Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.391
Filtrar
1.
Yakugaku Zasshi ; 144(5): 497-501, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-38692923

RESUMEN

Signal-transducing adaptor protein-2 (STAP-2) is a unique scaffold protein that regulates several immunological signaling pathways, including LIF/LIF receptor and LPS/TLR4 signals. STAP-2 is required for Fas/FasL-dependent T cell apoptosis and SDF-1α-induced T cell migration. Conversely, STAP-2 modulates integrin-mediated T cell adhesion, suggesting that STAP-2 is essential for several negative and positive T cell functions. However, whether STAP-2 is involved in T cell-antigen receptor (TCR)-mediated T cell activation is unknown. STAP-2 deficiency was recently reported to suppress TCR-mediated T cell activation by inhibiting LCK-mediated CD3ζ and ZAP-70 activation. Using STAP-2 deficient mice, it was demonstrated that STAP-2 is required for the pathogenesis of Propionibacterium acnes-induced granuloma formation and experimental autoimmune encephalomyelitis. Here, detailed functions of STAP-2 in TCR-mediated T cell activation, and how STAP-2 affects the pathogenesis of T cell-mediated inflammation and immune diseases, are reviewed.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T , Transducción de Señal , Linfocitos T , Proteína Tirosina Quinasa ZAP-70 , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Complejo CD3 , Adhesión Celular , Movimiento Celular , Quimiocina CXCL12/fisiología , Quimiocina CXCL12/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/etiología , Inflamación/inmunología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/fisiología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Propionibacterium acnes/fisiología , Propionibacterium acnes/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Proteína Tirosina Quinasa ZAP-70/metabolismo , Proteína Tirosina Quinasa ZAP-70/fisiología
2.
Int J Cancer ; 150(4): 688-704, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34716584

RESUMEN

The surface inhibitory receptor NKG2A forms heterodimers with the invariant CD94 chain and is expressed on a subset of activated CD8 T cells. As antibodies to block NKG2A are currently tested in several efficacy trials for different tumor indications, it is important to characterize the NKG2A+ CD8 T cell population in the context of other inhibitory receptors. Here we used a well-controlled culture system to study the kinetics of inhibitory receptor expression. Naïve mouse CD8 T cells were synchronously and repeatedly activated by artificial antigen presenting cells in the presence of the homeostatic cytokine IL-7. The results revealed NKG2A as a late inhibitory receptor, expressed after repeated cognate antigen stimulations. In contrast, the expression of PD-1, TIGIT and LAG-3 was rapidly induced, hours after first contact and subsequently down regulated during each resting phase. This late, but stable expression kinetics of NKG2A was most similar to that of TIM-3 and CD39. Importantly, single-cell transcriptomics of human tumor-infiltrating lymphocytes (TILs) showed indeed that these receptors were often coexpressed by the same CD8 T cell cluster. Furthermore, NKG2A expression was associated with cell division and was promoted by TGF-ß in vitro, although TGF-ß signaling was not necessary in a mouse tumor model in vivo. In summary, our data show that PD-1 reflects recent TCR triggering, but that NKG2A is induced after repeated antigen stimulations and represents a late inhibitory receptor. Together with TIM-3 and CD39, NKG2A might thus mark actively dividing tumor-specific TILs.


Asunto(s)
Proteínas de Punto de Control Inmunitario/fisiología , Subfamília C de Receptores Similares a Lectina de Células NK/fisiología , Animales , Antígenos CD/fisiología , Linfocitos T CD8-positivos/inmunología , División Celular , Receptor 2 Celular del Virus de la Hepatitis A/fisiología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/fisiología , Receptores Inmunológicos/fisiología , Factor de Crecimiento Transformador beta/farmacología , Microambiente Tumoral , Proteína del Gen 3 de Activación de Linfocitos
3.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34845016

RESUMEN

Unlike conventional αß T cells, γδ T cells typically recognize nonpeptide ligands independently of major histocompatibility complex (MHC) restriction. Accordingly, the γδ T cell receptor (TCR) can potentially recognize a wide array of ligands; however, few ligands have been described to date. While there is a growing appreciation of the molecular bases underpinning variable (V)δ1+ and Vδ2+ γδ TCR-mediated ligand recognition, the mode of Vδ3+ TCR ligand engagement is unknown. MHC class I-related protein, MR1, presents vitamin B metabolites to αß T cells known as mucosal-associated invariant T cells, diverse MR1-restricted T cells, and a subset of human γδ T cells. Here, we identify Vδ1/2- γδ T cells in the blood and duodenal biopsy specimens of children that showed metabolite-independent binding of MR1 tetramers. Characterization of one Vδ3Vγ8 TCR clone showed MR1 reactivity was independent of the presented antigen. Determination of two Vδ3Vγ8 TCR-MR1-antigen complex structures revealed a recognition mechanism by the Vδ3 TCR chain that mediated specific contacts to the side of the MR1 antigen-binding groove, representing a previously uncharacterized MR1 docking topology. The binding of the Vδ3+ TCR to MR1 did not involve contacts with the presented antigen, providing a basis for understanding its inherent MR1 autoreactivity. We provide molecular insight into antigen-independent recognition of MR1 by a Vδ3+ γδ TCR that strengthens an emerging paradigm of antibody-like ligand engagement by γδ TCRs.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Linfocitos Intraepiteliales/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Adulto , Presentación de Antígeno , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/fisiología , Humanos , Linfocitos Intraepiteliales/fisiología , Ligandos , Masculino , Antígenos de Histocompatibilidad Menor/química , Antígenos de Histocompatibilidad Menor/fisiología , Células T Invariantes Asociadas a Mucosa/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/fisiología
4.
Regul Toxicol Pharmacol ; 127: 105064, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34656748

RESUMEN

Over the last decade, immunotherapy has established itself as an important novel approach in the treatment of cancer, resulting in a growing importance in oncology. Engineered T cell therapies, namely chimeric antigen receptor (CAR) T cells and T cell receptor (TCR) T cell therapies, are platform technologies that have enabled the development of products with remarkable efficacy in several hematological malignancies and are thus the focus of intense research and development activity. While engineered T cell therapies offer promise in addressing currently intractable cancers, they also present unique challenges, including their nonclinical safety assessment. A workshop organized by HESI and the US Food and Drug Administration (FDA) was held to provide an interdisciplinary forum for representatives of industry, academia and regulatory authorities to share information and debate on current practices for the nonclinical safety evaluation of engineered T cell therapies. This manuscript leverages what was discussed at this workshop to provide an overview of the current important nonclinical safety assessment considerations for the development of these therapeutic modalities (cytokine release syndrome, neurotoxicity, on-target/off-tumor toxicities, off-target effects, gene editing or vector integration-associated genomic injury). The manuscript also discusses approaches used for hazard identification or risk assessment and provides a regulatory perspective on such aspects.


Asunto(s)
Ingeniería Celular/métodos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Linfocitos T/inmunología , Síndrome de Liberación de Citoquinas/fisiopatología , Edición Génica , Inmunoterapia Adoptiva/efectos adversos , Síndromes de Neurotoxicidad/fisiopatología , Receptores de Antígenos de Linfocitos T/fisiología , Medición de Riesgo
5.
Cell Biol Int ; 45(6): 1220-1230, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33527533

RESUMEN

Although T-cell receptors (TCRs) are related to the progression of breast cancer (BC), their prognostic values remain unclear. We downloaded the messenger RNA (mRNA) profiles and corresponding clinical information of 1413 BC patients from the Cancer Genome Atlas and Gene Expression Omnibus database, respectively. The different expression analysis of 104 TCRs in BC samples was performed, and the consensus clustering based on 104 TCRs was performed by using the K-mean method of R language. Univariate cox regression analysis was used to screen TCRs significantly associated with the prognosis of BC, and LASSO Cox analysis was applied to optimize key TCRs. The risk score was calculated using the prognostic model constructed based on six optimal TCRs, and multivariate Cox regression analysis was used to determine whether it was an independent prognostic signature. Finally, the nomogram was constructed to predict the overall survival of BC patients. Six optimal TCRs (ZAP70, GRAP2, NFKBIE, IFNG, NFKBIA, and PAK5), which were favorable for the prognosis of BC patients, were screened. Risk score could reliably predict the prognosis of BC patients as an independent prognostic signature. In addition, when bringing into two independent prognostic signatures, age and risk score, the nomogram model could better predict the overall survival of BC patients. Our results suggested that the poor prognosis of BC patients with high risk might be due to an immunosuppressive microenvironment. In summary, a prognostic risk model based on six TCRs was established and could efficiently predict the prognosis of BC patients.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Biomarcadores de Tumor/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Pronóstico , Microambiente Tumoral
6.
Mol Immunol ; 130: 64-68, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33360378

RESUMEN

The monomorphic MHC-class I-like molecule, MR1, presents small metabolites to T cells. MR1 is the restriction element for microbe-reactive mucosal-associated invariant T (MAIT) cells. MAIT cells have limited TCR usage, including a semi-invariant TCR alpha chain and express high levels of CD161 and CD26. In addition to microbial lumazine metabolites, recent studies have demonstrated that MR1 is able to capture a variety of diverse chemical entities including folate-derivatives, a number of drug-like and other synthetic small molecules, and as yet undefined compounds of self-origin. This capacity of MR1 to bind distinct ligands likely accounts for the recent identification of additional, non-canonical, subsets of MR1-restricted T (MR1T) cells. These subsets can be defined based on their ability to recognize diverse microbes as well as their reactivity to non-microbial cell-endogenous ligands, including tumor-associated antigens. Herein, we will discuss our current understanding of MR1T cell diversity in terms of TCR usage, ligand recognition and functional attributes.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Células T Invariantes Asociadas a Mucosa/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/fisiología , Subgrupos de Linfocitos T/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunidad Mucosa/inmunología , Antígenos de Histocompatibilidad Menor/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta
7.
Nat Immunol ; 22(1): 53-66, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33230330

RESUMEN

Regenerative stem cell-like memory (TSCM) CD8+ T cells persist longer and produce stronger effector functions. We found that MEK1/2 inhibition (MEKi) induces TSCM that have naive phenotype with self-renewability, enhanced multipotency and proliferative capacity. This is achieved by delaying cell division and enhancing mitochondrial biogenesis and fatty acid oxidation, without affecting T cell receptor-mediated activation. DNA methylation profiling revealed that MEKi-induced TSCM cells exhibited plasticity and loci-specific profiles similar to bona fide TSCM isolated from healthy donors, with intermediate characteristics compared to naive and central memory T cells. Ex vivo, antigenic rechallenge of MEKi-treated CD8+ T cells showed stronger recall responses. This strategy generated T cells with higher efficacy for adoptive cell therapy. Moreover, MEKi treatment of tumor-bearing mice also showed strong immune-mediated antitumor effects. In conclusion, we show that MEKi leads to CD8+ T cell reprogramming into TSCM that acts as a reservoir for effector T cells with potent therapeutic characteristics.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Inmunoterapia Adoptiva , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neoplasias/terapia , Células Madre/citología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Ciclo Celular/efectos de los fármacos , Humanos , Memoria Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/fisiología , Microambiente Tumoral
8.
Int J Mol Sci ; 21(21)2020 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-33171940

RESUMEN

T cell engineering with antigen-specific T cell receptors (TCRs) has allowed the generation of increasingly specific, reliable, and versatile T cell products with near-physiological features. However, a broad applicability of TCR-based therapies in cancer is still limited by the restricted number of TCRs, often also of suboptimal potency, available for clinical use. In addition, targeting of tumor neoantigens with TCR-engineered T cell therapy moves the field towards a highly personalized treatment, as tumor neoantigens derive from somatic mutations and are extremely patient-specific. Therefore, relevant TCRs have to be de novo identified for each patient and within a narrow time window. The naïve repertoire of healthy donors would represent a reliable source due to its huge diverse TCR repertoire, which theoretically entails T cells for any antigen specificity, including tumor neoantigens. As a challenge, antigen-specific naïve T cells are of extremely low frequency and mostly of low functionality, making the identification of highly functional TCRs finding a "needle in a haystack." In this review, we present the technological advancements achieved in high-throughput mapping of patient-specific neoantigens and corresponding cognate TCRs and how these platforms can be used to interrogate the naïve repertoire for a fast and efficient identification of rare but therapeutically valuable TCRs for personalized adoptive T cell therapy.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Inmunoterapia Adoptiva/tendencias , Neoplasias/genética , Medicina de Precisión/métodos , Receptores de Antígenos de Linfocitos T/fisiología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología
9.
Inflamm Res ; 69(9): 813-824, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32591922

RESUMEN

PURPOSE: It paradoxically seems counter-intuitive to consider treatments that activate the immune systems as a method to treat autoimmune diseases and chronic inflammations when these inflammatory conditions are themselves manifested by dysregulated activations of the immune responses. However, according to Newton's Third-Law of fundamental physics which formally states "For every action, there is an equal and opposite reaction", it can be reasonably argued that "For every activated pro-inflammatory response, there is an opposite and intrinsic anti-inflammatory response to follow." Therefore, harnessing these intrinsic self-regulated negative-feedbacks of anti-inflammatory and tolerogenic responses by activating the immune systems represents a novel therapeutic paradigm. METHODS: This review endeavoured to examine and discuss the current clinical and experimental evidences and therapeutic potentials of activating the innate and adaptive immune systems via their classical cell receptors, namely Toll-like receptors (TLRs), T-cell receptors (TCRs), and B cell receptors (BCRs), to modulate and suppress pathogenic inflammations. RESULTS: The evidence presented in this review illustrated the therapeutic potentials and the caveats of  recent approaches and advances in harnessing this unorthodox therapeutic paradigm in the treatments of autoimmune diseases, allergic and chronic inflammations. It highlighted the promising potentials of targeting BCR-activated tolergenic responses as a new approach in this new therapeutic paradigm.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inmunidad Adaptativa , Animales , Enfermedades Autoinmunes/inmunología , Homeostasis , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Inmunoterapia , Inflamación/inmunología , Receptores de Antígenos de Linfocitos B/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Receptor Toll-Like 4/fisiología
10.
Cancer Cell ; 37(6): 818-833.e9, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32516591

RESUMEN

T cells are central to all currently effective cancer immunotherapies, but the characteristics defining therapeutically effective anti-tumor T cells have not been comprehensively elucidated. Here, we delineate four phenotypic qualities of effective anti-tumor T cells: cell expansion, differentiation, oxidative stress, and genomic stress. Using a CRISPR-Cas9-based genetic screen of primary T cells we measured the multi-phenotypic impact of disrupting 25 T cell receptor-driven kinases. We identified p38 kinase as a central regulator of all four phenotypes and uncovered transcriptional and antioxidant pathways regulated by p38 in T cells. Pharmacological inhibition of p38 improved the efficacy of mouse anti-tumor T cells and enhanced the functionalities of human tumor-reactive and gene-engineered T cells, paving the way for clinically relevant interventions.


Asunto(s)
Neoplasias de la Mama/terapia , Sistemas CRISPR-Cas , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Fenotipo , Linfocitos T/trasplante , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Diferenciación Celular , Femenino , Ingeniería Genética , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/genética
11.
Mol Immunol ; 123: 64-73, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32422416

RESUMEN

T cells can help confer protective immunity by eliminating infections and tumors or drive immunopathology by damaging host cells. Both outcomes require a series of steps from the activation of naïve T cells to their clonal expansion, differentiation and migration to tissue sites. In addition to specific recognition of the antigen via the T cell receptor (TCR), multiple accessory signals from costimulatory molecules, cytokines and metabolites also influence each step along the progression of the T cell response. Current efforts to modify effector T cell function in many clinical contexts focus on the latter - which encompass antigen-independent and broad, contextual regulators. Not surprisingly, such approaches are often accompanied by adverse events, as they also affect T cells not relevant to the specific treatment. In contrast, fine tuning T cell responses by precisely targeting antigen-specific TCR signals has the potential to radically alter therapeutic strategies in a focused manner. Development of such approaches, however, requires a better understanding of functioning of the TCR and the biochemical signaling network coupled to it. In this article, we review some of the recent advances which highlight important roles of TCR signals throughout the activation and differentiation of T cells during an immune response. We discuss how, an appreciation of specific signaling modalities and variant ligands that influence the function of the TCR has the potential to influence design principles for the next generation of pharmacologic and cellular therapies, especially in the context of tumor immunotherapies involving adoptive cell transfers.


Asunto(s)
Traslado Adoptivo/métodos , Inmunidad Celular/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Células Presentadoras de Antígenos/inmunología , Diferenciación Celular/inmunología , Humanos , Inmunoterapia/métodos , Activación de Linfocitos , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal/inmunología
12.
J Biosci ; 452020.
Artículo en Inglés | MEDLINE | ID: mdl-32345776

RESUMEN

The hallmarks of the adaptive immune response are specificity and memory. The cellular response is mediated by T cells which express cell surface T cell receptors (TCRs) that recognize peptide antigens in complex with major histocompatibility complex (MHC) molecules on antigen presenting cells (APCs). However, binding of cognate TCRs with MHC-peptide complexes alone (signal 1) does not trigger optimal T cell activation. In addition to signal 1, the binding of positive and negative costimulatory receptors to their ligands modulates T cell activation. This complex signaling network prevents aberrant activation of T cells. CD28 is the main positive costimulatory receptor on nai¨ve T cells; upon activation, CTLA4 is induced but reduces T cell activation. Further studies led to the identification of additional negative costimulatory receptors known as checkpoints, e.g. PD1. This review chronicles the basic studies in T cell costimulation that led to the discovery of checkpoint inhibitors, i.e. antibodies to negative costimulatory receptors (e.g. CTLA4 and PD1) which reduce tumor growth. This discovery has been recognized with the award of the 2018 Nobel prize in Physiology/Medicine. This review highlights the structural and functional roles of costimulatory receptors, the mechanisms by which checkpoint inhibitors work, the challenges encountered and future prospects.


Asunto(s)
Receptores Coestimuladores e Inhibidores de Linfocitos T/fisiología , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Linfocitos T/inmunología , Células Presentadoras de Antígenos/inmunología , Antígenos CD28/química , Antígenos CD28/metabolismo , Antígeno CTLA-4/química , Antígeno CTLA-4/metabolismo , Receptores Coestimuladores e Inhibidores de Linfocitos T/química , Receptores Coestimuladores e Inhibidores de Linfocitos T/genética , Humanos , Activación de Linfocitos , Receptor de Muerte Celular Programada 1/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología
13.
Front Endocrinol (Lausanne) ; 11: 624122, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33597928

RESUMEN

The nuclear orphan receptors NR4A1, NR4A2, and NR4A3 are immediate early genes that are induced by various signals. They act as transcription factors and their activity is not regulated by ligand binding and are thus regulated via their expression levels. Their expression is transiently induced in T cells by triggering of the T cell receptor following antigen recognition during both thymic differentiation and peripheral T cell responses. In this review, we will discuss how NR4A family members impact different aspects of the life of a T cell from thymic differentiation to peripheral response against infections and cancer.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Receptores de Esteroides/fisiología , Receptores de Hormona Tiroidea/fisiología , Linfocitos T/fisiología , Timo/fisiología , Animales , Humanos , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/fisiología , Timo/citología
14.
Front Immunol ; 11: 615317, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33488626

RESUMEN

LAG3 is an important immune checkpoint with relevance in cancer, infectious disease and autoimmunity. However, despite LAG3's role in immune exhaustion and the great potential of LAG3 inhibition as treatment, much remains unknown about its biology, particularly its mechanism of action. This review describes the knowns, unknowns and controversies surrounding LAG3. This includes examination of how LAG3 is regulated transcriptionally and post-translationally by endocytosis and proteolytic cleavage. We also discuss the interactions of LAG3 with its ligands and the purpose thereof. Finally, we review LAG3's mechanism of action, including the roles of LAG3 intracellular motifs and the lack of a role for CD4 competition. Overall, understanding the biology of LAG3 can provide greater insight on LAG3 function, which may broaden the appreciation for LAG3's role in disease and potentially aid in the development of targeted therapies.


Asunto(s)
Antígenos CD/fisiología , Proteínas de Punto de Control Inmunitario/fisiología , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secuencias de Aminoácidos , Antígenos CD/química , Ensayos Clínicos como Asunto , Células Dendríticas/metabolismo , Endocitosis , Regulación de la Expresión Génica , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de Punto de Control Inmunitario/química , Ligandos , Subgrupos Linfocitarios/metabolismo , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neuronas/metabolismo , Biosíntesis de Proteínas , Procesamiento Proteico-Postraduccional , Receptores de Antígenos de Linfocitos T/fisiología , Transcripción Genética , Proteína del Gen 3 de Activación de Linfocitos
15.
J Hematol Oncol ; 12(1): 93, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492199

RESUMEN

Tumor neoantigen is the truly foreign protein and entirely absent from normal human organs/tissues. It could be specifically recognized by neoantigen-specific T cell receptors (TCRs) in the context of major histocompatibility complexes (MHCs) molecules. Emerging evidence has suggested that neoantigens play a critical role in tumor-specific T cell-mediated antitumor immune response and successful cancer immunotherapies. From a theoretical perspective, neoantigen is an ideal immunotherapy target because they are distinguished from germline and could be recognized as non-self by the host immune system. Neoantigen-based therapeutic personalized vaccines and adoptive T cell transfer have shown promising preliminary results. Furthermore, recent studies suggested the significant role of neoantigen in immune escape, immunoediting, and sensitivity to immune checkpoint inhibitors. In this review, we systematically summarize the recent advances of understanding and identification of tumor-specific neoantigens and its role on current cancer immunotherapies. We also discuss the ongoing development of strategies based on neoantigens and its future clinical applications.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Neoplasias/terapia , Vacunas contra el Cáncer/inmunología , Humanos , Inmunoterapia , Neoplasias/prevención & control , Receptores de Antígenos de Linfocitos T/fisiología
16.
Nat Med ; 25(8): 1251-1259, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31359002

RESUMEN

Immunotherapies that block inhibitory checkpoint receptors on T cells have transformed the clinical care of patients with cancer1. However, whether the T cell response to checkpoint blockade relies on reinvigoration of pre-existing tumor-infiltrating lymphocytes or on recruitment of novel T cells remains unclear2-4. Here we performed paired single-cell RNA and T cell receptor sequencing on 79,046 cells from site-matched tumors from patients with basal or squamous cell carcinoma before and after anti-PD-1 therapy. Tracking T cell receptor clones and transcriptional phenotypes revealed coupling of tumor recognition, clonal expansion and T cell dysfunction marked by clonal expansion of CD8+CD39+ T cells, which co-expressed markers of chronic T cell activation and exhaustion. However, the expansion of T cell clones did not derive from pre-existing tumor-infiltrating T lymphocytes; instead, the expanded clones consisted of novel clonotypes that had not previously been observed in the same tumor. Clonal replacement of T cells was preferentially observed in exhausted CD8+ T cells and evident in patients with basal or squamous cell carcinoma. These results demonstrate that pre-existing tumor-specific T cells may have limited reinvigoration capacity, and that the T cell response to checkpoint blockade derives from a distinct repertoire of T cell clones that may have just recently entered the tumor.


Asunto(s)
Carcinoma Basocelular/tratamiento farmacológico , Linfocitos Infiltrantes de Tumor/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/inmunología , Carcinoma Basocelular/inmunología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/inmunología , Humanos , Inmunoterapia , Receptores de Antígenos de Linfocitos T/fisiología , Análisis de Secuencia de ARN , Factor 1 de Transcripción de Linfocitos T/fisiología
17.
J Immunol ; 203(5): 1172-1188, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31350356

RESUMEN

T cells suddenly appeared in jawed fish ∼450 million years ago. Biological studies of fish T cells may provide helpful evidence to understand evolution of adaptive immune systems. To this end, using a Nile tilapia (Oreochromis niloticus) model, we revealed the regulatory mechanism of adaptive immunity mediated by ancestral T cells in jawed fish. Nile tilapia T cells as well as a tightly regulated mammalian/mechanistic target of rapamycin complex 1 (mTORC1) pathway participate in the cellular adaptive immune response during Streptococcus agalactiae infection. Blockade of mTORC1 signaling by rapamycin impairs T cell activation and Ag-induced proliferation in this early vertebrate. More critically, we show that signals from mTORC1 are indispensable for primordial effector T cells to eliminate infection by promoting the expression of proinflammatory cytokines, cytotoxic-related molecules, and proapoptotic genes. Mechanistically, teleost mTORC1 directs effector T cell function by coordinating multiple metabolic programs, including glycolysis, glutaminolysis, and lipogenesis through activating key transcription factors c-Myc, HIF-1α, and sterol regulatory element-binding proteins, and thus links immune signals to metabolic reprogramming in jawed fish. To our knowledge, these results represent the first description of the regulatory mechanism for T cell-mediated adaptive immunity in a fish species. From an evolutionary viewpoint, our study suggests that primordial T cells are armed with sophisticated regulatory strategies like those in modern T cells prior to the divergence of bony fish from the tetrapod lineage. Therefore, our findings fill in an important gap regarding evolution of the adaptive immune system.


Asunto(s)
Cíclidos/inmunología , Activación de Linfocitos , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Evolución Molecular , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/análisis , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal , Serina-Treonina Quinasas TOR/fisiología
18.
PLoS Biol ; 17(6): e3000314, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31194732

RESUMEN

Hypervariable T cell receptors (TCRs) play a key role in adaptive immunity, recognizing a vast diversity of pathogen-derived antigens. Our ability to extract clinically relevant information from large high-throughput sequencing of TCR repertoires (RepSeq) data is limited, because little is known about TCR-disease associations. We present Antigen-specific Lymphocyte Identification by Clustering of Expanded sequences (ALICE), a statistical approach that identifies TCR sequences actively involved in current immune responses from a single RepSeq sample and apply it to repertoires of patients with a variety of disorders - patients with autoimmune disease (ankylosing spondylitis [AS]), under cancer immunotherapy, or subject to an acute infection (live yellow fever [YF] vaccine). We validate the method with independent assays. ALICE requires no longitudinal data collection nor large cohorts, and it is directly applicable to most RepSeq datasets. Its results facilitate the identification of TCR variants associated with diseases and conditions, which can be used for diagnostics and rational vaccine design.


Asunto(s)
Inmunidad Adaptativa/genética , Regiones Determinantes de Complementariedad/genética , Receptores de Antígenos de Linfocitos T/fisiología , Análisis de Secuencia de ADN/métodos , Antígenos , Antígenos Virales , Análisis por Conglomerados , Regiones Determinantes de Complementariedad/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Inmunoterapia , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo
19.
Atherosclerosis ; 285: 71-78, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31048101

RESUMEN

BACKGROUND AND AIMS: CD8+ T-cells have been attributed both atherogenic and atheroprotective properties, but analysis of CD8+ T-cells has mostly been restricted to the circulation and secondary lymphoid organs. The atherosclerotic lesion, however, is a complex microenvironment containing a plethora of inflammatory signals, which may affect CD8+ T-cell activation. Here, we address how this environment affects the functionality of CD8+ T-cells. METHODS AND RESULTS: We compared the cytokine production of CD8+ T-cells derived from spleens and enzymatically digested aortas of apoE-/- mice with advanced atherosclerosis by flow cytometry. Aortic CD8+ T-cells produced decreased amounts of IFN-γ and TNF-α compared to their systemic counterparts. The observed dysfunctional phenotype of the lesion-derived CD8+ T-cells was not associated with classical exhaustion markers, but with increased expression of the ectonucleotidase CD39. Indeed, pharmacological inhibition of CD39 in apoE-/- mice partly restored cytokine production by CD8+ T-cells. Using a bone-marrow transplantation approach, we show that TCR signaling is required to induce CD39 expression on CD8+ T-cells in atherosclerotic lesions. Importantly, analysis of human endarterectomy samples showed a strong microenvironment specific upregulation of CD39 on CD8+ T-cells in the plaques of human patients compared to matched blood samples. CONCLUSIONS: Our results suggest that the continuous TCR signaling in the atherosclerotic environment in the vessel wall induces an immune regulatory CD8+ T-cell phenotype that is associated with decreased cytokine production through increased CD39 expression in both a murine atherosclerotic model and in atherosclerosis patients. This provides a new understanding of immune regulation by CD8+ T-cells in atherosclerosis.


Asunto(s)
Antígenos CD/fisiología , Apirasa/fisiología , Aterosclerosis/inmunología , Linfocitos T CD8-positivos/fisiología , Microambiente Celular/inmunología , Animales , Células Cultivadas , Humanos , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal
20.
Adv Mater ; 31(23): e1807359, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30968468

RESUMEN

T cell therapies require the removal and culture of T cells ex vivo to expand several thousand-fold. However, these cells often lose the phenotype and cytotoxic functionality for mediating effective therapeutic responses. The extracellular matrix (ECM) has been used to preserve and augment cell phenotype; however, it has not been applied to cellular immunotherapies. Here, a hyaluronic acid (HA)-based hydrogel is engineered to present the two stimulatory signals required for T-cell activation-termed an artificial T-cell stimulating matrix (aTM). It is found that biophysical properties of the aTM-stimulatory ligand density, stiffness, and ECM proteins-potentiate T cell signaling and skew phenotype of both murine and human T cells. Importantly, the combination of the ECM environment and mechanically sensitive TCR signaling from the aTM results in a rapid and robust expansion of rare, antigen-specific CD8+ T cells. Adoptive transfer of these tumor-specific cells significantly suppresses tumor growth and improves animal survival compared with T cells stimulated by traditional methods. Beyond immediate immunotherapeutic applications, demonstrating the environment influences the cellular therapeutic product delineates the importance of the ECM and provides a case study of how to engineer ECM-mimetic materials for therapeutic immune stimulation in the future.


Asunto(s)
Células Artificiales/citología , Ingeniería Celular/métodos , Inmunoterapia/métodos , Linfocitos T/citología , Traslado Adoptivo , Animales , Células Artificiales/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Matriz Extracelular/química , Humanos , Ácido Hialurónico/química , Hidrogeles , Ligandos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA