Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 182(2): 329-344.e19, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32589946

RESUMEN

Cell surface receptors and their interactions play a central role in physiological and pathological signaling. Despite its clinical relevance, the immunoglobulin superfamily (IgSF) remains uncharacterized and underrepresented in databases. Here, we present a systematic extracellular protein map, the IgSF interactome. Using a high-throughput technology to interrogate most single transmembrane receptors for binding to 445 IgSF proteins, we identify over 500 interactions, 82% previously undocumented, and confirm more than 60 receptor-ligand pairs using orthogonal assays. Our study reveals a map of cell-type-specific interactions and the landscape of dysregulated receptor-ligand crosstalk in cancer, including selective loss of function for tumor-associated mutations. Furthermore, investigation of the IgSF interactome in a large cohort of cancer patients identifies interacting protein signatures associated with clinical outcome. The IgSF interactome represents an important resource to fuel biological discoveries and a framework for understanding the functional organization of the surfaceome during homeostasis and disease, ultimately informing therapeutic development.


Asunto(s)
Inmunoglobulinas/metabolismo , Neoplasias/patología , Mapas de Interacción de Proteínas , Antígeno B7-H1/metabolismo , Antígeno Carcinoembrionario/metabolismo , Comunicación Celular , Análisis por Conglomerados , Medios de Cultivo Condicionados/química , Células HEK293 , Humanos , Inmunoglobulinas/química , Inmunoglobulinas/genética , Ligandos , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Unión Proteica , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
2.
Nat Immunol ; 22(5): 571-585, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33903764

RESUMEN

Fibroblastic reticular cells (FRCs) are specialized stromal cells that define tissue architecture and regulate lymphocyte compartmentalization, homeostasis, and innate and adaptive immunity in secondary lymphoid organs (SLOs). In the present study, we used single-cell RNA sequencing (scRNA-seq) of human and mouse lymph nodes (LNs) to identify a subset of T cell-zone FRCs defined by the expression of Gremlin1 (Grem1) in both species. Grem1-CreERT2 knock-in mice enabled localization, multi-omics characterization and genetic depletion of Grem1+ FRCs. Grem1+ FRCs primarily localize at T-B cell junctions of SLOs, neighboring pre-dendritic cells and conventional dendritic cells (cDCs). As such, their depletion resulted in preferential loss and decreased homeostatic proliferation and survival of resident cDCs and compromised T cell immunity. Trajectory analysis of human LN scRNA-seq data revealed expression similarities to murine FRCs, with GREM1+ cells marking the endpoint of both trajectories. These findings illuminate a new Grem1+ fibroblastic niche in LNs that functions to maintain the homeostasis of lymphoid tissue-resident cDCs.


Asunto(s)
Células Dendríticas Foliculares/inmunología , Fibroblastos/inmunología , Ganglios Linfáticos/inmunología , Células del Estroma/inmunología , Anciano , Animales , Apoptosis/genética , Apoptosis/inmunología , Proliferación Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Dendríticas Foliculares/metabolismo , Femenino , Fibroblastos/metabolismo , Regulación de la Expresión Génica/inmunología , Técnicas de Sustitución del Gen , Humanos , Inmunidad Celular/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ganglios Linfáticos/citología , Masculino , Ratones , Ratones Transgénicos , RNA-Seq , Análisis de la Célula Individual , Células del Estroma/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
Nat Immunol ; 21(4): 369-380, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32205888

RESUMEN

Lymph nodes (LNs) are strategically positioned at dedicated sites throughout the body to facilitate rapid and efficient immunity. Central to the structural integrity and framework of LNs, and the recruitment and positioning of leukocytes therein, are mesenchymal and endothelial lymph node stromal cells (LNSCs). Advances in the last decade have expanded our understanding and appreciation of LNSC heterogeneity, and the role they play in coordinating immunity has grown rapidly. In this review, we will highlight the functional contributions of distinct stromal cell populations during LN development in maintaining immune homeostasis and tolerance and in the activation and control of immune responses.


Asunto(s)
Sistema Inmunológico/inmunología , Ganglios Linfáticos/inmunología , Células del Estroma/inmunología , Animales , Células Endoteliales/inmunología , Homeostasis/inmunología , Humanos , Inmunidad/inmunología
4.
Immunity ; 56(10): 2188-2205, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37820582

RESUMEN

The cancer-immunity cycle provides a framework to understand the series of events that generate anti-cancer immune responses. It emphasizes the iterative nature of the response where the killing of tumor cells by T cells initiates subsequent rounds of antigen presentation and T cell stimulation, maintaining active immunity and adapting it to tumor evolution. Any step of the cycle can become rate-limiting, rendering the immune system unable to control tumor growth. Here, we update the cancer-immunity cycle based on the remarkable progress of the past decade. Understanding the mechanism of checkpoint inhibition has evolved, as has our view of dendritic cells in sustaining anti-tumor immunity. We additionally account for the role of the tumor microenvironment in facilitating, not just suppressing, the anti-cancer response, and discuss the importance of considering a tumor's immunological phenotype, the "immunotype". While these new insights add some complexity to the cycle, they also provide new targets for research and therapeutic intervention.


Asunto(s)
Inmunoterapia , Neoplasias , Humanos , Neoplasias/genética , Neoplasias/terapia , Neoplasias/metabolismo , Linfocitos T , Presentación de Antígeno , Genotipo , Microambiente Tumoral/genética
5.
Nat Immunol ; 20(11): 1506-1516, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31611698

RESUMEN

Fibroblastic reticular cells (FRCs) and their specialized collagen fibers termed 'conduits' form fundamental structural units supporting lymphoid tissues. In lymph nodes, conduits are known to transport interstitial fluid and small molecules from afferent lymphatics into the nodal parenchyma. However, the immunological contributions of conduit function have remained elusive. Here, we report that intestinal Peyer's patches (PPs) contain a specialized conduit system that directs the flow of water absorbed across the intestinal epithelium. Notably, PP FRCs responded to conduit fluid flow via the mechanosensitive ion channel Piezo1. Disruption of fluid flow or genetic deficiency of Piezo1 on CCL19-expressing stroma led to profound structural alterations in perivascular FRCs and associated high endothelial venules. This in turn impaired lymphocyte entry into PPs and initiation of mucosal antibody responses. These results identify a critical role for conduit-mediated fluid flow in the maintenance of PP homeostasis and mucosal immunity.


Asunto(s)
Inmunidad Mucosa , Mucosa Intestinal/inmunología , Linfocitos/inmunología , Mecanotransducción Celular/inmunología , Ganglios Linfáticos Agregados/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Movimiento Celular/inmunología , Quimiocina CCL19/metabolismo , Femenino , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Activación de Linfocitos , Linfocitos/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Ganglios Linfáticos Agregados/metabolismo , Agua/metabolismo
6.
Nat Immunol ; 20(12): 1668-1680, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31636464

RESUMEN

Lymph node fibroblastic reticular cells (FRCs) respond to signals from activated T cells by releasing nitric oxide, which inhibits T cell proliferation and restricts the size of the expanding T cell pool. Whether interactions with FRCs also support the function or differentiation of activated CD8+ T cells is not known. Here we report that encounters with FRCs enhanced cytokine production and remodeled chromatin accessibility in newly activated CD8+ T cells via interleukin-6. These epigenetic changes facilitated metabolic reprogramming and amplified the activity of pro-survival pathways through differential transcription factor activity. Accordingly, FRC conditioning significantly enhanced the persistence of virus-specific CD8+ T cells in vivo and augmented their differentiation into tissue-resident memory T cells. Our study demonstrates that FRCs play a role beyond restricting T cell expansion-they can also shape the fate and function of CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Fibroblastos/fisiología , Ganglios Linfáticos/inmunología , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Reprogramación Celular , Ensamble y Desensamble de Cromatina , Citotoxicidad Inmunológica , Epigénesis Genética , Regulación de la Expresión Génica , Memoria Inmunológica , Interleucina-6/genética , Interleucina-6/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo
7.
Immunity ; 54(5): 903-915, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979587

RESUMEN

Fibroblasts and macrophages are present in all tissues, and mounting evidence supports that these cells engage in direct communication to influence the overall tissue microenvironment and affect disease outcomes. Here, we review the current understanding of the molecular mechanisms that underlie fibroblast-macrophage interactions in health, fibrosis, and cancer. We present an integrated view of fibroblast-macrophage interactions that is centered on the CSF1-CSF1R axis and discuss how additional molecular programs linking these cell types can underpin disease onset, progression, and resolution. These programs may be tissue and context dependent, affected also by macrophage and fibroblast origin and state, as seen most clearly in cancer. Continued efforts to understand these cells and the means by which they interact may provide therapeutic approaches for the treatment of fibrosis and cancer.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis/metabolismo , Macrófagos/metabolismo , Neoplasias/metabolismo , Animales , Diferenciación Celular/fisiología , Humanos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Microambiente Tumoral/fisiología
8.
Immunity ; 54(7): 1511-1526.e8, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260887

RESUMEN

Myeloid cells encounter stromal cells and their matrix determinants on a continual basis during their residence in any given organ. Here, we examined the impact of the collagen receptor LAIR1 on myeloid cell homeostasis and function. LAIR1 was highly expressed in the myeloid lineage and enriched in non-classical monocytes. Proteomic definition of the LAIR1 interactome identified stromal factor Colec12 as a high-affinity LAIR1 ligand. Proteomic profiling of LAIR1 signaling triggered by Collagen1 and Colec12 highlighted pathways associated with survival, proliferation, and differentiation. Lair1-/- mice had reduced frequencies of Ly6C- monocytes, which were associated with altered proliferation and apoptosis of non-classical monocytes from bone marrow and altered heterogeneity of interstitial macrophages in lung. Myeloid-specific LAIR1 deficiency promoted metastatic growth in a melanoma model and LAIR1 expression associated with improved clinical outcomes in human metastatic melanoma. Thus, monocytes and macrophages rely on LAIR1 sensing of stromal determinants for fitness and function, with relevance in homeostasis and disease.


Asunto(s)
Homeostasis/fisiología , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Monocitos/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Apoptosis/fisiología , Médula Ósea/metabolismo , Médula Ósea/patología , Células COS , Diferenciación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Chlorocebus aethiops , Femenino , Humanos , Pulmón/patología , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/patología , Células Mieloides/metabolismo , Células Mieloides/patología , Metástasis de la Neoplasia/patología , Proteómica/métodos , Transducción de Señal/fisiología
9.
Immunity ; 52(4): 578-580, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294403

RESUMEN

The omentum, an adipose tissue rich in fat-associated lymphoid clusters in the peritoneal cavity, is associated with immune surveillance and protection against peritoneal contaminants. In this issue of Immunity, Jackson-Jones et al. reveal how omental stromal cells regulate neutrophil trafficking to control peritonitis.


Asunto(s)
Epiplón , Peritonitis , Tejido Adiposo , Humanos , Inflamación , Neutrófilos , Células del Estroma
10.
Nature ; 618(7966): 827-833, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37258670

RESUMEN

The immune phenotype of a tumour is a key predictor of its response to immunotherapy1-4. Patients who respond to checkpoint blockade generally present with immune-inflamed5-7 tumours that are highly infiltrated by T cells. However, not all inflamed tumours respond to therapy, and even lower response rates occur among tumours that lack T cells (immune desert) or that spatially exclude T cells to the periphery of the tumour lesion (immune excluded)8. Despite the importance of these tumour immune phenotypes in patients, little is known about their development, heterogeneity or dynamics owing to the technical difficulty of tracking these features in situ. Here we introduce skin tumour array by microporation (STAMP)-a preclinical approach that combines high-throughput time-lapse imaging with next-generation sequencing of tumour arrays. Using STAMP, we followed the development of thousands of arrayed tumours in vivo to show that tumour immune phenotypes and outcomes vary between adjacent tumours and are controlled by local factors within the tumour microenvironment. Particularly, the recruitment of T cells by fibroblasts and monocytes into the tumour core was supportive of T cell cytotoxic activity and tumour rejection. Tumour immune phenotypes were dynamic over time and an early conversion to an immune-inflamed phenotype was predictive of spontaneous or therapy-induced tumour rejection. Thus, STAMP captures the dynamic relationships of the spatial, cellular and molecular components of tumour rejection and has the potential to translate therapeutic concepts into successful clinical strategies.


Asunto(s)
Neoplasias , Linfocitos T , Microambiente Tumoral , Humanos , Inmunoterapia , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Linfocitos T/inmunología , Fenotipo , Fibroblastos , Monocitos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico
11.
Immunity ; 51(1): 119-130.e5, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31231034

RESUMEN

Tissue-resident macrophages require specific milieus for the maintenance of defining gene-expression programs. Expression of the transcription factor GATA6 is required for the homeostasis, function and localization of peritoneal cavity-resident macrophages. Gata6 expression is maintained in a non-cell autonomous manner and is elicited by the vitamin A metabolite, retinoic acid. Here, we found that the GATA6 transcriptional program is a common feature of macrophages residing in all visceral body cavities. Retinoic acid-dependent and -independent hallmark genes of GATA6+ macrophages were induced by mesothelial and fibroblastic stromal cells that express the transcription factor Wilms' Tumor 1 (WT1), which drives the expression of two rate-limiting enzymes in retinol metabolism. Depletion of Wt1+ stromal cells reduced the frequency of GATA6+ macrophages in the peritoneal, pleural and pericardial cavities. Thus, Wt1+ mesothelial and fibroblastic stromal cells constitute essential niche components supporting the tissue-specifying transcriptional landscape and homeostasis of cavity-resident macrophages.


Asunto(s)
Factor de Transcripción GATA6/metabolismo , Macrófagos/fisiología , Pericardio/inmunología , Cavidad Peritoneal/fisiología , Cavidad Pleural/inmunología , Proteínas Represoras/metabolismo , Células del Estroma/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Factor de Transcripción GATA6/genética , Homeostasis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Represoras/genética , Tretinoina/metabolismo , Proteínas WT1
12.
Nature ; 611(7934): 148-154, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36171287

RESUMEN

Recent single-cell studies of cancer in both mice and humans have identified the emergence of a myofibroblast population specifically marked by the highly restricted leucine-rich-repeat-containing protein 15 (LRRC15)1-3. However, the molecular signals that underlie the development of LRRC15+ cancer-associated fibroblasts (CAFs) and their direct impact on anti-tumour immunity are uncharacterized. Here in mouse models of pancreatic cancer, we provide in vivo genetic evidence that TGFß receptor type 2 signalling in healthy dermatopontin+ universal fibroblasts is essential for the development of cancer-associated LRRC15+ myofibroblasts. This axis also predominantly drives fibroblast lineage diversity in human cancers. Using newly developed Lrrc15-diphtheria toxin receptor knock-in mice to selectively deplete LRRC15+ CAFs, we show that depletion of this population markedly reduces the total tumour fibroblast content. Moreover, the CAF composition is recalibrated towards universal fibroblasts. This relieves direct suppression of tumour-infiltrating CD8+ T cells to enhance their effector function and augments tumour regression in response to anti-PDL1 immune checkpoint blockade. Collectively, these findings demonstrate that TGFß-dependent LRRC15+ CAFs dictate the tumour-fibroblast setpoint to promote tumour growth. These cells also directly suppress CD8+ T cell function and limit responsiveness to checkpoint blockade. Development of treatments that restore the homeostatic fibroblast setpoint by reducing the population of pro-disease LRRC15+ myofibroblasts may improve patient survival and response to immunotherapy.


Asunto(s)
Fibroblastos Asociados al Cáncer , Proteínas de la Membrana , Miofibroblastos , Neoplasias Pancreáticas , Células del Estroma , Animales , Humanos , Ratones , Fibroblastos Asociados al Cáncer/metabolismo , Linfocitos T CD8-positivos/inmunología , Proteínas de la Membrana/metabolismo , Miofibroblastos/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Receptores de Factores de Crecimiento Transformadores beta , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , Antígeno B7-H1
13.
Nat Immunol ; 16(1): 75-84, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25347465

RESUMEN

In lymph nodes, fibroblastic reticular cells (FRCs) form a collagen-based reticular network that supports migratory dendritic cells (DCs) and T cells and transports lymph. A hallmark of FRCs is their propensity to contract collagen, yet this function is poorly understood. Here we demonstrate that podoplanin (PDPN) regulates actomyosin contractility in FRCs. Under resting conditions, when FRCs are unlikely to encounter mature DCs expressing the PDPN receptor CLEC-2, PDPN endowed FRCs with contractile function and exerted tension within the reticulum. Upon inflammation, CLEC-2 on mature DCs potently attenuated PDPN-mediated contractility, which resulted in FRC relaxation and reduced tissue stiffness. Disrupting PDPN function altered the homeostasis and spacing of FRCs and T cells, which resulted in an expanded reticular network and enhanced immunity.


Asunto(s)
Colágeno/metabolismo , Fibroblastos/citología , Lectinas Tipo C/metabolismo , Ganglios Linfáticos/citología , Glicoproteínas de Membrana/metabolismo , Amidas/farmacología , Animales , Supervivencia Celular/inmunología , Colágeno/inmunología , Citoesqueleto/inmunología , Citoesqueleto/ultraestructura , Inhibidores Enzimáticos/farmacología , Femenino , Fibroblastos/inmunología , Fibroblastos/ultraestructura , Lectinas Tipo C/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/ultraestructura , Masculino , Glicoproteínas de Membrana/inmunología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Fosforilación , Piridinas/farmacología , Organismos Libres de Patógenos Específicos
14.
Nature ; 593(7860): 575-579, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33981032

RESUMEN

Fibroblasts are non-haematopoietic structural cells that define the architecture of organs, support the homeostasis of tissue-resident cells and have key roles in fibrosis, cancer, autoimmunity and wound healing1. Recent studies have described fibroblast heterogeneity within individual tissues1. However, the field lacks a characterization of fibroblasts at single-cell resolution across tissues in healthy and diseased organs. Here we constructed fibroblast atlases by integrating single-cell transcriptomic data from about 230,000 fibroblasts across 17 tissues, 50 datasets, 11 disease states and 2 species. Mouse fibroblast atlases and a DptIRESCreERT2 knock-in mouse identified two universal fibroblast transcriptional subtypes across tissues. Our analysis suggests that these cells can serve as a reservoir that can yield specialized fibroblasts across a broad range of steady-state tissues and activated fibroblasts in disease. Comparison to an atlas of human fibroblasts from perturbed states showed that fibroblast transcriptional states are conserved between mice and humans, including universal fibroblasts and activated phenotypes associated with pathogenicity in human cancer, fibrosis, arthritis and inflammation. In summary, a cross-species and pan-tissue approach to transcriptomics at single-cell resolution has identified key organizing principles of the fibroblast lineage in health and disease.


Asunto(s)
Fibroblastos/citología , Transcriptoma , Animales , Células Cultivadas , Enfermedad , Femenino , Fibroblastos/clasificación , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias , Especificidad de Órganos , Fenotipo , RNA-Seq , Análisis de la Célula Individual , Células del Estroma
15.
Nat Immunol ; 15(10): 973-81, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25151489

RESUMEN

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.


Asunto(s)
Linfocitos B/inmunología , Fibroblastos/inmunología , Homeostasis/inmunología , Linfocitos T/inmunología , Animales , Factor Activador de Células B/inmunología , Factor Activador de Células B/metabolismo , Linfocitos B/metabolismo , Movimiento Celular/inmunología , Supervivencia Celular/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Inmunidad Humoral/inmunología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Linfocitos T/metabolismo
16.
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
17.
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
18.
Nat Immunol ; 13(9): 888-99, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22797772

RESUMEN

Although much progress has been made in the understanding of the ontogeny and function of dendritic cells (DCs), the transcriptional regulation of the lineage commitment and functional specialization of DCs in vivo remains poorly understood. We made a comprehensive comparative analysis of CD8(+), CD103(+), CD11b(+) and plasmacytoid DC subsets, as well as macrophage DC precursors and common DC precursors, across the entire immune system. Here we characterized candidate transcriptional activators involved in the commitment of myeloid progenitor cells to the DC lineage and predicted regulators of DC functional diversity in tissues. We identified a molecular signature that distinguished tissue DCs from macrophages. We also identified a transcriptional program expressed specifically during the steady-state migration of tissue DCs to the draining lymph nodes that may control tolerance to self tissue antigens.


Asunto(s)
Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Células Dendríticas/inmunología , Transcripción Genética , Diferenciación Celular/genética , Células Dendríticas/citología , Perfilación de la Expresión Génica , Humanos
19.
Nat Immunol ; 13(11): 1118-28, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23023392

RESUMEN

We assessed gene expression in tissue macrophages from various mouse organs. The diversity in gene expression among different populations of macrophages was considerable. Only a few hundred mRNA transcripts were selectively expressed by macrophages rather than dendritic cells, and many of these were not present in all macrophages. Nonetheless, well-characterized surface markers, including MerTK and FcγR1 (CD64), along with a cluster of previously unidentified transcripts, were distinctly and universally associated with mature tissue macrophages. TCEF3, C/EBP-α, Bach1 and CREG-1 were among the transcriptional regulators predicted to regulate these core macrophage-associated genes. The mRNA encoding other transcription factors, such as Gata6, was associated with single macrophage populations. We further identified how these transcripts and the proteins they encode facilitated distinguishing macrophages from dendritic cells.


Asunto(s)
Antígenos CD/genética , Macrófagos/metabolismo , ARN Mensajero/genética , Factores de Transcripción/genética , Transcripción Genética , Animales , Antígenos CD/inmunología , Diferenciación Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Variación Genética , Hígado/citología , Hígado/inmunología , Hígado/metabolismo , Pulmón/citología , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Ratones , Microglía/citología , Microglía/inmunología , Microglía/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , ARN Mensajero/inmunología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Factores de Transcripción/inmunología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA