Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 4734, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34354077

RESUMO

The tumor microenvironment (TME) is a complex amalgam of tumor cells, immune cells, endothelial cells and fibroblastic stromal cells (FSC). Cancer-associated fibroblasts are generally seen as tumor-promoting entity. However, it is conceivable that particular FSC populations within the TME contribute to immune-mediated tumor control. Here, we show that intratumoral treatment of mice with a recombinant lymphocytic choriomeningitis virus-based vaccine vector expressing a melanocyte differentiation antigen resulted in T cell-dependent long-term control of melanomas. Using single-cell RNA-seq analysis, we demonstrate that viral vector-mediated transduction reprogrammed and activated a Cxcl13-expressing FSC subset that show a pronounced immunostimulatory signature and increased expression of the inflammatory cytokine IL-33. Ablation of Il33 gene expression in Cxcl13-Cre-positive FSCs reduces the functionality of intratumoral T cells and unleashes tumor growth. Thus, reprogramming of FSCs by a self-antigen-expressing viral vector in the TME is critical for curative melanoma treatment by locally sustaining the activity of tumor-specific T cells.


Assuntos
Melanoma Experimental/terapia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/patologia , Técnicas de Reprogramação Celular/métodos , Quimiocina CXCL13/genética , Quimiocina CXCL13/imunologia , Feminino , Vetores Genéticos , Interleucina-33/deficiência , Interleucina-33/genética , Interleucina-33/imunologia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/imunologia , Vírus da Coriomeningite Linfocítica/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Estromais/imunologia , Células Estromais/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Microambiente Tumoral/imunologia
2.
Nat Immunol ; 22(8): 1042-1051, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34267375

RESUMO

Pathogens and vaccines that produce persisting antigens can generate expanded pools of effector memory CD8+ T cells, described as memory inflation. While properties of inflating memory CD8+ T cells have been characterized, the specific cell types and tissue factors responsible for their maintenance remain elusive. Here, we show that clinically applied adenovirus vectors preferentially target fibroblastic stromal cells in cultured human tissues. Moreover, we used cell-type-specific antigen targeting to define critical cells and molecules that sustain long-term antigen presentation and T cell activity after adenovirus vector immunization in mice. While antigen targeting to myeloid cells was insufficient to activate antigen-specific CD8+ T cells, genetic activation of antigen expression in Ccl19-cre-expressing fibroblastic stromal cells induced inflating CD8+ T cells. Local ablation of vector-targeted cells revealed that lung fibroblasts support the protective function and metabolic fitness of inflating memory CD8+ T cells in an interleukin (IL)-33-dependent manner. Collectively, these data define a critical fibroblastic niche that underpins robust protective immunity operating in a clinically important vaccine platform.


Assuntos
Adenoviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Interleucina-33/imunologia , Ativação Linfocitária/imunologia , Células Estromais/imunologia , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Quimiocina CCL19/metabolismo , Quimera/genética , Epitopos de Linfócito T/imunologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Vetores Genéticos/imunologia , Humanos , Pulmão/citologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vacinação
3.
J Exp Med ; 218(5)2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33765133

RESUMO

Tissue-resident macrophages play a crucial role in maintaining homeostasis. Macrophage progenitors migrate to tissues perinatally, where environmental cues shape their identity and unique functions. Here, we show that the absence of PPARγ affects neonatal development and VCAM-1 expression of splenic iron-recycling red pulp macrophages (RPMs) and bone marrow erythroblastic island macrophages (EIMs). Transcriptome analysis of the few remaining Pparg-deficient RPM-like and EIM-like cells suggests that PPARγ is required for RPM and EIM identity, cell cycling, migration, and localization, but not function in mature RPMs. Notably, Spi-C, another transcription factor implicated in RPM development, was not essential for neonatal expansion of RPMs, even though the transcriptome of Spic-deficient RPMs was strongly affected and indicated a loss of identity. Similarities shared by Pparg- and Spic-deficient RPM-like cells allowed us to identify pathways that rely on both factors. PPARγ and Spi-C collaborate in inducing transcriptional changes, including VCAM-1 and integrin αD expression, which could be required for progenitor retention in the tissue, allowing access to niche-related signals that finalize differentiation.


Assuntos
Medula Óssea/imunologia , Eritroblastos/imunologia , Macrófagos/imunologia , PPAR gama/imunologia , Baço/imunologia , Animais , Medula Óssea/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Eritroblastos/citologia , Eritroblastos/metabolismo , Eritrócitos/citologia , Eritrócitos/imunologia , Eritrócitos/metabolismo , Regulação da Expressão Gênica , Ferro/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monócitos/imunologia , Monócitos/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Baço/citologia , Baço/metabolismo
4.
Sci Immunol ; 5(51)2020 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-32917792

RESUMO

Fibroblastic reticular cells (FRCs) are stromal cells that actively promote the induction of immune responses by coordinating the interaction of innate and adaptive immune cells. However, whether and to which extent immune cell activation is determined by lymph node FRC reprogramming during acute viral infection has remained unexplored. Here, we genetically ablated expression of the type I interferon-α receptor (Ifnar) in Ccl19-Cre+ cells and found that sensing of type I interferon imprints an antiviral state in FRCs and thereby preserves myeloid cell composition in lymph nodes of naive mice. During localized lymphocytic choriomeningitis virus infection, IFNAR signaling precipitated profound phenotypic adaptation of all FRC subsets enhancing antigen presentation, chemokine-driven immune cell recruitment, and immune regulation. The IFNAR-dependent shift of all FRC subsets toward an immunostimulatory state reduced exhaustive CD8+ T cell activation. In sum, these results unveil intricate circuits underlying type I IFN sensing in lymph node FRCs that enable protective antiviral immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Fibroblastos/imunologia , Interferon Tipo I/imunologia , Coriomeningite Linfocítica/imunologia , Células Estromais/imunologia , Animais , Linhagem Celular , Interferon gama/imunologia , Vírus da Coriomeningite Linfocítica , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/imunologia
5.
Nat Immunol ; 21(6): 649-659, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32424359

RESUMO

Efficient generation of germinal center (GC) responses requires directed movement of B cells between distinct microenvironments underpinned by specialized B cell-interacting reticular cells (BRCs). How BRCs are reprogrammed to cater to the developing GC remains unclear, and studying this process is largely hindered by incomplete resolution of the cellular composition of the B cell follicle. Here we used genetic targeting of Cxcl13-expressing cells to define the molecular identity of the BRC landscape. Single-cell transcriptomic analysis revealed that BRC subset specification was predetermined in the primary B cell follicle. Further topological remodeling of light and dark zone follicular dendritic cells required CXCL12-dependent crosstalk with B cells and dictated GC output by retaining B cells in the follicle and steering their interaction with follicular helper T cells. Together, our results reveal that poised BRC-defined microenvironments establish a feed-forward system that determines the efficacy of the GC reaction.


Assuntos
Escuridão , Células Dendríticas Foliculares/imunologia , Células Dendríticas Foliculares/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Imunomodulação/efeitos da radiação , Luz , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores , Comunicação Celular , Quimiocina CXCL12/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Análise de Célula Única , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
6.
Cell Rep ; 30(3): 893-904.e6, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31968261

RESUMO

Fibroblastic reticular cells (FRCs) form a road-like cellular network in lymph nodes (LNs) that provides essential chemotactic, survival, and regulatory signals for immune cells. While the topological characteristics of the FRC network have been elaborated, the network properties of the micro-tubular conduit system generated by FRCs, which drains lymph fluid through a pipeline-like system to distribute small molecules and antigens, has remained unexplored. Here, we quantify the crucial 3D morphometric parameters and determine the topological properties governing the structural organization of the intertwined networks. We find that the conduit system exhibits lesser small-worldness and lower resilience to perturbation compared to the FRC network, while the robust topological organization of both networks is maintained in a lymphotoxin-ß-receptor-independent manner. Overall, the high-resolution topological analysis of the "roads-and-pipes" networks highlights essential parameters underlying the functional organization of LN micro-environments and will, hence, advance the development of multi-scale LN models.


Assuntos
Linfonodos/anatomia & histologia , Linfonodos/fisiologia , Animais , Fibroblastos/citologia , Cinética , Receptor beta de Linfotoxina/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Transdução de Sinais
7.
Nat Commun ; 11(1): 519, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980640

RESUMO

Fibroblastic reticular cells (FRCs) are immunologically specialized myofibroblasts of lymphoid organ, and FRC maturation is essential for structural and functional properties of lymph nodes (LNs). Here we show that YAP and TAZ (YAP/TAZ), the final effectors of Hippo signaling, regulate FRC commitment and maturation. Selective depletion of YAP/TAZ in FRCs impairs FRC growth and differentiation and compromises the structural organization of LNs, whereas hyperactivation of YAP/TAZ enhances myofibroblastic characteristics of FRCs and aggravates LN fibrosis. Mechanistically, the interaction between YAP/TAZ and p52 promotes chemokine expression that is required for commitment of FRC lineage prior to lymphotoxin-ß receptor (LTßR) engagement, whereas LTßR activation suppresses YAP/TAZ activity for FRC maturation. Our findings thus present YAP/TAZ as critical regulators of commitment and maturation of FRCs, and hold promise for better understanding of FRC-mediated pathophysiologic processes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Fibroblastos/metabolismo , Linfonodos/citologia , Transativadores/metabolismo , Adipócitos/metabolismo , Animais , Quimiocinas/metabolismo , Fibroblastos/ultraestrutura , Linfonodos/ultraestrutura , Receptor beta de Linfotoxina/metabolismo , Mesoderma/metabolismo , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Proteínas de Sinalização YAP
8.
Science ; 366(6467): 881-886, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31727837

RESUMO

Myocarditis can develop into inflammatory cardiomyopathy through chronic stimulation of myosin heavy chain 6-specific T helper (TH)1 and TH17 cells. However, mechanisms governing the cardiotoxicity programming of heart-specific T cells have remained elusive. Using a mouse model of spontaneous autoimmune myocarditis, we show that progression of myocarditis to lethal heart disease depends on cardiac myosin-specific TH17 cells imprinted in the intestine by a commensal Bacteroides species peptide mimic. Both the successful prevention of lethal disease in mice by antibiotic therapy and the significantly elevated Bacteroides-specific CD4+ T cell and B cell responses observed in human myocarditis patients suggest that mimic peptides from commensal bacteria can promote inflammatory cardiomyopathy in genetically susceptible individuals. The ability to restrain cardiotoxic T cells through manipulation of the microbiome thereby transforms inflammatory cardiomyopathy into a targetable disease.


Assuntos
Doenças Autoimunes/complicações , Bacteroides/imunologia , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/microbiologia , Microbioma Gastrointestinal/imunologia , Miocardite/complicações , Peptídeos/imunologia , beta-Galactosidase/imunologia , Animais , Doenças Autoimunes/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Humanos , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Miocardite/imunologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/imunologia , Células Th17/imunologia
9.
Immunity ; 50(6): 1467-1481.e6, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31201093

RESUMO

Tissue-resident macrophages are receptive to specific signals concentrated in cellular niches that direct their cell differentiation and maintenance genetic programs. Here, we found that deficiency of the cytokine RANKL in lymphoid tissue organizers and marginal reticular stromal cells of lymph nodes resulted in the loss of the CD169+ sinusoidal macrophages (SMs) comprising the subcapsular and the medullary subtypes. Subcapsular SM differentiation was impaired in mice with targeted RANK deficiency in SMs. Temporally controlled RANK removal in lymphatic endothelial cells (LECs) revealed that lymphatic RANK activation during embryogenesis and shortly after birth was required for the differentiation of both SM subtypes. Moreover, RANK expression by LECs was necessary for SM restoration after inflammation-induced cell loss. Thus, cooperation between mesenchymal cells and LECs shapes a niche environment that supports SM differentiation and reconstitution after inflammation.


Assuntos
Citocinas/metabolismo , Linfonodos/citologia , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Células Estromais/metabolismo , Animais , Biomarcadores , Diferenciação Celular , Microambiente Celular , Imunofenotipagem , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Transdução de Sinais
10.
Trends Immunol ; 39(10): 775-787, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30150089

RESUMO

Lymph nodes (LNs) are strategically positioned outposts of the immune system that underpin regional immune surveillance. The current model describing LN formation in mice is based on a two cell-type interaction scheme with lymphoid tissue inducer cells regulating the activation of mesenchymal lymphoid tissue organizer cells. We highlight here the key role of lymphatic endothelial cells during the initiation of LN formation. The involvement of lymphatic endothelial cells as an additional organizer cell type in LN organogenesis unveils multiple control levels that govern the generation of lymphoid organs. Moreover, the linkage between lymphangiogenic and lymphvasculogenic processes and guidance of the accumulation and activation of lymphoid tissue inducer cells in the embryo suggests that LN formation may be driven on demand by developing organ systems.


Assuntos
Células Endoteliais/imunologia , Linfonodos/fisiologia , Células-Tronco Mesenquimais/imunologia , Modelos Imunológicos , Organogênese/fisiologia , Animais , Diferenciação Celular , Humanos , Vigilância Imunológica , Camundongos
11.
Sci Immunol ; 3(26)2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-30097537

RESUMO

Immune protection of the body cavities depends on the swift activation of innate and adaptive immune responses in nonclassical secondary lymphoid organs known as fat-associated lymphoid clusters (FALCs). Compared with classical secondary lymphoid organs such as lymph nodes and Peyer's patches, FALCs develop along distinct differentiation trajectories and display a reduced structural complexity. Although it is well established that fibroblastic reticular cells (FRCs) are an integral component of the immune-stimulating infrastructure of classical secondary lymphoid organs, the role of FRCs in FALC-dependent peritoneal immunity remains unclear. Using FRC-specific gene targeting, we found that FRCs play an essential role in FALC-driven immune responses. Specifically, we report that initiation of peritoneal immunity was governed through FRC activation in a myeloid differentiation primary response 88 (MYD88)-dependent manner. FRC-specific ablation of MYD88 blocked recruitment of inflammatory monocytes into FALCs and subsequent CD4+ T cell-dependent B-cell activation and IgG class switching. Moreover, containment of Salmonella infection was compromised in mice lacking MYD88 expression in FRCs, indicating that FRCs in FALCs function as an initial checkpoint in the orchestration of protective immune responses in the peritoneal cavity.


Assuntos
Fibroblastos/citologia , Fibroblastos/imunologia , Gordura Intra-Abdominal/imunologia , Cavidade Peritoneal/fisiologia , Animais , Quimiocina CCL2/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium , Fator de Necrose Tumoral alfa/imunologia
12.
Oncoimmunology ; 7(4): e1414129, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29632733

RESUMO

The tumor microenvironment harbors cancer-associated fibroblasts that function as major modulators of cancer progression. Here, we assessed to which extent distinct cancer-associated fibroblast subsets impact mammary carcinoma growth and cancer cell stemness in an orthotopic murine model. We found that fibroblasts expressing the Cre recombinase under the control of the interleukin 7 promoter occupied mainly the tumor margin where they physically interacted with tumor cells. Intratumoral ablation of interleukin 7-expressing fibroblasts impaired breast tumor growth and reduced the clonogenic potential of cancer cells. Moreover, cDNA expression profiling revealed a distinct oncogenic signature of interleukin 7-producing fibroblasts. In particular, Cxcl12 expression was strongly enhanced in interleukin 7-producing fibroblasts and cell type-specific genetic ablation and systemic pharmacological inhibition revealed that the CXCL12/CXCR4 axis impacts breast tumor cell stemness. Elevated expression of CXCL12 and other stem cell factors in primary human breast cancer-associated fibroblasts indicates that certain fibroblast populations support tumor cell stemness and thereby promote breast cancer growth.

13.
J Allergy Clin Immunol ; 142(4): 1257-1271.e4, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29391257

RESUMO

BACKGROUND: A particular characteristic of non-small cell lung cancer is the composition of the tumor microenvironment with a very high proportion of fibroblastic stromal cells (FSCs). OBJECTIVE: Lapses in our basic knowledge of fibroblast phenotype and function in the tumor microenvironment make it difficult to define whether FSC subsets exist that exhibit either tumor-promoting or tumor-suppressive properties. METHODS: We used gene expression profiling of lung versus tumor FSCs from patients with non-small cell lung cancer. Moreover, CCL19-expressing FSCs were studied in transgenic mouse models by using a lung cancer metastasis model. RESULTS: CCL19 mRNA expression in human tumor FSCs correlates with immune cell infiltration and intratumoral accumulation of CD8+ T cells. Mechanistic dissection in murine lung carcinoma models revealed that CCL19-expressing FSCs form perivascular niches to promote accumulation of CD8+ T cells in the tumor. Targeted ablation of CCL19-expressing tumor FSCs reduced immune cell recruitment and resulted in unleashed tumor growth. CONCLUSION: These data suggest that a distinct population of CCL19-producing FSCs fosters the development of an immune-stimulating intratumoral niche for immune cells to control cancer growth.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Quimiocina CCL19/imunologia , Fibroblastos/imunologia , Neoplasias Pulmonares/imunologia , Células Estromais/imunologia , Animais , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Quimiocina CCL19/genética , Humanos , Neoplasias Pulmonares/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T/transplante , Transcriptoma , Microambiente Tumoral/imunologia
14.
Immunity ; 47(1): 80-92.e4, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28709801

RESUMO

Lymph nodes (LNs) are strategically situated throughout the body at junctures of the blood vascular and lymphatic systems to direct immune responses against antigens draining from peripheral tissues. The current paradigm describes LN development as a programmed process that is governed through the interaction between mesenchymal lymphoid tissue organizer (LTo) cells and hematopoietic lymphoid tissue inducer (LTi) cells. Using cell-type-specific ablation of key molecules involved in lymphoid organogenesis, we found that initiation of LN development is dependent on LTi-cell-mediated activation of lymphatic endothelial cells (LECs) and that engagement of mesenchymal stromal cells is a succeeding event. LEC activation was mediated mainly by signaling through receptor activator of NF-κB (RANK) and the non-canonical NF-κB pathway and was steered by sphingosine-1-phosphate-receptor-dependent retention of LTi cells in the LN anlage. Finally, the finding that pharmacologically enforced interaction between LTi cells and LECs promotes ectopic LN formation underscores the central LTo function of LECs.


Assuntos
Células Endoteliais/fisiologia , Linfonodos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Organogênese , Animais , Diferenciação Celular , Células Cultivadas , Coristoma , Embrião de Mamíferos , Receptor beta de Linfotoxina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais
15.
J Clin Invest ; 127(4): 1574-1588, 2017 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-28319044

RESUMO

Alloimmune T cell responses induce graft-versus-host disease (GVHD), a serious complication of allogeneic bone marrow transplantation (allo-BMT). Although Notch signaling mediated by Delta-like 1/4 (DLL1/4) Notch ligands has emerged as a major regulator of GVHD pathogenesis, little is known about the timing of essential Notch signals and the cellular source of Notch ligands after allo-BMT. Here, we have shown that critical DLL1/4-mediated Notch signals are delivered to donor T cells during a short 48-hour window after transplantation in a mouse allo-BMT model. Stromal, but not hematopoietic, cells were the essential source of Notch ligands during in vivo priming of alloreactive T cells. GVHD could be prevented by selective inactivation of Dll1 and Dll4 in subsets of fibroblastic stromal cells that were derived from chemokine Ccl19-expressing host cells, including fibroblastic reticular cells and follicular dendritic cells. However, neither T cell recruitment into secondary lymphoid organs nor initial T cell activation was affected by Dll1/4 loss. Thus, we have uncovered a pathogenic function for fibroblastic stromal cells in alloimmune reactivity that can be dissociated from their homeostatic functions. Our results reveal what we believe to be a previously unrecognized Notch-mediated immunopathogenic role for stromal cell niches in secondary lymphoid organs after allo-BMT and define a framework of early cellular and molecular interactions that regulate T cell alloimmunity.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Linfonodos/patologia , Receptores Notch/fisiologia , Baço/patologia , Linfócitos T/imunologia , Aloenxertos , Animais , Transplante de Medula Óssea , Proteínas de Ligação ao Cálcio , Células Cultivadas , Feminino , Fibroblastos/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/patologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Ligantes , Linfonodos/imunologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Baço/imunologia , Linfócitos T/metabolismo
16.
Nat Immunol ; 17(12): 1388-1396, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27798617

RESUMO

Fibroblastic reticular cells (FRCs) of secondary lymphoid organs form distinct niches for interaction with hematopoietic cells. We found here that production of the cytokine IL-15 by FRCs was essential for the maintenance of group 1 innate lymphoid cells (ILCs) in Peyer's patches and mesenteric lymph nodes. Moreover, FRC-specific ablation of the innate immunological sensing adaptor MyD88 unleashed IL-15 production by FRCs during infection with an enteropathogenic virus, which led to hyperactivation of group 1 ILCs and substantially altered the differentiation of helper T cells. Accelerated clearance of virus by group 1 ILCs precipitated severe intestinal inflammatory disease with commensal dysbiosis, loss of intestinal barrier function and diminished resistance to colonization. In sum, FRCs act as an 'on-demand' immunological 'rheostat' by restraining activation of group 1 ILCs and thereby preventing immunopathological damage in the intestine.


Assuntos
Citrobacter rodentium/imunologia , Infecções por Coronavirus/imunologia , Infecções por Enterobacteriaceae/imunologia , Fibroblastos/imunologia , Interleucina-15/metabolismo , Linfócitos/imunologia , Vírus da Hepatite Murina/imunologia , Animais , Células Cultivadas , Imunidade Inata , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Nódulos Linfáticos Agregados/patologia , Células Th1/imunologia , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo
17.
Immunity ; 42(6): 981-3, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26084016

RESUMO

Atherosclerosis is a lipid-storage disease of arteries that is exacerbated by chronic inflammatory processes. In this issue of Immunity, Hu et al. (2015) demonstrate that T cell responses in atherosclerotic lesions are controlled in tertiary lymphoid organs in the arterial wall.


Assuntos
Envelhecimento/imunologia , Aterosclerose/imunologia , Receptor beta de Linfotoxina/metabolismo , Miócitos de Músculo Liso/fisiologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais
18.
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
19.
J Exp Med ; 211(11): 2265-79, 2014 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-25311507

RESUMO

Fibroblast-like cells of secondary lymphoid organs (SLO) are important for tissue architecture. In addition, they regulate lymphocyte compartmentalization through the secretion of chemokines, and participate in the orchestration of appropriate cell-cell interactions required for adaptive immunity. Here, we provide data demonstrating the functional importance of SLO fibroblasts during Notch-mediated lineage specification and immune response. Genetic ablation of the Notch ligand Delta-like (DL)1 identified splenic fibroblasts rather than hematopoietic or endothelial cells as niche cells, allowing Notch 2-driven differentiation of marginal zone B cells and of Esam(+) dendritic cells. Moreover, conditional inactivation of DL4 in lymph node fibroblasts resulted in impaired follicular helper T cell differentiation and, consequently, in reduced numbers of germinal center B cells and absence of high-affinity antibodies. Our data demonstrate previously unknown roles for DL ligand-expressing fibroblasts in SLO niches as drivers of multiple Notch-mediated immune differentiation processes.


Assuntos
Microambiente Celular/imunologia , Fibroblastos/metabolismo , Imunidade , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Receptores Notch/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores/metabolismo , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Quimiocina CCL19/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Imunofenotipagem , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Baço/imunologia , Baço/metabolismo , Células Estromais/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
20.
Nat Immunol ; 15(10): 973-81, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25151489

RESUMO

Fibroblastic reticular cells (FRCs) are known to inhabit T cell-rich areas of lymphoid organs, where they function to facilitate interactions between T cells and dendritic cells. However, in vivo manipulation of FRCs has been limited by a dearth of genetic tools that target this lineage. Here, using a mouse model to conditionally ablate FRCs, we demonstrated their indispensable role in antiviral T cell responses. Unexpectedly, loss of FRCs also attenuated humoral immunity due to impaired B cell viability and follicular organization. Follicle-resident FRCs established a favorable niche for B lymphocytes via production of the cytokine BAFF. Thus, our study indicates that adaptive immunity requires an intact FRC network and identifies a subset of FRCs that control B cell homeostasis and follicle identity.


Assuntos
Linfócitos B/imunologia , Fibroblastos/imunologia , Homeostase/imunologia , Linfócitos T/imunologia , Animais , Fator Ativador de Células B/imunologia , Fator Ativador de Células B/metabolismo , Linfócitos B/metabolismo , Movimento Celular/imunologia , Sobrevivência Celular/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fibroblastos/metabolismo , Citometria de Fluxo , Imunidade Humoral/imunologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA