Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 184(15): 4016-4031.e22, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34081922

RESUMO

Cross-presentation of antigens from dead tumor cells by type 1 conventional dendritic cells (cDC1s) is thought to underlie priming of anti-cancer CD8+ T cells. cDC1 express high levels of DNGR-1 (a.k.a. CLEC9A), a receptor that binds to F-actin exposed by dead cell debris and promotes cross-presentation of associated antigens. Here, we show that secreted gelsolin (sGSN), an extracellular protein, decreases DNGR-1 binding to F-actin and cross-presentation of dead cell-associated antigens by cDC1s. Mice deficient in sGsn display increased DNGR-1-dependent resistance to transplantable tumors, especially ones expressing neoantigens associated with the actin cytoskeleton, and exhibit greater responsiveness to cancer immunotherapy. In human cancers, lower levels of intratumoral sGSN transcripts, as well as presence of mutations in proteins associated with the actin cytoskeleton, are associated with signatures of anti-cancer immunity and increased patient survival. Our results reveal a natural barrier to cross-presentation of cancer antigens that dampens anti-tumor CD8+ T cell responses.


Assuntos
Apresentação Cruzada/imunologia , Gelsolina/metabolismo , Imunidade , Lectinas Tipo C/metabolismo , Neoplasias/imunologia , Receptores Imunológicos/metabolismo , Receptores Mitogênicos/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Apresentação Cruzada/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Gelsolina/química , Gelsolina/deficiência , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunidade/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Mutação/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Ligação Proteica/efeitos dos fármacos , Análise de Sobrevida
2.
Nat Immunol ; 22(2): 140-153, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33349708

RESUMO

Type 1 conventional dendritic (cDC1) cells are necessary for cross-presentation of many viral and tumor antigens to CD8+ T cells. cDC1 cells can be identified in mice and humans by high expression of DNGR-1 (also known as CLEC9A), a receptor that binds dead-cell debris and facilitates XP of corpse-associated antigens. Here, we show that DNGR-1 is a dedicated XP receptor that signals upon ligand engagement to promote phagosomal rupture. This allows escape of phagosomal contents into the cytosol, where they access the endogenous major histocompatibility complex class I antigen processing pathway. The activity of DNGR-1 maps to its signaling domain, which activates SYK and NADPH oxidase to cause phagosomal damage even when spliced into a heterologous receptor and expressed in heterologous cells. Our data reveal the existence of innate immune receptors that couple ligand binding to endocytic vesicle damage to permit MHC class I antigen presentation of exogenous antigens and to regulate adaptive immunity.


Assuntos
Apresentação de Antígeno , Apresentação Cruzada , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Fagossomos/metabolismo , Receptores Imunológicos/metabolismo , Receptores Mitogênicos/metabolismo , Linfócitos T/metabolismo , Animais , Morte Celular , Técnicas de Cocultura , Células Dendríticas/imunologia , Células HEK293 , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Lectinas Tipo C/genética , Ligantes , Camundongos , NADPH Oxidases/metabolismo , Fagossomos/genética , Fagossomos/imunologia , Fosforilação , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Receptores Imunológicos/genética , Receptores Mitogênicos/genética , Transdução de Sinais , Quinase Syk/metabolismo , Linfócitos T/imunologia
3.
Cell ; 169(4): 570-586, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28475890

RESUMO

Choices have consequences. Immune cells survey and migrate throughout the body and sometimes take residence in niche environments with distinct communities of cells, extracellular matrix, and nutrients that may differ from those in which they matured. Imbedded in immune cell physiology are metabolic pathways and metabolites that not only provide energy and substrates for growth and survival, but also instruct effector functions, differentiation, and gene expression. This review of immunometabolism will reference the most recent literature to cover the choices that environments impose on the metabolism and function of immune cells and highlight their consequences during homeostasis and disease.


Assuntos
Leucócitos/citologia , Leucócitos/imunologia , Animais , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Humanos , Leucócitos/metabolismo , Linfócitos T/imunologia , Microambiente Tumoral
4.
Cell ; 171(2): 385-397.e11, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28919076

RESUMO

T cell receptor (TCR) signaling without CD28 can elicit primary effector T cells, but memory T cells generated during this process are anergic, failing to respond to secondary antigen exposure. We show that, upon T cell activation, CD28 transiently promotes expression of carnitine palmitoyltransferase 1a (Cpt1a), an enzyme that facilitates mitochondrial fatty acid oxidation (FAO), before the first cell division, coinciding with mitochondrial elongation and enhanced spare respiratory capacity (SRC). microRNA-33 (miR33), a target of thioredoxin-interacting protein (TXNIP), attenuates Cpt1a expression in the absence of CD28, resulting in cells that thereafter are metabolically compromised during reactivation or periods of increased bioenergetic demand. Early CD28-dependent mitochondrial engagement is needed for T cells to remodel cristae, develop SRC, and rapidly produce cytokines upon restimulation-cardinal features of protective memory T cells. Our data show that initial CD28 signals during T cell activation prime mitochondria with latent metabolic capacity that is essential for future T cell responses.


Assuntos
Antígenos CD28/metabolismo , Ativação Linfocitária , Mitocôndrias/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Carnitina O-Palmitoiltransferase , Inibidores Enzimáticos/farmacologia , Compostos de Epóxi/farmacologia , Humanos , Interleucina-15/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/metabolismo , Estresse Fisiológico , Linfócitos T/metabolismo
5.
Cell ; 166(1): 63-76, 2016 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-27293185

RESUMO

Activated effector T (TE) cells augment anabolic pathways of metabolism, such as aerobic glycolysis, while memory T (TM) cells engage catabolic pathways, like fatty acid oxidation (FAO). However, signals that drive these differences remain unclear. Mitochondria are metabolic organelles that actively transform their ultrastructure. Therefore, we questioned whether mitochondrial dynamics controls T cell metabolism. We show that TE cells have punctate mitochondria, while TM cells maintain fused networks. The fusion protein Opa1 is required for TM, but not TE cells after infection, and enforcing fusion in TE cells imposes TM cell characteristics and enhances antitumor function. Our data suggest that, by altering cristae morphology, fusion in TM cells configures electron transport chain (ETC) complex associations favoring oxidative phosphorylation (OXPHOS) and FAO, while fission in TE cells leads to cristae expansion, reducing ETC efficiency and promoting aerobic glycolysis. Thus, mitochondrial remodeling is a signaling mechanism that instructs T cell metabolic programming.


Assuntos
Dinâmica Mitocondrial , Linfócitos T/citologia , Linfócitos T/metabolismo , Animais , Diferenciação Celular , Transporte de Elétrons , Ácidos Graxos/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Glicólise , Humanos , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Transdução de Sinais , Linfócitos T/imunologia
6.
Cell ; 162(6): 1229-41, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26321679

RESUMO

Failure of T cells to protect against cancer is thought to result from lack of antigen recognition, chronic activation, and/or suppression by other cells. Using a mouse sarcoma model, we show that glucose consumption by tumors metabolically restricts T cells, leading to their dampened mTOR activity, glycolytic capacity, and IFN-γ production, thereby allowing tumor progression. We show that enhancing glycolysis in an antigenic "regressor" tumor is sufficient to override the protective ability of T cells to control tumor growth. We also show that checkpoint blockade antibodies against CTLA-4, PD-1, and PD-L1, which are used clinically, restore glucose in tumor microenvironment, permitting T cell glycolysis and IFN-γ production. Furthermore, we found that blocking PD-L1 directly on tumors dampens glycolysis by inhibiting mTOR activity and decreasing expression of glycolysis enzymes, reflecting a role for PD-L1 in tumor glucose utilization. Our results establish that tumor-imposed metabolic restrictions can mediate T cell hyporesponsiveness during cancer.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Glicólise , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral , Animais , Anticorpos Monoclonais/administração & dosagem , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Interferon gama/imunologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia
7.
Nature ; 615(7953): 705-711, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36922598

RESUMO

Artificial sweeteners are used as calorie-free sugar substitutes in many food products and their consumption has increased substantially over the past years1. Although generally regarded as safe, some concerns have been raised about the long-term safety of the consumption of certain sweeteners2-5. In this study, we show that the intake of high doses of sucralose in mice results in immunomodulatory effects by limiting T cell proliferation and T cell differentiation. Mechanistically, sucralose affects the membrane order of T cells, accompanied by a reduced efficiency of T cell receptor signalling and intracellular calcium mobilization. Mice given sucralose show decreased CD8+ T cell antigen-specific responses in subcutaneous cancer models and bacterial infection models, and reduced T cell function in models of T cell-mediated autoimmunity. Overall, these findings suggest that a high intake of sucralose can dampen T cell-mediated responses, an effect that could be used in therapy to mitigate T cell-dependent autoimmune disorders.


Assuntos
Sacarose , Edulcorantes , Linfócitos T , Animais , Camundongos , Sacarose/análogos & derivados , Edulcorantes/administração & dosagem , Edulcorantes/efeitos adversos , Edulcorantes/farmacologia , Edulcorantes/uso terapêutico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia , Inocuidade dos Alimentos , Sinalização do Cálcio/efeitos dos fármacos , Receptores de Antígenos de Linfócitos T/efeitos dos fármacos , Receptores de Antígenos de Linfócitos T/imunologia , Infecções Bacterianas/imunologia , Neoplasias/imunologia , Autoimunidade/efeitos dos fármacos , Autoimunidade/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia
8.
Nat Immunol ; 17(6): 656-65, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27043409

RESUMO

Group 2 innate lymphoid cells (ILC2s) regulate tissue inflammation and repair after activation by cell-extrinsic factors such as host-derived cytokines. However, the cell-intrinsic metabolic pathways that control ILC2 function are undefined. Here we demonstrate that expression of the enzyme arginase-1 (Arg1) during acute or chronic lung inflammation is a conserved trait of mouse and human ILC2s. Deletion of mouse ILC-intrinsic Arg1 abrogated type 2 lung inflammation by restraining ILC2 proliferation and dampening cytokine production. Mechanistically, inhibition of Arg1 enzymatic activity disrupted multiple components of ILC2 metabolic programming by altering arginine catabolism, impairing polyamine biosynthesis and reducing aerobic glycolysis. These data identify Arg1 as a key regulator of ILC2 bioenergetics that controls proliferative capacity and proinflammatory functions promoting type 2 inflammation.


Assuntos
Arginase/metabolismo , Linfócitos/fisiologia , Pneumonia/imunologia , Animais , Arginase/genética , Proliferação de Células/genética , Células Cultivadas , Citocinas/metabolismo , Glicólise/genética , Humanos , Imunidade Inata , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Poliaminas/metabolismo , Células Th2/imunologia
10.
Immunity ; 49(6): 1021-1033.e6, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30566880

RESUMO

Metabolic engagement is intrinsic to immune cell function. Prostaglandin E2 (PGE2) has been shown to modulate macrophage activation, yet how PGE2 might affect metabolism is unclear. Here, we show that PGE2 caused mitochondrial membrane potential (Δψm) to dissipate in interleukin-4-activated (M(IL-4)) macrophages. Effects on Δψm were a consequence of PGE2-initiated transcriptional regulation of genes, particularly Got1, in the malate-aspartate shuttle (MAS). Reduced Δψm caused alterations in the expression of 126 voltage-regulated genes (VRGs), including those encoding resistin-like molecule α (RELMα), a key marker of M(IL-4) cells, and genes that regulate the cell cycle. The transcription factor ETS variant 1 (ETV1) played a role in the regulation of 38% of the VRGs. These results reveal ETV1 as a Δψm-sensitive transcription factor and Δψm as a mediator of mitochondrial-directed nuclear gene expression.


Assuntos
Núcleo Celular/efeitos dos fármacos , Dinoprostona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Animais , Núcleo Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Interleucina-4/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Immunity ; 44(6): 1325-36, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27332732

RESUMO

Greater understanding of the complex host responses induced by type 1 interferon (IFN) cytokines could allow new therapeutic approaches for diseases in which these cytokines are implicated. We found that in response to the Toll-like receptor-9 agonist CpGA, plasmacytoid dendritic cells (pDC) produced type 1 IFNs, which, through an autocrine type 1 IFN receptor-dependent pathway, induced changes in cellular metabolism characterized by increased fatty acid oxidation (FAO) and oxidative phosphorylation (OXPHOS). Direct inhibition of FAO and of pathways that support this process, such as fatty acid synthesis, prevented full pDC activation. Type 1 IFNs also induced increased FAO and OXPHOS in non-hematopoietic cells and were found to be responsible for increased FAO and OXPHOS in virus-infected cells. Increased FAO and OXPHOS in response to type 1 IFNs was regulated by PPARα. Our findings reveal FAO, OXPHOS and PPARα as potential targets to therapeutically modulate downstream effects of type 1 IFNs.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/metabolismo , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , PPAR alfa/metabolismo , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Acetil-CoA C-Aciltransferase/metabolismo , Animais , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Diferenciação Celular , Células Cultivadas , Ilhas de CpG/imunologia , Enoil-CoA Hidratase/metabolismo , Regulação da Expressão Gênica , Imunidade , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/imunologia , Fosforilação Oxidativa , Racemases e Epimerases/metabolismo , Receptores de Interferon/metabolismo , Transdução de Sinais , Receptor Toll-Like 9/metabolismo
12.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34161266

RESUMO

Fever can provide a survival advantage during infection. Metabolic processes are sensitive to environmental conditions, but the effect of fever on T cell metabolism is not well characterized. We show that in activated CD8+ T cells, exposure to febrile temperature (39 °C) augmented metabolic activity and T cell effector functions, despite having a limited effect on proliferation or activation marker expression. Transcriptional profiling revealed an up-regulation of mitochondrial pathways, which was consistent with increased mass and metabolism observed in T cells exposed to 39 °C. Through in vitro and in vivo models, we determined that mitochondrial translation is integral to the enhanced metabolic activity and function of CD8+ T cells exposed to febrile temperature. Transiently exposing donor lymphocytes to 39 °C prior to infusion in a myeloid leukemia mouse model conferred enhanced therapeutic efficacy, raising the possibility that exposure of T cells to febrile temperatures could have clinical potential.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Febre/imunologia , Mitocôndrias/metabolismo , Biossíntese de Proteínas , Animais , Antineoplásicos/metabolismo , Linfócitos T CD8-Positivos/ultraestrutura , Citocinas/biossíntese , Glucose/metabolismo , Leucemia Mieloide/imunologia , Leucemia Mieloide/patologia , Leucemia Mieloide/prevenção & controle , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitocôndrias/ultraestrutura , Modelos Biológicos , Temperatura
13.
Immunity ; 41(1): 75-88, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25001241

RESUMO

Generation of CD8(+) memory T cells requires metabolic reprogramming that is characterized by enhanced mitochondrial fatty-acid oxidation (FAO). However, where the fatty acids (FA) that fuel this process come from remains unclear. While CD8(+) memory T cells engage FAO to a greater extent, we found that they acquired substantially fewer long-chain FA from their external environment than CD8(+) effector T (Teff) cells. Rather than using extracellular FA directly, memory T cells used extracellular glucose to support FAO and oxidative phosphorylation (OXPHOS), suggesting that lipids must be synthesized to generate the substrates needed for FAO. We have demonstrated that memory T cells rely on cell intrinsic expression of the lysosomal hydrolase LAL (lysosomal acid lipase) to mobilize FA for FAO and memory T cell development. Our observations link LAL to metabolic reprogramming in lymphocytes and show that cell intrinsic lipolysis is deterministic for memory T cell fate.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Ácidos Graxos/metabolismo , Memória Imunológica/imunologia , Lipólise/imunologia , Esterol Esterase/metabolismo , 4-Butirolactona/análogos & derivados , 4-Butirolactona/farmacologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ácido Graxo Sintases/antagonistas & inibidores , Ácido Graxo Sintases/genética , Ácidos Graxos/biossíntese , Glucose/metabolismo , Interleucina-15/imunologia , Interleucina-2/imunologia , Lipólise/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/metabolismo , Oxirredução , Fosforilação Oxidativa , Oxigênio/metabolismo , Proteínas Quinases/genética , Interferência de RNA , RNA Interferente Pequeno , Esterol Esterase/biossíntese
14.
J Cell Sci ; 133(5)2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-32144194

RESUMO

The importance of cancer-cell-autonomous functions of the tumour suppressor p53 (encoded by TP53) has been established in many studies, but it is now clear that the p53 status of the cancer cell also has a profound impact on the immune response. Loss or mutation of p53 in cancers can affect the recruitment and activity of myeloid and T cells, allowing immune evasion and promoting cancer progression. p53 can also function in immune cells, resulting in various outcomes that can impede or support tumour development. Understanding the role of p53 in tumour and immune cells will help in the development of therapeutic approaches that can harness the differential p53 status of cancers compared with most normal tissue.


Assuntos
Neoplasias , Proteína Supressora de Tumor p53 , Humanos , Imunidade , Mutação , Neoplasias/genética , Proteína Supressora de Tumor p53/genética
16.
J Virol ; 89(10): 5592-601, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25762732

RESUMO

UNLABELLED: The host-targeted antiviral drug UV-4B reduces viral replication and promotes survival in a mouse model of experimental dengue virus (DENV) infection. UV-4B is an iminosugar that inhibits the α-glucosidase family of enzymes and subsequently the folding of glycosylated proteins, both viral and host. Here, we utilized next-generation sequencing to investigate evolution of a flavivirus under selective pressure by a host-targeted antiviral in vivo. In viral populations recovered from UV-4B-treated mice, there was a significant increase in the number of single-nucleotide polymorphisms (SNPs) and the ratio of nonsynonymous to synonymous SNPs compared to findings in viral populations from vehicle-treated mice. The strongest evidence of positive selection was in the glycosylated membrane protein, thereby providing in vivo validation of the mechanism of action of an iminosugar. In addition, mutations in glycosylated proteins were present only in drug-treated mice after a single passage. However, the bulk of the other mutations were present in both populations, indicating nonspecific selective pressure. Together with the continued control of viremia by UV-4B, these findings are consistent with the previously predicted high genetic barrier to escape mutations in host-targeted antivirals. IMPORTANCE: Although hundreds of millions of people are infected with DENV every year, there is currently no approved vaccine or antiviral therapy. UV-4B has demonstrated antiviral activity against DENV and is expected to enter clinical trials soon. Therefore, it is important to understand the mechanisms of DENV resistance to UV-4B. Host-targeted antivirals are thought to have a higher genetic barrier to escape mutants than directly acting antivirals, yet there are very few published studies of viral evolution under host-targeted antivirals. No study to date has described flavivirus evolution in vivo under selective pressure by a host-based antiviral drug. We present the first in vivo study of the sequential progression of viral evolution under selective pressure by a host-targeted antiviral compound. This study bolsters support for the clinical development of UV-4B as an antiviral drug against DENV, and it provides a framework to compare how treatment with other host-targeted antiflaviviral drugs in humans and different animal models influence viral genetic diversity.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/genética , Dengue/tratamento farmacológico , Dengue/virologia , Animais , Vírus da Dengue/fisiologia , Modelos Animais de Doenças , Evolução Molecular , Variação Genética , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imino Açúcares/farmacologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Mutação , Polimorfismo de Nucleotídeo Único , Seleção Genética , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos
17.
Proc Natl Acad Sci U S A ; 110(35): 14336-41, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23940348

RESUMO

A characteristic of memory T (TM) cells is their ability to mount faster and stronger responses to reinfection than naïve T (TN) cells do in response to an initial infection. However, the mechanisms that allow this rapid recall are not completely understood. We found that CD8 TM cells have more mitochondrial mass than CD8 TN cells and, that upon activation, the resulting secondary effector T (TE) cells proliferate more quickly, produce more cytokines, and maintain greater ATP levels than primary effector T cells. We also found that after activation, TM cells increase oxidative phosphorylation and aerobic glycolysis and sustain this increase to a greater extent than TN cells, suggesting that greater mitochondrial mass in TM cells not only promotes oxidative capacity, but also glycolytic capacity. We show that mitochondrial ATP is essential for the rapid induction of glycolysis in response to activation and the initiation of proliferation of both TN and TM cells. We also found that fatty acid oxidation is needed for TM cells to rapidly respond upon restimulation. Finally, we show that dissociation of the glycolysis enzyme hexokinase from mitochondria impairs proliferation and blocks the rapid induction of glycolysis upon T-cell receptor stimulation in TM cells. Our results demonstrate that greater mitochondrial mass endows TM cells with a bioenergetic advantage that underlies their ability to rapidly recall in response to reinfection.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Metabolismo Energético , Ativação Linfocitária , Camundongos , Tamanho Mitocondrial
18.
Science ; 384(6694): 428-437, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38662827

RESUMO

A role for vitamin D in immune modulation and in cancer has been suggested. In this work, we report that mice with increased availability of vitamin D display greater immune-dependent resistance to transplantable cancers and augmented responses to checkpoint blockade immunotherapies. Similarly, in humans, vitamin D-induced genes correlate with improved responses to immune checkpoint inhibitor treatment as well as with immunity to cancer and increased overall survival. In mice, resistance is attributable to the activity of vitamin D on intestinal epithelial cells, which alters microbiome composition in favor of Bacteroides fragilis, which positively regulates cancer immunity. Our findings indicate a previously unappreciated connection between vitamin D, microbial commensal communities, and immune responses to cancer. Collectively, they highlight vitamin D levels as a potential determinant of cancer immunity and immunotherapy success.


Assuntos
Bacteroides fragilis , Microbioma Gastrointestinal , Inibidores de Checkpoint Imunológico , Neoplasias , Vitamina D , Animais , Feminino , Humanos , Masculino , Camundongos , Bacteroides fragilis/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Neoplasias/microbiologia , Neoplasias/terapia , Vitamina D/administração & dosagem , Vitamina D/metabolismo , Dieta , Linhagem Celular Tumoral , Calcifediol/administração & dosagem , Calcifediol/metabolismo , Proteína de Ligação a Vitamina D/genética , Proteína de Ligação a Vitamina D/metabolismo
19.
PLoS Pathog ; 7(2): e1001297, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21379341

RESUMO

Dengue virus (DENV) is a mosquito-borne flavivirus, and symptoms of infection range from asymptomatic to the severe dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). High viral loads correlate with disease severity, and both type I & II interferons (IFNs) are crucial for controlling viral replication. We have previously reported that signal transducer and activator of transcription (STAT) 1-deficient mice are resistant to DENV-induced disease, but little is known about this STAT1-independent mechanism of protection. To determine the molecular basis of the STAT1-independent pathway, mice lacking STAT1, STAT2, or both STAT1 and STAT2 were infected with a virulent mouse-adapted strain of DENV2. In the first 72 hours of infection, the single-deficient mice lacking STAT1 or STAT2 possessed 50-100 fold higher levels of viral RNA than wild type mice in the serum, spleen, and other visceral tissues, but remained resistant to DENV-induced death. In contrast, the double-deficient mice exhibited the early death phenotype previously observed in type I and II IFN receptor knockout mice (AG129), indicating that STAT2 is the mediator of the STAT1-independent host defense mechanism. Further studies demonstrated that this STAT2-dependent STAT1-independent mechanism requires the type I IFN receptor, and contributes to the autocrine amplification of type I IFN expression. Examination of gene expression in the spleen and bone marrow-derived macrophages following DENV infection revealed STAT2-dependent pathways can induce the transcription of a subset of interferon stimulated genes even in the absence of STAT1. Collectively, these results help elucidate the nature of the poorly understood STAT1-independent host defense mechanism against viruses by identifying a functional type I IFN/STAT2 signaling pathway following DENV infection in vivo.


Assuntos
Vírus da Dengue/patogenicidade , Dengue/imunologia , Dengue/virologia , Imunidade Inata , Receptor de Interferon alfa e beta/metabolismo , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT2/fisiologia , Animais , Western Blotting , Imunoprecipitação da Cromatina , DNA Viral/genética , Dengue/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imunofluorescência , Macrófagos , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , RNA Viral/genética , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Replicação Viral
20.
Cell Rep ; 42(12): 113506, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38019655

RESUMO

Cross-presentation of dead cell-associated antigens by conventional dendritic cells type 1 (cDC1s) is critical for CD8+ T cells response against many tumors and viral infections. It is facilitated by DNGR-1 (CLEC9A), an SYK-coupled cDC1 receptor that detects dead cell debris. Here, we report that DNGR-1 engagement leads to rapid activation of CBL and CBL-B E3 ligases to cause K63-linked ubiquitination of SYK and terminate signaling. Genetic deletion of CBL E3 ligases or charge-conserved mutation of target lysines within SYK abolishes SYK ubiquitination and results in enhanced DNGR-1-dependent antigen cross-presentation. We also find that cDC1 deficient in CBL E3 ligases are more efficient at cross-priming CD8+ T cells to dead cell-associated antigens and promoting host resistance to tumors. Our findings reveal a role for CBL-dependent ubiquitination in limiting cross-presentation of dead cell-associated antigens and highlight an axis of negative regulation of cDC1 activity that could be exploited to increase anti-tumor immunity.


Assuntos
Apresentação Cruzada , Ubiquitina-Proteína Ligases , Linfócitos T CD8-Positivos , Proteínas Proto-Oncogênicas c-cbl , Ubiquitinação , Células Dendríticas , Quinase Syk
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA