Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2826: 219-230, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39017896

RESUMO

One way memory B cells provide protection is by rapidly differentiating into plasma cells. Plasma cells are vital in providing long-term protection against pathogens; however, they can also be detrimental to health in the case of antibody-mediated autoimmunity. Therefore, compounds which modulate the survival of plasma cells have been of interest for therapeutic intervention. Investigation of ex vivo plasma cell survival has previously been limited by the low frequency of plasma cells in the blood. Here we describe a novel ex vivo culture system that only requires 3000-5000 cells per condition. This method permits the assessment of human plasma cell survival derived from blood and can assess the impact of small molecule inhibitors on plasma cell viability.


Assuntos
Sobrevivência Celular , Plasmócitos , Humanos , Plasmócitos/imunologia , Plasmócitos/citologia , Plasmócitos/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cultura de Células/métodos , Células Cultivadas , Citometria de Fluxo/métodos
2.
Cell Metab ; 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38959897

RESUMO

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.

3.
Curr Opin Immunol ; 86: 102410, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38237251

RESUMO

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.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias , Humanos , Linfócitos T , Fibroblastos/patologia , Fibroblastos Associados a Câncer/patologia , Imunidade Celular , Microambiente Tumoral
4.
Clin Transl Immunology ; 12(10): e1470, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37799772

RESUMO

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.

6.
Eur J Immunol ; 53(9): e2250355, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36991561

RESUMO

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.


Assuntos
Fibroblastos , Transdução de Sinais , Camundongos , Humanos , Animais , Macrófagos , Linfonodos
8.
Front Digit Health ; 3: 704584, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34713176

RESUMO

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.

9.
Immunity ; 54(8): 1628-1630, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34380060

RESUMO

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.


Assuntos
Linfoma Folicular , Fibroblastos , Humanos , Linfonodos , Tonsila Palatina
11.
Immunol Rev ; 302(1): 299-320, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34164824

RESUMO

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.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias , Biomarcadores , Fibroblastos , Humanos , Neoplasias/genética , Neoplasias/terapia , Microambiente Tumoral
12.
Int J Mol Sci ; 22(4)2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673197

RESUMO

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.


Assuntos
Fibroblastos Associados a Câncer/imunologia , Proliferação de Células , Neoplasias/imunologia , Linfócitos T/imunologia , Fibroblastos Associados a Câncer/patologia , Linhagem Celular Transformada , Humanos , Neoplasias/patologia , Linfócitos T/patologia
13.
J Immunol ; 206(2): 310-320, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33397745

RESUMO

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.


Assuntos
Fibroblastos Associados a Câncer/imunologia , Imunoterapia/tendências , Neoplasias/imunologia , Linfócitos T/imunologia , Animais , Carcinogênese , Humanos , Metástase Neoplásica
14.
Curr Opin Immunol ; 64: 110-116, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32497868

RESUMO

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.


Assuntos
Fibroblastos , Tecido Linfoide , Linfócitos B , Humanos , Sistema Linfático , Linfócitos T
15.
Cancer Immunol Res ; 6(12): 1472-1485, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30266714

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Gelatinases/metabolismo , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Células Estromais/metabolismo , Microambiente Tumoral/imunologia , Animais , Neoplasias da Mama/imunologia , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Proliferação de Células , Endopeptidases , Feminino , Regulação da Expressão Gênica , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Pericitos/metabolismo , Pericitos/patologia , Células Estromais/patologia , Linfócitos T/patologia
16.
PLoS Biol ; 16(9): e2005046, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30180168

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Microambiente Celular , Linfonodos/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/citologia , Adulto , Proliferação de Células , Criança , Fibroblastos/citologia , Humanos , Memória Imunológica , Fenótipo
17.
J Exp Med ; 215(4): 1227-1243, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29549115

RESUMO

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.


Assuntos
Centro Germinativo/citologia , Plasmócitos/metabolismo , Transdução de Sinais , Células Estromais/citologia , Linfócitos T Auxiliares-Indutores/citologia , Animais , Antígenos/metabolismo , Diferenciação Celular , Movimento Celular , Quimiocinas/metabolismo , Regulação da Expressão Gênica , Imunidade , Fatores Reguladores de Interferon/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Ligantes , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Estromais/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
19.
Cell Death Differ ; 23(12): 1899-1901, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27689877
20.
Nat Rev Immunol ; 15(6): 350-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25998961

RESUMO

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.


Assuntos
Linfócitos B/imunologia , Células Dendríticas Foliculares/imunologia , Infecções/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Miofibroblastos/imunologia , Linfócitos T/imunologia , Animais , Comunicação Celular/fisiologia , Movimento Celular , Proliferação de Células/fisiologia , Humanos , Tolerância Imunológica/imunologia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Modelos Imunológicos , Miofibroblastos/citologia , Viroses/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA