, L. S., R. Y. Shimizu, and D. A. Bruckner. 1994. Detection of
tients. Folia Parasitol. (Prague) 40:261–266. microsporidial spores in fecal specimens from patients diagnosed with cryp-
138. Field, A. S., D. J. Marriott, S. T. Milliken, B. J. Brew, E. U. Canning, J. G. tosporidiosis. J. Clin. Microbiol. 32:1739–1741.
Kench, P. Darveniza, and J. L. Harkness. 1996. Myositis associated with a 158. Garvey, M. J., P. G. Ambrose, and J. L. Ulmer. 1995. Topical fumagillin in
newly described microsporidian, Trachipleistophora hominis, in a patient the treatment of microsporidial keratoconjunctivitis in AIDS. Ann. Phar-
with AIDS. J. Clin. Microbiol. 34:2803–2811. macother. 29:872–874.
139. Flepp, M., B. Sauer, R. Lüthy, and R. Weber. 1996. Human microsporidi- 158a.Gatehouse, H. S., and L. A. Malone. 1998. The ribosomal RNA gene region
osis in HIV-seronegative, immunocompetent patients, abstr. LM25, p. 331. of Nosema apis (Microspora): DNA sequence for small and large subunit
In Program and Abstracts of the 35th Interscience Conference on Antimi- rRNA genes and evidence of a large tandem repeat unit size. J. Invertebr.
crobial Agents and Chemotherapy. American Society for Microbiology, Pathol. 71:97–105.
Washington, D.C. 159. Germot, A., H. Philippe, and H. Le Guyader. 1997. Evidence for loss of
140. Foudraine, N. A., G. J. Weverling, T. van Gool, M. T. L. Roos, F. de Wolf, mitochondria in microsporidia from a mitochondrial-type HSP70 in
P. P. Koopmans, P. J. van den Broek, P. L. Meenhorst, R. van Leeuwen, Nosema locustae. Mol. Biochem. Parasitol. 87:159–168.
J. M. A. Lange, and P. Reiss. 1998. Improvement of chronic diarrhoea in 160. Giang, T. T., D. P. Kotler, M. L. Garro, and J. M. Orenstein. 1993. Tissue
patients with advanced HIV-1 infection during potent antiretroviral ther- diagnosis of intestinal microsporidiosis using the chromotrope-2R modified
apy. AIDS 12:35–41. trichrome stain. Arch. Pathol. Lab. Med. 117:1249–1251.
141. Franssen, F. F., J. T. Lumeij, and F. van Knapen. 1995. Susceptibility of 160a.Goguel, J., C. Katlama, C. Sarfati, C. Maslo, C. Leport, and J. M. Molina.
Encephalitozoon cuniculi to several drugs in vitro. Antimicrob. Agents Che- 1997. Remission of AIDS-associated intestinal microsporidiosis with highly
mother. 39:1265–1268. active antiretroviral therapy. AIDS 11:1658–1659.
142. Franzen, C., G. Fätkenheuer, B. Salzberger, A. Müller, G. Mahrle, V. Diehl, 161. Grau, A., M. E. Valls, J. E. Williams, D. S. Ellis, M. J. Muntané, and C.
and M. Schrappe. 1994. Intestinal microsporidiosis in patients with ac- Nadal. 1996. Miositis por Pleistophora en un paciente con sida. Med. Clin.
quired immunodeficiency syndrome—report of three more German cases. (Barcalona) 107:779–781.
Infection 22:417–419. 162. Gunnarsson, G., D. Hurlbut, P. C. DeGirolami, M. Federman, and C.
143. Franzen, C., A. Müller, A. Schwenk, B. Salzberger, G. Fätkenheuer, G. Wanke. 1995. Multiorgan microsporidiosis: report of five cases and review.
Mahrle, V. Diehl, and M. Schrappe. 1995. Intestinal microsporidiosis with Clin. Infect. Dis. 21:37–44.
Septata intestinalis in a patient with AIDS—response to albendazole. J. In- 163. Haque, A., W. S. Hollister, A. Willcox, and E. U. Canning. 1993. The
fect. 31:237–239. antimicrosporidial activity of albendazole. J. Invertebr. Pathol. 62:171–177.
144. Franzen, C., D. A. Schwartz, G. S. Visvesvara, A. Müller, A. Schwenk, B. 164. Hartskeerl, R. A., A. R. Schuitema, and R. deWachter. 1993. Secondary
Salzberger, G. Fätkenheuer, P. Hartmann, G. Mahrle, V. Diehl, and M. structure of the small subunit ribosomal RNA sequence of the microspo-
Schrappe. 1995. Immunologically confirmed disseminated, asymptomatic ridium Encephalitozoon cuniculi. Nucleic Acids Res. 21:1489.
Encephalitozoon cuniculi infection of the gastrointestinal tract in a patient 165. Hartskeerl, R. A., A. R. Schuitema, T. Van Gool, and W. J. Terpstra. 1993.
with AIDS. Clin. Infect. Dis. 21:1480–1484. Genetic evidence for the occurrence of extra-intestinal Enterocytozoon bie-
145. Franzen, C., A. Müller, P. Hegener, B. Salzberger, P. Hartmann, G. Fät- neusi infections. Nucleic Acids Res. 21:4150.
kenheuer, V. Diehl, and M. Schrappe. 1995. Detection of microsporidia 166. Hartskeerl, R. A., T. Van Gool, A. R. Schuitema, E. S. Didier, and W. J.
(Enterocytozoon bieneusi) in intestinal biopsy specimens from human im- Terpstra. 1995. Genetic and immunological characterization of the micro-
munodeficiency virus-infected patients by PCR. J. Clin. Microbiol. 33:2294– sporidian Septata intestinalis Cali, Kotler and Orenstein, 1993: reclassifica-
2296. tion to Encephalitozoon intestinalis. Parasitology 110:277–285.
280 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

166a.Hartskeerl, R. A. Unpublished data. Antiprotozoal activities of benzimidazoles and correlations with beta-tubu-
167. Hautvast, J. L., J. J. Tolboom, T. J. Derks, P. Beckers, and R. W. Sauer- lin sequence. Antimicrob. Agents Chemother. 38:2086–2090.
wein. 1997. Asymptomatic intestinal microsporidiosis in a human immuno- 189. Katiyar, S. K., G. S. Visvesvara, and T. D. Edlind. 1995. Comparisons of
deficiency virus-seronegative, immunocompetent Zambian child. Pediatr. ribosomal RNA sequences from amitochondrial protozoa: implications for
Infect. Dis. J. 16:415–416. processing, mRNA binding and paromomycin susceptibility. Gene 152:27–
168. He, Q., G. J. Leitch, G. S. Visvesvara, and S. Wallace. 1996. Effects of 33.
nifedipine, metronidazole, and nitric oxide donors on spore germination 190. Katzwinkel-Wladarsch, S., M. Lieb, W. Helse, T. Löscher, and H. Rinder.
and cell culture infection of the microsporidia Encephalitozoon hellem and 1996. Direct amplification and species determination of microsporidian
Encephalitozoon cuniculi. Antimicrob. Agents Chemother. 40:179–185. DNA from stool specimens. Trop. Med. Int. Health 1:373–378.
169. Hedrick, R. P., J. M. Groff, and D. V. Baxa. 1991. Experimental infection 191. Katzwinkel-Wladarsch, S., P. Deplazes, R. Weber, T. Löscher, and H.
with Nucleospora salmonis n.g., n.sp.: an intranuclear microsporidium from Rinder. 1997. Comparison of polymerase chain reaction with light micros-
chinook salmon (Oncorhynchus tshawytscha). Fish Health Sect./Am. Fish. copy for detection of microsporidia in clinical specimens. Eur. J. Clin.
Soc. Newsl. 19:5. Microbiol. Infect. Dis. 16:7–10.
170. Henriksen, P. 1986. The prevalence of encephalitozoonosis in Danish 192. Keeling, P. J., and W. F. Doolittle. 1996. Alpha-tubulin from early-diverging
farmed foxes. Nord. Veterinaermed. 38:167–172. eukaryotic lineages and the evolution of the tubulin family. Mol. Biol. Evol.
171. Hewan-Lowe, K., B. Furlong, M. Sims, and D. A. Schwartz. 1997. Coinfec- 13:1297–1305.
tion with Giardia lamblia and Enterocytozoon bieneusi in a patient with 193. Keeling, P. J., and G. I. McFadden. 1998. Origins of microsporidia. Trends
acquired immunodeficiency syndrome and chronic diarrhea. Arch. Pathol. Microbiol. 6:19–23.
Lab. Med. 121:417–422. 194. Kelkar, R., P. S. R. K. Sastry, S. S. Kulkarni, T. K. Saikia, P. M. Parikh,
172. Hing, M., D. Marriott, J. Verre, and A. Field. 1992. Enteric microsporidi- and S. H. Advani. 1997. Pulmonary microsporidial infection in a patient

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


osis—response to azithromycin therapy, p. 23. In Workshop on Intestinal with CML undergoing allogeneic marrow transplant. Bone Marrow Trans-
Microsporidiosis in HIV Infection. plant. 19:179–182.
172a.Hinkle, G., H. G. Morrison, and M. L. Sogin. 1997. Genes coding for 195. Kelly, P., G. McPhail, B. Ngwenya, N. Luo, A. H. Karew, C. Pankhurst, F.
reverse transcriptase, DNA-directed RNA polymerase, and chitin synthase Drobniewski, and M. Farthing. 1994. Septata intestinalis: a new microspo-
from the microsporidian Spraguea lophii. Biol. Bull. 193:250–251. ridian in Africa. Lancet 344:271–272.
173. Hirt, R. P., B. Healy, C. R. Vossbrinck, E. U. Canning, and T. M. Embley. 196. Kemp, R. L., and J. P. Kluge. 1975. Encephalitozoon sp. in the blue-masked
1997. A mitochondrial Hsp70 orthologue in Vairimorpha necatrix: molecular lovebird, Agapornis personata (Reichenow): first confirmed report of micro-
evidence that microsporidia once contained mitochondria. Curr. Biol. sporidian infection in birds. J. Protozool. 22:489–491.
7:995–998. 197. Kent, M. L., D. M. L. Hervio, M. F. Docker, and R. H. Devlin. 1996.
174. Hollister, W. S., and E. U. Canning. 1987. An enzyme-linked immunosor- Taxonomy studies and diagnostic tests for myxosporan and microsporidian
bent assay (ELISA) for detection of antibodies to Encephalitozoon cuniculi pathogens of salmonid fishes utilising ribosomal DNA sequence. J. Eu-
and its use in determination of infections in man. Parasitology 94:209–219. karyot. Microbiol. 43:98S–99S.
175. Hollister, W. S., E. U. Canning, and M. Viney. 1989. Prevalence of anti- 198. Keohane, E. M., P. M. Takvorian, A. Cali, H. B. Tanowitz, M. Wittner, and
bodies to Encephalitozoon cuniculi in stray dogs as determined by an L. M. Weiss. 1996. Identification of a microsporidian polar tube protein
ELISA. Vet. Rec. 124:332–336. reactive monoclonal antibody. J. Eukaryot. Microbiol. 43:26–31.
176. Hollister, W. S., E. U. Canning, and A. Willcox. 1991. Evidence for wide- 199. Keohane, E. M., G. A. Orr, P. M. Takvorian, A. Cali, H. B. Tanowitz, M.
spread occurrence of antibodies to Encephalitozoon cuniculi (Microspora) Wittner, and L. M. Weiss. 1996. Purification and characterization of a
in man provided by ELISA and other serological tests. Parasitology 102: microsporidian polar tube protein. Mol. Biochem. Parasitol. 79:255–259.
33–43. 200. Keohane, E. M., G. A. Orr, H. S. Zhang, P. M. Takvorian, A. Cali, H. B.
177. Hollister, W. S., E. U. Canning, and N. I. Colbourn. 1993. A species of Tanowitz, M. Wittner, and L. M. Weiss. 1998. The molecular characteriza-
Encephalitozoon isolated from an AIDS patient: criteria for species differ- tion of the major polar tube protein gene from Encephalitozoon hellem, a
entiation. Folia Parasitol. (Prague) 40:293–295. microsporidian parasite of humans. Mol. Biochem. Parasitol. 94:227–236.
178. Hollister, W. S., E. U. Canning, N. I. Colbourn, A. Curry, and C. J. Lacey. 200a.Kester, K. E., G. W. Turiansky, and P. L. McEvoy. 1998. Nodular cutaneous
1993. Characterization of Encephalitozoon hellem (Microspora) isolated microsporidiosis in a patient with AIDS and successful treatment with
from the nasal mucosa of a patient with AIDS. Parasitology 107:351–358. long-term oral clindamycin therapy. Ann. Intern. Med. 128:911–914.
179. Hollister, W. S., E. U. Canning, N. I. Colbourn, and E. J. Aarons. 1995. 201. Knapp, P. E., J. R. Saltzmann, and P. Fairchild. 1996. Acalculous chole-
Encephalitozoon cuniculi isolated from the urine of an AIDS patient, which cystitis associated with microsporidial infection in a patient with AIDS.
differs from canine and murine isolates. J. Eukaryot. Microbiol. 42:367–372. Clin. Infect. Dis. 22:195–196.
180. Hollister, W. S., E. U. Canning, E. Weidner, A. S. Field, J. Kench, and D. J. 202. Kock, N. P., H. Petersen, T. Fenner, I. Sobottka, C. Schmetz, P. Deplazes,
Marriott. 1996. Development and ultrastructure of Trachipleistophora N. J. Pieniazek, H. Albrecht, and J. Schottelius. 1997. Species-specific
hominis n.g., n.sp. after in vitro isolation from an AIDS patient and inoc- identification of microsporidia in stool and intestinal biopsy specimens by
ulation into athymic mice. Parasitology 112:143–154. the polymerase chain reaction. Eur. J. Clin. Microbiol. Infect. Dis. 16:369–
181. Hollister, W. S., E. U. Canning, and C. L. Anderson. 1996. Identification of 379.
microsporidia causing human disease. J. Eukaryot. Microbiol. 43:104S– 203. Kokoskin, E., T. W. Gyorkos, A. Camus, L. Cedilotte, T. Purtill, and B.
105S. Ward. 1994. Modified technique for efficient detection of microsporidia.
181a.Hutin, Y. J. F., M.-N. Sombardier, O. Liguory, C. Sarfati, F. Derouin, J. J. Clin. Microbiol. 32:1074–1075.
Modai, and J. M. Molina. 1998. Risk factors for intestinal microsporidiosis 204. Kotler, D. P., A. Francisco, F. Clayton, J. V. Scholes, and J. M. Orenstein.
in patients with human immunodeficiency virus infection: a case-control 1990. Small intestinal injury and parasitic diseases in AIDS. Ann. Intern.
study. J. Infect. Dis. 178:904–907. Med. 113:444–449.
182. Ignatius, R., S. Henschel, O. Liesenfeld, U. Mansmann, W. Schmidt, S. 205. Kotler, D. P., S. Reka, K. Chow, and J. M. Orenstein. 1993. Effects of
Koppe, T. Schneider, W. Heise, U. Futh, E. O. Riecken, H. Hahn, and R. enteric parasitoses and HIV infection upon small intestinal structure and
Ullrich. 1997. Comparative evaluation of modified trichrome and Uvitex 2B function in patients with AIDS. J. Clin. Gastroenterol. 16:10–15.
stains for detection of low numbers of microsporidial spores in stool spec- 206. Kotler, D. P., T. T. Giang, M. L. Garro, and J. M. Orenstein. 1994. Light
imens. J. Clin. Microbiol. 35:2266–2269. microscopic diagnosis of microsporidiosis in patients with AIDS. Am. J.
183. Issi, I. V. 1986. Microsporidia as a phylum of parasitic protozoa. Acad. Sci. Gastroenterol. 89:540–544.
USSR (Leningrad) 10:6–136. 207. Kotler, D. P., and J. M. Orenstein. 1994. Prevalence of intestinal micro-
184. Josephson, G. D., J. Sarlin, J. Reidy, and R. Pincus. 1996. Microsporidial sporidiosis in HIV-infected individuals referred for gastroenterological
rhinosinusitis: is this the next pathogen to infect the sinuses of the immu- evaluation. Am. J. Gastroenterol. 89:1998–2002.
nocompromised host? Otolaryngol. Head Neck Surg. 114:137–139. 208. Kotler, D. P., and J. M. Orenstein. 1995. Microsporidia, p. 1129–1140. In
185. Joste, N. E., J. D. Rich, K. J. Busam, and D. A. Schwartz. 1996. Autopsy M. J. Blaser, P. D. Smith, J. I. Ravdin, H. B. Greenberg, and R. L. Guerrant
verification of Encephalitozoon intestinalis (microsporidiosis) eradication (ed.), Infections of the gastrointestinal tract. Raven Press, New York, N.Y.
following albendazole therapy. Arch. Pathol. Lab. Med. 120:199–203. 209. Koudela, B., J. Vı́tovec, Z. Kucerová, O. Ditrich, and J. Trávnı́cek. 1993.
186. Kamaishi, T., T. Hashimoto, Y. Nakamura, F. Nakamura, S. Mureta, N. The severe combined immunodeficient mouse as a model for Encephalito-
Okada, K. Okamoto, M. Shimizu, and M. Hasegawa. 1996. Protein phy- zoon cuniculi microsporidiosis. Fol. Parasitol. (Prague) 40:279–286.
logeny of translation elongation factor EF-1a suggests microsporidians are 210. Koudela, B., J. Lom, J. Vı́tovec, Z. Kucerová, O. Ditrich, and J. Trávnı́cek.
extremely ancient eukaryotes. J. Mol. Evol. 42:257–263. 1994. In vivo efficacy of albendazole against Encephalitozoon cuniculi in
187. Kamaishi, T., T. Hashimoto, Y. Nakamura, Y. Masuda, F. Nakamura, K. SCID mice. J. Eukaryot. Microbiol. 41:49S–50S.
Okamoto, M. Shimizu, and M. Hasegawa. 1996. Complete nucleotide se- 211. Lacey, C. J., A. M. Clarke, P. Fraser, T. Metcalfe, G. Bonsor, and A. Curry.
quences of the genes encoding translation elongation factors 1alpha and 2 1992. Chronic microsporidian infection of the nasal mucosae, sinuses and
from a microsporidian parasite, Glugea plecoglossi: implications for the conjunctivae in HIV disease. Genitourin. Med. 68:179–181.
deepest branching of eukaryotes. J. Biochem. 120:1095–1103. 212. Lambl, B. B., M. Federman, D. Pleskow, and C. A. Wanke. 1996. Malab-
188. Katiyar, S. K., V. R. Gordon, G. L. McLaughlin, and T. D. Edlind. 1994. sorption and wasting in AIDS patients with microsporidia and pathogen-
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 281

negative diarrhea. AIDS 10:739–744. 236a.Mathis, A., et al. Unpublished data.


213. Lanzafame, M., S. Bonora, G. Di Perri, B. Allegranzi, I. Guasparri, A. 237. Matsubayashi, H., T. Koike, T. Mikata, and S. Hagiwara. 1959. A case of
Cazzadori, S. Ferrari, S. Vento, and E. Concia. 1997. Microsporidium Encephalitozoon-like-body infection in man. Arch. Pathol. 67:181–187.
species in pulmonary cavitary lesions of AIDS patients infected with Rhodo- 238. McCluskey, P. J., P. V. Goonan, D. J. Marriott, and A. S. Field. 1993.
coccus equi. Clin. Infect. Dis. 25:926–927. Microsporidial keratoconjunctivitis in AIDS. Eye 7:80–83.
214. Larsson, R. 1986. Ultrastructure, function, and classification of microspo- 239. McDougall, R. J., M. W. Tandy, R. E. Boreham, D. J. Stenzel, and P. J.
ridia. Prog. Protistol. 1:325–390. O’Donoghue. 1993. Incidental finding of a microsporidian parasite from an
215. Lecuit, M., E. Oksenhendler, and C. Sarfati. 1994. Use of albendazole for AIDS patient. J. Clin. Microbiol. 31:436–439.
disseminated microsporidian infection in a patient with AIDS. Clin. Infect. 239a.McInnes, E. F., and C. G. Stewart. 1991. The pathology of subclinical
Dis. 19:332–333. infection of Encephalitozoon cuniculi in canine dams producing pups with
216. Ledford, D. K., M. D. Overman, A. Gonzalo, A. Cali, W. Mester, and R. F. overt encephalitozoonosis. J. S. Afr. Vet. Assoc. 62:51–54.
Lockey. 1985. Microsporidiosis myositis in a patient with acquired immu- 240. McWhinney, P. H., D. Nathwani, S. T. Green, J. F. Boyd, and J. A. Forrest.
nodeficiency syndrome. Ann. Intern. Med. 102:628–630. 1991. Microsporidiosis detected in association with AIDS-related sclerosing
217. Leipe, D. D., J. H. Gunderson, T. A. Nerad, and M. L. Sogin. 1993. Small cholangitis. AIDS 5:1394–1395.
subunit ribosomal RNA1 of Hexamita inflata and the quest for the first 241. McWilliam, L. J., and A. Curry. 1990. Intestinal microsporidiosis in
branch in the eukaryotic tree. Mol. Biochem. Parasitol. 59:41–48. AIDS. J. Clin. Pathol. 43:173–174.
218. Leitch, G. J., Q. He, S. Wallace, and G. S. Visvesvara. 1993. Inhibition of 242. Mertens, R. B., E. S. Didier, M. C. Fishbein, D. C. Bertucci, L. B. Rogers,
the spore polar filament extrusion of the microsporidium, Encephalitozoon and J. M. Orenstein. 1997. Encepahlitozoon cuniculi microsporidiosis: in-
hellem, isolated from an AIDS patient. J. Eukaryot. Microbiol. 40:711–717. fection of the brain, heart, kidneys, trachea, adrenal glands, and urinary
219. Leitch, G. J., M. Scanlon, A. Shaw, G. S. Visvesvara, and S. Wallace. 1997. bladder in a patient with AIDS. Mod. Pathol. 10:68–77.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


Use of a fluorescent probe to assess the activities of candidate agents 243. Metcalfe, T. W., R. M. Doran, P. L. Rowlands, A. Curry, and C. J. Lacey.
against intracellular forms of Encephalitozoon microsporidia. Antimicrob. 1992. Microsporidal keratoconjunctivitis in a patient with AIDS. Br. J.
Agents Chemother. 41:337–344. Ophthalmol. 76:177–178.
220. Levaditi, C., S. Nicolau, and R. Schoen. 1923. L’agent étiologique de 244. Michiels, J. F., P. Hofman, M. C. Saint Paul, R. Loubiere, E. Bernard, and
l’encéphalite épizootique du lapin (Encephalitozoon cuniculi). C. R. Soc. Y. LeFichoux. 1993. Pathological features of intestinal microsporidiosis in
Biol. Paris 89:984–986. HIV positive patients. A report of 13 new cases. Pathol. Res. Pract. 189:
221. Li, J., S. K. Katiyar, A. Hamelin, G. S. Visvesvara, and T. D. Edlind. 1998. 377–383.
Tubulin genes from AIDS-associated microsporidia and implications for 245. Modigliani, R., C. Bories, Y. Le Charpentier, M. Salmeron, B. Messing, A.
phylogeny and benzimidazole sensitivity. Mol. Biochem. Parasitol. 78:289– Galian, J. C. Rambaud, A. Lavergne, B. Cochand Priollet, and I. Desportes.
295. 1985. Diarrhoea and malabsorption in acquired immune deficiency syn-
222. Liguory, O., F. David, C. Sarfati, A. R. J. Schuitema, R. A. Hartskeerl, F. drome: a study of four cases with special emphasis on opportunistic proto-
Derouin, J. Modai, and J. M. Molina. 1997. Diagnosis of infections caused zoan infestations. Gut 26:179–187.
by Enterocytozoon bieneusi and Encephalitozoon intestinalis using polymer- 246. Molina, J. M., C. Sarfati, B. Beauvais, M. Lémann, A. Lesourd, F. Ferchal,
ase chain reaction in stool specimens. AIDS 11:723–726. I. Casin, P. Lagrange, R. Modigliani, F. Derouin, and J. Modai. 1993.
222a.Liguory, O., F. David, C. Sarfati, F. Derouin, and J. M. Molina. 1998. Intestinal microsporidiosis in human immunodeficiency virus-infected pa-
Determination of types of Enterocytozoon bieneusi strains isolated from tients with chronic unexplained diarrhea: prevalence and clinical and bio-
patients with intestinal microsporidiosis. J. Clin. Microbiol. 36:1882–1885. logic features. J. Infect. Dis. 167:217–221.
223. Lom, J. 1993. Introductory remarks on microsporidia in the AIDS era. Folia 247. Molina, J. M., E. Oksenhendler, B. Beauvais, C. Sarfati, A. Jaccard, F.
Parasitol. (Prague) 40:255–256. Derouin, and J. Modai. 1995. Disseminated microsporidiosis due to Septata
224. Lowder, C. Y., D. M. Meisler, J. T. McMahon, D. L. Longworth, and I. intestinalis in patients with AIDS: clinical features and response to albenda-
Rutherford. 1990. Microsporidia infection of the cornea in a man seropos- zole therapy. J. Infect. Dis. 171:245–249.
itive for human immunodeficiency virus. Am. J. Ophthalmol. 109:242–244. 248. Molina, J. M., J. Goguel, C. Sarfati, C. Chastang, I. Desportes-Livage, J. F.
225. Lowder, C. Y. 1993. Ocular microsporidiosis. Int. Ophthalmol. Clin. 33:145– Michiels, C. Maslo, C. Katlama, L. Cotte, C. Leport, F. Raffi, F. Derouin,
151. and J. Modai. 1997. Potential efficacy of fumagillin in intestinal microspo-
226. Lowder, C. Y., J. T. McMahon, D. M. Meisler, E. M. Dodds, L. H. Cala- ridiosis due to Enterocytozoon bieneusi in patients with HIV infection:
brese, E. S. Didier, and A. Cali. 1996. Microsporidial keratoconjunctivitis results of a drug screening study. The French Microsporidiosis Study
caused by Septata intestinalis in a patient with acquired immunodeficiency Group. AIDS 11:1603–1610.
syndrome. Am. J. Ophtalmol 121:715–717. 248a.Molina, J. M., C. Chastang, J. Goguel, J. F. Michiels, C. Sarfati, I. Des-
227. Lucas, S. B., L. Papadaki, C. Conlon, N. Sewankambo, R. Goodgame, and portes-Livage, J. Horton, F. Derouin, and J. Modai. 1998. Albendazole for
D. Serwadda. 1989. Diagnosis of intestinal microsporidiosis in patients with treatment and prophylaxis of microsporidiosis due to Encephalitozoon in-
AIDS. J. Clin. Pathol. 42:885–887. testinalis in patients with AIDS: a randomized double-blind controlled trial.
227a.Luján, H. D., J. T. Conrad, C. G. Clark, M. C. Touz, F. Delbac, C. P. J. Infect. Dis. 177:1373–1377.
Vivarès, and T. E. Nash. 1998. Detection of microsporidia spore-specific 249. Monneret, G., M. Rabodonirina, L. Cotte, I. Desportes-Livage, S. Paulus,
antigens by monoclonal antibodies. Hybridoma 17:237–243. O. Bastien, J. Troncy, A. Lachaux, A. Boibieux, and F. Roumanet-Dubois.
228. Luna, V. A., B. K. Stewart, D. L. Bergeron, C. R. Clausen, J. J. Plorde, and 1995. Detection of intestinal microsporidian spores in non-human immu-
T. R. Fritsche. 1995. Use of the fluorochrome calcofluor white in the nodeficiency virus infected population. Ann. Biol. Clin. (Paris) 53:563–564.
screening of stool specimens for spores of microsporidia. Am. J. Clin. 250. Moss, R. B., L. M. Beaudet, B. M. Wenig, A. M. Nelson, A. Firpo, U. Punja,
Pathol. 103:656–659. T. S. Scott, and M. A. Kaliner. 1997. Microsporidium-associated sinusitis.
229. Macher, A. M., R. Neafie, P. Angritt, and S. M. Masur. 1988. Microspo- Ear Nose Throat J. 76:95–101.
ridial myositis and the acquired immunodeficiency syndrome (AIDS): a 251. Moura, H., D. A. Schwartz, F. Bornay-Llinares, F. C. Sodre, S. Wallace, and
four-year follow-up. Ann. Intern. Med. 109:343. G. S. Visvesvara. 1997. A new and improved “quick-hot Gram-chromo-
230. Macher, A. M., R. Neafie, P. Angritt, S. M. Tuur, R. P. Nelson, and A. Cali. trope” technique that differentially stains microsporidian spores in clinical
1988. Case for diagnosis - Military Medicine - July 1988. Mil. Med. 153: samples, including paraffin-embedded tissue sections. Arch. Pathol. Lab.
M41–M48. Med. 121:888–893.
231. Malone, L. A., and C. A. McIvor. 1993. Pulsed-field gel electrophoresis from 251a.Müller, M. 1997. What are the microsporidia? Parasitol. Today 13:455–456.
four microsporidian isolates. J. Invertebr. Pathol. 61:203–205. 252. Munderloh, U. G., T. J. Kurtti, and S. E. Ross. 1990. Electrophoretic
232. Malone, L. A., and C. A. McIvor. 1995. DNA probes for two microsporidia, characterization of chromosomal DNA from two microsporidia. J. Inver-
Nosema bombycis and Nosema costelytrae. J. Invertebr. Pathol. 65:269–273. tebr. Pathol. 56:243–248.
233. Malone, L. A., and C. A. McIvor. 1996. Use of nucleotide sequence data to 253. Muscat, I. 1990. Human microsporidiosis. J. Infect. 21:125–129.
identify a microsporidian pathogen of Pieris rapae (Lepidoptera, Pieridae). 254. Naegeli, K. W. 1857. Über die neue Krankheit der Seidenraupe und ver-
J. Invertebr. Pathol. 68:231–238. wandte Organismen. Bot. Z. 15:760–761.
234. Mansfield, K. G., A. Carville, D. Shvetz, J. MacKey, S. Tzipori, and A. A. 255. Nagano, H., C. Satoh, and K. Furuya. 1996. Nucleotide sequences of DNA
Lackner. 1997. Identification of an Enterocytozoon bieneusi-like microspo- fragments of Encephalitozoon cuniculi amplified by polymerase chain reac-
ridian parasite in simian-immunodeficiency-virus-inoculated macaques with tion with primers regarded as specific for Echinococcus. J. Eukaryot. Mi-
hepatobiliary disease. Am. J. Pathol. 150:1395–1405. crobiol. 43:217–221.
234a.Mansfield, K. G., et al. Unpublished data. 256. Niederkorn, J. Y., J. A. Shadduck, and E. Weidner. 1980. Antigenic cross-
235. Margileth, A. M., A. J. Strano, R. Chandra, R. Neafie, M. Blum, and R. M. reactivity amoung different microsporidian spores as determined by immu-
McCully. 1973. Dissiminated nosematosis in an immunologically compro- nofluorescence. J. Parasitol. 66:675–677.
mised infant. Arch. Pathol. 95:145–150. 257. Oldfield, E. C. 1995. Albendazole: new hope for treatment of microspo-
236. Mathis, A., M. Michel, H. Kuster, C. Muller, R. Weber, and P. Deplazes. ridiosis in AIDS. Am. J. Gastroenterol. 90:159–160.
1997. Two Encephalitozoon cuniculi strains of human origin are infectious 258. Olsen, G. J., and C. R. Woese. 1993. Ribosomal RNA: a key to phylogeny.
to rabbits. Parasitology 114:29–35. FASEB J. 7:113–123.
282 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

259. Ombrouck, C., B. Romestand, J. M. da Costa, I. Desportes Livage, A. 280. Pomport-Castillon, C., B. Romestand, and J. F. De Jonckheere. 1997.
Datry, F. Coste, G. Bouix, and M. Gentilini. 1995. Use of cross-reactive Identification and phylogenetic relationships of microsporidia by riboprint-
antigens of the microsporidian Glugea atherinae for the possible detection ing. J. Eukaryot. Microbiol. 44:540–544.
of Enterocytozoon bieneusi by western blot. Am. J. Trop. Med. Hyg. 52:89– 281. Preston, D. C. 1993. Electrophysiology of microsporidia myositis in an
93. AIDS patient. Muscle Nerve 16:1420–1422.
260. Ombrouck, C., L. Ciceron, and I. Desportes-Livage. 1996. Specific and 281a.Pulparampil, N., D. Graham, D. Phalen, and K. Snowden. 1998. Enceph-
rapid detection of microsporidia in stool specimens from AIDS patients by alitozoon hellem in two electus parrots (Electus roratus): identification from
PCR. Parasite 3:85–86. archival tissues. J. Eukaryot. Microbiol. 45:651–655.
261. Ombrouck, C., I. Desportes-Livage, A. Achbarou, and M. Gentilini. 1996. 282. Rabeneck, L., F. Gyorkey, R. M. Genta, P. Gyorkey, L. W. Foote, and J. M.
Specific detection of the microsporidia Encephalitozoon intestinalis in AIDS Risser. 1993. The role of microsporidia in the pathogenesis of HIV-related
patients. C. R. Acad. Sci. 319:39–43. chronic diarrhea. Ann. Intern. Med. 119:895–899.
262. Ombrouck, C., L. Ciceron, S. Biligui, S. Brown, P. Marechal, T. Van Gool, 283. Rabeneck, L., R. M. Genta, F. Gyorkey, J. E. Clarridge, P. Gyorkey, and
A. Datry, M. Danis, and I. Desportes-Livage. 1997. Specific PCR assay for L. W. Foote. 1995. Observations on the pathological spectrum and clinical
direct detection of intestinal microsporidia Enterocytozoon bieneusi and course of microsporidiosis in men infected with the human immunodefi-
Encephalitozoon intestinalis in fecal specimens from human immunodefi- ciency virus: follow-up study. Clin. Infect. Dis. 20:1229–1235.
ciency virus-infected patients. J. Clin. Microbiol. 35:652–655. 284. Rabodonirina, M., M. Bertocchi, I. Desportes-Livage, L. Cotte, H. Levrey,
263. Orenstein, J. M., J. Chiang, W. Steinberg, P. D. Smith, H. Rotterdam, and M. A. Piens, G. Monneret, M. Celard, J. F. Mornex, and M. Mojon. 1996.
D. P. Kotler. 1990. Intestinal microsporidiosis as a cause of diarrhea in Enterocytozoon bieneusi as a cause of chronic diarrhea in a heart-lung
human immunodeficiency virus-infected patients: a report of 20 cases. transplant recipient who was seronegative for human immunodeficiency
Hum. Pathol. 21:475–481. virus. Clin. Infect. Dis. 23:114–117.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


264. Orenstein, J. M., W. Zierdt, C. Zierdt, and D. P. Kotler. 1990. Identification 285. Ragan, M. A. 1988. Ribosomal RNA and the major lines of evolution: a
of spores of Enterocytozoon bieneusi in stool and duodenal fluid from AIDS perspective. Biosystems 21:177–187.
patients. Lancet 336:1127–1128. 286. Raynaud, L., F. Delbac, V. Broussolle, M. Rabodonirina, V. Girault, M.
265. Orenstein, J. M. 1991. Microsporidiosis in the acquired immunodeficiency Wallon, G. Cozon, C. P. Vivares, and F. Peyron. 1998. Identification of
syndrome. J. Parasitol. 77:843–864. Encephalitozoon intestinalis in travelers with chronic diarrhea by specific
266. Orenstein, J. M., M. Tenner, A. Cali, and D. P. Kotler. 1992. A microspo- PCR amplification. J. Clin. Microbiol. 36:37–40.
ridian previously undescribed in humans, infecting enterocytes and macro- 287. Remadi, S., J. Dumais, K. Wafa, and W. MacGee. 1995. Pulmonary micro-
phages, and associated with diarrhea in an acquired immunodeficiency sporidiosis in a patient with the acquired immunodeficiency syndrome. A
syndrome patient. Hum. Pathol. 23:722–728. case report. Acta Cytol. 39:1112–1116.
267. Orenstein, J. M., M. Tenner, and D. P. Kotler. 1992. Localization of 288. Rijpstra, A. C., E. U. Canning, R. J. van Ketel, J. K. Eeftinck Schattenkerk,
infection by the microsporidian Enterocytozoon bieneusi in the gastrointes- and J. J. Laarman. 1988. Use of light microscopy to diagnose small-intes-
tinal tract of AIDS patients with diarrhea. AIDS 6:195–197. tinal microsporidiosis in patients with AIDS. J. Infect. Dis. 157:827–831.
268. Orenstein, J. M., D. T. Dieterich, and D. P. Kotler. 1992. Systemic dissem- 289. Rinder, H., S. Katzwinkel-Wladarsch, and T. Löscher. 1997. Evidence for
ination by a newly recognized intestinal microsporidia species in AIDS. the existence of genetically distinct strains of Enterocytozoon bieneusi. Para-
AIDS 6:1143–1150. sitol. Res. 83:670–672.
269. Orenstein, J. M., D. T. Dieterich, and D. P. Kotler. 1993. Albendazole as a 290. Rinder, H., K. Janitschke, H. Aspöck, A. J. da Silva, P. Deplazes, D. P.
treatment for intestinal and disseminated microsporidiosis due to Septata Fedorko, C. Franzen, U. Futh, F. Hünger, A. Lehmacher, C. G. Meyer, J. M.
intestinalis in AIDS patients: a report of four cases. AIDS 7(Suppl. 3):S40– Molina, J. Sandford, R. Weber, T. Löscher, and the Diagnostic Multicenter
S42. Study Group on Microsporidia. 1998. Blinded, externally controlled mul-
270. Orenstein, J. M., E. Lew, M. A. Poles, and D. Dieterich. 1995. The endo- ticenter evaluation of light microscopy and PCR for detection of microspo-
scopic brush cytology specimen in the diagnosis of intestinal microspo- ridia in stool specimens. J. Clin. Microbiol. 36:1814–1818.
ridiosis. AIDS 9:1199–1201. 291. Rosberger, D. F., O. N. Serdarevic, R. A. Erlandson, R. T. Bryan, D. A.
271. Orenstein, J. M., H. P. Gaetz, A. T. Yachnis, S. S. Frankel, R. B. Mertens, Schwartz, G. S. Visvesvara, and P. C. Keenan. 1993. Successful treatment
and E. S. Didier. 1997. Disseminated microsporidiosis in AIDS: are any of microsporidial keratoconjunctivitis with topical fumagillin in a patient
organs spared. AIDS 11:385–386. with AIDS. Cornea 12:261–265.
272. Owen, R. L. 1997. Polymerase chain reaction of stool: a powerful tool for 292. Rossi, R. M., C. Wanke, and M. Federman. 1996. Microsporidian sinusitis
specific diagnosis and epidemiologic investigation of enteric microsporidia in patients with the acquired immunodeficiency syndrome. Laryngoscope
infections. AIDS 11:817–818. 106:966–971.
273. Peacock, C. S., C. Blanshard, D. G. Tovey, D. S. Ellis, and B. G. Gazzard. 293. Rotterdam, H. 1993. The acquired immunodeficiency syndrome and the
1991. Histological diagnosis of intestinal microsporidiosis in patients with evolution of new micro-organisms: a pathologist’s view. Hum. Pathol. 24:
AIDS. J. Clin. Pathol. 44:558–563. 935–936.
274. Pedro-de-Lelis, F. J., V. Sabater-Marco, and A. Herrera-Ballester. 1995. 294. Ryan, N. J., G. Sutherland, K. Coughlan, M. Globan, J. Doultree, J. Mar-
Necrotizing maxillary sinus mucormycosis related to candidiasis and micro- shall, R. W. Baird, J. Pedersen, and B. Dwyer. 1993. A new trichrome-blue
sporidiosis in an AIDS patient. AIDS 9:1386–1388. stain for detection of microsporidial species in urine, stool, and nasopha-
275. Peterson, N., J. A. Liu, and J. Shadduck. 1988. Encephalitozoon cuniculi: ryngeal specimens. J. Clin. Microbiol. 31:3264–3269.
quantitation of parasites and evaluation of viability. J. Protozool. 35:430– 295. Sandfort, J., A. Hannemann, H. Gelderblom, K. Stark, R. L. Owen, and B.
434. Ruf. 1994. Enterocytozoon bieneusi infection in an immunocompetent pa-
275a.Peyretaillade, E., V. Broussolle, P. Peyret, G. Méténier, M. Gouy, and C. P. tient who had acute diarrhea and who was not infected with the human
Vivarès. 1998. Microsporidia, amitochondrial protists, possess a 70-kDa immunodeficiency virus. Clin. Infect. Dis. 19:514–516.
heat shock protein gene of mitochondrial evolutionary origin. Mol. Biol. 296. Sax, P. E., J. D. Rich, W. S. Pieciak, and Y. M. Trnka. 1995. Intestinal
Evol. 15:683–689. microsporidiosis occurring in a liver transplant recipient. Transplantation
275b.Peyretaillade, E., C. Biderre, P. Peyret, F. Duffieux, G. Méténier, M. Gouy, 60:617–618.
B. Michot, and C. P. Vivarès. 1998. Microsporidian Encephalitozoon cunic- 297. Scaglia, M., L. Sacchi, S. Gatti, A. M. Bernuzzi, P. Polver, I. Piacentini, E.
uli, a unicellular eukaryote, with a unusual chromosomal dispersion of Concia, G. P. Croppo, A. J. da Silva, N. J. Pieniazek, S. B. Slemenda, S.
ribosomal genes and a LSU rRNA reduced to the universal core. Nucleic Wallace, G. J. Leitch, and G. S. Visvesvara. 1994. Isolation and identifica-
Acids Res. 26:3513–3520. tion of Encephalitozoon hellem from an Italian AIDS patient with dissem-
276. Pieniazek, N. J., A. J. da Silva, S. B. Slemenda, G. S. Visvesvara, T. J. inated microsporidiosis. APMIS 102:817–827.
Kurtti, and C. Yasunaga. 1996. Nosema trichoplusiae is a synonym of 298. Scaglia, M., L. Sacchi, G. P. Croppo, A. da Silva, S. Gatti, S. Corona, A.
Nosema bombycis based on the sequence of the small subunit ribosomal Orani, A. M. Bernuzzi, N. J. Pieniazek, S. B. Slemenda, S. Wallace, and
RNA coding region. J. Invertebr. Pathol. 67:316–317. G. S. Visvesvara. 1997. Pulmonary microsporidiosis due to Encephalitozoon
276a.Pieniazek, N. J. Unpublished data. hellem in a patient with AIDS. J. Infect. 34:119–126.
277. Pinnolis, M., P. R. Egbert, R. L. Font, and F. C. Winter. 1981. Nosematosis 299. Scaglia, M., S. Gatti, L. Sacchi, S. Corona, G. Chichina, A. M. Bernuzzi, G.
of the cornea. Case report, including electron microscopic studies. Arch. Barbarini, G. P. Croppo, A. J. da Silva, N. J. Pieniazek, and G. S. Visves-
Ophthalmol. 99:1044–1047. vara. 1998. Asymptomatic respiratory tract microsporidiosis due to Enceph-
278. Pol, S., C. Romana, S. Richard, F. Carnot, J. L. Dumont, H. Bouche, G. alitozoon hellem in three patients with AIDS. Clin. Infect. Dis. 26:174–176.
Pialoux, M. Stern, J. F. Pays, and P. Berthelot. 1992. Enterocytozoon bie- 300. Schmidt, E., and J. A. Shadduck. 1983. Murine encephalitozoonosis model
neusi infection in acquired immunodeficiency syndrome-related sclerosing for studying the host-parasite relationship of a chronic infection. Infect.
cholangitis. Gastroenterology 102:1778–1781. Immun. 40:936–942.
279. Pol, S., C. A. Romana, S. Richard, P. Amouyal, I. Desportes Livage, F. 301. Schmidt, W., T. Schneider, W. Heise, J. D. Schulzke, T. Weinke, R. Igna-
Carnot, J. F. Pays, and P. Berthelot. 1993. Microsporidia infection in tius, R. L. Owen, M. Zeitz, E. O. Riecken, and R. Ullrich. 1997. Mucosal
patients with the human immunodeficiency virus and unexplained cholan- abnormalities in microsporidiosis. AIDS 11:1589–1594.
gitis. N. Engl. J. Med. 328:95–99. 301a.Schnittger, L., et al. Unpublished data.
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 283

302. Schottelius, J., Y. Lo, and C. Schmetz. 1995. Septata intestinalis and En- 328. Sobottka, I., H. Albrecht, H. Schafer, J. Schottelius, G. S. Visvesvara, R.
cephalitozoon cuniculi: cross-reactivity between two microsporidian species. Laufs, and D. A. Schwartz. 1995. Disseminated Encephalitozoon (Septata)
Folia Parasitol. (Prague) 42:169–172. intestinalis infection in a patient with AIDS: novel diagnostic approaches
303. Schuitema, A. R. J., R. A. Hartskeerl, T. van Gool, R. Laxminarayan, and and autopsy-confirmed parasitological cure following treatment with al-
W. J. Terpstra. 1993. Application of the polymerase chain reaction for the bendazole. J. Clin. Microbiol. 33:2948–2952.
diagnosis of microsporidiosis. AIDS 7(Suppl. 3):S62–S63. 329. Sobottka, I., H. Albrecht, J. Schottelius, C. Schmetz, M. Bentfeld, R. Laufs,
304. Schwartz, D. A., R. T. Bryan, K. O. Hewan Lowe, G. S. Visvesvara, R. and D. A. Schwartz. 1995. Self-limited traveller’s diarrhea due to a dual
Weber, A. Cali, and P. Angritt. 1992. Disseminated microsporidiosis (En- infection with Enterocytozoon bieneusi and Cryptosporidium parvum in an
cephalitozoon hellem) and acquired immunodeficiency syndrome. Autopsy immunocompetent HIV-negative child. Eur. J. Clin. Microbiol. Infect. Dis.
evidence for respiratory acquisition. Arch. Pathol. Lab. Med. 116:660–668. 14:919–920.
305. Schwartz, D. A., G. S. Visvesvara, G. J. Leitch, L. Tashjian, M. Pollack, J. 330. Sorvillo, F., and P. Kerndt. 1995. Pathogenicity of the microsporidia. AIDS
Holden, and R. T. Bryan. 1993. Pathology of symptomatic microsporidial 9:215.
(Encephalitozoon hellem) bronchiolitis in the acquired immunodeficiency 331. Soule, J. B., A. L. Halverson, R. B. Becker, M. C. Pistole, and J. M.
syndrome: a new respiratory pathogen diagnosed from lung biopsy, bron- Orenstein. 1997. A patient with acquired immunodeficiency syndrome and
choalveolar lavage, sputum, and tissue culture. Hum. Pathol. 24:937–943. untreated Encephalitozoon (Septata) intestinalis microsporidiosis leading to
306. Schwartz, D. A., G. S. Visvesvara, M. C. Diesenhouse, R. Weber, R. L. Font, small bowel perforation. Response to albendazole. Arch. Pathol. Lab. Med.
L. A. Wilson, G. Corrent, O. N. Serdarevic, D. F. Rosberger, P. C. Keenen, 121:880–887.
H. E. Grossniklaus, K. Hewan-Lowe, and R. T. Bryan. 1993. Pathologic 332. Sowerby, T. M., C. N. Conteas, O. G. Berlin, and J. Donovan. 1995. Mi-
features and immunofluorescent antibody demonstration of ocular micro- crosporidiosis in patients with relatively preserved CD4 counts. AIDS
sporidiosis (Encephalitozoon hellem) in seven patients with acquired immu- 9:975.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


nodeficiency syndrome. Am. J. Ophthalmol. 115:285–292. 332a.Sparfel, J. M., C. Sarfati, O. Liguory, B. Caroff, N. Dumoutier, B. Gueglio,
307. Schwartz, D. A., R. T. Bryan, and G. S. Visvesvara. 1994. Diagnostic ap- E. Billaud, F. Raffi, J. M. Molina, M. Miegeville, and F. Derouin. 1997.
proaches for Encephalitozoon infections in patients with AIDS. J. Eukaryot. Detection of microsporidia and identification of Enterocytozoon bieneusi in
Microbiol. 41:59S–60S. surface water by filtration followed by specific PCR. J. Eukaryot. Microbiol.
308. Schwartz, D. A., G. Visvesvara, R. Weber, and R. T. Bryan. 1994. Male 44:78S.
genital tract microsporidiosis and AIDS: prostatic abscess due to Enceph- 333. Sprague, V., and S. H. Vernick. 1971. The ultrastructure of Encephalitozoon
alitozoon hellem. J. Eukaryot. Microbiol. 41:61S. cuniculi (Microsporidia, Nosematidae) and ist taxonomic significance. J.
309. Schwartz, D. A., R. T. Bryan, R. Weber, and G. S. Visvesvara. 1994. Mi- Protozool. 18:560–569.
crosporidiosis in HIV positive patients: current methods for diagnosis using 334. Sprague, V. 1974. Nosema connori n. sp., a microsporidian parasite of man.
biopsy, cytologic, ultrastructural, immunological, and tissue culture tech- Trans. Am. Microsc. Soc. 93:400–403.
niques. Folia Parasitol. (Prague) 41:101–109. 335. Sprague, V., and J. Vávra. 1977. Systematics of the microsporidia, p. 1–510.
310. Schwartz, D. A., A. Abou Elella, C. M. Wilcox, L. Gorelkin, G. S. Visves- In L. A. Bulla, Jr., and T. C. Cheng (ed.), Comparative pathobiology, vol.
vara, S. E. Thompson, R. Weber, and R. T. Bryan. 1995. The presence of 2. Plenum Press, New York, N.Y.
Enterocytozoon bieneusi spores in the lamina propria of small bowel biopsies 336. Sprague, V. 1982. Microspora, p. 589–594. In S. P. Parker (ed.), Synopsis
with no evidence of disseminated microsporidiosis. Arch. Pathol. Lab. Med. and classification of living organisms, vol. 1. McGraw Hill Book Co., New
119:424–428. York, N.Y.
311. Schwartz, D. A., I. Sobottka, G. J. Leitch, A. Cali, and G. S. Visvesvara. 337. Sprague, V., J. J. Bencnel, and E. L. Hazard. 1992. Taxonomy of phylum
1996. Pathology of microsporidiosis: emerging parasitic infections in pa- microspora. Crit. Rev. Microbiol. 18:285–395.
tients with acquired immunodeficiency syndrome. Arch. Pathol. Lab. Med. 338. Sun, T., M. H. Kaplan, S. Teichberg, G. Weissman, T. Smilari, and C.
120:173–188. Urmacher. 1994. Intestinal microsporidiosis. Report of five cases. Ann.
312. Shadduck, J. A. 1969. Nosema cuniculi: In vitro isolation. Science 166:516– Clin. Lab. Sci. 24:521–532.
517. 339. Svenson, J., J. D. MacLean, E. Kokoskin-Nelson, J. Szabo, J. Lough, and
313. Shadduck, J. A., W. T. Watson, S. P. Pakes, and A. Cali. 1979. Animal M. J. Gill. 1993. Microsporidiosis in AIDS patients. Can. Commun. Dis.
infectivity of Encephalitozoon cuniculi. J. Parasitol. 65:123–129. Rep. 19:13–15.
314. Shadduck, J. A. 1980. Effect of fumagillin on in vitro multiplication of 339a.Talal, A. H., D. P. Kotler, J. M. Orenstein, and L. M. Weiss. 1998. Detec-
Encephalitozoon cuniculi. J. Protozool. 27:202–208. tion of Enterocytozoon bieneusi in fecal specimens by polymerase chain
315. Shadduck, J. A. 1989. Human microsporidiosis and AIDS. Rev. Infect. Dis. reaction analysis with primers to the small-subunit rRNA. Clin. Infect. Dis.
11:203–207. 26:673–675.
316. Shadduck, J. A., and E. Greeley. 1989. Microsporidia and human infections. 340. Terada, S., K. R. Reddy, L. J. Jeffers, A. Cali, and E. R. Schiff. 1987.
Clin. Microbiol. Rev. 2:158–165. Microsporidian hepatitis in the acquired immunodeficiency syndrome. Ann.
317. Shadduck, J. A., R. A. Meccoli, R. Davis, and R. L. Font. 1990. Isolation of Intern. Med. 107:61–62.
a microsporidian from a human patient. J. Infect. Dis. 162:773–776. 341. Torres, C. M. 1927. Sur une nouvelle maladie de l’homme, caracterisee par
318. Shadduck, J. A., and J. M. Orenstein. 1993. Comparative pathology of la presence d’un parasite intracellulaire, tres proche de Toxoplasma et de
microsporidiosis. Arch. Pathol. Lab. Med. 117:1215–1219. l’Encephalitozoon dans le tissu musculaire cardique, les muscles du
319. Shah, G. K., D. Pfister, L. E. Probst, P. Ferrieri, and E. Holland. 1996. squelette, le tissu celluaire sous-cutane et le tissu nereux. C. R. Seances Soc.
Diagnosis of microsporidial keratitis by confocal microscopy and the chro- Biol. Paris 97:1778–1781.
motrope stain. Am. J. Ophthalmol. 121:89–91. 342. Trammer, T., F. Dombrowski, M. Doehring, W. A. Maier, and H. M. Seitz.
320. Sharpstone, D., A. Rowbottom, M. Nelson, and B. Gazzard. 1995. The 1997. Opportunistic properties of Nosema algerae (Microspora), a mosquito
treatment of microsporidial diarrhoea with thalidomide. AIDS 9:658–659. parasite, in immunocompromised mice. J. Eukaryot. Microbiol. 44:258–262.
321. Sharpstone, D., A. Rowbottom, N. Francis, G. Tovey, D. Ellis, M. Barrett, 343. Tzipori, S., A. Carville, G. Widmer, D. Kotler, K. Mansfield, and A. Lack-
and B. Gazzard. Thalidomide: anovel therapy for microsporidiosis. Gastro- ner. 1997. Transmission and establishment of a persistent infection of
enterology 112:1823–1829. Enterocytozoon bieneusi, derived from a human with AIDS, in simian im-
322. Sheth, S. G., C. Bates, M. Federman, and S. Chopra. 1997. Fulminant munodeficiency virus-infected rhesus monkeys. J. Infect. Dis. 175:1016–
hepatic failure caused by microsporidial infection in a patient with AIDS. 1020.
AIDS 11:553–554. 344. Ullrich, R., M. Zeitz, C. Bergs, K. Janitschke, and E. O. Riecken. 1991.
323. Silveira, H., E. U. Canning, and J. A. Shadduck. 1993. Experimental infec- Intestinal microsporidiosis in a German patient with AIDS. Klin. Wochen-
tion of athymic mice with the human microsporidian Nosema corneum. schr. 69:443–445.
Parasitology 107:489–496. 345. Undeen, A. H., and N. E. Alger. 1976. Infection of the white mouse by a
324. Silveira, H., and E. U. Canning. 1995. Vittaforma corneae n. comb. for the mosquito parasite. Exp. Parasitol. 40:86–88.
human microsporidium Nosema corneum Shadduck, Meccoli, Davis & 346. Van den Bergh Weerman, M. A., T. Van Gool, J. K. Eeftinck Schattenkerk,
Font, 1990, based on its ultrastructure in the liver of experimentally infected and K. P. Dingemans. 1993. Electron microscopy as an essential technique
athymic mice. J. Eukaryot. Microbiol. 42:158–165. for the identification of parasites in AIDS patients. Eur. J. Morphol. 31:1–2.
325. Simon, D., L. M. Weiss, H. B. Tanowitz, A. Cali, J. Jones, and M. Wittner. 347. Van Gool, T., W. S. Hollister, W. E. Schattenkerk, M. A. van den Bergh
1991. Light microscopic diagnosis of human microsporidiosis and variable Weerman, W. J. Terpstra, R. J. van Ketel, P. Reiss, and E. U. Canning.
response to octreotide. Gastroenterology 100:271–273. 1990. Diagnosis of Enterocytozoon bieneusi microsporidiosis in AIDS pa-
326. Simon, D. 1994. Microsporidia in HIV diarrhea: present but pathogenic? tients by recovery of spores from faeces. Lancet 336:697–698.
Am. J. Gastroenterol. 89:636–638. 348. Van Gool, T., F. Snijders, P. Reiss, J. K. Eeftinck Schattenkerk, M. A. van
327. Singh, M., G. J. Kane, L. Mackinlay, I. Quaki, E. H. Yap, B. C. Ho, L. C. den Bergh Weerman, J. F. Bartelsman, J. J. Bruins, E. U. Canning, and J.
Ho, and K. C. Lim. 1982. Detection of antibodies to Nosema cuniculi Dankert. 1993. Diagnosis of intestinal and disseminated microsporidial
(Protozoa: Microscoporidia) in human and animal sera by the indirect infections in patients with HIV by a new rapid fluorescence technique.
fluorescent antibody technique. Southeast Asian J. Trop. Med. Public J. Clin. Pathol. 46:694–699.
Health 13:110–113. 349. Van Gool, T., E. U. Canning, and J. Dankert. 1994. An improved practical
284 FRANZEN AND MÜLLER CLIN. MICROBIOL. REV.

and sensitive technique for the detection of microsporidian spores in stool low up. Clin. Investig. 70:1019–1023.
samples. Trans. R. Soc. Trop. Med. Hyg. 88:189–190. 369. Weber, R., H. Kuster, G. S. Visvesvara, R. T. Bryan, D. A. Schwartz, and R.
350. Van Gool, T., E. U. Canning, H. Gilis, M. A. van den Bergh Weerman, J. K. Lüthy. 1993. Disseminated microsporidiosis due to Encephalitozoon hellem:
Eeftinck Schattenkerk, and J. Dankert. 1994. Septata intestinalis frequently pulmonary colonization, microhematuria, and mild conjunctivitis in a pa-
isolated from stool of AIDS patients with a new cultivation method. Para- tient with AIDS. Clin. Infect. Dis. 17:415–419.
sitology 109:281–289. 370. Weber, R., B. Sauer, R. Lüthy, and D. Nadal. 1993. Intestinal coinfection
351. Van Gool, T., E. Luderhoff, K. J. Nathoo, C. F. Kiire, J. Dankert, and P. R. with Enterocytozoon bieneusi and Cryptosporidium in a human immunode-
Mason. 1995. High prevalence of Enterocytozoon bieneusi infections among ficiency virus-infected child with chronic diarrhea. Clin. Infect. Dis. 17:480–
HIV-positive individuals with persistent diarrhoea in Harare, Zimbabwe. 483.
Trans. R. Soc. Trop. Med. Hyg. 89:478–480. 371. Weber, R., and R. T. Bryan. 1994. Microsporidial infections in immunode-
352. Van Gool, T., J. C. M. Vetter, A. P. Van Dam, G. A. C. Belling, H. Gilis, and ficient and immunocompetent patients. Clin. Infect. Dis. 19:517–521.
J. Dankert. 1995. Serological diagnosis of Septata intestinalis (Si) infections, 372. Weber, R., R. T. Bryan, D. A. Schwartz, and R. L. Owen. 1994. Human
abstr. D79, p. 80. In Program and Abstracts of the 35th Interscience Con- microsporidial infections. Clin. Microbiol. Rev. 7:426–461.
ference on Antimicrobial Agents and Chemotherapy. American Society for 373. Weber, R., B. Sauer, M. A. Spycher, P. Deplazes, R. Keller, R. Ammann, J.
Microbiology, Washington, D.C. Briner, and R. Lüthy. 1994. Detection of Septata intestinalis in stool spec-
353. Van Gool, T., J. C. M. Vetter, B. Weinmayr, A. Van Dam, F. Derouin, and imens and coprodiagnostic monitoring of successful treatment with al-
J. Dankert. 1997. High seroprevalence of Encephalitozoon species in im- bendazole. Clin. Infect. Dis. 19:342–345.
munocompetent subjects. J. Infect. Dis. 175:1020–1024. 374. Weber, R., P. Deplazes, M. Flepp, A. Mathis, R. Baumann, B. Sauer, H.
354. Vávra, J., P. Bedrnı́k, and J. Cintál. 1972. Isolation and in vitro cultivation Kuster, and R. Lüthy. 1997. Cerebral microsporidiosis due to Encephalito-
of the mammalian microsporidian Encephalitozoon cuniculi. Folia Parasitol. zoon cuniculi in a patient with human immunodeficient virus infection.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest


(Prague) 19:349–354. N. Engl. J. Med. 336:474–478.
355. Vávra, J., R. Dahbiová, W. S. Hollister, and E. U. Canning. 1993. Staining 375. Weber, R., A. Mathis, S. Zimmerli, and P. Deplazes. 1997. Epidemiology
of microsporidian spores by optical brighteners with remarks on the use of and clinical manifestations of HIV-associated microsporidiosis. Second
brighteners for the diagnosis of AIDS associated human microsporidioses. Workshop on Microsporidiosis and Cryptosporidiosis in Immunodeficient
Folia Parasitol. (Prague) 40:267–272. Patients.
356. Vávra, J., E. Nohynkova, L. Machala, and J. Spala. 1993. An extremely 376. Weiser, J. 1977. Contribution to the classification of microsporidia. Vestn.
rapid method for detection of microsporidia in biopsy materials from AIDS Cesk. Spol. Zool. 41:308–320.
patients. Folia Parasitol. (Prague) 40:273–274. 377. Weiser, J. 1993. Early experiences with microsporidia of man and mam-
356a.Vávra, J., A. T. Yachnis, J. A. Shadduck, and J. M. Orenstein. 1998. mals. Folia Parasitol. (Prague) 40:257–260.
Microsporidia of the genus Trachipleistophora—causative agents of human 378. Weiss, J. B. 1995. DNA probes and PCR for diagnosis of parasitic infec-
microsporidiosis: description of Trachipleistophora anthropophthera n. sp. tions. Clin. Microbiol. Rev. 8:113–130.
(Protozoa: Microsporidia). J. Eukaryot. Microbiol. 45:273–283. 379. Weiss, L. M., A. Cali, E. Levee, D. LaPlace, H. Tanowitz, D. Simon, and M.
357. Velasquez, J. N., S. Carnevale, E. A. Guarnera, J. H. Labbe, A. Chertcoff, Wittner. 1992. Diagnosis of Encephalitozoon cuniculi infection by western
M. G. Cabrera, and M. I. Rodriguez. 1996. Detection of the microsporidian blot and the use of cross-reactive antigens for the possible detection of
parasite Enterocytozoon bieneusi in specimens from patients with AIDS by microsporidiosis in humans. Am. J. Trop. Med. Hyg. 47:456–462.
PCR. J. Clin. Microbiol. 43:3230–3232. 380. Weiss, L. M., E. Michalakakis, C. M. Coyle, H. B. Tanowitz, and M.
358. Visvesvara, G. S., G. J. Leitch, H. Moura, S. Wallace, R. Weber, and R. T. Wittner. 1994. The in vitro activity of albendazole against Encephalitozoon
Bryan. 1991. Culture, electron microscopy, and immunoblot studies on a cuniculi. J. Eukaryot. Microbiol. 41:65S.
microsporidian parasite isolated from the urine of a patient with AIDS. J. 381. Weiss, L. M., X. Zhu, A. Cali, H. B. Tanowitz, and M. Wittner. 1994. Utility
Protozool. 38:105S–111S. of microsporidian rRNA in diagnosis and phylogeny: a review. Folia Para-
359. Visvesvara, G. S., G. J. Leitch, A. J. da Silva, G. P. Croppo, H. Moura, S. sitol. (Prague) 41:81–90.
Wallace, S. B. Slemenda, D. A. Schwartz, D. Moss, R. T. Bryan, and N. J. 381a.Weiss, L. M., and C. R. Vossbrinck. 1998. Microsporidiosis: molecular and
Pieniazek. 1994. Polyclonal and monoclonal antibody and PCR-amplified diagnostic aspects. Adv. Parasitol. 40:351–395.
small-subunit rRNA identification of a microsporidian, Encephalitozoon 382. Weiss, L. M. 1995. . . .and now microsporidiosis. Ann. Intern. Med. 123:
hellem, isolated from an AIDS patient with disseminated infection. J. Clin. 954–956.
Microbiol. 32:2760–2768. 383. WHO Parasitic Diseases Surveillance. 1983. Antibody to Encephalitozoon
360. Visvesvara, G. S., A. J. da Silva, G. P. Croppo, N. J. Pieniazek, G. J. Leitch, cuniculi in man. W. H. O. Weekly Epidemiol. Rec. 58:30–32.
D. Ferguson, H. de Moura, S. Wallace, S. B. Slemenda, I. Tyrrell, D. F. 384. Wicher, V., R. E. Baughn, C. Fuentealba, J. A. Shadduck, F. Abbruscato,
Moore, and J. Meador. 1995. In vitro culture and serologic and molecular and K. Wicher. 1991. Enteric infection with an obligate intracellular para-
identification of Septata intestinalis isolated from urine of a patient with site, Encephalitozoon cuniculi, in an experimental model. Infect. Immun.
AIDS. J. Clin. Microbiol. 33:930–936. 59:2225–2231.
361. Visvesvara, G., G. J. Leitch, N. J. Pieniazek, A. J. da Silva, S. Wallace, S. B. 385. Willson, R., R. Harrington, B. Stewart, and T. Fritsche. 1995. Human
Slemenda, R. Weber, D. A. Schwartz, L. Gorelkin, C. M. Wilcox, and R. T. immunodeficiency virus 1-associated necrotizing cholangitis caused by in-
Bryan. 1995. Short-term in vitro culture and molecular analysis of the fection with Septata intestinalis. Gastroenterology 108:247–251.
microsporidian, Enterocytozoon bieneusi. J. Eukaryot. Microbiol. 42:506– 386. Wongtavatchai, J., P. A. Conrad, and R. P. Hedrick. 1995. In vitro charac-
510. teristics of the microsporidian: Enterocytozoon salmonis. J. Eukaryot. Mi-
362. Vossbrinck, C. R., and C. R. Woese. 1986. Eukaryotic ribosomes that lack crobiol. 42:401–405.
a 5.8S RNA. Nature 320:287–288. 387. Woese, C. R. 1987. Bacterial evolution. Microbiol. Rev. 51:221–271.
363. Vossbrinck, C. R., J. V. Maddox, S. Friedman, B. A. Debrunner-Vossbrinck, 388. Wright, J. H., and E. M. Craighead. 1922. Infectious motor paralysis in
and C. R. Woese. 1987. Ribosomal RNA sequence suggests microsporidia young rabbits. J. Exp. Med. 36:135–140.
are extremely ancient eukaryotes. Nature 326:411–414. 389. Wuhib, T., T. M. J. Silva, R. D. Newman, L. S. Garcia, M. L. D. Pereira,
364. Vossbrinck, C. R., M. D. Baker, E. S. Didier, B. A. Debrunner Vossbrinck, C. S. Chaves, S. P. Wahlquist, R. T. Bryan, R. L. Guerrant, A. de Q. Sousa,
and J. A. Shadduck. 1993. Ribosomal DNA sequences of Encephalitozoon T. R. B. S. de Queiroz, and C. L. Sears. 1994. Cryptosporidial and micro-
hellem and Encephalitozoon cuniculi: species identification and phylogenetic sporidial infections in human immunodeficiency virus-infected patients in
construction. J. Eukaryot. Microbiol. 40:354–362. northeastern Brazil. J. Infect. Dis. 170:494–497.
365. Vossbrinck, C. R., M. D. Baker, and E. S. Didier. 1996. Comparative rRNA 390. Yachnis, A. T., J. Berg, A. Martinez-Salazar, B. S. Bender, L. Diaz, A. M.
analysis of microsporidia including AIDS related species. J. Eukaryot. Mi- Rojiani, T. A. Eskin, and J. M. Orenstein. 1996. Disseminated microspo-
crobiol. 43:110S. ridiosis especially infecting the brain, heart, and kidneys: report of a newly
365a.Wanke, C. A., P. DeGirolami, and M. Federman. 1996. Enterocytozoon recognized pansporoblastic species in two symptomatic AIDS patients.
bieneusi and diarrheal disease in patients who were not infected with human Am. J. Clin. Pathol. 106:535–543.
immunodeficiency virus: case report and review. Clin. Infect. Dis. 23:816– 391. Yee, R. W., F. O. Tio, J. A. Martinez, K. S. Held, J. A. Shadduck, and E. S.
818. Didier. 1991. Resolution of microsporidial epithelial keratopathy in a pa-
366. Weber, R., R. T. Bryan, R. L. Owen, C. M. Wilcox, L. Gorelkin, and G. S. tient with AIDS. Ophthalmology 98:196–201.
Visvesvara. 1992. Improved light-microscopical detection of microsporidia 392. Zender, H. O., E. Arrigoni, J. Eckert, and Y. Kapanci. 1989. A case of
spores in stool and duodenal aspirates. N. Engl. J. Med. 326:161–166. Encephalitozoon cuniculi peritonitis in a patient with AIDS. Am. J. Clin.
367. Weber, R., H. Kuster, R. Keller, T. Bachi, M. A. Spycher, J. Briner, E. Pathol. 92:352–356.
Russi, and R. Lüthy. 1992. Pulmonary and intestinal microsporidiosis in a 393. Zhu, X., M. Wittner, H. B. Tanowitz, A. Cali, and L. M. Weiss. 1993.
patient with the acquired immunodeficiency syndrome. Am. Rev. Respir. Nucleotide sequence of the small ribosomal RNA of Encephalitozoon cu-
Dis. 146:1603–1605. niculi. Nucleic Acids Res. 21:1315.
368. Weber, R., A. Müller, M. A. Spycher, M. Opravil, R. Ammann, and J. 394. Zhu, X., M. Wittner, H. B. Tanowitz, A. Cali, and L. M. Weiss. 1993.
Briner. 1992. Intestinal Enterocytozoon bieneusi microsporidiosis in an HIV- Nucleotide sequence of the small subunit rRNA of Septata intestinalis.
infected patient: diagnosis by ileo-colonoscopic biopsies and long-term fol- Nucleic Acids Res. 21:4846.
VOL. 12, 1999 MOLECULAR TECHNIQUES FOR ANALYSIS OF MICROSPORIDIA 285

395. Zhu, X., M. Wittner, H. B. Tanowitz, D. Kotler, A. Cali, and L. M. Weiss. and Ameson michaelis: phylogenetic construction and structural correspon-
1993. Small subunit rRNA sequence of Enterocytozoon bieneusi and its dence. J. Eukaryot. Microbiol. 41:204–209.
potential diagnostic role with use of the polymerase chain reaction. J. In- 397. Zierdt, C. H., V. J. Gill, and W. S. Zierdt. 1993. Detection of microsporidian
fect. Dis. 168:1570–1575. spores in clinical samples by indirect fluorescent-antibody assay using
396. Zhu, X., M. Wittner, H. B. Tanowitz, A. Cali, and L. M. Weiss. 1994. whole-cell antisera to Encephalitozoon cuniculi and Encephalitozoon hellem.
Ribosomal RNA sequences of Enterocytozoon bieneusi, Septata intestinalis J. Clin. Microbiol. 31:3071–3074.

Downloaded from http://cmr.asm.org/ on January 23, 2015 by guest

Das könnte Ihnen auch gefallen