Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Biomacromolecules ; 25(9): 5670-5701, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39177507

RESUMO

Fungal pathogens cause over 6.5 million life-threatening systemic infections annually, with mortality rates ranging from 20 to 95%, even with medical intervention. The World Health Organization has recently emphasized the urgent need for new antifungal drugs. However, the range of effective antifungal agents remains limited and resistance is increasing. This Review explores the current landscape of fungal infections and antifungal drugs, focusing on synthetic polymeric nanomaterials like nanoparticles that enhance the physicochemical properties of existing drugs. Additionally, we examine intrinsically antifungal polymers that mimic naturally occurring peptides. Advances in polymer characterization and synthesis now allow precise design and screening for antifungal activity, biocompatibility, and drug interactions. These antifungal polymers represent a promising new class of drugs for combating fungal infections.


Assuntos
Antifúngicos , Micoses , Nanoestruturas , Polímeros , Antifúngicos/farmacologia , Antifúngicos/química , Micoses/tratamento farmacológico , Humanos , Polímeros/química , Polímeros/farmacologia , Nanoestruturas/química , Fungos/efeitos dos fármacos , Nanopartículas/química , Animais
2.
Biomacromolecules ; 25(2): 871-889, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38165721

RESUMO

Invasive fungal infections impose a substantial global health burden. They cause more than 1.5 million deaths annually and are insufficiently met by the currently approved antifungal drugs. Antifungal peptides are a promising alternative to existing antifungal drugs; however, they can be challenging to synthesize, and are often susceptible to proteases in vivo. Synthetic polymers which mimic the properties of natural antifungal peptides can circumvent these limitations. In this study, we developed a library of 29 amphiphilic polyacrylamides with different charged units, namely, amines, guanidinium, imidazole, and carboxylic acid groups, representative of the natural amino acids lysine, arginine, histidine, and glutamic acid. Ternary polymers incorporating primary ammonium (lysine-like) or imidazole (histidine-like) groups demonstrated superior activity against Candida albicans and biocompatibility with mammalian cells compared to the polymers containing the other charged groups. Furthermore, a combination of primary ammonium, imidazole, and guanidinium (arginine-like) within the same polymer outperformed the antifungal drug amphotericin B in terms of therapeutic index and exhibited fast C. albicans-killing activity. The most promising polymer compositions showed synergistic effects in combination with caspofungin and fluconazole against C. albicans and additionally demonstrated activity against other clinically relevant fungi. Collectively, these results indicate the strong potential of these easily producible polymers to be used as antifungals.


Assuntos
Compostos de Amônio , Antifúngicos , Animais , Antifúngicos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Polímeros/farmacologia , Histidina , Guanidina/farmacologia , Lisina , Candida albicans , Imidazóis/farmacologia , Arginina/farmacologia , Testes de Sensibilidade Microbiana , Mamíferos
3.
J Cell Sci ; 134(24)2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34792152

RESUMO

Echinocandins such as caspofungin are frontline antifungal drugs that compromise ß-1,3 glucan synthesis in the cell wall. Recent reports have shown that fungal cells can resist killing by caspofungin by upregulation of chitin synthesis, thereby sustaining cell wall integrity (CWI). When echinocandins are removed, the chitin content of cells quickly returns to basal levels, suggesting that there is a fitness cost associated with having elevated levels of chitin in the cell wall. We show here that simultaneous activation of the calcineurin and CWI pathways generates a subpopulation of Candida albicans yeast cells that have supra-normal chitin levels interspersed throughout the inner and outer cell wall, and that these cells are non-viable, perhaps due to loss of wall elasticity required for cell expansion and growth. Mutations in the Ca2+-calcineurin pathway prevented the formation of these non-viable supra-high chitin cells by negatively regulating chitin synthesis driven by the CWI pathway. The Ca2+-calcineurin pathway may therefore act as an attenuator that prevents the overproduction of chitin by coordinating both chitin upregulation and negative regulation of the CWI signaling pathway. This article has an associated First Person interview with the first author of the paper.


Assuntos
Calcineurina , Candida albicans , Calcineurina/genética , Candida albicans/genética , Parede Celular , Quitina , Proteínas Fúngicas , Humanos , Lipopeptídeos/farmacologia
4.
PLoS Pathog ; 13(5): e1006403, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28542528

RESUMO

Candida albicans is able to proliferate in environments that vary dramatically in ambient pH, a trait required for colonising niches such as the stomach, vaginal mucosal and the GI tract. Here we show that growth in acidic environments involves cell wall remodelling which results in enhanced chitin and ß-glucan exposure at the cell wall periphery. Unmasking of the underlying immuno-stimulatory ß-glucan in acidic environments enhanced innate immune recognition of C. albicans by macrophages and neutrophils, and induced a stronger proinflammatory cytokine response, driven through the C-type lectin-like receptor, Dectin-1. This enhanced inflammatory response resulted in significant recruitment of neutrophils in an intraperitoneal model of infection, a hallmark of symptomatic vaginal colonisation. Enhanced chitin exposure resulted from reduced expression of the cell wall chitinase Cht2, via a Bcr1-Rim101 dependent signalling cascade, while increased ß-glucan exposure was regulated via a non-canonical signalling pathway. We propose that this "unmasking" of the cell wall may induce non-protective hyper activation of the immune system during growth in acidic niches, and may attribute to symptomatic vaginal infection.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Parede Celular/imunologia , Animais , Candida albicans/fisiologia , Candidíase/microbiologia , Parede Celular/química , Humanos , Concentração de Íons de Hidrogênio , Imunidade Inata , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos
5.
PLoS Pathog ; 12(4): e1005566, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27073846

RESUMO

Efficient carbon assimilation is critical for microbial growth and pathogenesis. The environmental yeast Saccharomyces cerevisiae is "Crabtree positive", displaying a rapid metabolic switch from the assimilation of alternative carbon sources to sugars. Following exposure to sugars, this switch is mediated by the transcriptional repression of genes (carbon catabolite repression) and the turnover (catabolite inactivation) of enzymes involved in the assimilation of alternative carbon sources. The pathogenic yeast Candida albicans is Crabtree negative. It has retained carbon catabolite repression mechanisms, but has undergone posttranscriptional rewiring such that gluconeogenic and glyoxylate cycle enzymes are not subject to ubiquitin-mediated catabolite inactivation. Consequently, when glucose becomes available, C. albicans can continue to assimilate alternative carbon sources alongside the glucose. We show that this metabolic flexibility promotes host colonization and virulence. The glyoxylate cycle enzyme isocitrate lyase (CaIcl1) was rendered sensitive to ubiquitin-mediated catabolite inactivation in C. albicans by addition of a ubiquitination site. This mutation, which inhibits lactate assimilation in the presence of glucose, reduces the ability of C. albicans cells to withstand macrophage killing, colonize the gastrointestinal tract and cause systemic infections in mice. Interestingly, most S. cerevisiae clinical isolates we examined (67%) have acquired the ability to assimilate lactate in the presence of glucose (i.e. they have become Crabtree negative). These S. cerevisiae strains are more resistant to macrophage killing than Crabtree positive clinical isolates. Moreover, Crabtree negative S. cerevisiae mutants that lack Gid8, a key component of the Glucose-Induced Degradation complex, are more resistant to macrophage killing and display increased virulence in immunocompromised mice. Thus, while Crabtree positivity might impart a fitness advantage for yeasts in environmental niches, the more flexible carbon assimilation strategies offered by Crabtree negativity enhance the ability of yeasts to colonize and infect the mammalian host.


Assuntos
Candida albicans/metabolismo , Candida albicans/patogenicidade , Candidíase/metabolismo , Macrófagos/microbiologia , Saccharomyces cerevisiae/metabolismo , Virulência/fisiologia , Animais , Western Blotting , Metabolismo dos Carboidratos , Linhagem Celular , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Ubiquitinação
6.
PLoS Pathog ; 10(4): e1004050, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24722226

RESUMO

Chitin is an essential structural polysaccharide of fungal pathogens and parasites, but its role in human immune responses remains largely unknown. It is the second most abundant polysaccharide in nature after cellulose and its derivatives today are widely used for medical and industrial purposes. We analysed the immunological properties of purified chitin particles derived from the opportunistic human fungal pathogen Candida albicans, which led to the selective secretion of the anti-inflammatory cytokine IL-10. We identified NOD2, TLR9 and the mannose receptor as essential fungal chitin-recognition receptors for the induction of this response. Chitin reduced LPS-induced inflammation in vivo and may therefore contribute to the resolution of the immune response once the pathogen has been defeated. Fungal chitin also induced eosinophilia in vivo, underpinning its ability to induce asthma. Polymorphisms in the identified chitin receptors, NOD2 and TLR9, predispose individuals to inflammatory conditions and dysregulated expression of chitinases and chitinase-like binding proteins, whose activity is essential to generate IL-10-inducing fungal chitin particles in vitro, have also been linked to inflammatory conditions and asthma. Chitin recognition is therefore critical for immune homeostasis and is likely to have a significant role in infectious and allergic disease.


Assuntos
Candida albicans/química , Quitina/imunologia , Interleucina-10/imunologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Receptor Toll-Like 9/imunologia , Animais , Asma/genética , Asma/imunologia , Asma/patologia , Candida albicans/imunologia , Quitina/química , Feminino , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-10/genética , Masculino , Camundongos , Proteína Adaptadora de Sinalização NOD2/genética , Receptor Toll-Like 9/genética
7.
J Cell Sci ; 126(Pt 12): 2668-77, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23606739

RESUMO

In fungi, as with all walled organisms, cytokinesis followed by septation marks the end of the cell cycle and is essential for cell division and viability. For yeasts, the septal cross-wall comprises a ring and primary septal plate composed of chitin, and a secondary septum thickened with ß(1,3)-glucan. In the human pathogen Candida albicans, chitin synthase enzyme Chs1 builds the primary septum that is surrounded by a chitin ring made by Chs3. Here we show that the lethal phenotype induced by repression of CHS1 was abrogated by stress-induced synthesis of alternative and novel septal types synthesized by other chitin synthase enzymes that have never before been implicated in septation. Chs2 and Chs8 formed a functional salvage septum, even in the absence of both Chs1 and Chs3. A second type of salvage septum formed by Chs2 in combination with Chs3 or Chs8 was proximally offset in the mother-bud neck. Chs3 alone or in combination with Chs8 formed a greatly thickened third type of salvage septum. Therefore, cell wall stress induced alternative forms of septation that rescued cell division in the absence of Chs1, demonstrating that fungi have previously unsuspected redundant strategies to enable septation and cell division to be maintained, even under potentially lethal environmental conditions.


Assuntos
Candida albicans/fisiologia , Citocinese/fisiologia , Estresse Fisiológico/fisiologia , Candida albicans/metabolismo , Divisão Celular/fisiologia , Parede Celular/metabolismo , Parede Celular/fisiologia , Quitina/metabolismo , Quitina Sintase/metabolismo
8.
Fungal Genet Biol ; 82: 264-76, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26257018

RESUMO

Candida albicans has four chitin synthases from three different enzyme classes which deposit chitin in the cell wall, including at the polarized tips of growing buds and hyphae, and sites of septation. The two class I enzymes, Chs2 and Chs8, are responsible for most of the measurable chitin synthase activity in vitro, but their precise biological functions in vivo remain obscure. In this work, detailed phenotypic analyses of a chs2Δchs8Δ mutant have shown that C. albicans class I chitin synthases promote cell integrity during early polarized growth in yeast and hyphal cells. This was supported by live cell imaging of YFP-tagged versions of the class I chitin synthases which revealed that Chs2-YFP was localized at sites of polarized growth. Furthermore, a unique and dynamic pattern of localization of the class I enzymes at septa of yeast and hyphae was revealed. Phosphorylation of Chs2 on the serine at position 222 was shown to regulate the amount of Chs2 that is localized to sites of polarized growth and septation. Independently from this post-translational modification, specific cell wall stresses were also shown to regulate the amount of Chs2 that localizes to specific sites in cells, and this was linked to the ability of the class I enzymes to reinforce cell wall integrity during early polarized growth in the presence of these stresses.


Assuntos
Candida albicans/metabolismo , Parede Celular/metabolismo , Quitina Sintase/metabolismo , Candida albicans/genética , Quitina Sintase/genética , Expressão Gênica , Genes Reporter , Hifas , Mutação , Fenótipo , Fosforilação , Transporte Proteico , Proteínas Recombinantes de Fusão , Estresse Fisiológico/genética
9.
PLoS Pathog ; 9(4): e1003276, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23633946

RESUMO

The fungal cell wall is the first point of interaction between an invading fungal pathogen and the host immune system. The outer layer of the cell wall is comprised of GPI anchored proteins, which are post-translationally modified by both N- and O-linked glycans. These glycans are important pathogen associated molecular patterns (PAMPs) recognised by the innate immune system. Glycan synthesis is mediated by a series of glycosyl transferases, located in the endoplasmic reticulum and Golgi apparatus. Mnn2 is responsible for the addition of the initial α1,2-mannose residue onto the α1,6-mannose backbone, forming the N-mannan outer chain branches. In Candida albicans, the MNN2 gene family is comprised of six members (MNN2, MNN21, MNN22, MNN23, MNN24 and MNN26). Using a series of single, double, triple, quintuple and sextuple mutants, we show, for the first time, that addition of α1,2-mannose is required for stabilisation of the α1,6-mannose backbone and hence regulates mannan fibril length. Sequential deletion of members of the MNN2 gene family resulted in the synthesis of lower molecular weight, less complex and more uniform N-glycans, with the sextuple mutant displaying only un-substituted α1,6-mannose. TEM images confirmed that the sextuple mutant was completely devoid of the outer mannan fibril layer, while deletion of two MNN2 orthologues resulted in short mannan fibrils. These changes in cell wall architecture correlated with decreased proinflammatory cytokine induction from monocytes and a decrease in fungal virulence in two animal models. Therefore, α1,2-mannose of N-mannan is important for both immune recognition and virulence of C. albicans.


Assuntos
Candida albicans/imunologia , Candida albicans/patogenicidade , Mananas/imunologia , Manose/metabolismo , Manosiltransferases/metabolismo , Glicoproteínas de Membrana/imunologia , Animais , Candida albicans/enzimologia , Candidíase/imunologia , Parede Celular/química , Parede Celular/imunologia , Feminino , Proteínas Fúngicas/genética , Proteínas Fúngicas/imunologia , Proteínas Fúngicas/metabolismo , Humanos , Mananas/química , Manose/química , Manosiltransferases/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Polissacarídeos/metabolismo , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Alinhamento de Sequência , Deleção de Sequência
10.
Nat Commun ; 15(1): 6818, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39122699

RESUMO

More than two million people worldwide are affected by life-threatening, invasive fungal infections annually. Candida species are the most common cause of nosocomial, invasive fungal infections and are associated with mortality rates above 40%. Despite the increasing incidence of drug-resistance, the development of novel antifungal formulations has been limited. Here we investigate the antifungal mode of action and therapeutic potential of positively charged, synthetic peptide mimics to combat Candida albicans infections. Our data indicates that these synthetic polymers cause endoplasmic reticulum stress and affect protein glycosylation, a mode of action distinct from currently approved antifungal drugs. The most promising polymer composition damaged the mannan layer of the cell wall, with additional membrane-disrupting activity. The synergistic combination of the polymer with caspofungin prevented infection of human epithelial cells in vitro, improved fungal clearance by human macrophages, and significantly increased host survival in a Galleria mellonella model of systemic candidiasis. Additionally, prolonged exposure of C. albicans to the synergistic combination of polymer and caspofungin did not lead to the evolution of tolerant strains in vitro. Together, this work highlights the enormous potential of these synthetic peptide mimics to be used as novel antifungal formulations as well as adjunctive antifungal therapy.


Assuntos
Antifúngicos , Candida albicans , Candidíase , Caspofungina , Sinergismo Farmacológico , Peptídeos , Candida albicans/efeitos dos fármacos , Antifúngicos/farmacologia , Humanos , Caspofungina/farmacologia , Animais , Candidíase/tratamento farmacológico , Candidíase/microbiologia , Peptídeos/farmacologia , Peptídeos/química , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Mananas/farmacologia , Mananas/química , Mariposas/microbiologia , Mariposas/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Polímeros/farmacologia , Polímeros/química
11.
Nat Rev Microbiol ; 21(4): 248-259, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36266346

RESUMO

The fungal cell wall is essential for growth and survival, and is a key target for antifungal drugs and the immune system. The cell wall must be robust but flexible, protective and shielding yet porous to nutrients and membrane vesicles and receptive to exogenous signals. Most fungi have a common inner wall skeleton of chitin and ß-glucans that functions as a flexible viscoelastic frame to which a more diverse set of outer cell wall polymers and glycosylated proteins are attached. Whereas the inner wall largely determines shape and strength, the outer wall confers properties of hydrophobicity, adhesiveness, and chemical and immunological heterogeneity. The spatial organization and dynamic regulation of the wall in response to prevailing growth conditions enable fungi to thrive within changing, diverse and often hostile environments. Understanding this architecture provides opportunities to develop diagnostics and drugs to combat life-threatening fungal infections.


Assuntos
Glucanos , beta-Glucanas , Glucanos/análise , Glucanos/química , Glucanos/metabolismo , Parede Celular/metabolismo , Antifúngicos , beta-Glucanas/análise , beta-Glucanas/metabolismo , Quitina/análise , Quitina/química , Quitina/metabolismo , Proteínas Fúngicas/análise , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo
12.
J Cell Sci ; 123(Pt 13): 2199-206, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20530569

RESUMO

The ability to undergo polarised cell growth is fundamental to the development of almost all walled organisms. Fungi are characterised by yeasts and moulds, and both cellular forms have been studied extensively as tractable models of cell polarity. Chitin is a hallmark component of fungal cell walls. Chitin synthesis is essential for growth, viability and rescue from many conditions that impair cell-wall integrity. In the polymorphic human pathogen Candida albicans, chitin synthase 3 (Chs3) synthesises the majority of chitin in the cell wall and is localised at the tips of growing buds and hyphae, and at the septum. An analysis of the C. albicans phospho-proteome revealed that Chs3 can be phosphorylated at Ser139. Mutation of this site showed that both phosphorylation and dephosphorylation are required for the correct localisation and function of Chs3. The kinase Pkc1 was not required to target Chs3 to sites of polarised growth. This is the first report demonstrating an essential role for chitin synthase phosphorylation in the polarised biosynthesis of fungal cell walls and suggests a new mechanism for the regulation of this class of glycosyl-transferase enzyme.


Assuntos
Candida albicans/metabolismo , Polaridade Celular , Quitina/biossíntese , Sequência de Aminoácidos , Candida albicans/citologia , Ciclo Celular/fisiologia , Quitina Sintase/genética , Quitina Sintase/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Hifas/metabolismo , Hifas/ultraestrutura , Isoenzimas/genética , Isoenzimas/metabolismo , Dados de Sequência Molecular , Fosforilação , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
13.
FEMS Microbiol Ecol ; 98(10)2022 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-36007932

RESUMO

The human gut microbiota protects the host from invading pathogens and the overgrowth of indigenous opportunistic species via a process called colonization resistance. Here, we investigated the antagonistic activity of human gut bacteria towards Candida albicans, an opportunistic fungal pathogen that can cause severe infections in susceptible individuals. Coculture batch incubations of C. albicans in the presence of faecal microbiota from six healthy individuals revealed varying levels of inhibitory activity against C. albicans. 16S rRNA gene amplicon profiling of these faecal coculture bacterial communities showed that the Bifidobacteriaceae family, and Bifidobacterium adolescentis in particular, were most correlated with antagonistic activity against C. albicans. Follow-up mechanistic studies performed under anaerobic conditions confirmed that culture supernatants of Bifidobacterium species, particularly B. adolescentis, inhibited C. albicans in vitro. Fermentation acids (FA), including acetate and lactate, present in the bifidobacterial supernatants were important contributors to inhibitory activity. However, increasing the pH of both bacterial supernatants and mixtures of FA reduced their anti-Candida effects, indicating a combinatorial effect of prevailing pH and FA. This work, therefore, demonstrates potential mechanisms underpinning gut microbiome-mediated colonization resistance against C. albicans, and identifies particularly inhibitory components such as bifidobacteria and FA as targets for further study.


Assuntos
Candida albicans , Microbioma Gastrointestinal , Bactérias , Bifidobacterium , Humanos , Lactatos/farmacologia , RNA Ribossômico 16S/genética
14.
Infect Immun ; 79(5): 1961-70, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21357722

RESUMO

Chitin is a skeletal cell wall polysaccharide of the inner cell wall of fungal pathogens. As yet, little about its role during fungus-host immune cell interactions is known. We show here that ultrapurified chitin from Candida albicans cell walls did not stimulate cytokine production directly but blocked the recognition of C. albicans by human peripheral blood mononuclear cells (PBMCs) and murine macrophages, leading to significant reductions in cytokine production. Chitin did not affect the induction of cytokines stimulated by bacterial cells or lipopolysaccharide (LPS), indicating that blocking was not due to steric masking of specific receptors. Toll-like receptor 2 (TLR2), TLR4, and Mincle (the macrophage-inducible C-type lectin) were not required for interactions with chitin. Dectin-1 was required for immune blocking but did not bind chitin directly. Cytokine stimulation was significantly reduced upon stimulation of PBMCs with heat-killed chitin-deficient C. albicans cells but not with live cells. Therefore, chitin is normally not exposed to cells of the innate immune system but is capable of influencing immune recognition by blocking dectin-1-mediated engagement with fungal cell walls.


Assuntos
Candidíase/imunologia , Quitina/imunologia , Interações Hospedeiro-Parasita/imunologia , Imunidade Inata/imunologia , Macrófagos/imunologia , Animais , Candida albicans/imunologia , Citocinas/biossíntese , Citocinas/imunologia , Humanos , Lectinas Tipo C , Leucócitos Mononucleares/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/imunologia
15.
PLoS Pathog ; 4(4): e1000040, 2008 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-18389063

RESUMO

Echinocandins are a new generation of novel antifungal agent that inhibit cell wall beta(1,3)-glucan synthesis and are normally cidal for the human pathogen Candida albicans. Treatment of C. albicans with low levels of echinocandins stimulated chitin synthase (CHS) gene expression, increased Chs activity, elevated chitin content and reduced efficacy of these drugs. Elevation of chitin synthesis was mediated via the PKC, HOG, and Ca(2+)-calcineurin signalling pathways. Stimulation of Chs2p and Chs8p by activators of these pathways enabled cells to survive otherwise lethal concentrations of echinocandins, even in the absence of Chs3p and the normally essential Chs1p, which synthesize the chitinous septal ring and primary septum of the fungus. Under such conditions, a novel proximally offset septum was synthesized that restored the capacity for cell division, sustained the viability of the cell, and abrogated morphological and growth defects associated with echinocandin treatment and the chs mutations. These findings anticipate potential resistance mechanisms to echinocandins. However, echinocandins and chitin synthase inhibitors synergized strongly, highlighting the potential for combination therapies with greatly enhanced cidal activity.


Assuntos
Antifúngicos/farmacologia , Candida albicans/efeitos dos fármacos , Quitina Sintase/biossíntese , Quitina/biossíntese , Equinocandinas/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Benzenossulfonatos/farmacologia , Cloreto de Cálcio/farmacologia , Candida albicans/metabolismo , Quitina Sintase/antagonistas & inibidores , Quitina Sintase/genética , Relação Dose-Resposta a Droga , Antagonismo de Drogas , Quimioterapia Combinada , Ativadores de Enzimas/farmacologia
16.
Cell Surf ; 6: 100047, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33294751

RESUMO

Despite the importance of fungal cell walls as the principle determinant of fungal morphology and the defining element determining fungal interactions with other cells, few scalar models have been developed that reconcile chemical and microscopic attributes of its structure. The cell wall of the fungal pathogen Candida albicans is comprised of an amorphous inner skeletal layer of ß(1,3)- and ß(1,6)-glucan and chitin and an outer fibrillar layer thought to be dominated by highly mannosylated cell wall proteins. The architecture of these two layers can be resolved at the electron microscopy level, but the visualised structure of the wall has not yet been defined precisely in chemical terms. We have therefore examined the precise structure, location and molecular sizes of the cell wall components using transmission electron microscopy and tomography and tested predictions of the cell wall models using mutants and agents that perturb the normal cell wall structure. We demonstrate that the fibrils are comprised of a frond of N-linked outer chain mannans linked to a basal layer of GPI-proteins concentrated in the mid-wall region and that the non-elastic chitin microfibrils are cantilevered with sufficient lengths of non-fibrillar chitin and/or ß-glucan to enable the chitin-glucan cage to flex, e.g. during morphogenesis and osmotic swelling. We present the first three-dimensional nano-scalar model of the C. albicans cell wall which can be used to test hypotheses relating to the structure-function relationships that underpin the pathobiology of this fungal pathogen.

17.
Mol Genet Genomics ; 281(4): 459-71, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19153767

RESUMO

Maintenance of the integrity of the cell wall in fungi is essential. One mechanism that cells use to maintain cell wall integrity in response to cell wall damage is to up-regulate chitin synthesis. In Candida albicans, the PKC cell wall integrity, Ca(2+)/calcineurin and high osmolarity glycerol (HOG) signalling pathways co-ordinately regulate chitin synthesis in response to cell wall stress. The transcription factors downstream of these pathways and their DNA binding sites within the promoters of target genes are well characterised in Saccharomyces cerevisiae, but not in C. albicans. The promoters of the C. albicans class I CHS genes (CaCHS2 and CaCHS8) were functionally dissected with the aim of identifying and characterising the transcription factors and promoter elements that mediate the transcriptional up-regulation of CaCHS2 and CaCHS8 in response to cell wall stress. This analysis provided evidence that the PKC cell wall integrity pathway may operate through RLM1-elements in the CaCHS2 and CaCHS8 promoters, but that promoter sequences that respond to the Ca(2+)/calcineurin and HOG signalling pathways in S. cerevisiae did not directly regulate chitin synthase 2 and 8 gene transcription in C. albicans.


Assuntos
Candida albicans/enzimologia , Candida albicans/genética , Quitina Sintase/genética , Genes Fúngicos , Regiões Promotoras Genéticas , Sequência de Bases , Sítios de Ligação , Parede Celular/enzimologia , Quitina Sintase/classificação , Primers do DNA/genética , DNA Fúngico/genética , DNA Fúngico/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Transdução de Sinais , Fatores de Transcrição/metabolismo
18.
mBio ; 9(1)2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29437927

RESUMO

The fungal cell wall is a critically important structure that represents a permeability barrier and protective shield. We probed Candida albicans and Cryptococcus neoformans with liposomes containing amphotericin B (AmBisome), with or without 15-nm colloidal gold particles. The liposomes have a diameter of 60 to 80 nm, and yet their mode of action requires them to penetrate the fungal cell wall to deliver amphotericin B to the cell membrane, where it binds to ergosterol. Surprisingly, using cryofixation techniques with electron microscopy, we observed that the liposomes remained intact during transit through the cell wall of both yeast species, even though the predicted porosity of the cell wall (pore size, ~5.8 nm) is theoretically too small to allow these liposomes to pass through intact. C. albicans mutants with altered cell wall thickness and composition were similar in both their in vitro AmBisome susceptibility and the ability of liposomes to penetrate the cell wall. AmBisome exposed to ergosterol-deficient C. albicans failed to penetrate beyond the mannoprotein-rich outer cell wall layer. Melanization of C. neoformans and the absence of amphotericin B in the liposomes were also associated with a significant reduction in liposome penetration. Therefore, AmBisome can reach cell membranes intact, implying that fungal cell wall viscoelastic properties are permissive to vesicular structures. The fact that AmBisome can transit through chemically diverse cell wall matrices when these liposomes are larger than the theoretical cell wall porosity suggests that the wall is capable of rapid remodeling, which may also be the mechanism for release of extracellular vesicles.IMPORTANCE AmBisome is a broad-spectrum fungicidal antifungal agent in which the hydrophobic polyene antibiotic amphotericin B is packaged within a 60- to 80-nm liposome. The mode of action involves perturbation of the fungal cell membrane by selectively binding to ergosterol, thereby disrupting membrane function. We report that the AmBisome liposome transits through the cell walls of both Candida albicans and Cryptococcus neoformans intact, despite the fact that the liposome is larger than the theoretical cell wall porosity. This implies that the cell wall has deformable, viscoelastic properties that are permissive to transwall vesicular traffic. These observations help explain the low toxicity of AmBisome, which can deliver its payload directly to the cell membrane without unloading the polyene in the cell wall. In addition, these findings suggest that extracellular vesicles may also be able to pass through the cell wall to deliver soluble and membrane-bound effectors and other molecules to the extracellular space.


Assuntos
Anfotericina B/metabolismo , Antifúngicos/metabolismo , Candida albicans/química , Parede Celular/química , Cryptococcus neoformans/química , Elasticidade , Viscosidade , Candida albicans/efeitos dos fármacos , Parede Celular/metabolismo , Microscopia Crioeletrônica , Cryptococcus neoformans/efeitos dos fármacos
19.
Methods Mol Biol ; 845: 41-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22328366

RESUMO

Techniques used to generate mutants in Candida albicans commonly result in additional and undesired genetic rearrangements. Detection of aneuploidy is, therefore, an important step forward in the quality control of mutant phenotypes. In this chapter, we describe how to extract genomic DNA and perform a quantitative multiplex PCR to compare the karyotype of any mutant strain to that of its parent and allow the detection of any unwanted aneuploidy.


Assuntos
Aneuploidia , Candida albicans/genética , Cariotipagem/métodos , Mutagênese/genética , DNA Fúngico/genética , DNA Fúngico/isolamento & purificação , Genoma Fúngico/genética , Reação em Cadeia da Polimerase Multiplex
20.
BMC Res Notes ; 5: 258, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22631601

RESUMO

BACKGROUND: Saccharomyces cerevisiae senses hyperosmotic conditions via the HOG signaling network that activates the stress-activated protein kinase, Hog1, and modulates metabolic fluxes and gene expression to generate appropriate adaptive responses. The integral control mechanism by which Hog1 modulates glycerol production remains uncharacterized. An additional Hog1-independent mechanism retains intracellular glycerol for adaptation. Candida albicans also adapts to hyperosmolarity via a HOG signaling network. However, it remains unknown whether Hog1 exerts integral or proportional control over glycerol production in C. albicans. RESULTS: We combined modeling and experimental approaches to study osmotic stress responses in S. cerevisiae and C. albicans. We propose a simple ordinary differential equation (ODE) model that highlights the integral control that Hog1 exerts over glycerol biosynthesis in these species. If integral control arises from a separation of time scales (i.e. rapid HOG activation of glycerol production capacity which decays slowly under hyperosmotic conditions), then the model predicts that glycerol production rates elevate upon adaptation to a first stress and this makes the cell adapts faster to a second hyperosmotic stress. It appears as if the cell is able to remember the stress history that is longer than the timescale of signal transduction. This is termed the long-term stress memory. Our experimental data verify this. Like S. cerevisiae, C. albicans mimimizes glycerol efflux during adaptation to hyperosmolarity. Also, transient activation of intermediate kinases in the HOG pathway results in a short-term memory in the signaling pathway. This determines the amplitude of Hog1 phosphorylation under a periodic sequence of stress and non-stressed intervals. Our model suggests that the long-term memory also affects the way a cell responds to periodic stress conditions. Hence, during osmohomeostasis, short-term memory is dependent upon long-term memory. This is relevant in the context of fungal responses to dynamic and changing environments. CONCLUSIONS: Our experiments and modeling have provided an example of identifying integral control that arises from time-scale separation in different processes, which is an important functional module in various contexts.


Assuntos
Candida albicans/enzimologia , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Estresse Fisiológico , Biologia de Sistemas , Adaptação Fisiológica , Ativação Enzimática , Glicerol/metabolismo , Modelos Biológicos , Pressão Osmótica , Fosforilação , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa