Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunohorizons ; 7(5): 366-379, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37219538

RESUMEN

CD39 (ENTPD1) is a key enzyme responsible for degradation of extracellular ATP and is upregulated in the tumor microenvironment (TME). Extracellular ATP accumulates in the TME from tissue damage and immunogenic cell death, potentially initiating proinflammatory responses that are reduced by the enzymatic activity of CD39. Degradation of ATP by CD39 and other ectonucleotidases (e.g., CD73) results in extracellular adenosine accumulation, constituting an important mechanism for tumor immune escape, angiogenesis induction, and metastasis. Thus, inhibiting CD39 enzymatic activity can inhibit tumor growth by converting a suppressive TME to a proinflammatory environment. SRF617 is an investigational, anti-CD39, fully human IgG4 Ab that binds to human CD39 with nanomolar affinity and potently inhibits its ATPase activity. In vitro functional assays using primary human immune cells demonstrate that inhibiting CD39 enhances T-cell proliferation, dendritic cell maturation/activation, and release of IL-1ß and IL-18 from macrophages. In vivo, SRF617 has significant single-agent antitumor activity in human cell line-derived xenograft models that express CD39. Pharmacodynamic studies demonstrate that target engagement of CD39 by SRF617 in the TME inhibits ATPase activity, inducing proinflammatory mechanistic changes in tumor-infiltrating leukocytes. Syngeneic tumor studies using human CD39 knock-in mice show that SRF617 can modulate CD39 levels on immune cells in vivo and can penetrate the TME of an orthotopic tumor, leading to increased CD8+ T-cell infiltration. Targeting CD39 is an attractive approach for treating cancer, and, as such, the properties of SRF617 make it an excellent drug development candidate.


Asunto(s)
Inmunoglobulina G , Activación de Linfocitos , Humanos , Animales , Ratones , Anticuerpos Monoclonales , Adenosina Trifosfatasas , Adenosina Trifosfato
2.
Cell Rep ; 33(9): 108436, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264610

RESUMEN

The phosphatidylinositol 3-kinase (PI3K) signaling cascade downstream of the B cell receptor (BCR) signalosome is essential for B cell maturation. Proper signaling strength is maintained through the PI3K negative regulator phosphatase and tensin homolog (PTEN). Although a role for microRNA (miRNA)-dependent control of the PTEN-PI3K axis has been described, the contribution of individual miRNAs to the regulation of this crucial signaling modality in mature B lymphocytes remains to be elucidated. Our analyses reveal that ablation of miR-29 specifically in B lymphocytes results in an increase in PTEN expression and dampening of the PI3K pathway in mature B cells. This dysregulation has a profound impact on the survival of B lymphocytes and results in increased class switch recombination and decreased plasma cell differentiation. Furthermore, we demonstrate that ablation of one copy of Pten is sufficient to ameliorate the phenotypes associated with miR-29 loss. Our data suggest a critical role for the miR-29-PTEN-PI3K regulatory axis in mature B lymphocytes.


Asunto(s)
Linfocitos B/metabolismo , MicroARNs/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Diferenciación Celular , Humanos , Ratones , Transducción de Señal , Análisis de Supervivencia
3.
Cell Rep ; 28(8): 2169-2181.e4, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31433990

RESUMEN

Coordinate control of T cell proliferation, survival, and differentiation are essential for host protection from pathogens and cancer. Long-lived memory cells, whose precursors are formed during the initial immunological insult, provide protection from future encounters, and their generation is the goal of many vaccination strategies. microRNAs (miRNAs) are key nodes in regulatory networks that shape effective T cell responses through the fine-tuning of thousands of genes. Here, using compound conditional mutant mice to eliminate miR-15/16 family miRNAs in T cells, we show that miR-15/16 restrict T cell cycle, survival, and memory T cell differentiation. High throughput sequencing of RNA isolated by cross-linking immunoprecipitation of AGO2 combined with gene expression analysis in miR-15/16-deficient T cells indicates that these effects are mediated through the direct inhibition of an extensive network of target genes within pathways critical to cell cycle, survival, and memory.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Memoria Inmunológica , MicroARNs/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Animales , Antígenos/metabolismo , Ciclo Celular/genética , Diferenciación Celular/genética , Supervivencia Celular/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Sitios Genéticos , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Transgénicos , MicroARNs/genética
4.
Proc Natl Acad Sci U S A ; 115(11): E2585-E2593, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29476012

RESUMEN

Skeletal muscle regeneration is a highly orchestrated process that depends on multiple immune-system cell types, notably macrophages (MFs) and Foxp3+CD4+ regulatory T (Treg) cells. This study addressed how Treg cells rein in MFs during regeneration of murine muscle after acute injury with cardiotoxin. We first delineated and characterized two subsets of MFs according to their expression of major histocompatibility complex class II (MHCII) molecules, i.e., their ability to present antigens. Then, we assessed the impact of Treg cells on these MF subsets by punctually depleting Foxp3+ cells during the regenerative process. Treg cells controlled both the accumulation and phenotype of the two types of MFs. Their absence after injury promoted IFN-γ production, primarily by NK and effector T cells, which ultimately resulted in MF dysregulation and increased inflammation and fibrosis, pointing to compromised muscle repair. Thus, we uncovered an IFN-γ-centered regulatory layer by which Treg cells keep MFs in check and dampen inflammation during regeneration of skeletal muscle.


Asunto(s)
Interferón gamma/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Regeneración/fisiología , Linfocitos T Reguladores/metabolismo , Animales , Cardiotoxinas/toxicidad , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Fenotipo , Linfocitos T Reguladores/fisiología
5.
Annu Rev Immunol ; 34: 609-33, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27168246

RESUMEN

The immune system is responsible for defending an organism against the myriad of microbial invaders it constantly confronts. It has become increasingly clear that the immune system has a second major function: the maintenance of organismal homeostasis. Foxp3(+)CD4(+) regulatory T cells (Tregs) are important contributors to both of these critical activities, defense being the primary purview of Tregs circulating through lymphoid organs, and homeostasis ensured mainly by their counterparts residing in parenchymal tissues. This review focuses on so-called tissue Tregs. We first survey existing information on the phenotype, function, sustaining factors, and human equivalents of the three best-characterized tissue-Treg populations-those operating in visceral adipose tissue, skeletal muscle, and the colonic lamina propria. We then attempt to distill general principles from this body of work-as concerns the provenance, local adaptation, molecular sustenance, and targets of action of tissue Tregs, in particular.


Asunto(s)
Tejido Adiposo/inmunología , Colon/inmunología , Membrana Mucosa/inmunología , Músculo Esquelético/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/metabolismo , Homeostasis , Humanos , Especificidad de Órganos
6.
Immunity ; 44(2): 355-67, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26872699

RESUMEN

Normal repair of skeletal muscle requires local expansion of a special population of Foxp3(+)CD4(+) regulatory T (Treg) cells. Such cells failed to accumulate in acutely injured muscle of old mice, known to undergo ineffectual repair. This defect reflected reduced recruitment of Treg cells to injured muscle, as well as less proliferation and retention therein. Interleukin-33 (IL-33) regulated muscle Treg cell homeostasis in young mice, and its administration to old mice ameliorated their deficits in Treg cell accumulation and muscle regeneration. The major IL-33-expressing cells in skeletal muscle displayed a constellation of markers diagnostic of fibro/adipogenic progenitor cells and were often associated with neural structures, including nerve fibers, nerve bundles, and muscle spindles, which are stretch-sensitive mechanoreceptors important for proprioception. IL-33(+) cells were more frequent after muscle injury and were reduced in old mice. IL-33 is well situated to relay signals between the nervous and immune systems within the muscle context.


Asunto(s)
Envejecimiento/inmunología , Interleucina-33/metabolismo , Músculo Esquelético/fisiología , Células Madre/inmunología , Linfocitos T Reguladores/inmunología , Adipogénesis/inmunología , Animales , Biomarcadores/metabolismo , Proliferación Celular , Células Cultivadas , Homeostasis , Humanos , Mecanotransducción Celular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/inervación , Fibras Nerviosas/fisiología , Neuroinmunomodulación , Regeneración/inmunología , Cicatrización de Heridas
7.
Nat Immunol ; 15(12): 1162-70, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25362490

RESUMEN

MicroRNAs (miRNAs) exert powerful effects on immunological function by tuning networks of target genes that orchestrate cell activity. We sought to identify miRNAs and miRNA-regulated pathways that control the type 2 helper T cell (TH2 cell) responses that drive pathogenic inflammation in asthma. Profiling miRNA expression in human airway-infiltrating T cells revealed elevated expression of the miRNA miR-19a in asthma. Modulating miR-19 activity altered TH2 cytokine production in both human and mouse T cells, and TH2 cell responses were markedly impaired in cells lacking the entire miR-17∼92 cluster. miR-19 promoted TH2 cytokine production and amplified inflammatory signaling by direct targeting of the inositol phosphatase PTEN, the signaling inhibitor SOCS1 and the deubiquitinase A20. Thus, upregulation of miR-19a in asthma may be an indicator and a cause of increased TH2 cytokine production in the airways.


Asunto(s)
Asma/inmunología , Citocinas/biosíntesis , MicroARNs/inmunología , Células Th2/inmunología , Animales , Asma/genética , Asma/metabolismo , Líquido del Lavado Bronquioalveolar/citología , Ensayos Clínicos como Asunto , Citometría de Flujo , Ensayos Analíticos de Alto Rendimiento , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa Multiplex , Células Th2/metabolismo , Regulación hacia Arriba
8.
Blood ; 120(1): 130-42, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22613798

RESUMEN

Natural killer (NK) cells play a critical role in early host defense to infected and transformed cells. Here, we show that mice deficient in Eri1, a conserved 3'-to-5' exoribonuclease that represses RNA interference, have a cell-intrinsic defect in NK-cell development and maturation. Eri1(-/-) NK cells displayed delayed acquisition of Ly49 receptors in the bone marrow (BM) and a selective reduction in Ly49D and Ly49H activating receptors in the periphery. Eri1 was required for immune-mediated control of mouse CMV (MCMV) infection. Ly49H(+) NK cells deficient in Eri1 failed to expand efficiently during MCMV infection, and virus-specific responses were also diminished among Eri1(-/-) T cells. We identified miRNAs as the major endogenous small RNA target of Eri1 in mouse lymphocytes. Both NK and T cells deficient in Eri1 displayed a global, sequence-independent increase in miRNA abundance. Ectopic Eri1 expression rescued defective miRNA expression in mature Eri1(-/-) T cells. Thus, mouse Eri1 regulates miRNA homeostasis in lymphocytes and is required for normal NK-cell development and antiviral immunity.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Exonucleasas/genética , Exonucleasas/inmunología , Células Asesinas Naturales/inmunología , MicroARNs/inmunología , Traslado Adoptivo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Exorribonucleasas , Homeostasis/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/virología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Mutantes , MicroARNs/genética , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo
9.
Immunity ; 36(2): 175-87, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22326582

RESUMEN

Follicular helper T cells (Tfh cells) are the major producers of interleukin-4 (IL-4) in secondary lymphoid organs where humoral immune responses develop. Il4 regulation in Tfh cells appears distinct from the classical T helper 2 (Th2) cell pathway, but the underlying molecular mechanisms remain largely unknown. We found that hypersensitivity site V (HS V; also known as CNS2), a 3' enhancer in the Il4 locus, is essential for IL-4 production by Tfh cells. Mice lacking HS V display marked defects in type 2 humoral immune responses, as evidenced by abrogated IgE and sharply reduced IgG1 production in vivo. In contrast, effector Th2 cells that are involved in tissue responses were far less dependent on HS V. HS V facilitated removal of repressive chromatin marks during Th2 and Tfh cell differentiation and increased accessibility of the Il4 promoter. Thus, Tfh and Th2 cells utilize distinct but overlapping molecular mechanisms to regulate Il4, a finding with important implications for understanding the molecular basis of allergic diseases.


Asunto(s)
Interleucina-4/biosíntesis , Interleucina-4/genética , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Sitios de Unión/genética , Secuencia Conservada , Citocinas/genética , Elementos de Facilitación Genéticos , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Inmunidad Humoral/genética , Interleucina-4/deficiencia , Pulmón/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Regiones Promotoras Genéticas , Eliminación de Secuencia , Subgrupos de Linfocitos T/inmunología , Células Th2/inmunología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...