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








Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 208(12): 2749-2760, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35867676

RESUMO

T-bet+ B cells have emerged as a major B cell subset associated with both protective immunity and immunopathogenesis. T-bet is a transcription factor associated with the type I adaptive immune response to intracellular pathogens, driving an effector program characterized by the production of IFN-γ. Murine infection with the intracellular bacterium, Ehrlichia muris, generates protective extrafollicular T cell-independent T-bet+ IgM-secreting plasmablasts, as well as T-bet+ IgM memory cells. Although T-bet is a signature transcription factor for this subset, it is dispensable for splenic CD11c+ memory B cell development, but not for class switching to IgG2c. In addition to the T-bet+ plasmablasts found in the spleen, we show that Ab-secreting cells can also be found within the mouse peritoneal cavity; these cells, as well as their CD138- counterparts, also expressed T-bet. A large fraction of the T-bet+ peritoneal B cells detected during early infection were highly proliferative and expressed CXCR3 and CD11b, but, unlike in the spleen, they did not express CD11c. T-bet+ CD11b+ memory B cells were the dominant B cell population in the peritoneal cavity at 30 d postinfection, and although they expressed high levels of T-bet, they did not require B cell-intrinsic T-bet expression for their generation. Our data uncover a niche for T-bet+ B cells within the peritoneal cavity during intracellular bacterial infection, and they identify this site as a reservoir for T-bet+ B cell memory.


Assuntos
Infecções Bacterianas , Cavidade Peritoneal , Animais , Linfócitos B , Antígeno CD11c/metabolismo , Imunoglobulina M , Camundongos , Camundongos Endogâmicos C57BL , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição
2.
Nat Commun ; 13(1): 452, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35064115

RESUMO

CD11c+T-bet+ B cells are recognized as an important component of humoral immunity and autoimmunity. These cells can be distinguished from other B cells by their higher expression of the adenosine receptor 2a. Here we address whether A2A receptor activation can affect CD11c+T-bet+ B cells. We show that administration of the A2A receptor agonist CGS-21680 depletes established CD11c+T-bet+ B cells in ehrlichial-infected mice, in a B cell-intrinsic manner. Agonist treatment similarly depletes CD11c+T-bet+ B cells and CD138+ B cells and reduces anti-nuclear antibodies in lupus-prone mice. Agonist treatment is also associated with reduced kidney pathology and lymphadenopathy. Moreover, A2A receptor stimulation depletes pathogenic lymphocytes and ameliorates disease even after disease onset, highlighting the therapeutic potential of this treatment. This study suggests that targeting the adenosine signaling pathway may provide a method for the treatment of lupus and other autoimmune diseases mediated by T-bet+ B cells.


Assuntos
Autoimunidade , Linfócitos B/imunologia , Antígeno CD11c/metabolismo , Infecções/imunologia , Agonistas do Receptor Purinérgico P1/farmacologia , Receptor A2A de Adenosina/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Autoimunidade/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Modelos Animais de Doenças , Ehrlichia , Feminino , Infecções/patologia , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos Endogâmicos C57BL
3.
PLoS One ; 16(1): e0244855, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33507994

RESUMO

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the pandemic human respiratory illness COVID-19, is a global health emergency. While severe acute disease has been linked to an expansion of antibody-secreting plasmablasts, we sought to identify B cell responses that correlated with positive clinical outcomes in convalescent patients. We characterized the peripheral blood B cell immunophenotype and plasma antibody responses in 40 recovered non-hospitalized COVID-19 subjects that were enrolled as donors in a convalescent plasma treatment study. We observed a significant negative correlation between the frequency of peripheral blood memory B cells and the duration of symptoms for convalescent subjects. Memory B cell subsets in convalescent subjects were composed of classical CD24+ class-switched memory B cells, but also activated CD24-negative and natural unswitched CD27+ IgD+ IgM+ subsets. Memory B cell frequency was significantly correlated with both IgG1 and IgM responses to the SARS-CoV-2 spike protein receptor binding domain (RBD) in most seropositive subjects. IgM+ memory, but not switched memory, directly correlated with virus-specific antibody responses, and remained stable over 3 months. Our findings suggest that the frequency of memory B cells is a critical indicator of disease resolution, and that IgM+ memory B cells may play an important role in SARS-CoV-2 immunity.


Assuntos
Subpopulações de Linfócitos B/imunologia , COVID-19/imunologia , Adulto , Idoso , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Formação de Anticorpos , Linfócitos B/imunologia , Convalescença , Progressão da Doença , Feminino , Humanos , Imunidade/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/metabolismo , Imunofenotipagem/métodos , Masculino , Pessoa de Meia-Idade , Recuperação de Função Fisiológica/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação
4.
medRxiv ; 2020 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-32908991

RESUMO

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the pandemic human respiratory illness COVID-19, is a global health emergency. While severe acute disease has been linked to an expansion of antibody-secreting plasmablasts, we sought to identify B cell responses that correlated with positive clinical outcomes in convalescent patients. We characterized the peripheral blood B cell immunophenotype and plasma antibody responses in 40 recovered non-hospitalized COVID-19 subjects that were enrolled as donors in a convalescent plasma treatment study. We observed a significant negative correlation between the frequency of peripheral blood memory B cells and the duration of symptoms for convalescent subjects. Memory B cell subsets in convalescent subjects were composed of classical CD24+ class-switched memory B cells, but also activated CD24-negative and natural unswitched CD27+ IgD+ IgM+ subsets. Memory B cell frequency was significantly correlated with both IgG1 and IgM responses to the SARS-CoV-2 spike protein receptor binding domain (RBD). IgM+ memory, but not switched memory, directly correlated with virus-specific antibody responses, and remained stable over time. Our findings suggest that the frequency of memory B cells is a critical indicator of disease resolution, and that IgM+ memory B cells play an important role in SARS-CoV-2 immunity.

5.
J Immunol ; 205(4): 1050-1058, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32680956

RESUMO

CD11c+ T-bet+ B cells generated during ehrlichial infection require CD4+ T cell help and IL-21 signaling for their development, but the exact T cell subset required had not been known. In this study, we show in a mouse model of Ehrlichia muris that type 1 T follicular helper (TFH1) cells provide help to CD11c+ T-bet+ B cells via the dual secretion of IL-21 and IFN-γ in a CD40/CD40L-dependent manner. TFH1 cell help was delivered in two phases: IFN-γ signals were provided early in infection, whereas CD40/CD40L help was provided late in infection. In contrast to T-bet+ T cells, T-bet+ B cells did not develop in the absence of B cell-intrinsic Bcl-6 but were generated in the absence of T-bet. T-bet-deficient memory B cells were largely indistinguishable from their wild-type counterparts, although they no longer underwent switching to IgG2c. These data suggest that a primary function of T-bet in B cells during ehrlichial infection is to promote appropriate class switching, not lineage specification. Thus, CD11c+ memory B cells develop normally without T-bet but require Bcl-6 and specialized help from dual cytokine-producing TFH1 cells.


Assuntos
Antígenos CD11/metabolismo , Interferon gama/metabolismo , Interleucinas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Células T Auxiliares Foliculares/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos CD11/imunologia , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Ligante de CD40/imunologia , Ligante de CD40/metabolismo , Ehrlichia/imunologia , Ehrlichia/metabolismo , Feminino , Memória Imunológica/imunologia , Interferon gama/imunologia , Interleucinas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-bcl-6/imunologia , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Células T Auxiliares Foliculares/imunologia , Proteínas com Domínio T/imunologia
6.
J Immunol ; 203(9): 2415-2424, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31570507

RESUMO

Bacterial, parasitic, and viral infections are well-known causes of lymphoid tissue disorganization, although the factors, both host and/or pathogen derived, that mediate these changes are largely unknown. Ehrlichia muris infection in mice causes a loss of germinal center (GC) B cells that is accompanied by the generation of extrafollicular T-bet+ CD11c+ plasmablasts and IgM memory B cells. We addressed a possible role for TNF-α in this process because this cytokine has been shown to regulate GC development. Ablation of TNF-α during infection resulted in an 8-fold expansion of GL7+ CD38lo CD95+ GC B cells, and a 2.5- and 5-fold expansion of CD138+ plasmablasts and T-bet+ memory cells, respectively. These changes were accompanied by a reduction in splenomegaly, more organized T and B cell zones, and an improved response to Ag challenge. CXCL13, the ligand for CXCR5, was detected at 6-fold higher levels following infection but was much reduced following TNF-α ablation, suggesting that CXCL13 dysregulation also contributes to loss of lymphoid tissue organization. T follicular helper cells, which also underwent expansion in infected TNF-α--deficient mice, may also have contributed to the expansion of T-bet+ B cells, as the latter are known to require T cell help. Our findings contrast with previously described roles for TNF-α in GCs and reveal how host-pathogen interactions can induce profound changes in cytokine and chemokine production that can alter lymphoid tissue organization, GC B cell development, and extrafollicular T-bet+ B cell generation.


Assuntos
Linfócitos B/imunologia , Infecções Bacterianas/imunologia , Centro Germinativo/imunologia , Tecido Linfoide/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Quimiocina CXCL13/fisiologia , Tolerância Imunológica , Memória Imunológica , Tecido Linfoide/imunologia , Camundongos , Camundongos Endogâmicos C57BL
7.
Infect Immun ; 87(12)2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31548319

RESUMO

Antibodies are essential for immunity against Ehrlichia chaffeensis, and protective mechanisms involve blocking of ehrlichial attachment or complement and Fcγ-receptor-dependent destruction. In this study, we determined that major outer membrane protein 1 (OMP-19) hypervariable region 1 (HVR1)-specific human monoclonal antibodies (huMAbs) are protective through conventional extracellular neutralization and, more significantly, through a novel intracellular TRIM21-mediated mechanism. Addition of OMP-1-specific huMAb EHRL-15 (IgG1) prevented infection by blocking attachment/entry, a mechanism previously reported; conversely, OMP-1-specific huMAb EHRL-4 (IgG3) engaged intracellular TRIM21 and initiated an immediate innate immune response and rapid intracellular degradation of ehrlichiae. EHRL-4-TRIM21-mediated inhibition was significantly impaired in TRIM21 knockout THP-1 cells. EHRL-4 interacted with cytosolic Fc receptor TRIM21, observed by confocal microscopy and confirmed by co-immunoprecipitation. E. chaffeensis-EHRL-4-TRIM21 complexes caused significant upregulation of proinflammatory cytokine/chemokine transcripts and resulted in rapid (<30 min) nuclear accumulation of NF-κB and TRIM21 and ehrlichial destruction. We investigated the role of TRIM21 in the autophagic clearance of ehrlichiae in the presence of EHRL-4. Colocalization between EHRL-4-opsonized ehrlichiae, polyubiquitinated TRIM21, autophagy regulators (ULK1 and beclin 1) and effectors (LC3 and p62), and lysosome-associated membrane protein 2 (LAMP2) was observed. Moreover, autophagic flux defined by conversion of LC3I to LC3II and accumulation and degradation of p62 was detected, and EHRL-4-mediated degradation of E. chaffeensis was abrogated by the autophagy inhibitor 3-methyladenine. Our results demonstrate that huMAbs are capable of inhibiting E. chaffeensis infection by distinct effector mechanisms: extracellularly by neutralization and intracellularly by engaging TRIM21, which mediates a rapid innate immune response that mobilizes the core autophagy components, triggering localized selective autophagic degradation of ehrlichiae.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Ehrlichia chaffeensis/imunologia , Ribonucleoproteínas/genética , Adenina/análogos & derivados , Adenina/farmacologia , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Antígenos de Bactérias/genética , Autofagia/imunologia , Aderência Bacteriana/genética , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Ehrlichia chaffeensis/genética , Técnicas de Inativação de Genes , Humanos , Imunidade Humoral/imunologia , NF-kappa B/genética , Células THP-1
8.
Cell Rep ; 24(4): 824-837.e3, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-30044980

RESUMO

Immunoglobulin M (IgM) memory cells undergo differentiation in germinal centers following antigen challenge, but the full effector cell potential of these cells is unknown. We monitored the differentiation of enhanced yellow fluorescent protein (eYFP)-labeled CD11c+ and CD11cneg T-bet+ IgM memory cells after their transfer into naive recipient mice. Following challenge infection, many memory cells differentiated into IgM-producing plasmablasts. Other donor B cells entered germinal centers, downregulated CD11c, underwent class switch recombination, and became switched memory cells. Yet other donor cells were maintained as IgM memory cells, and these IgM memory cells retained their multi-lineage potential following serial transfer. These findings were corroborated at the molecular level using immune repertoire analyses. Thus, IgM memory cells can differentiate into all effector B cell lineages and undergo self-renewal, properties that are characteristic of stem cells. We propose that these memory cells exist to provide long-term multi-functional immunity and act primarily to maintain the production of protective antibodies.


Assuntos
Linfócitos B/imunologia , Memória Imunológica/imunologia , Animais , Diferenciação Celular/imunologia , Ehrlichia/imunologia , Ehrlichiose/imunologia , Feminino , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos C57BL
9.
Cell Immunol ; 321: 8-17, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28838763

RESUMO

CD11c+ T-bet+ B cells have now been detected and characterized in different experimental and clinical settings, in both mice and humans. Whether such cells are monolithic, or define subsets of B cells with different functions is not yet known. Our studies have identified CD11c+ IgM+ CD19hi splenic IgM memory B cells that appear at approximately three weeks post-ehrlichial infection, and persist indefinitely, during low-level chronic infection. Although the CD11c+ T-bet+ B cells we have described are distinct, they appear to share many features with similar cells detected under diverse conditions, including viral infections, aging, and autoimmunity. We propose that CD11c+ T-bet+ B cells as a group share characteristics of memory B cells that are maintained under conditions of inflammation and/or low-level chronic antigen stimulation. In some cases, these cells may be advantageous, by providing immunity to re-infection, but in others may be deleterious, by contributing to aged-associated autoimmune responses.


Assuntos
Linfócitos B/imunologia , Infecções Bacterianas/imunologia , Antígeno CD11c/imunologia , Memória Imunológica/imunologia , Inflamação/imunologia , Proteínas com Domínio T/imunologia , Animais , Linfócitos B/metabolismo , Infecções Bacterianas/genética , Infecções Bacterianas/microbiologia , Antígeno CD11c/metabolismo , Doença Crônica , Perfilação da Expressão Gênica/métodos , Humanos , Memória Imunológica/genética , Inflamação/genética , Proteínas com Domínio T/metabolismo
10.
PLoS One ; 12(6): e0178853, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28575114

RESUMO

IgM memory cells are recognized as an important component of B cell memory in mice and humans. Our studies of B cells elicited in response to ehrlichial infection identified a population of CD11c-positive IgM memory cells, and an IgM bone marrow antibody-secreting cell population. The origin of these cells was unknown, although an early T-independent spleen CD11c- and T-bet-positive IgM plasmablast population precedes both, suggesting a linear relationship. A majority of the IgM memory cells detected after day 30 post-infection, also T-bet-positive, had undergone somatic hypermutation, indicating they expressed activation-induced cytidine deaminase (AID). Therefore, to identify early AID-expressing precursor B cells, we infected an AID-regulated tamoxifen-inducible Cre-recombinase-EYFP reporter strain. Tamoxifen administration led to the labeling of both IgM memory cells and bone marrow ASCs on day 30 and later post-infection. High frequencies of labeled cells were identified on day 30 post-infection, following tamoxifen administration on day 10 post-infection, although IgM memory cells were marked when tamoxifen was administered as early as day 4 post-infection. Transcription of Aicda in the early plasmablasts was not detected in the absence of CD4 T cells, but occurred independently of TLR signaling. Unlike the IgM memory cells, the bone marrow IgM ASCs were elicited independent of T cell help. Moreover, Aicda was constitutively expressed in IgM memory cells, but not in bone marrow ASCs. These studies demonstrate that two distinct long-term IgM-positive B cell populations are generated early in response to infection, but are maintained via separate mechanisms.


Assuntos
Células da Medula Óssea/citologia , Imunoglobulina M/imunologia , Memória Imunológica , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL
11.
Dev Comp Immunol ; 65: 114-123, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27377583

RESUMO

Bat immunity has received increasing attention because some bat species are being decimated by the fungal disease, White Nose Syndrome, while other species are potential reservoirs of zoonotic viruses. Identifying specific immune processes requires new specific tools and reagents. In this study, we describe a new mouse monoclonal antibody (mAb) reactive with Eptesicus fuscus immunoglobulins. The epitope recognized by mAb BT1-4F10 was localized to immunoglobulin light (lambda) chains; hence, the mAb recognized serum immunoglobulins and B lymphocytes. The BT1-4F10 epitope appeared to be restricted to Microchiropteran immunoglobulins and absent from Megachiropteran immunoglobulins. Analyses of sera and other E. fuscus fluids showed that most, if not all, secreted immunoglobulins utilized lambda light chains. Finally, mAb BT1-4F10 permitted the identification of B cell follicles in splenic white pulp. This Microchiropteran-specific mAb has potential utility in seroassays; hence, this reagent may have both basic and practical applications for studying immune process.


Assuntos
Anticorpos Antifúngicos/isolamento & purificação , Anticorpos Monoclonais/isolamento & purificação , Linfócitos B/imunologia , Quirópteros/imunologia , Micoses/imunologia , Zoonoses/imunologia , Animais , Linhagem Celular , Separação Celular , Mapeamento de Epitopos , Citometria de Fluxo , Cadeias lambda de Imunoglobulina/imunologia , Imunofenotipagem , Camundongos , Microscopia
12.
Infect Immun ; 83(5): 2139-47, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25776744

RESUMO

Infection of mice with the bacterium Ehrlichia muris elicits a protective T cell-independent (TI) IgM response mediated primarily by a population of CD11c-expressing plasmablasts in the spleen. Although splenic marginal zone (MZ) B cells are considered to be important for TI responses to blood-borne pathogens, MZ B cells were not responsible for generating plasmablasts in response to Ehrlichia muris. Moreover, antigen-specific serum IgM was decreased only modestly in splenectomized mice and in mice that lacked spleen, lymph nodes, and Peyer's patches (SLP mice). Both splenectomized and SLP mice were protected against lethal ehrlichial challenge infection. Moreover, we found a high frequency of Ehrlichia-specific plasmablasts in the omentum of both conventional and SLP mice. Omental plasmablasts elicited during Ehrlichia infection lacked expression of CD138 but expressed CD11c in a manner similar to that of their splenic counterparts. Selective ablation of CD11c-expressing B cells nearly eliminated the omental Ehrlichia-specific plasmablasts and reduced antigen-specific serum IgM, identifying the omental B cells as a source of IgM production in the SLP mice. Generation of the omental plasmablasts was route dependent, as they were detected following peritoneal infection but not following intravenous infection. Our data identify the omentum as an important auxiliary site of IgM production during intracellular bacterial infection.


Assuntos
Anticorpos Antibacterianos/metabolismo , Ehrlichiose/imunologia , Imunoglobulina M/metabolismo , Omento/imunologia , Plasmócitos/imunologia , Animais , Antígeno CD11c/análise , Ehrlichia/imunologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Omento/patologia , Sindecana-1/análise
13.
Infect Immun ; 81(12): 4490-7, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24082070

RESUMO

B cell activating factor of the tumor necrosis factor family (BAFF) is an essential survival factor for B cells and has been shown to regulate T cell-independent (TI) IgM production. During Ehrlichia muris infection, TI IgM secretion in the spleen was BAFF dependent, and antibody-mediated BAFF neutralization led to an impairment of IgM-mediated host defense. The failure of TI plasmablasts to secrete IgM was not a consequence of alterations in their generation, survival, or early differentiation, since all occurred normally in infected mice following BAFF neutralization. Gene expression characteristic of plasma cell differentiation was also unaffected by BAFF neutralization in vivo, and except for CD138, plasmablast cell surface marker expression was unaffected. IgM was produced, since it was detected intracellularly, and impaired secretion was not due to a failure to express the IgM secretory exon. Addition of BAFF to plasmablasts in vitro rescued IgM secretion, suggesting that BAFF signaling can directly regulate secretory processes. Our findings indicate that BAFF signaling can modulate TI host defense by acting at a late stage in B cell differentiation, via its regulation of terminal plasmablast differentiation and/or IgM secretion.


Assuntos
Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Ehrlichia/imunologia , Ehrlichiose/imunologia , Imunoglobulina M/imunologia , Animais , Fator Ativador de Células B/antagonistas & inibidores , Fator Ativador de Células B/metabolismo , Diferenciação Celular , Imunoglobulina M/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/imunologia , Plasmócitos/metabolismo , Transdução de Sinais/imunologia , Sindecana-1/imunologia , Linfócitos T/imunologia
14.
J Immunol ; 191(3): 1240-9, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23804710

RESUMO

Immunological memory has long considered to be harbored in B cells that express high-affinity class-switched IgG. IgM-positive memory B cells can also be generated following immunization, although their physiological role has been unclear. In this study, we show that bacterial infection elicited a relatively large population of IgM memory B cells that were uniquely identified by their surface expression of CD11c, CD73, and programmed death-ligand 2. The cells lacked expression of cell surface markers typically expressed by germinal center B cells, were CD138 negative, and did not secrete Ab ex vivo. The population was also largely quiescent and accumulated somatic mutations. The IgM memory B cells were located in the region of the splenic marginal zone and were not detected in blood or other secondary lymphoid organs. Generation of the memory cells was CD4 T cell dependent and required IL-21R signaling. In vivo depletion of the IgM memory B cells abrogated the IgG recall responses to specific Ag challenge, demonstrating that the cell population was required for humoral memory, and underwent class-switch recombination following Ag encounter. Our findings demonstrate that T cell-dependent IgM memory B cells can be elicited at high frequency and can play an important role in maintaining long-term immunity during bacterial infection.


Assuntos
Linfócitos B/imunologia , Switching de Imunoglobulina/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Memória Imunológica , 5'-Nucleotidase/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Proteínas da Membrana Bacteriana Externa/imunologia , Antígeno CD11c/metabolismo , Linfócitos T CD4-Positivos/imunologia , Ehrlichia/imunologia , Ehrlichiose/imunologia , Centro Germinativo/imunologia , Imunização , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Receptores de Interleucina-21/metabolismo , Sindecana-1/metabolismo
15.
J Immunol ; 190(9): 4725-35, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23526822

RESUMO

Hematopoietic stem and progenitor cell (HSPC) phenotype and function can change in response to infectious challenge. These changes can be mediated by cytokines, IFNs, and pathogen-associated molecules, via TLR, and are thought to promote tailored immune responses for particular pathogens. In this study, we investigated the signals that activate HSPCs during ehrlichiosis, a disease characterized by profound hematopoietic dysfunction in both humans and mice. In a mouse model of ehrlichiosis, we observed that infection-induced proliferation of bone marrow HSPCs was dependent on IFN-γ signaling and was partially dependent on MyD88. However, MyD88 was not required in HSPCs for their expansion during infection, because similar frequencies of MyD88-deficient and wild-type HSPCs proliferated in mixed bone marrow chimeric mice. MyD88-deficient mice exhibited low serum and bone marrow concentration of IFN-γ compared with wild-type mice. We next identified CD4 T cells as the primary cells producing IFN-γ in the bone marrow and demonstrated a nonredundant role for CD4-derived IFN-γ in increased HSPCs. Using mixed bone marrow chimeric mice, we identified a requirement for MyD88 in CD4 T cells for increased T-bet expression, optimal IFN-γ production, and CD4 T cell proliferation. Our data demonstrate an essential role for CD4 T cells in mediating HSPC activation in response to bacterial infection and illustrate a novel role for MyD88 signaling in CD4 T cells in this process. These findings further support the idea that IFN-γ production is essential for HSPC activation and hematopoietic responses to infection.


Assuntos
Infecções Bacterianas/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Interferon gama/biossíntese , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/microbiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Proliferação de Células , Ehrlichia/imunologia , Ehrlichiose/imunologia , Ehrlichiose/metabolismo , Ehrlichiose/microbiologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/microbiologia , Interferon gama/imunologia , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia
16.
PLoS Pathog ; 9(12): e1003837, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24385909

RESUMO

T cell responses to enteric bacteria are important in inflammatory bowel disease. I2, encoded by the pfiT gene of Pseudomonas fluorescens, is a T-cell superantigen associated with human Crohn's disease. Here we report the crystal structure of pfiT at 1.7Å resolution and provide a functional analysis of the interaction of pfiT and its homolog, PA2885, with human class II MHC. Both pfiT and PA2885 bound to mammalian cells and stimulated the proliferation of human lymphocytes. This binding was greatly inhibited by anti-class II MHC HLA-DR antibodies, and to a lesser extent, by anti HLA-DQ and DP antibodies, indicating that the binding was class II MHC-specific. GST-pfiT efficiently precipitated both endogenous and in vitro purified recombinant HLA-DR1 molecules, indicating that pfiT directly interacted with HLA-DR1. Competition studies revealed that pfiT and the superantigen Mycoplasma arthritidis mitogen (MAM) competed for binding to HLA-DR, indicating that their binding sites overlap. Structural analyses established that pfiT belongs to the TetR-family of DNA-binding transcription regulators. The distinct structure of pfiT indicates that it represents a new family of T cell superantigens.


Assuntos
Doença de Crohn/imunologia , Pseudomonas fluorescens/imunologia , Superantígenos/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Cultivadas , Cristalografia por Raios X , Antígeno HLA-DR1/metabolismo , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Pseudomonas aeruginosa/imunologia , Homologia de Sequência , Superantígenos/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
17.
J Immunol ; 189(9): 4451-8, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23028057

RESUMO

Fully functional T cells are necessary for the maintenance of protective immunity during chronic infections. However, activated T cells often undergo apoptosis or exhaustion upon chronic stimulation mediated by Ag or inflammation. T cell attrition can be compensated for by the production of thymus-derived T cells, although the new naive T cells must undergo T cell priming and differentiation under conditions different from those encountered during acute infection. We used a murine model of Mycobacterium tuberculosis infection to address how the activation and differentiation of new thymic emigrants is affected by chronic inflammation, as well as whether the newly developed effector T cells help to maintain peripheral T cell responses. Although new thymic emigrants contributed to the peripheral T cell response early during acute M. tuberculosis infection, the relative contribution of new effector T cells to the peripheral CD4 and CD8 T cell pools declined during chronic infection. The decline in new T cell recruitment was a consequence of quantitative and/or qualitative changes in Ag presentation, because during chronic infection both the priming and expansion of naive T cells were inefficient. Thus, although thymic tolerance is not a major factor that limits protective T cell responses, the chronic environment does not efficiently support naive T cell priming and accumulation during M. tuberculosis infection. These studies support our previous findings that long-term protective T cell responses can be maintained indefinitely in the periphery, but also suggest that the perturbation of homeostasis during chronic inflammatory responses may elicit immune pathology mediated by new T cells.


Assuntos
Mycobacterium tuberculosis/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Subpopulações de Linfócitos T/microbiologia , Timo/citologia , Timo/imunologia , Tuberculose Pulmonar/patologia
18.
J Immunol ; 189(3): 1440-7, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22730531

RESUMO

Polyreactivity is well known as a property of natural IgM produced by B-1 cells. We demonstrate that polyreactive IgM is also generated during infection of mice with Ehrlichia muris, a tick-borne intracellular bacterial pathogen. The polyreactive IgM bound self and foreign Ags, including single-stranded and double-stranded DNA, insulin, thyroglobulin, LPS, influenza virus, and Borrelia burgdorferi. Production of polyreactive IgM during infection was Ag driven, not due to polyclonal B cell activation, as the majority of polyreactive IgM recognized ehrlichial Ag(s), including an immunodominant outer membrane protein. Monoclonal polyreactive IgM derived from T cell-independent spleen plasmablasts, which was germline-encoded, also bound cytoplasmic and nuclear Ags in HEp-2 cells. Polyreactive IgM protected immunocompromised mice against lethal bacterial challenge infection. Serum from human ehrlichiosis patients also contained polyreactive and self-reactive IgM. We propose that polyreactivity increases IgM efficacy during infection but may also exacerbate or mollify the response to foreign and self Ags.


Assuntos
Antígenos de Bactérias/imunologia , Imunoglobulina M/biossíntese , Líquido Intracelular/imunologia , Líquido Intracelular/microbiologia , Animais , Antígenos T-Independentes/imunologia , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Ehrlichia/imunologia , Ehrlichiose/sangue , Ehrlichiose/imunologia , Ehrlichiose/metabolismo , Epitopos Imunodominantes/imunologia , Líquido Intracelular/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Plasmócitos/imunologia , Plasmócitos/metabolismo , Baço/imunologia , Baço/metabolismo , Baço/patologia
19.
PLoS One ; 6(12): e28669, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22194881

RESUMO

How hematopoietic stem cells (HSCs) respond to inflammatory signals during infections is not well understood. Our studies have used a murine model of ehrlichiosis, an emerging tick-born disease, to address how infection impacts hematopoietic function. Infection of C57BL/6 mice with the intracellular bacterium, Ehrlichia muris, results in anemia and thrombocytopenia, similar to what is observed in human ehrlichiosis patients. In the mouse, infection promotes myelopoiesis, a process that is critically dependent on interferon gamma (IFNγ) signaling. In the present study, we demonstrate that E. muris infection also drives the transient proliferation and expansion of bone marrow Lin-negative Sca-1(+) cKit(+) (LSK) cells, a population of progenitor cells that contains HSCs. Expansion of the LSK population in the bone marrow was associated with a loss of dormant, long-term repopulating HSCs, reduced engraftment, and a bias towards myeloid lineage differentiation within that population. The reduced engraftment and myeloid bias of the infection-induced LSK cells was transient, and was most pronounced on day 8 post-infection. The infection-induced changes were accompanied by an expansion of more differentiated multipotent progenitor cells, and required IFNγ signaling. Thus, in response to inflammatory signals elicited during acute infection, HSCs can undergo a rapid, IFNγ-dependent, transient shift from dormancy to activity, ostensibly, to provide the host with additional or better-armed innate cells for host defense. Similar changes in hematopoietic function likely underlie many different infections of public health importance.


Assuntos
Infecções Bacterianas/imunologia , Células-Tronco Hematopoéticas/citologia , Interferon gama/metabolismo , Doença Aguda , Animais , Ataxina-1 , Ataxinas , Proliferação de Células , Separação Celular , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais
20.
J Immunol ; 187(12): 6180-4, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22079983

RESUMO

CD4 T cells are essential for immune control of γ-herpesvirus latency. We previously identified a murine MHC class II-restricted epitope in γ-herpesvirus-68 gp150 (gp150(67-83)I-A(b)) that elicits CD4 T cells that are maintained throughout long-term infection. However, it is unknown whether naive cells can be recruited into the antiviral CD4 T cell pool during latency. In this study, we generate a mouse transgenic for a gp150-specific TCR and show epitope-specific activation of transgenic CD4 T cells during acute and latent infections. Furthermore, although only dendritic cells can stimulate virus-specific CD8 T cells during latency, we show that both dendritic cells and B cells stimulate transgenic CD4 T cells. These studies demonstrate that naive CD4 T cells specific for a viral glycoprotein can be stimulated throughout infection, even during quiescent latency, suggesting that CD4 T cell memory is maintained in part by the continual recruitment of naive cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Epitopos de Linfócito T/imunologia , Ativação Linfocitária/imunologia , Rhadinovirus/imunologia , Latência Viral/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Epitopos de Linfócito T/genética , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Ativação Viral/genética , Ativação Viral/imunologia , Latência Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA