Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Elife ; 112022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35050850

RESUMEN

T cell activation requires engagement of a cognate antigen by the T cell receptor (TCR) and the co-stimulatory signal of CD28. Both TCR and CD28 aggregate into clusters at the plasma membrane of activated T cells. While the role of TCR clustering in T cell activation has been extensively investigated, little is known about how CD28 clustering contributes to CD28 signalling. Here, we report that upon CD28 triggering, the BAR-domain protein sorting nexin 9 (SNX9) is recruited to CD28 clusters at the immunological synapse. Using three-dimensional correlative light and electron microscopy, we show that SNX9 generates membrane tubulation out of CD28 clusters. Our data further reveal that CD28 clusters are in fact dynamic structures and that SNX9 regulates their stability as well as CD28 phosphorylation and the resulting production of the cytokine IL-2. In summary, our work suggests a model in which SNX9-mediated tubulation generates a membrane environment that promotes CD28 triggering and downstream signalling events.


Asunto(s)
Antígenos CD28 , Membrana Celular , Transducción de Señal/genética , Nexinas de Clasificación , Animales , Antígenos CD28/genética , Antígenos CD28/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Humanos , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Células Jurkat , Activación de Linfocitos/genética , Ratones , Ratones Transgénicos , Fosforilación , Receptores de Antígenos de Linfocitos T/metabolismo , Nexinas de Clasificación/genética , Nexinas de Clasificación/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo
2.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768945

RESUMEN

CRAC, which plays important role in Ca2+-dependent T-lymphocyte activation, is composed of the ER-resident STIM1 and the plasma membrane Orai1 pore-forming subunit. Both accumulate at the immunological synapse (IS) between a T cell and an antigen-presenting cell (APC). We hypothesized that adapter/interacting proteins regulate Orai1 residence in the IS. We could show that mGFP-tagged Orai1-Full channels expressed in Jurkat cells had a biphasic IS-accumulation kinetics peaked at 15 min. To understand the background of Orai1 IS-redistribution we knocked down STIM1 and SAP97 (adaptor protein with a short IS-residency (15 min) and ability to bind Orai1 N-terminus): the mGFP-Orai1-Full channels kept on accumulating in the IS up to the 60th minute in the STIM1- and SAP97-lacking Jurkat cells. Deletion of Orai1 N terminus (mGFP-Orai1-Δ72) resulted in the same time course as described for STIM1/SAP97 knock-down cells. Ca2+-imaging of IS-engaged T-cells revealed that of Orai1 residency modifies the Ca2+-response: cells expressing mGFP-Orai1-Δ72 construct or mGFP-Orai1-Full in SAP-97 knock-down cells showed higher number of Ca2+-oscillation up to the 90th minute after IS formation. Overall, these data suggest that SAP97 may contribute to the short-lived IS-residency of Orai1 and binding of STIM1 to Orai1 N-terminus is necessary for SAP97-Orai1 interaction.


Asunto(s)
Señalización del Calcio/inmunología , Sinapsis Inmunológicas/metabolismo , Proteína ORAI1/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Inmunidad Adaptativa , Homólogo 1 de la Proteína Discs Large/antagonistas & inhibidores , Homólogo 1 de la Proteína Discs Large/genética , Homólogo 1 de la Proteína Discs Large/metabolismo , Retículo Endoplásmico/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/inmunología , Células Jurkat , Cinética , Activación de Linfocitos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/química , Proteína ORAI1/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Molécula de Interacción Estromal 1/antagonistas & inhibidores , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34006637

RESUMEN

The liver X receptor (LXR) is a key transcriptional regulator of cholesterol, fatty acid, and phospholipid metabolism. Dynamic remodeling of immunometabolic pathways, including lipid metabolism, is a crucial step in T cell activation. Here, we explored the role of LXR-regulated metabolic processes in primary human CD4+ T cells and their role in controlling plasma membrane lipids (glycosphingolipids and cholesterol), which strongly influence T cell immune signaling and function. Crucially, we identified the glycosphingolipid biosynthesis enzyme glucosylceramide synthase as a direct transcriptional LXR target. LXR activation by agonist GW3965 or endogenous oxysterol ligands significantly altered the glycosphingolipid:cholesterol balance in the plasma membrane by increasing glycosphingolipid levels and reducing cholesterol. Consequently, LXR activation lowered plasma membrane lipid order (stability), and an LXR antagonist could block this effect. LXR stimulation also reduced lipid order at the immune synapse and accelerated activation of proximal T cell signaling molecules. Ultimately, LXR activation dampened proinflammatory T cell function. Finally, compared with responder T cells, regulatory T cells had a distinct pattern of LXR target gene expression corresponding to reduced lipid order. This suggests LXR-driven lipid metabolism could contribute to functional specialization of these T cell subsets. Overall, we report a mode of action for LXR in T cells involving the regulation of glycosphingolipid and cholesterol metabolism and demonstrate its relevance in modulating T cell function.


Asunto(s)
Colesterol/genética , Glicoesfingolípidos/genética , Receptores X del Hígado/inmunología , Linfocitos T/inmunología , Adolescente , Adulto , Benzoatos/farmacología , Bencilaminas/farmacología , Membrana Celular , Colesterol/inmunología , Femenino , Glucosiltransferasas/genética , Glicoesfingolípidos/biosíntesis , Glicoesfingolípidos/inmunología , Humanos , Sinapsis Inmunológicas/efectos de los fármacos , Sinapsis Inmunológicas/genética , Ligandos , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/inmunología , Receptores X del Hígado/agonistas , Receptores X del Hígado/antagonistas & inhibidores , Receptores X del Hígado/genética , Masculino , Redes y Vías Metabólicas/inmunología , Persona de Mediana Edad , Oxiesteroles/farmacología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Linfocitos T/metabolismo , Adulto Joven
4.
Int J Mol Sci ; 22(5)2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33806327

RESUMEN

Personalised medicine is the future and hope for many patients, including those with cancers. Early detection, as well as rapid, well-selected treatment, are key factors leading to a good prognosis. MicroRNA mediated gene regulation is a promising area of development for new diagnostic and therapeutic methods, crucial for better prospects for patients. Bladder cancer is a frequent neoplasm, with high lethality and lacking modern, advanced therapeutic modalities, such as immunotherapy. MicroRNAs are involved in bladder cancer pathogenesis, proliferation, control and response to treatment, which we summarise in this perspective in response to lack of recent review publications in this field. We further performed a correlation-based analysis of microRNA and gene expression data in bladder cancer (BLCA) TCGA dataset. We identified 27 microRNAs hits with opposite expression profiles to genes involved in immune response in bladder cancer, and 24 microRNAs hits with similar expression profiles. We discuss previous studies linking the functions of these microRNAs to bladder cancer and assess if they are good candidates for personalised medicine therapeutics and diagnostics. The discussed functions include regulation of gene expression, interplay with transcription factors, response to treatment, apoptosis, cell proliferation and angiogenesis, initiation and development of cancer, genome instability and tumour-associated inflammatory reaction.


Asunto(s)
Proteínas de Punto de Control Inmunitario/genética , MicroARNs/genética , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/inmunología , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Bases de Datos Genéticas , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Sinapsis Inmunológicas/genética , Modelos Genéticos , ARN Mensajero/genética , ARN Neoplásico/genética
5.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33903232

RESUMEN

The development of follicular helper CD4 T (TFH) cells is a dynamic process resulting in a heterogenous pool of TFH subsets. However, the cellular and molecular determinants of this heterogeneity and the possible mechanistic links between them is not clear. We found that human TFH differentiation is associated with significant changes in phenotypic, chemokine, functional, metabolic and transcriptional profile. Furthermore, this differentiation was associated with distinct positioning to follicular proliferating B cells. Single-cell T cell receptor (TCR) clonotype analysis indicated the transitioning toward PD-1hiCD57hi phenotype. Furthermore, the differentiation of TFH cells was associated with significant reduction in TCR level and drastic changes in immunological synapse formation. TFH synapse lacks a tight cSMAC (central supra molecular activation Cluster) but displays the TCR in peripheral microclusters, which are potentially advantageous in the ability of germinal center (GC) B cells to receive necessary help. Our data reveal significant aspects of human TFH heterogeneity and suggest that the PD-1hiCD57hi TFH cells, in particular, are endowed with distinctive programming and spatial positioning for optimal GC B cell help.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/inmunología , Receptores de Antígenos de Linfocitos T/genética , Células T Auxiliares Foliculares/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos CD57/genética , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Quimiocinas/genética , Centro Germinal/inmunología , Centro Germinal/metabolismo , Humanos , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/inmunología , Activación de Linfocitos/inmunología , Fenotipo , Receptor de Muerte Celular Programada 1/genética , Receptores de Antígenos de Linfocitos T/inmunología , Células T Auxiliares Foliculares/metabolismo , Subgrupos de Linfocitos T/inmunología
6.
Nat Commun ; 12(1): 2163, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846331

RESUMEN

γδ T cells are a distinct subgroup of T cells that bridge the innate and adaptive immune system and can attack cancer cells in an MHC-unrestricted manner. Trials of adoptive γδ T cell transfer in solid tumors have had limited success. Here, we show that DNA methyltransferase inhibitors (DNMTis) upregulate surface molecules on cancer cells related to γδ T cell activation using quantitative surface proteomics. DNMTi treatment of human lung cancer potentiates tumor lysis by ex vivo-expanded Vδ1-enriched γδ T cells. Mechanistically, DNMTi enhances immune synapse formation and mediates cytoskeletal reorganization via coordinated alterations of DNA methylation and chromatin accessibility. Genetic depletion of adhesion molecules or pharmacological inhibition of actin polymerization abolishes the potentiating effect of DNMTi. Clinically, the DNMTi-associated cytoskeleton signature stratifies lung cancer patients prognostically. These results support a combinatorial strategy of DNMTis and γδ T cell-based immunotherapy in lung cancer management.


Asunto(s)
Citoesqueleto/metabolismo , Citotoxicidad Inmunológica/genética , Epigénesis Genética , Sinapsis Inmunológicas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Línea Celular Tumoral , Citoesqueleto/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Decitabina/farmacología , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sinapsis Inmunológicas/efectos de los fármacos , Marcaje Isotópico , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/metabolismo , Masculino , Ratones Endogámicos NOD , Fosfotirosina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Cell Biol ; 220(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33533935

RESUMEN

Dendritic cells (DCs) are crucial for the priming of naive T cells and the initiation of adaptive immunity. Priming is initiated at a heterologous cell-cell contact, the immunological synapse (IS). While it is established that F-actin dynamics regulates signaling at the T cell side of the contact, little is known about the cytoskeletal contribution on the DC side. Here, we show that the DC actin cytoskeleton is decisive for the formation of a multifocal synaptic structure, which correlates with T cell priming efficiency. DC actin at the IS appears in transient foci that are dynamized by the WAVE regulatory complex (WRC). The absence of the WRC in DCs leads to stabilized contacts with T cells, caused by an increase in ICAM1-integrin-mediated cell-cell adhesion. This results in lower numbers of activated and proliferating T cells, demonstrating an important role for DC actin in the regulation of immune synapse functionality.


Asunto(s)
Actinas/inmunología , Comunicación Celular/inmunología , Células Dendríticas/inmunología , Sinapsis Inmunológicas/inmunología , Linfocitos T/inmunología , Actinas/genética , Animales , Adhesión Celular/genética , Adhesión Celular/inmunología , Comunicación Celular/genética , Proliferación Celular/genética , Femenino , Sinapsis Inmunológicas/genética , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Masculino , Ratones , Ratones Noqueados
8.
Front Immunol ; 11: 600012, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33343572

RESUMEN

IL4I1 is an immunoregulatory enzyme that inhibits CD8 T-cell proliferation in vitro and in the tumoral context. Here, we dissected the effect of IL4I1 on CD8 T-cell priming by studying the differentiation of a transgenic CD8 T-cell clone and the endogenous repertoire in a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection. Unexpectedly, we show that IL4I1 accelerates the expansion of functional effector CD8 T cells during the first several days after infection and increases the average affinity of the elicited repertoire, supporting more efficient LCMV clearance in WT mice than IL4I1-deficient mice. Conversely, IL4I1 restrains the differentiation of CD8 T-cells into long-lived memory precursors and favors the memory response to the most immunodominant peptides. IL4I1 expression does not affect the phenotype or antigen-presenting functions of dendritic cells (DCs), but directly reduces the stability of T-DC immune synapses in vitro, thus dampening T-cell activation. Overall, our results support a model in which IL4I1 increases the threshold of T-cell activation, indirectly promoting the priming of high-affinity clones while limiting memory T-cell differentiation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Memoria Inmunológica , L-Aminoácido Oxidasa/inmunología , Activación de Linfocitos , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/patología , Células Dendríticas/inmunología , Células Dendríticas/patología , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/inmunología , Sinapsis Inmunológicas/patología , L-Aminoácido Oxidasa/genética , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/genética , Ratones , Ratones Noqueados
9.
Front Immunol ; 11: 581119, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240268

RESUMEN

Congenital defects of the immune system called primary immunodeficiency disorders (PID) describe a group of diseases characterized by a decrease, an absence, or a malfunction of at least one part of the immune system. As a result, PID patients are more prone to develop life-threatening complications, including cancer. PID currently include over 400 different disorders, however, the variety of PID-related cancers is narrow. We discuss here reasons for this clinical phenotype. Namely, PID can lead to cell intrinsic failure to control cell transformation, failure to activate tumor surveillance by cytotoxic cells or both. As the most frequent tumors seen among PID patients stem from faulty lymphocyte development leading to leukemia and lymphoma, we focus on the extensive genomic alterations needed to create the vast diversity of B and T lymphocytes with potential to recognize any pathogen and why defects in these processes lead to malignancies in the immunodeficient environment of PID patients. In the second part of the review, we discuss PID affecting tumor surveillance and especially membrane trafficking defects caused by altered exocytosis and regulation of the actin cytoskeleton. As an impairment of these membrane trafficking pathways often results in dysfunctional effector immune cells, tumor cell immune evasion is elevated in PID. By considering new anti-cancer treatment concepts, such as transfer of genetically engineered immune cells, restoration of anti-tumor immunity in PID patients could be an approach to complement standard therapies.


Asunto(s)
Leucemia de Células B/etiología , Linfoma de Células B/etiología , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Reparación del ADN/genética , Reparación del ADN/inmunología , Exocitosis/genética , Exocitosis/inmunología , Inestabilidad Genómica , Humanos , Sinapsis Inmunológicas/genética , Leucemia de Células B/genética , Leucemia de Células B/inmunología , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Modelos Inmunológicos , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/inmunología , Factores de Riesgo , Escape del Tumor/genética
10.
EMBO J ; 39(16): e104730, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32643825

RESUMEN

The chimeric antigen receptor (CAR) directs T cells to target and kill specific cancer cells. Despite the success of CAR T therapy in clinics, the intracellular signaling pathways that lead to CAR T cell activation remain unclear. Using CD19 CAR as a model, we report that, similar to the endogenous T cell receptor (TCR), antigen engagement triggers the formation of CAR microclusters that transduce downstream signaling. However, CAR microclusters do not coalesce into a stable central supramolecular activation cluster (cSMAC). Moreover, LAT, an essential scaffold protein for TCR signaling, is not required for microcluster formation, immunological synapse formation, nor actin remodeling following CAR activation. However, CAR T cells still require LAT for an optimal production of the cytokine IL-2. Together, these data show that CAR T cells can bypass LAT for a subset of downstream signaling outputs, thus revealing a rewired signaling pathway as compared to native T cells.


Asunto(s)
Sinapsis Inmunológicas/inmunología , Interleucina-2/inmunología , Receptores Quiméricos de Antígenos/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Células HEK293 , Humanos , Sinapsis Inmunológicas/genética , Interleucina-2/genética , Células Jurkat , Receptores Quiméricos de Antígenos/genética , Transducción de Señal/genética
11.
Cancer Lett ; 487: 1-9, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32454143

RESUMEN

Chimeric antigen receptor T (CAR-T) therapy faces at least two major obstacles in solid tumors, including to find specific antigen among the heterogeneous tumor mass and to overcome the inhibitory microenvironment. Developing novel strategies to overcome these difficulties has been the burning issue in immunotherapy. Here we came up with the concept of tagging cancer cells by tumor-targeting adenoviruses (Ad). We constructed recombinant Ads expressing CD19 tag driven by tumor-specific promoters, which could label antigenically different tumors for single anti-CD19 CAR-T recognition. One Ad, namely AdC68-TMC-tCD19 could mediate universal tag expression and functional immunological synapse formation between CAR-T and cancer cells. In premixed mice model, all tagged mice survived after CAR-T infusion and tumor volume were inhibited by 91.78%. Furthermore, we combined the tumor tagging ability with oncolysis and generated the replicative AdC68-Sur-E1A-TMC-tCD19. Oncolytic tagging system could diminish established tumors in vivo and prolong mice survival significantly. Therefore, we suggest the universal oncolytic Ad-tagging system in combination with single target CAR-T cells could be a powerful complement in immunotherapy against antigenically mismatched solid tumors.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Adenoviridae/genética , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Antígenos CD19/uso terapéutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/uso terapéutico , Línea Celular Tumoral , Terapia Combinada , Humanos , Sinapsis Inmunológicas/efectos de los fármacos , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/inmunología , Ratones , Viroterapia Oncolítica/tendencias , Virus Oncolíticos/genética , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/uso terapéutico , Linfocitos T/inmunología , Linfocitos T/virología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Signal ; 13(627)2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32291315

RESUMEN

Diacylglycerol kinases (DGKs) limit antigen receptor signaling in immune cells by consuming the second messenger diacylglycerol (DAG) to generate phosphatidic acid (PA). Here, we showed that DGKζ promotes lymphocyte function-associated antigen 1 (LFA-1)-mediated adhesion and F-actin generation at the immune synapse of B cells with antigen-presenting cells (APCs), mostly in a PA-dependent manner. Measurement of single-cell mechanical force generation indicated that DGKζ-deficient B cells exerted lower forces at the immune synapse than did wild-type B cells. Nonmuscle myosin activation and translocation of the microtubule-organizing center (MTOC) to the immune synapse were also impaired in DGKζ-deficient B cells. These functional defects correlated with the decreased ability of B cells to present antigen and activate T cells in vitro. The in vivo germinal center response of DGKζ-deficient B cells was also reduced compared with that of wild-type B cells, indicating that loss of DGKζ in B cells impaired T cell help. Together, our data suggest that DGKζ shapes B cell responses by regulating actin remodeling, force generation, and antigen uptake-related events at the immune synapse. Hence, an appropriate balance in the amounts of DAG and PA is required for optimal B cell function.


Asunto(s)
Linfocitos B/metabolismo , Citoesqueleto/inmunología , Diacilglicerol Quinasa/inmunología , Sinapsis Inmunológicas/inmunología , Animales , Citoesqueleto/genética , Diacilglicerol Quinasa/genética , Sinapsis Inmunológicas/genética , Ratones , Ratones Noqueados
13.
J Cell Biol ; 219(2)2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31977034

RESUMEN

Activation of naive T cells by antigen-presenting cells (APCs) is an essential step in mounting an adaptive immune response. It is known that antigen recognition and T cell receptor (TCR) signaling depend on forces applied by the T cell actin cytoskeleton, but until recently, the underlying mechanisms have been poorly defined. Here, we review recent advances in the field, which show that specific actin-dependent structures contribute to the process in distinct ways. In essence, T cell priming involves a tug-of-war between the cytoskeletons of the T cell and the APC, where the actin cytoskeleton serves as a mechanical intermediate that integrates force-dependent signals. We consider each of the relevant actin-rich T cell structures separately and address how they work together at the topologically and temporally complex cell-cell interface. In addition, we address how this mechanobiology can be incorporated into canonical immunological models to improve how these models explain T cell sensitivity and antigenic specificity.


Asunto(s)
Citoesqueleto de Actina/genética , Actinas/genética , Células Presentadoras de Antígenos/inmunología , Sinapsis Inmunológicas/genética , Mecanotransducción Celular , Citoesqueleto de Actina/inmunología , Actinas/inmunología , Inmunidad Adaptativa/inmunología , Comunicación Celular/inmunología , Citoesqueleto/genética , Citoesqueleto/inmunología , Humanos , Sinapsis Inmunológicas/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Modelos Inmunológicos , Seudópodos/inmunología , Seudópodos/ultraestructura , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/genética , Linfocitos T/inmunología
14.
J Allergy Clin Immunol ; 145(1): 345-357.e9, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31600547

RESUMEN

BACKGROUND: Patients with signal transducer and activator of transcription 5b (STAT5b) deficiency have impairment in T-cell homeostasis and natural killer (NK) cells which leads to autoimmunity, recurrent infections, and combined immune deficiency. OBJECTIVE: In this study we characterized the NK cell defect in STAT5b-deficient human NK cells, as well as Stat5b-/- mice. METHODS: We used multiparametric flow cytometry, functional NK cell assays, microscopy, and a Stat5b-/- mouse model to elucidate the effect of impaired and/or absent STAT5b on NK cell development and function. RESULTS: This alteration generated a nonfunctional CD56bright NK cell subset characterized by low cytokine production. The CD56dim NK cell subset had decreased expression of perforin and CD16 and a greater frequency of cells expressing markers of immature NK cells. We observed low NK cell numbers and impaired NK cell maturation, suggesting that STAT5b is involved in terminal NK cell maturation in Stat5b-/- mice. Furthermore, human STAT5b-deficient NK cells had low cytolytic capacity, and fixed-cell microscopy showed poor convergence of lytic granules. This was accompanied by decreased expression of costimulatory and activating receptors. Interestingly, granule convergence and cytolytic function were restored after IL-2 stimulation. CONCLUSIONS: Our results show that in addition to the impaired terminal maturation of NK cells, human STAT5b mutation leads to impairments in early activation events in NK cell lytic synapse formation. Our data provide further insight into NK cell defects caused by STAT5b deficiency.


Asunto(s)
Inmunidad Celular , Sinapsis Inmunológicas/inmunología , Células Asesinas Naturales/inmunología , Mutación , Factor de Transcripción STAT5/inmunología , Animales , Femenino , Humanos , Sinapsis Inmunológicas/genética , Células Asesinas Naturales/patología , Masculino , Ratones , Ratones Noqueados , Factor de Transcripción STAT5/genética
15.
J Clin Invest ; 130(2): 974-980, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31714899

RESUMEN

Cancer immune evasion is achieved through multiple layers of immune tolerance mechanisms including immune editing, recruitment of tolerogenic immune cells, and secretion of immunosuppressive cytokines. Recent success with immune checkpoint inhibitors in cancer immunotherapy suggests a dysfunctional immune synapse as a pivotal tolerogenic mechanism. Tumor cells express immune synapse proteins to suppress the immune system, which is often modulated by epigenetic mechanisms. When the methylation status of key immune synapse genes was interrogated, we observed disproportionately hypermethylated costimulatory genes and hypomethylation of immune checkpoint genes, which were negatively associated with functional T cell recruitment to the tumor microenvironment. Therefore, the methylation status of immune synapse genes reflects tumor immunogenicity and correlates with survival.


Asunto(s)
Metilación de ADN , ADN de Neoplasias/inmunología , Epigénesis Genética/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Genes Relacionados con las Neoplasias , Sinapsis Inmunológicas/inmunología , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Línea Celular Tumoral , ADN de Neoplasias/genética , Humanos , Sinapsis Inmunológicas/genética , Neoplasias/genética , Neoplasias/patología , Linfocitos T/inmunología , Microambiente Tumoral/genética
16.
Sci Signal ; 12(612)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31848320

RESUMEN

Genetic diversity in human natural killer (NK) cell receptors is linked to resistance and susceptibility to many diseases. Here, we tested the effect of this diversity on the nanoscale organization of killer cell immunoglobulin-like receptors (KIRs). Using superresolution microscopy, we found that inhibitory KIRs encoded by different genes and alleles were organized differently at the surface of primary human NK cells. KIRs that were found at low abundance assembled into smaller clusters than those formed by KIRs that were more highly abundant, and at low abundance, there was a greater proportion of KIRs in clusters. Upon receptor triggering, a structured interface called the immune synapse assembles, which facilitates signal integration and controls NK cell responses. Here, triggering of low-abundance receptors resulted in less phosphorylation of the downstream phosphatase SHP-1 but more phosphorylation of the adaptor protein Crk than did triggering of high-abundance receptors. In cells with greater KIR abundance, SHP-1 dephosphorylated Crk, which potentiated NK cell spreading during activation. Thus, genetic variation modulates both the abundance and nanoscale organization of inhibitory KIRs. That is, as well as the number of receptors at the cell surface varying with genotype, the way in which these receptors are organized in the membrane also varies. Essentially, a change in the average surface abundance of a protein at the cell surface is a coarse descriptor entwined with changes in local nanoscale clustering. Together, our data indicate that genetic diversity in inhibitory KIRs affects membrane-proximal signaling and, unexpectedly, the formation of activating immune synapses.


Asunto(s)
Variación Genética , Sinapsis Inmunológicas , Células Asesinas Naturales/inmunología , Receptores KIR , Transducción de Señal , Línea Celular Tumoral , Humanos , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Proteínas Proto-Oncogénicas c-crk/genética , Proteínas Proto-Oncogénicas c-crk/inmunología , Receptores KIR/genética , Receptores KIR/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología
17.
Adv Immunol ; 144: 23-63, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31699219

RESUMEN

B cells are essential to the adaptive immune system for providing the humoral immunity against cohorts of pathogens. The presentation of antigen to the B cell receptor (BCR) leads to the initiation of B cell activation, which is a process sensitive to the stiffness features of the substrates presenting the antigens. Mechanosensing of the B cells, potentiated through BCR signaling and the adhesion molecules, efficiently regulates B cell activation, proliferation and subsequent antibody responses. Defects in sensing of the antigen-presenting substrates can lead to the activation of autoreactive B cells in autoimmune diseases. The use of high-resolution, high-speed live-cell imaging along with the sophisticated biophysical materials, has uncovered the mechanisms underlying the initiation of B cell activation within seconds of its engagement with the antigen presenting substrates. In this chapter, we reviewed studies that have contributed to uncover the molecular mechanisms of B cell mechanosensing during the initiation of B cell activation.


Asunto(s)
Formación de Anticuerpos , Presentación de Antígeno , Linfocitos B/inmunología , Mecanotransducción Celular/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología , Animales , Enfermedades Autoinmunes/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/inmunología , Humanos , Sinapsis Inmunológicas/química , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/patología , Integrinas/inmunología , Proteínas Motoras Moleculares/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo
18.
PLoS One ; 14(7): e0210377, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31269031

RESUMEN

Immunological synapse (IS) formation between a T cell and an antigen-presenting cell is accompanied by the reorientation of the T cell centrosome toward the interface. This polarization response is thought to enhance the specificity of T cell effector function by enabling the directional secretion of cytokines and cytotoxic factors toward the antigen-presenting cell. Centrosome reorientation is controlled by polarized signaling through diacylglycerol (DAG) and protein kinase C (PKC). This drives the recruitment of the motor protein dynein to the IS, where it pulls on microtubules to reorient the centrosome. Here, we used T cell receptor photoactivation and imaging methodology to investigate the mechanisms controlling dynein accumulation at the synapse. Our results revealed a remarkable spatiotemporal correlation between dynein recruitment to the synaptic membrane and the depletion of cortical filamentous actin (F-actin) from the same region, suggesting that the two events were causally related. Consistent with this hypothesis, we found that pharmacological disruption of F-actin dynamics in T cells impaired both dynein accumulation and centrosome reorientation. DAG and PKC signaling were necessary for synaptic F-actin clearance and dynein accumulation, while calcium signaling and microtubules were dispensable for both responses. Taken together, these data provide mechanistic insight into the polarization of cytoskeletal regulators and highlight the close coordination between microtubule and F-actin architecture at the IS.


Asunto(s)
Actinas/inmunología , Células Presentadoras de Antígenos/inmunología , Dineínas/inmunología , Sinapsis Inmunológicas/inmunología , Linfocitos T/inmunología , Actinas/genética , Animales , Células Presentadoras de Antígenos/citología , Centrosoma/inmunología , Dineínas/genética , Sinapsis Inmunológicas/genética , Ratones , Ratones Transgénicos , Microtúbulos/genética , Microtúbulos/inmunología , Linfocitos T/citología
19.
J Cell Biol ; 218(7): 2247-2264, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31197029

RESUMEN

B lymphocytes capture antigens from the surface of presenting cells by forming an immune synapse. Local secretion of lysosomes, which are guided to the synaptic membrane by centrosome repositioning, can facilitate the extraction of immobilized antigens. However, the molecular basis underlying their delivery to precise domains of the plasma membrane remains elusive. Here we show that microtubule stabilization, triggered by engagement of the B cell receptor, acts as a cue to release centrosome-associated Exo70, which is redistributed to the immune synapse. This process is coupled to the recruitment and activation of GEF-H1, which is required for assembly of the exocyst complex, used to promote tethering and fusion of lysosomes at the immune synapse. B cells silenced for GEF-H1 or Exo70 display defective lysosome secretion, which results in impaired antigen extraction and presentation. Thus, centrosome repositioning coupled to changes in microtubule stability orchestrates the spatial-temporal distribution of the exocyst complex to promote polarized lysosome secretion at the immune synapse.


Asunto(s)
Presentación de Antígeno/genética , Linfocitos B/inmunología , Sinapsis Inmunológicas/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Proteínas de Transporte Vesicular/genética , Animales , Presentación de Antígeno/inmunología , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Membrana Celular/inmunología , Polaridad Celular/genética , Polaridad Celular/inmunología , Centrosoma/inmunología , Exocitosis/genética , Exocitosis/inmunología , Lisosomas/genética , Lisosomas/inmunología , Ratones , Microtúbulos/genética , Microtúbulos/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología
20.
Cell Immunol ; 341: 103919, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31047647

RESUMEN

Wiskott-Aldrich syndrome (WAS) is a form of primary immunodeficiency (PIDs) resulting from mutations of the gene that encodes Wiskott-Aldrich syndrome protein (WASp). WASp is the first identified and most widely studied protein belonging to the actin nucleation-promoting factor family and plays significant role in integrating and transforming signals from critical receptors on the cell surface to actin remodeling. WASp functions in immune defense and homeostasis through the regulation of actin cytoskeleton-dependent cellular processes as well as processes uncoupled with actin polymerization like nuclear transcription programs. In this article, we review the mechanisms of WASp activation through an understanding of its structure. We further discuss the role of WASp in adaptive immunity, paying special attention to some recent findings on the crucial role of WASp in the formation of immunological synapse, the regulation of T follicular helper (Tfh) cells and in the prevention of autoimmunity.


Asunto(s)
Citoesqueleto de Actina/inmunología , Linfocitos B/inmunología , Homeostasis/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Síndrome de Wiskott-Aldrich/inmunología , Citoesqueleto de Actina/genética , Inmunidad Adaptativa , Animales , Autoinmunidad/genética , Linfocitos B/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Homeostasis/genética , Humanos , Inmunidad Innata , Sinapsis Inmunológicas/genética , Ratones , Transducción de Señal , Linfocitos T Colaboradores-Inductores/patología , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/patología , Proteína del Síndrome de Wiskott-Aldrich/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA