Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-32094132

RESUMO

Lactoferrin (LF) is a multifunctional milk protein with antimicrobial activity against a range of pathogens. While numerous studies report that LF is active against fungi, there are considerable differences in the level of antifungal activity and the capacity of LF to interact with other drugs. Here we undertook a comprehensive evaluation of the antifungal spectrum of activity of three defined sources of LF across 22 yeast and 24 mold species and assessed its interactions with six widely used antifungal drugs. LF was broadly and consistently active against all yeast species tested (MICs, 8 to 64 µg/ml), with the extent of activity being strongly affected by iron saturation. LF was synergistic with amphotericin B (AMB) against 19 out of 22 yeast species tested, and synergy was unaffected by iron saturation but was affected by the extent of LF digestion. LF-AMB combination therapy significantly prolonged the survival of Galleria mellonella wax moth larvae infected with Candida albicans or Cryptococcus neoformans and decreased the fungal burden 12- to 25-fold. Evidence that LF directly interacts with the fungal cell surface was seen via scanning electron microscopy, which showed pore formation, hyphal thinning, and major cell collapse in response to LF-AMB synergy. Important virulence mechanisms were disrupted by LF-AMB treatment, which significantly prevented biofilms in C. albicans and C. glabrata, inhibited hyphal development in C. albicans, and reduced cell and capsule size and phenotypic diversity in Cryptococcus Our results demonstrate the potential of LF-AMB as an antifungal treatment that is broadly synergistic against important yeast pathogens, with the synergy being attributed to the presence of one or more LF peptides.


Assuntos
Anfotericina B/farmacologia , Antifúngicos/farmacologia , Lactoferrina/farmacologia , Leveduras/efeitos dos fármacos , Animais , Biofilmes/efeitos dos fármacos , Candida albicans/efeitos dos fármacos , Candida albicans/ultraestrutura , Cryptococcus neoformans/efeitos dos fármacos , Cryptococcus neoformans/ultraestrutura , Sinergismo Farmacológico , Cápsulas Fúngicas/efeitos dos fármacos , Hifas/efeitos dos fármacos , Larva/microbiologia , Testes de Sensibilidade Microbiana , Mariposas , Leveduras/ultraestrutura
2.
J Mycol Med ; 30(1): 100905, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31706700

RESUMO

INTRODUCTION: Iron chelator has previously demonstrated fungicidal effects. This study aimed to investigate the antifungal activity of the iron chelators deferoxamine (DFO) and deferasirox (DSX) against Cryptococcus. MATERIALS AND METHODS: Cryptococcus neoformans and Cryptococcus gattii were used to determine the minimal inhibitory concentrations (MICs) of DFO and DSX, and the fractional inhibitory concentration index (FICI) of DFO and DSX when combined with amphotericin B (AMB). Expression of cryptococcal CFT1, CFT2, and CIR1 genes was determined using real-time polymerase chain reaction (PCR). RESULTS: Neither DFO nor DSX alone showed antifungal activity against Cryptococcus strains. When combined with AMB, the MICs of DFO and DSX decreased from>200µg/mL to 6.25 or 12.5µg/mL. The MIC of AMB decreased one-fold dilution in most strains when combined with iron chelators. The FICI of DFO+AMB and DSX+AMB was 0.5 and 1, respectively. C. neoformans showed significant growth retardation when incubated with a combination of sub-MIC concentrations of AMB and DFO; whereas, C. gattii demonstrated lesser growth retardation in DFO+AMB. No cryptococcal growth retardation was observed when DSX was combined with AMB. When C. neoformans was grown in DFO, the CFT1, CFT2, and CIR1 proteins were expressed 1.7, 2.0, and 0.9 times, respectively. When C. neoformans was grown in DSX, the CFT1, CFT2, and CIR1 genes were expressed 0.5, 0.6, and 0.3 times, respectively. CONCLUSION: Synergistic antifungal activity of combination DFO and AMB was observed in Cryptococcus. Relatively increased CFT1 and CFT2 expression may be associated with the effect of DFO that inhibits the growth of fungi.


Assuntos
Cryptococcus/efeitos dos fármacos , Cryptococcus/crescimento & desenvolvimento , Cryptococcus/genética , Quelantes de Ferro/farmacologia , Ferro/metabolismo , Anfotericina B/farmacologia , Antifúngicos/farmacologia , Criptococose/tratamento farmacológico , Criptococose/microbiologia , Cryptococcus/metabolismo , Cryptococcus neoformans/efeitos dos fármacos , Cryptococcus neoformans/genética , Cryptococcus neoformans/crescimento & desenvolvimento , Cryptococcus neoformans/metabolismo , Deferasirox/farmacologia , Desferroxamina/farmacologia , Sinergismo Farmacológico , Cápsulas Fúngicas/efeitos dos fármacos , Cápsulas Fúngicas/genética , Cápsulas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Humanos , Infecções Fúngicas Invasivas/complicações , Infecções Fúngicas Invasivas/tratamento farmacológico , Infecções Fúngicas Invasivas/microbiologia , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/microbiologia , Redes e Vias Metabólicas/efeitos dos fármacos , Redes e Vias Metabólicas/genética , Testes de Sensibilidade Microbiana , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
3.
mSphere ; 4(3)2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31167944

RESUMO

Rhodotorula yeasts are pink, encapsulated basidiomycetes isolated from a variety of environments and clinical settings. They are increasingly linked with disease, particularly central venous catheter infections and meningitis, in immunocompromised patients. Eight clinical and eight environmental strains molecularly typed as Rhodotorula mucilaginosa were compared to six Cryptococcus neoformans strains for phenotypic variability. Growth on cell integrity-challenging media suggested that R. mucilaginosa cells possess differences in signaling pathways, cell wall composition, or assembly and that their membranes are more susceptible to perturbations than those of C. neoformans All 16 R. mucilaginosa strains produced urease, while none produced melanin with l-3,4-dihydroxyphenylalanine (l-DOPA) as a substrate. India ink staining reveals that clinical R. mucilaginosa capsules are larger than environmental capsules but that both are generally smaller than C. neoformans capsules. All R. mucilaginosa strains were resistant to fluconazole. Only two clinical strains were susceptible to voriconazole; all of the environmental strains were resistant. We generated an anticapsular antibody (Rh1) to R. mucilaginosa; Rh1 did not bind C. neoformans control strains, was specific to Rhodotorula species, and bound to all tested Rhodotorula strains. Binding assays performed with wheat germ agglutinin (WGA), concanavalin A (ConA), calcofluor white (CFW), and eosin Y dye (EY) cell surface probes suggested that chitin may be more accessible in R. mucilaginosa but that the total abundance of chitooligomers is less than in C. neoformans This report describes a novel reagent that can be used to identify Rhodotorula species and lays the foundation for future cell envelope composition analysis.IMPORTANCE Currently, there is very little known about the phenotypic variability within species of Rhodotorula strains and the role of their capsule. Cryptococcus neoformans has been considered the only encapsulated human fungal pathogen, but as more individuals come to live in states of immunocompromised health, they are more susceptible to fungal infections, including those by RhodotorulaR. mucilaginosa species are some of those most commonly associated with clinical infections. We wanted to know if clinical and environmental strains of R. mucilaginosa demonstrated disparate capsule phenotypes. With limited antifungal options available and clinical Rhodotorula spp. often resistant to common antifungal drugs such as fluconazole, caspofungin (1, 2), and voriconazole (2), a better understanding of the fungal biology could inform the design and use of future antifungal drugs. The generation of an antibody specific to Rhodotorula fungi could be a useful diagnostic tool, and this work presents the first mention of such in the literature.


Assuntos
Parede Celular/química , Cápsulas Fúngicas/química , Rhodotorula/química , Animais , Anticorpos Antifúngicos/imunologia , Antifúngicos/farmacologia , Parede Celular/efeitos dos fármacos , Cryptococcus neoformans/química , Cryptococcus neoformans/efeitos dos fármacos , Cápsulas Fúngicas/efeitos dos fármacos , Humanos , Melaninas , Fenótipo , Coelhos , Rhodotorula/efeitos dos fármacos , Transdução de Sinais , Urease/biossíntese
4.
Microb Pathog ; 123: 296-303, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30041002

RESUMO

The present study was conducted to investigate the effects of a natural product from honeybees, named propolis, against Cryptococcus neoformans and its effect in the expression of putative virulence factors, such as capsular polysaccharides, melanin production and urease enzyme. Ethanol extract propolis (EEP) was first tested for its anti-cryptococcal activity and explored its impact on virulence factors in both phenotypes and enzyme activities. Moreover, the cryptococcal virulence genes were investigated using real time RT-PCR. The MIC value of EEP, 1 mg ml-1, displayed potent inhibition of C. neoformans cell viability. Of note is the high efficacy of sub-MIC concentrations (ranging from 0.5 to 0.125 mg ml-1) in decreasing the production of capsule, melanin, as well as laccase and urease enzyme activities. Importantly, EEP exhibited statistically decrease in the expression of gene-encoded virulence factors. In conclusion, EEP mediates C. neoformans growth inhibition and virulence factors by reducing the gene-encoding virulence-associated proteins and, thereby, disrupting the morphologic presence and attenuating their virulence. This study introduced EEP as regards anti-cryptococcal virulence factors activities; therefore, EEP would provide alternative ways of controlling the pathogenicity.


Assuntos
Antifúngicos/farmacologia , Cryptococcus neoformans/efeitos dos fármacos , Própole/farmacologia , Fatores de Virulência/metabolismo , Animais , Antifúngicos/química , Abelhas/química , Criptococose/tratamento farmacológico , Cryptococcus neoformans/citologia , Cryptococcus neoformans/genética , Cryptococcus neoformans/metabolismo , Cápsulas Fúngicas/efeitos dos fármacos , Polissacarídeos Fúngicos/metabolismo , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Cinética , Lacase/metabolismo , Melaninas/metabolismo , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Fenótipo , Própole/química , Tailândia , Urease/metabolismo , Virulência/efeitos dos fármacos , Virulência/genética , Fatores de Virulência/genética
5.
Artigo em Inglês | MEDLINE | ID: mdl-29844051

RESUMO

Cryptococcus spp. are common opportunistic fungal pathogens, particularly in HIV patients. The approved drug miltefosine (MFS) has potential as an alternative antifungal against cryptococcosis; however, the mechanism of action of MFS in Cryptococcus is poorly understood. Here, we examined the effects of MFS on C. neoformans and C. gattii yeasts (planktonic and biofilm lifestyles) to clarify its mechanism of action. MFS presented inhibitory and fungicidal effects against planktonic Cryptococcus cells, with similar activities against dispersion biofilm cells, while sessile biofilm cells were less sensitive to MFS. Interestingly, MFS had postantifungal effect on Cryptococcus, with a proliferation delay of up to 8.15 h after a short exposure to fungicidal doses. MFS at fungicidal concentrations increased the plasma membrane permeability, likely due to a direct interaction with ergosterol, as suggested by competition assays with exogenous ergosterol. Moreover, MFS reduced the mitochondrial membrane potential, increased reactive oxygen species (ROS) production, and induced DNA fragmentation and condensation, all of which are hallmarks of apoptosis. Transmission electron microscopy analysis showed that MFS-treated yeasts had a reduced mucopolysaccharide capsule (confirmed by morphometry with light microscopy), plasma membrane irregularities, mitochondrial swelling, and a less conspicuous cell wall. Our results suggest that MFS increases the plasma membrane permeability in Cryptococcus via an interaction with ergosterol and also affects the mitochondrial membrane, eventually leading to apoptosis, in line with its fungicidal activity. These findings confirm the potential of MFS as an antifungal against C. neoformans and C. gattii and warrant further studies to establish clinical protocols for MFS use against cryptococcosis.


Assuntos
Antifúngicos/farmacologia , Apoptose/efeitos dos fármacos , Cryptococcus gattii/efeitos dos fármacos , Cryptococcus neoformans/efeitos dos fármacos , Fosforilcolina/análogos & derivados , Anfotericina B/farmacologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Criptococose/tratamento farmacológico , Criptococose/microbiologia , Cryptococcus gattii/metabolismo , Cryptococcus gattii/ultraestrutura , Cryptococcus neoformans/metabolismo , Cryptococcus neoformans/ultraestrutura , Fragmentação do DNA/efeitos dos fármacos , Ergosterol/metabolismo , Cápsulas Fúngicas/efeitos dos fármacos , Cápsulas Fúngicas/metabolismo , Cápsulas Fúngicas/ultraestrutura , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Infecções Oportunistas/tratamento farmacológico , Infecções Oportunistas/microbiologia , Fosforilcolina/farmacologia , Plâncton/efeitos dos fármacos , Plâncton/crescimento & desenvolvimento , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-28031206

RESUMO

Cryptococcus neoformans is an environmentally ubiquitous fungal pathogen that primarily causes disease in people with compromised immune systems, particularly those with advanced AIDS. There are estimated to be almost 1 million cases per year of cryptococcal meningitis in patients infected with human immunodeficiency virus, leading to over 600,000 annual deaths, with a particular burden in sub-Saharan Africa. Amphotericin B (AMB) and fluconazole (FLC) are key components of cryptococcal meningitis treatment: AMB is used for induction, and FLC is for consolidation, maintenance and, for occasional individuals, prophylaxis. However, the results of standard antifungal susceptibility testing (AFST) for AMB and FLC do not correlate well with therapeutic outcomes and, consequently, no clinical breakpoints have been established. While a number of explanations for this absence of correlation have been proffered, one potential reason that has not been adequately explored is the possibility that the physiological differences between the in vivo infection environment and the in vitro AFST environment lead to disparate drug susceptibilities. These susceptibility-influencing factors include melanization, which does not occur during AFST, the size of the polysaccharide capsule, which is larger in infecting cells than in those grown under normal laboratory conditions, and the presence of large polyploid "titan cells," which rarely occur under laboratory conditions. Understanding whether and how C. neoformans differentially expresses mechanisms of resistance to AMB and FLC in the AFST environment compared to the in vivo environment could enhance our ability to interpret AFST results and possibly lead to the development of more applicable testing methods.


Assuntos
Antifúngicos/farmacologia , Cryptococcus neoformans/efeitos dos fármacos , Cápsulas Fúngicas/efeitos dos fármacos , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Melaninas/biossíntese , Anfotericina B/farmacologia , Cryptococcus neoformans/patogenicidade , Cryptococcus neoformans/fisiologia , Farmacorresistência Fúngica/fisiologia , Fluconazol/farmacologia , Cápsulas Fúngicas/fisiologia , Polissacarídeos Fúngicos/biossíntese , Proteínas Fúngicas/biossíntese , Infecções por HIV/complicações , Infecções por HIV/microbiologia , Infecções por HIV/patologia , Infecções por HIV/virologia , Humanos , Melaninas/genética , Meningite Criptocócica/complicações , Meningite Criptocócica/microbiologia , Meningite Criptocócica/patologia , Meningite Criptocócica/virologia , Testes de Sensibilidade Microbiana , Pigmentação/fisiologia , Poliploidia
7.
Virulence ; 6(6): 618-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26103530

RESUMO

Cryptococcus neoformans undergoes phenotypical changes during host infection in order to promote persistence and survival. Studies have demonstrated that such adaptations require alterations in gene transcription networks by distinct mechanisms. Drugs such as the histone deacetylases inhibitors (HDACi) Sodium Butyrate (NaBut) and Trichostatin A (TSA) can alter the chromatin conformation and have been used to modulate epigenetic states in the treatment of diseases such as cancer. In this work, we have studied the effect of NaBut and TSA on the expression of C. neoformans major virulence phenotypes and on the survival rate of an animal model infected with drugs-treated yeasts. Both drugs affected fungal growth at 37°C more intensely than at 30°C; nonetheless, drugs did not affect cell viability at the concentrations we studied. HDACi also provoked the reduction of the fungal capsule expansion. Phospholipases enzyme activity decreased; mating process and melanin synthesis were also affected by both inhibitors. NaBut led to an increase in the population of cells in G2/M. Treated yeast cells, which were washed in order to remove the drugs from the culture medium prior to the inoculation in the Galleria mellonela infection model, did not cause significant difference at the host survival curve when compared to non-treated cells. Overall, NaBut effects on the impairment of C. neoformans main virulence factors were more intense and stable than the TSA effects.


Assuntos
Cryptococcus neoformans/efeitos dos fármacos , Cryptococcus neoformans/fisiologia , Inibidores de Histona Desacetilases/metabolismo , Animais , Ácido Butírico/metabolismo , Divisão Celular/efeitos dos fármacos , Cryptococcus neoformans/crescimento & desenvolvimento , Modelos Animais de Doenças , Cápsulas Fúngicas/efeitos dos fármacos , Cápsulas Fúngicas/metabolismo , Ácidos Hidroxâmicos/metabolismo , Lepidópteros , Melaninas/metabolismo , Viabilidade Microbiana/efeitos dos fármacos , Fenótipo , Fosfolipases/análise , Análise de Sobrevida , Temperatura , Virulência/efeitos dos fármacos
8.
PLoS One ; 9(11): e112669, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25392951

RESUMO

Cryptococcus gattii is an emergent human pathogen. Fluconazole is commonly used for treatment of cryptococcosis, but the emergence of less susceptible strains to this azole is a global problem and also the data regarding fluconazole-resistant cryptococcosis are scarce. We evaluate the influence of fluconazole on murine cryptococcosis and whether this azole alters the polysaccharide (PS) from cryptococcal cells. L27/01 strain of C. gattii was cultivated in high fluconazole concentrations and developed decreased drug susceptibility. This phenotype was named L27/01F, that was less virulent than L27/01 in mice. The physical, structural and electrophoretic properties of the PS capsule of L27/01F were altered by fluconazole. L27/01F presented lower antiphagocytic properties and reduced survival inside macrophages. The L27/01F did not affect the central nervous system, while the effect in brain caused by L27/01 strain began after only 12 hours. Mice infected with L27/01F presented lower production of the pro-inflammatory cytokines, with increased cellular recruitment in the lungs and severe pulmonary disease. The behavioral alterations were affected by L27/01, but no effects were detected after infection with L27/01F. Our results suggest that stress to fluconazole alters the capsule of C. gattii and influences the clinical manifestations of cryptococcosis.


Assuntos
Antifúngicos/farmacologia , Criptococose/tratamento farmacológico , Cryptococcus gattii/efeitos dos fármacos , Fluconazol/farmacologia , Cápsulas Fúngicas/efeitos dos fármacos , Polissacarídeos Fúngicos/química , Animais , Criptococose/microbiologia , Criptococose/mortalidade , Criptococose/patologia , Cryptococcus gattii/química , Cryptococcus gattii/patogenicidade , Farmacorresistência Fúngica/efeitos dos fármacos , Cápsulas Fúngicas/química , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Fenótipo , Índice de Gravidade de Doença , Análise de Sobrevida
9.
Carbohydr Res ; 389: 57-65, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24612865

RESUMO

In a project targeting the synthesis of large oligosaccharide structures corresponding to the Cryptococcus neoformans GXM capsular polysaccharide, an easy access to thiodisaccharide building blocks comprising a ß-linked glucuronic acid moiety and a 6-O-acetyl group was required. Several pathways to such building blocks have been investigated, addressing the problem of constructing a ß-linked glucuronic acid residue protected with groups that are orthogonal to a primary acetyl group. Two efficient routes have been developed, one using benzoylated glucosyl donors to form the ß-linkage followed by a change of protecting groups to benzyls and subsequent introduction of the carboxyl function and the acetyl group. The second route explored the possibility to achieve ß-selectivity using glucuronyl donors without acyl protecting groups. BF3-etherate promoted glycosylations with benzyl (2,3,4-tri-O-benzyl-α-D-glucupyranosyl)uronate trichloroacetimidate in the presence of nitrile solvents and at low temperatures reproducibly gave good yields of disaccharides with high ß-selectivity. Furthermore, the use of recently reported glucuronyl thioglycoside donors protected with a cyclic 2,4-silylene acetal was found to represent another efficient and completely ß-selective way to desired disaccharide building blocks.


Assuntos
Cryptococcus neoformans/metabolismo , Cápsulas Fúngicas/efeitos dos fármacos , Cápsulas Fúngicas/metabolismo , Polissacarídeos/química , Polissacarídeos/metabolismo , Tioglicosídeos/síntese química , Tioglicosídeos/farmacologia , Configuração de Carboidratos , Técnicas de Química Sintética , Cryptococcus neoformans/efeitos dos fármacos , Glicosilação , Tioglicosídeos/química
10.
Fungal Genet Biol ; 60: 64-73, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23608320

RESUMO

The principal capsular component of Cryptococcus neoformans, glucuronoxylomannan (GXM), interacts with surface glycans, including chitin-like oligomers. Although the role of GXM in cryptococcal infection has been well explored, there is no information on how chitooligomers affect fungal pathogenesis. In this study, surface chitooligomers of C. neoformans were blocked through the use of the wheat germ lectin (WGA) and the effects on animal pathogenesis, interaction with host cells, fungal growth and capsule formation were analyzed. Treatment of C. neoformans cells with WGA followed by infection of mice delayed mortality relative to animals infected with untreated fungal cells. This observation was associated with reduced brain colonization by lectin-treated cryptococci. Blocking chitooligomers also rendered yeast cells less efficient in their ability to associate with phagocytes. WGA did not affect fungal viability, but inhibited GXM release to the extracellular space and capsule formation. In WGA-treated yeast cells, genes that are involved in capsule formation and GXM traffic had their transcription levels decreased in comparison with untreated cells. Our results suggest that cellular pathways required for capsule formation and pathogenic mechanisms are affected by blocking chitin-derived structures at the cell surface of C. neoformans. Targeting chitooligomers with specific ligands may reveal new therapeutic alternatives to control cryptococcosis.


Assuntos
Cryptococcus neoformans/patogenicidade , Cápsulas Fúngicas/metabolismo , Fagocitose/efeitos dos fármacos , Polissacarídeos/metabolismo , Aglutininas do Germe de Trigo/farmacologia , Animais , Encéfalo/microbiologia , Quitina/metabolismo , Criptococose/tratamento farmacológico , Criptococose/patologia , Cryptococcus neoformans/crescimento & desenvolvimento , Cryptococcus neoformans/metabolismo , Cápsulas Fúngicas/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Aglutininas do Germe de Trigo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA