Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Immunity ; 44(3): 622-633, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26921107

RESUMO

Stromal cells generate a complex cellular scaffold that provides specialized microenvironments for lymphocyte activation in secondary lymphoid organs. Here, we assessed whether local activation of stromal cells in the central nervous system (CNS) is mandatory to transfer immune recognition from secondary lymphoid organs into the infected tissue. We report that neurotropic virus infection in mice triggered the establishment of such stromal cell niches in the CNS. CNS stromal cell activation was dominated by a rapid and vigorous production of CC-motif chemokine receptor (CCR) 7 ligands CCL19 and CCL21 by vascular endothelial cells and adjacent fibroblastic reticular cell (FRC)-like cells in the perivascular space. Moreover, CCR7 ligands produced by CNS stromal cells were crucial to support recruitment and local re-activation of antiviral CD8(+) T cells and to protect the host from lethal neuroinflammatory disease, indicating that CNS stromal cells generate confined microenvironments that control protective T cell immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Sistema Nervoso Central/imunologia , Endotélio Vascular/imunologia , Vírus da Hepatite A/imunologia , Hepatite A/imunologia , Inflamação Neurogênica/parasitologia , Receptores CCR7/metabolismo , Células Estromais/imunologia , Animais , Movimento Celular , Microambiente Celular , Sistema Nervoso Central/virologia , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Endotélio Vascular/virologia , Hepatite A/complicações , Imunidade Celular , Imunomodulação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Inflamação Neurogênica/etiologia , Receptores CCR7/genética , Células Estromais/virologia , Tropismo Viral
2.
J Immunol ; 210(6): 774-785, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36715496

RESUMO

Hallmarks of life-threatening, coronavirus-induced disease include dysregulated antiviral immunity and immunopathological tissue injury. Nevertheless, the sampling of symptomatic patients overlooks the initial inflammatory sequela culminating in severe coronavirus-induced disease, leaving a fundamental gap in our understanding of the early mechanisms regulating anticoronavirus immunity and preservation of tissue integrity. In this study, we delineate the innate regulators controlling pulmonary infection using a natural mouse coronavirus. Within hours of infection, the cellular landscape of the lung was transcriptionally remodeled altering host metabolism, protein synthesis, and macrophage maturation. Genetic perturbation revealed that these transcriptional programs were type I IFN dependent and critically controlled both host cell survival and viral spread. Unrestricted viral replication overshooting protective IFN responses culminated in increased IL-1ß and alarmin production and triggered compensatory neutrophilia, interstitial inflammation, and vascular injury. Thus, type I IFNs critically regulate early viral burden, which serves as an innate checkpoint determining the trajectory of coronavirus dissemination and immunopathology.


Assuntos
Infecções por Coronavirus , Interferon Tipo I , Vírus da Hepatite Murina , Pneumonia , Animais , Camundongos , Imunidade Inata , Antivirais/farmacologia , Replicação Viral
3.
Immunity ; 32(5): 714-25, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20451411

RESUMO

The transcription factor early B cell factor-1 (Ebf1) is a key determinant of B lineage specification and differentiation. To gain insight into the molecular basis of Ebf1 function in early-stage B cells, we combined a genome-wide ChIP sequencing analysis with gain- and loss-of-function transcriptome analyses. Among 565 genes that are occupied and transcriptionally regulated by Ebf1, we identified large sets involved in (pre)-B cell receptor and Akt signaling, cell adhesion, and migration. Interestingly, a third of previously described Pax5 targets was found to be occupied by Ebf1. In addition to Ebf1-activated and -repressed genes, we identified targets at which Ebf1 induces chromatin changes that poise the genes for expression at subsequent stages of differentiation. Poised chromatin states on specific targets could also be established by Ebf1 expression in T cells but not in NIH 3T3 cells, suggesting that Ebf1 acts as a "pioneer" factor in a hematopoietic chromatin context.


Assuntos
Linfócitos B/imunologia , Cromatina/genética , Redes Reguladoras de Genes , Ativação Linfocitária/imunologia , Fator de Transcrição PAX5/metabolismo , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Linhagem da Célula , Cromatina/química , Cromatina/metabolismo , Mapeamento Cromossômico , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Fator de Transcrição PAX5/genética , Regiões Promotoras Genéticas , Ligação Proteica , Transdução de Sinais
4.
Eur J Immunol ; 45(8): 2218-31, 2015 08.
Artigo em Inglês | MEDLINE | ID: mdl-25973789

RESUMO

The thymic epithelium forms specialized niches to enable thymocyte differentiation. While the common epithelial progenitor of medullary and cortical thymic epithelial cells (mTECs and cTECs) is well defined, early stages of mTEC lineage specification have remained elusive. Here, we utilized in vivo targeting of mTECs to resolve their differentiation pathways and to determine whether mTEC progenitors participate in thymocyte education. We found that mTECs descend from a lineage committed, podoplanin (PDPN)-expressing progenitor located at the cortico-medullary junction. PDPN(+) junctional TECs (jTECs) represent a distinct TEC population that builds the thymic medulla, but only partially supports negative selection and thymocyte differentiation. Moreover, conditional gene targeting revealed that abrogation of alternative NF-κB pathway signaling in the jTEC stage completely blocked mTEC development. Taken together, this study identifies jTECs as lineage-committed mTEC progenitors and shows that NF-κB-dependent progression of jTECs to mTECs is critical to secure central tolerance.


Assuntos
Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Glicoproteínas de Membrana/imunologia , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Células-Tronco/imunologia , Timo/imunologia , Animais , Diferenciação Celular/genética , Células Epiteliais/citologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , NF-kappa B/genética , Transdução de Sinais/genética , Células-Tronco/citologia , Timo/citologia
5.
J Immunol ; 190(8): 4162-74, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23509359

RESUMO

CD8(+) T cell memory inflation, first described in murine CMV (MCMV) infection, is characterized by the accumulation of high-frequency, functional Ag-specific CD8(+) T cell pools with an effector-memory phenotype and enrichment in peripheral organs. Although persistence of Ag is considered essential, the rules underpinning memory inflation are still unclear. The MCMV model is, however, complicated by the virus's low-level persistence and stochastic reactivation. We developed a new model of memory inflation based on a ß-galactosidase (ßgal)-recombinant adenovirus vector. After i.v. administration in C57BL/6 mice, we observed marked memory inflation in the ßgal96 epitope, whereas a second epitope, ßgal497, undergoes classical memory formation. The inflationary T cell responses show kinetics, distribution, phenotype, and functions similar to those seen in MCMV and are reproduced using alternative routes of administration. Memory inflation in this model is dependent on MHC class II. As in MCMV, only the inflating epitope showed immunoproteasome independence. These data define a new model for memory inflation, which is fully replication independent, internally controlled, and reproduces the key immunologic features of the CD8(+) T cell response. This model provides insight into the mechanisms responsible for memory inflation and, because it is based on a vaccine vector, also is relevant to novel T cell-inducing vaccines in humans.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Memória Imunológica , Transtornos da Memória/imunologia , Transtornos da Memória/virologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Linfócitos T CD8-Positivos/patologia , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/imunologia , Feminino , Vetores Genéticos/administração & dosagem , Memória Imunológica/genética , Masculino , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Muromegalovirus/genética , Muromegalovirus/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
6.
Proc Natl Acad Sci U S A ; 109(8): 3012-7, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22315415

RESUMO

Infections with persistent viruses are a frequent cause of immunosuppression, autoimmune sequelae, and/or neoplastic disease. Plasmacytoid dendritic cells (pDCs) are innate immune cells that produce type I interferon (IFN-I) and other cytokines in response to virus-derived nucleic acids. Persistent viruses often cause depletion or functional impairment of pDCs, but the role of pDCs in the control of these viruses remains unclear. We used conditional targeting of pDC-specific transcription factor E2-2 to generate mice that constitutively lack pDCs in peripheral lymphoid organs and tissues. The profound impact of pDC deficiency on innate antiviral responses was revealed by the failure to control acute infection with the cytopathic mouse hepatitis virus. Furthermore, pDC-deficient animals failed to clear lymphocytic choriomeningitis virus (LCMV) from hematopoietic organs during persistent LCMV infection. This failure was associated with reduced numbers and functionality of LCMV-specific CD4(+) helper T cells and impaired antiviral CD8(+) T-cell responses. Adoptive transfer of LCMV-specific T cells revealed that both CD4(+) and CD8(+) T cells required IFN-I for expansion, but only CD4(+) T cells required the presence of pDCs. In contrast, mice with pDC-specific loss of MHC class II expression supported normal CD4(+) T-cell response to LCMV. These data suggest that pDCs facilitate CD4(+) helper T-cell responses to persistent viruses independently of direct antigen presentation. Thus pDCs provide an essential link between innate and adaptive immunity to chronic viral infection, likely through the secretion of IFN-I and other cytokines.


Assuntos
Células Dendríticas/imunologia , Linfócitos T/imunologia , Viroses/imunologia , Viroses/virologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Doença Crônica , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Modelos Animais de Doenças , Marcação de Genes , Coriomeningite Linfocítica/sangue , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/prevenção & controle , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/imunologia , Fator de Transcrição 4
7.
Proc Natl Acad Sci U S A ; 109(4): 1233-8, 2012 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-22232667

RESUMO

Generation of antiviral IgM is usually considered as a marker of a short-lived initial antibody response that is replaced by hypermutated and more-efficient IgG. However, once viruses have established a particular niche for their persistence (e.g., within the CNS), the immune system has to specifically mobilize a broad range of antimicrobial effectors to contain the pathogen in the long term. Infection of the CNS with the mouse hepatitis virus (MHV) provides a unique model situation in which the extent of inflammatory CNS disease is determined by the balance between antiviral immune control, viral replication, and immune-mediated damage. We show here that whereas antibody- or B cell-deficient mice failed to contain MHV CNS infection and developed progressive demyelinating disease, germline IgM produced in activation-induced cytidine deaminase-deficient mice (aicda(-/-)) provided long-term protection against the chronic multiple sclerosis-like disease. Furthermore, we found that appropriate B-cell activation within the CNS-draining lymph node and subsequent CXCR3-mediated migration of antiviral IgM-secreting cells to the infected CNS was dependent on CD40-mediated interaction of B cells with T helper cells. These data indicate that the CD40-mediated collaboration of T and B cells is critical to secure neuroprotective IgM responses during viral CNS infection.


Assuntos
Antígenos CD40/imunologia , Sistema Nervoso Central/virologia , Doenças Desmielinizantes/virologia , Imunoglobulina M/imunologia , Vírus da Hepatite Murina/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Análise de Variância , Animais , Linfócitos B/imunologia , Sistema Nervoso Central/imunologia , Citidina Desaminase/deficiência , Doenças Desmielinizantes/imunologia , ELISPOT , Citometria de Fluxo , Imunofluorescência , Imunoglobulina M/genética , Camundongos , Camundongos Knockout
8.
Eur J Immunol ; 43(12): 3233-43, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23963995

RESUMO

Attenuation of T cell-mediated damage of blood endothelial cells (BECs) in transplanted organs is important to prevent transplant vasculopathy (TV) and chronic rejection. Here, we assessed the importance of minor histocompatibility antigen (mHA) distribution and different coinhibitory molecules for T cell-BEC interaction. A transgenic mHA was directed specifically to BECs using the Tie2 promoter and cellular interactions were assessed in graft-versus-host disease-like and heterotopic heart transplantation settings. We found that cognate CD4(+) T-cell help was critical for the activation of BEC-specific CD8(+) T cells. However, systemic mHA expression on BECs efficiently attenuated adoptively transferred, BEC-specific CD4(+) and CD8(+) T cells and hence prevented tissue damage, whereas restriction of mHA expression to heart BECs precipitated the development of TV. Importantly, the lack of the coinhibitory molecules programmed death-1 (PD-1) and B and T lymphocyte attenuator fostered the initial activation of BEC-specific CD4(+) T cells, but did not affect development of TV. In contrast, TV was significantly augmented in the absence of PD-1 on BEC-specific CD8(+) T cells. Taken together, these results indicate that antigen distribution in the vascular bed determines the impact of coinhibition and, as a consequence, critically impinges on T cell-mediated vascular immunopathology.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Endoteliais/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração , Antígenos de Histocompatibilidade Menor/imunologia , Doenças Vasculares/imunologia , Aloenxertos , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Rejeição de Enxerto/genética , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Camundongos , Camundongos Knockout , Antígenos de Histocompatibilidade Menor/biossíntese , Antígenos de Histocompatibilidade Menor/genética , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
9.
J Immunol ; 188(8): 3678-85, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22407917

RESUMO

Regulatory T cells (Tregs) are important for the attenuation of immune reactions. During viral CNS infections, however, an indiscriminate maintenance of CNS immune privilege through Treg-mediated negative regulation could prevent autoimmune sequelae but impair the control of viral replication. We analyzed in this study the impact of Tregs on the development of acute viral encephalomyelitis, T cell-mediated antiviral protection, and prevention of CNS autoimmunity following intranasal infection with the gliatropic mouse hepatitis virus strain A59. To assess the contribution of Tregs in vivo, we specifically depleted CD4(+)Foxp3(+) T cells in a diphtheria toxin-dependent manner. We found that depletion of Tregs had no impact on viral distribution and clearance and did not significantly alter virus-specific CD4(+) and CD8(+) T cell responses. However, Treg depletion led to a more severe CNS inflammation associated with neuronal damage. Dissection of the underlying immunopathological mechanisms revealed the elaborate Treg-dependent regulation of self-reactive CD4(+) T cell proliferation within the CNS-draining lymph node and downtuning of CXCR3 expression on T cells. Taken together, these results suggest that Tregs preserve CNS immune privilege through selective control of CNS-specific Th cells while keeping protective antiviral immunity fully operative.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções do Sistema Nervoso Central/imunologia , Infecções por Coronavirus/imunologia , Encefalomielite/imunologia , Vírus da Hepatite Murina/imunologia , Linfócitos T Reguladores/imunologia , Doença Aguda , Administração Intranasal , Animais , Autoantígenos/imunologia , Autoimunidade , Antígenos CD4/imunologia , Linfócitos T CD8-Positivos/patologia , Proliferação de Células , Infecções do Sistema Nervoso Central/virologia , Infecções por Coronavirus/virologia , Encefalomielite/virologia , Fatores de Transcrição Forkhead/imunologia , Humanos , Imunidade Celular , Linfonodos/imunologia , Linfonodos/patologia , Ativação Linfocitária , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Receptores CXCR3/imunologia , Linfócitos T Reguladores/patologia
10.
Eur J Immunol ; 40(3): 733-43, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20049875

RESUMO

Occlusive transplant vasculopathy (TV) is the major cause for chronic graft rejection. Since endothelial cells (EC) are the first graft cells encountered by activated host lymphocytes, it is important to delineate the molecular mechanisms that coordinate the interaction of EC with activated T cells. Here, the interaction of CD8(+) T cells with Ag-presenting EC in vivo was examined using a transgenic heart transplantation model with beta-galactosidase (beta-gal) expression exclusively in EC (Tie2-LacZ hearts). We found that priming with beta-gal peptide-loaded DC failed to generate a strong systemic IFN-gamma response, but elicited pronounced TV in both IFN-gamma receptor (IFNGR)-competent, and ifngr(-/-) Tie2-LacZ hearts. In contrast, stimulation of EC-specific CD8(+) T cells with beta-gal-recombinant mouse cytomegalovirus (MCMV-LacZ) in recipients of ifngr(+/+) Tie2-LacZ hearts did not precipitate significant TV. However, MCMV-LacZ infection of recipients of ifngr(-/-) Tie2-LacZ hearts led to massive activation of beta-gal-specific CD8 T cells, and led to development of fulminant TV. Further analyses revealed that the strong systemic IFN-gamma "storm" associated with MCMV infection induced upregulation of programmed death-1 ligand 1 (PD-L1) on EC, and subsequent attenuation of programmed death-1 (PD-1)-expressing EC-specific CD8(+) T cells. Thus, IFNGR signaling in ECs activates a potent peripheral negative feedback circuit that protects vascularized grafts from occlusive TV.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Endoteliais/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Receptores de Interferon/imunologia , Transdução de Sinais/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Separação Celular , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Ensaio de Imunoadsorção Enzimática , Retroalimentação Fisiológica , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Interferon/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor de Interferon gama
11.
PLoS One ; 9(10): e110576, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25333902

RESUMO

Reversible lysine acetylation plays an important role in the regulation of T cell responses. HDAC1 has been shown to control peripheral T helper cells, however the role of HDAC1 in CD8+ T cell function remains elusive. By using conditional gene targeting approaches, we show that LckCre-mediated deletion of HDAC1 led to reduced numbers of thymocytes as well as peripheral T cells, and to an increased fraction of CD8+CD4- cells within the CD3/TCRßlo population, indicating that HDAC1 is essential for the efficient progression of immature CD8+CD4- cells to the DP stage. Moreover, CD44hi effector CD8+ T cells were enhanced in mice with a T cell-specific deletion of HDAC1 under homeostatic conditions and HDAC1-deficient CD44hi CD8+ T cells produced more IFNγ upon ex vivo PMA/ionomycin stimulation in comparison to wild-type cells. Naïve (CD44l°CD62L+) HDAC1-null CD8+ T cells displayed a normal proliferative response, produced similar amounts of IL-2 and TNFα, slightly enhanced amounts of IFNγ, and their in vivo cytotoxicity was normal in the absence of HDAC1. However, T cell-specific loss of HDAC1 led to a reduced anti-viral CD8+ T cell response upon LCMV infection and impaired expansion of virus-specific CD8+ T cells. Taken together, our data indicate that HDAC1 is required for the efficient generation of thymocytes and peripheral T cells, for proper CD8+ T cell homeostasis and for an efficient in vivo expansion and activation of CD8+ T cells in response to LCMV infection.


Assuntos
Antivirais/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Histona Desacetilase 1/metabolismo , Animais , Antivirais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células , Receptores de Hialuronatos/metabolismo , Interferon gama/metabolismo , Interleucina-2/metabolismo , Ionomicina/farmacologia , Coriomeningite Linfocítica/tratamento farmacológico , Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Necrose Tumoral alfa/metabolismo
12.
J Exp Med ; 210(3): 465-73, 2013 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-23420877

RESUMO

The development of lymph nodes (LNs) and formation of LN stromal cell microenvironments is dependent on lymphotoxin-ß receptor (LTßR) signaling. In particular, the LTßR-dependent crosstalk between mesenchymal lymphoid tissue organizer and hematopoietic lymphoid tissue inducer cells has been regarded as critical for these processes. Here, we assessed whether endothelial cell (EC)-restricted LTßR signaling impacts on LN development and the vascular LN microenvironment. Using EC-specific ablation of LTßR in mice, we found that conditionally LTßR-deficient animals failed to develop a significant proportion of their peripheral LNs. However, remnant LNs showed impaired formation of high endothelial venules (HEVs). Venules had lost their cuboidal shape, showed reduced segment length and branching points, and reduced adhesion molecule and constitutive chemokine expression. Due to the altered EC-lymphocyte interaction, homing of lymphocytes to peripheral LNs was significantly impaired. Thus, this study identifies ECs as an important LTßR-dependent lymphoid tissue organizer cell population and indicates that continuous triggering of the LTßR on LN ECs is critical for lymphocyte homeostasis.


Assuntos
Células Endoteliais/fisiologia , Linfonodos/fisiologia , Receptor beta de Linfotoxina/fisiologia , Transdução de Sinais/fisiologia , Vênulas/fisiologia , Animais , Caderinas/fisiologia , Homeostase , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Transgenes
13.
Front Immunol ; 3: 279, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22969771

RESUMO

Vascular endothelial cells (ECs) form the inner layer of blood vessels and exert crucial functions during immune reactions including coagulation, inflammation, and regulation of innate immunity. Importantly, ECs can interact with T cells in an antigen-specific, i.e., T cell receptor-dependent manner. In this review, we will discuss EC actions and reactions during acute inflammation and focus on the interaction of T cells with ECs at two vascular sites: the high endothelial venule (HEV) of lymph nodes, and the vascular lesion during transplant vasculopathy (TV). HEVs are characterized by a highly active endothelium that produces chemoattracting factors and expresses adhesion molecules to facilitate transit of lymphocytes into the lymph node (LN) parenchyma. Yet, T cell-EC interaction at this anatomical location results neither in T cell activation nor tolerization. In contrast, the endothelium at sites of chronic inflammation, such as solid organ transplants, can promote T cell activation by upregulation of major histocompatibility complex (MHC) and costimulatory molecules. Importantly, a major function of ECs in inflamed tissues must be the maintenance of vascular integrity including the efficient attenuation of effector T cells that may damage the vascular bed. Thus, antigen-specific T cell-EC interaction is characterized by a tightly controlled balance between immunological ignorance, immune activation, and tolerization.

14.
Front Immunol ; 2: 50, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22566840

RESUMO

Stromal cells provide the structural foundation of secondary lymphoid organs (SLOs), and regulate leukocyte access and cell migration within the different compartments of spleen and lymph nodes (LNs). Furthermore, several stromal cell subsets have been implied in shaping of T cell responses through direct presentation of antigen. Despite significant gain of knowledge on the biology of different SLO-resident stromal cell subsets, their molecular and functional characterization has remained incomplete. To address this need, we have generated a bacterial artificial chromosome-transgenic mouse model that utilizes the podoplanin (pdpn) promoter to express the Cre-recombinase exclusively in stromal cells of SLOs. The characterization of the Pdpn-Cre mouse revealed transgene expression in subsets of fibroblastic reticular cells and lymphatic endothelial cells in LNs. Furthermore, the transgene facilitated the identification of a novel splenic perivascular stromal cell subpopulation that forms web-like structures around central arterioles. Assessment of the in vivo antigen expression in the genetically tagged stromal cells in Pdpn-Cre mice revealed activation of both MHC I and II-restricted TCR transgenic T cells. Taken together, stromal pdpn-Cre expression is well-suited to characterize the phenotype and to dissect the function of lymphoid organ stromal cells.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA