Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(3): e1011272, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36972308

RESUMO

The signaling pathways and networks regulating expression of chondroitin sulfate proteoglycan 4 (CSPG4), a cancer-related protein that serves as a receptor for Clostridiodes difficile TcdB, are poorly defined. In this study, TcdB-resistant/CSPG4-negative HeLa cells were generated by exposure to increasing concentrations of the toxin. The cells that emerged (HeLa R5) lost expression of CSPG4 mRNA and were resistant to binding by TcdB. mRNA expression profiles paired with integrated pathway analysis correlated changes in the Hippo and estrogen signaling pathways with a CSPG4 decrease in HeLa R5 cells. Both signaling pathways altered CSPG4 expression when modulated chemically or through CRISPR-mediated deletion of key transcriptional regulators in the Hippo pathway. Based on the in vitro findings, we predicted and experimentally confirmed that a Hippo pathway inactivating drug (XMU-MP-1) provides protection from C. difficile disease in a mouse model. These results provide insights into key regulators of CSPG4 expression and identify a therapeutic for C. difficile disease.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Humanos , Animais , Camundongos , Clostridioides difficile/genética , Via de Sinalização Hippo , Toxinas Bacterianas/metabolismo , Células HeLa , Clostridioides , RNA Mensageiro/metabolismo , Proteínas de Membrana/metabolismo , Proteoglicanas de Sulfatos de Condroitina/metabolismo
2.
Curr Opin Microbiol ; 65: 81-86, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34773906

RESUMO

The autoinducing peptide-based Agr system in Clostridioides difficile is involved in virulence factor expression, motility, and sporulation. This review highlights several of the recent discoveries regarding C. difficile Agr. Typical Agr systems rely on the combined activities of four proteins involved in peptide expression, peptide processing, peptide sensing, and transcriptional regulation. As emphasized in this review, at least two C. difficile Agr systems (Agr1 and Agr3) lack the set of proteins associated with this regulatory network. In line with this, recent finding indicate Agr1 can function in ways that may not depend on accumulation of extracellular peptide. Also, described are the similarities and differences in Agr systems within the pathogenic Clostridia.


Assuntos
Clostridioides difficile , Clostridioides , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Clostridioides difficile/genética , Regulação Bacteriana da Expressão Gênica , Peptídeos/metabolismo , Percepção de Quorum , Fatores de Virulência/genética
3.
Sci Rep ; 8(1): 16931, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30446701

RESUMO

CREB and C/EBP ß signaling pathways are modulated during inflammation and also targeted by Bacillus anthracis edema toxin (ET), but how these factors individually and jointly contribute to changes in immune cell function is poorly understood. Using CRISPR/Cas9 gene editing, macrophage cell lines lacking CREB and isoforms of C/EBP ß were generated and analyzed for changes in responses to LPS, ET, and IL-4. Macrophages lacking C/EBP ß suppressed induction of IL-10 and Arg1, while IL-6 was increased in these cells following exposure to LPS. Examination of C/EBP ß isoforms indicated the 38 kDa isoform was necessary for the expression of IL-10 and Arg1. ChIP-Seq analysis of CREB and C/EBP ß binding to targets on the chromosome of human PBMC identified several regions where both factors overlapped in their binding, suggesting similar gene targeting or cooperative effects. Based on the ChIP-Seq data, a panel of previously unknown targets of CREB and C/EBP ß was identified and includes genes such as VNN2, GINS4, CTNNBL1, and SULF2. Isoforms of a transcriptional corepressor, transducin-like enhancer of Split (TLE), were also found to have CREB and C/EBP ß binding their promoter and were up regulated by ET. Finally, we explore a possible layer of C/EBP ß regulation by a protein complex consisting of adenomatous polyposis coli (APC) and PKA. Collectively, these data provide new insights into the role of CREB and C/EBP ß as immunosignaling regulators and targets of an important bacterial virulence factor.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Imunidade Inata , Leucócitos/imunologia , Leucócitos/metabolismo , Animais , Linhagem Celular , AMP Cíclico , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Imunomodulação/genética , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Células RAW 264.7
4.
Front Immunol ; 9: 3064, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30697210

RESUMO

Memory B cells that are generated during an infection or following vaccination act as sentinels to guard against future infections. Upon repeat antigen exposure memory B cells differentiate into new antibody-secreting plasma cells to provide rapid and sustained protection. Some pathogens evade or suppress the humoral immune system, or induce memory B cells with a diminished ability to differentiate into new plasma cells. This leaves the host vulnerable to chronic or recurrent infections. Single cell approaches coupled with next generation antibody gene sequencing facilitate a detailed analysis of the pathogen-specific memory B cell repertoire. Monoclonal antibodies that are generated from antibody gene sequences allow a functional analysis of the repertoire. This review discusses what has been learned thus far from analysis of diverse pathogen-specific memory B cell compartments and describes major differences in their repertoires. Such information may illuminate ways to advance the goal of improving vaccine and therapeutic antibody design.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Memória Imunológica , Plasmócitos/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Antígenos/imunologia , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Humoral/imunologia , Plasmócitos/metabolismo
5.
mBio ; 8(3)2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28512094

RESUMO

Clostridium difficile infection (CDI) is a major cause of hospital-associated, antibiotic-induced diarrhea, which is largely mediated by the production of two large multidomain clostridial toxins, TcdA and TcdB. Both toxins coordinate the action of specific domains to bind receptors, enter cells, and deliver a catalytic fragment into the cytosol. This results in GTPase inactivation, actin disassembly, and cytotoxicity. TcdB in particular has been shown to encode a region covering amino acids 1753 to 1851 that affects epitope exposure and cytotoxicity. Surprisingly, studies here show that several peptides derived from this region, which share the consensus sequence 1769NVFKGNTISDK1779, protect cells from the action of TcdB. One peptide, PepB2, forms multiple interactions with the carboxy-terminal region of TcdB, destabilizes TcdB structure, and disrupts cell binding. We further show that these effects require PepB2 to form a higher-order polymeric complex, a process that requires the central GN amino acid pair. These data suggest that TcdB1769-1779 interacts with repeat sequences in the proximal carboxy-terminal domain of TcdB (i.e., the CROP domain) to alter the conformation of TcdB. Furthermore, these studies provide insights into TcdB structure and functions that can be exploited to inactivate this critical virulence factor and ameliorate the course of CDI.IMPORTANCEClostridium difficile is a leading cause of hospital-associated illness that is often associated with antibiotic treatment. To cause disease, C. difficile secretes toxins, including TcdB, which is a multidomain intracellular bacterial toxin that undergoes conformational changes during cellular intoxication. This study describes the development of peptide-based inhibitors that target a region of TcdB thought to be critical for structural integrity of the toxin. The results show that peptides derived from a structurally important region of TcdB can be used to destabilize the toxin and prevent cellular intoxication. Importantly, this work provides a novel means of toxin inhibition that could in the future develop into a C. difficile treatment.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Clostridioides difficile/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Animais , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Células CHO , Linhagem Celular Tumoral , Infecções por Clostridium/microbiologia , Cricetulus , Epitopos , Humanos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fatores de Virulência
6.
J Biol Chem ; 288(30): 21526-36, 2013 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-23775085

RESUMO

In cells of the innate immune system, pathological increases in intracellular cAMP attenuate immune responses and contribute to infections by bacteria such as Bacillus anthracis. In this work, cAMP from B. anthracis edema toxin (ET) is found to activate the Notch signaling pathway in both mouse macrophages and human monocytes. ET as well as a cell-permeable activator of PKA induce Notch target genes (HES1, HEY1, IL2RA, and IL7R) and are able to significantly enhance the induction of these Notch target genes by a Toll-like receptor ligand. Elevated cAMP also resulted in increased levels of Groucho/transducin-like enhancer of Split (TLE) and led to increased amounts of a transcriptional repressor complex consisting of TLE and the Notch target Hes1. To address the mechanism used by ET to activate Notch signaling, components of Notch signaling were examined, and results revealed that ET increased levels of recombinant recognition sequence binding protein at the Jκ site (RBP-J), a DNA binding protein and principal transcriptional regulator of Notch signaling. Overexpression studies indicated that RBP-J was sufficient to activate Notch signaling and potentiate LPS-induced Notch signaling. Further examination of the mechanism used by ET to activate Notch signaling revealed that C/EBP ß, a transcription factor activated by cAMP, helped activate Notch signaling and up-regulated RBP-J. These studies demonstrate that cAMP activates Notch signaling and increases the expression of TLE, which could be an important mechanism utilized by cAMP to suppress immune responses.


Assuntos
AMP Cíclico/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Monócitos/metabolismo , Receptor Notch1/metabolismo , Proteínas Repressoras/metabolismo , Animais , Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bucladesina/análogos & derivados , Bucladesina/farmacologia , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Linhagem Celular , Células Cultivadas , Proteínas Correpressoras , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Immunoblotting , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Lipopolissacarídeos/farmacologia , Luciferases/genética , Luciferases/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Monócitos/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Receptor Notch1/genética , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição HES-1
7.
Environ Sci Pollut Res Int ; 19(3): 619-27, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21881907

RESUMO

PURPOSE: The aim of this study is to determine whether exposure to hazardous chemicals alters chemokine or cytokine production in macrophages and link these events to changes in intracellular signaling pathways and activation of specific gene promoters. METHODS: RAW 264.7 mouse macrophages were treated with selected toxic industrial chemicals (TICs) and examined for changes in immune function. Luminex multiplex technology was used to assess changes in cytokine/chemokine expression and activation of kinase signaling pathways. In addition, a panel of macrophage cell lines with promoter-specific luciferase reporter genes were generated and treated with the TICs, and transcriptional responses to these chemicals were detected by changes in luminescence. RESULTS: Changes in expression of cytokines and chemokines were linked to changes in the activation state of intracellular signaling pathways. Overall, the findings reveal that sublytic levels of TICs can alter the profile of cytokines and chemokines expressed by macrophages, with a pattern that suggests immunosuppression. The data demonstrate that critical changes in immune function correlate with activation of kinase signaling pathways in macrophages. CONCLUSIONS: These data provide insight into the effects of sublytic doses of selected TICs on macrophage function, with a particular emphasis on identifying changes in expression of cytokines and chemokines. These altered patterns in immune function were linked to changes in the activation state of intracellular signaling pathways. The data strongly suggest that small amounts of TICs can have subtle, yet very critical, effects on macrophages.


Assuntos
Citocinas/metabolismo , Substâncias Perigosas/farmacologia , Imunossupressores/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Transformada , Quimiocinas/metabolismo , Cloretos/química , Cloretos/farmacologia , Genes Reporter/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Hidrocarbonetos Clorados/química , Hidrocarbonetos Clorados/farmacologia , Imunossupressores/química , Macrófagos/imunologia , Camundongos , Peso Molecular , Concentração Osmolar , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Compostos de Enxofre/química , Compostos de Enxofre/farmacologia , Transcrição Gênica/efeitos dos fármacos
8.
Cell Immunol ; 272(2): 175-81, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22082566

RESUMO

Lethal toxin, a key virulence factor produced by Bacillus anthracis, induces cell death, in part by disrupting numerous signaling pathways, in mouse macrophages. However, exposure to sublethal doses of lethal toxin allows some cells to survive. Because these pro-survival signaling events occur within a few hours after exposure to sublethal doses, we hypothesized that acute phase proteins might influence macrophage survival. Our data show that serum amyloid A (SAA) is produced in response to lethal toxin treatment. Moreover, pre-treatment of macrophages with exogenous SAA protected macrophages from lethal toxin-mediated death. Exogenous SAA activated the p38 mitogen activated protein kinase (MAP) kinase pathway, while lethal toxin mutants incapable of p38 activation were incapable of causing cell death. Chemical inhibition of the p38 activation pathway abrogated the protective effects of SAA. These data show that SAA affords protection against lethal toxin in mouse macrophages and link this response to the p38 pathway.


Assuntos
Toxinas Bacterianas/farmacologia , Macrófagos/efeitos dos fármacos , Proteína Amiloide A Sérica/metabolismo , Animais , Antígenos de Bactérias/metabolismo , Bacillus anthracis/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação/genética , Substâncias Protetoras/metabolismo , Proteínas Recombinantes/metabolismo , Proteína Amiloide A Sérica/farmacologia , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Infect Immun ; 79(8): 3302-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21576335

RESUMO

Anthrax edema toxin (ET) is one of two binary toxins produced by Bacillus anthracis that contributes to the virulence of this pathogen. ET is an adenylate cyclase that generates high levels of cyclic AMP (cAMP), causing alterations in multiple host cell signaling pathways. We previously demonstrated that ET increases cell surface expression of the anthrax toxin receptors (ANTXR) in monocyte-derived cells and promotes dendritic cell (DC) migration toward the lymph node-homing chemokine MIP-3ß. In this work, we sought to determine if glycogen synthase kinase 3 (GSK-3) is important for ET-induced modulation of macrophage and DC function. We demonstrate that inhibition of GSK-3 dampens ET-induced maturation and migration processes of monocyte-derived dendritic cells (MDDCs). Additional studies reveal that the ET-induced expression of ANTXR in macrophages was decreased when GSK-3 activity was disrupted with chemical inhibitors or with small interfering RNA (siRNA) targeting GSK-3. Further examination of the ET induction of ANTXR revealed that a dominant negative form of CREB could block the ET induction of ANTXR, suggesting that CREB or a related family member was involved in the upregulation of ANTXR. Because CREB and GSK-3 activity appeared to be important for ET-induced ANTXR expression, the impact of GSK-3 on ET-induced CREB activity was examined in RAW 264.7 cells possessing a CRE-luciferase reporter. As with ANTXR expression, the ET induction of the CRE reporter was decreased by reducing GSK-3 activity. These studies not only provide insight into host pathways targeted by ET but also shed light on interactions between GSK-3 and CREB pathways in host immune cells.


Assuntos
Antraz/imunologia , Antraz/patologia , Antígenos de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Células Dendríticas/imunologia , Quinase 3 da Glicogênio Sintase/metabolismo , Macrófagos/imunologia , Receptores de Peptídeos/metabolismo , Animais , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Camundongos
10.
J Biol Chem ; 286(22): 19364-72, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21487015

RESUMO

The production of cAMP from Bacillus anthracis edema toxin (ET) activates gene expression in macrophages through a complex array of signaling pathways, most of which remain poorly defined. In this study, the tumor suppressor protein adenomatous polyposis coli (APC) was found to be important for the up-regulation of previously defined ET-stimulated genes (Vegfa, Ptgs2, Arg2, Cxcl2, Sdc1, and Cebpb). A reduction in the expression of these genes after ET exposure was observed when APC was disrupted in macrophages using siRNA or in bone marrow-derived macrophages obtained from C57BL/6J-Apc(Min) mice, which are heterozygous for a truncated form of APC. In line with this observation, ET increased the expression of APC at the transcriptional level, leading to increased amounts of APC in the nucleus. The mechanism utilized by APC to increase ET-induced gene expression was determined to depend on the ability of APC to interact with C/EBP ß, which is a transcription factor activated by cAMP. Coimmunoprecipitation experiments found that APC associated with C/EBP ß and that levels of this complex increase after ET exposure. A further connection was uncovered when silencing APC was determined to reduce the ET-induced phosphorylation of C/EBP ß at Thr-188. This ET-mediated phosphorylation of C/EBP ß was blocked by glycogen synthase kinase 3 (GSK-3) inhibitors, suggesting that GSK-3 is involved in the activation of C/EBP ß and supporting the idea of APC helping direct interactions between GSK-3 and C/EBP ß. These results indicate that ET stimulates gene expression by promoting the formation of an inducible protein complex consisting of APC and C/EBP ß.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Antígenos de Bactérias/farmacologia , Bacillus anthracis , Toxinas Bacterianas/farmacologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/metabolismo , Complexos Multiproteicos/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/genética , Linhagem Celular , AMP Cíclico , Inativação Gênica , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Camundongos , Complexos Multiproteicos/genética
11.
J Infect Dis ; 202(2): 251-60, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20533877

RESUMO

Bacillus anthracis remains a serious bioterrorism concern, and the currently licensed vaccine remains an incomplete solution for population protection from inhalation anthrax and has been associated with concerns regarding efficacy and safety. Thus, understanding how to generate long-lasting protective immunity with reduced immunizations or provide protection through postexposure immunotherapeutics are long-sought goals. Through evaluation of a large military cohort, we characterized the levels of antibodies against protective antigen and found that over half of anthrax vaccinees had low serum levels of in vitro toxin neutralization capacity. Using solid-phase epitope mapping and confirmatory assays, we identified several neutralization-associated humoral epitopes and demonstrated that select antipeptide responses mediated protection in vitro. Finally, passively transferred antibodies specific for select epitopes provided protection in an in vivo lethal toxin mouse model. Identification of these antigenic regions has important implications for vaccine design and the development of directed immunotherapeutics.


Assuntos
Antraz/imunologia , Anticorpos Antibacterianos/imunologia , Bacillus anthracis/imunologia , Adulto , Sequência de Aminoácidos , Animais , Antraz/epidemiologia , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/química , Anticorpos Antibacterianos/genética , Antígenos de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Bioterrorismo , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Feminino , Humanos , Imunização/métodos , Imunoglobulina G/sangue , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Oklahoma , Peptídeos/análise , Peptídeos/química , Grupos Raciais , Vacinação/métodos
12.
Infect Immun ; 78(6): 2418-28, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20308305

RESUMO

During advanced stages of inhalation anthrax, Bacillus anthracis accumulates at high levels in the bloodstream of the infected host. This bacteremia leads to sepsis during late-stage anthrax; however, the mechanisms through which B. anthracis-derived factors contribute to the pathology of infected hosts are poorly defined. Peptidoglycan, a major component of the cell wall of Gram-positive bacteria, can provoke symptoms of sepsis in animal models. We have previously shown that peptidoglycan of B. anthracis can induce the production of proinflammatory cytokines by cells in human blood. Here, we show that biologically active peptidoglycan is shed from an active culture of encapsulated B. anthracis strain Ames in blood. Peptidoglycan is able to bind to surfaces of responding cells, and internalization of peptidoglycan is required for the production of inflammatory cytokines. We also show that the peptidoglycan traffics to lysosomes, and lysosomal function is required for cytokine production. We conclude that peptidoglycan of B. anthracis is initially bound by an unknown extracellular receptor, is phagocytosed, and traffics to lysosomes, where it is degraded to a product recognized by an intracellular receptor. Binding of the peptidoglycan product to the intracellular receptor causes a proinflammatory response. These findings provide new insight into the mechanism by which B. anthracis triggers sepsis during a critical stage of anthrax disease.


Assuntos
Bacillus anthracis/imunologia , Citocinas/imunologia , Lisossomos/metabolismo , Peptidoglicano/imunologia , Fagocitose , Animais , Sangue/microbiologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovinos
13.
Infect Immun ; 78(4): 1610-7, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20123711

RESUMO

The current Bacillus anthracis vaccine consists largely of protective antigen (PA), the protein of anthrax toxin that mediates entry of edema factor (EF) or lethal factor (LF) into cells. PA induces protective antibody (Ab)-mediated immunity against Bacillus anthracis but has limited efficacy and duration. We previously demonstrated that activation of CD1d-restricted natural killer-like T cells (NKT) with a CD1d-binding glycolipid led to enhanced Ab titers specific for foreign antigen (Ag). We therefore tested the hypothesis that activation of NKT cells with the CD1d ligand (alpha-galactosylceramide [alpha-GC]) at the time of immunization improves PA-specific Ab responses. We observed that alpha-GC enhanced PA-specific Ab titers in C57BL/6 mice. In CD1d(-/-) mice deficient in type I and type II NKT cells the anti-PA Ab response was diminished. In Jalpha281(-/-) mice expressing CD1d but lacking type I alpha-GC-reactive NKT cells, alpha-GC did not enhance the Ab response. In vitro neutralization assays were performed and showed that the Ab titers correlated with protection of macrophages against anthrax lethal toxin (LT). The neutralization capacity of the Ab was further tested in lethal challenge studies, which revealed that NKT activation leads to enhanced in vivo protection against LT. Anti-PA Ab titers, neutralization, and protection were then measured over a period of several months, and this revealed that NKT activation leads to a sustained protective Ab response. These results suggest that NKT-activating CD1d ligands could be exploited for the development of improved vaccines for Bacillus anthracis that increase not only neutralizing Ab titers but also the duration of the protection afforded by Ab.


Assuntos
Vacinas contra Antraz/imunologia , Anticorpos Neutralizantes/biossíntese , Antígenos CD1d/imunologia , Linfócitos B/imunologia , Bacillus anthracis/imunologia , Toxinas Bacterianas/antagonistas & inibidores , Linfócitos T/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Antígenos CD1d/genética , Toxinas Bacterianas/imunologia , Modelos Animais de Doenças , Feminino , Galactosilceramidas/administração & dosagem , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Testes de Neutralização , Análise de Sobrevida
14.
PLoS Pathog ; 5(9): e1000588, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19779559

RESUMO

Exogenous CD1d-binding glycolipid (alpha-Galactosylceramide, alpha-GC) stimulates TCR signaling and activation of type-1 natural killer-like T (NKT) cells. Activated NKT cells play a central role in the regulation of adaptive and protective immune responses against pathogens and tumors. In the present study, we tested the effect of Bacillus anthracis lethal toxin (LT) on NKT cells both in vivo and in vitro. LT is a binary toxin known to suppress host immune responses during anthrax disease and intoxicates cells by protective antigen (PA)-mediated intracellular delivery of lethal factor (LF), a potent metalloprotease. We observed that NKT cells expressed anthrax toxin receptors (CMG-2 and TEM-8) and bound more PA than other immune cell types. A sub-lethal dose of LT administered in vivo in C57BL/6 mice decreased expression of the activation receptor NKG2D by NKT cells but not by NK cells. The in vivo administration of LT led to decreased TCR-induced cytokine secretion but did not affect TCR expression. Further analysis revealed LT-dependent inhibition of TCR-stimulated MAP kinase signaling in NKT cells attributable to LT cleavage of the MAP kinase kinase MEK-2. We propose that Bacillus anthracis-derived LT causes a novel form of functional anergy in NKT cells and therefore has potential for contributing to immune evasion by the pathogen.


Assuntos
Antígenos de Bactérias/farmacologia , Antígenos CD1d/metabolismo , Toxinas Bacterianas/farmacologia , Células T Matadoras Naturais/efeitos dos fármacos , Células T Matadoras Naturais/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Antígenos CD1d/imunologia , Bacillus anthracis/imunologia , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Galactosilceramidas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Células T Matadoras Naturais/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Peptídeos/biossíntese , Receptores de Peptídeos/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Infect Immun ; 77(1): 162-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18981257

RESUMO

The bipartite anthrax lethal toxin (LeTx) consisting of protective antigen (PA) and lethal factor (LF) is a major virulence factor contributing to death from systemic Bacillus anthracis infection. The current vaccine elicits antibodies directed primarily to PA; however, in experimental settings serologic responses to LF can neutralize LeTx and contribute to protection against infection. The goals of the present study were to identify sequential B-cell epitopes of LF and to determine the capacity of these determinants to bind neutralizing antibodies. Sera of recombinant LF-immunized A/J mice exhibited high titers of immunoglobulin G anti-LF reactivity that neutralized LeTx in vitro 78 days after the final booster immunization and protected the mice from in vivo challenge with 3 50% lethal doses of LeTx. These sera bound multiple discontinuous epitopes, and there were major clusters of reactivity on native LF. Strikingly, all three neutralizing, LF-specific monoclonal antibodies tested bound specific peptide sequences that coincided with sequential epitopes identified in polyclonal antisera from recombinant LF-immunized mice. This study confirms that LF induces high-titer protective antibodies in vitro and in vivo. Moreover, the binding of short LF peptides by LF-specific neutralizing monoclonal antibodies suggests that generation of protective antibodies by peptide vaccination may be feasible for this antigen. This study paves the way for a more effective anthrax vaccine by identifying discontinuous peptide epitopes of LF.


Assuntos
Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Bacillus anthracis/imunologia , Toxinas Bacterianas/imunologia , Epitopos de Linfócito B/imunologia , Animais , Antraz/prevenção & controle , Vacinas contra Antraz/imunologia , Anticorpos Antibacterianos/sangue , Anticorpos Monoclonais/imunologia , Linhagem Celular , Mapeamento de Epitopos , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos A , Testes de Neutralização
16.
Infect Immun ; 77(1): 23-31, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18936179

RESUMO

In an effort to better understand the mechanisms by which Bacillus anthracis establishes disease, experiments were undertaken to identify the genes essential for intracellular germination. Eighteen diverse genetic loci were identified via an enrichment protocol using a transposon-mutated library of B. anthracis spores, which was screened for mutants delayed in intracellular germination. Fourteen transposon mutants were identified in genes not previously associated with B. anthracis germination and included disruption of factors involved in membrane transport, transcriptional regulation, and intracellular signaling. Four mutants contained transposon insertions in gerHA, gerHB, gerHC, and pagA, respectively, each of which has been previously associated with germination or survival of B. anthracis within macrophages. Strain MIGD101 (named for macrophage intracellular germination defective 101) was of particular interest, since this mutant contained a transposon insertion in an intergenic region between BAs2807 and BAs2808, and was the most highly represented mutant in the enrichment. Analysis of B. anthracis MIGD101 by confocal microscopy and differential heat sensitivity following macrophage infection revealed ungerminated spores within the cell. Moreover, B. anthracis MIGD101 was attenuated in cell killing relative to the parent strain. Further experimental analysis found that B. anthracis MIGD101 was defective in five known B. anthracis germination pathways, supporting a mechanism wherein the intergenic region between BAs2807 and BAs2808 has a global affect on germination of this pathogen. Collectively, these findings provide insight into the mechanisms supporting B. anthracis germination within host cells.


Assuntos
Bacillus anthracis/patogenicidade , Macrófagos/microbiologia , Esporos Bacterianos/patogenicidade , Animais , Bacillus anthracis/genética , Bacillus anthracis/crescimento & desenvolvimento , Proteínas de Bactérias/genética , Linhagem Celular , Sobrevivência Celular , Contagem de Colônia Microbiana , Elementos de DNA Transponíveis , Ordem dos Genes , Genes Bacterianos , Camundongos , Mutagênese Insercional , Esporos Bacterianos/genética , Esporos Bacterianos/crescimento & desenvolvimento , Virulência , Fatores de Virulência/genética
17.
Infect Immun ; 76(11): 4895-904, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18765729

RESUMO

Bacillus anthracis edema toxin (ET) generates high levels of cyclic AMP and impacts a complex network of signaling pathways in targeted cells. In the current study, we sought to identify kinase signaling pathways modulated by ET to better understand how this toxin alters cell physiology. Using a panel of small-molecule inhibitors of mammalian kinases, we found that inhibitors of glycogen synthase kinase 3 beta (GSK-3beta) protected cells from ET-induced changes in the cell cycle. GSK-3beta inhibitors prevented declines in cellular levels of cyclin D1 and c-Jun following treatment of macrophages with ET. Strikingly, cell fractionation experiments and confocal immunofluorescence microscopy revealed that ET activates a compartmentalized pool of GSK-3beta residing in the nuclei, but not in the cytoplasm, of macrophages. To investigate the outcome of this event, we examined the cellular location and activation state of beta-catenin, a critical substrate of GSK-3beta, and found that the protein was inactivated within the nucleus following intoxication with ET. To determine if ET could overcome the effects of stimuli that inactivate GSK-3beta, we examined the impact of the toxin on the Wnt signaling pathway. The results of these experiments revealed that by targeting GSK-3beta residing in the nucleus, ET circumvents the upstream cytoplasmic inactivation of GSK-3beta, which occurs following exposure to Wnt-3A. These findings suggest ET arrests the cell cycle by a mechanism involving activation of GSK-3beta residing in the nucleus, and by using this novel mechanism of intoxication, ET avoids cellular systems that would otherwise reverse the effects of the toxin.


Assuntos
Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Núcleo Celular/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Inibidores Enzimáticos/farmacologia , Imunofluorescência , Glicogênio Sintase Quinase 3 beta , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
18.
Appl Environ Microbiol ; 74(16): 5201-10, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18552183

RESUMO

The engulfment of Bacillus anthracis spores by macrophages is an important step in the pathogenesis of inhalational anthrax. However, from a quantitative standpoint, the magnitude to which macrophages interact with and engulf spores remains poorly understood, in part due to inherent limitations associated with commonly used assays. To analyze phagocytosis of spores by RAW264.7 macrophage-like cells in a high-throughput, nonsubjective manner, we labeled B. anthracis Sterne 7702 spores prior to infection with an Alexa Fluor 488 amine-reactive dye in a manner that did not alter their germination, growth kinetics, and heat resistance. Using flow cytometry, large numbers of cells exposed to labeled spores were screened to concurrently discriminate infected from uninfected cells and surface-associated from internalized spores. These experiments revealed that spore uptake was not uniform, but instead, highly heterogeneous and characterized by subpopulations of infected and uninfected cells, as well as considerable variation in the number of spores associated with individual cells. Flow cytometry analysis of infections demonstrated that spore uptake was independent of the presence or absence of fetal bovine serum, a germinant that, while routinely used in vitro, complicates the interpretation of the outcome of infections. Two commonly used macrophage cell lines, RAW264.7 and J774A.1 cells, were compared, revealing significant disparity between these two models in the rates of phagocytosis of labeled spores. These studies provide the experimental framework for investigating mechanisms of spore phagocytosis, as well as quantitatively evaluating strategies for interfering with macrophage binding and uptake of spores.


Assuntos
Antraz/imunologia , Bacillus anthracis/crescimento & desenvolvimento , Bacillus anthracis/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Animais , Linhagem Celular , Citometria de Fluxo , Interações Hospedeiro-Patógeno , Macrófagos/microbiologia , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Esporos Bacterianos/imunologia , Succinimidas
19.
Biochem Biophys Res Commun ; 363(4): 959-64, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17910886

RESUMO

Clostridium sordellii lethal toxin (TcsL) inactivates small GTPases via glucosylation and induces apoptosis in mammalian cells; however, signaling events that link substrate modification with modulation of the mitochondria in these cells has not been determined. Experiments in the current study examined TcsL modulation of the Akt signaling pathway and related downstream targets. Early in TcsL intoxication, cells demonstrated a dramatic decrease in phosphorylated Akt, and this event required toxin enzymatic activity. The decrease in phosphorylated Akt was followed by caspase-dependent processing of Bcl-x(L) and Bid, revealing the connection between GTPase inactivation and mitochondrial-mediated apoptosis observed in TcsL-intoxicated cells. Levels of glycogen synthase kinase-3beta declined during later times of TcsL intoxication, suggesting a second intermediate step in apoptosis. Collectively, these data provide insight into the cascade of signaling events that lead to apoptotic death of TcsL-intoxicated cells.


Assuntos
Apoptose/efeitos dos fármacos , Toxinas Bacterianas/toxicidade , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caspases/metabolismo , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Glucosiltransferases/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Proteínas Mitocondriais/metabolismo , Fosforilação/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína bcl-X/metabolismo
20.
Infect Immun ; 75(12): 5726-34, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17923523

RESUMO

Bacillus anthracis transitions from a dormant spore to a vegetative bacillus through a series of structural and biochemical changes collectively referred to as germination. The timing of germination is important during early steps in infection and may determine if B. anthracis survives or succumbs to responsive macrophages. In the current study experiments determined the contribution of endogenous D-alanine production to the efficiency and timing of B. anthracis spore germination under in vitro and in vivo conditions. Racemase-mediated production of endogenous D-alanine by B. anthracis altered the kinetics for initiation of germination over a range of spore densities and exhibited a threshold effect wherein small changes in spore number resulted in major changes in germination efficiency. This threshold effect correlated with D-alanine production, was prevented by an alanine racemase inhibitor, and required L-alanine. Interestingly, endogenous production of inhibitory levels of D-alanine was detected under experimental conditions that did not support germination and in a germination-deficient mutant of B. anthracis. Racemase-dependent production of D-alanine enhanced survival of B. anthracis during interaction with murine macrophages, suggesting a role for inhibition of germination during interaction with these cells. Finally, in vivo experiments revealed an approximately twofold decrease in the 50% lethal dose of B. anthracis spores administered in the presence of D-alanine, indicating that rates of germination may be directly influenced by the levels of this amino acid during early stages of disease.


Assuntos
Alanina/biossíntese , Antraz/metabolismo , Antraz/microbiologia , Bacillus anthracis/fisiologia , Alanina/antagonistas & inibidores , Alanina Racemase/química , Animais , Bacillus anthracis/química , Bacillus anthracis/metabolismo , Feminino , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Esporos Bacterianos/química , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/metabolismo , Estereoisomerismo , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA