Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
mSphere ; : e0025324, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38814077

RESUMO

Aspergillus fumigatus is the leading cause of severe mold infections in immunocompromised patients. This common fungus possesses innate attributes that allow it to evade the immune system, including its ability to survive the high copper (Cu) levels in phagosomes. Our previous work has revealed that under high Cu levels, the A. fumigatus transcription factor AceA is activated, inducing the expression of the copper exporter CrpA to expel excess Cu. To identify additional elements in Cu resistance, we evolved A. fumigatus wild-type and mutant ΔaceA or ΔcrpA strains under increasing Cu concentrations. Sequencing of the resultant resistant strains identified both shared and unique evolutionary pathways to resistance. Reintroduction of three of the most common mutations in genes encoding Pma1 (plasma membrane H+-ATPase), Gcs1 (glutamate cysteine-ligase), and Cpa1 (carbamoyl-phosphate synthetase), alone and in combination, into wild-type A. fumigatus confirmed their additive role in conferring Cu resistance. Detailed analysis indicated that the pma1 mutation L424I preserves Pma1 H+-ATPase activity under high Cu concentrations and that the cpa1 mutation A37V confers a survival advantage to conidia in the presence of Cu. Interestingly, simultaneous mutations of all three genes did not alter virulence in infected mice. Our work has identified novel Cu-resistance pathways and provides an evolutionary approach for dissecting the molecular basis of A. fumigatus adaptation to diverse environmental challenges.IMPORTANCEAspergillus fumigatus is the most common mold infecting patients with weakened immunity. Infection is caused by the inhalation of mold spores into the lungs and is often fatal. In healthy individuals, spores are engulfed by lung immune cells and destroyed by a combination of enzymes, oxidants, and high levels of copper. However, the mold can protect itself by pumping out excess copper with specific transporters. Here, we evolved A. fumigatus under high copper levels and identified new genetic mutations that help it resist the toxic effects of copper. We studied how these mutations affect the mold's ability to resist copper and how they impact its ability to cause disease. This is the first such study in a pathogenic mold, and it gives us a better understanding of how it manages to bypass our body's defenses during an infection.

2.
Nat Commun ; 13(1): 2463, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35513379

RESUMO

Candida albicans is the most common fungal pathogen and a prevalent cause of deadly bloodstream infections. Better understanding of the immune response against it, and the ways by which it evades immunity, are crucial for developing new therapeutics against it. Natural Killer (NK) cells are innate lymphocytes best known for their role against viruses and tumors. In recent years it became clear that NK cells also play an important role in anti-fungal immunity. Here we show that while NK cells recognize and eliminate C. albicans, the fungal cells inhibit NK cells by manipulating the immune checkpoint receptor TIGIT (T cell immunoreceptor with Ig and ITIM domains) in both humans and mice. We identify the responsible fungal ligands as members of the Als (Agglutinin-Like Sequences) protein family. Furthermore, we show that blocking this interaction using immunotherapy with a TIGIT-blocking antibody can re-establish anti-Candida immunity and serve as a potential therapeutic tool.


Assuntos
Aglutininas , Candida albicans , Aglutininas/metabolismo , Animais , Candida albicans/metabolismo , Imunoterapia , Células Matadoras Naturais , Camundongos , Receptores Imunológicos/metabolismo
3.
Virulence ; 10(1): 925-934, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31694453

RESUMO

In contrast to mammalia, fungi are able to synthesize the branched-chain amino acid leucine de novo. Recently, the transcription factor LeuB has been shown to cross-regulate leucine biosynthesis, nitrogen metabolism and iron homeostasis in Aspergillus fumigatus, the most common human mold pathogen. Moreover, the leucine biosynthetic pathway intermediate α-isopropylmalate (α-IPM) has previously been shown to posttranslationally activate LeuB homologs in S. cerevisiae and A. nidulans. Here, we demonstrate that in A. fumigatus inactivation of both leucine biosynthetic enzymes α-IPM synthase (LeuC), which disrupts α-IPM synthesis, and α-IPM isomerase (LeuA), which causes cellular α-IPM accumulation, results in leucine auxotrophy. However, compared to lack of LeuA, lack of LeuC resulted in increased leucine dependence, a growth defect during iron starvation and decreased expression of LeuB-regulated genes including genes involved in iron acquisition. Lack of either LeuA or LeuC decreased virulence in an insect infection model, and inactivation of LeuC rendered A. fumigatus avirulent in a pulmonary aspergillosis mouse model. Taken together, we demonstrate that the lack of two leucine biosynthetic enzymes, LeuA and LeuC, results in significant phenotypic consequences indicating that the regulator LeuB is activated by α-IPM in A. fumigatus and that the leucine biosynthetic pathway is an attractive target for the development of antifungal drugs.


Assuntos
Aspergillus fumigatus/genética , Aspergillus fumigatus/patogenicidade , Proteínas Fúngicas/genética , Ferro/metabolismo , Leucina/biossíntese , Virulência , Adaptação Fisiológica , Animais , Aspergillus fumigatus/metabolismo , Proteínas de Bactérias/genética , Vias Biossintéticas , Feminino , Regulação Fúngica da Expressão Gênica , Homeostase , Hidroliases/genética , Larva/microbiologia , Camundongos , Camundongos Endogâmicos ICR , Mariposas/microbiologia , Aspergilose Pulmonar/microbiologia
4.
Virulence ; 9(1): 1036-1049, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30052132

RESUMO

BACKGROUND: Aspergillus fumigatus is the most prevalent airborne fungal pathogen, causing invasive fungal infections mainly in immunosuppressed individuals. Death rates from invasive aspergillosis remain high because of limited treatment options and increasing antifungal resistance. The aim of this study was to identify key fungal-specific genes participating in vitamin B biosynthesis in A. fumigatus. Because these genes are absent in humans they can serve as possible novel targets for antifungal drug development. METHODS: By sequence homology we identified, deleted and analysed four key A. fumigatus genes (riboB, panA, pyroA, thiB) involved respectively in the biosynthesis of riboflavin (vitamin B2), pantothenic acid (vitamin B5), pyridoxine (vitamin B6) and thiamine (vitamin B1). RESULTS: Deletion of riboB, panA, pyroA or thiB resulted in respective vitamin auxotrophy. Lack of riboflavin and pantothenic acid biosynthesis perturbed many cellular processes including iron homeostasis. Virulence in murine pulmonary and systemic models of infection was severely attenuated following deletion of riboB and panA, strongly reduced after pyroA deletion and weakly attenuated after thiB deletion. CONCLUSIONS: This study reveals the biosynthetic pathways of the vitamins riboflavin and pantothenic acid as attractive targets for novel antifungal therapy. Moreover, the virulence studies with auxotrophic mutants serve to identify the availability of nutrients to pathogens in host niches. ABBREVIATIONS: BPS: bathophenanthrolinedisulfonate; BSA: bovine serum albumin; CFU: colony forming unit; -Fe: iron starvation; +Fe: iron sufficiency; hFe: high iron; NRPSs: nonribosomal peptide synthetases; PKSs: polyketide synthaseses; wt: wild type.


Assuntos
Aspergilose/microbiologia , Aspergillus fumigatus/enzimologia , Aspergillus fumigatus/patogenicidade , Proteínas Fúngicas/metabolismo , Ferro/metabolismo , Ácido Pantotênico/biossíntese , Riboflavina/biossíntese , Animais , Aspergillus fumigatus/genética , Aspergillus fumigatus/metabolismo , Vias Biossintéticas , Feminino , Proteínas Fúngicas/genética , GTP Cicloidrolase/genética , GTP Cicloidrolase/metabolismo , Deleção de Genes , Homeostase , Humanos , Camundongos , Camundongos Endogâmicos ICR , Fosforilases/genética , Fosforilases/metabolismo , Virulência
5.
Virulence ; 9(1): 1273-1286, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30027796

RESUMO

The ubiquitous fungus Aspergillus flavus is notorious for contaminating many important crops and food-stuffs with the carcinogenic mycotoxin, aflatoxin. This fungus is also the second most frequent Aspergillus pathogen after A. fumigatus infecting immunosuppressed patients. In many human fungal pathogens including A. fumigatus, the ability to defend from toxic levels of copper (Cu) is essential in pathogenesis. In A. fumigatus, the Cu-fist DNA binding protein, AceA, and the Cu ATPase transporter, CrpA, play critical roles in Cu defense. Here, we show that A. flavus tolerates higher concentrations of Cu than A. fumigatus and other Aspergillus spp. associated with the presence of two homologs of A. fumigatus CrpA termed CrpA and CrpB. Both crpA and crpB are transcriptionally induced by increasing Cu concentrations via AceA activity. Deletion of crpA or crpB alone did not alter high Cu tolerance, suggesting they are redundant. Deletion of both genes resulted in extreme Cu sensitivity that was greater than that following deletion of the regulatory transcription factor aceA. The ΔcrpAΔcrpB and ΔaceA strains were also sensitive to ROI stress. Compared to wild type, these mutants were impaired in the ability to colonize maize seed treated with Cu fungicide but showed no difference in virulence on non-treated seed. A mouse model of invasive aspergillosis showed ΔcrpAΔcrpB and to a lesser degree ΔaceA to be significantly reduced in virulence, following the greater sensitivity of ΔcrpAΔcrpB to Cu than ΔaceA.


Assuntos
Aspergillus flavus/patogenicidade , ATPases Transportadoras de Cobre/metabolismo , Cobre/farmacologia , Proteínas Fúngicas/genética , Zea mays/microbiologia , Animais , Aspergilose/enzimologia , Aspergillus flavus/efeitos dos fármacos , Aspergillus fumigatus/efeitos dos fármacos , ATPases Transportadoras de Cobre/genética , Feminino , Deleção de Genes , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos ICR , Fatores de Transcrição/genética , Virulência , Zea mays/enzimologia
6.
Cell Rep ; 19(5): 1008-1021, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28467895

RESUMO

The Fenton-chemistry-generating properties of copper ions are considered a potent phagolysosome defense against pathogenic microbes, yet our understanding of underlying host/microbe dynamics remains unclear. We address this issue in invasive aspergillosis and demonstrate that host and fungal responses inextricably connect copper and reactive oxygen intermediate (ROI) mechanisms. Loss of the copper-binding transcription factor AceA yields an Aspergillus fumigatus strain displaying increased sensitivity to copper and ROI in vitro, increased intracellular copper concentrations, decreased survival in challenge with murine alveolar macrophages (AMΦs), and reduced virulence in a non-neutropenic murine model. ΔaceA survival is remediated by dampening of host ROI (chemically or genetically) or enhancement of copper-exporting activity (CrpA) in A. fumigatus. Our study exposes a complex host/microbe multifactorial interplay that highlights the importance of host immune status and reveals key targetable A. fumigatus counter-defenses.


Assuntos
Aspergillus/metabolismo , Cobre/metabolismo , Interações Hospedeiro-Patógeno , Espécies Reativas de Oxigênio/metabolismo , Animais , Aspergillus/genética , Aspergillus/patogenicidade , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Cultivadas , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , ATPases do Tipo-P/genética , ATPases do Tipo-P/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Virulência/genética
7.
Med Mycol ; 55(1): 118-124, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27562862

RESUMO

Aspergillosis has emerged as important human mycoses, in view of the ever expanding population at risk. The emergence of resistance to the most commonly used drugs for aspergillosis, the azoles, the mediocre activity, and frequent toxicity of the current antifungal armamentarium, support the need for development of novel antifungals for treatment of this disease. In this minireview, we describe recent efforts by small drug companies and University research labs to develop novel therapies for invasive aspergillus infections. We specifically discuss four small-molecule antifungals (T-2307, E1210/APX001, ASP2397, and F901318) with novel modes-of-action, which are currently entering phase I clinical trials. In addition, we provide a nonexhaustive discussion of some interesting, yet early developments in the quest for improved therapeutic strategies such as (i) novel formulations of amphotericin B including AMB nanoparticle suspensions and AMB-arabinogalactan or AMB-PEG conjugates that show low toxicity and high efficacy in preclinical animal models, (ii) repurposed drugs that synergize with existing antifungals (clozafimine, trichostatin A, MGCD290, geldanamycin, tacrolimus, cyclosporin), (iii) natural products (psoriasin, humidimycin), and (iv) immunotherapy using adoptive transfer of activated immune cells with antifungal activity. We argue that despite the plethora of candidates, the extremely low success rates of drug development leading to clinically useful drugs reinforces the need for continued clinical reliance on mainstream antifungals and their improved derivatives.


Assuntos
Antifúngicos/isolamento & purificação , Antifúngicos/farmacologia , Aspergilose/terapia , Aspergillus/efeitos dos fármacos , Aspergillus/imunologia , Descoberta de Drogas/tendências , Animais , Produtos Biológicos/isolamento & purificação , Produtos Biológicos/farmacologia , Ensaios Clínicos como Assunto , Composição de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Tratamento Farmacológico/métodos , Humanos , Imunoterapia/métodos
9.
Antimicrob Agents Chemother ; 56(1): 1-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22006001

RESUMO

Many natural broad-spectrum cationic antimicrobial peptides (AMPs) possess a general mode of action that is dependent on lipophilicity and charge. Modulating the lipophilicity of AMPs by the addition of a fatty acid has been an effective strategy to increase the lytic activity and can further broaden the spectrum of AMPs. However, lipophilic modifications that narrow the spectrum of activity and exclusively direct peptides to fungi are less common. Here, we show that short peptide sequences can be targeted to fungi with structured lipophilic biomolecules, such as vitamin E and cholesterol. The conjugates were active against Aspergillus fumigatus, Cryptococcus neoformans, and Candida albicans but not against bacteria and were observed to cause membrane perturbation by transmission electron microscopy and in membrane permeability studies. However, for C. albicans, selected compounds were effective without the perturbation of the cell membrane, and synergism was seen with a vitamin E conjugate and amphotericin B. Moreover, in combination with ß-cyclodextrin, antibacterial activity emerged in selected compounds. Biocompatibility for selected active compounds was tested in vitro and in vivo using toxicity assays on erythrocytes, macrophages, and mice. In vitro cytotoxicity experiments led to selective toxicity ratios (50% lethal concentration/MIC) of up to 64 for highly active antifungal compounds, and no in vivo murine toxicity was seen. Taken together, these results highlight the importance of the conjugated lipophilic structure and suggest that the modulation of other biologically relevant peptides with hydrophobic moieties, such as cholesterol and vitamin E, generate compounds with unique bioactivity.


Assuntos
Anfotericina B/farmacologia , Peptídeos Catiônicos Antimicrobianos/síntese química , Colesterol/química , Vitamina E/química , beta-Ciclodextrinas/farmacologia , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Aspergillus fumigatus/efeitos dos fármacos , Aspergillus fumigatus/crescimento & desenvolvimento , Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Candida albicans/efeitos dos fármacos , Candida albicans/crescimento & desenvolvimento , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Colesterol/farmacologia , Cryptococcus neoformans/efeitos dos fármacos , Cryptococcus neoformans/crescimento & desenvolvimento , Sinergismo Farmacológico , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Transmissão , Micoses/tratamento farmacológico , Micoses/microbiologia , Especificidade da Espécie , Eletricidade Estática , Vitamina E/farmacologia
10.
PLoS One ; 6(3): e17509, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21412410

RESUMO

Aspergillus fumigatus is the most commonly encountered mold pathogen of humans, predominantly infecting the respiratory system. Colonization and penetration of the lung alveolar epithelium is a key but poorly understood step in the infection process. This study focused on identifying the transcriptional and cell-signaling responses activated in A549 alveolar carcinoma cells incubated in the presence of A. fumigatus wild-type and ΔPrtT protease-deficient germinating conidia and culture filtrates (CF). Microarray analysis of exposed A549 cells identified distinct classes of genes whose expression is altered in the presence of germinating conidia and CF and suggested the involvement of both NFkB and MAPK signaling pathways in mediating the cellular response. Phosphoprotein analysis of A549 cells confirmed that JNK and ERK1/2 are phosphorylated in response to CF from wild-type A. fumigatus and not phosphorylated in response to CF from the ΔPrtT protease-deficient strain. Inhibition of JNK or ERK1/2 kinase activity substantially decreased CF-induced cell damage, including cell peeling, actin-cytoskeleton damage, and reduction in metabolic activity and necrotic death. These results suggest that inhibition of MAPK-mediated host responses to treatment with A. fumigatus CF decreases cellular damage, a finding with possible clinical implications.


Assuntos
Aspergillus fumigatus/metabolismo , Proteínas Fúngicas/metabolismo , Sistema de Sinalização das MAP Quinases , Necrose/patologia , Actinas/metabolismo , Aspergillus fumigatus/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Citoproteção/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Filtração , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mutação/genética , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeo Hidrolases/metabolismo , Polimerização/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Reprodutibilidade dos Testes , Esporos Fúngicos/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
11.
Infect Immun ; 77(9): 4051-60, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19564385

RESUMO

The role of secreted proteases in the virulence of the pathogenic fungus Aspergillus fumigatus remains controversial. Recently, the Aspergillus niger transcription factor PrtT was shown to control the expression of multiple secreted proteases. In this work, the gene which encodes the PrtT homolog in A. fumigatus was cloned and its function analyzed using a deletion mutant strain. Deletion of A. fumigatus prtT resulted in the loss of secreted protease activity. The expression of six secreted proteases (ALP, MEP, Dpp4, CpdS, AFUA_2G17330, and AFUA_7G06220) was markedly reduced. Culture filtrates from the prtT deletion strain exhibited reduced killing of lung epithelial cells and lysis of erythrocytes. However, the prtT deletion strain did not exhibit altered virulence in lung-infected mice. These results suggest that PrtT is not a significant virulence factor in A. fumigatus.


Assuntos
Aspergillus fumigatus/enzimologia , Proteínas Fúngicas/fisiologia , Peptídeo Hidrolases/metabolismo , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Animais , Aspergillus fumigatus/patogenicidade , Linhagem Celular Tumoral , Feminino , Hemólise , Humanos , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Peptídeo Hidrolases/genética , Transcrição Gênica , Virulência
12.
FEMS Yeast Res ; 7(6): 879-86, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17559411

RESUMO

We have previously reported that growth on alcohol vapors confers hemolytic properties on certain yeast species and strains ('microbial alcohol conferred hemolysis'; MACH). Here, a Saccharomyces cerevisiae deletion library consisting of c. 4800 clones was screened for MACH mutants in the presence of n-butanol vapors; 136 mutants were MACH-negative, and 325 exhibited reduced hemolysis and/or growth. Of the MACH-negative mutants, 35.3% were affected in mitochondrial-related genes. The data suggest that intact mitochondrial and respiratory chain functions are critical for the observed MACH phenomenon. We propose that the uncontrolled cellular uptake of alcohol results in yeast 'hyper-respiration', leading to elaboration of hemolytic molecules such as hydrogen peroxide and hemolysis-causing lipids. To support this premise, we showed that: (1) exogenous catalase and glutathione reduce alcohol-conferred hemolysis in S. cerevisiae BY4741 and Candida tropicalis 59445; (2) C. tropicalis produces hydrogen peroxide following growth on ethanol and n-butanol, as shown using xylenol orange; and (3) a lysophospholipid-containing lipid extract from alcohol-grown C. tropicalis specifically causes hemolysis.


Assuntos
Álcoois/farmacologia , Hemólise/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Catalase/metabolismo , Citocromos/análise , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação Fúngica da Expressão Gênica , Glutationa/metabolismo , Peróxido de Hidrogênio/toxicidade , Lisofosfolipídeos/metabolismo , Mutação/genética , Consumo de Oxigênio/efeitos dos fármacos , Fenóis , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Sulfóxidos , Xilenos/metabolismo
13.
J Antimicrob Chemother ; 57(2): 230-5, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16352735

RESUMO

OBJECTIVES: We have previously shown that moxifloxacin conferred protective anti-inflammatory effects against Candida pneumonia in immunosuppressed mice. Further in vitro studies showed anti-inflammatory effects of moxifloxacin in LPS and cytokine-stimulated monocytic and epithelial cells. In the present study, concentrating on a more challenging pathogen of immunosuppressed hosts, we studied the effect of moxifloxacin on cytokine secretion and signal transduction mechanisms in monocytic cells stimulated with Aspergillus fumigatus. METHODS: Human peripheral blood monocytes (PBMCs) and a human monocytic cell line (THP-1) were incubated with 1.5x10(6)/mL conidia of a clinical isolate of A. fumigatus. Cytokine secretion and activation of NFkappaB and the MAP-kinases ERK1/2 and p38 were measured with and without the addition of moxifloxacin (5-20 mg/L). RESULTS: Stimulation of PBMCs and THP-1 cells with A. fumigatus increased IL-8, IL-1beta and TNF-alpha secretion (4.1-, 8.3- and 7-fold, and 5.4-, 3.7- and 17.8-fold, respectively). Addition of moxifloxacin (5-20 mg/L) inhibited cytokine secretion up to 45.7+/-5%, 72+/-13% and 73+/-10% in PBMCs and up to 35.6+/-0.5%, 30+/-2.4% and 19+/-4% in THP-1 cells (P<0.05). Signal transduction studies showed that incubation of THP-1 cells with A. fumigatus increased ERK1/2 and p38 phosphorylation and p65-NFkappaB protein expression by 1.6-, 1.3- and 1.8-fold, respectively. Addition of moxifloxacin inhibited ERK1/2, p38 and p65-NFkappaB by up to 69+/-14%, 58+/-3% and 75+/-15%, respectively. CONCLUSIONS: Our results indicate that moxifloxacin acts as an anti-inflammatory agent in monocytic cells stimulated with A. fumigatus conidia. Whether these effects may be protective as in the Candida pneumonia model is unknown and merits in vivo studies in models of pulmonary aspergillosis.


Assuntos
Antibacterianos/farmacologia , Anti-Inflamatórios não Esteroides , Aspergillus fumigatus/fisiologia , Compostos Aza/farmacologia , Interleucina-1/metabolismo , Interleucina-8/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/metabolismo , NF-kappa B/metabolismo , Quinolinas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Aspergilose/microbiologia , Western Blotting , Ativação Enzimática/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Fluoroquinolonas , Humanos , Terapia de Imunossupressão , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Moxifloxacina , Proteína Oncogênica p65(gag-jun)/genética , Proteína Oncogênica p65(gag-jun)/fisiologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Antimicrob Agents Chemother ; 48(9): 3279-83, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15328085

RESUMO

Caspofungin (CAS) inhibits fungal cell wall synthesis. Sulfamethoxazole (SMX) inhibits folate biosynthesis and is active in vitro against Aspergillus spp. We studied the activities of the combination of CAS and SMX against 31 Aspergillus isolates and compared them with that of SMX combined with amphotericin B (AMB) or itraconazole (ITC). MICs and minimal effective concentrations (MECs) were determined by the NCCLS broth microdilution method. With MIC endpoints, the combination of SMX and CAS showed synergy or synergy to additivity against 29 of 31 isolates. With MEC endpoints, synergy to additivity was found against 12 of 31 isolates and indifference was displayed against the rest of them. SMX in combination with AMB or ITC was not truly synergistic, while synergy to additivity was found for SMX-AMB and SMX-ITC against 17 of 31 and 3 of 12 isolates, respectively. No antagonism was found with any of the drug combinations. Further analysis of the synergy of CAS and SMX was performed by detailed measurement of hyphal length by microscopy and time-dependent 2,3-bis(2-methoxy-4-nitro-5-[(sulfenylamino)carbonyl]-2H-tetrazolium hydroxide (XTT)-based hyphal damage experiments. With MEC endpoints, the combination of CAS and SMX was characterized by a greater than 50% decrease in hyphal length compared to the hyphal lengths achieved with double the concentration of each drug alone. The XTT-based hyphal damage studies showed a statistically significant (P < 0.05) reduction in viability with CAS and SMX in combination compared to the viabilities achieved with double the concentration of each drug alone. These findings support the synergy results found by using MIC endpoints and suggest that visual MEC measurements may not be sufficient to identify the synergistic interactions seen by more sensitive, quantitative methods. Animal models are required to validate the significance of the synergy of CAS and SMX against Aspergillus spp. observed in vitro.


Assuntos
Antifúngicos/farmacologia , Aspergilose/microbiologia , Aspergillus/efeitos dos fármacos , Peptídeos Cíclicos , Peptídeos/farmacologia , Sulfametoxazol/farmacologia , Anfotericina B/farmacologia , Aspergillus/ultraestrutura , Caspofungina , Combinação de Medicamentos , Sinergismo Farmacológico , Equinocandinas , Corantes Fluorescentes , Humanos , Hifas/efeitos dos fármacos , Hifas/ultraestrutura , Itraconazol/farmacologia , Lipopeptídeos , Testes de Sensibilidade Microbiana , Microscopia de Fluorescência , Sais de Tetrazólio
15.
J Infect Dis ; 189(11): 1965-73, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15143461

RESUMO

Aspergillus fumigatus is an opportunistic pathogenic fungus that predominantly infects the respiratory system. Penetration of the lung alveolar epithelium is a key step in the infectious process. The cytoskeleton of alveolar epithelial cells forms the cellular basis for the formation of a physical barrier between the cells and their surroundings. This study focused on the distinct effects of A. fumigatus on the actin cytoskeleton of A549 lung pneumocytes. Of the 3 major classes of cytoskeletal fibers--actin microfilaments, microtubules, and intermediate filaments--only the actin cytoskeleton was found to undergo major structural changes in response to infection, including loss of actin stress fibers, formation of actin aggregates, disruption of focal adhesion sites, and cell blebbing. These changes could be specifically blocked in wild-type strains of A. fumigatus by the addition of antipain, a serine and cysteine protease inhibitor, and were not induced by an alkaline serine protease-deficient strain of A. fumigatus. Antipain also reduced, by approximately 50%, fungal-induced A549 cell detachment from the plates and reduction in viability. Our findings suggest that A. fumigatus breaches the alveolar epithelial cell barrier by secreting proteases that act together to disorganize the actin cytoskeleton and destroy cell attachment to the substrate by disrupting focal adhesions.


Assuntos
Actinas/metabolismo , Aspergilose/microbiologia , Aspergillus fumigatus/enzimologia , Citoesqueleto/metabolismo , Endopeptidases/metabolismo , Antipaína/farmacologia , Aspergillus fumigatus/fisiologia , Benzenossulfonatos/química , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Formazans/química , Humanos , Imuno-Histoquímica , Pulmão/microbiologia , Pulmão/ultraestrutura , Microscopia Confocal , Microtúbulos , Inibidores de Proteases/farmacologia , Vinculina/fisiologia
16.
Antimicrob Agents Chemother ; 47(4): 1416-8, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12654681

RESUMO

Caspofungin and itraconazole were studied alone and in combination against 31 clinical isolates of Aspergillus spp. according to NCCLS M38-P guidelines. MICs and microscopic minimal effective concentrations (MECs) were recorded, and synergy was calculated by using both end points. Synergy or synergy to additivity was found in 30 of 31 isolates by using MIC end points. With MEC end points no synergy was found and indifference was detected in 26 of 31 strains.


Assuntos
Antibacterianos/farmacologia , Antifúngicos/farmacologia , Aspergillus/efeitos dos fármacos , Itraconazol/farmacologia , Peptídeos Cíclicos , Peptídeos , Caspofungina , Sinergismo Farmacológico , Equinocandinas , Lipopeptídeos , Testes de Sensibilidade Microbiana
17.
J Antimicrob Chemother ; 51(2): 313-6, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12562696

RESUMO

Work in Saccharomyces cerevisiae and Cryptococcus neoformans suggests that caspofungin could interact with the calcineurin pathway. We examined the in vitro interaction of caspofungin with calcineurin inhibitors (FK506, cyclosporin, FK520 and L685,818) and the TOR inhibitor rapamycin in 13 isolates of Aspergillus species. Caspofungin activity was enhanced by calcineurin/TOR inhibitors for all Aspergillus isolates studied. Further investigation of this pathway is warranted.


Assuntos
Antibacterianos/farmacologia , Antifúngicos/farmacologia , Aspergillus/efeitos dos fármacos , Aspergillus/crescimento & desenvolvimento , Inibidores de Calcineurina , Peptídeos Cíclicos , Peptídeos , Tacrolimo/análogos & derivados , Caspofungina , Combinação de Medicamentos , Equinocandinas , Glucosiltransferases/genética , Imunossupressores/farmacologia , Lipopeptídeos , Proteínas de Membrana/genética , Testes de Sensibilidade Microbiana , Saccharomyces cerevisiae/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/genética , Sirolimo/farmacologia , Tacrolimo/farmacologia
18.
Antimicrob Agents Chemother ; 46(8): 2462-9, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12121919

RESUMO

Caspofungin inhibits the synthesis of 1, 3-beta-D-glucan, an essential cell wall target in fungi. Genetic studies in the model yeast Saccharomyces cerevisiae have shown that mutations in FKS1 and FKS2 genes result in caspofungin resistance. However, direct demonstration of the role of gene overexpression in caspofungin resistance has been lacking. We transformed wild-type S. cerevisiae with an S. cerevisiae URA3-based GAL1 cDNA library and selected transformants in glucose synthetic complete plates lacking uracil (glucose SC minus uracil plates). We then moved the transformants to galactose SC minus uracil plates containing caspofungin (1 microg/ml) and looked for caspofungin-resistant colonies. We retested the candidates (true positives were sensitive on glucose caspofungin and resistant on galactose caspofungin media, respectively). We identified 16 caspofungin-resistant candidates. Restriction analysis and hybridization confirmed that 15 of the 16 clones were identical. We sequenced one of the cDNA clones and found that it contained the cDNA for SBE2. SBE2 has been described in S. cerevisiae to encode a Golgi protein involved in the transport of cell wall components (B. Santos and M. Snyder, Mol. Biol. Cell, 11:435-452, 2000). The SBE2 cDNA plasmid conferred again galactose-dependent caspofungin resistance when transformed back into the wild-type S. cerevisiae. Finally, the SBE2 deletion mutant was hypersensitive to caspofungin. In conclusion, overexpression of Sbe2p under the regulated control of the GAL1 promoter results in caspofungin resistance in S. cerevisiae. This transport pathway may provide insight into the tolerance or lack of sensitivity to caspofungin of some pathogenic fungi.


Assuntos
Antibacterianos/farmacologia , Antifúngicos/farmacologia , Proteínas Fúngicas/biossíntese , Complexo de Golgi/metabolismo , Glicoproteínas de Membrana/biossíntese , Peptídeos Cíclicos , Peptídeos , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Caspofungina , Parede Celular/efeitos dos fármacos , Parede Celular/ultraestrutura , DNA Fúngico/genética , Resistência Microbiana a Medicamentos/genética , Equinocandinas , Galactose/metabolismo , Glucose/metabolismo , Lipopeptídeos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica , Microscopia de Contraste de Fase , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Transporte Vesicular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA