Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Cell Metab ; 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38959897

RESUMEN

A mechanistic connection between aging and development is largely unexplored. Through profiling age-related chromatin and transcriptional changes across 22 murine cell types, analyzed alongside previous mouse and human organismal maturation datasets, we uncovered a transcription factor binding site (TFBS) signature common to both processes. Early-life candidate cis-regulatory elements (cCREs), progressively losing accessibility during maturation and aging, are enriched for cell-type identity TFBSs. Conversely, cCREs gaining accessibility throughout life have a lower abundance of cell identity TFBSs but elevated activator protein 1 (AP-1) levels. We implicate TF redistribution toward these AP-1 TFBS-rich cCREs, in synergy with mild downregulation of cell identity TFs, as driving early-life cCRE accessibility loss and altering developmental and metabolic gene expression. Such remodeling can be triggered by elevating AP-1 or depleting repressive H3K27me3. We propose that AP-1-linked chromatin opening drives organismal maturation by disrupting cell identity TFBS-rich cCREs, thereby reprogramming transcriptome and cell function, a mechanism hijacked in aging through ongoing chromatin opening.

2.
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
3.
Clin Transl Immunology ; 12(10): e1470, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37799772

RESUMEN

Objectives: B cells drive the production of autoreactive antibody-secreting cells (ASCs) in autoimmune diseases such as Systemic Lupus Erythematosus (SLE) and Sjögren's syndrome, causing long-term organ damage. Current treatments for antibody-mediated autoimmune diseases target B cells or broadly suppress the immune system. However, pre-existing long-lived ASCs are often refractory to treatment, leaving a reservoir of autoreactive cells that continue to produce antibodies. Therefore, the development of novel treatment methods targeting ASCs is vital to improve patient outcomes. Our objective was to test whether targeting the epigenetic regulator BMI-1 could deplete ASCs in autoimmune conditions in vivo and in vitro. Methods: Use of a BMI-1 inhibitor in both mouse and human autoimmune settings was investigated. Lyn -/- mice, a model of SLE, were treated with the BMI-1 small molecule inhibitor PTC-028, before assessment of ASCs, serum antibody and immune complexes. To examine human ASC survival, a novel human fibroblast-based assay was established, and the impact of PTC-028 on ASCs derived from Sjögren's syndrome patients was evaluated. Results: BMI-1 inhibition significantly decreased splenic and bone marrow ASCs in Lyn -/- mice. The decline in ASCs was linked to aberrant cell cycle gene expression and led to a significant decrease in serum IgG3, immune complexes and anti-DNA IgG. PTC-028 was also efficacious in reducing ex vivo plasma cell survival from both Sjögren's syndrome patients and age-matched healthy donors. Conclusion: These data provide evidence that inhibiting BMI-1 can deplete ASC in a variety of contexts and thus BMI-1 is a viable therapeutic target for antibody-mediated autoimmune diseases.

4.
Nat Immunol ; 24(7): 1054-1055, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37322180

Asunto(s)
Encéfalo , Linfocitos
5.
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
7.
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.

8.
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
10.
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
11.
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
12.
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
13.
Curr Opin Immunol ; 64: 110-116, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32497868

RESUMEN

Fibroblastic reticular cells (FRCs) are a necessary immunological component for T cell health. These myofibroblasts are specialized for immune cell support and develop in locations where T and B lymphocyte priming occurs, usually secondary lymphoid organs, but also tertiary lymphoid structures and sites of chronic inflammation. This review describes their dual supportive and suppressive functions and emerging evidence on the co-ordination required to balance these competing roles.


Asunto(s)
Fibroblastos , Tejido Linfoide , Linfocitos B , Humanos , Sistema Linfático , Linfocitos T
14.
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
15.
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
16.
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
18.
19.
Nat Rev Immunol ; 15(6): 350-61, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25998961

RESUMEN

Over the past decade, a series of discoveries relating to fibroblastic reticular cells (FRCs) ­ immunologically specialized myofibroblasts found in lymphoid tissue ­ has promoted these cells from benign bystanders to major players in the immune response. In this Review, we focus on recent advances regarding the immunobiology of lymph node-derived FRCs, presenting an updated view of crucial checkpoints during their development and their dynamic control of lymph node expansion and contraction during infection. We highlight the robust effects of FRCs on systemic B cell and T cell responses, and we present an emerging view of FRCs as drivers of pathology following acute and chronic viral infections. Lastly, we review emerging therapeutic advances that harness the immunoregulatory properties of FRCs.


Asunto(s)
Linfocitos B/inmunología , Células Dendríticas Foliculares/inmunología , Infecciones/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Miofibroblastos/inmunología , Linfocitos T/inmunología , Animales , Comunicación Celular/fisiología , Movimiento Celular , Proliferación Celular/fisiología , Humanos , Tolerancia Inmunológica/inmunología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Modelos Inmunológicos , Miofibroblastos/citología , Virosis/inmunología
20.
Sci Transl Med ; 6(249): 249ra109, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25122637

RESUMEN

Sepsis is an aggressive inflammatory syndrome and a global health burden estimated to kill 7.3 million people annually. Single-target molecular therapies have not addressed the multiple disease pathways triggered by septic injury. Cell therapies might offer a broader set of mechanisms of action that benefit complex, multifocal disease processes. We describe a population of immune-specialized myofibroblasts derived from lymph node tissue, termed fibroblastic reticular cells (FRCs). Because FRCs have an immunoregulatory function in lymph nodes, we hypothesized that ex vivo-expanded FRCs would control inflammation when administered therapeutically. Indeed, a single injection of ex vivo-expanded allogeneic FRCs reduced mortality in mouse models of sepsis when administered at early or late time points after septic onset. Mice treated with FRCs exhibited lower local and systemic concentrations of proinflammatory cytokines and reduced bacteremia. When administered 4 hours after induction of lipopolysaccharide endotoxemia, or cecal ligation and puncture (CLP) sepsis in mice, FRCs reduced deaths by at least 70%. When administered late in disease (16 hours after CLP), FRCs still conveyed a robust survival advantage (44% survival compared to 0% for controls). FRC therapy was dependent on the metabolic activity of nitric oxide synthase 2 (NOS2) as the primary molecular mechanism of drug action in the mice. Together, these data describe a new anti-inflammatory cell type and provide preclinical evidence for therapeutic efficacy in severe sepsis that warrants further translational study.


Asunto(s)
Fibroblastos/trasplante , Ganglios Linfáticos/citología , Sepsis/terapia , Animales , Bacteriemia/patología , Ciego/patología , Movimiento Celular , Citocinas/sangre , Modelos Animales de Enfermedad , Endotoxemia/patología , Endotoxemia/terapia , Femenino , Ligadura , Lipopolisacáridos , Ratones , Óxido Nítrico Sintasa de Tipo II/metabolismo , Peritoneo/patología , Punciones , Sepsis/patología , Bazo/patología , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA