Acessibilidade / Reportar erro

Current relevance of fungal and trypanosomatid glycolipids and sphingolipids: studies defining structures conspicuously absent in mammals

Abstracts

Recently, glycosphingolipids have been attracting attention due to their role on biological systems as second messengers or modulators of signal transduction, affecting several events, which range from apoptosis to regulation of the cell cycle. In pathogenic fungi, glycolipids are expressed in two classes: neutral monohexosylceramides (glucosyl-or galactosylceramide) and acidic glycosylinositol phosphorylceramides (the latter class carries longer glycan chains). It is worth to mention that monohexosylceramides exhibit significant structural differences in their lipid moieties compared to their mammalian counterparts, whereas the glycosylinositol phosphorylceramides exhibit remarkable structural differences in their carbohydrate moieties in comparison to mammal glycosphingolipids counterpart. We observed that glycosylinositol phosphorylceramides are capable of promoting immune response in infected humans. In addition, inhibiting fungal glycosphingolipid biosynthetic pathways leads to an inhibition of colony formation, spore germination, cell cycle, dimorphism and hyphal growth. Other pathogens, such as trypanosomatids, also present unique glycolipids, which may have an important role for the parasite development and/or disease establishment. Regarding host-pathogen interaction, cell membrane rafts, which are enriched in sphingolipids and sterols, participate in parasite/fungal infection. In this review, it is discussed the different biological roles of (glyco) (sphingo)lipids of pathogenic/opportunistic fungi and trypanosomatids.

glycosphingolipids; glycosylinositol phosphorylceramides; inositol phosphorylceramide; Leishmania; membrane rafts; pathogenic fungi


Recentemente, glicoesfingolipídeos têm atraído atenção devido ao seu papel na biologia celular como segundo-mensageiro ou moduladores da transdução de sinal, afetando vários eventos, desde apoptose até a regulação do ciclo celular. Em fungos patogênicos, existem duas classes de glicolipídeos: monohexosil ceramidas neutras (glucosil-ou galactosilceramida) e glicosilinositol fosforilceramidas (os quais apresentam cadeias de carboidratos mais longas). É importante enfatizar que as monohexosil ceramidas exibem diferenças estruturais nas suas porções lipídicas quando comparadas às de mamíferos, enquanto que glicosilinositol fosforilceramidas exibem diferenças estruturais marcantes em suas porções carboidratos em comparação aos glicoesfingolipídeos de mamíferos. Observamos também que glicosilinositol fosforilceramidas são capazes de promover resposta imune em indíviduos infectados. Além do mais, inibição das vias biossintéticas de glicoesfingolipídeos de fungos acarreta a inibição da formação de colônias, germinação de esporos, ciclo celular, dimorfismo e crescimento de hifas. Outros patógenos, como os tripanosomatídeos, também apresentam glicolipídeos únicos, os quais apresentam um papel importante para o desenvolvimento do parasita e/ou para o estabelecimento da doença. Em relação à interação hospedeiro-patógeno, os "membrane rafts", estruturas da membrana plasmática enriquecidas em esfingolipídeos e esteróis, têm participação fundamental na infecção do parasita/fungo. Nesta revisão, discutimos os diferentes papéis biológicos dos (glico) (esfingo)lipídeos de fungos patogênicos/oportunistas e de tripanosomatídeos.

glicoesfingolipídeos; glicosilinositol-fosforilceramidas; inositol fosforilceramidas; Leishmania; "membrane rafts"; fungos patogênicos


BIOMEDICAL AND MEDICAL SCIENCES

Current relevance of fungal and trypanosomatid glycolipids and sphingolipids: studies defining structures conspicuously absent in mammals

Helio K. TakahashiI; Marcos S. ToledoI; Erika SuzukiII; Loriane TagliariI; Anita H. StrausI

ISetor de Imunoquímica de Glicoconjugados, Departamento de Bioquímica Ed. J. L. Prado, Rua Botucatu, 862, 04023-900 São Paulo, SP, Brasil

IIDepartamento de Microbiologia, Imunologia e Parasitologia, Rua Botucatu, 862, 04023-900 São Paulo, SP, Brasil

Correspondence to Correspondence to: Dr. Anita H. Straus E-mail: straus.bioq@epm.br

ABSTRACT

Recently, glycosphingolipids have been attracting attention due to their role on biological systems as second messengers or modulators of signal transduction, affecting several events, which range from apoptosis to regulation of the cell cycle. In pathogenic fungi, glycolipids are expressed in two classes: neutral monohexosylceramides (glucosyl-or galactosylceramide) and acidic glycosylinositol phosphorylceramides (the latter class carries longer glycan chains). It is worth to mention that monohexosylceramides exhibit significant structural differences in their lipid moieties compared to their mammalian counterparts, whereas the glycosylinositol phosphorylceramides exhibit remarkable structural differences in their carbohydrate moieties in comparison to mammal glycosphingolipids counterpart. We observed that glycosylinositol phosphorylceramides are capable of promoting immune response in infected humans. In addition, inhibiting fungal glycosphingolipid biosynthetic pathways leads to an inhibition of colony formation, spore germination, cell cycle, dimorphism and hyphal growth. Other pathogens, such as trypanosomatids, also present unique glycolipids, which may have an important role for the parasite development and/or disease establishment. Regarding host-pathogen interaction, cell membrane rafts, which are enriched in sphingolipids and sterols, participate in parasite/fungal infection. In this review, it is discussed the different biological roles of (glyco) (sphingo)lipids of pathogenic/opportunistic fungi and trypanosomatids.

Key words: glycosphingolipids, glycosylinositol phosphorylceramides, inositol phosphorylceramide, Leishmania, membrane rafts, pathogenic fungi.

RESUMO

Recentemente, glicoesfingolipídeos têm atraído atenção devido ao seu papel na biologia celular como segundo-mensageiro ou moduladores da transdução de sinal, afetando vários eventos, desde apoptose até a regulação do ciclo celular. Em fungos patogênicos, existem duas classes de glicolipídeos: monohexosil ceramidas neutras (glucosil-ou galactosilceramida) e glicosilinositol fosforilceramidas (os quais apresentam cadeias de carboidratos mais longas). É importante enfatizar que as monohexosil ceramidas exibem diferenças estruturais nas suas porções lipídicas quando comparadas às de mamíferos, enquanto que glicosilinositol fosforilceramidas exibem diferenças estruturais marcantes em suas porções carboidratos em comparação aos glicoesfingolipídeos de mamíferos. Observamos também que glicosilinositol fosforilceramidas são capazes de promover resposta imune em indíviduos infectados. Além do mais, inibição das vias biossintéticas de glicoesfingolipídeos de fungos acarreta a inibição da formação de colônias, germinação de esporos, ciclo celular, dimorfismo e crescimento de hifas. Outros patógenos, como os tripanosomatídeos, também apresentam glicolipídeos únicos, os quais apresentam um papel importante para o desenvolvimento do parasita e/ou para o estabelecimento da doença. Em relação à interação hospedeiro-patógeno, os "membrane rafts", estruturas da membrana plasmática enriquecidas em esfingolipídeos e esteróis, têm participação fundamental na infecção do parasita/fungo. Nesta revisão, discutimos os diferentes papéis biológicos dos (glico) (esfingo)lipídeos de fungos patogênicos/oportunistas e de tripanosomatídeos.

Palavras-chave: glicoesfingolipídeos, glicosilinositol-fosforilceramidas, inositol fosforilceramidas, Leishmania, "membrane rafts", fungos patogênicos.

INTRODUCTION

This review is part of the celebration on the 75 years of Escola Paulista de Medicina/Universidade Federal de São Paulo. The development and establishment of research on different areas of the carbohydrate chemistry and biology in this University can be attributed to the great effort of Professor Carl P. Dietrich, who dedicated himself to fostering the growth of young scientists and the glycobiology field in Brazil. His pioneering contributions in mid 1970's on the structure and biological role of glycosaminoglycans and proteoglycans provided then a new concept of carbohydrate-to-carbohydrate interaction in eukaryotic cells that may define various cellular aspects, including oncogenic transformation and development (Dietrich et al. 1977, 1983, Dietrich 1984). However, it took close to 20 years, through the independent studies from several laboratories, to substantiate the functional significance of carbohydrate-to-carbohydrate interaction in the cell recognition and growth (Kojima and Hakomori 1991, Haseley et al. 2001, Matsuura and Kobayashi 2004). That concept, although greeted by considerable skepticism at the time, finds its place in the current notion on the importance of glycoconjugates in cellular function. A typical example is the key role of carbohydrate moieties of glycosphingolipid-enriched microdomains (GEMs) in cell-cell interaction and cellmatrix interaction (Hakomori 2000). In this context, this review is focused on structural studies of fungal and trypanosomatid glycosylinositol phosphorylceramides and neutral glycosphingolipids, as well as their functional roles in the fungus/parasite-host interaction.

GLYCOSPHINGOLIPIDS FROM PATHOGENIC FUNGI

In 1969, Steiner et al. described for the first time in Saccharomyces cerevisiae a glycosylated-inositol phosphorylceramide found in a fungus. Since then, especially in the last decades, finding new structures and defining the biosynthesis of these and other glycolipids from pathogenic or opportunistic fungi have become crucial due to the increasing mortality of immunosupressed patients associated with fungal infections. In several cases, death is a direct consequence of current antifungal therapy ineffectiveness (Odds et al. 2003, Moore et al. 2000), leading therefore for a compelling search for development of antifungal therapeutic agents.

In the last 15 years, our laboratory has purified and characterized over 20 glycosphingolipids (GSL) from several pathogenic and opportunistic fungi, such as Paracoccidioides brasiliensis, Histoplasma capsulatum, Sporothrix schenckii, Aspergillus fumigatus, Candida albicans, Cryptococcus neoformans, Cr. laurentii and Cr. albidus (Tables I and II). Our group and other investigators found two major classes of GSLs present in fungi, namely monohexosylceramides (CMHs) and glycosylinositol phosphorylceramides (GIPCs). CMH class comprises glucosylceramide (GlcCer) and galactosylceramide (GalCer), having (4E, 8E)-9-methyl-4, 8-sphingadienine as long chain base (d19:2) in combination with either N-2'-hydroxyoctadecanoate (h18:0) or N -2'-hydroxy- (E)-3'-octadecenoate (h18:1) [ (E3-unsaturated fatty acid]. Table I summarizes the expression of GlcCer and GalCer in several fungi, and it is noteworthy that CMHs from mycelium forms of P. brasiliensis and H. capsulatum present a higher percentage of unsaturated fatty acids (Toledo et al. 1999, 2001c). In addition, we previously proposed that activation/deactivation of a desaturase, responsible for the unsaturation modification of P. brasiliensis GlcCer, could represent a key step in a signaling cascade leading to transition from yeast to mycelium (or vice versa) that is initiatedby change in temperature (Toledo et al. 2001c). Moreover, while GlcCer was detected in all analyzed fungi, with exception of S. cerevisiae and Candida glabrata (Saito et al. 2006), GalCer was only detected in A. fumigatus and yeast forms of S. schenckii (Table I). Interestingly, for these two fungi, we observed a higher percentage of GalCer bearing unsaturated fatty acid compared to GlcCer, suggesting that galactosyltransferase poorly uses saturated form of ceramide as acceptor. On the other hand, glucosyltransferases may use equally well saturated and unsaturated ceramides as acceptors for GlcCer biosynthesis (Toledo et al. 1999) (Table I). Regarding only S. schenckii, results suggest that activation of GalCer-synthase enzyme follows the transition of mycelium to yeast, or on the contrary, suppression of this activity follows the transition of yeast to mycelium (Toledo et al. 2000), inferring that some fungi have the ability to use a wide range of ceramide and glycosyl structures in order to survive and proliferate in a variety of substrata and growth conditions, as well as in infectivity processes.

The other class of GSLs found in fungi comprises about 20 distinct GIPC structures, with the discovery rate increasing over the last 10 years. GIPCs are acidic glycosphingolipids, widely distributed among fungal species (Dickson and Lester 2002). These molecules present a ceramide (N -acylsphingosine) moiety linked to a glycan chain of variable length and structure through an inositol-phosphate linker. Compared to CMHs, GIPCs present a less variable ceramide portion, containing t18:0 and t20:0 4-hydroxysphinganines (phytosphingosines), which are only found in fungi, and an h24:0 fatty acid (Scheme 1, Product '1'). These results suggest a dichotomy at the biosynthesis level for GIPCs and CMHs (Leipelt et al. 2001).


At the monosaccharide level, three distinct cores were determined in fungal GIPCs: i) GlcNα1-2Ins, ii) Manα1-6Ins, and iii) Manα1-2Ins (Scheme 1, Products '3', '5' and '7', respectively). The differential expression of these glycan cores seems to be dependent on fungal species and, apparently, strongly regulated during morphogenesis (Barr et al. 1984, Barr and Lester 1984, Levery et al. 1998, Toledo et al. 2001a, b, 2007, Loureiro y Penha et al. 2001). While structure "iii" (Manα2IPC) is the most common core, found from S. cerevisiae to a wide range of mycophatogens, to date, only S. schenckii seems to express all three cores in its GIPC structures (Toledo et al. 2001a, b), as seen on Table II. In fact, several GIPCs present long glycan moieties, which are built up, for example, on core "iii" (Scheme 1, Product 7), as it wasdescribed for P. brasiliensis, H. capsulatum, S. schenckii, A. fumigatus, A. nidulans, C. albicans and Cr. neoformans. Structure details are shown on Table II, and Scheme 1, which present a biosynthetic tree for the GIPCs known to date in fungi. Furthermore, recently, more complex structures, built up on cores "i" (GlcNα1-2Ins - product '4', Scheme 1) and "iii" (Manα1-2Ins - products '11' to '17', Scheme 1) have been found in A. fumigatus (Toledo et al. 2007, Simenel et al. 2008). As a result, together, these glycoconjugates constitute an extensive structural diversity that provides several evidences to propose different fungal glycolipids biosynthetic pathways, which can be inhibited by specific drugs without affecting mammal biosynthetic pathways.

INHIBITION OF FUNGAL GLYCOSPHINGOLIPID BIOSYNTHETIC PATHWAYS

Since fungal GSLs have unique structures not found in mammals and also are implicated in many fundamental cellular processes in fungi, including growth, differentiation and morphogenesis (Barr et al. 1984, Barr and Lester 1984, Levery et al. 1998, 2002, Toledo et al. 2001a, b, 2007, Loureiro y Penha et al. 2001), blocking the biosynthesis of these GSLs has become a target for therapy development and understanding the fungal biology. Inhibitors for inositol phosphoceramide-synthase (IPC-synthase), for example, have been described in recent years. These enzymes catalyze the transfer of myo-inositol-1-phosphate from phosphatidylinositol to ceramide, resulting in synthesis of IPC (product '2', Scheme 1). Moreover, IPC-synthases are believed to be encoded by orthologous AUR1/IPC1 genes, which were found in S. cerevisiae, Schizosaccharomyces pombe, Candida spp., Aspergillus spp. and Cr. neoformans (Nagiec et al. 1997, Hashida-Okado et al. 1996, 1998, Kuroda et al. 1999, Heidler and Radding 1995, 2000). Aureobasidin-A, khafrefungin and rustmicin are examples of IPC-synthase inhibitors, which present potent and selective anti-fungal activities, and also low toxicity in mammals, making them possible candidates as antifungal drugs (Takesako et al. 1993, Mandala et al. 1997, 1998, Georgopapadakou 2000).

As shown by Levery et al. (2002), fungi expressing GlcCer are also highly susceptible to GlcCer-synthase inhibitors. D-threo-1-Phenyl -2-decanoylamino -3-morpholino -1-propanol (D-threo PDMP) (Radin et al. 1993) and its analogs, which have been widely applied for inhibition of mammalian GlcCer-synthase (GCS, UDP-Glc:ceramide β-glucosyltransferase) and are also capable to inhibit GlcCer-synthase in A. fumigatus and A. nidulans, affecting spore germination, cell cycle and hyphal growth. Furthermore, it is worth to mention that A. fumigatus expresses both GlcCer and GalCer and, therefore, theoretically, this fungus could prevent build up of free ceramide by utilizing it for synthesis of GalCer in the presence of D-threo-PDMP. However, it was observed that A. fumigatus susceptibility to this GlcCer-synthase inhibitor is similar to fungi expressing GlcCer only, suggesting that GlcCer performs other relevant functional roles in fungi which are not simply related to maintaining normal ceramide levels. Recently, we also observed that GlcCer-synthase inhibitors are able to strongly inhibit colony formation, spore germination, cell cycle and yeast to mycelium transformation in P. brasiliensis, H. capsulatum, S. schenckii and C. neoformans, clearly showing the importance of glucosylceramide on fungal biology. Therefore, it should be emphasized that these results indicate that the metabolic pathway of GlcCer synthesis represents a viable way to block fungal growth and differentiation, and since several structural aspects in fungal CMH lipid moiety differ of those from mammals (Levery et al. 2000), this glycolipid biosynthetic pathway should be explored with drugs that do not affect mammal GlcCer expression.

IMMUNE-RESPONSES TO FUNGAL GSLs

In addition to the importance of these glycolipids on the biology of the fungus, some of these glycoconjugates have shown to be highly antigenic. By HPTLC immunostaining, it was detected several GIPCs from H. capsulatum, P. brasiliensis and A. fumigatus reactive with sera of patients with histoplasmosis, paracoccidioidomycosis and aspergillosis (Barr and Lester 1984, Toledo et al. 1995, 2007, Bertini et al. 2007). By using immunochemical assays, it was observed that high antigenicity was due to presence of terminal galactofuranose residues in these glycolipids (Toledo et al. 1995, 2007, Bertini et al. 2007). Our group also demonstrated that GIPC Pb-3 (former Pb-1), which bears a terminal galactofuranose residue, is able to promote, in patients with paracoccidioidomycosis, a primary immune response with IgM production and subsequent switching to IgG1 (Bertini et al. 2007). We also observed that, after 5 months of antifungal treatment, there was a decline of symptoms and IgG1 titers, which makes Pb-3 a good marker for monitoring disease treatment. Since these antibodies may bind specifically to Fcγ receptors of phagocytes and as Pb-3 is expressed on yeast surface (Suzuki et al. 1997), IgG1 antibodies could play an important role in fungal clearance by promoting phagocytes to remove P. brasiliensis from the organism.

Regarding the cellular immune response, we analyzed the effect of purified preparations of glycosphingolipids from P. brasiliensis, A. fumigatus and S. schenckii on murine T lymphocytes. Four pure GIPCs (Pb-3, Pb-2, Af-4 and Ss-Y6) and two pure CMHs (GlcCer and GalCer) from different fungi were tested by measuring concanavalin A (Con A)-induced [3H]-thymidine uptake by normal BALB/c mouse lymphonode cells (Giorgio et al. 1992, 2003). Our analysis revealed that all four GIPCs inhibited T lymphocyte proliferation in vitro with IC50 values < 6µM. It was observed that the most efficient inhibitors were Pb-3, Pb-2, Af-4 and Ss-Y6, reaching 50% of inhibition with a concentration of ~ 5µM. On the other hand, neutral GSLs as CMH (GlcCer and GalCer) from fungi and from mammals reached 50% of inhibition in a concentration of ~ 20µM. These results clearly demonstrate that fungal glycolipids are able to inhibit T lymphocyte proliferation efficiently in a dose-dependent manner without inducing cell death, and also to elicit antibody production, suggesting that these GSLs may play an important role in modulating the host immune response.

FUNGI-HOST INTERACTION MECHANISMS: ROLE OF MEMBRANE RAFTS

For infection establishment in vertebrate host, some fungi also trigger host cell signaling pathways, which may lead to fungal invasion into host cells, host cell apoptosis or modulation of host immune response (see reviews Mendes-Giannini et al. 2005, Filler and Sheppard 2006, Netea et al. 2008). Belanger et al. (2002) and Monteiro da Silva et al. (2007), for example, showed that C. albicans and P. brasiliensis invasion was associated to activation of host tyrosine kinases activation. On the other hand, some events, such as apoptosis, may be activated or blocked depending on the species of fungi studied (Filler and Sheppard 2006, Mendes-Giannini et al. 2005). These final events may probably be due to the interaction between fungi and host molecules, such as extracellular matrix proteins, receptors, or even cell domains, such as membrane rafts (Maza et al. 2008, Netea et al. 2008, Filler and Sheppard 2006, Mendes-Giannini et al. 2005).

Recently, membrane rafts (also known as lipid rafts) were defined as small, cholesterol-and sphingolipid-enriched cell membrane domains (10-200 nm), which are highly dynamic, and where lipids and proteins move in and out, leading to cell signaling modulation events (Pike 2006). In addition, several membrane rafts present different lipid or protein compositions, leading consequently to distinct cell functions (Rajendran and Simons 2005, Zajchowski and Robbins 2002, Pike 2003).

Regarding pathogen-host relationship, in the last decade, several groups have demonstrated that bacteria, virus and protozoa, such as Listeria monocytogenes, Pseudomonas aeruginosa, HIV and Plasmodium falciparum, interact with mammal host cell through membrane rafts. This leads to infection in the host organism by manipulating cell signaling events like rearrangement of the cytoskeleton or even fusion of phagosomes with lysosomes (see review by Mañes et al. 2003). Concerning to fungi, by using several approaches, such as membrane raft disruption with methyl-β-cyclodextrin (mβCD) and localization in cell membrane of the ganglioside GM1, a membrane raft marker, we demonstrated for the first time that epithelial cell membrane rafts are essential for P. brasiliensis adhesion to host cells, and as well as for activation of host cell protein kinases, such as Src-family kinases (Maza et al. 2008). The role of these and other kinases activation in P. brasiliensis-epithelial cell interaction is currently under investigation in ourlaboratory.

Membrane rafts are also present in fungi. We observed that membrane domain fractions from H. capsulatum and P. brasiliensis showed an enrichment of GIPCs, CMH, ergosterol and proteins, such as Pma1p, a known yeast membrane raft marker. In addition, it was verified that disruption of fungal membrane rafts promoted an inhibition by 40% of H. capsulatum infection in alveolar macrophages, suggesting that proteins and lipids organization integrity of H. capsulatum membrane rafts is essential to yeast-host cell interaction. These results indicate that the integrity of membrane microdomains of H. capsulatum would be essential in the process of macrophage infectivity by this fungus. Besides, the cell membrane organization and the identification of the components of these microdomains will contribute to clarify the mechanisms involved in the adhesion and late invasion of macrophages by H. capsulatum (L. Tagliari et al. unpublished data).

LEISHMANIA (GLYCO) (SPHINGO)LIPIDS: BIOLOGICAL ROLE AND ORGANIZATION

Our group has also been working extensively in characterizing and studying the organization and biological role of (glyco) (sphingo)lipids of Leishmania spp., the ethiologic agent of leishmaniasis, which may present cutaneous lesions (localized or diffuse) or fatal visceral infection, depending on the Leishmania species. Leishmania comprises two forms in its life cycle: promastigotes, found in the midgut of sandflies, and amastigotes, the obligatory intracellular form that proliferate inside of an acidified vacuole of mammal macrophages. Amastigotes of L. (L.) amazonensis, species that causes human cutaneous leishmaniasis, present on the parasite surface several GSLs, which are recognized by three monoclonal antibodies (mAbs), termed ST-3, ST-4 and ST-5. These mAbs were able to inhibit up to 80% of macrophage invasion by amastigotes, suggesting that GSLs are essential for L. (L.) amazonensis binding to macrophages (Straus et al. 1993, 1997). Moreover, Galpβ1-3Galp disaccharide, which is part of one of the L. (L.) amazonensis amastigote GSLs, Galpβ1-3Galpα1-3Galpβ1-4Glcpβ1-1Cer (referred herein to as β-Gal-globotriaosylceramide), recognized by mAb ST-3, was also able to inhibit ~40% of macrophage invasion by these parasites, corroborating therefore the importance of these GSLs in L. (L.) amazonensis amastigote infection (Straus et al. 1993, 1997, Tanaka et al. 2007). Recently, a possible receptor for L. (L.) amazonensis amastigote GSLs was identified as being a 30 kDa protein from macrophage (Tanaka et al. 2007). Interestingly, this protein was not able to bind to Leishmania (Leishmania) chagasi amastigotes, which cause a visceral type of leishmaniasis and lack β-Gal-globotriaosylceramide, suggesting that GSLs of L. (L.) amazonensis amastigotes confer a particular specificity towards the host (Tanakaet al. 2007).

On the other hand, GSLs or glycoinositolphospholipid (GIPL) reactive with mAbs ST-3, ST-4 and ST-5 are absent in L. (L.) amazonensis promastigotes. Instead, these forms present other glycoconjugates, known as lipophosphoglycan (LPG) and proteophosphoglycan (PPG) which have epitopes recognized by mAbs ST-3, ST-4 and ST-5 (V. B. Valero et al., unpublished data). LPG and PPGs, along with glycosylphosphatidylinositol (GPI)-anchored proteins, and GIPLs are the major glycoconjugates found among several species of Leishmania (Descoteaux and Turco 1999, Guha-Niyogi et al. 2001, Handman 1999, Rittig and Bogdan 2000, Silveira 'et al. 2005, Suzuki et al. 2002, Spath et al. 2003). Recently, our group has demonstrated for L. (L.) amazonensis that GSLs and sterols of amastigotes, and GIPLs, sterols and IPC of promastigotes are preferentially distributed in membrane rafts (Fig. 1) (Suzuki et al. 2008). In a similar fashion, different species of Leishmania, L. (V.) braziliensis also presents membrane rafts enriched in sterol and IPC, in addition to phosphatidylinositol (PI) and phosphatidylethanolamine (PE) (Yoneyama et al. 2006). Treatment of L. (V.) braziliensis promastigotes with mβCD, a membrane raft disruptor, inhibited 50% of macrophage infection when compared to non-treated parasites, suggesting that Leishmania membrane rafts participate in this interaction.


One major sphingolipid found in promastigotes of Leishmania is IPC (Kaneshiro et al. 1986, McConville and Bacic 1989). Recently, we demonstrated that Aureobasidin-A (AbA), an IPC synthase inhibitor, was able to decrease growth rate of promastigotes of L. (L.) amazonensis, L. (L.) major and L. (V.) braziliensis, and to decrease dramatically the number of amastigotes of L. (L.) amazonensis per infected macrophage (Tanaka et al. 2007, Suzuki et al. 2008). In addition, AbA-treated promastigotes were viable, suggesting that IPC is important for cell division events. On the other hand, as IPC were not detected in L. (L.) amazonensis amastigotes by conventional methods, and since these forms were highly susceptible to this drug, one could speculate that IPC, even in very low amounts, could be a key compound for the parasite survival in the host cell.

As for fungi, glycolipids containing terminal galactofuranose (Gal f ) units are also present in L. (L.) major, and in other trypanosomatids, as Trypanosoma cruzi. In addition to being highly antigenic (patients with Chagas' disease present anti-Gal f antibodies) (Golgher et al. 1993), these glycolipids play an important role in parasite-host interaction. For instance, Fab fragments of mAb MEST-1, which recognizes terminal Gal f on GIPL-1 of L. (L.) major promastigotes, were able to inhibit the promastigotes and amastigotes invasion into murine macrophages (Suzuki et al. 2002). Moreover, for T. cruzi, Nogueira et al. (2007) demonstrated that Gal f -containing GIPCs are important for epimastigoteforms-midgut interaction with host cells, differentiation and proliferation, and Bertini el al. (2003) showed that trypomastigotes Gal f -containing glycoproteins are important for Vero cells invasion by this form of T. cruzi.

Therefore, as discussed in this review, (glyco)-(sphingo)lipids, present in trypanosomatids and in pathogenic/opportunistic fungi, may be crucial for their interaction with host cells, differentiation, proliferation and host immune response. Understanding the mechanisms involved in these events, such as membrane raft formation, since its initial assembly at Golgi complex and endoplasmic reticulum, dynamic organization and glycolipid biosynthetic pathways may contribute to open new vistas leading to the discovery of new and more efficien ttherapies.

CONCLUDING REMARKS

More than 20 glycosylinositol phosphorylceramides with new glycan sequences, as well as monohexosyl-ceramides with unique modifications in the ceramide moiety, were described in pathogenic/opportunistic fungi in the last 10 years by our laboratory and other groups. The remarkable structural diversity of GIPCs observed in fungi is built up from three different cores: GlcNα1-2Ins, Manα1-2Ins and Manα1-6Ins. Since carbohydrate determinants are synthesized in stepwise fashion in which each monosaccharide is transferred from a sugar nucleotide donor to the acceptor molecule (Tolmasky et al. 1982), it will be possible to assign glycosyltransferases not being present in mammals as potential targets for more efficient therapies. In a similar fashion, since several structural aspects of the lipid moiety of fungal CMH differ significantly of those from mammals (Levery et al. 2000), this glycolipid biosynthetic pathway is an attractive target to be explored aiming the development of new drugs that will block fungal growth, but will not affect mammalian GlcCer expression. It is worth to mention that, similarly to mammal cells, fungal GIPCs and CMHs are not scattered in the plasma membrane but preferentially located in the glycosphingolipid enriched membrane microdomains (GEMs).

We also described that glycolipids play an important role in macrophage invasion by different species of Leishmania. These glycolipids are located in membrane rafts and disruption of such membrane microdomains with methyl-β-cyclodextrin reduces significantly parasite infectivity. The importance of IPC-containing glycolipids on L. (L.) amazonensis, L. (L.) major and L. (V.) braziliensis assessed using AbA promoted a significant decrease in the parasite growth and a striking effect in the macrophage infection by L. (L.) amazonensis.

Based on the present results, it is possible to conclude that fungal and parasite membrane microdomains preferentially accumulate specific glyco (sphingo)lipids and other proteins/glycoproteins involved in adhesion, signal transduction, modulation of cellular and humoral responses of the host and other vital activities. Thus, a detailed knowledge on the structure of fungal and parasite glycolipids may provide ground to develop more specific and sensitive reagents/drugs for immunodiagnosis and treatment of systemic mycosis and leishmaniasis.

ACKNOWLEDGMENTS

This work was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Fundo de Auxílio aos Docentes e Alunos da Universidade Federal de São Paulo (FADA-UNIFESP).

Manuscript received on July 31 2008; accepted for publication on February 20, 2009; presented by LUIZ R. TRAVASSOS

In commemoration of the 75th anniversary of Escola Paulista de Medicina/ Universidade Federal de São Paulo. Dedicated to the memory of Prof. Carl P. Dietrich.

  • AOKI K, UCHIYAMA R, ITONORI S, SUGITA S, CHE FS, ISOGAI A, HADA N, TAKEDA T, KUMAGAI H AND YAMAMOTO K. 2004. Structural elucidation of novel phosphocholine-containing glycosylinositol-phosphoceramide in filamentous fungi and their induction of cell death of cultured rice cells. Biochem J 378: 461-472.
  • BARR K AND LESTER RL. 1984. Occurrence of novel antigenic phosphoinositol-containing sphingolipids in the pathogenic yeast Histoplasma capsulatum Biochemistry 23: 5581-5588.
  • BARR K, LAINE RA AND LESTER RL. 1984. Carbohydrate structures of three novel phosphoinositol-containing sphingolipids from the yeast Histoplasma capsulatum Biochemistry 23: 5589-5596.
  • BELANGER PH, JOHNSTON DA, FRATTI RA, ZHANG M AND FILLER SG. 2002. Endocytosis of Candida albicans by vascular endothelial cells is associated with tyrosine phosphorylation of specific host cell proteins. Cell Microbiol 4: 805-812.
  • BENNION B, PARK C, FULLER M, LINDSEY R, MOMANY M, JENNEMANN R AND LEVERY SB. 2003. Glycosphingolipids of the model fungus Aspergillus nidulans: Characterization of GIPCs with oligo-α-mannose-type glycans. J Lipid Res 44: 2073-2088.
  • BERTINI S, TAKAHASHI HK AND STRAUS AH. 2003. Inhibition of trypomastigotes infectivity by monoclonal antibody BST-1, directed to epimastigotes GIPCs. Cross-reactivity with high molecular weight antigen presentin trypomastigotes. Rev Inst Med Trop S Paulo 45: 83.
  • BERTINI S, COLOMBO AL, TAKAHASHI HK AND STRAUSAH. 2007. Expression of antibodies directed to Paracoccidioides brasiliensis glycosphingolipids during the course of paracoccidioidomycosis treatment. Clin Vaccine Immunol 14: 150-156.
  • CARDOSO DBS, ANGLUSTER J, TRAVASSOS LR AND ALVIANO CS. 1987. Isolation and characterization of a glucocerebroside (monoglucosylceramide) from Sporothrix schenckii FEMS Microbiol Lett 43: 279-282.
  • DESCOTEAUX A AND TURCO SJ. 1999. Glycoconjugates in Leishmania infectivity. Biochim Biophys Acta 1455: 341-352.
  • DICKSON RC AND LESTER RL. 2002. Sphingolipid functions in Saccharomyces cerevisiae Biochim Biophys Acta 1583: 13-25.
  • DIETRICH CP. 1984. A model for cell-cell recognition and control of cell growth mediated by sulfated glycosaminoglycans. Braz J Med Biol Res 17: 5-15.
  • DIETRICH CP, SAMPAIO LO, TOLEDO OMS AND CÁSSARO CMF. 1977. Cell recognition and adhesiveness. A possible biological role for the sulfated mucopolysaccharides. Biochem Biophys Res Commun 75: 329-336.
  • DIETRICH CP, NADER HB AND STRAUS AH. 1983. Structural differences of heparan sulfates according to the tissue and species of origin. Biochem Biophys Res Commun 111: 865-871.
  • FILLER SG AND SHEPPARD DC. 2006. Fungal invasion of normally non-phagocytic host cells. PLoS Pathog 2: e129.
  • GEORGOPAPADAKOU NH. 2000. Antifungals targeted to sphingolipid synthesis: Focus on inositol phosphorylceramide synthase. Expert Opin Investig Drugs 9: 1787-1796.
  • GIORGIO S, JASIULIONIS MG, STRAUS AH, TAKAHASHI HK AND BARBIERI CL. 1992. Inhibition of mouse lymphocyte proliferative response by glycosphingolipids from Leishmania (L.) amazonensis Exp Parasitol 75: 119-125.
  • GIORGIO S, SANTOS MR, STRAUS AH, TAKAHASHI HK AND BARBIERI CL. 2003. Effect of glycosphingolipids purified from Leishmania (Leishmania) amazonensis amastigotes on human peripheral lymphocytes. Clin Diagn Lab Immunol 10: 469-472.
  • GOLGHER DB, COLLI W, SOUTO-PADRÓN T AND ZINGALES B. 1993. Galactofuranose-containing glycoconjugates of epimastigote and trypomastigote forms of Trypanosoma cruzi Mol Biochem Parasitol 60: 249-264.
  • GUHA-NIYOGI A, SULLIVAN DR AND TURCO SJ. 2001. Glycoconjugate structures of parasitic protozoa. Glycobiology 11: 45-59.
  • HAKOMORI S. 2000. Cell adhesion/recognition and signaltransduction through glycosphingolipid microdomain. Glycoconj J 17: 143-151.
  • HANDMAN E. 1999. Cell biology of Leishmania Adv Parasitol 44: 1-39.
  • HASELEY SR, VERMEER HJ, KAMERLING JP AND VLIEGENTHART JF. 2001. Carbohydrate self-recognition mediates marine sponge cellular adhesion. Proc Natl Acad Sci USA 98: 9419-9424.
  • HASHIDA-OKADO T, OGAWA A, ENDO M, YASUMOTO R, TAKESAKO K AND KATO I. 1996. AUR1, a novel gene conferring aureobasidin resistance on Saccharomyces cerevisiae: a study of defective morphologies in Aur1p-depleted cells. Mol Gen Genet 251: 236-244.
  • HASHIDA-OKADO T, YASUMOTO R, ENDO M, TAKESAKO K AND KATO I. 1998. Isolation and characterization of the aureobasidin A-resistant gene, aur1R, on Schizosaccharomyces pombe: roles of Aur1p+ in cell morphogenesis. Curr Genet 33: 38-45.
  • HEIDLER SA AND RADDING JA. 1995. The AUR1 gene in Saccharomyces cerevisiae encodes a dominant resistance to the antifungal agent Aureobasidin A (LY295337). Antimicrob Agents Chemother 39: 2765-2769.
  • HEIDLER SA AND RADDING JA. 2000. Inositol phosphoryl transferases from human pathogenic fungi. Biochim Biophys Acta 1500: 147-152.
  • HEISE N, GUTIERREZ ALS, MATTOS KA, JONES C, WAIT R, PREVIATO JO AND MENDONÇA-PREVIATO L. 2002. Molecular analysis of a novel family of complex glycoinositolphosphoryl ceramides from Cryptococcus neoformans: Structural differences between encapsulated and acapsular yeast forms. Glycobiology 12: 409-420.
  • KANESHIRO ES, JAYASIMHULU K AND LESTER RL. 1986. Characterization of inositol lipids from Leishmania donovani promastigotes: identification of an inositol sphingophospholipid. J Lipid Res 27: 1294-1303.
  • KOJIMA N AND HAKOMORI S. 1991. Cell adhesion, spreading, and motility of GM3-expressing cells based on glycolipid-glycolipid interaction. J Biol Chem 266: 17552- 17558.
  • KURODA M, HASHIDA-OKADO T, YASUMOTO R, GOMI K, KATO I AND TAKESAKO K. 1999. An aureobasidin A resistance gene isolated from Aspergillus is a homolog of yeast AUR1, a gene responsible for inositol phosphorylceramide (IPC) synthase activity. Mol Gen Genet 261:290-296.
  • LEIPELT M, WARNECKE D, ZAHRINGER U, OTT C, MULLER F, HUBE B AND HEINZ E. 2001. Glucosylceramide synthases, a gene family responsible for the biosynthesis of glucosphingolipids in animals, plants, and fungi. J Biol Chem 276: 33621-33629.
  • LEVERY SB, TOLEDO MS, STRAUS AH AND TAKAHASHI HK. 1998. Structure elucidation of sphingolipids from the mycopathogen Paracoccidioides brasiliensis: An immunodominant β-galactofuranose residue is carried by a novel glycosylinositol phosphorylceramide antigen. Biochemistry 37: 8764-8775.
  • LEVERY SB, TOLEDO MS, DOONG RL, STRAUS AH AND TAKAHASHI HK. 2000. Comparative analysis of ceramide structural modification found in fungal cerebrosides by electrospray tandem mass spectrometry with low energy collision-induced dissociation of Li+ adduct ions. Rapid Commun Mass Spectrom 14: 551-563.
  • LEVERY SB, MOMANY M, LINDSEY R, TOLEDO MS, SHAYMAN JA, FULLER M, BROOKS K, DOONG RL, STRAUS AH AND TAKAHASHI HK. 2002. Disruption of the glucosylceramide biosynthesis pathway in Aspergillus nidulans and Aspergillus fumigatus by inhibitors of UDP-Glc: ceramide glucosyltransferase strongly affects spore germination, cell cycle, and hyphal growth. FEBS Lett 525: 59-64.
  • LOUREIRO Y PENHA CV, TODESCHINI AR, LOPES-BEZERRA LM, WAIT R, JONES C, MATTOS KA, HEISE N, MENDONÇA-PREVIATO L AND PREVIATO JO. 2001. Characterization of novel structures of mannosylinositol phosphorylceramides from the yeast forms of Sporothrix schenckii Eur J Biochem 268: 4243-4250.
  • MANDALA SM, THORNTON RA, ROSENBACH M, MILLIGAN J, GARCIA-CALVO M, BULL HG AND KURTZ MB. 1997. Khafrefungin, a novel inhibitor of sphingolipid synthesis. J Biol Chem 272: 32709-32714.
  • MANDALA SM ET AL. 1998. Rustmicin, a potent antifungal agent, inhibits sphingolipid synthesis at the inositol phosphoceramide synthase. J Biol Chem 273: 14942-14949.
  • MAÑES S, DEL REAL G AND MARTÍNEZ-A C. 2003. Pathogens: raft hijackers. Nat Rev Immunol 3: 557-568.
  • MATSUURA K AND KOBAYASHI K. 2004. Analysis of GM3-Gg3 interaction using clustered gycoconjugate models constructed from glycolipid monolayers and artificial glycoconjugate polymers. Glycoconj J 21: 139-148.
  • MAZA PK, STRAUS AH, TOLEDO MS, TAKAHASHI HK AND SUZUKI E. 2008. Interaction of epithelial cell membrane rafts with Paracoccidioides brasiliensis leads to fungal adhesion and Src-family kinase activation. Microbes Infect 10: 540-547.
  • MCCONVILLE MJ AND BACIC A. 1989. A family of glycoinositol phospholipids from Leishmania major, Isolation, characterization, and antigenicity. J Biol Chem 264: 757-766.
  • MENDES-GIANNINI MJ, SOARES CP, DA SILVA JL AND ANDREOTTI PF. 2005 Interaction of pathogenic fungi with host cells: Molecular and cellular approaches. FEMS Immunol Med Microbiol 45: 383-394.
  • MONTEIRO DA SILVA JL, ANDREOTTI PF, BENARD G, SOARES CP, MIRANDA ET AND MENDES-GIANNINI MJ. 2007. Epithelial cells treated with genistein inhibit adhesion and endocytosis of Paracoccidioides brasiliensis Antonie Van Leeuwenhoek 92: 129-135.
  • MOORE CB, SAYERS N, MOSQUERA J, SLAVEN J AND DENNING DW. 2000. Antifungal drug resistance in Aspergillus J Infect 41: 203-220.
  • NAGIEC MM, NAGIEC EE, BALTISBERGER JA, WELLS GB, LESTER RL AND DICKSON RC. 1997. Sphingolipid synthesis as a target for antifungal drugs. Complementation of the inositol phosphorylceramide synthase defect in a mutant strain of Saccharomyces cerevisiae by the AUR1 gene. J Biol Chem 272: 9809-9817.
  • NETEA MG, BROWN GD, KULLBERG BJ AND GOW NA. 2008. An integrated model of the recognition of Candida albicans by the innate immune system. Nat Rev Microbiol 6: 67-78.
  • NOGUEIRA NFS, GONZALEZ MS, GOMES JE, SOUZA W, GARCIA ES, AZAMBUJA P, NOHARA LL, ALMEIDA IC, ZINGALES B AND COLLI W. 2007. Trypanosoma cruzi: Involvement of glycoinositolphospholipids in the attachment to the luminal midgut surface of Rhodnius prolixus Exp Parasitol 116: 120-128.
  • ODDS FC, BROWN AJP AND GOW NAR. 2003. Antifungal agents: mechanisms of action. Trends Microbiol 11: 272- 279.
  • PIKE LJ. 2003. Lipid rafts: bringing order to chaos. J Lipid Res 44: 655-667.
  • PIKE LJ. 2006. Rafts defined: a report on the Keystone Symposium on Lipid Rafts and Cell Function. J Lipid Res 47: 1597-1598.
  • RADIN NS, SHAYMAN JA AND INOKUCHI J. 1993. Metabolic effects of inhibiting glucosylceramide synthesis with PDMP and other substances. Adv Lipid Res 26: 183-213.
  • RAJENDRAN L AND SIMONS K. 2005. Lipid rafts and membrane dynamics. J Cell Sci 118: 1099-1102.
  • RITTIG MG AND BOGDAN C. 2000. Leishmania-host-cell interaction: complexities and alternative views. Parasitol Today 16: 292-297.
  • SAITO K, TAKAKUWA N, OHNISHI M AND ODA Y. 2006. Presence of glucosylceramide in yeast and its relation to alkali tolerance of yeast. Appl Microbiol Biotechnol 71: 515-521.
  • SILVEIRA TGV, YONEYAMA KAG, TAKAHASHI HK AND STRAUS AH. 2005. Isolation of Leishmania (Viannia) braziliensis glycolipid antigens and their reactivity with mAb SST-1, specific for parasites of Viannia subgenus. Parasitology 131: 737-745.
  • SIMENEL C, CODDEVILLE B, DELEPIERRE M, LATGÉ JP AND FONTAINE T. 2008. Glycosylinositolphosphoceramides in Aspergillus fumigatus Glycobiology 18: 84-96.
  • SPATH GF, GARRAWAY LA, TURCO SJ AND BEVERLEY SM. 2003. The role (s) of lipophosphoglycan (LPG) in the establishment of Leishmania major infections in mammalian hosts. Proc Natl Acad Sci USA 100: 9536-9541.
  • STEINER S, SMITH S, WAECHTER CJ AND LESTER RL. 1969. Isolation and partial characterization of a major inositol-containing lipid in Baker's yeast, mannosyl-diinositol, di-phosphoryl-ceramide. Proc Natl Acad SciUSA 64: 1042-1048.
  • STRAUS AH, LEVERY SB, JASIULIONIS MG, SALYAN ME, STEELE SJ, TRAVASSOS LR, HAKOMORI S AND TAKAHASHI HK. 1993. Stage-specific glycosphingolipids from amastigote forms of Leishmania (L.) amazonensis Immunogenicity and role in parasite binding and invasion of macrophages. J Biol Chem 268: 13723-13730.
  • STRAUS AH, VALERO VB, TAKIZAWA CM, LEVERY SB, TOLEDO MS, SUZUKI E, SALYAN ME, HAKOMORI S, BARBIERI CL AND TAKAHASHI HK. 1997. Glycosphingolipid antigens from Leishmania (L.) amazonensis amastigotes. Binding of anti-glycosphingolipid monoclonal antibodies in vitro and in vivo Braz J Med Biol Res 30: 395-399.
  • SUZUKI E, TOLEDO MS, TAKAHASHI HK AND STRAUS AH. 1997. A monoclonal antibody directed to terminal residue of β-galactofuranose of a glycolipid antigen isolated from Paracoccidioides brasiliensis: cross-reactivity with Leishmania major and Trypanosoma cruzi Glycobiology 7: 463-468.
  • SUZUKI E, TANAKA AK, TOLEDO MS, TAKAHASHI HK AND STRAUS AH. 2002. Role of β-d-galactofuranose in Leishmania major macrophage invasion. Infect Immun 70: 6592-6596.
  • SUZUKI E, TANAKA AK, TOLEDO MS, LEVERY SB, TAKAHASHI HK AND STRAUS AH. 2008. Trypanosomatidand fungal glycolipids and sphingolipids as infectivity factors and potential targets for development of new therapeutic strategies. Biochim Biophys Acta 1780: 362-369.
  • TAKESAKO K, KURODA H, INOUE T, HARUNA F, YOSHIKAWA Y AND KATO I. 1993. Biological properties of Aureobasidin A, a cyclic depsipeptide antifungal antibiotic. J Antibiot 49: 1414-1420.
  • TANAKA AK, GORIN PAJ, TAKAHASHI HK AND STRAUS AH. 2007. Role of Leishmania (Leishmania) amazonensis amastigote glycosphingolipids in macrophage infectivity. Braz J Med Biol Res 40: 799-806.
  • TOLEDO MS, SUZUKI E, STRAUS AH AND TAKAHASHI HK. 1995. Glycolipids from Paracoccidioides brasiliensis Isolation of a galactofuranose-containing glycolipid reactive with sera of patients with paracoccidioidomycosis. J Med Vet Mycol 33: 247-251.
  • TOLEDO MS, LEVERY SB, STRAUS AH, SUZUKI E, MOMANY M, GLUSHKA J, MOULTON JM AND TAKAHASHI HK. 1999. Characterization of sphingolipids from mycopathogens: Factors correlating with expression of 2-hydroxy fatty acyl (E)-Δ3-unsaturation in cerebrosides of Paracoccidioides brasiliensis and Aspergillus fumigatus Biochemistry 38: 7294-7306.
  • TOLEDO MS, LEVERY SB, STRAUS AH AND TAKAHASHI HK. 2000. Dimorphic expression of cerebrosides in the mycopathogen Sporothrix schenckii J Lipid Res 41: 797-806.
  • TOLEDO MS, LEVERY SB, STRAUS AH AND TAKAHASHI HK. 2001a. Sphingolipids of the mycopathogen Sporothrix schenckii: Identification of a glycosylinositol phosphorylceramide novel core GlcNH2α
  • TOLEDO MS, LEVERY SB, GLUSHKA J, STRAUS AH AND TAKAHASHI HK. 2001b. Structure elucidation of sphingolipids from the mycopathogen Sporothrix schenckii: Identification of novel glycosylinositol phosphorylceramides with core Manα
  • TOLEDO MS, LEVERY SB, SUZUKI E, STRAUS AH AND TAKAHASHI HK. 2001c. Characterization of cerebrosides from the thermally dimorphic mycopathogen Histoplasma capsulatum: expression of 2-hydroxy fatty N-acyl (E)-Delta (3)-unsaturation correlates with the yeast-mycelium phase transition. Glycobiology 11: 113-124.
  • TOLEDO MS, LEVERY SB, BENNION B, GUIMARÃES LL, CASTLE SA, LINDSEY R, MOMANY M, PARK C, STRAUS AH AND TAKAHASHI HK. 2007. Analysis of glycosylinositol phosphorylceramides expressed by the opportunistic mycopathogen Aspergillus fumigatus J Lipid Res 48: 1801-1824.
  • TOLMASKY ME, TAKAHASHI HK, STANELONI RJ AND LELOIR LF. 1982. N-glycosylation of proteins in plants. An Asoc Quim Argent 70: 405-411.
  • VILLAS-BOAS MH, EGGE H, POHLENTZ G, HARTMANN R AND BERGTER EB. 1994. Structural determination of N-2'-hydroxyoctadecenoyl-1-O-beta-D-glucopyranosyl-9-methyl-4, 8-sphingadienine from species of Aspergillus. Chem Phys Lipids 70: 11-19.
  • YONEYAMA KAG, TANAKA AK, SILVEIRA TGV, TAKAHASHI HK AND STRAUS AH. 2006. Characterization of Leishmania (Viannia) braziliensis membrane micro-domains, and their role in macrophage infectivity. J Lipid Res 47: 2171-2178.
  • ZAJCHOWSKI LD AND ROBBINS SM. 2002. Lipid rafts and little caves. Compartmentalized signaling in membrane microdomains. Eur J Biochem 269: 737-752.
  • Correspondence to:

    Dr. Anita H. Straus
    E-mail:
  • Publication Dates

    • Publication in this collection
      20 Aug 2009
    • Date of issue
      Sept 2009

    History

    • Accepted
      20 Feb 2009
    • Received
      31 July 2008
    Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
    E-mail: aabc@abc.org.br