Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Immunity ; 57(6): 1345-1359.e5, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38692280

RESUMEN

Regulatory T (Treg) cells in epidydimal visceral adipose tissue (eVAT) of lean mice and humans regulate metabolic homeostasis. We found that constitutive or punctual depletion of eVAT-Treg cells reined in the differentiation of stromal adipocyte precursors. Co-culture of these precursors with conditional medium from eVAT-Treg cells limited their differentiation in vitro, suggesting a direct effect. Transcriptional comparison of adipocyte precursors, matured in the presence or absence of the eVAT-Treg-conditioned medium, identified the oncostatin-M (OSM) signaling pathway as a key distinction. Addition of OSM to in vitro cultures blocked the differentiation of adipocyte precursors, while co-addition of anti-OSM antibodies reversed the ability of the eVAT-Treg-conditioned medium to inhibit in vitro adipogenesis. Genetic depletion of OSM (specifically in Treg) cells or of the OSM receptor (specifically on stromal cells) strongly impaired insulin sensitivity and related metabolic indices. Thus, Treg-cell-mediated control of local progenitor cells maintains adipose tissue and metabolic homeostasis, a regulatory axis seemingly conserved in humans.


Asunto(s)
Adipocitos , Diferenciación Celular , Homeostasis , Resistencia a la Insulina , Linfocitos T Reguladores , Animales , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Humanos , Ratones , Adipocitos/metabolismo , Diferenciación Celular/inmunología , Oncostatina M/metabolismo , Transducción de Señal , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/citología , Grasa Intraabdominal/inmunología , Células del Estroma/metabolismo , Ratones Endogámicos C57BL , Técnicas de Cocultivo , Adipogénesis , Células Cultivadas , Masculino , Tejido Adiposo/metabolismo , Tejido Adiposo/citología , Medios de Cultivo Condicionados/farmacología
2.
Immunity ; 56(4): 829-846.e8, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36822206

RESUMEN

Specific microbial signals induce the differentiation of a distinct pool of RORγ+ regulatory T (Treg) cells crucial for intestinal homeostasis. We discovered highly analogous populations of microbiota-dependent Treg cells that promoted tissue regeneration at extra-gut sites, notably acutely injured skeletal muscle and fatty liver. Inflammatory meditators elicited by tissue damage combined with MHC-class-II-dependent T cell activation to drive the accumulation of gut-derived RORγ+ Treg cells in injured muscle, wherein they regulated the dynamics and tenor of early inflammation and helped balance the proliferation vs. differentiation of local stem cells. Reining in IL-17A-producing T cells was a major mechanism underlying the rheostatic functions of RORγ+ Treg cells in compromised tissues. Our findings highlight the importance of gut-trained Treg cell emissaries in controlling the response to sterile injury of non-mucosal tissues.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Ratones , Linfocitos T Reguladores , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Ratones Endogámicos C57BL
3.
Proc Natl Acad Sci U S A ; 121(4): e2320602121, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38227656

RESUMEN

Foxp3+CD4+ regulatory T (Treg) cells found within tissues regulate local immunity, inflammation, and homeostasis. Tregs in epididymal visceral adipose tissue (eVAT) are critical regulators of local and systemic inflammation and metabolism. During aging and under obesogenic conditions, eVAT Tregs undergo transcriptional and phenotypic changes and are important for containing inflammation and normalizing metabolic indices. We have employed single-cell RNA sequencing, single-cell Tra and Trb sequencing, adoptive transfers, photoconvertible mice, cellular interaction analyses, and in vitro cultures to dissect the evolving heterogeneity of eVAT Tregs with aging and obesity. Distinct Treg subtypes with distinguishable gene expression profiles and functional roles were enriched at differing ages and with differing diets. Like those in lean mice, eVAT Tregs in obese mice were not primarily recruited from the circulation but instead underwent local expansion and had a distinct and diversified T cell receptor repertoire. The different eVAT-Treg subtypes were specialized in different functions; for example, the subtypes enriched in lean, but not obese, mice suppressed adipogenesis. The existence of functionally divergent eVAT-Treg subtypes in response to obesogenic conditions presents possibilities for precision therapeutics in the context of obesity.


Asunto(s)
Tejido Adiposo , Linfocitos T Reguladores , Ratones , Animales , Tejido Adiposo/metabolismo , Dieta , Obesidad/metabolismo , Ratones Obesos , Inflamación/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33753509

RESUMEN

Foxp3+CD4+ regulatory T cells (Tregs) regulate most types of immune response as well as several processes important for tissue homeostasis, for example, metabolism and repair. Dedicated Treg compartments-with distinct transcriptomes, T cell receptor repertoires, and growth/survival factor dependencies-have been identified in several nonlymphoid tissues. These Tregs are specifically adapted to function and operate in their home tissue-When, where, and how do they take on their specialized characteristics? We recently reported that a splenic Treg population expressing low levels of the transcription factor PPARγ (peroxisome proliferator-activated receptor gamma) contains precursors of Tregs residing in visceral adipose tissue. This finding made sense given that PPARγ, the "master regulator" of adipocyte differentiation, is required for the accumulation and function of Tregs in visceral adipose tissue but not in lymphoid tissues. Here we use single-cell RNA sequencing, single-cell Tcra and Tcrb sequencing, and adoptive-transfer experiments to show that, unexpectedly, the splenic PPARγlo Treg population is transcriptionally heterogeneous and engenders Tregs in multiple nonlymphoid tissues beyond visceral adipose tissue, such as skin and liver. The existence of a general pool of splenic precursors for nonlymphoid-tissue Tregs opens possibilities for regulating their emergence experimentally or therapeutically.


Asunto(s)
Grasa Intraabdominal/inmunología , PPAR alfa/metabolismo , Bazo/inmunología , Linfocitos T Reguladores/inmunología , Transcriptoma , Traslado Adoptivo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR alfa/genética , RNA-Seq , Análisis de la Célula Individual
5.
J Cell Sci ; 127(Pt 16): 3555-67, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24951116

RESUMEN

Information from multiple signaling axes is integrated in the determination of cellular phenotypes. Here, we demonstrate this aspect of cellular decision making in glioblastoma multiforme (GBM) cells by investigating the multivariate signaling regulatory functions of the protein tyrosine phosphatase SHP2 (also known as PTPN11). Specifically, we demonstrate that the ability of SHP2 to simultaneously drive ERK1/2 and antagonize STAT3 pathway activities produces qualitatively different effects on the phenotypes of proliferation and resistance to EGFR and c-MET co-inhibition. Whereas the ERK1/2 and STAT3 pathways independently promote proliferation and resistance to EGFR and c-MET co-inhibition, SHP2-driven ERK1/2 activity is dominant in driving cellular proliferation and SHP2-mediated antagonism of STAT3 phosphorylation prevails in the promotion of GBM cell death in response to EGFR and c-MET co-inhibition. Interestingly, the extent of these SHP2 signaling regulatory functions is diminished in glioblastoma cells that express sufficiently high levels of the EGFR variant III (EGFRvIII) mutant, which is commonly expressed in GBM. In cells and tumors that express EGFRvIII, SHP2 also antagonizes the phosphorylation of EGFRvIII and c-MET and drives expression of HIF-1α and HIF-2α, adding complexity to the evolving understanding of the regulatory functions of SHP2 in GBM.


Asunto(s)
Proliferación Celular , Glioblastoma/enzimología , Sistema de Señalización de MAP Quinasas , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Femenino , Gefitinib , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/fisiopatología , Humanos , Indoles/administración & dosificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Desnudos , Fosforilación/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Quinazolinas/administración & dosificación , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Sulfonas/administración & dosificación
6.
J Exp Med ; 219(5)2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35380608

RESUMEN

Subsequent to acute injury, skeletal muscle undergoes a stereotypic regenerative process that reestablishes homeostasis. Various types of innate and adaptive immunocytes exert positive or negative influences at specific stages along the course of muscle regeneration. We describe an unanticipated role for γδT cells in promoting healthy tissue recovery after injection of cardiotoxin into murine hindlimb muscle. Within a few days of injury, IL-17A-producing γδT cells displaying primarily Vγ6+ antigen receptors accumulated at the wound site. Punctual ablation experiments showed that these cells boosted early inflammatory events, notably recruitment of neutrophils; fostered the proliferation of muscle stem and progenitor cells; and thereby promoted tissue regeneration. Supplementation of mice harboring low numbers of IL-17A+ γδT cells with recombinant IL-17A largely reversed their inflammatory and reparative defects. Unexpectedly, the accumulation and influences of γδT cells in this experimental context were microbiota dependent, unveiling an orthogonal perspective on the treatment of skeletal muscle pathologies such as catastrophic wounds, wasting, muscular dystrophies, and myositides.


Asunto(s)
Interleucina-17 , Microbiota , Desarrollo de Músculos , Regeneración , Linfocitos T , Animales , Ratones , Ratones Endogámicos C57BL , Músculos , Receptores de Antígenos de Linfocitos T gamma-delta
7.
Sci Immunol ; 7(75): eabl7641, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36179011

RESUMEN

Regulatory T cells (Tregs) in nonlymphoid organs provide critical brakes on inflammation and regulate tissue homeostasis. Although so-called "tissue Tregs" are phenotypically and functionally diverse, serving to optimize their performance and survival, up-regulation of pathways related to circadian rhythms is a feature they share. Yet the diurnal regulation of Tregs and its consequences are controversial and poorly understood. Here, we profiled diurnal variations in visceral adipose tissue (VAT) and splenic Tregs in the presence and absence of core-clock genes. VAT, but not splenic, Tregs up-regulated their cell-intrinsic circadian program and exhibited diurnal variations in their activation and metabolic state. BMAL1 deficiency specifically in Tregs led to constitutive activation and poor oxidative metabolism in VAT, but not splenic, Tregs. Disruption of core-clock components resulted in loss of fitness: BMAL1-deficient VAT Tregs were preferentially lost during competitive transfers and in heterozygous TregBmal1Δ females. After 16 weeks of high-fat diet feeding, VAT inflammation was increased in mice harboring BMAL1-deficient Tregs, and the remaining cells lost the transcriptomic signature of bona fide VAT Tregs. Unexpectedly, VAT Tregs suppressed adipocyte lipolysis, and BMAL1 deficiency specifically in Tregs abrogated the characteristic diurnal variation in adipose tissue lipolysis, resulting in enhanced suppression of lipolysis throughout the day. These findings argue for the importance of the cell-intrinsic clock program in optimizing VAT Treg function and fitness.


Asunto(s)
Ritmo Circadiano , Grasa Intraabdominal , Factores de Transcripción ARNTL/genética , Animales , Femenino , Inflamación , Lipólisis , Ratones , Linfocitos T Reguladores
8.
Nat Rev Immunol ; 21(9): 597-611, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33772242

RESUMEN

The FOXP3+CD4+ regulatory T (Treg) cells located in non-lymphoid tissues differ in phenotype and function from their lymphoid organ counterparts. Tissue Treg cells have distinct transcriptomes, T cell receptor repertoires and growth and survival factor dependencies that arm them to survive and operate in their home tissue. Their functions extend beyond immune surveillance to tissue homeostasis, including regulation of local and systemic metabolism, promotion of tissue repair and regeneration, and control of the proliferation, differentiation and fate of non-lymphoid cell progenitors. Treg cells in diverse tissues share a common FOXP3+CD4+ precursor located within lymphoid organs. This precursor undergoes definitive specialization once in the home tissue, following a multilayered array of common and tissue-distinct transcriptional programmes. Our deepening knowledge of tissue Treg cell biology will inform ongoing attempts to harness Treg cells for precision immunotherapeutics.


Asunto(s)
Linfocitos T Reguladores/inmunología , Animales , Femenino , Homeostasis/inmunología , Humanos , Grasa Intraabdominal/citología , Grasa Intraabdominal/inmunología , Masculino , Ratones , Modelos Inmunológicos , Músculo Esquelético/citología , Músculo Esquelético/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Piel/citología , Piel/inmunología , Linfocitos T Reguladores/fisiología , Transcriptoma
9.
Nat Commun ; 12(1): 301, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436596

RESUMEN

Macrophages are innate immune cells that contribute to fighting infections, tissue repair, and maintaining tissue homeostasis. To enable such functional diversity, macrophages resolve potentially conflicting cues in the microenvironment via mechanisms that are unclear. Here, we use single-cell RNA sequencing to explore how individual macrophages respond when co-stimulated with inflammatory stimuli LPS and IFN-γ and the resolving cytokine IL-4. These co-stimulated macrophages display a distinct global transcriptional program. However, variable negative cross-regulation between some LPS + IFN-γ-specific and IL-4-specific genes results in cell-to-cell heterogeneity in transcription. Interestingly, negative cross-regulation leads to mutually exclusive expression of the T-cell-polarizing cytokine genes Il6 and Il12b versus the IL-4-associated factors Arg1 and Chil3 in single co-stimulated macrophages, and single-cell secretion measurements show that these specialized functions are maintained for at least 48 h. This study suggests that increasing functional diversity in the population is one strategy macrophages use to respond to conflicting environmental cues.


Asunto(s)
Polaridad Celular , Macrófagos/citología , Animales , Arginasa/metabolismo , Polaridad Celular/efectos de los fármacos , Polaridad Celular/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Aprendizaje Automático , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Redes Neurales de la Computación , Oportunidad Relativa , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos
10.
Integr Biol (Camb) ; 11(4): 142-153, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31242304

RESUMEN

Innate immune cells, including macrophages and dendritic cells, protect the host from pathogenic assaults in part through secretion of a program of cytokines and chemokines (C/Cs). Cell-to-cell variability in C/C secretion appears to contribute to the regulation of the immune response, but the sources of secretion variability are largely unknown. To begin to track the biological sources that control secretion variability, we developed and validated a microfluidic device to integrate live-cell imaging of fluorescent reporter proteins with a single-cell assay of protein secretion. We used this device to image NF-κB RelA nuclear translocation dynamics and Tnf transcription dynamics in macrophages in response to stimulation with the bacterial component lipopolysaccharide (LPS), followed by quantification of secretion of TNF, CCL2, CCL3, and CCL5. We found that the timing of the initial peak of RelA signaling in part determined the relative level of TNF and CCL3 secretion, but not CCL2 and CCL5 secretion. Our results support evidence that differences in timing across cell processes partly account for cell-to-cell variability in downstream responses, but that other factors introduce variability at each biological step.


Asunto(s)
Dispositivos Laboratorio en un Chip , Macrófagos/metabolismo , Transcripción Genética , Animales , Anticuerpos , Comunicación Celular , Quimiocina CCL2/metabolismo , Quimiocina CCL3/metabolismo , Quimiocina CCL5/metabolismo , Diseño de Equipo , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microfluídica , Células RAW 264.7 , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Curr Opin Biotechnol ; 52: 109-115, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29656236

RESUMEN

Systems biology provides an effective approach to decipher, predict, and ultimately manipulate the complex and inter-connected networks that regulate the immune system. Advances in high-throughput, multiplexed experimental techniques have increased the availability of proteomic and transcriptomic immunological datasets, and as a result, have also accelerated the development of new data-driven computational algorithms to extract biological insight from these data. This review highlights how data-driven statistical models have been used to characterize immune cell subsets and their functions, to map the signaling and intercellular networks that regulate immune responses, and to connect immune cell states to disease outcomes to generate hypotheses for novel therapeutic strategies. We focus on recent advances in evaluating immune cell responses following viral infection and in the tumor microenvironment, which hold promise for improving vaccines, antiviral and cancer immunotherapy.


Asunto(s)
Análisis de Datos , Sistema Inmunológico/metabolismo , Estadística como Asunto , Biología de Sistemas , Animales , Humanos , Sistema Inmunológico/citología , Neoplasias/patología , Transducción de Señal
12.
Science ; 362(6417)2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30467144

RESUMEN

During tissue repair, myofibroblasts produce extracellular matrix (ECM) molecules for tissue resilience and strength. Altered ECM deposition can lead to tissue dysfunction and disease. Identification of distinct myofibroblast subsets is necessary to develop treatments for these disorders. We analyzed profibrotic cells during mouse skin wound healing, fibrosis, and aging and identified distinct subpopulations of myofibroblasts, including adipocyte precursors (APs). Multiple mouse models and transplantation assays demonstrate that proliferation of APs but not other myofibroblasts is activated by CD301b-expressing macrophages through insulin-like growth factor 1 and platelet-derived growth factor C. With age, wound bed APs and differential gene expression between myofibroblast subsets are reduced. Our findings identify multiple fibrotic cell populations and suggest that the environment dictates functional myofibroblast heterogeneity, which is driven by fibroblast-immune interactions after wounding.


Asunto(s)
Macrófagos/fisiología , Miofibroblastos/fisiología , Repitelización/fisiología , Piel/lesiones , Cicatrización de Heridas/fisiología , Adipocitos/fisiología , Animales , Proliferación Celular , Matriz Extracelular/metabolismo , Fibrosis , Integrina beta1/genética , Queloide/patología , Lectinas Tipo C/análisis , Lectinas Tipo C/metabolismo , Linfocinas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Repitelización/genética , Piel/inmunología , Piel/patología , Envejecimiento de la Piel/fisiología , Transcriptoma , Cicatrización de Heridas/genética
13.
J Exp Med ; 215(3): 877-893, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29436395

RESUMEN

Eliciting effective antitumor immune responses in patients who fail checkpoint inhibitor therapy is a critical challenge in cancer immunotherapy, and in such patients, tumor-associated myeloid cells and macrophages (TAMs) are promising therapeutic targets. We demonstrate in an autochthonous, poorly immunogenic mouse model of melanoma that combination therapy with an agonistic anti-CD40 mAb and CSF-1R inhibitor potently suppressed tumor growth. Microwell assays to measure multiplex protein secretion by single cells identified that untreated tumors have distinct TAM subpopulations secreting MMP9 or cosecreting CCL17/22, characteristic of an M2-like state. Combination therapy reduced the frequency of these subsets, while simultaneously inducing a separate polyfunctional inflammatory TAM subset cosecreting TNF-α, IL-6, and IL-12. Tumor suppression by this combined therapy was partially dependent on T cells, and on TNF-α and IFN-γ. Together, this study demonstrates the potential for targeting TAMs to convert a "cold" into an "inflamed" tumor microenvironment capable of eliciting protective T cell responses.


Asunto(s)
Inmunoterapia , Inflamación/patología , Células Mieloides/patología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Antígenos CD40/agonistas , Antígenos CD40/metabolismo , Proliferación Celular , Interferón gamma/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Melanoma Experimental/patología , Ratones , Neoplasias/patología , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas B-raf/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Análisis de Supervivencia , Linfocitos T/inmunología , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo
14.
Evolution ; 70(2): 270-81, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26882316

RESUMEN

Virus-host coevolution has selected for generalized host defense against viruses, exemplified by interferon production/signaling and other innate immune function in eukaryotes such as humans. Although cell-surface binding primarily limits virus infection success, generalized adaptation to counteract innate immunity across disparate hosts may contribute to RNA virus emergence potential. We examined this idea using vesicular stomatitis virus (VSV) populations previously evolved on strictly immune-deficient (HeLa) cells, strictly immune competent (MDCK) cells, or on alternating deficient/competent cells. By measuring viral fitness in unselected human cancer cells of differing innate immunity, we confirmed that HeLa-adapted populations were specialized for innate immune-deficient hosts, whereas MDCK-adapted populations were relatively more generalized for fitness on hosts of differing innate immune capacity and of different species origin. We also confirmed that HeLa-evolved populations maintained fitness in immune-deficient nonhuman primate cells. These results suggest that innate immunity is more prominent than host species in determining viral fitness at the host-cell level. Finally, our prediction was inexact that selection on alternating deficient/competent hosts should produce innate viral generalists. Rather, fitness differences among alternating host-evolved VSV populations indicated variable capacities to evade innate immunity. Our results suggest that the evolutionary history of innate immune selection can affect whether RNA viruses evolve greater host-breadth.


Asunto(s)
Especificidad del Huésped , Inmunidad Innata , Células de Riñón Canino Madin Darby , Selección Genética , Vesiculovirus/genética , Animales , Perros , Evolución Molecular , Células HeLa , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA