Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35165181

RESUMO

Staphylococcus aureus is a foremost bacterial pathogen responsible for a vast array of human diseases. Staphylococcal superantigens (SAgs) constitute a family of exotoxins from S. aureus that bind directly to major histocompatibility complex (MHC) class II and T cell receptors to drive extensive T cell activation and cytokine release. Although these toxins have been implicated in serious disease, including toxic shock syndrome, the specific pathological mechanisms remain unclear. Herein, we aimed to elucidate how SAgs contribute to pathogenesis during bloodstream infections and utilized transgenic mice encoding human MHC class II to render mice susceptible to SAg activity. We demonstrate that SAgs contribute to S. aureus bacteremia by massively increasing bacterial burden in the liver, and this was mediated by CD4+ T cells that produced interferon gamma (IFN-γ) to high levels in a SAg-dependent manner. Bacterial burdens were reduced by blocking IFN-γ, phenocopying SAg-deletion mutant strains, and inhibiting a proinflammatory response. Infection kinetics and flow cytometry analyses suggested that this was a macrophage-driven mechanism, which was confirmed through macrophage-depletion experiments. Experiments in human cells demonstrated that excessive IFN-γ allowed S. aureus to replicate efficiently within macrophages. This indicates that SAgs promote bacterial survival by manipulating the immune response to inhibit effective clearing of S. aureus Altogether, this work implicates SAg toxins as critical therapeutic targets for preventing persistent or severe S. aureus disease.


Assuntos
Interferon gama/imunologia , Infecções Estafilocócicas/imunologia , Superantígenos/imunologia , Animais , Bacteriemia , Enterotoxinas/imunologia , Exotoxinas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/imunologia , Staphylococcus aureus/patogenicidade , Linfócitos T/imunologia , Fatores de Virulência/imunologia
2.
Cytotherapy ; 25(5): 490-501, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36781360

RESUMO

B-cell maturation antigen (BCMA) is a clinically validated target for multiple myeloma. T-cell engineered with chimeric antigen receptors (CARs) directed against BCMA have demonstrated robust therapeutic activity in clinical trials, but toxicities remain a significant concern for a subset of patients, supporting continued investigation of other engineered T-cell platforms that may offer equal efficacy with an improved toxicity profile. The authors recently described a BCMA-specific, T-cell-centric synthetic antigen receptor, the T-cell antigen coupler (TAC) receptor, that can be used to engineer T-cell with robust anti-myeloma activity. Here the authors describe the creation of a fully humanized BCMA-specific TAC receptor. Single-chain variable fragments (scFvs) were developed from BCMA-specific F(ab)s that were identified in a fully human phage display library. Twenty-four configurations of the F(ab)s were evaluated in a medium-throughput screening using primary T-cell, and a single F(ab), TRAC 3625, emerged as the most robust following in vitro and in vivo evaluation. An optimized BCMA-specific TAC receptor was developed through iterations of the BCMA-TAC design that evaluated a next-generation TAC scaffold sequence, different domains connecting the TAC to the 3625 scFv and different orientations of the TRAC 3625 heavy and light variable regions.


Assuntos
Mieloma Múltiplo , Linfócitos T , Humanos , Mieloma Múltiplo/terapia , Antígeno de Maturação de Linfócitos B , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T
3.
J Infect Dis ; 224(12 Suppl 2): S56-S63, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34396410

RESUMO

While infection by Neisseria gonorrhoeae is often asymptomatic in women, undetected infections can ascend into the upper genital tract to elicit an inflammatory response that manifests as pelvic inflammatory disease, with the outcomes depending on the intensity and duration of inflammation and whether it is localized to the endometrial, fallopian tube, ovarian, and/or other tissues. This review examines the contribution of N. gonorrhoeae versus other potential causes of pelvic inflammatory disease by considering new insights gained through molecular, immunological, and microbiome-based analyses, and the current epidemiological burden of infection, with an aim to highlighting key areas for future study.


Assuntos
Infecções por Chlamydia/epidemiologia , Gonorreia/epidemiologia , Neisseria gonorrhoeae/isolamento & purificação , Doença Inflamatória Pélvica/epidemiologia , Infecções por Chlamydia/complicações , Endometrite/microbiologia , Endométrio/microbiologia , Endométrio/patologia , Tubas Uterinas/microbiologia , Feminino , Gonorreia/diagnóstico , Humanos , Doença Inflamatória Pélvica/diagnóstico , Doença Inflamatória Pélvica/microbiologia
4.
Infect Immun ; 86(8)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29760215

RESUMO

The gonococcal Opa proteins are an antigenically variable family of surface adhesins that bind human carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), CEACAM3, CEACAM5, and/or CEACAM6, cell surface glycoproteins that are differentially expressed on a broad spectrum of human cells and tissues. While they are presumed to be important for infection, the significance of various Opa-CEACAM-mediated cellular interactions in the context of the genital tract has remained unclear. Here, we observed that CEACAM1 and CEACAM5 are differentially expressed on epithelia lining the upper and lower portions of the human female genital tract, respectively. Using transgenic mouse lines expressing human CEACAMs in a manner that reflects this differential pattern, we considered the impact of Opa-CEACAM interactions during uncomplicated lower genital tract infections versus during pelvic inflammatory disease. Our results demonstrate that Opa-CEACAM5 binding on vaginal epithelia facilitates the long-term colonization of the lower genital tract, while Opa protein binding to CEACAM1 on uterine epithelia enhances gonococcal association and penetration into these tissues. While these Opa-dependent interactions with CEACAM-expressing epithelial surfaces promote infection, Opa binding by neutrophil-expressed CEACAMs counterbalances this by facilitating more effective gonococcal clearance. Furthermore, during uterine infections, CEACAM-dependent tissue invasion aggravates disease pathology by increasing the acute inflammatory response. Together, these findings demonstrate that the outcome of infection is determined by both the cell type-specific expression of human CEACAMs and the CEACAM specificity of the Opa variants expressed, which combine to determine the level of gonococcal association with the genital mucosa versus the extent of CEACAM-dependent inflammation and gonococcal clearance by neutrophils.


Assuntos
Antígenos CD/metabolismo , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/metabolismo , Antígeno Carcinoembrionário/metabolismo , Moléculas de Adesão Celular/metabolismo , Genitália Feminina/patologia , Gonorreia/fisiopatologia , Infecções do Sistema Genital/fisiopatologia , Animais , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Proteínas Ligadas por GPI/metabolismo , Perfilação da Expressão Gênica , Genitália Feminina/microbiologia , Gonorreia/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neisseria gonorrhoeae/fisiologia , Infecções do Sistema Genital/microbiologia , Resultado do Tratamento , Útero/microbiologia , Útero/patologia , Vagina/microbiologia , Vagina/patologia
5.
J Infect Dis ; 215(5): 824-829, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28035011

RESUMO

During toxic shock syndrome (TSS), bacterial superantigens trigger a polyclonal T -cell response leading to a potentially catastrophic "cytokine storm". Whether innate-like invariant natural killer T (iNKT) cells, with remarkable immunomodulatory properties, participate in TSS is unclear. Using genetic and cell depletion approaches, we generated iNKT cell-deficient, superantigen-sensitive HLA-DR4-transgenic (DR4tg) mice, which were compared with their iNKT-sufficient counterparts for responsiveness to staphylococcal enterotoxin B (SEB). Both approaches indicate that iNKT cells are pathogenic in TSS. Importantly, treating DR4tg mice with a TH2-polarizing glycolipid agonist of iNKT cells reduced SEB-inflicted morbidity/mortality. Therefore, iNKT cells may constitute an attractive therapeutic target in superantigen-mediated illnesses.


Assuntos
Antígeno HLA-DR4/genética , Células T Matadoras Naturais/imunologia , Choque Séptico/imunologia , Choque Séptico/prevenção & controle , Animais , Modelos Animais de Doenças , Enterotoxinas/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Superantígenos/sangue , Superantígenos/imunologia
6.
J Bacteriol ; 198(19): 2732-42, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27457715

RESUMO

UNLABELLED: Toxic shock syndrome toxin 1 (TSST-1) is a Staphylococcus aureus superantigen that has been implicated in both menstrual and nonmenstrual toxic shock syndrome (TSS). Despite the important role of TSST-1 in severe human disease, a comprehensive understanding of staphylococcal regulatory factors that control TSST-1 expression remains incomplete. The S. aureus exotoxin expression (Sae) operon contains a well-characterized two-component system that regulates a number of important exotoxins in S. aureus, although regulation of TSST-1 by the Sae system has not been investigated. We generated a defined deletion mutant of the Sae histidine kinase sensor (saeS) in the prototypic menstrual TSS strain S. aureus MN8. Mutation of saeS resulted in a complete loss of TSST-1 expression. Using both luciferase reporter experiments and quantitative real-time PCR, we demonstrate that the Sae system is an important transcriptional activator of TSST-1 expression. Recombinant SaeR was able to bind directly to the tst promoter to a region containing two SaeR consensus binding sites. Although the stand-alone SarA transcriptional regulator has been shown to be both a positive and a negative regulator of TSST-1, deletion of sarA in S. aureus MN8 resulted in a dramatic overexpression of TSST-1. As expected, mutation of agr also reduced TSST-1 expression, but this phenotype appeared to be independent of Sae. A double mutation of saeS and sarA resulted in the loss of TSST-1 expression. This work indicates that the Sae system is a dominant and direct transcriptional activator that is required for expression of TSST-1. IMPORTANCE: The TSST-1 superantigen is an exotoxin, produced by some strains of S. aureus, that has a clear role in both menstrual and nonmenstrual TSS. Although the well-characterized agr quorum sensing system is a known positive regulator of TSST-1, the molecular mechanisms that directly control TSST-1 expression are only partially understood. Our studies demonstrate that the Sae two-component regulatory system is a positive transcriptional regulator that binds directly to the TSST-1 promoter, and furthermore, our data suggest that Sae is required for expression of TSST-1. This work highlights how major regulatory circuits can converge to fine-tune exotoxin expression and suggests that the Sae regulatory system may be an important target for antivirulence strategies.


Assuntos
Toxinas Bacterianas/metabolismo , Enterotoxinas/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas Quinases/metabolismo , Staphylococcus aureus/metabolismo , Superantígenos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Enterotoxinas/genética , Regiões Promotoras Genéticas , Proteínas Quinases/genética , Staphylococcus aureus/genética , Superantígenos/genética
7.
PLoS Pathog ; 10(5): e1004155, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24875883

RESUMO

Establishing the genetic determinants of niche adaptation by microbial pathogens to specific hosts is important for the management and control of infectious disease. Streptococcus pyogenes is a globally prominent human-specific bacterial pathogen that secretes superantigens (SAgs) as 'trademark' virulence factors. SAgs function to force the activation of T lymphocytes through direct binding to lateral surfaces of T cell receptors and class II major histocompatibility complex (MHC-II) molecules. S. pyogenes invariably encodes multiple SAgs, often within putative mobile genetic elements, and although SAgs are documented virulence factors for diseases such as scarlet fever and the streptococcal toxic shock syndrome (STSS), how these exotoxins contribute to the fitness and evolution of S. pyogenes is unknown. Here we show that acute infection in the nasopharynx is dependent upon both bacterial SAgs and host MHC-II molecules. S. pyogenes was rapidly cleared from the nasal cavity of wild-type C57BL/6 (B6) mice, whereas infection was enhanced up to ∼10,000-fold in B6 mice that express human MHC-II. This phenotype required the SpeA superantigen, and vaccination with an MHC -II binding mutant toxoid of SpeA dramatically inhibited infection. Our findings indicate that streptococcal SAgs are critical for the establishment of nasopharyngeal infection, thus providing an explanation as to why S. pyogenes produces these potent toxins. This work also highlights that SAg redundancy exists to avoid host anti-SAg humoral immune responses and to potentially overcome host MHC-II polymorphisms.


Assuntos
Proteínas de Bactérias/metabolismo , Exotoxinas/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Proteínas de Membrana/metabolismo , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Superantígenos/imunologia , Doença Aguda , Animais , Proteínas de Bactérias/imunologia , Exotoxinas/imunologia , Humanos , Proteínas de Membrana/imunologia , Camundongos Endogâmicos C57BL , Nasofaringe/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Infecções Estreptocócicas/prevenção & controle , Streptococcus pyogenes/genética , Superantígenos/genética , Linfócitos T/imunologia
8.
FASEB J ; 29(2): 711-23, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25416549

RESUMO

Staphylococcus aureus is a major component of the skin microbiota and causes a large number of serious infections. S. aureus first interacts with epidermal keratinocytes to breach the epidermal barrier through mechanisms not fully understood. By use of primary keratinocytes from mice with epidermis-restricted Ilk gene inactivation and control integrin-linked kinase (ILK)-expressing littermates, we investigated the role of ILK in epidermal S. aureus invasion. Heat-killed, but not live, bacteria were internalized to Rab5- and Rab7-positive phagosomes, and incubation with keratinocyte growth factor increased their uptake 2.5-fold. ILK-deficient mouse keratinocytes internalized bacteria 2- to 4-fold less efficiently than normal cells. The reduced invasion by live S. aureus of ILK-deficient cells was restored in the presence of exogenous, constitutively active Rac1. Thus, Rac1 functions downstream from ILK during invasion. Further, invasion by S. aureus of Rac1-deficient cells was 2.5-fold lower than in normal cells. Paradoxically, staphylococcal cutaneous penetration of mouse skin explants with ILK-deficient epidermis was 35-fold higher than that of normal skin, indicating defects in epidermal barrier function in the absence of ILK. Thus, we identified an ILK-Rac1 pathway essential for bacterial invasion of keratinocytes, and established ILK as a key contributor to prevent invasive staphylococcal cutaneous infection.


Assuntos
Queratinócitos/microbiologia , Neuropeptídeos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Staphylococcus aureus/patogenicidade , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Separação Celular , Epiderme/metabolismo , Fator 7 de Crescimento de Fibroblastos/metabolismo , Citometria de Fluxo , Gentamicinas/química , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Microbiota , Microscopia de Fluorescência , Fagocitose , Proteínas Recombinantes/metabolismo , Pele/microbiologia , Infecções Estafilocócicas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
9.
Infect Immun ; 82(9): 3588-98, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24914221

RESUMO

Staphylococcus aureus is a versatile bacterial pathogen that produces T cell-activating toxins known as superantigens (SAgs). Although excessive immune activation by SAgs can induce a dysregulated cytokine storm as a component of what is known as toxic shock syndrome (TSS), the contribution of SAgs to the staphylococcal infection process is not well defined. Here, we evaluated the role of the bacterial superantigen staphylococcal enterotoxin A (SEA) in a bacteremia model using humanized transgenic mice expressing SAg-responsive HLA-DR4 molecules. Infection with S. aureus Newman induced SEA-dependent Vß skewing of T cells and enhanced bacterial survival in the liver compared with infection by sea knockout strain. SEA-induced gamma interferon, interleukin-12, and chemokine responses resulted in increased infiltration of CD11b(+) Ly6G(+) neutrophils into the liver, promoting the formation of abscesses that contained large numbers of viable staphylococci. Hepatic abscesses occurred significantly more frequently in S. aureus Newman-infected livers than in livers infected with the Newman sea knockout strain, promoting the survival of S. aureus in vivo. This represents a novel mechanism during infection whereby S. aureus utilizes SAgs to form a specialized niche and manipulate the immune system.


Assuntos
Abscesso/imunologia , Neutrófilos/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Superantígenos/imunologia , Abscesso/microbiologia , Animais , Antígenos Ly/imunologia , Antígeno CD11b/imunologia , Enterotoxinas/imunologia , Antígeno HLA-DR4/imunologia , Interferon gama/imunologia , Interleucina-12/imunologia , Fígado/imunologia , Fígado/microbiologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Neutrófilos/microbiologia , Infecções Estafilocócicas/microbiologia , Linfócitos T/imunologia , Linfócitos T/microbiologia
10.
Proc Natl Acad Sci U S A ; 108(8): 3360-5, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21282650

RESUMO

The production of the staphylococcal exotoxin toxic shock syndrome toxin-1 (TSST-1) by Staphylococcus aureus has been associated with essentially all cases of menstruation-associated toxic shock syndrome (TSS). In this work, we show that the human vaginal isolate Lactobacillus reuteri RC-14 produces small signaling molecules that are able to interfere with the staphylococcal quorum-sensing system agr, a key regulator of virulence genes, and repress the expression of TSST-1 in S. aureus MN8, a prototype of menstrual TSS S. aureus strains. Quantitative real-time PCR data showed that transcription from the Ptst promoter, as well as the P2 and P3 promoters of the agr system from all four agr subgroups of S. aureus, was strongly inhibited in response to growth with L. reuteri RC-14 cultural supernatant. Alterations in the transcriptional levels of two other virulence-associated regulators sarA and saeRS were also observed, indicating a potential overall influence of L. reuteri RC-14 signals on the production of virulence factors in S. aureus. S. aureus promoter-lux reporter strains were used to screen biochemically fractionated L. reuteri RC-14 supernatant, and the cyclic dipeptides cyclo(L-Phe-L-Pro) and cyclo(L-Tyr-L-Pro) were identified as the signaling molecules. The results from this work contribute to a better understanding of interspecies cell-to-cell communication between Lactobacillus and Staphylococcus, and provide a unique mechanism by which endogenous or probiotic strains may attenuate virulence factor production by bacterial pathogens.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Toxinas Bacterianas/antagonistas & inibidores , Dipeptídeos/farmacologia , Enterotoxinas/antagonistas & inibidores , Limosilactobacillus reuteri/metabolismo , Choque Séptico/microbiologia , Staphylococcus aureus/patogenicidade , Transativadores/antagonistas & inibidores , Vagina/microbiologia , Proteínas de Bactérias/genética , Comunicação Celular , Dipeptídeos/biossíntese , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Menstruação , Probióticos , Percepção de Quorum , Superantígenos , Transativadores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA