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











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 2307, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37085516

RESUMO

The intestinal lamina propria contains a diverse network of fibroblasts that provide key support functions to cells within their local environment. Despite this, our understanding of the diversity, location and ontogeny of fibroblasts within and along the length of the intestine remains incomplete. Here we show that the small and large intestinal lamina propria contain similar fibroblast subsets that locate in specific anatomical niches. Nevertheless, we find that the transcriptional profile of similar fibroblast subsets differs markedly between the small intestine and colon suggesting region specific functions. We perform in vivo transplantation and lineage-tracing experiments to demonstrate that adult intestinal fibroblast subsets, smooth muscle cells and pericytes derive from Gli1-expressing precursors present in embryonic day 12.5 intestine. Trajectory analysis of single cell RNA-seq datasets of E12.5 and adult mesenchymal cells suggest that adult smooth muscle cells and fibroblasts derive from distinct embryonic intermediates and that adult fibroblast subsets develop in a linear trajectory from CD81+ fibroblasts. Finally, we provide evidence that colonic subepithelial PDGFRαhi fibroblasts comprise several functionally distinct populations that originate from an Fgfr2-expressing fibroblast intermediate. Our results provide insights into intestinal stromal cell diversity, location, function, and ontogeny, with implications for intestinal development and homeostasis.


Assuntos
Intestino Grosso , Células-Tronco Mesenquimais , Colo , Fibroblastos/metabolismo , Intestino Grosso/anatomia & histologia , Intestino Grosso/citologia , Intestino Delgado , Intestinos/anatomia & histologia , Intestinos/citologia , Proteína GLI1 em Dedos de Zinco/genética , Células-Tronco Mesenquimais/metabolismo
2.
Nat Protoc ; 16(4): 2051-2067, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33619391

RESUMO

Gut-associated lymphoid tissues (GALTs) comprise key intestinal immune inductive sites, including the Peyer's patches of the small intestine and different types of isolated lymphoid follicle (ILF) found along the length of the gut. Our understanding of human GALT is limited due to a lack of protocols for their isolation. Here we describe a technique that, uniquely among intestinal cell isolation protocols, allows identification and isolation of all human GALT, as well as GALT-free intestinal lamina propria (LP). The technique involves the mechanical separation of intestinal mucosa from the submucosa, allowing the identification and isolation of submucosal ILF (SM-ILF), LP-embedded mucosal ILF (M-ILF) and LP free of contaminating lymphoid tissue. Individual SM-ILF, M-ILF and Peyer's patch follicles can be subsequently digested for downstream cellular and molecular characterization. The technique, which takes 4-10 h, will be useful for researchers interested in intestinal immune development and function in health and disease.


Assuntos
Trato Gastrointestinal/fisiologia , Tecido Linfoide/fisiologia , Técnicas de Cultura de Tecidos/métodos , Contagem de Células , Sobrevivência Celular , Colo/fisiologia , Doença de Crohn/patologia , Humanos , Imunidade Inata , Mucosa Intestinal/citologia , Antígenos Comuns de Leucócito/metabolismo
3.
Sci Signal ; 12(596)2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31455723

RESUMO

Second mitochondria-derived activator of caspase (SMAC) mimetics (SMs) are selective antagonists of the inhibitor of apoptosis proteins (IAPs), which activate noncanonical NF-κB signaling and promote tumor cell death. Through gene expression analysis, we found that treatment of CD4+ T cells with SMs during T helper 17 (TH17) cell differentiation disrupted the balance between two antagonistic transcription factor modules. Moreover, proteomics analysis revealed that SMs altered the abundance of proteins associated with cell cycle, mitochondrial activity, and the balance between canonical and noncanonical NF-κB signaling. Whereas SMs inhibited interleukin-17 (IL-17) production and ameliorated TH17 cell-driven inflammation, they stimulated IL-22 secretion. Mechanistically, SM-mediated activation of NF-κB-inducing kinase (NIK) and the transcription factors RelB and p52 directly suppressed Il17a expression and IL-17A protein production, as well as the expression of a number of other immune genes. Induction of IL-22 production correlated with the NIK-dependent reduction in cMAF protein abundance and the enhanced activity of the aryl hydrocarbon receptor. Last, SMs also increased IL-9 and IL-13 production and, under competing conditions, favored the differentiation of naïve CD4+ T cells into TH2 cells rather than TH17 cells. These results demonstrate that SMs shape the gene expression and protein profiles of TH17 cells and inhibit TH17 cell-driven autoimmunity.


Assuntos
Proteínas Reguladoras de Apoptose , Materiais Biomiméticos/farmacologia , Diferenciação Celular/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Mitocondriais , Proteínas Serina-Treonina Quinases/imunologia , Células Th17/imunologia , Animais , Regulação da Expressão Gênica/imunologia , Camundongos , Camundongos Transgênicos , Células Th17/citologia , Células Th2/citologia , Células Th2/imunologia , Quinase Induzida por NF-kappaB
4.
J Immunol ; 201(2): 524-532, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29848752

RESUMO

Despite the essential role of thymic epithelial cells (TEC) in T cell development, the signals regulating TEC differentiation and homeostasis remain incompletely understood. In this study, we show a key in vivo role for the vitamin A metabolite, retinoic acid (RA), in TEC homeostasis. In the absence of RA signaling in TEC, cortical TEC (cTEC) and CD80loMHC class IIlo medullary TEC displayed subset-specific alterations in gene expression, which in cTEC included genes involved in epithelial proliferation, development, and differentiation. Mice whose TEC were unable to respond to RA showed increased cTEC proliferation, an accumulation of stem cell Ag-1hi cTEC, and, in early life, a decrease in medullary TEC numbers. These alterations resulted in reduced thymic cellularity in early life, a reduction in CD4 single-positive and CD8 single-positive numbers in both young and adult mice, and enhanced peripheral CD8+ T cell survival upon TCR stimulation. Collectively, our results identify RA as a regulator of TEC homeostasis that is essential for TEC function and normal thymopoiesis.


Assuntos
Células Epiteliais/imunologia , Transdução de Sinais/imunologia , Timo/imunologia , Tretinoína/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Proliferação de Células/fisiologia , Feminino , Homeostase/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Gut ; 66(12): 2110-2120, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28615301

RESUMO

OBJECTIVE: Postoperative ileus (POI), the most frequent complication after intestinal surgery, depends on dendritic cells (DCs) and macrophages. Here, we have investigated the mechanism that activates these cells and the contribution of the intestinal microbiota for POI induction. DESIGN: POI was induced by manipulating the intestine of mice, which selectively lack DCs, monocytes or macrophages. The disease severity in the small and large intestine was analysed by determining the distribution of orally applied fluorescein isothiocyanate-dextran and by measuring the excretion time of a retrogradely inserted glass ball. The impact of the microbiota on intestinal peristalsis was evaluated after oral antibiotic treatment. RESULTS: We found that Cd11c-Cre+ Irf4flox/flox mice lack CD103+CD11b+ DCs, a DC subset unique to the intestine whose function is poorly understood. Their absence in the intestinal muscularis reduced pathogenic inducible nitric oxide synthase (iNOS) production by monocytes and macrophages and ameliorated POI. Pathogenic iNOS was produced in the jejunum by resident Ly6C- macrophages and infiltrating chemokine receptor 2-dependent Ly6C+ monocytes, but in the colon only by the latter demonstrating differential tolerance mechanisms along the intestinal tract. Consistently, depletion of both cell subsets reduced small intestinal POI, whereas the depletion of Ly6C+ monocytes alone was sufficient to prevent large intestinal POI. The differential role of monocytes and macrophages in small and large intestinal POI suggested a potential role of the intestinal microbiota. Indeed, antibiotic treatment reduced iNOS levels and ameliorated POI. CONCLUSIONS: Our findings reveal that CD103+CD11b+ DCs and the intestinal microbiome are a prerequisite for the activation of intestinal monocytes and macrophages and for dysregulating intestinal motility in POI.


Assuntos
Células Dendríticas/citologia , Microbioma Gastrointestinal , Íleus/imunologia , Íleus/microbiologia , Ativação de Macrófagos , Monócitos/imunologia , Peristaltismo/imunologia , Complicações Pós-Operatórias/imunologia , Complicações Pós-Operatórias/microbiologia , Animais , Antígenos CD/imunologia , Antígeno CD11b/imunologia , Modelos Animais de Doenças , Trânsito Gastrointestinal , Íleus/fisiopatologia , Cadeias alfa de Integrinas/imunologia , Camundongos , Camundongos Transgênicos , Complicações Pós-Operatórias/fisiopatologia
6.
Thorax ; 71(11): 1006-1011, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27329043

RESUMO

BACKGROUND: Alveolar macrophages (AMFs) are critical regulators of lung function, and may participate in graft rejection following lung transplantation. Recent studies in experimental animals suggest that most AMFs are self-maintaining cells of embryonic origin, but knowledge about the ontogeny and life span of human AMFs is scarce. METHODS: To follow the origin and longevity of AMFs in patients with lung transplantation for more than 100 weeks, we obtained transbronchial biopsies from 10 gender-mismatched patients with lung transplantation. These were subjected to combined in situ hybridisation for X/Y chromosomes and immunofluorescence staining for macrophage markers. Moreover, development of AMFs in humanised mice reconstituted with CD34+ umbilical cord-derived cells was assessed. RESULTS: The number of donor-derived AMFs was unchanged during the 2 year post-transplantation period. A fraction of the AMFs proliferated locally, demonstrating that at least a subset of human AMFs have the capacity to self-renew. Lungs of humanised mice were found to abundantly contain populations of human AMFs expressing markers compatible with a monocyte origin. Moreover, in patients with lung transplantation we found that recipient monocytes seeded the alveoli early after transplantation, and showed subsequent phenotypical changes consistent with differentiation into proliferating mature AMFs. This resulted in a stable mixed chimerism between donor and recipient AMFs throughout the 2-year period. CONCLUSIONS: The finding that human AMFs are maintained in the lung parenchyma for several years indicates that pulmonary macrophage transplantation can be a feasible therapeutic option for patients with diseases caused by dysfunctional AMFs. Moreover, in a lung transplantation setting, long-term persistence of donor AMFs may be important for the development of chronic graft rejection.


Assuntos
Transplante de Pulmão , Macrófagos Alveolares/patologia , Transplantados , Adulto , Animais , Biópsia , Feminino , Imunofluorescência , Rejeição de Enxerto/patologia , Humanos , Hibridização In Situ , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade
7.
Cell Rep ; 14(10): 2375-88, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26947077

RESUMO

Despite the key role of primary and secondary lymphoid organ stroma in immunity, our understanding of the heterogeneity and ontogeny of these cells remains limited. Here, we identify a functionally distinct subset of BP3(-)PDPN(+)PDGFRß(+)/α(+)CD34(+) stromal adventitial cells in both lymph nodes (LNs) and thymus that is located within the vascular niche surrounding PDPN(-)PDGFRß(+)/α(-)Esam-1(+)ITGA7(+) pericytes. CD34(+) adventitial cells developed in late embryonic thymus and in postnatal LNs and in the thymus originated, along with pericytes, from a common anlage-seeding progenitor population. Using lymphoid organ re-aggregate grafts, we demonstrate that adult CD34(+) adventitial cells are capable of differentiating into multiple lymphoid stroma-like subsets including pericyte-, FRC-, MRC-, and FDC-like cells, the development of which was lymphoid environment-dependent. These findings extend the current understanding of lymphoid mesenchymal cell heterogeneity and highlight a role of the CD34(+) adventitia as a potential ubiquitous source of lymphoid stromal precursors in postnatal tissues.


Assuntos
Antígenos CD34/metabolismo , Células Estromais/metabolismo , ADP-Ribosil Ciclase/genética , ADP-Ribosil Ciclase/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Citometria de Fluxo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Imuno-Histoquímica , Linfonodos/citologia , Linfonodos/metabolismo , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pericitos/citologia , Pericitos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Células Estromais/citologia , Timo/citologia , Timo/metabolismo
8.
Eur J Immunol ; 45(2): 574-83, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25521433

RESUMO

Thymus colonisation and thymocyte positioning are regulated by interactions between CCR7 and CCR9, and their respective ligands, CCL19/CCL21 and CCL25. The ligands of CCR7 and CCR9 also interact with the atypical receptor CCRL1 (also known as ACKR4), which is expressed in the thymus and has recently been reported to play an important role in normal αßT-cell development. Here, we show that CCRL1 is expressed within the thymic cortex, predominantly by MHC-II(low) CD40(-) cortical thymic epithelial cells and at the subcapsular zone by a population of podoplanin(+) thymic epithelial cells in mice. Interestingly, CCRL1 is also expressed by stromal cells which surround the pericytes of vessels at the corticomedullary junction, the site for progenitor cell entry and mature thymocyte egress from the thymus. We show that CCRL1 suppresses thymocyte progenitor entry into the thymus, however, the thymus size and cellularity are the same in adult WT and CCRL1(-/-) mice. Moreover, CCRL1(-/-) mice have no major perturbations in T-cell populations at different stages of thymic differentiation and development, and have a similar rate of thymocyte migration into the blood. Collectively, our findings argue against a major role for CCRL1 in normal thymus development and function.


Assuntos
Células Epiteliais/metabolismo , Linfopoese/genética , Receptores CCR/genética , Células Estromais/metabolismo , Timócitos/metabolismo , Timo/metabolismo , Animais , Antígenos CD40/deficiência , Antígenos CD40/genética , Antígenos CD40/imunologia , Diferenciação Celular , Movimento Celular , Microambiente Celular , Células Epiteliais/citologia , Células Epiteliais/imunologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Pericitos/citologia , Pericitos/imunologia , Receptores CCR/deficiência , Receptores CCR/imunologia , Receptores CCR7/genética , Receptores CCR7/imunologia , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/imunologia , Células Estromais/citologia , Células Estromais/imunologia , Timócitos/citologia , Timócitos/imunologia , Timo/citologia , Timo/crescimento & desenvolvimento , Timo/imunologia
9.
Eur J Immunol ; 43(12): 3098-107, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23966272

RESUMO

The intestinal mucosa is exposed to large amounts of foreign antigen (Ag) derived from commensal bacteria, dietary Ags, and intestinal pathogens. Dendritic cells (DCs) are believed to be involved in the induction of tolerance to harmless Ags and in mounting protective immune responses to pathogens and, as such, to play key roles in regulating intestinal immune homeostasis. The characterization of classical DCs (cDCs) in the intestinal lamina propria has been under intense investigation in recent years but the use of markers (including CD11c, CD11b, MHC class II), which are also expressed by intestinal MΦs, has led to some controversy regarding their definition. Here we review recent studies that help to distinguish cDCs subsets from monocyte-derived cells in the intestinal mucosa. We address the phenotype and ontogeny of these cDC subsets and highlight recent findings indicating that these subsets play distinct roles in the regulation of mucosal immune responses in vivo.


Assuntos
Antígenos de Diferenciação/imunologia , Células Dendríticas/imunologia , Imunidade nas Mucosas/fisiologia , Macrófagos/imunologia , Monócitos/imunologia , Mucosa/imunologia , Animais , Antígenos/imunologia , Células Dendríticas/citologia , Humanos , Tolerância Imunológica/fisiologia , Macrófagos/citologia , Monócitos/citologia , Mucosa/citologia
10.
J Immunol ; 188(10): 4801-9, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22504647

RESUMO

The vitamin A metabolite and transcriptional modulator retinoic acid (RA) is recognized as an important regulator of epithelial cell homeostasis in several tissues. Despite the known importance of the epithelial compartment of the thymus in T cell development and selection, the potential role of RA in the regulation of thymic cortical and medullary epithelial cell homeostasis has yet to be addressed. In this study, using fetal thymus organ cultures, we demonstrate that endogenous RA signaling promotes thymic epithelial cell (TEC) cell-cycle exit and restricts TEC cellularity preferentially in the cortical TEC compartment. Combined gene expression, biochemical, and functional analyses identified mesenchymal cells as the major source of RA in the embryonic thymus. In reaggregate culture experiments, thymic mesenchyme was required for RA-dependent regulation of TEC expansion, highlighting the importance of mesenchyme-derived RA in modulating TEC turnover. The RA-generating potential of mesenchymal cells was selectively maintained within a discrete Ly51(int)gp38(+) subset of Ly51(+) mesenchyme in the adult thymus, suggesting a continual role for mesenchymal cell-derived RA in postnatal TEC homeostasis. These findings identify RA signaling as a novel mechanism by which thymic mesenchyme influences TEC development.


Assuntos
Células Epiteliais/imunologia , Homeostase/imunologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Receptores do Ácido Retinoico/fisiologia , Timo/citologia , Timo/imunologia , Animais , Ciclo Celular/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/embriologia
11.
Blood ; 119(25): 6052-62, 2012 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-22442345

RESUMO

Mouse CD8α(+) dendritic cells (DCs) in lymphoid organs and CD103(+) CD11b(-) DCs in nonlymphoid tissues share phenotypic and functional similarities, as well as a unique shared developmental dependence on the transcription factor Batf3. Human DCs resembling mouse CD8α(+) DCs in phenotype and function have been identified in human blood, spleen, and tonsil. However, it is not clear whether such cells are also present in human nonlymphoid organs, and their equivalence to mouse CD8α(+) DC has recently been questioned. Furthermore, the identification of "CD8α(+) DC-like" cells across different tissues and species remains problematic because of the lack of a unique marker that can be used to unambiguously define lineage members. Here we show that mouse CD8α(+) DCs and CD103(+) CD11b(-) DCs can be defined by shared high expression of DNGR-1 (CLEC9A). We further show that DNGR-1 uniquely marks a CD11b(-) human DC population present in both lymphoid and nonlymphoid tissues of humans and humanized mice. Finally, we demonstrate that knockdown of Batf3 selectively prevents the development of DNGR-1(+) human DCs in vitro. Thus, high expression of DNGR-1 specifically and universally identifies a unique DC subset in mouse and humans. Evolutionarily conserved Batf3 dependence justifies classification of DNGR-1(hi) DCs as a distinct DC lineage.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Células Dendríticas/metabolismo , Lectinas Tipo C/fisiologia , Tecido Linfoide/metabolismo , Receptores Imunológicos/fisiologia , Proteínas Repressoras/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Biomarcadores/análise , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Células Dendríticas/fisiologia , Feminino , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Tecido Linfoide/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Especificidade de Órgãos/genética , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Especificidade da Espécie
12.
Immunity ; 33(1): 71-83, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20643338

RESUMO

In the Peyer's patches (PPs), germinal centers (GCs) are chronically induced by bacteria and are the major sites for generation of gut immunoglobulin A (IgA) immune responses. Whether follicular dendritic cells (FDCs) within the GCs directly contribute to the IgA production in PPs is unknown. We showed here that direct stimulation of FDCs by bacterial products and retinoic acid synergistically enhanced the expression of the chemokine CXCL13, the survival factor BAFF, and molecules that facilitate the secretion and activation of the cytokine TGF-beta1. A reduced production of these molecules by PP FDCs associated with deficiencies in the Toll-like receptor pathway or vitamin A resulted in decreased numbers of GC B cells and defective generation of IgA(+) B cells within PP GCs. Our data indicate that PP FDCs are conditioned by environmental stimuli to express key factors for B cell migration, survival, and preferential generation of IgA in gut.


Assuntos
Quimiocina CXCL13/metabolismo , Células Dendríticas Foliculares/metabolismo , Imunidade nas Mucosas , Imunoglobulina A/biossíntese , Fatores Imunológicos/farmacologia , Animais , Formação de Anticorpos , Fator Ativador de Células B/genética , Fator Ativador de Células B/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL13/genética , Células Dendríticas Foliculares/imunologia , Células Dendríticas Foliculares/patologia , Perfilação da Expressão Gênica , Centro Germinativo/patologia , Imunoglobulina A/genética , Camundongos , Nódulos Linfáticos Agregados/patologia , Transdução de Sinais , Receptores Toll-Like/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Deficiência de Vitamina A
13.
Immunobiology ; 215(9-10): 692-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20580119

RESUMO

Intestinal dendritic cell and macrophage subsets are believed to play key roles in maintaining intestinal homeostasis in the steady state and in driving protective immune responses in the setting of intestinal infection. This mini-review focuses on recent progress regarding the ontogeny and function of small intestinal lamina propria dendritic cell/macrophage subsets. In particular we discuss recent findings suggesting that small intestinal CD103(+) dendritic cells and Cx3cr1(+) cells derive from distinct precursor populations and that CD103(+) dendritic cells represent the major migratory population of cells with a key role in initiating adaptive immune responses in the draining mesenteric lymph node. In contrast, Cx3cr1(+) cells appear to represent a tissue resident population, phenotypically indistinguishable from tissue resident macrophages. These latter observations suggest an important division of labour between dendritic cell/macrophage subsets in the regulation of intestinal immune responses in the steady state.


Assuntos
Células Dendríticas/imunologia , Intestino Delgado/imunologia , Macrófagos/imunologia , Animais , Antígenos CD/biossíntese , Antígenos de Diferenciação/biossíntese , Receptor 1 de Quimiocina CX3C , Diferenciação Celular/imunologia , Linhagem da Célula , Movimento Celular , Imunidade nas Mucosas , Cadeias alfa de Integrinas/biossíntese , Camundongos , Receptores de Quimiocinas/biossíntese
14.
J Immunol ; 183(8): 5079-84, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19786532

RESUMO

Although CD4(+) memory T cells reside within secondary lymphoid tissue, the major reservoir of these cells is in the lamina propria of the intestine. In this study, we demonstrate that, in the absence of signals through both OX40 and CD30, CD4(+) T cells are comprehensively depleted from the lamina propria. Deficiency in either CD30 or OX40 alone reduced CD4(+) T cell numbers, however, in mice deficient in both OX40 and CD30, CD4(+) T cell loss was greatly exacerbated. This loss of CD4(+) T cells was not due to a homing defect because CD30 x OX40-deficient OTII cells were not impaired in their ability to express CCR9 and alpha(4)beta(7) or traffic to the small intestine. There was also no difference in the priming of wild-type (WT) and CD30 x OX40-deficient OTII cells in the mesenteric lymph node after oral immunization. However, following oral immunization, CD30 x OX40-deficient OTII cells trafficked to the lamina propria but failed to persist compared with WT OTII cells. This was not due to reduced levels of Bcl-2 or Bcl-XL, because expression of these was comparable between WT and double knockout OTII cells. Collectively, these data demonstrate that signals through CD30 and OX40 are required for the survival of CD4(+) T cells within the small intestine lamina propria.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Mucosa Intestinal/imunologia , Antígeno Ki-1/imunologia , Receptores OX40/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Sobrevivência Celular/imunologia , Mucosa Intestinal/metabolismo , Intestino Delgado/citologia , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Antígeno Ki-1/genética , Antígeno Ki-1/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores OX40/genética , Receptores OX40/metabolismo , Transdução de Sinais/imunologia , Baço/imunologia , Baço/metabolismo , Proteína bcl-X/imunologia , Proteína bcl-X/metabolismo
15.
J Leukoc Biol ; 83(1): 156-64, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17911179

RESUMO

The chemokine CCL25 is constitutively expressed in the thymus, and its receptor CCR9 is expressed on subsets of developing thymocytes. Nevertheless, the function of CCL25/CCR9 in adult thymopoiesis remains unclear. Here, we demonstrate that purified CCR9(-/-) hematopoietic stem cells are deficient in their ability to generate all major thymocyte subsets including double-negative 1 (DN1) cells in competitive transfers. CCR9(-/-) bone marrow contained normal numbers of lineage(-) Sca-1+c-kit+, common lymphoid progenitors, and lymphoid-primed multipotent progenitors (LMPP), and CCR9(-/-) LMPP showed similar T cell potential as their wild-type (WT) counterparts when cultured on OP9-delta-like 1 stromal cells. In contrast, early thymic progenitor and DN2 thymocyte numbers were reduced in the thymus of adult CCR9(-/-) mice. In fetal thymic organ cultures (FTOC), CCR9(-/-) DN1 cells were as efficient as WT DN1 cells in generating double-positive (DP) thymocytes; however, under competitive FTOC, CCR9(-/-) DP cell numbers were reduced significantly. Similarly, following intrathymic injection into sublethally irradiated recipients, CCR9(-/-) DN cells were out-competed by WT DN cells in generating DP thymocytes. Finally, in competitive reaggregation thymic organ cultures, CCR9(-/-) preselection DP thymocytes were disadvantaged significantly in their ability to generate CD4 single-positive (SP) thymocytes, a finding that correlated with a reduced ability to form TCR-MHC-dependent conjugates with thymic epithelial cells. Together, these results highlight a role for CCR9 at several stages of adult thymopoiesis: in hematopoietic progenitor seeding of the thymus, in the DN-DP thymocyte transition, and in the generation of CD4 SP thymocytes.


Assuntos
Linfopoese/imunologia , Receptores CCR/metabolismo , Linfócitos T/imunologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CCR/deficiência , Timo/citologia , Timo/imunologia
16.
Blood ; 110(8): 2955-64, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17540845

RESUMO

The molecular pathways regulating lymphoid priming, fate, and development of multipotent bone marrow (BM) stem/progenitor cells that continuously replace thymic progenitors remain largely unknown. Herein, we show that fms-like tyrosine kinase 3 (Flt3) ligand (Fl)-deficient mice have distinct reductions in the earliest thymic progenitors in fetal, postnatal, and adult thymus. A critical role of FL in thymopoiesis was particularly evident in the absence of interleukin-7 receptor alpha (IL-7Ralpha) signaling. Fl-/-Il-7r-/- mice have extensive reductions in fetal and postnatal thymic progenitors that result in a loss of active thymopoiesis in adult mice, demonstrating an indispensable role of FL in IL-7Ralpha-independent fetal and adult T lymphopoiesis. Moreover, we establish a unique and critical role of FL, distinct from that of IL-7Ralpha, in regulation of the earliest lineage-negative (Lin(-)) Lin(-)SCA1+KIT+ (LSK) FLT3(hi) lymphoid-primed multipotent progenitors in BM, demonstrating a key role of FLT3 signaling in regulating the very earliest stages of lymphoid progenitors.


Assuntos
Células-Tronco Hematopoéticas/citologia , Linfopoese/fisiologia , Proteínas de Membrana/metabolismo , Receptores de Interleucina-7/metabolismo , Linfócitos T/citologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Sequência de Bases , Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Feto , Citometria de Fluxo , Imunofluorescência , Expressão Gênica/imunologia , Células-Tronco Hematopoéticas/imunologia , Processamento de Imagem Assistida por Computador , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Timo/embriologia , Timo/imunologia , Timo/patologia
17.
Eur J Immunol ; 37(8): 2090-6, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17578846

RESUMO

Chemokine signaling has been implicated in directing colonization of the fetal thymus by hematopoietic precursors. However, the patterns of expression of the chemokine receptors responsible for directing thymic colonization by the earliest thymic migrants remain unknown. We have identified heterogeneity within the earliest thymus seeding cells based on chemokine receptor expression. By analyzing the first wave of progenitors to colonize the thymus at E12 of gestation, we show that multiple chemokine receptors are expressed by T-lymphoid precursors present within perithymic mesenchyme, while expression of chemokine ligands is limited to CCL21, CCL25 and CXCL12, which are located in distinct epithelial and mesenchymal compartments of the thymic/parathyroid anlagen. Collectively, these results identify multiple populations of T-lymphoid precursors colonizing the fetal thymus and provide evidence for several potential pathways mediating migration of precursors into the embryonic thymus.


Assuntos
Quimiotaxia de Leucócito/imunologia , Células-Tronco Hematopoéticas/imunologia , Receptores de Quimiocinas/biossíntese , Linfócitos T/imunologia , Timo/embriologia , Animais , Feto , Citometria de Fluxo , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Imuno-Histoquímica , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/citologia , Timo/imunologia
18.
Immunol Rev ; 215: 226-42, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17291292

RESUMO

The intestinal mucosa represents the largest body surface toward the external environment and harbors numerous T lymphocytes that take up resident within the intestinal epithelium or in the underlying lamina propria (LP). The intraepithelial lymphocytes include subsets of 'unconventional' T cells with unclear ontogeny and reactivity that localize to this site independently of antigen-specific activation in secondary lymphoid organs. In contrast, the majority of the 'conventional' gut T cells are recruited into the intestinal mucosa subsequent to their activation in intestinal inductive sites, including Peyer's patches (PPs) and mesenteric lymph nodes (MLNs). T cells homing to the small intestine express a distinct pattern of homing molecules, allowing them to interact with and transmigrate across intestinal postcapillary endothelium. At least some of these homing molecules, including the integrin alpha(4)beta(7) and the chemokine receptor CCR9, are induced on T cells during their activation in PPs or MLNs. Mucosal dendritic cells (DCs) play a key role in this process, but not all intestinal DCs possess the ability to confer a gut-homing capacity to T cells. Instead, functionally specialized CD103(+) DCs derived from the small intestinal LP appear to selectively regulate T-cell homing to the small intestine.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Quimiotaxia de Leucócito/imunologia , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Células Dendríticas/imunologia , Células Epiteliais/imunologia , Humanos , Mucosa Intestinal/citologia , Subpopulações de Linfócitos T/citologia
19.
Eur J Immunol ; 32(12): 3488-97, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12442331

RESUMO

The CC chemokine CCL25 (TECK) is selectively expressed in the thymus and small intestine, indicating a potential role in T lymphocyte development. In the present study we examined the role of CCL25 in the generation of the small-intestinal CD8alpha alpha(+)CD3(+) intraepithelial lymphocyte (IEL) compartment. CCL25 mRNA expression in the murine small intestine increased at three weeks of age and corresponded with the appearance of CD8alpha alpha(+)CD3(+) lymphocytes in the small-intestinal epithelium. Administration of monoclonal neutralizing anti-CCL25 antibody to two-week-old mice led to a approximately 50% reduction in the total number of CD8alpha alpha(+)TCRgamma delta(+) and CD8alpha alpha(+)TCRalpha beta(+) IEL at four weeks of age. Freshly isolated murine CD8alpha alpha(+)CD3(+) IEL migrated in response to CCL25 and expressed the CCL25 receptor, CCR9. Analysis of CCR9 expression on putative IEL precursor populations demonstrated the presence of both CCR9(-) and CCR9(+) cells and indicated that up-regulation of this receptor occurred during IEL precursor differentiation. Finally, data from wild-type and RAG(-/-) mice suggested that the reduction in CD8alpha alpha(+)CD3(+) IEL in anti-CCL25 antibody treated mice resulted primarily from defective maintenance and/or development of IEL precursors rather than a direct effect on mature CD8alpha alpha(+)CD3(+) IEL.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Quimiocinas CC/metabolismo , Intestino Delgado/citologia , Intestino Delgado/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Complexo CD3/metabolismo , Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/citologia , Sobrevivência Celular , Quimiocinas CC/antagonistas & inibidores , Quimiocinas CC/genética , Quimiotaxia de Leucócito , Células Epiteliais/citologia , Células Epiteliais/imunologia , Genes RAG-1 , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Subpopulações de Linfócitos T/citologia
20.
Eur J Immunol ; 32(1): 97-103, 2002 01.
Artigo em Inglês | MEDLINE | ID: mdl-11754008

RESUMO

Plasma cell differentiation is induced in vitro by lipopolysaccharide (LPS) stimulation but can be blocked by including anti-CD40 antibodies. Using subtractive cDNA hybridization we have identified the cell surface protein Ly6C as differentially expressed on B cells stimulated with LPS only. Ly6C has been shown to be expressed on certain T cell subsets and on subsets of macrophages and NK cells, but not on resting B cells. We show that Ly6C is up-regulated upon LPS stimulation of B cells in vitro and that this up-regulation is blocked by anti-CD40 or anti-Ig antibodies. Furthermore, ELISPOT analysis of cells sorted by magnetic-activated cell sorting show that Ly6C is expressed on ex vivo plasma cells from the spleen and bone marrow. Flow cytometric analysis showed that Ly6C is expressed on splenic plasma cells as well as on lamina propria plasma cells. Finally, Ly6C cross-linking positively up-regulated the amount of immunoglobulin produced by LPS-stimulated splenic B cells in vitro.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Expressão Gênica , Glicoproteínas de Membrana/genética , Fosfatidilinositóis/metabolismo , Animais , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/efeitos dos fármacos , Biomarcadores , Células da Medula Óssea/citologia , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Reagentes de Ligações Cruzadas , Intestino Delgado/citologia , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitógenos/farmacologia , Plasmócitos , Baço/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA