Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Trends Immunol ; 44(11): 890-901, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37827864

RESUMEN

The therapeutic potential of interleukin (IL)-2 in cancer treatment has been known for decades, yet its widespread adoption in clinical practice remains limited. Recently, chimeric proteins of an anti-PD-1 antibody and suboptimal IL-2 variants were shown to stimulate potent antitumor and antiviral immunity by inducing unique effector CD8+ T cells in mice. A similar subset of cytotoxic T cells is induced by depletion of regulatory T cells (Tregs), suggesting IL-2 sequestration as a major mechanism through which regulatory T cells suppress activated CD8+ T cells. Here, we present our view of how IL-2-based biologicals can boost the antitumor response at a cellular level, and propose that the role of Tregs following such treatments may have been previously overestimated.


Asunto(s)
Linfocitos T CD8-positivos , Interleucina-2 , Ratones , Animales , Interleucina-2/uso terapéutico , Interleucina-2/metabolismo , Inmunoterapia , Linfocitos T Citotóxicos , Linfocitos T Reguladores , Fenotipo
2.
Elife ; 122023 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-36705564

RESUMEN

Regulatory T cells (Tregs) are indispensable for maintaining self-tolerance by suppressing conventional T cells. On the other hand, Tregs promote tumor growth by inhibiting anticancer immunity. In this study, we identified that Tregs increase the quorum of self-reactive CD8+ T cells required for the induction of experimental autoimmune diabetes in mice. Their major suppression mechanism is limiting available IL-2, an essential T-cell cytokine. Specifically, Tregs inhibit the formation of a previously uncharacterized subset of antigen-stimulated KLRK1+ IL-7R+ (KILR) CD8+ effector T cells, which are distinct from conventional effector CD8+ T cells. KILR CD8+ T cells show superior cell-killing abilities in vivo. The administration of agonistic IL-2 immunocomplexes phenocopies the absence of Tregs, i.e., it induces KILR CD8+ T cells, promotes autoimmunity, and enhances antitumor responses in mice. Counterparts of KILR CD8+ T cells were found in the human blood, revealing them as a potential target for immunotherapy.


As well as protecting us from invading pathogens, like bacteria or viruses, our immune system can also identify dangerous cells of our own that may cause the body harm, such as cancer cells. Once detected, a population of immune cells called cytotoxic T cells launch into action to kill the potentially harmful cell. However, sometimes the immune system makes mistakes and attacks healthy cells which it misidentifies as being dangerous, leading to autoimmune diseases. Special immune cells called T regulatory lymphocytes, or 'Tregs', can suppress the activity of cytotoxic T cells, preventing them from hurting the body's own cells. While this can have a positive impact and reduce the effects of autoimmunity, Tregs can also make the immune system less responsive to cancer cells and allow tumors to grow. But how Tregs alter the behavior of cytotoxic T cells during autoimmune diseases and cancer is poorly understood. While multiple mechanisms have been proposed, none of these have been tested in living animal models of these diseases. To address this, Tsyklauri et al. studied Tregs in laboratory mice which had been modified to have autoimmune diabetes, which is when the body attacks the cells responsible for producing insulin. The experiments revealed that Tregs take up a critical signaling molecule called IL-2 which cytotoxic T cells need to survive and multiply. As a result, there is less IL-2 molecules available in the environment, inhibiting the cytotoxic T cells' activity. Furthermore, if Tregs are absent and there is an excess of IL-2, this causes cytotoxic T cells to transition into a previously unknown subset of T cells with superior killing abilities. Tsyklauri et al. were able to replicate these findings in two different groups of laboratory mice which had been modified to have cancer. This suggests that Tregs suppress the immune response to cancer cells and prevent autoimmunity using the same mechanism. In the future, this work could help researchers to develop therapies that alter the behavior of cytotoxic T cells and/or Tregs to either counteract autoimmune diseases, or help the body fight off cancer.


Asunto(s)
Diabetes Mellitus Tipo 1 , Linfocitos T Reguladores , Animales , Humanos , Ratones , Linfocitos T CD8-positivos , Diabetes Mellitus Tipo 1/patología , Tolerancia Inmunológica , Interleucina-2 , Subfamilia K de Receptores Similares a Lectina de Células NK , Receptores de Interleucina-7
3.
Nat Immunol ; 24(1): 174-185, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36564464

RESUMEN

The kinase LCK and CD4/CD8 co-receptors are crucial components of the T cell antigen receptor (TCR) signaling machinery, leading to key T cell fate decisions. Despite decades of research, the roles of CD4-LCK and CD8-LCK interactions in TCR triggering in vivo remain unknown. In this study, we created animal models expressing endogenous levels of modified LCK to resolve whether and how co-receptor-bound LCK drives TCR signaling. We demonstrated that the role of LCK depends on the co-receptor to which it is bound. The CD8-bound LCK is largely dispensable for antiviral and antitumor activity of cytotoxic T cells in mice; however, it facilitates CD8+ T cell responses to suboptimal antigens in a kinase-dependent manner. By contrast, the CD4-bound LCK is required for efficient development and function of helper T cells via a kinase-independent stabilization of surface CD4. Overall, our findings reveal the role of co-receptor-bound LCK in T cell biology, show that CD4- and CD8-bound LCK drive T cell development and effector immune responses using qualitatively different mechanisms and identify the co-receptor-LCK interactions as promising targets for immunomodulation.


Asunto(s)
Proteína Tirosina Quinasa p56(lck) Específica de Linfocito , Linfocitos T Citotóxicos , Ratones , Animales , Linfocitos T Citotóxicos/metabolismo , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Antígenos CD4 , Transducción de Señal , Receptores de Antígenos de Linfocitos T/metabolismo , Antígenos CD8/metabolismo
4.
Nat Immunol ; 23(11): 1644-1652, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36271145

RESUMEN

Interleukin-17A (IL-17A) is a key mediator of protective immunity to yeast and bacterial infections but also drives the pathogenesis of several autoimmune diseases, such as psoriasis or psoriatic arthritis. Here we show that the tetra-transmembrane protein CMTM4 is a subunit of the IL-17 receptor (IL-17R). CMTM4 constitutively associated with IL-17R subunit C to mediate its stability, glycosylation and plasma membrane localization. Both mouse and human cell lines deficient in CMTM4 were largely unresponsive to IL-17A, due to their inability to assemble the IL-17R signaling complex. Accordingly, CMTM4-deficient mice had a severe defect in the recruitment of immune cells following IL-17A administration and were largely resistant to experimental psoriasis, but not to experimental autoimmune encephalomyelitis. Collectively, our data identified CMTM4 as an essential component of IL-17R and a potential therapeutic target for treating IL-17-mediated autoimmune diseases.


Asunto(s)
Artritis Psoriásica , Encefalomielitis Autoinmune Experimental , Psoriasis , Humanos , Ratones , Animales , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Interleucina-17/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Proteínas con Dominio MARVEL/genética
5.
Eur J Immunol ; 51(3): 512-530, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33501647

RESUMEN

Regulatory T cells (Tregs) play a key role in the peripheral self-tolerance and preventing autoimmunity. While classical CD4+ Foxp3+ Tregs are well established, their CD8+ counterparts are still controversial in many aspects including their phenotypic identity and their mechanisms of suppression. Because of these controversies and because of only a limited number of studies documenting the immunoregulatory function of CD8+ Tregs in vivo, the concept of CD8+ Tregs is still not unanimously accepted. We propose that any T-cell subset considered as true regulatory must be distinguishable from other cell types and must suppress in vivo immune responses via a known mechanism. In this article, we revisit the concept of CD8+ Tregs by focusing on the characterization of individual CD8+ T-cell subsets with proposed regulatory capacity separately. Therefore, we review the phenotype and function of CD8+ FOXP3+ T cells, CD8+ CD122+ T cells, CD8+ CD28low/- T cells, CD8+ CD45RClow T cells, T cells expressing CD8αα homodimer and Qa-1-restricted CD8+ T cells to show whether there is sufficient evidence to establish these subsets as bona fide Tregs. Based on the intrinsic ability of CD8+ Treg subsets to promote immune tolerance in animal models, we elaborate on their potential use in clinics.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD28/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Fenotipo , Subgrupos de Linfocitos T/inmunología
6.
EMBO Rep ; 22(2): e50785, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33426789

RESUMEN

Bardet-Biedl Syndrome (BBS) is a pleiotropic genetic disease caused by the dysfunction of primary cilia. The immune system of patients with ciliopathies has not been investigated. However, there are multiple indications that the impairment of the processes typically associated with cilia may have influence on the hematopoietic compartment and immunity. In this study, we analyze clinical data of BBS patients and corresponding mouse models carrying mutations in Bbs4 or Bbs18. We find that BBS patients have a higher prevalence of certain autoimmune diseases. Both BBS patients and animal models have altered red blood cell and platelet compartments, as well as elevated white blood cell levels. Some of the hematopoietic system alterations are associated with BBS-induced obesity. Moreover, we observe that the development and homeostasis of B cells in mice is regulated by the transport complex BBSome, whose dysfunction is a common cause of BBS. The BBSome limits canonical WNT signaling and increases CXCL12 levels in bone marrow stromal cells. Taken together, our study reveals a connection between a ciliopathy and dysregulated immune and hematopoietic systems.


Asunto(s)
Enfermedades Autoinmunes , Síndrome de Bardet-Biedl , Hematopoyesis , Animales , Síndrome de Bardet-Biedl/complicaciones , Síndrome de Bardet-Biedl/genética , Cilios , Modelos Animales de Enfermedad , Hematopoyesis/genética , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Mutación
7.
Nat Commun ; 11(1): 2361, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32398640

RESUMEN

The development of thymic regulatory T cells (Treg) is mediated by Aire-regulated self-antigen presentation on medullary thymic epithelial cells (mTECs) and dendritic cells (DCs), but the cooperation between these cells is still poorly understood. Here we show that signaling through Toll-like receptors (TLR) expressed on mTECs regulates the production of specific chemokines and other genes associated with post-Aire mTEC development. Using single-cell RNA-sequencing, we identify a new thymic CD14+Sirpα+ population of monocyte-derived dendritic cells (CD14+moDC) that are enriched in the thymic medulla and effectively acquire mTEC-derived antigens in response to the above chemokines. Consistently, the cellularity of CD14+moDC is diminished in mice with MyD88-deficient TECs, in which the frequency and functionality of thymic CD25+Foxp3+ Tregs are decreased, leading to aggravated mouse experimental colitis. Thus, our findings describe a TLR-dependent function of mTECs for the recruitment of CD14+moDC, the generation of Tregs, and thereby the establishment of central tolerance.


Asunto(s)
Colitis/inmunología , Células Dendríticas/inmunología , Células Epiteliales/inmunología , Linfocitos T Reguladores/inmunología , Timo/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno , Autoantígenos/inmunología , Separación Celular , Quimiocinas/inmunología , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Citometría de Flujo , Receptores de Lipopolisacáridos/metabolismo , Ratones , Receptores Inmunológicos/metabolismo , Autotolerancia , Análisis de Secuencia de ARN , Transducción de Señal/inmunología , Análisis de la Célula Individual , Linfocitos T Reguladores/trasplante , Timo/citología , Receptores Toll-Like/metabolismo , Regulación hacia Arriba
8.
Hum Mutat ; 40(11): 2068-2087, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31283077

RESUMEN

Bardet-Biedl syndrome (BBS) is a recessive genetic disease causing multiple organ anomalies. Most patients carry mutations in genes encoding for the subunits of the BBSome, an octameric ciliary transport complex, or accessory proteins involved in the BBSome assembly or function. BBS proteins have been extensively studied using in vitro, cellular, and animal models. However, the molecular functions of particular BBS proteins and the etiology of the BBS symptoms are still largely elusive. In this study, we applied a meta-analysis approach to study the genotype-phenotype association in humans using our database of all reported BBS patients. The analysis revealed that the identity of the causative gene and the character of the mutation partially predict the clinical outcome of the disease. Besides their potential use for clinical prognosis, our analysis revealed functional differences of particular BBS genes in humans. Core BBSome subunits BBS2, BBS7, and BBS9 manifest as more critical for the function and development of kidneys than peripheral subunits BBS1, BBS4, and BBS8/TTC8, suggesting that incomplete BBSome retains residual function at least in the kidney.


Asunto(s)
Síndrome de Bardet-Biedl/diagnóstico , Síndrome de Bardet-Biedl/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Fenotipo , Factores de Ribosilacion-ADP/genética , Disfunción Cognitiva/genética , Anomalías Congénitas/genética , Estudios de Asociación Genética/métodos , Humanos , Riñón/anomalías , Enfermedades Renales/congénito , Enfermedades Renales/genética , Mutación , Penetrancia , Proteínas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...