Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Cell Chem Biol ; 31(5): 862-883, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38428418

RESUMEN

The immune system shapes tumor development and progression. Although immunotherapy has transformed cancer treatment, its overall efficacy remains limited, underscoring the need to uncover mechanisms to improve therapeutic effects. Metabolism-associated processes, including intracellular metabolic reprogramming and intercellular metabolic crosstalk, are emerging as instructive signals for anti-tumor immunity. Here, we first summarize the roles of intracellular metabolic pathways in controlling immune cell function in the tumor microenvironment. How intercellular metabolic communication regulates anti-tumor immunity, and the impact of metabolites or nutrients on signaling events, are also discussed. We then describe how targeting metabolic pathways in tumor cells or intratumoral immune cells or via nutrient-based interventions may boost cancer immunotherapies. Finally, we conclude with discussions on profiling and functional perturbation methods of metabolic activity in intratumoral immune cells, and perspectives on future directions. Uncovering the mechanisms for metabolic rewiring and communication in the tumor microenvironment may enable development of novel cancer immunotherapies.


Asunto(s)
Inmunoterapia , Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Redes y Vías Metabólicas , Comunicación Celular/inmunología , Transducción de Señal
2.
J Clin Invest ; 133(24)2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-38099491

RESUMEN

Foxp3-expressing Tregs employ multiple suppressive mechanisms to curtail conventional T cell (Tconv) responses and establish tissue homeostasis. How Foxp3 coordinates Treg contact-dependent suppressive function is not fully resolved. In this issue of the JCI, Wang and colleagues revealed that Foxp3-mediated inhibition of ryanodine receptor 2 (RyR2) led to strong Treg-DC interactions and enhanced immunosuppression. RyR2 depletion in Tconvs phenocopied this effect and equipped Tconvs with Treg-like suppressive function in multiple inflammatory or autoimmune contexts. This study provides molecular and therapeutic insights underlying how cell-cell contact limits immune reactivity.


Asunto(s)
Canal Liberador de Calcio Receptor de Rianodina , Linfocitos T Reguladores , Ratones , Animales , Ratones Endogámicos C57BL , Terapia de Inmunosupresión , Factores de Transcripción Forkhead
3.
Nature ; 624(7990): 154-163, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37968405

RESUMEN

CD8+ cytotoxic T cells (CTLs) orchestrate antitumour immunity and exhibit inherent heterogeneity1,2, with precursor exhausted T (Tpex) cells but not terminally exhausted T (Tex) cells capable of responding to existing immunotherapies3-7. The gene regulatory network that underlies CTL differentiation and whether Tex cell responses can be functionally reinvigorated are incompletely understood. Here we systematically mapped causal gene regulatory networks using single-cell CRISPR screens in vivo and discovered checkpoints for CTL differentiation. First, the exit from quiescence of Tpex cells initiated successive differentiation into intermediate Tex cells. This process is differentially regulated by IKAROS and ETS1, the deficiencies of which dampened and increased mTORC1-associated metabolic activities, respectively. IKAROS-deficient cells accumulated as a metabolically quiescent Tpex cell population with limited differentiation potential following immune checkpoint blockade (ICB). Conversely, targeting ETS1 improved antitumour immunity and ICB efficacy by boosting differentiation of Tpex to intermediate Tex cells and metabolic rewiring. Mechanistically, TCF-1 and BATF are the targets for IKAROS and ETS1, respectively. Second, the RBPJ-IRF1 axis promoted differentiation of intermediate Tex to terminal Tex cells. Accordingly, targeting RBPJ enhanced functional and epigenetic reprogramming of Tex cells towards the proliferative state and improved therapeutic effects and ICB efficacy. Collectively, our study reveals that promoting the exit from quiescence of Tpex cells and enriching the proliferative Tex cell state act as key modalities for antitumour effects and provides a systemic framework to integrate cell fate regulomes and reprogrammable functional determinants for cancer immunity.


Asunto(s)
Diferenciación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Edición Génica , Mutagénesis , Neoplasias , Análisis de la Célula Individual , Linfocitos T Citotóxicos , Humanos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Inhibidores de Puntos de Control Inmunológico/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias/genética , Neoplasias/inmunología , Análisis de la Célula Individual/métodos , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
4.
Nat Immunol ; 24(10): 1735-1747, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37679549

RESUMEN

Neurodegenerative diseases, including Alzheimer's disease (AD), are characterized by innate immune-mediated inflammation, but functional and mechanistic effects of the adaptive immune system remain unclear. Here we identify brain-resident CD8+ T cells that coexpress CXCR6 and PD-1 and are in proximity to plaque-associated microglia in human and mouse AD brains. We also establish that CD8+ T cells restrict AD pathologies, including ß-amyloid deposition and cognitive decline. Ligand-receptor interaction analysis identifies CXCL16-CXCR6 intercellular communication between microglia and CD8+ T cells. Further, Cxcr6 deficiency impairs accumulation, tissue residency programming and clonal expansion of brain PD-1+CD8+ T cells. Ablation of Cxcr6 or CD8+ T cells ultimately increases proinflammatory cytokine production from microglia, with CXCR6 orchestrating brain CD8+ T cell-microglia colocalization. Collectively, our study reveals protective roles for brain CD8+ T cells and CXCR6 in mouse AD pathogenesis and highlights that microenvironment-specific, intercellular communication orchestrates tissue homeostasis and protection from neuroinflammation.

5.
Nature ; 620(7972): 200-208, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37407815

RESUMEN

Cancer cells evade T cell-mediated killing through tumour-immune interactions whose mechanisms are not well understood1,2. Dendritic cells (DCs), especially type-1 conventional DCs (cDC1s), mediate T cell priming and therapeutic efficacy against tumours3. DC functions are orchestrated by pattern recognition receptors3-5, although other signals involved remain incompletely defined. Nutrients are emerging mediators of adaptive immunity6-8, but whether nutrients affect DC function or communication between innate and adaptive immune cells is largely unresolved. Here we establish glutamine as an intercellular metabolic checkpoint that dictates tumour-cDC1 crosstalk and licenses cDC1 function in activating cytotoxic T cells. Intratumoral glutamine supplementation inhibits tumour growth by augmenting cDC1-mediated CD8+ T cell immunity, and overcomes therapeutic resistance to checkpoint blockade and T cell-mediated immunotherapies. Mechanistically, tumour cells and cDC1s compete for glutamine uptake via the transporter SLC38A2 to tune anti-tumour immunity. Nutrient screening and integrative analyses show that glutamine is the dominant amino acid in promoting cDC1 function. Further, glutamine signalling via FLCN impinges on TFEB function. Loss of FLCN in DCs selectively impairs cDC1 function in vivo in a TFEB-dependent manner and phenocopies SLC38A2 deficiency by eliminating the anti-tumour therapeutic effect of glutamine supplementation. Our findings establish glutamine-mediated intercellular metabolic crosstalk between tumour cells and cDC1s that underpins tumour immune evasion, and reveal glutamine acquisition and signalling in cDC1s as limiting events for DC activation and putative targets for cancer treatment.


Asunto(s)
Sistema de Transporte de Aminoácidos A , Células Dendríticas , Glutamina , Neoplasias , Transducción de Señal , Sistema de Transporte de Aminoácidos A/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Glutamina/metabolismo , Neoplasias/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
6.
Nat Cell Biol ; 24(11): 1642-1654, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36302969

RESUMEN

Phosphatase and tensin homologue (PTEN) is frequently mutated in human cancer, but its roles in lymphopoiesis and tissue homeostasis remain poorly defined. Here we show that PTEN orchestrates a two-step developmental process linking antigen receptor and IL-23-Stat3 signalling to type-17 innate-like T cell generation. Loss of PTEN leads to pronounced accumulation of mature IL-17-producing innate-like T cells in the thymus. IL-23 is essential for their accumulation, and ablation of IL-23 or IL-17 signalling rectifies the reduced survival of female PTEN-haploinsufficient mice that model human patients with PTEN mutations. Single-cell transcriptome and network analyses revealed the dynamic regulation of PTEN, mTOR and metabolic activities that accompanied type-17 cell programming. Furthermore, deletion of mTORC1 or mTORC2 blocks PTEN loss-driven type-17 cell accumulation, and this is further shaped by the Foxo1 and Stat3 pathways. Collectively, our study establishes developmental and metabolic signalling networks underpinning type-17 cell fate decisions and their functional effects at coordinating PTEN-dependent tissue homeostasis.


Asunto(s)
Interleucina-17 , Linfocitos T , Humanos , Femenino , Ratones , Animales , Linfocitos T/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Homeostasis , Interleucina-23
7.
Nat Chem Biol ; 18(5): 470-481, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35484263

RESUMEN

T cells orchestrate adaptive immunity against pathogens and other immune challenges, but their dysfunction can also mediate the pathogenesis of cancer and autoimmunity. Metabolic adaptation in response to immunological and microenvironmental signals contributes to T cell function and fate decision. Lipid metabolism has emerged as a key regulator of T cell responses, with selective lipid metabolites serving as metabolic rheostats to integrate environmental cues and interplay with intracellular signaling processes. Here, we discuss how extracellular, de novo synthesized and membrane lipids orchestrate T cell biology. We also describe the roles of lipids as regulators of intracellular signaling at the levels of transcriptional, epigenetic and post-translational regulation in T cells. Finally, we summarize therapeutic targeting of lipid metabolism and signaling, and conclude with a discussion of important future directions. Understanding the molecular and functional interplay between lipid metabolism and T cell biology will ultimately inform therapeutic intervention for human disease.


Asunto(s)
Metabolismo de los Lípidos , Neoplasias , Humanos , Metabolismo de los Lípidos/fisiología , Lípidos de la Membrana , Neoplasias/metabolismo , Transducción de Señal/fisiología , Linfocitos T/metabolismo
8.
Immunity ; 55(1): 14-30, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021054

RESUMEN

Adaptive immune responses mediated by T cells and B cells are crucial for protective immunity against pathogens and tumors. Differentiation and function of immune cells require dynamic reprogramming of cellular metabolism. Metabolic inputs, pathways, and enzymes display remarkable flexibility and heterogeneity, especially in vivo. How metabolic plasticity and adaptation dictate functional specialization of immune cells is fundamental to our understanding and therapeutic modulation of the immune system. Extensive progress has been made in characterizing the effects of metabolic networks on immune cell fate and function in discrete microenvironments or immunological contexts. In this review, we summarize how rewiring of cellular metabolism determines the outcome of adaptive immunity in vivo, with a focus on how metabolites, nutrients, and driver genes in immunometabolism instruct cellular programming and immune responses during infection, inflammation, and cancer in mice and humans. Understanding context-dependent metabolic remodeling will manifest legitimate opportunities for therapeutic intervention of human disease.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Enfermedades del Sistema Inmune/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Microambiente Celular , Reprogramación Celular , Humanos
9.
Nature ; 600(7888): 308-313, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34795452

RESUMEN

Nutrients are emerging regulators of adaptive immunity1. Selective nutrients interplay with immunological signals to activate mechanistic target of rapamycin complex 1 (mTORC1), a key driver of cell metabolism2-4, but how these environmental signals are integrated for immune regulation remains unclear. Here we use genome-wide CRISPR screening combined with protein-protein interaction networks to identify regulatory modules that mediate immune receptor- and nutrient-dependent signalling to mTORC1 in mouse regulatory T (Treg) cells. SEC31A is identified to promote mTORC1 activation by interacting with the GATOR2 component SEC13 to protect it from SKP1-dependent proteasomal degradation. Accordingly, loss of SEC31A impairs T cell priming and Treg suppressive function in mice. In addition, the SWI/SNF complex restricts expression of the amino acid sensor CASTOR1, thereby enhancing mTORC1 activation. Moreover, we reveal that the CCDC101-associated SAGA complex is a potent inhibitor of mTORC1, which limits the expression of glucose and amino acid transporters and maintains T cell quiescence in vivo. Specific deletion of Ccdc101 in mouse Treg cells results in uncontrolled inflammation but improved antitumour immunity. Collectively, our results establish epigenetic and post-translational mechanisms that underpin how nutrient transporters, sensors and transducers interplay with immune signals for three-tiered regulation of mTORC1 activity and identify their pivotal roles in licensing T cell immunity and immune tolerance.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Nutrientes , Mapas de Interacción de Proteínas , Linfocitos T Reguladores , Animales , Femenino , Masculino , Ratones , Proteínas Portadoras/metabolismo , Sistemas CRISPR-Cas/genética , Factores de Transcripción Forkhead/metabolismo , Genoma/genética , Homeostasis , Tolerancia Inmunológica , Inflamación/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neoplasias/inmunología , Proteínas Nucleares/metabolismo , Nutrientes/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transactivadores/metabolismo
10.
Nature ; 595(7869): 724-729, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34234346

RESUMEN

T follicular helper (TFH) cells are crucial for B cell-mediated humoral immunity1. Although transcription factors such as BCL6 drive the differentiation of TFH cells2,3, it is unclear whether and how post-transcriptional and metabolic programs enforce TFH cell programming. Here we show that the cytidine diphosphate (CDP)-ethanolamine pathway co-ordinates the expression and localization of CXCR5 with the responses of TFH cells and humoral immunity. Using in vivo CRISPR-Cas9 screening and functional validation in mice, we identify ETNK1, PCYT2, and SELENOI-enzymes in the CDP-ethanolamine pathway for de novo synthesis of phosphatidylethanolamine (PE)-as selective post-transcriptional regulators of TFH cell differentiation that act by promoting the surface expression and functional effects of CXCR5. TFH cells exhibit unique lipid metabolic programs and PE is distributed to the outer layer of the plasma membrane, where it colocalizes with CXCR5. De novo synthesis of PE through the CDP-ethanolamine pathway co-ordinates these events to prevent the internalization and degradation of CXCR5. Genetic deletion of Pcyt2, but not of Pcyt1a (which mediates the CDP-choline pathway), in activated T cells impairs the differentiation of TFH cells, and this is associated with reduced humoral immune responses. Surface levels of PE and CXCR5 expression on B cells also depend on Pcyt2. Our results reveal that phospholipid metabolism orchestrates post-transcriptional mechanisms for TFH cell differentiation and humoral immunity, highlighting the metabolic control of context-dependent immune signalling and effector programs.


Asunto(s)
Inmunidad Humoral , Fosfatidiletanolaminas/metabolismo , Receptores CXCR5/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/inmunología , Sistemas CRISPR-Cas , Diferenciación Celular , Citidina Difosfato , Femenino , Regulación de la Expresión Génica , Humanos , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfotransferasas (Aceptor de Grupo Alcohol) , ARN Nucleotidiltransferasas , Transducción de Señal
11.
Artículo en Inglés | MEDLINE | ID: mdl-33820774

RESUMEN

The formation of long-lived memory T cells is a critical feature of the adaptive immune response. T cells undergo metabolic reprogramming to establish a functional memory population. While initial studies characterized key metabolic pathways necessary for memory T-cell development, recent findings highlight that metabolic regulation of memory T-cell subsets is diverse. Here we describe the different requirements for metabolic programs and metabolism-related signaling pathways in memory T-cell development. We further discuss the contribution of cellular metabolism to memory T-cell functional reprogramming and stemness within acute and chronic inflammatory environments. Last, we highlight knowledge gaps and propose approaches to determine the roles of metabolites and metabolic enzymes in memory T-cell fate. Understanding how cellular metabolism regulates a functionally diverse memory population will undoubtedly provide new therapeutic insights to modulate protective T-cell immunity in human disease.


Asunto(s)
Reprogramación Celular , Células T de Memoria/metabolismo , Animales , Humanos , Transducción de Señal
12.
Curr Opin Biotechnol ; 68: 240-250, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33676144

RESUMEN

T cells shape immune responses in cancer, autoimmunity and infection, in which CD4+ T helper (Th) and CD8+ T cells mediate effector responses that are suppressed by regulatory T (Treg) cells. The balance between effector T cell and Treg cell function orchestrates immune homeostasis and functional programming, with important contributions to the onset and progression of cancer. Cellular metabolism is dynamically rewired in T cells in response to environmental cues and dictates various aspects of T cell function. In this review, we summarize recent findings on how cellular metabolism modulates effector T cell and Treg cell functional fitness in homeostasis and cancer immunity, and highlight the therapeutic implications of targeting immunometabolic pathways for cancer and other diseases.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Autoinmunidad , Homeostasis , Humanos , Linfocitos T Reguladores
13.
Nature ; 591(7849): 306-311, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33627871

RESUMEN

Regulatory T cells (Treg cells) are essential for immune tolerance1, but also drive immunosuppression in the tumour microenvironment2. Therapeutic targeting of Treg cells in cancer will therefore require the identification of context-specific mechanisms that affect their function. Here we show that inhibiting lipid synthesis and metabolic signalling that are dependent on sterol-regulatory-element-binding proteins (SREBPs) in Treg cells unleashes effective antitumour immune responses without autoimmune toxicity. We find that the activity of SREBPs is upregulated in intratumoral Treg cells. Moreover, deletion of SREBP-cleavage-activating protein (SCAP)-a factor required for SREBP activity-in these cells inhibits tumour growth and boosts immunotherapy that is triggered by targeting the immune-checkpoint protein PD-1. These effects of SCAP deletion are associated with uncontrolled production of interferon-γ and impaired function of intratumoral Treg cells. Mechanistically, signalling through SCAP and SREBPs coordinates cellular programs for lipid synthesis and inhibitory receptor signalling in these cells. First, de novo fatty-acid synthesis mediated by fatty-acid synthase (FASN) contributes to functional maturation of Treg cells, and loss of FASN from Treg cells inhibits tumour growth. Second, Treg cells in tumours show enhanced expression of the PD-1 gene, through a process that depends on SREBP activity and signals via mevalonate metabolism to protein geranylgeranylation. Blocking PD-1 or SREBP signalling results in dysregulated activation of phosphatidylinositol-3-kinase in intratumoral Treg cells. Our findings show that metabolic reprogramming enforces the functional specialization of Treg cells in tumours, pointing to new ways of targeting these cells for cancer therapy.


Asunto(s)
Metabolismo de los Lípidos , Neoplasias/inmunología , Neoplasias/metabolismo , Transducción de Señal , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Colesterol/metabolismo , Ácido Graso Sintasas/metabolismo , Ácidos Grasos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ácido Mevalónico/metabolismo , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Linfocitos T Reguladores/enzimología , Regulación hacia Arriba
14.
Cell ; 184(5): 1245-1261.e21, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636132

RESUMEN

How early events in effector T cell (TEFF) subsets tune memory T cell (TMEM) responses remains incompletely understood. Here, we systematically investigated metabolic factors in fate determination of TEFF and TMEM cells using in vivo pooled CRISPR screening, focusing on negative regulators of TMEM responses. We found that amino acid transporters Slc7a1 and Slc38a2 dampened the magnitude of TMEM differentiation, in part through modulating mTORC1 signaling. By integrating genetic and systems approaches, we identified cellular and metabolic heterogeneity among TEFF cells, with terminal effector differentiation associated with establishment of metabolic quiescence and exit from the cell cycle. Importantly, Pofut1 (protein-O-fucosyltransferase-1) linked GDP-fucose availability to downstream Notch-Rbpj signaling, and perturbation of this nutrient signaling axis blocked terminal effector differentiation but drove context-dependent TEFF proliferation and TMEM development. Our study establishes that nutrient uptake and signaling are key determinants of T cell fate and shape the quantity and quality of TMEM responses.


Asunto(s)
Aminoácidos/metabolismo , Linfocitos T CD8-positivos/citología , Memoria Inmunológica , Transducción de Señal , Sistemas de Transporte de Aminoácidos/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Sistemas CRISPR-Cas , Ciclo Celular , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Transgénicos , Células Precursoras de Linfocitos T/citología
15.
Cell Metab ; 32(6): 996-1011.e7, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33207246

RESUMEN

Effector regulatory T (eTreg) cells are essential for immune tolerance and depend upon T cell receptor (TCR) signals for generation. The immunometabolic signaling mechanisms that promote the differentiation and maintenance of eTreg cells remain unclear. Here, we show that isoprenoid-dependent posttranslational lipid modifications dictate eTreg cell accumulation and function by intersecting with TCR-induced intracellular signaling. We find that isoprenoids are essential for activated Treg cell suppressive activity, and Treg cell-specific deletion of the respective farnesylation- and geranylgeranylation-promoting enzymes Fntb or Pggt1b leads to the development of fatal autoimmunity, associated with reduced eTreg cell accumulation. Mechanistically, Fntb promotes eTreg cell maintenance by regulating mTORC1 activity and ICOS expression. In contrast, Pggt1b acts as a rheostat of TCR-dependent transcriptional programming and Rac-mediated signaling for establishment of eTreg cell differentiation and immune tolerance. Therefore, our results identify bidirectional metabolic signaling, specifically between immunoreceptor signaling and metabolism-mediated posttranslational lipid modifications, for the differentiation and maintenance of eTreg cells.


Asunto(s)
Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Linfocitos T Reguladores , Terpenos , Animales , Femenino , Tolerancia Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Prenilación de Proteína , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Terpenos/inmunología , Terpenos/metabolismo
16.
Immunity ; 53(2): 243-245, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814023

RESUMEN

Chimeric antigen receptor (CAR) T cells are potent drivers of antitumor immunity, but promoting durable CAR T cell responses remains challenging. In this issue of Immunity, Li et al. (2020) show that blockade of CAR ubiquitination induces CAR recycling to the cell surface, leading to increased CAR T cell cytotoxicity and longevity by amplifying 41BB-dependent signaling and mitochondrial metabolism.


Asunto(s)
Receptores Quiméricos de Antígenos , Línea Celular Tumoral , Regulación hacia Abajo , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Exp Med ; 217(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32289155

RESUMEN

Invariant natural killer T (iNKT) cells acquire effector functions during development by mechanisms that remain poorly understood. Here, we show that the Hippo kinases Mst1 and Mst2 act as molecular rheostats for the terminal maturation and effector differentiation programs of iNKT cells. Loss of Mst1 alone or together with Mst2 impedes iNKT cell development, associated with defective IL-15-dependent cell survival. Mechanistically, Mst1 enforces iNKT cellular and transcriptional quiescence associated with maturation and commitment to iNKT1 cells by suppressing proliferation and Opa1-related mitochondrial metabolism that are dynamically regulated during iNKT cell development. Furthermore, Mst1 shapes the reciprocal fate decisions between iNKT1 and iNKT17 effector cells, which respectively depend upon mitochondrial dynamics and ICOS-mTORC2 signaling. Collectively, these findings establish Mst1 as a crucial regulator of mitochondrial homeostasis and quiescence in iNKT cell development and effector lineage differentiation and highlight that establishment of quiescence programs underlies iNKT cell development and effector maturation.


Asunto(s)
Ciclo Celular , Linaje de la Célula , Factor de Crecimiento de Hepatocito/metabolismo , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Supervivencia Celular , Regulación de la Expresión Génica , Vía de Señalización Hippo , Homeostasis , Interleucina-15/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Serina-Treonina Quinasa 3 , Transcripción Genética
18.
Cell Res ; 30(4): 328-342, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32203134

RESUMEN

Adaptive immunity is essential for pathogen and tumor eradication, but may also trigger uncontrolled or pathological inflammation. T cell receptor, co-stimulatory and cytokine signals coordinately dictate specific signaling networks that trigger the activation and functional programming of T cells. In addition, cellular metabolism promotes T cell responses and is dynamically regulated through the interplay of serine/threonine kinases, immunological cues and nutrient signaling networks. In this review, we summarize the upstream regulators and signaling effectors of key serine/threonine kinase-mediated signaling networks, including PI3K-AGC kinases, mTOR and LKB1-AMPK pathways that regulate metabolism, especially in T cells. We also provide our perspectives about the pending questions and clinical applicability of immunometabolic signaling. Understanding the regulators and effectors of immunometabolic signaling networks may uncover therapeutic targets to modulate metabolic programming and T cell responses in human disease.


Asunto(s)
Proteínas Quinasas Activadas por AMP/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Linfocitos T , Serina-Treonina Quinasas TOR/inmunología , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Humanos , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
19.
Immunol Rev ; 295(1): 15-38, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32212344

RESUMEN

The evolutionarily conserved serine/threonine kinase mTOR (mechanistic target of rapamycin) forms the distinct protein complexes mTORC1 and mTORC2 and integrates signals from the environment to coordinate downstream signaling events and various cellular processes. T cells rely on mTOR activity for their development and to establish their homeostasis and functional fitness. Here, we review recent progress in our understanding of the upstream signaling and downstream targets of mTOR. We also provide an updated overview of the roles of mTOR in T-cell development, homeostasis, activation, and effector-cell fate decisions, as well as its important impacts on the suppressive activity of regulatory T cells. Moreover, we summarize the emerging roles of mTOR in T-cell exhaustion and transdifferentiation. A better understanding of the contribution of mTOR to T-cell fate decisions will ultimately aid in the therapeutic targeting of mTOR in human disease.


Asunto(s)
Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Transdiferenciación Celular/inmunología , Citocinas/metabolismo , Citoesqueleto/metabolismo , Metabolismo Energético , Humanos , Memoria Inmunológica , Activación de Linfocitos/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
20.
Sci Adv ; 6(1): eaaw6443, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31911938

RESUMEN

Regulatory T cell (Treg) activation and expansion occur during neonatal life and inflammation to establish immunosuppression, yet the mechanisms governing these events are incompletely understood. We report that the transcriptional regulator c-Myc (Myc) controls immune homeostasis through regulation of Treg accumulation and functional activation. Myc activity is enriched in Tregs generated during neonatal life and responding to inflammation. Myc-deficient Tregs show defects in accumulation and ability to transition to an activated state. Consequently, loss of Myc in Tregs results in an early-onset autoimmune disorder accompanied by uncontrolled effector CD4+ and CD8+ T cell responses. Mechanistically, Myc regulates mitochondrial oxidative metabolism but is dispensable for fatty acid oxidation (FAO). Indeed, Treg-specific deletion of Cox10, which promotes oxidative phosphorylation, but not Cpt1a, the rate-limiting enzyme for FAO, results in impaired Treg function and maturation. Thus, Myc coordinates Treg accumulation, transitional activation, and metabolic programming to orchestrate immune homeostasis.


Asunto(s)
Ácidos Grasos/metabolismo , Terapia de Inmunosupresión , Inflamación/inmunología , Proteínas Proto-Oncogénicas c-myc/genética , Linfocitos T Reguladores/inmunología , Transferasas Alquil y Aril/inmunología , Animales , Animales Recién Nacidos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Citometría de Flujo , Homeostasis/inmunología , Inflamación/genética , Proteínas de la Membrana/inmunología , Ratones , Oxidación-Reducción , Fosforilación Oxidativa , Proteínas Proto-Oncogénicas c-myc/inmunología , Linfocitos T Reguladores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA