Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Curr Opin Immunol ; 86: 102410, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38237251

RESUMEN

T-cell immunotherapy is now a first-line cancer treatment for metastatic melanoma and some lung cancer subtypes, which is a welcome clinical success. However, the response rates observed in these diseases are not yet replicated across other prominent solid tumour types, particularly stromal-rich subtypes with a complex microenvironment that suppresses infiltrating T cells. Cancer-associated fibroblasts (CAFs) are one of the most abundant and pro-pathogenic players in the tumour microenvironment, promoting tumour neogenesis, persistence and metastasis. Accumulating evidence is clear that CAFs subdue anti-tumour T-cell immunity and interfere with immunotherapy. CAFs can be grouped into different subtypes that operate synergistically to suppress T-cell function, including myofibroblastic CAFs, inflammatory CAFs and antigen-presenting CAFs, among other nomenclatures. Here, we review the mechanisms used by CAFs to induce T- cell tolerance and how these functions are likely to affect immunotherapy outcomes.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias , Humanos , Linfocitos T , Fibroblastos/patología , Fibroblastos Asociados al Cáncer/patología , Inmunidad Celular , Microambiente Tumoral
2.
Eur J Immunol ; 53(9): e2250355, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36991561

RESUMEN

The lymph node (LN) is home to resident macrophage populations that are essential for immune function and homeostasis, but key factors controlling this niche are undefined. Here, we show that fibroblastic reticular cells (FRCs) are an essential component of the LN macrophage niche. Genetic ablation of FRCs caused rapid loss of macrophages and monocytes from LNs across two in vivo models. Macrophages co-localized with FRCs in human LNs, and murine single-cell RNA-sequencing revealed that FRC subsets broadly expressed master macrophage regulator CSF1. Functional assays containing purified FRCs and monocytes showed that CSF1R signaling was sufficient to support macrophage development. These effects were conserved between mouse and human systems. These data indicate an important role for FRCs in maintaining the LN parenchymal macrophage niche.


Asunto(s)
Fibroblastos , Transducción de Señal , Ratones , Humanos , Animales , Macrófagos , Ganglios Linfáticos
3.
Front Digit Health ; 3: 704584, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34713176

RESUMEN

Three-dimensional (3D) cancer models are invaluable tools designed to study tumour biology and new treatments. Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest types of cancer, has been progressively explored with bioengineered 3D approaches by deconstructing elements of its tumour microenvironment. Here, we investigated the suitability of collagen-nanocellulose hydrogels to mimic the extracellular matrix of PDAC and to promote the formation of tumour spheroids and multicellular 3D cultures with stromal cells. Blending of type I collagen fibrils and cellulose nanofibres formed a matrix of controllable stiffness, which resembled the lower profile of pancreatic tumour tissues. Collagen-nanocellulose hydrogels supported the growth of tumour spheroids and multicellular 3D cultures, with increased metabolic activity and matrix stiffness. To validate our 3D cancer model, we tested the individual and combined effects of the anti-cancer compound triptolide and the chemotherapeutics gemcitabine and paclitaxel, resulting in differential cell responses. Our blended 3D matrices with tuneable mechanical properties consistently maintain the growth of PDAC cells and its cellular microenvironment and allow the screening of anti-cancer treatments.

4.
Immunity ; 54(8): 1628-1630, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34380060

RESUMEN

Fibroblasts are the immunological architects of lymph nodes. In this issue of Immunity, Mourcin et al. describe the human tonsil fibroblast landscape and predicted T and B cell interactions. Transcriptomic changes in follicular lymphoma could provide untapped clinical targets.


Asunto(s)
Linfoma Folicular , Fibroblastos , Humanos , Ganglios Linfáticos , Tonsila Palatina
5.
Immunol Rev ; 302(1): 299-320, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34164824

RESUMEN

Fibroblasts, custodians of tissue architecture and function, are no longer considered a monolithic entity across tissues and disease indications. Recent advances in single-cell technologies provide an unrestricted, high-resolution view of fibroblast heterogeneity that exists within and across tissues. In this review, we summarize a compendium of single-cell transcriptomic studies and provide a comprehensive accounting of fibroblast subsets, many of which have been described to occupy specific niches in tissues at homeostatic and pathologic states. Understanding this heterogeneity is particularly important in the context of cancer, as the diverse cancer-associated fibroblast (CAF) phenotypes in the tumor microenvironment (TME) are directly impacted by the expression phenotypes of their predecessors. Relationships between these heterogeneous populations often accompany and influence response to therapy in cancer and fibrosis. We further highlight the importance of integrating single-cell studies to deduce common fibroblast phenotypes across disease states, which will facilitate the identification of common signaling pathways, gene regulatory programs, and cell surface markers that are going to advance drug discovery and targeting.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias , Biomarcadores , Fibroblastos , Humanos , Neoplasias/genética , Neoplasias/terapia , Microambiente Tumoral
6.
Int J Mol Sci ; 22(4)2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33673197

RESUMEN

T cell immunotherapy is now a mainstay therapy for several blood-borne cancers as well as metastatic melanoma. Unfortunately, many epithelial tumors respond poorly to immunotherapy, and the reasons for this are not well understood. Cancer-associated fibroblasts (CAFs) are the most frequent non-neoplastic cell type in most solid tumors, and they are emerging as a key player in immunotherapy resistance. A range of immortalized CAF lines will be essential tools that will allow us to understand immune responses against cancer and develop novel strategies for cancer immunotherapy. To study the effect of CAFs on T cell proliferation, we created and characterized a number of novel immortalized human CAFs lines (Im-CAFs) from human breast, colon, and pancreatic carcinomas. Im-CAFs shared similar phenotypes, matrix remodeling and contraction capabilities, and growth and migration rates compared to the primary CAFs. Using primary isolates from breast carcinoma, colorectal carcinoma, and pancreatic ductal adenocarcinoma, we report that CAFs across major tumor types are able to potently suppress T cell proliferation in vitro. Im-CAFs retained this property. Im-CAFs are a key tool that will provide important insights into the mechanisms of CAF-mediated T cell suppression through techniques such as CRISPR-Cas9 modification, molecular screens, and pipeline drug testing.


Asunto(s)
Fibroblastos Asociados al Cáncer/inmunología , Proliferación Celular , Neoplasias/inmunología , Linfocitos T/inmunología , Fibroblastos Asociados al Cáncer/patología , Línea Celular Transformada , Humanos , Neoplasias/patología , Linfocitos T/patología
7.
J Immunol ; 206(2): 310-320, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33397745

RESUMEN

Over the past decade, T cell immunotherapy has changed the face of cancer treatment, providing robust treatment options for several previously intractable cancers. Unfortunately, many epithelial tumors with high mortality rates respond poorly to immunotherapy, and an understanding of the key impediments is urgently required. Cancer-associated fibroblasts (CAFs) comprise the most frequent nonneoplastic cellular component in most solid tumors. Far from an inert scaffold, CAFs significantly influence tumor neogenesis, persistence, and metastasis and are emerging as a key player in immunotherapy resistance. In this review, we discuss the physical and chemical barriers that CAFs place between effector T cells and their tumor cell targets, and the therapies poised to target them.


Asunto(s)
Fibroblastos Asociados al Cáncer/inmunología , Inmunoterapia/tendencias , Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Carcinogénesis , Humanos , Metástasis de la Neoplasia
8.
Cancer Immunol Res ; 6(12): 1472-1485, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30266714

RESUMEN

Cancer-associated fibroblasts (CAFs) are generally associated with poor clinical outcome. CAFs support tumor growth in a variety of ways and can suppress antitumor immunity and response to immunotherapy. However, a precise understanding of CAF contributions to tumor growth and therapeutic response is lacking. Discrepancies in this field of study may stem from heterogeneity in the composition and function of fibroblasts in the tumor microenvironment. Furthermore, it remains unclear whether CAFs directly interact with and suppress T cells. Here, mouse and human breast tumors were used to examine stromal cells expressing fibroblast activation protein (FAP), a surface marker for CAFs. Two discrete populations of FAP+ mesenchymal cells were identified on the basis of podoplanin (PDPN) expression: a FAP+PDPN+ population of CAFs and a FAP+PDPN- population of cancer-associated pericytes (CAPs). Although both subsets expressed extracellular matrix molecules, the CAF transcriptome was enriched in genes associated with TGFß signaling and fibrosis compared with CAPs. In addition, CAFs were enriched at the outer edge of the tumor, in close contact with T cells, whereas CAPs were localized around vessels. Finally, FAP+PDPN+ CAFs suppressed the proliferation of T cells in a nitric oxide-dependent manner, whereas FAP+PDPN- pericytes were not immunosuppressive. Collectively, these findings demonstrate that breast tumors contain multiple populations of FAP-expressing stromal cells of dichotomous function, phenotype, and location.


Asunto(s)
Neoplasias de la Mama/patología , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Células del Estroma/metabolismo , Microambiente Tumoral/inmunología , Animales , Neoplasias de la Mama/inmunología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Proliferación Celular , Endopeptidasas , Femenino , Regulación de la Expresión Génica , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Pericitos/metabolismo , Pericitos/patología , Células del Estroma/patología , Linfocitos T/patología
9.
PLoS Biol ; 16(9): e2005046, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30180168

RESUMEN

The microenvironment of lymphoid organs can aid healthy immune function through provision of both structural and molecular support. In mice, fibroblastic reticular cells (FRCs) create an essential T-cell support structure within lymph nodes, while human FRCs are largely unstudied. Here, we show that FRCs create a regulatory checkpoint in human peripheral T-cell activation through 4 mechanisms simultaneously utilised. Human tonsil and lymph node-derived FRCs constrained the proliferation of both naïve and pre-activated T cells, skewing their differentiation away from a central memory T-cell phenotype. FRCs acted unilaterally without requiring T-cell feedback, imposing suppression via indoleamine-2,3-dioxygenase, adenosine 2A Receptor, prostaglandin E2, and transforming growth factor beta receptor (TGFßR). Each mechanistic pathway was druggable, and a cocktail of inhibitors, targeting all 4 mechanisms, entirely reversed the suppressive effect of FRCs. T cells were not permanently anergised by FRCs, and studies using chimeric antigen receptor (CAR) T cells showed that immunotherapeutic T cells retained effector functions in the presence of FRCs. Since mice were not suitable as a proof-of-concept model, we instead developed a novel human tissue-based in situ assay. Human T cells stimulated using standard methods within fresh tonsil slices did not proliferate except in the presence of inhibitors described above. Collectively, we define a 4-part molecular mechanism by which FRCs regulate the T-cell response to strongly activating events in secondary lymphoid organs while permitting activated and CAR T cells to utilise effector functions. Our results define 4 feasible strategies, used alone or in combinations, to boost primary T-cell responses to infection or cancer by pharmacologically targeting FRCs.


Asunto(s)
Diferenciación Celular/inmunología , Microambiente Celular , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/citología , Adulto , Proliferación Celular , Niño , Fibroblastos/citología , Humanos , Memoria Inmunológica , Fenotipo
10.
J Exp Med ; 215(4): 1227-1243, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29549115

RESUMEN

Germinal centers (GCs) are the sites where B cells undergo affinity maturation. The regulation of cellular output from the GC is not well understood. Here, we show that from the earliest stages of the GC response, plasmablasts emerge at the GC-T zone interface (GTI). We define two main factors that regulate this process: Tfh-derived IL-21, which supports production of plasmablasts from the GC, and TNFSF13 (APRIL), which is produced by a population of podoplanin+ CD157high fibroblastic reticular cells located in the GTI that are also rich in message for IL-6 and chemokines CXCL12, CCL19, and CCL21. Plasmablasts in the GTI express the APRIL receptor TNFRSF13B (TACI), and blocking TACI interactions specifically reduces the numbers of plasmablasts appearing in the GTI. Plasma cells generated in the GTI may provide an early source of affinity-matured antibodies that may neutralize pathogens or provide feedback regulating GC B cell selection.


Asunto(s)
Centro Germinal/citología , Células Plasmáticas/metabolismo , Transducción de Señal , Células del Estroma/citología , Linfocitos T Colaboradores-Inductores/citología , Animales , Antígenos/metabolismo , Diferenciación Celular , Movimiento Celular , Quimiocinas/metabolismo , Regulación de la Expresión Génica , Inmunidad , Factores Reguladores del Interferón/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Ligandos , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células del Estroma/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
11.
12.
Mol Ther ; 22(5): 999-1007, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24496384

RESUMEN

The secreted proteins from a cell constitute a natural biologic library that can offer significant insight into human health and disease. Discovering new secreted proteins from cells is bounded by the limitations of traditional separation and detection tools to physically fractionate and analyze samples. Here, we present a new method to systematically identify bioactive cell-secreted proteins that circumvent traditional proteomic methods by first enriching for protein candidates by differential gene expression profiling. The bone marrow stromal cell secretome was analyzed using enriched gene expression datasets in combination with potency assay testing. Four proteins expressed by stromal cells with previously unknown anti-inflammatory properties were identified, two of which provided a significant survival benefit to mice challenged with lethal endotoxic shock. Greater than 85% of secreted factors were recaptured that were otherwise undetected by proteomic methods, and remarkable hit rates of 18% in vitro and 9% in vivo were achieved.


Asunto(s)
Proteínas Contráctiles/genética , Proteínas Contráctiles/metabolismo , Encefalinas/genética , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/genética , Interleucina-10/metabolismo , Precursores de Proteínas/genética , Proteínas/metabolismo , Choque Séptico/terapia , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Encefalinas/metabolismo , Proteínas de la Matriz Extracelular/genética , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular , Interleucina-10/genética , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Biosíntesis de Proteínas/genética , Precursores de Proteínas/metabolismo , Proteínas/genética , Proteómica , Factores de Empalme de ARN , Choque Séptico/genética
13.
Cell Mol Life Sci ; 71(7): 1305-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23955570

RESUMEN

Ablation of tetraspanin protein TSPAN12 from human MDA-MB-231 cells significantly decreased primary tumor xenograft growth, while increasing tumor apoptosis. Furthermore, TSPAN12 removal markedly enhanced tumor-endothelial interactions and increased metastasis to mouse lungs. TSPAN12 removal from human MDA-MB-231 cells also caused diminished association between FZD4 (a key canonical Wnt pathway receptor) and its co-receptor LRP5. The result likely explains substantially enhanced proteosomal degradation of ß-catenin, a key effecter of canonical Wnt signaling. Consistent with disrupted canonical Wnt signaling, TSPAN12 ablation altered expression of LRP5, Naked 1 and 2, DVL2, DVL3, Axin 1, and GSKß3 proteins. TSPAN12 ablation also altered expression of several genes regulated by ß-catenin (e.g. CCNA1, CCNE2, WISP1, ID4, SFN, ME1) that may help to explain altered tumor growth and metastasis. In conclusion, these results provide the first evidence for TSPAN12 playing a role in supporting primary tumor growth and suppressing metastasis. TSPAN12 appears to function by stabilizing FZD4-LRP5 association, in support of canonical Wnt-pathway signaling, leading to enhanced ß-catenin expression and function.


Asunto(s)
Neoplasias de la Mama/patología , Tetraspaninas/fisiología , beta Catenina/metabolismo , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Femenino , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Ratones SCID , Metástasis de la Neoplasia/genética , Tetraspaninas/genética , Tetraspaninas/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt
14.
Immunol Rev ; 251(1): 160-76, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23278748

RESUMEN

Secondary lymphoid organs (SLOs), including lymph nodes, Peyer's patches, and the spleen, have evolved to bring cells of the immune system together. In these collaborative environments, lymphocytes scan the surfaces of antigen-presenting cells for cognate antigens, while moving along stromal networks. The cell-cell interactions between stromal and hematopoietic cells in SLOs are therefore integral to the normal functioning of these tissues. Not only do stromal cells physically construct SLO architecture but they are essential for regulating hematopoietic populations within these domains. Stromal cells interact closely with lymphocytes and dendritic cells, providing scaffolds on which these cells migrate, and recruiting them into niches by secreting chemokines. Within lymph nodes, stromal cell-ensheathed conduit networks transport small antigens deep into the SLO parenchyma. More recently, stromal cells have been found to induce peripheral CD8(+) T-cell tolerance and control the extent to which newly activated T cells proliferate within lymph nodes. Thus, stromal-hematopoietic crosstalk has important consequences for regulating immune cell function within SLOs. In addition, stromal cell interactions with hematopoietic cells, other stroma, and the inflammatory milieu have profound effects on key stromal functions. Here, we examine ways in which these interactions within the lymph node environment influence the adaptive immune response.


Asunto(s)
Inmunidad Adaptativa , Células Madre Hematopoyéticas/inmunología , Ganglios Linfáticos/inmunología , Animales , Presentación de Antígeno , Comunicación Celular/inmunología , Movimiento Celular/inmunología , Quimiocinas/inmunología , Humanos , Activación de Linfocitos , Células del Estroma/inmunología
15.
Immunity ; 37(2): 276-89, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22884313

RESUMEN

To initiate adaptive immunity, dendritic cells (DCs) move from parenchymal tissues to lymphoid organs by migrating along stromal scaffolds that display the glycoprotein podoplanin (PDPN). PDPN is expressed by lymphatic endothelial and fibroblastic reticular cells and promotes blood-lymph separation during development by activating the C-type lectin receptor, CLEC-2, on platelets. Here, we describe a role for CLEC-2 in the morphodynamic behavior and motility of DCs. CLEC-2 deficiency in DCs impaired their entry into lymphatics and trafficking to and within lymph nodes, thereby reducing T cell priming. CLEC-2 engagement of PDPN was necessary for DCs to spread and migrate along stromal surfaces and sufficient to induce membrane protrusions. CLEC-2 activation triggered cell spreading via downregulation of RhoA activity and myosin light-chain phosphorylation and triggered F-actin-rich protrusions via Vav signaling and Rac1 activation. Thus, activation of CLEC-2 by PDPN rearranges the actin cytoskeleton in DCs to promote efficient motility along stromal surfaces.


Asunto(s)
Movimiento Celular/fisiología , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Actinas/metabolismo , Inmunidad Adaptativa/fisiología , Animales , Células Presentadoras de Antígenos/metabolismo , Plaquetas/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Embrión de Mamíferos , Células Endoteliales/metabolismo , Endotelio Linfático/citología , Endotelio Linfático/metabolismo , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Cadenas Ligeras de Miosina/metabolismo , Activación Plaquetaria , Embarazo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal/fisiología , Piel/citología , Piel/metabolismo , Técnicas de Cultivo de Tejidos , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
16.
PLoS One ; 7(8): e42677, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22880080

RESUMEN

Recent evidence suggests that the decline in resistance to viral infections with age occurs predominantly as a result of a gradual loss of naïve antigen-specific T cells. As such, restoration of the naïve T cell repertoire to levels seen in young healthy adults may improve defence against infection in the aged. We have previously shown that sex steroid ablation (SSA) rejuvenates the ageing thymus and increases thymic export of naïve T cells, but it remains unclear whether T cell responses are improved. Using mouse models of clinically relevant diseases, we now demonstrate that SSA increases the number of naïve T cells able to respond to antigen, thereby enhancing effector responses in aged mice. Specifically, aged mice exhibit a delay in clearing influenza A virus, which correlates with diminished specific cytotoxic activity. This is due to a decreased magnitude of response and not an intrinsic defect in effector T cell function. Upon SSA, aged mice exhibit increased T cell responsiveness that restores efficient viral clearance. We further demonstrate that SSA decreases the incidence of an inducible tumour in aged mice and can potentially increase their responsiveness to a low-dose human papillomavirus vaccine in clearing pre-formed tumours. As thymectomy abrogates the increase in T cell numbers and responsiveness following SSA, we propose that the T cell effects of SSA are dependent on thymic reactivation and subsequent replenishment of the peripheral T cell pool with newly emigrated naïve T cells. These findings have important implications for strategies to improve protection from infection and responsiveness to vaccination in the aged.


Asunto(s)
Envejecimiento/inmunología , Vacunas contra el Cáncer/inmunología , Hormonas Esteroides Gonadales/deficiencia , Vacunas contra la Influenza/inmunología , Neoplasias/inmunología , Animales , Castración , Citotoxicidad Inmunológica , Relación Dosis-Respuesta Inmunológica , Hormonas Esteroides Gonadales/metabolismo , Humanos , Memoria Inmunológica/inmunología , Virus de la Influenza A/inmunología , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Regeneración , Linfocitos T Citotóxicos/inmunología , Timo/inmunología , Timo/patología , Vacunación
17.
Nat Immunol ; 13(5): 499-510, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22466668

RESUMEN

Lymph node stromal cells (LNSCs) closely regulate immunity and self-tolerance, yet key aspects of their biology remain poorly elucidated. Here, comparative transcriptomic analyses of mouse LNSC subsets demonstrated the expression of important immune mediators, growth factors and previously unknown structural components. Pairwise analyses of ligands and cognate receptors across hematopoietic and stromal subsets suggested a complex web of crosstalk. Fibroblastic reticular cells (FRCs) showed enrichment for higher expression of genes relevant to cytokine signaling, relative to their expression in skin and thymic fibroblasts. LNSCs from inflamed lymph nodes upregulated expression of genes encoding chemokines and molecules involved in the acute-phase response and the antigen-processing and antigen-presentation machinery. Poorly studied podoplanin (gp38)-negative CD31(-) LNSCs showed similarities to FRCs but lacked expression of interleukin 7 (IL-7) and were identified as myofibroblastic pericytes that expressed integrin α(7). Together our data comprehensively describe the transcriptional characteristics of LNSC subsets.


Asunto(s)
Expresión Génica/inmunología , Inflamación/inmunología , Ganglios Linfáticos/inmunología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Transcriptoma , Reacción de Fase Aguda/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos CD/inmunología , Antígenos CD/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Homeostasis/inmunología , Inflamación/genética , Cadenas alfa de Integrinas/inmunología , Cadenas alfa de Integrinas/metabolismo , Interleucina-7/inmunología , Interleucina-7/metabolismo , Ganglios Linfáticos/citología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Pericitos/inmunología , Pericitos/metabolismo , Autotolerancia/inmunología , Análisis de Matrices Tisulares/métodos
18.
Mol Ther ; 20(1): 178-86, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21952165

RESUMEN

Mesenchymal stem cells (MSCs) are emerging as a promising immunotherapeutic, based largely on their overt suppression of T lymphocytes under inflammatory and autoimmune conditions. While paracrine cross-talk between MSCs and T cells has been well-studied, an intrinsic transcriptional switch that programs MSCs for immunomodulation has remained undefined. Here we show that bone marrow-derived MSCs require the transcriptional regulator Aire to suppress T cell-mediated pathogenesis in a mouse model of chronic colitis. Surprisingly, Aire did not control MSC suppression of T cell proliferation in vitro. Instead, Aire reduced T cell mitochondrial reductase by negatively regulating a proinflammatory cytokine, early T cell activation factor (Eta)-1. Neutralization of Eta-1 enabled Aire(-/-) MSCs to ameliorate colitis, reducing the number of infiltrating effector T cells in the colon, and normalizing T cell reductase levels. We propose that Aire represents an early molecular switch imposing a suppressive MSC phenotype via regulation of Eta-1. Monitoring Aire expression in MSCs may thus be a critical parameter for clinical use.


Asunto(s)
Enfermedad de Crohn/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factores de Transcripción/metabolismo , Animales , Técnicas de Cocultivo , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Femenino , Humanos , Terapia de Inmunosupresión , Inflamación/inmunología , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteopontina/genética , Osteopontina/metabolismo , Oxidación-Reducción , Linfocitos T/inmunología , Factores de Transcripción/genética , Transcripción Genética , Proteína AIRE
19.
Nat Immunol ; 12(11): 1096-104, 2011 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-21926986

RESUMEN

Fibroblastic reticular cells (FRCs) and lymphatic endothelial cells (LECs) are nonhematopoietic stromal cells of lymphoid organs. They influence the migration and homeostasis of naive T cells; however, their influence on activated T cells remains undescribed. Here we report that FRCs and LECs inhibited T cell proliferation through a tightly regulated mechanism dependent on nitric oxide synthase 2 (NOS2). Expression of NOS2 and production of nitric oxide paralleled the activation of T cells and required a tripartite synergism of interferon-γ, tumor necrosis factor and direct contact with activated T cells. Notably, in vivo expression of NOS2 by FRCs and LECs regulated the size of the activated T cell pool. Our study elucidates an as-yet-unrecognized role for the lymph node stromal niche in controlling T cell responses.


Asunto(s)
Selección Clonal Mediada por Antígenos , Endotelio Linfático/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Células del Estroma/metabolismo , Linfocitos T/metabolismo , Animales , Procesos de Crecimiento Celular/genética , Movimiento Celular/genética , Células Cultivadas , Endotelio Linfático/inmunología , Endotelio Linfático/patología , Uniones Intercelulares/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ganglios Linfáticos/patología , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Células del Estroma/inmunología , Células del Estroma/patología , Linfocitos T/inmunología , Linfocitos T/patología , Transgenes/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
20.
Crit Rev Immunol ; 31(3): 171-87, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21740349

RESUMEN

In essence, normal thymus function involves the production of a broad repertoire of αßT cells capable of responding to foreign antigens with low risk of autoreactivity. Thymic epithelial cells are an essential component of the thymic stromal microenvironment, promoting the growth and export of self-tolerant thymocytes. Autoimmune disease, resulting from a loss of self-tolerance, is clinically and genetically complex, and accordingly has many potential etiological origins. However, it is commonly linked to defects in the thymic epithelial microenvironment. The study of autoimmune-linked thymic stromal dysfunction has indisputably advanced our understanding of T cell tolerance; notably, a field-wide paradigm shift occurred when autoimmune regulator (Aire) was found to drive expression of a multitude of peripheral tissue-restricted antigens in medullary thymic epithelial cells. Many other associations with polygenically controlled autoimmune diseases have been reported but are more difficult to definitively dissect. Paradoxically, immunodeficiency and age-related immunosenescence are also linked with increased autoimmunity. Here we discuss the theoretical basis and the evidence gathered thus far to support these associations.


Asunto(s)
Envejecimiento/inmunología , Enfermedades Autoinmunes/inmunología , Células Epiteliales/inmunología , Autotolerancia/inmunología , Células del Estroma/inmunología , Timo , Factores de Transcripción/metabolismo , Envejecimiento/metabolismo , Antígenos/inmunología , Antígenos/metabolismo , Enfermedades Autoinmunes/metabolismo , Autoinmunidad/inmunología , Inmunodeficiencia Variable Común/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Células Epiteliales/metabolismo , Humanos , Células del Estroma/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Timo/inmunología , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Proteína AIRE
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA