Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mSphere ; 3(2)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29600281

RESUMO

Candida glabrata is the second most common species causing candidiasis. C. glabrata can also readily acquire resistance to azole drugs, complicating its treatment. Here we add to the collection of disruption markers to aid in genetic analysis of this yeast. This new construct is marked with a nourseothricin resistance cassette that produces an estrogen-activated form of Cre recombinase in a methionine-regulated manner. This allows eviction and reuse of this cassette in a facile manner. Using this new disruption marker, we have constructed a series of strains lacking different members of the major facilitator superfamily (MFS) of membrane transporter proteins. The presence of 15 MFS proteins that may contribute to drug resistance in C. glabrata placed a premium on development of a marker that could easily be reused to construct multiple gene-disrupted strains. Employing this recyclable marker, we found that loss of the MFS transporter-encoding gene FLR1 caused a dramatic increase in diamide resistance (as seen before), and deletion of two other MFS-encoding genes did not influence this phenotype. Interestingly, loss of FLR1 led to an increase in levels of oxidized glutathione, suggesting a possible molecular explanation for this enhanced oxidant sensitivity. We also found that while overproduction of the transcription factor Yap1 could suppress the fluconazole sensitivity caused by loss of the important ATP-binding cassette transporter protein Cdr1, this required the presence of FLR1. IMPORTANCE Export of drugs is a problem for chemotherapy of infectious organisms. A class of membrane proteins called the major facilitator superfamily contains a large number of proteins that often elevate drug resistance when overproduced but do not impact this phenotype when the gene is removed. We wondered if this absence of a phenotype for a disruption allele might be due to the redundancy of this group of membrane proteins. We describe the production of an easy-to-use recyclable marker cassette that will allow construction of strains lacking multiple members of the MFS family of transporter proteins.


Assuntos
Candida glabrata/genética , Farmacorresistência Fúngica/genética , Proteínas Fúngicas/genética , Transportadores de Ânions Orgânicos/genética , Transportadores de Cassetes de Ligação de ATP/genética , Antifúngicos/farmacologia , Candida glabrata/efeitos dos fármacos , Fluconazol/farmacologia , Regulação Fúngica da Expressão Gênica , Técnicas de Inativação de Genes , Engenharia Genética , Marcadores Genéticos , Glutationa/metabolismo , Integrases/genética , Metionina/metabolismo , Testes de Sensibilidade Microbiana , Fenótipo , Estreptotricinas/farmacologia , Fatores de Transcrição/genética
2.
mSphere ; 1(3)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27303750

RESUMO

Staphylococcus aureus diseases affect ~500,000 individuals per year in the United States. Worldwide, the USA100, USA200, USA400, and USA600 lineages cause many of the life-threatening S. aureus infections, such as bacteremia, infective endocarditis, pneumonia, toxic shock syndrome, and surgical site infections. However, the virulence mechanisms associated with these clonal lineages, in particular the USA100 and USA600 isolates, have been severely understudied. We investigated the virulence of these strains, in addition to strains in the USA200, USA300, and USA400 types, in well-established in vitro assays and in vivo in the rabbit model of infective endocarditis and sepsis. We show in the infective endocarditis and sepsis model that strains in the USA100 and USA600 lineages cause high lethality and are proficient in causing native valve infective endocarditis. Strains with high cytolytic activity or producing toxic shock syndrome toxin 1 (TSST-1) or staphylococcal enterotoxin C (SEC) caused lethal sepsis, even with low cytolytic activity. Strains in the USA100, USA200, USA400, and USA600 lineages consistently contained genes that encode for the enterotoxin gene cluster proteins, SEC, or TSST-1 and were proficient at causing infective endocarditis, while the USA300 strains lacked these toxins and were deficient in promoting vegetation growth. The USA100, USA200, and USA400 strains in our collection formed strong biofilms in vitro, whereas the USA200 and USA600 strains exhibited increased blood survival. Hence, infective endocarditis and lethal sepsis are multifactorial and not intrinsic to any one individual clonal group, further highlighting the importance of expanding our knowledge of S. aureus pathogenesis to clonal lineages causative of invasive disease. IMPORTANCE S. aureus is the leading cause of infective endocarditis in the developed world, affecting ~40,000 individuals each year in the United States, and the second leading cause of bacteremia (D. R. Murdoch et al., Arch Intern Med 169:463-473, 2009, http://dx.doi.org/10.1001/archinternmed.2008.603, and H. Wisplinghoff et al., Clin Infect Dis 39:309-317, 2004, http://dx.doi.org/10.1086/421946). Even with current medical advances, S. aureus bloodstream infections and infective endocarditis carry mortality rates of 20 to 66% (S. Y. Tong et al., Clin Microbiol Rev 28:603-661, 2015, http://dx.doi.org/10.1128/CMR.00134-14). S. aureus lineages associated with human disease worldwide include clonal complex 5 (CC5)/USA100, CC30/USA200, CC8/USA300, CC1/USA400, and CC45/USA600. The CC5/USA100, CC30/USA200, and CC45/USA600 lineages cause invasive disease yet remain poorly characterized. USA300 and cytotoxins are central to most S. aureus virulence studies, and yet, we find evidence that clonal groups are quite heterogeneous in parameters canonically used to measure virulence, including cytotoxicity, biofilm formation, and blood survival, and that the superantigen profile is an important parameter to consider when defining the virulence of S. aureus strains.

3.
PLoS One ; 11(4): e0154762, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27124393

RESUMO

BACKGROUND: Superantigens are indispensable virulence factors for Staphylococcus aureus in disease causation. Superantigens stimulate massive immune cell activation, leading to toxic shock syndrome (TSS) and contributing to other illnesses. However, superantigens differ in their capacities to induce body-wide effects. For many, their production, at least as tested in vitro, is not high enough to reach the circulation, or the proteins are not efficient in crossing epithelial and endothelial barriers, thus remaining within tissues or localized on mucosal surfaces where they exert only local effects. In this study, we address the role of TSS toxin-1 (TSST-1) and most importantly the enterotoxin gene cluster (egc) in infective endocarditis and sepsis, gaining insights into the body-wide versus local effects of superantigens. METHODS: We examined S. aureus TSST-1 gene (tstH) and egc deletion strains in the rabbit model of infective endocarditis and sepsis. Importantly, we also assessed the ability of commercial human intravenous immunoglobulin (IVIG) plus vancomycin to alter the course of infective endocarditis and sepsis. RESULTS: TSST-1 contributed to infective endocarditis vegetations and lethal sepsis, while superantigens of the egc, a cluster with uncharacterized functions in S. aureus infections, promoted vegetation formation in infective endocarditis. IVIG plus vancomycin prevented lethality and stroke development in infective endocarditis and sepsis. CONCLUSIONS: Our studies support the local tissue effects of egc superantigens for establishment and progression of infective endocarditis providing evidence for their role in life-threatening illnesses. In contrast, TSST-1 contributes to both infective endocarditis and lethal sepsis. IVIG may be a useful adjunct therapy for infective endocarditis and sepsis.


Assuntos
Toxinas Bacterianas/genética , Endocardite Bacteriana/microbiologia , Enterotoxinas/genética , Sepse/microbiologia , Choque Séptico/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Superantígenos/genética , Animais , Antibacterianos/uso terapêutico , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Masculino , Coelhos , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Vancomicina/uso terapêutico
4.
Biochemistry ; 55(17): 2510-7, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27015018

RESUMO

ß-Toxin is an important virulence factor of Staphylococcus aureus, contributing to colonization and development of disease [Salgado-Pabon, W., et al. (2014) J. Infect. Dis. 210, 784-792; Huseby, M. J., et al. (2010) Proc. Natl. Acad. Sci. U.S.A. 107, 14407-14412; Katayama, Y., et al. (2013) J. Bacteriol. 195, 1194-1203]. This cytotoxin has two distinct mechanisms of action: sphingomyelinase activity and DNA biofilm ligase activity. However, the distinct mechanism that is most important for its role in infective endocarditis is unknown. We characterized the active site of ß-toxin DNA biofilm ligase activity by examining deficiencies in site-directed mutants through in vitro DNA precipitation and biofilm formation assays. Possible conformational changes in mutant structure compared to that of wild-type toxin were assessed preliminarily by trypsin digestion analysis, retention of sphingomyelinase activity, and predicted structures based on the native toxin structure. We addressed the contribution of each mechanism of action to producing infective endocarditis and sepsis in vivo in a rabbit model. The H289N ß-toxin mutant, lacking sphingomyelinase activity, exhibited lower sepsis lethality and infective endocarditis vegetation formation compared to those of the wild-type toxin. ß-Toxin mutants with disrupted biofilm ligase activity did not exhibit decreased sepsis lethality but were deficient in infective endocarditis vegetation formation compared to the wild-type protein. Our study begins to characterize the DNA biofilm ligase active site of ß-toxin and suggests ß-toxin functions importantly in infective endocarditis through both of its mechanisms of action.


Assuntos
Toxinas Bacterianas/efeitos adversos , Biofilmes/efeitos dos fármacos , Endocardite/etiologia , Proteínas Hemolisinas/efeitos adversos , Ligases/deficiência , Sepse/etiologia , Esfingomielina Fosfodiesterase/deficiência , Staphylococcus aureus/enzimologia , Sequência de Aminoácidos , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Biofilmes/crescimento & desenvolvimento , Endocardite/enzimologia , Endocardite/patologia , Feminino , Proteínas Hemolisinas/química , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Masculino , Conformação Proteica , Coelhos , Sepse/enzimologia , Sepse/patologia , Esfingomielina Fosfodiesterase/efeitos adversos , Esfingomielina Fosfodiesterase/química , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo , Infecções Estafilocócicas/complicações , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética
5.
PLoS One ; 10(11): e0143341, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26599862

RESUMO

Superantigens (SAgs) are important virulence factors in S. aureus. Recent studies identified their presence in animal coagulase-negative staphylococci (CNS). The emergence of human-associated SAg+ CNS would mark a prodigious shift in virulence capabilities. We examined CNS isolates from healthy human nares and diseased individuals, and determined that no known SAgs were present.


Assuntos
Staphylococcus aureus/imunologia , Superantígenos/análise , Voluntários Saudáveis , Humanos , Staphylococcus aureus/isolamento & purificação , Superantígenos/imunologia , Fatores de Virulência/análise , Fatores de Virulência/imunologia
6.
mBio ; 6(2): e02554, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25714716

RESUMO

UNLABELLED: Excessive weight and obesity are associated with the development of diabetes mellitus type 2 (DMII) in humans. They also pose high risks of Staphylococcus aureus colonization and overt infections. S. aureus causes a wide range of severe illnesses in both healthy and immunocompromised individuals. Among S. aureus virulence factors, superantigens are essential for pathogenicity. In this study, we show that rabbits that are chronically exposed to S. aureus superantigen toxic shock syndrome toxin-1 (TSST-1) experience impaired glucose tolerance, systemic inflammation, and elevated endotoxin levels in the bloodstream, all of which are common findings in DMII. Additionally, such DMII-associated findings are also seen through effects of TSST-1 on isolated adipocytes. Collectively, our findings suggest that chronic exposure to S. aureus superantigens facilitates the development of DMII, which may lead to therapeutic targeting of S. aureus and its superantigens. IMPORTANCE: Obesity has a strong correlation with type 2 diabetes, in which fatty tissue, containing adipocytes, contributes to the development of the illness through altered metabolism and chronic inflammation. The human microbiome changes in persons with obesity and type 2 diabetes, including increases in Staphylococcus aureus colonization and overt infections. While the microbiome is essential for human wellness, there is little understanding of the role of microbes in obesity or the development of diabetes. Here, we demonstrate that the S. aureus superantigen toxic shock syndrome toxin-1 (TSST-1), an essential exotoxin in pathogenesis, induces inflammation, lipolysis, and insulin resistance in adipocytes both in vitro and in vivo. Chronic stimulation of rabbits with TSST-1 results in impaired systemic glucose tolerance, the hallmark finding in type 2 diabetes in humans, suggesting a role of S. aureus and its superantigens in the progression to type 2 diabetes.


Assuntos
Toxinas Bacterianas/sangue , Diabetes Mellitus Tipo 2/etiologia , Endotoxinas/sangue , Enterotoxinas/sangue , Inflamação/patologia , Infecções Estafilocócicas/complicações , Superantígenos/sangue , Animais , Diabetes Mellitus Tipo 2/fisiopatologia , Enterotoxinas/metabolismo , Teste de Tolerância a Glucose , Coelhos
8.
J Infect Dis ; 210(12): 1920-7, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24951827

RESUMO

BACKGROUND: Diabetic foot ulcer (DFU) infections are challenging. Staphylococcus aureus is the most commonly isolated pathogen in DFUs. Superantigens (SAgs) are causative in many S. aureus infections. We hypothesized both that DFU S. aureus will produce large SAg numbers, consistent with skin infections, and that certain SAgs will be overrepresented. We assessed the SAg and α-toxin profile of isolates from patients with DFU, compared with profiles of isolates from other sources. MATERIALS: Twenty-five S. aureus isolates from patients with DFU were characterized. Polymerase chain reaction was used to detect genes for methicillin-resistance and SAgs. Some SAgs and the α-toxin were quantified. We compared the SAg profile of DFU isolates with SAg profiles of S. aureus isolates from skin lesions of patients with atopic dermatitis and from vaginal mucosa of healthy individuals. RESULTS: Most DFU isolates were methicillin susceptible (64%), with USA100 the most common clonal group. The SAg gene profile of DFU isolates most closely resembled that of isolates from patients with atopic dermatitis, with the highest number of different SAg genes per isolate and a high prevalence of staphylococcal enterotoxin D and the enterotoxin gene cluster. DFU isolates also had a high prevalence of staphylococcal enterotoxin-like X. CONCLUSIONS: Comparison of the SAg profile of DFU isolates to SAg profiles of skin lesion isolates and vaginal mucosa isolates revealed that the SAg profile of DFU isolates was more similar to that of skin lesion isolates. SAgs offer selective advantages in facilitating DFU infections and suggest that therapies to neutralize or reduce SAg production by S. aureus may be beneficial in management of patients with DFU.


Assuntos
Pé Diabético/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Superantígenos/genética , Toxinas Bacterianas/análise , Toxinas Bacterianas/genética , Feminino , Proteínas Hemolisinas/análise , Proteínas Hemolisinas/genética , Humanos , Masculino , Reação em Cadeia da Polimerase , Staphylococcus aureus/química , Staphylococcus aureus/isolamento & purificação , Superantígenos/análise , Úlcera/microbiologia
9.
J Infect Dis ; 210(5): 784-92, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24620023

RESUMO

BACKGROUND: Staphylococcus aureus causes life-threatening infections, including infective endocarditis, sepsis, and pneumonia. ß-toxin is a sphingomyelinase encoded for by virtually all S. aureus strains and exhibits human immune cell cytotoxicity. The toxin enhances S. aureus phenol-soluble modulin activity, and its activity is enhanced by superantigens. The bacteriophage φSa3 inserts into the ß-toxin gene in human strains, inactivating it in the majority of S. aureus clonal groups. Hence, most strains are reported not to secrete ß-toxin. METHODS: This dynamic was investigated by examining ß-toxin production by multiple clonal groups of S. aureus, both in vitro and in vivo during infections in rabbit models of infective endocarditis, sepsis, and pneumonia. RESULTS: ß-toxin phenotypic variants are common among strains containing φSa3. In vivo, φSa3 is differentially induced in heart vegetations, kidney abscesses, and ischemic liver compared to spleen and blood, and in vitro growth in liquid culture. Furthermore, in pneumonia, wild-type ß-toxin production leads to development of large caseous lesions, and in infective endocarditis, increases the size of pathognomonic vegetations. CONCLUSIONS: This study demonstrates the dynamic interaction between S. aureus and the infected host, where φSa3 serves as a regulator of virulence gene expression, and increased fitness and virulence in new environments.


Assuntos
Inativação Gênica , Proteínas Hemolisinas/metabolismo , Prófagos/genética , Esfingomielina Fosfodiesterase/metabolismo , Fagos de Staphylococcus/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus/virologia , Animais , Toxinas Bacterianas/genética , Modelos Animais de Doenças , Endocardite Bacteriana/microbiologia , Endocardite Bacteriana/patologia , Proteínas Hemolisinas/genética , Mutagênese Insercional , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/patologia , Coelhos , Recombinação Genética , Sepse/microbiologia , Sepse/patologia , Esfingomielina Fosfodiesterase/genética , Staphylococcus aureus/genética
10.
PLoS One ; 8(10): e77988, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24205055

RESUMO

BACKGROUND: Human adipocytes may have significant functions in wound healing and the development of diabetes through production of pro-inflammatory cytokines after stimulation by gram-negative bacterial endotoxin. Diabetic foot ulcers are most often associated with staphylococcal infections. Adipocyte responses in the area of the wound may play a role in persistence and pathology. We studied the effect of staphylococcal superantigens (SAgs) on immortalized human adipocytes, alone and in the presence of bacterial endotoxin or staphylococcal α-toxin. METHODOLOGY/PRINCIPAL FINDINGS: Primary non-diabetic and diabetic human preadipocytes were immortalized by the reverse transcriptase component of telomerase (TERT) and the E6/E7 genes of human papillomavirus. The immortal cells were demonstrated to have properties of non-immortalized pre-adipocytes and could be differentiated into mature and functional adipocytes. Differentiated adipocytes exposed to staphylococcal SAgs produced robust levels of cytokines IL-6 and IL-8, but there were no significant differences in levels between the non-diabetic and diabetic cells. Cytokine production was increased by co-incubation of adipocytes with SAgs and endotoxin together. In contrast, α-toxin alone was cytotoxic at high concentrations, but, at sub-cytotoxic doses, did not stimulate production of IL-6 and IL-8. CONCLUSIONS/SIGNIFICANCE: Endotoxin has been proposed to contribute to diabetes through enhanced insulin resistance after chronic exposure and stimulation of adipocytes to produce cytokines. Our data indicate staphylococcal SAgs TSST-1 and SEB alone and in combination with bacterial endotoxin also stimulate adipocytes to produce cytokines and thus may contribute to the inflammatory response found in chronic diabetic ulcers and in the systemic inflammation that is associated with the development and persistence of diabetes. The immortal human pre-adipocytes reported here will be useful for studies to understand further the mechanism by which toxins are involved in wound healing and the development and clinical manifestations of obesity and diabetes.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Quimiocinas/metabolismo , Superantígenos/farmacologia , Toxinas Bacterianas/farmacologia , Células Cultivadas , Enterotoxinas/farmacologia , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Cicatrização/efeitos dos fármacos
11.
Antimicrob Agents Chemother ; 57(11): 5432-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23959313

RESUMO

Gram-positive bacteria cause serious human illnesses through combinations of cell surface and secreted virulence factors. We initiated studies with four of these organisms to develop novel topical antibacterial agents that interfere with growth and exotoxin production, focusing on menaquinone analogs. Menadione, 1,4-naphthoquinone, and coenzymes Q1 to Q3 but not menaquinone, phylloquinone, or coenzyme Q10 inhibited the growth and to a greater extent exotoxin production of Staphylococcus aureus, Bacillus anthracis, Streptococcus pyogenes, and Streptococcus agalactiae at concentrations of 10 to 200 µg/ml. Coenzyme Q1 reduced the ability of S. aureus to cause toxic shock syndrome in a rabbit model, inhibited the growth of four Gram-negative bacteria, and synergized with another antimicrobial agent, glycerol monolaurate, to inhibit S. aureus growth. The staphylococcal two-component system SrrA/B was shown to be an antibacterial target of coenzyme Q1. We hypothesize that menaquinone analogs both induce toxic reactive oxygen species and affect bacterial plasma membranes and biosynthetic machinery to interfere with two-component systems, respiration, and macromolecular synthesis. These compounds represent a novel class of potential topical therapeutic agents.


Assuntos
Antibacterianos/farmacologia , Bacillus anthracis/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Streptococcus agalactiae/efeitos dos fármacos , Streptococcus pyogenes/efeitos dos fármacos , Vitamina K 2/farmacologia , Administração Tópica , Animais , Bacillus anthracis/crescimento & desenvolvimento , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Membrana Celular/efeitos dos fármacos , Sinergismo Farmacológico , Exotoxinas/antagonistas & inibidores , Exotoxinas/metabolismo , Humanos , Lauratos/farmacologia , Monoglicerídeos/farmacologia , Coelhos , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Choque Séptico/tratamento farmacológico , Choque Séptico/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/metabolismo , Streptococcus agalactiae/crescimento & desenvolvimento , Streptococcus pyogenes/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA