Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 213(3): 373-383, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38884660

RESUMO

Conventionally, immune responses are studied in the context of inflamed tissues and their corresponding draining lymph nodes (LNs). However, little is known about the effects of systemic inflammatory signals generated during local inflammation on distal tissues and nondraining LNs. Using a mouse model of cutaneous immunization, we found that systemic inflammatory stimuli triggered a rapid and selective distal response in the small intestine and the mesenteric LN (mesLN). This consisted of increased permeability of intestinal blood vessels and lymphatic drainage of bloodborne solutes into the mesLN, enhanced activation and migration of intestinal dendritic cells, as well as amplified T cell responses in the mesLNs to systemic but not orally derived Ags. Mechanistically, we found that the small intestine endothelial cells preferentially expressed molecules involved in TNF-α signaling and that TNF-α blockade markedly diminished distal intestinal responses to cutaneous immunization. Together, these findings reveal that the intestinal immune system is rapidly and selectively activated in response to inflammatory cues regardless of their origin, thus identifying an additional layer of defense and enhanced surveillance of a key barrier organ at constant risk of pathogen encounter.


Assuntos
Imunização , Linfonodos , Animais , Camundongos , Linfonodos/imunologia , Imunização/métodos , Camundongos Endogâmicos C57BL , Citocinas/imunologia , Citocinas/metabolismo , Intestino Delgado/imunologia , Células Dendríticas/imunologia , Inflamação/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Linfócitos T/imunologia , Mucosa Intestinal/imunologia
2.
J Immunol ; 212(6): 1022-1028, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38294253

RESUMO

Plasma cells secrete an abundance of Abs and are a crucial component of our immune system. The intestinal lamina propria harbors the largest population of plasma cells, most of which produce IgA. These Abs can bind to beneficial gut bacteria to reinforce intestinal homeostasis and provide protection against enteric pathogens. Plasma cells downregulate many cell-surface proteins commonly used to identify B cells. In mice, expression of the surface marker CD138 has been widely used to identify plasma cells in lymph nodes, bone marrow, and spleen. Intestinal plasma cells require liberation via extensive tissue processing involving treatment with collagenase. We report that detection of CD138 surface expression is reduced following collagenase treatment. Using a mouse in which yellow fluorescent protein expression is controlled by the plasma cell requisite transcription factor Blimp-1, we show that surface detection of transmembrane activator and CAML interactor captures a significant proportion of Ab-secreting plasma cells in the intestinal lamina propria and gut-draining mesenteric lymph nodes. Additionally, we describe a flow cytometry panel based on the detection of surface markers to identify murine B cell subsets in the intestinal lamina propria and, as a proof of concept, combine it with a cutting-edge fate-tracking system to characterize the fate of germinal center B cells activated in early life. By identifying plasma cells and other key intestinal B subsets in a manner compatible with several downstream applications, including sorting and culturing and in vitro manipulations, this efficient and powerful approach can enhance studies of mucosal immunity.


Assuntos
Imunoglobulina A , Plasmócitos , Animais , Camundongos , Linfócitos B , Colagenases/metabolismo , Mucosa , Mucosa Intestinal
3.
Cancer Res Commun ; 3(9): 1756-1769, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37674528

RESUMO

Mechanisms for Helicobacter pylori (Hp)-driven stomach cancer are not fully understood. In a transgenic mouse model of gastric preneoplasia, concomitant Hp infection and induction of constitutively active KRAS (Hp+KRAS+) alters metaplasia phenotypes and elicits greater inflammation than either perturbation alone. Gastric single-cell RNA sequencing showed that Hp+KRAS+ mice had a large population of metaplastic pit cells that expressed the intestinal mucin Muc4 and the growth factor amphiregulin. Flow cytometry and IHC-based immune profiling revealed that metaplastic pit cells were associated with macrophage and T-cell inflammation. Accordingly, expansion of metaplastic pit cells was prevented by gastric immunosuppression and reversed by antibiotic eradication of Hp. Finally, MUC4 expression was significantly associated with proliferation in human gastric cancer samples. These studies identify an Hp-associated metaplastic pit cell lineage, also found in human gastric cancer tissues, whose expansion is driven by Hp-dependent inflammation. Significance: Using a mouse model, we have delineated metaplastic pit cells as a precancerous cell type whose expansion requires Hp-driven inflammation. In humans, metaplastic pit cells show enhanced proliferation as well as enrichment in precancer and early cancer tissues, highlighting an early step in the gastric metaplasia to cancer cascade.


Assuntos
Helicobacter pylori , Neoplasias Gástricas , Humanos , Animais , Camundongos , Proteínas Proto-Oncogênicas p21(ras) , Modelos Animais de Doenças , Inflamação
5.
Cell Rep Med ; 2(6): 100324, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34195687

RESUMO

Necrotizing enterocolitis (NEC) is an intestinal disorder that disproportionately affects premature infants and lacks in effective therapeutics. Mihi and colleagues1 demonstrated that the cytokine interleukin-22 promotes intestinal epithelial regeneration and reduces disease severity in an experimental model of NEC.


Assuntos
Enterocolite Necrosante , Doenças Fetais , Doenças do Prematuro , Citocinas , Enterocolite Necrosante/tratamento farmacológico , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/tratamento farmacológico
6.
Cell Host Microbe ; 27(6): 849-851, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32526178

RESUMO

Childhood undernutrition is associated with dysbiosis and dampened vaccine responses. Understanding how nutrients influence the microbiota and immunity is critical for vaccine efficacy. In this issue of Cell Host & Microbe, Di Luccia et al. and Huus et al. reveal that nutrition affects IgA responses to the microbiota and oral vaccines.


Assuntos
Imunidade nas Mucosas , Microbiota , Bactérias , Criança , Disbiose , Humanos , Imunoglobulina A
7.
Elife ; 82019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31433298

RESUMO

B-1a cells play an important role in mediating tissue homeostasis and protecting against infections. They are the main producers of 'natural' IgM, spontaneously secreted serum antibodies predominately reactive to self antigens, like phosphatidylcholine (PtC), or antigens expressed by the intestinal microbiota. The mechanisms that regulate the B-1a immunoglobulin (Ig) repertoire and their antibody secretion remain poorly understood. Here, we use a novel reporter mouse to demonstrate that production of self- and microbiota-reactive antibodies is linked to BCR signaling in B-1a cells. Moreover, we show that Toll-like receptors (TLRs) are critical for shaping the Ig repertoire of B-1a cells as well as regulating their antibody production. Strikingly, we find that both the colonization of a microbiota as well as microbial-sensing TLRs are required for anti-microbiota B-1a responses, whereas nucleic-acid sensing TLRs are required for anti-PtC responses, demonstrating that linked activation of BCR and TLRs controls steady state B-1a responses to both self and microbiota-derived antigens.


Assuntos
Autoantígenos/imunologia , Subpopulações de Linfócitos B/imunologia , Fatores Imunológicos/metabolismo , Microbiota/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Animais , Camundongos , Proteínas Proto-Oncogênicas c-bcr/metabolismo
8.
Science ; 364(6446): 1179-1184, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31221858

RESUMO

Intestinal adaptive immune responses influence host health, yet only a few intestinal bacteria species that induce cognate adaptive immune responses during homeostasis have been identified. Here, we show that Akkermansia muciniphila, an intestinal bacterium associated with systemic effects on host metabolism and PD-1 checkpoint immunotherapy, induces immunoglobulin G1 (IgG1) antibodies and antigen-specific T cell responses in mice. Unlike previously characterized mucosal responses, T cell responses to A. muciniphila are limited to T follicular helper cells in a gnotobiotic setting, without appreciable induction of other T helper fates or migration to the lamina propria. However, A. muciniphila-specific responses are context dependent and adopt other fates in conventional mice. These findings suggest that, during homeostasis, contextual signals influence T cell responses to the microbiota and modulate host immune function.


Assuntos
Imunidade Adaptativa , Microbioma Gastrointestinal/imunologia , Homeostase , Intestinos/imunologia , Verrucomicrobia/imunologia , Animais , Movimento Celular/imunologia , Feminino , Vida Livre de Germes , Imunidade nas Mucosas , Imunoglobulina G/imunologia , Mucosa Intestinal/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Auxiliares-Indutores/imunologia
9.
Cell Host Microbe ; 24(5): 613-615, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30439335

RESUMO

Though critical for preventing fatal sepsis, the mechanisms mediating the capture of bloodstream bacteria are incompletely understood. New work by Zeng et al. (2018) demonstrates that estrogen-regulated innate antibodies protect females and newborns from death following bloodstream infection with enteropathogenic Eschericia coli.


Assuntos
Imunidade Inata/imunologia , Sepse/prevenção & controle , Fatores Sexuais , Animais , Anticorpos Antibacterianos/imunologia , Complemento C3/imunologia , Escherichia coli Enteropatogênica , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Estrogênios/imunologia , Feminino , Microbioma Gastrointestinal/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Fígado/imunologia , Fígado/metabolismo , Masculino , Troca Materno-Fetal/imunologia , Camundongos , Camundongos Knockout , Gravidez , Sepse/imunologia , Sepse/microbiologia , Soro/imunologia
10.
Cell Host Microbe ; 21(4): 419-420, 2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28407479

RESUMO

Commensal microbes colonize the skin where they promote immune development and prevent infection without inducing damaging inflammatory responses. In this issue of Cell Host & Microbe, Scharschmidt et al. (2017) show that during hair follicle development, commensals induce regulatory T cell migration to the skin to ensure cutaneous homeostasis.


Assuntos
Microbiota/imunologia , Simbiose/imunologia , Homeostase , Humanos , Pele/imunologia , Linfócitos T Reguladores/imunologia
11.
Cell ; 165(4): 827-41, 2016 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-27153495

RESUMO

To maintain a symbiotic relationship between the host and its resident intestinal microbiota, appropriate mucosal T cell responses to commensal antigens must be established. Mice acquire both IgG and IgA maternally; the former has primarily been implicated in passive immunity to pathogens while the latter mediates host-commensal mutualism. Here, we report the surprising observation that mice generate T cell-independent and largely Toll-like receptor (TLR)-dependent IgG2b and IgG3 antibody responses against their gut microbiota. We demonstrate that maternal acquisition of these antibodies dampens mucosal T follicular helper responses and subsequent germinal center B cell responses following birth. This work reveals a feedback loop whereby T cell-independent, TLR-dependent antibodies limit mucosal adaptive immune responses to newly acquired commensal antigens and uncovers a broader function for maternal IgG.


Assuntos
Animais Recém-Nascidos/imunologia , Microbioma Gastrointestinal , Imunidade nas Mucosas , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Leite Humano/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Animais Recém-Nascidos/microbiologia , Linfócitos B/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Receptores Toll-Like/imunologia
12.
J Exp Med ; 211(5): 961-74, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24711580

RESUMO

Regulatory T (T reg) cells play an essential role in preventing autoimmunity but can also impair clearance of foreign pathogens. Paradoxically, signals known to promote T reg cell function are abundant during infection and could inappropriately enhance T reg cell activity. How T reg cell function is restrained during infection to allow the generation of effective antiviral responses remains largely unclear. We demonstrate that the potent antiviral type I interferons (IFNs) directly inhibit co-stimulation-dependent T reg cell activation and proliferation, both in vitro and in vivo during acute infection with lymphocytic choriomeningitis virus (LCMV). Loss of the type I IFN receptor specifically in T reg cells results in functional impairment of virus-specific CD8(+) and CD4(+) T cells and inefficient viral clearance. Together, these data demonstrate that inhibition of T reg cells by IFNs is necessary for the generation of optimal antiviral T cell responses during acute LCMV infection.


Assuntos
Infecções por Arenaviridae/imunologia , Imunidade Celular/imunologia , Interferon Tipo I/metabolismo , Vírus da Coriomeningite Linfocítica/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T/imunologia , Transferência Adotiva , Animais , Primers do DNA/genética , Citometria de Fluxo , Imunidade Celular/efeitos dos fármacos , Interferon Tipo I/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/efeitos dos fármacos
13.
Cell Host Microbe ; 14(5): 488-90, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24237694

RESUMO

IgA antibodies help maintain intestinal immune homeostasis with resident commensal species; however, the precise mechanisms regulating IgA induction and the epitopes recognized by these antibodies remain incompletely understood. In this issue of Cell Host & Microbe, Tyler et al. (2013) demonstrate that TLR5-dependent induction of anti-flagellin antibodies prevents commensal association with the intestinal mucosa by limiting bacterial motility.


Assuntos
Flagelina/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunidade nas Mucosas , Locomoção , Microbiota/imunologia , Animais
14.
Immunity ; 37(3): 501-10, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22960221

RESUMO

Foxp3(+) regulatory T (Treg) cells limit inflammatory responses and maintain immune homeostasis. Although comprised of several phenotypically and functionally distinct subsets, the differentiation of specialized Treg cell populations within the periphery is poorly characterized. We demonstrate that the development of T-bet(+) Treg cells that potently inhibit T helper 1 (Th1) cell responses was dependent on the transcription factor STAT1 and occurred directly in response to interferon-γ produced by effector T cells. Additionally, delayed induction of the IL-12Rß2 receptor component after STAT1 activation helped ensure that Treg cells do not readily complete STAT4-dependent Th1 cell development and lose their ability to suppress effector T cell proliferation. Thus, we define a pathway of abortive Th1 cell development that results in the specialization of peripheral Treg cells and demonstrate that impaired expression of a single cytokine receptor helps maintain Treg cell-suppressive function in the context of inflammatory Th1 cell responses.


Assuntos
Diferenciação Celular , Subunidade beta 2 de Receptor de Interleucina-12/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Interferon gama/farmacologia , Interleucina-12/farmacologia , Subunidade beta 2 de Receptor de Interleucina-12/genética , Subunidade beta 2 de Receptor de Interleucina-12/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores CXCR3/genética , Receptores CXCR3/imunologia , Receptores CXCR3/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT4/imunologia , Fator de Transcrição STAT4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia , Proteínas com Domínio T/metabolismo , Linfócitos T Reguladores/metabolismo , Células Th1/metabolismo , Fatores de Tempo
16.
Nat Rev Immunol ; 11(2): 119-30, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21267013

RESUMO

Forkhead box P3 (FOXP3)(+) regulatory T (T(Reg)) cells prevent autoimmune disease, maintain immune homeostasis and modulate immune responses during infection. To accomplish these tasks, T(Reg) cell activity is precisely controlled, and this requires T(Reg) cells to alter their migratory, functional and homeostatic properties in response to specific cues in the immune environment. We review progress in understanding the diversity of T(Reg) cells, T(Reg) cell function in different anatomical and inflammatory settings, and the influence of the immune environment on T(Reg) cell activity. We also consider how these factors affect immune-mediated disease in the contexts of infection, autoimmunity, cancer and transplantation.


Assuntos
Imunidade Ativa , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Movimento Celular , Citocinas/metabolismo , Fatores de Transcrição Forkhead/análise , Homeostase , Humanos , Tecido Linfoide/imunologia
17.
Nat Immunol ; 10(6): 595-602, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19412181

RESUMO

Several subsets of Foxp3(+) regulatory T cells (T(reg) cells) work in concert to maintain immune homeostasis. However, the molecular bases underlying the phenotypic and functional diversity of T(reg) cells remain obscure. We show that in response to interferon-gamma, Foxp3(+) T(reg) cells upregulated the T helper type 1 (T(H)1)-specifying transcription factor T-bet. T-bet promoted expression of the chemokine receptor CXCR3 on T(reg) cells, and T-bet(+) T(reg) cells accumulated at sites of T(H)1 cell-mediated inflammation. Furthermore, T-bet expression was required for the homeostasis and function of T(reg) cells during type 1 inflammation. Thus, in a subset of CD4(+) T cells, the activities of the transcription factors Foxp3 and T-bet are overlaid, which results in T(reg) cells with unique homeostatic and migratory properties optimized for the suppression of T(H)1 responses in vivo.


Assuntos
Inflamação/imunologia , Proteínas com Domínio T/metabolismo , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Homeostase , Inflamação/metabolismo , Interferon gama/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CXCR3/metabolismo , Proteínas com Domínio T/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA