Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 343
Filtrar
1.
Cancer Lett ; 526: 248-258, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34875341

RESUMEN

OTU domain-containing ubiquitin aldehyde-binding proteins 1 (OTUB1) is the most important element of the deubiquitinase OTU superfamily, which has been identified as an essential regulator of diverse physiological processes, such as DNA damage repair and cytokines secretion. Recently, we found that the pro-carcinogenesis role of OTUB1 and the relationship between OTUB1 and immune response have gradually become the research hot-spot. OTUB1 regulates NK/CD8 T cell activation, autoimmune diseases, PD-L1 mediated immune evasion, viral or bacterial infection related immune response and the occurrence and progression of various cancers via deubiquitinating and stabilizing related proteins. This review provides a comprehensive description about the role and regulatory axis of OTUB1. We can explore the balance between immune response and defense via regulating the level of OTUB1, and targeting OTUB1 might restrain the progression of cancers. This review highlights the experimental evidence that OTUB1 is a feasible and potential therapeutic target against various cancers progression and immune diseases or disorder.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Neoplasias/inmunología , Animales , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Humanos , Neoplasias/enzimología
2.
Front Immunol ; 12: 730342, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721394

RESUMEN

Background and Objectives: Inhibition of de novo pyrimidine synthesis in proliferating T and B lymphocytes by teriflunomide, a pharmacological inhibitor of dihydroorotate dehydrogenase (DHODH), has been shown to be an effective therapy to treat patients with MS in placebo-controlled phase 3 trials. Nevertheless, the underlying mechanism contributing to the efficacy of DHODH inhibition has been only partially elucidated. Here, we aimed to determine the impact of teriflunomide on the immune compartment in a longitudinal high-dimensional follow-up of patients with relapse-remitting MS (RRMS) treated with teriflunomide. Methods: High-dimensional spectral flow cytometry was used to analyze the phenotype and the function of innate and adaptive immune system of patients with RRMS before and 12 months after teriflunomide treatment. In addition, we assessed the impact of teriflunomide on the migration of memory CD8 T cells in patients with RRMS, and we defined patient immune metabolic profiles. Results: We found that 12 months of treatment with teriflunomide in patients with RRMS does not affect the B cell or CD4 T cell compartments, including regulatory TREG follicular helper TFH cell and helper TH cell subsets. In contrast, we observed a specific impact of teriflunomide on the CD8 T cell compartment, which was characterized by decreased homeostatic proliferation and reduced production of TNFα and IFNγ. Furthermore, we showed that DHODH inhibition also had a negative impact on the migratory velocity of memory CD8 T cells in patients with RRMS. Finally, we showed that the susceptibility of memory CD8 T cells to DHODH inhibition was not related to impaired metabolism. Discussion: Overall, these findings demonstrate that the clinical efficacy of teriflunomide results partially in the specific susceptibility of memory CD8 T cells to DHODH inhibition in patients with RRMS and strengthens active roles for these T cells in the pathophysiological process of MS.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Crotonatos/uso terapéutico , Dihidroorotato Deshidrogenasa/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Hidroxibutiratos/uso terapéutico , Memoria Inmunológica/efectos de los fármacos , Inmunosupresores/uso terapéutico , Células T de Memoria/efectos de los fármacos , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Nitrilos/uso terapéutico , Toluidinas/uso terapéutico , Adulto , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Crotonatos/efectos adversos , Dihidroorotato Deshidrogenasa/metabolismo , Inhibidores Enzimáticos/efectos adversos , Femenino , Humanos , Hidroxibutiratos/efectos adversos , Inmunosupresores/efectos adversos , Interferón gamma/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Células T de Memoria/enzimología , Células T de Memoria/inmunología , Esclerosis Múltiple Recurrente-Remitente/diagnóstico , Esclerosis Múltiple Recurrente-Remitente/enzimología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Nitrilos/efectos adversos , Fenotipo , Factores de Tiempo , Toluidinas/efectos adversos , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/metabolismo
3.
Cell Rep ; 37(2): 109804, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34644563

RESUMEN

Patients with activated phosphatidylinositol 3-kinase delta (PI3Kδ) syndrome (APDS) present with sinopulmonary infections, lymphadenopathy, and cytomegalvirus (CMV) and/or Epstein-Barr virus (EBV) viremia, yet why patients fail to clear certain chronic viral infections remains incompletely understood. Using patient samples and a mouse model (Pik3cdE1020K/+ mice), we demonstrate that, upon activation, Pik3cdE1020K/+ CD8+ T cells exhibit exaggerated features of effector populations both in vitro and after viral infection that are associated with increased Fas-mediated apoptosis due to sustained FoxO1 phosphorylation and Fasl derepression, enhanced mTORC1 and c-Myc signatures, metabolic perturbations, and an altered chromatin landscape. Conversely, Pik3cdE1020K/+ CD8+ cells fail to sustain expression of proteins critical for central memory, including TCF1. Strikingly, activated Pik3cdE1020K/+ CD8+ cells exhibit altered transcriptional and epigenetic circuits characterized by pronounced interleukin-2 (IL-2)/STAT5 signatures and heightened IL-2 responses that prevent differentiation to memory-like cells in IL-15. Our data position PI3Kδ as integrating multiple signaling nodes that promote CD8+ T cell effector differentiation, providing insight into phenotypes of patients with APDS.


Asunto(s)
Linfocitos T CD8-positivos/enzimología , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Memoria Inmunológica , Enfermedades de Inmunodeficiencia Primaria/enzimología , Transcripción Genética , Virosis/enzimología , Adolescente , Adulto , Animales , Apoptosis , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Estudios de Casos y Controles , Niño , Cromatina/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/inmunología , Modelos Animales de Enfermedad , Activación Enzimática , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/inmunología , Transducción de Señal , Virosis/genética , Virosis/inmunología
4.
Cell Rep ; 37(2): 109796, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34644568

RESUMEN

To gain insight into the signaling determinants of effector-associated DNA methylation programming among CD8 T cells, we explore the role of interleukin (IL)-12 in the imprinting of IFNg expression during CD8 T cell priming. We observe that anti-CD3/CD28-mediated stimulation of human naive CD8 T cells is not sufficient to induce substantial demethylation of the IFNg promoter. However, anti-CD3/CD28 stimulation in the presence of the inflammatory cytokine, IL-12, results in stable demethylation of the IFNg locus that is commensurate with IFNg expression. IL-12-associated demethylation of the IFNg locus is coupled to cell division through TET2-dependent demethylation in an ex vivo human chimeric antigen receptor T cell model system and an in vivo immunologically competent murine system. Collectively, these data illustrate that IL-12 signaling promotes TET2-mediated effector DNA demethylation programming in CD8 T cells and serve as proof of concept that cytokines can guide induction of epigenetically regulated traits for T cell-based immunotherapies.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Interferón gamma/metabolismo , Interleucina-12/farmacología , Coriomeningitis Linfocítica/enzimología , Células T de Memoria/efectos de los fármacos , Animales , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Modelos Animales de Enfermedad , Humanos , Memoria Inmunológica/efectos de los fármacos , Interferón gamma/genética , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Células T de Memoria/enzimología , Células T de Memoria/inmunología , Células T de Memoria/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Prueba de Estudio Conceptual , Transducción de Señal
5.
Ann Vasc Surg ; 77: 255-262, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34411666

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is a chronic inflammatory disease characterized by localized progressive dilatation. Currently, paeonol has been shown to possess anti-inflammatory and protective cardiovascular properties. Our study aimed to investigate the potential influences of paeonol on AAA progression. METHODS: Experimental AAAs were created in C57BL/6J mice by intra-aortic infusion of porcine pancreatic elastase, and then intragastrically administered paeonol (20 mg/kg/day) for 14 days. The effects of paeonol on experimental AAA were measured by ultrasound imaging, histopathology, and western blot analyses. RESULTS: Paeonol treatment limited the enlargement of the aneurysmal diameter and alleviated the depletion of elastic fibers and vascular smooth muscle cells (VSMCs). Furthermore, the infiltration of CD68+ macrophages and CD8+ lymphocytes was obviously attenuated after paeonol administration, along with mural neoangiogenesis. Western blot results showed that paeonol inhibited the expression of matrix metalloproteinase (MMP) and the NF-κB pathway activation. CONCLUSIONS: Paeonol might prevent experimental AAA progression by inhibiting the NF-κB pathway, which suggests that it is a potential drug for AAA.


Asunto(s)
Acetofenonas/farmacología , Antiinflamatorios/farmacología , Aorta Abdominal/efectos de los fármacos , Aneurisma de la Aorta Abdominal/prevención & control , FN-kappa B/metabolismo , Animales , Aorta Abdominal/inmunología , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Neovascularización Patológica , Transducción de Señal
6.
Nat Commun ; 12(1): 5147, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34446716

RESUMEN

PTEN is frequently mutated in human cancers and PTEN mutants promote tumor progression and metastasis. PTEN mutations have been implicated in immune regulation, however, the underlying mechanism is largely unknown. Here, we report that PTENα, the isoform of PTEN, remains active in cancer bearing stop-gained PTEN mutations. Through counteraction of CD8+ T cell-mediated cytotoxicity, PTENα leads to T cell dysfunction and accelerates immune-resistant cancer progression. Clinical analysis further uncovers that PTENα-active mutations suppress host immune responses and result in poor prognosis in cancer as relative to PTENα-inactive mutations. Furthermore, germline deletion of Ptenα in mice increases cell susceptibility to immune attack through augmenting stress granule formation and limiting synthesis of peroxidases, leading to massive oxidative cell death and severe inflammatory damage. We propose that PTENα protects tumor from T cell killing and thus PTENα is a potential target in antitumor immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias/enzimología , Neoplasias/inmunología , Fosfohidrolasa PTEN/inmunología , Animales , Linfocitos T CD8-positivos/enzimología , Femenino , Humanos , Terapia de Inmunosupresión , Isoenzimas/genética , Isoenzimas/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Neoplasias/genética , Fosfohidrolasa PTEN/genética , Escape del Tumor
7.
Int Immunopharmacol ; 97: 107685, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33951560

RESUMEN

BACKGROUND: The 2019 Coronavirus (COVID-19) pandemic poses a huge threat internationally; however, the role of the host immune system in the pathogenesis of COVID-19 is not well understood. METHODS: Cytokine and chemokine levels and characterisation of immune cell subsets from 20 COVID-19 cases after hospital admission (17 critically ill and 3 severe patients) and 16 convalescent patients were determined using a multiplex immunoassay and flow cytometry, respectively. RESULTS: IP-10, MCP-1, MIG, IL-6, and IL-10 levels were significantly higher in acute severe/critically ill patients with COVID-19, whereas were normal in patients who had reached convalescence. CD8 T cells in severe and critically ill COVID-19 patients expressed high levels of cytotoxic granules (granzyme B and perforin)and was hyperactivated as evidenced by the high proportions of CD38. Furthermore, the cytotoxic potential of natural killer (NK) cells, and the frequencies of myeloid dendritic cells and plasmacytoid dendritic cells was reduced in patients with severe and critical COVID-19; however, these dysregulations were found to be restored in convalescent phases. CONCLUSION: Thus, elicitation of the hyperactive cytokine-mediated inflammatory response, dysregulation of CD8 T and NK cells, and deficiency of host myeloid and plasmacytoid DCs, may contribute to COVID-19 pathogenesis and provide insights into potential therapeutic targets and strategies.


Asunto(s)
COVID-19/sangre , COVID-19/inmunología , Convalecencia , Inflamación/etiología , ADP-Ribosil Ciclasa 1/sangre , Enfermedad Aguda , Adulto , Anciano , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Quimiocina CCL2/sangre , Quimiocina CXCL10/sangre , Quimiocina CXCL9/sangre , Enfermedad Crítica , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/inmunología , Células Dendríticas/inmunología , Femenino , Granzimas/metabolismo , Humanos , Interleucina-10/sangre , Interleucina-6/sangre , Células Asesinas Naturales/enzimología , Células Asesinas Naturales/inmunología , Masculino , Glicoproteínas de Membrana/sangre , Persona de Mediana Edad , Perforina/metabolismo
8.
Aging Cell ; 20(5): e13344, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33939265

RESUMEN

Aging leads to a progressive functional decline of the immune system, rendering the elderly increasingly susceptible to disease and infection. The degree to which immune cell senescence contributes to this decline remains unclear, however, since markers that label immune cells with classical features of cellular senescence accurately and comprehensively have not been identified. Using a second-generation fluorogenic substrate for ß-galactosidase and multi-parameter flow cytometry, we demonstrate here that peripheral blood mononuclear cells (PBMCs) isolated from healthy humans increasingly display cells with high senescence-associated ß-galactosidase (SA-ßGal) activity with advancing donor age. The greatest age-associated increases were observed in CD8+ T-cell populations, in which the fraction of cells with high SA-ßGal activity reached average levels of 64% in donors in their 60s. CD8+ T cells with high SA-ßGal activity, but not those with low SA-ßGal activity, were found to exhibit features of telomere dysfunction-induced senescence and p16-mediated senescence, were impaired in their ability to proliferate, developed in various T-cell differentiation states, and had a gene expression signature consistent with the senescence state previously observed in human fibroblasts. Based on these results, we propose that senescent CD8+ T cells with classical features of cellular senescence accumulate to levels that are significantly higher than previously reported and additionally provide a simple yet robust method for the isolation and characterization of senescent CD8+ T cells with predictive potential for biological age.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD8-positivos/citología , Senescencia Celular/inmunología , beta-Galactosidasa/metabolismo , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Separación Celular , Células Cultivadas , Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Citometría de Flujo , Expresión Génica , Humanos , Telómero
9.
J Immunother Cancer ; 9(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33883257

RESUMEN

BACKGROUND: In the tumor microenvironment, tumor cells are able to suppress antitumor immunity by competing for essential nutrients, including amino acids. However, whether amino acid depletion modulates the activity of CD8+ tumor-infiltrating lymphocytes (TILs) is unclear. METHOD: In this study, we evaluated the roles of amino acids and the Rag complex in regulating mammalian target of rapamycin complex 1 (mTORC1) signaling in CD8+ TILs. RESULTS: We discovered that the Rag complex, particularly RagD, was crucial for CD8+ T-cell antitumor immunity. RagD expression was positively correlated with the antitumor response of CD8+ TILs in both murine syngeneic tumor xenografts and clinical human colon cancer samples. On RagD deficiency, CD8+ T cells were rendered more dysfunctional, as demonstrated by attenuation of mTORC1 signaling and reductions in proliferation and cytokine secretion. Amino acids maintained RagD-mediated mTORC1 translocation to the lysosome, thereby achieving maximal mTORC1 activity in CD8+ T cells. Moreover, the limited T-cell access to leucine (LEU), overshadowed by tumor cell amino acid consumption, led to impaired RagD-dependent mTORC1 activity. Finally, combined with antiprogrammed cell death protein 1 antibody, LEU supplementation improved T-cell immunity in MC38 tumor-bearing mice in vivo. CONCLUSION: Our results revealed that robust signaling of amino acids by RagD and downstream mTORC1 signaling were crucial for T-cell receptor-initiated antitumor immunity. The characterization the role of RagD and LEU in nutrient mTORC1 signaling in TILs might suggest potential therapeutic strategies based on the manipulation of RagD and its upstream pathway.


Asunto(s)
Linfocitos T CD8-positivos/enzimología , Leucina/metabolismo , Linfocitos Infiltrantes de Tumor/enzimología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Melanoma Experimental/enzimología , Proteínas de Unión al GTP Monoméricas/metabolismo , Neoplasias Cutáneas/enzimología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Activación Enzimática , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Escape del Tumor , Microambiente Tumoral
10.
Nat Immunol ; 22(4): 460-470, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33767425

RESUMEN

Targeting the p53-MDM2 pathway to reactivate tumor p53 is a chemotherapeutic approach. However, the involvement of this pathway in CD8+ T cell-mediated antitumor immunity is unknown. Here, we report that mice with MDM2 deficiency in T cells exhibit accelerated tumor progression and a decrease in tumor-infiltrating CD8+ T cell survival and function. Mechanistically, MDM2 competes with c-Cbl for STAT5 binding, reduces c-Cbl-mediated STAT5 degradation and enhances STAT5 stability in tumor-infiltrating CD8+ T cells. Targeting the p53-MDM2 interaction with a pharmacological agent, APG-115, augmented MDM2 in T cells, thereby stabilizing STAT5, boosting T cell immunity and synergizing with cancer immunotherapy. Unexpectedly, these effects of APG-115 were dependent on p53 and MDM2 in T cells. Clinically, MDM2 abundance correlated with T cell function and interferon-γ signature in patients with cancer. Thus, the p53-MDM2 pathway controls T cell immunity, and targeting this pathway may treat patients with cancer regardless of tumor p53 status.


Asunto(s)
Linfocitos T CD8-positivos/enzimología , Linfocitos Infiltrantes de Tumor/enzimología , Neoplasias/enzimología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Antineoplásicos/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Terapia Combinada , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/trasplante , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Estabilidad Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/genética , Factor de Transcripción STAT5/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Nat Commun ; 12(1): 741, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531485

RESUMEN

The heterogeneous nature of tumour microenvironment (TME) underlying diverse treatment responses remains unclear in nasopharyngeal carcinoma (NPC). Here, we profile 176,447 cells from 10 NPC tumour-blood pairs, using single-cell transcriptome coupled with T cell receptor sequencing. Our analyses reveal 53 cell subtypes, including tumour-infiltrating CD8+ T, regulatory T (Treg), and dendritic cells (DCs), as well as malignant cells with different Epstein-Barr virus infection status. Trajectory analyses reveal exhausted CD8+ T and immune-suppressive TNFRSF4+ Treg cells in tumours might derive from peripheral CX3CR1+CD8+ T and naïve Treg cells, respectively. Moreover, we identify immune-regulatory and tolerogenic LAMP3+ DCs. Noteworthily, we observe intensive inter-cell interactions among LAMP3+ DCs, Treg, exhausted CD8+ T, and malignant cells, suggesting potential cross-talks to foster an immune-suppressive niche for the TME. Collectively, our study uncovers the heterogeneity and interacting molecules of the TME in NPC at single-cell resolution, which provide insights into the mechanisms underlying NPC progression and the development of precise therapies for NPC.


Asunto(s)
Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/metabolismo , Carcinoma Nasofaríngeo/inmunología , Carcinoma Nasofaríngeo/metabolismo , Microambiente Tumoral/fisiología , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Microambiente Tumoral/inmunología
12.
Nat Immunol ; 22(2): 193-204, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33398181

RESUMEN

Metabolic reprograming toward aerobic glycolysis is a pivotal mechanism shaping immune responses. Here we show that deficiency in NF-κB-inducing kinase (NIK) impairs glycolysis induction, rendering CD8+ effector T cells hypofunctional in the tumor microenvironment. Conversely, ectopic expression of NIK promotes CD8+ T cell metabolism and effector function, thereby profoundly enhancing antitumor immunity and improving the efficacy of T cell adoptive therapy. NIK regulates T cell metabolism via a NF-κB-independent mechanism that involves stabilization of hexokinase 2 (HK2), a rate-limiting enzyme of the glycolytic pathway. NIK prevents autophagic degradation of HK2 through controlling cellular reactive oxygen species levels, which in turn involves modulation of glucose-6-phosphate dehydrogenase (G6PD), an enzyme that mediates production of the antioxidant NADPH. We show that the G6PD-NADPH redox system is important for HK2 stability and metabolism in activated T cells. These findings establish NIK as a pivotal regulator of T cell metabolism and highlight a post-translational mechanism of metabolic regulation.


Asunto(s)
Linfocitos T CD8-positivos/enzimología , Neoplasias del Colon/enzimología , Metabolismo Energético , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/enzimología , Melanoma Experimental/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Citotoxicidad Inmunológica , Estabilidad de Enzimas , Femenino , Glucosafosfato Deshidrogenasa/metabolismo , Glucólisis , Hexoquinasa/genética , Hexoquinasa/metabolismo , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/trasplante , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Ratones Noqueados , NADP/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Microambiente Tumoral , Quinasa de Factor Nuclear kappa B
13.
Science ; 371(6527): 405-410, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33479154

RESUMEN

Infection triggers expansion and effector differentiation of T cells specific for microbial antigens in association with metabolic reprograming. We found that the glycolytic enzyme lactate dehydrogenase A (LDHA) is induced in CD8+ T effector cells through phosphoinositide 3-kinase (PI3K) signaling. In turn, ablation of LDHA inhibits PI3K-dependent phosphorylation of Akt and its transcription factor target Foxo1, causing defective antimicrobial immunity. LDHA deficiency cripples cellular redox control and diminishes adenosine triphosphate (ATP) production in effector T cells, resulting in attenuated PI3K signaling. Thus, nutrient metabolism and growth factor signaling are highly integrated processes, with glycolytic ATP serving as a rheostat to gauge PI3K-Akt-Foxo1 signaling in the control of T cell immunity. Such a bioenergetic mechanism for the regulation of signaling may explain the Warburg effect.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Glucólisis , Lactato Deshidrogenasa 5/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Animales , Linfocitos T CD8-positivos/enzimología , Proteína Forkhead Box O1/metabolismo , Humanos , Lactato Deshidrogenasa 5/genética , Listeria monocytogenes , Listeriosis/enzimología , Listeriosis/inmunología , Ratones , Ratones Mutantes , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Efecto Warburg en Oncología
14.
Respir Res ; 21(1): 188, 2020 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-32677970

RESUMEN

BACKGROUND: A disintegrin and metalloproteinase domain-15 (ADAM15) is expressed by activated leukocytes, and fibroblasts in vitro. Whether ADAM15 expression is increased in the lungs of COPD patients is not known. METHODS: ADAM15 gene expression and/or protein levels were measured in whole lung and bronchoalveolar lavage (BAL) macrophage samples obtained from COPD patients, smokers, and non-smokers. Soluble ADAM15 protein levels were measured in BAL fluid (BALF) and plasma samples from COPD patients and controls. Cells expressing ADAM15 in the lungs were identified using immunostaining. Staining for ADAM15 in different cells in the lungs was related to forced expiratory volume in 1 s (FEV1), ratio of FEV1 to forced vital capacity (FEV1/FVC), and pack-years of smoking history. RESULTS: ADAM15 gene expression and/or protein levels were increased in alveolar macrophages and whole lung samples from COPD patients versus smokers and non-smokers. Soluble ADAM15 protein levels were similar in BALF and plasma samples from COPD patients and controls. ADAM15 immunostaining was increased in macrophages, CD8+ T cells, epithelial cells, and airway α-smooth muscle (α-SMA)-positive cells in the lungs of COPD patients. ADAM15 immunostaining in macrophages, CD8+ T cells and bronchial (but not alveolar) epithelial cells was related inversely to FEV1 and FEV1/FVC, but not to pack-years of smoking history. ADAM15 staining levels in airway α-SMA-positive cells was directly related to FEV1/FVC. Over-expressing ADAM15 in THP-1 cells reduced their release of matrix metalloproteinases and CCL2. CONCLUSIONS: These results link increased ADAM15 expression especially in lung leukocytes and bronchial epithelial cells to the pathogenesis of COPD.


Asunto(s)
Proteínas ADAM/metabolismo , Bronquios/enzimología , Linfocitos T CD8-positivos/enzimología , Células Epiteliales/enzimología , Macrófagos Alveolares/enzimología , Proteínas de la Membrana/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Beijing , Biomarcadores/metabolismo , Boston , Bronquios/fisiopatología , Estudios de Casos y Controles , Inglaterra , Femenino , Volumen Espiratorio Forzado , Humanos , Masculino , Persona de Mediana Edad , No Fumadores , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Fumadores , Células THP-1 , Regulación hacia Arriba , Capacidad Vital , Adulto Joven
15.
Front Immunol ; 11: 1638, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32695123

RESUMEN

The SARS-CoV2 (COVID-19) pandemic and uncertainties in developing a vaccine have created an urgent need for new therapeutic approaches. A key question is whether it is possible to make rational predictions of new therapies based on the presently available scientific and medical information. In this regard, I have noticed an omission in the present analysis in the literature related to the exploitation of glycogen synthase kinase 3 (GSK-3) as a therapeutic approach. This is based on two key observations, that GSK-3 inhibitors can simultaneously block SARs viral replication, while boosting CD8+ adaptive T-cell and innate natural killer (NK) responses. Firstly, it is already clear that GSK-3 phosphorylation of SARs CoV1 N protein on key serine residues is needed for viral replication such that small molecule inhibitors (SMIs) of GSK-3 can inhibit viral replication. In comparing protein sequences, I show here that the key sites in the N protein of SARs CoV1 N for replication are conserved in SARs CoV2. This strongly suggests that GSK-3 SMIs will also inhibit SARs Cov2 replication. Secondly, we and others have previously documented that GSK-3 SMIs markedly enhance CD8+ cytolytic T-cell (CTL) and NK cell anti-viral effector functions leading to a reduction in both acute and chronic viral infections in mice. My hypothesis is that the repurposing of low-cost inhibitors of GSK-3 such as lithium will limit SARS-CoV2 infections by both reducing viral replication and potentiating the immune response against the virus. To date, there has been no mention of this dual connection between GSK-3 and SARs CoV2 in the literature. To my knowledge, no other drugs exist with the potential to simultaneously target both viral replication and immune response against SARs CoV2.


Asunto(s)
Betacoronavirus/fisiología , Linfocitos T CD8-positivos , Inhibidores Enzimáticos/uso terapéutico , Glucógeno Sintasa Quinasa 3 , Inmunidad Celular/efectos de los fármacos , Células Asesinas Naturales , Replicación Viral/efectos de los fármacos , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/enzimología , Infecciones por Coronavirus/inmunología , Humanos , Células Asesinas Naturales/enzimología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/enzimología , Neumonía Viral/inmunología , SARS-CoV-2
16.
Ther Adv Respir Dis ; 14: 1753466620905280, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32270742

RESUMEN

BACKGROUND: The class III NAD-dependent histone deacetylase (HDAC) sirtuin 1 (SIRT1) is an important regulator of senescence, aging, and inflammation. SIRT1de-acetylates chromatin histones, thereby silencing inflammatory gene transcription. We have reported increased steroid-resistant senescent pro-inflammatory CD28nullCD8+ T cells in patients with chronic obstructive pulmonary disease (COPD). We hypothesized that SIRT1 is reduced in these cells in COPD, and that treatment with SIRT1 activators (resveratrol, curcumin) and agents preventing NAD depletion (theophylline) would upregulate SIRT1 and reduce pro-inflammatory cytokine expression in these steroid-resistant cells. METHODS: Blood was collected from n = 10 COPD and n = 10 aged-matched controls. Expression of CD28, SIRT1, and pro-inflammatory cytokines was determined in CD8+ and CD8- T and natural killer T (NKT)-like cells cultured in the presence of ±1 µM prednisolone, ±5 mg/L theophylline, ±1 µM curcumin, ±25 µM resveratrol, using flow cytometry and immunofluorescence. RESULTS: There was an increase in the percentage of CD28nullCD8+ T and NKT-like cells in COPD patients compared with controls. Decreased SIRT1 expression was identified in CD28nullCD8+T and NKT-like cells compared with CD28+ counterparts from both patients and controls (e.g. CD28null 11 ± 3% versus CD28+ 57 ± 9%). Loss of SIRT1 was associated with increased production of IFNγ and TNFα, steroid resistance, and disease severity. SIRT1 expression was upregulated in the presence of all drugs and was associated with a decrease in steroid resistance and IFNγ and TNFα production by CD28nullCD8+T and NKT-like cells. The presence of the SIRT1 inhibitor, EX-527 negated [by 92 ± 12% (median ± SEM)] the effect of the SIRT1 activator SRT720 on the percentage of CD8+ T cells producing IFNγ and TNFα. CONCLUSIONS: Steroid resistance in pro-inflammatory CD28nullCD8+ T and NKT-like cells is associated with decreased SIRT1 expression. Treatment with prednisolone, in combination with theophylline, curcumin or resveratrol increases SIRT1 expression, restores steroid sensitivity, and inhibits pro-inflammatory cytokine production from these cells and may reduce systemic inflammation in COPD. The reviews of this paper are available via the supplemental material section.


Asunto(s)
Linfocitos T CD8-positivos/enzimología , Citocinas/metabolismo , Inmunosenescencia , Mediadores de Inflamación/metabolismo , Células T Asesinas Naturales/enzimología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Sirtuina 1/metabolismo , Anciano , Antiinflamatorios/farmacología , Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Estudios de Casos y Controles , Células Cultivadas , Curcumina/farmacología , Resistencia a Medicamentos , Femenino , Glucocorticoides/farmacología , Humanos , Inmunosenescencia/efectos de los fármacos , Masculino , Persona de Mediana Edad , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/inmunología , Fenotipo , Prednisolona/farmacología , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Resveratrol/farmacología , Teofilina/farmacología
17.
Mol Oncol ; 14(5): 1028-1044, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32157792

RESUMEN

B-cell non-Hodgkin's lymphoma (NHL) is a class of heterogeneous diseases with variable clinical outcomes. Immunosuppression is particularly common in the subtypes of lymphoma with poor prognosis, but the underlying mechanism remains unclear. Using a RT-PCR array analysis, we have identified that glycosyltransferase 1 domain-containing 1 (GLT1D1), an enzyme that transfers glycosyl groups to proteins, is highly upregulated in the incurable subtype of B-cell NHL and in early relapse diffuse large B-cell lymphoma. Analysis of clinical specimens revealed that GLT1D1 expression was positively correlated with the level of glycosylated programmed cell death-ligand 1 (PD-L1) in B-cell NHL and that high GLT1D1 expression was associated with poor prognosis. Mechanistically, we showed that GLT1D1 transferred N-linked glycans to PD-L1, thus promoting the immunosuppressive function of glycosylated PD-L1. Downregulation of GLT1D1 resulted in a decrease of glycosylated PD-L1 and enhanced cytotoxic T-cell function against lymphoma cells. In vivo, overexpression of GLT1D1 promoted tumor growth by facilitating tumor immune escape through increased levels of PD-L1. Our work has identified GLT1D1 as a predictive biomarker for B-cell NHL. It has also shown that this enzyme enhances PD-L1 stabilization via N-glycosylation, thus promoting immunosuppression and tumor growth. As such, GLT1D1 might be a novel therapeutic target for the treatment of B-NHL.


Asunto(s)
Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Carcinogénesis/genética , Terapia de Inmunosupresión , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Linfocitos T CD8-positivos/enzimología , Sistemas CRISPR-Cas , Carcinogénesis/inmunología , Línea Celular Tumoral , Supervivencia Celular/genética , Técnicas de Cocultivo , Bases de Datos Genéticas , Femenino , Técnicas de Silenciamiento del Gen , Glicosilación , Humanos , Linfoma de Células B/genética , Linfoma de Células B/mortalidad , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/mortalidad , Ratones , Ratones Endogámicos C57BL , Polisacáridos/metabolismo , Pronóstico , ARN Interferente Pequeño , Linfocitos T/inmunología , Tunicamicina/farmacología
18.
JCI Insight ; 4(24)2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31751318

RESUMEN

As sufficient extracellular arginine is crucial for T cell function, depletion of extracellular arginine by elevated arginase 1 (Arg1) activity has emerged as a hallmark immunosuppressive mechanism. However, the potential cell-autonomous roles of arginases in T cells have remained unexplored. Here, we show that the arginase isoform expressed by T cells, the mitochondrial Arg2, is a cell-intrinsic regulator of CD8+ T cell activity. Both germline Arg2 deletion and adoptive transfer of Arg2-/- CD8+ T cells significantly reduced tumor growth in preclinical cancer models by enhancing CD8+ T cell activation, effector function, and persistence. Transcriptomic, proteomic, and high-dimensional flow cytometry characterization revealed a CD8+ T cell-intrinsic role of Arg2 in modulating T cell activation, antitumor cytoxicity, and memory formation, independently of extracellular arginine availability. Furthermore, specific deletion of Arg2 in CD8+ T cells strongly synergized with PD-1 blockade for the control of tumor growth and animal survival. These observations, coupled with the finding that pharmacologic arginase inhibition accelerates activation of ex vivo human T cells, unveil Arg2 as a potentially new therapeutic target for T cell-based cancer immunotherapies.


Asunto(s)
Arginasa/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Colorrectales/inmunología , Melanoma Experimental/inmunología , Mitocondrias/enzimología , Animales , Arginasa/genética , Arginina/metabolismo , Linfocitos T CD8-positivos/enzimología , Línea Celular Tumoral , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Citotoxicidad Inmunológica , Femenino , Eliminación de Gen , Humanos , Tolerancia Inmunológica/inmunología , Memoria Inmunológica/inmunología , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Melanoma Experimental/enzimología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Trasplante de Neoplasias , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores
19.
Nutrients ; 11(4)2019 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-30959753

RESUMEN

Telomerase in T lymphocytes is dynamic and limited evidence from epidemiological studies indicates that the enzyme can be modulated in peripheral lymphocytes by dietary and lifestyle factors. The differential effect of dietary intervention on T cell subsets has not been investigated so far. Brassica vegetables are known for their multiple beneficial effects on human health, and here, the effect of a five-day short-term intervention with raw or cooked leaves of Brassica carinata on telomerase activity in CD4+ and CD8+ T cells from 22 healthy volunteers was investigated in a randomized single-blind, controlled crossover study. Blood samples were collected before and after intervention, and CD4+/CD8+ T lymphocytes were isolated. Telomerase activity was quantified using the TRAP-ELISA assay. Intervention with both preparations led to a marginal increase in telomerase activity of CD4+ cells compared to the baseline level. In CD8+ cells, a significant increase in telomerase activity (25%, p < 0.05) was seen after intervention with the cooked material. An increase in telomerase activity in CD8+ cells of healthy volunteers could be regarded as beneficial in terms of helping with the cell-mediated immune response. Whether a Brassica intervention has long-term effects on telomere extension in specific T cell subsets needs to be determined.


Asunto(s)
Brassica/química , Linfocitos T CD8-positivos/enzimología , Dieta , Telomerasa/metabolismo , Linfocitos T CD4-Positivos/enzimología , Culinaria , Estudios Cruzados , Análisis de los Alimentos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Fitoquímicos/química
20.
Immunity ; 50(5): 1249-1261.e5, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30952606

RESUMEN

Regulated activation of the cytokine TGF-ß by integrins αvß6 and αvß8 expressed on keratinocytes is required for residence of epidermal-resident memory T cells, but whether skin-derived signals also affect recirculating memory cells in the skin remains unclear. Here, we show that after resolution of skin vaccinia virus (VV) infection, antigen-specific circulating memory CD8+ T cells migrated into skin. In mice lacking αvß6 and αvß8 integrins (Itgb6-/-Itgb8fl/fl-K14-cre), the absence of epidermal-activated TGF-ß resulted in a gradual loss of E- or P-selectin-binding central and peripheral memory populations, which were rescued when skin entry was inhibited. Skin recirculating memory cells were required for optimal host defense against skin VV infection. These data demonstrate that skin migration can persist after resolution of local skin infection and that the cytokine environment within this nonlymphoid tissue shapes the differentiation state and persistence of the central and peripheral memory-T-cell pool.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Integrinas/metabolismo , Queratinocitos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Virus Vaccinia/inmunología , Animales , Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/enzimología , Diferenciación Celular/inmunología , Citocinas/inmunología , Activación Enzimática , Femenino , Integrinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Piel/citología , Piel/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...