Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 15: 1293723, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38690263

RESUMO

T cells must adapt to variations in tissue microenvironments; these adaptations include the degree of oxygen availability. The hypoxia-inducible factor (HIF) transcription factors control much of this adaptation, and thus regulate many aspects of T cell activation and function. The HIFs are in turn regulated by oxygen-dependent hydroxylases: both the prolyl hydroxylases (PHDs) which interact with the VHL tumour suppressor and control HIF turnover, and the asparaginyl hydroxylase known as the Factor inhibiting HIF (FIH), which modulates HIF transcriptional activity. To determine the role of this latter factor in T cell function, we generated T cell-specific FIH knockout mice. We found that FIH regulates T cell fate and function in a HIF-dependent manner and show that the effects of FIH activity occur predominantly at physiological oxygen concentrations. T cell-specific loss of FIH boosts T cell cytotoxicity, augments T cell expansion in vivo, and improves anti-tumour immunotherapy in mice. Specifically inhibiting FIH in T cells may therefore represent a promising strategy for cancer immunotherapy.


Assuntos
Diferenciação Celular , Camundongos Knockout , Animais , Camundongos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ativação Linfocitária/imunologia , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Camundongos Endogâmicos C57BL
2.
Cell Rep ; 42(9): 113013, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37632752

RESUMO

2-Hydroxyglutarate (2HG) is a byproduct of the tricarboxylic acid (TCA) cycle and is readily detected in the tissues of healthy individuals. 2HG is found in two enantiomeric forms: S-2HG and R-2HG. Here, we investigate the differential roles of these two enantiomers in cluster of differentiation (CD)8+ T cell biology, where we find they have highly divergent effects on proliferation, differentiation, and T cell function. We show here an analysis of structural determinants that likely underlie these differential effects on specific α-ketoglutarate (αKG)-dependent enzymes. Treatment of CD8+ T cells with exogenous S-2HG, but not R-2HG, increased CD8+ T cell fitness in vivo and enhanced anti-tumor activity. These data show that S-2HG and R-2HG should be considered as two distinct and important actors in the regulation of T cell function.


Assuntos
Neoplasias , Linfócitos T Citotóxicos , Humanos , Linfócitos T Citotóxicos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Glutaratos/metabolismo , Neoplasias/metabolismo , Isocitrato Desidrogenase
3.
Nat Metab ; 5(10): 1747-1764, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37605057

RESUMO

T cell function and fate can be influenced by several metabolites: in some cases, acting through enzymatic inhibition of α-ketoglutarate-dependent dioxygenases, in others, through post-translational modification of lysines in important targets. We show here that glutarate, a product of amino acid catabolism, has the capacity to do both, and has potent effects on T cell function and differentiation. We found that glutarate exerts those effects both through α-ketoglutarate-dependent dioxygenase inhibition, and through direct regulation of T cell metabolism via glutarylation of the pyruvate dehydrogenase E2 subunit. Administration of diethyl glutarate, a cell-permeable form of glutarate, alters CD8+ T cell differentiation and increases cytotoxicity against target cells. In vivo administration of the compound is correlated with increased levels of both peripheral and intratumoural cytotoxic CD8+ T cells. These results demonstrate that glutarate is an important regulator of T cell metabolism and differentiation with a potential role in the improvement of T cell immunotherapy.


Assuntos
Fenômenos Bioquímicos , Linfócitos T CD8-Positivos , Linfócitos T CD8-Positivos/metabolismo , Glutaratos/metabolismo
4.
Elife ; 122023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37166103

RESUMO

Oxygenation levels are a determinative factor in T cell function. Here, we describe how oxygen tensions sensed by mouse and human T cells at the moment of activation act to persistently modulate both differentiation and function. We found that in a protocol of CAR-T cell generation, 24 hr of low oxygen levels during initial CD8+ T cell priming is sufficient to enhance antitumour cytotoxicity in a preclinical model. This is the case even when CAR-T cells are subsequently cultured under high oxygen tensions prior to adoptive transfer. Increased hypoxia-inducible transcription factor (HIF) expression was able to alter T cell fate in a similar manner to exposure to low oxygen tensions; however, only a controlled or temporary increase in HIF signalling was able to consistently improve cytotoxic function of T cells. These data show that oxygenation levels during and immediately after T cell activation play an essential role in regulating T cell function.


Assuntos
Linfócitos T CD8-Positivos , Oxigênio , Camundongos , Humanos , Animais , Oxigênio/metabolismo , Transdução de Sinais , Ativação Linfocitária , Transferência Adotiva
5.
Life (Basel) ; 13(2)2023 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-36836944

RESUMO

Canine vaccination is the main tool for preventing dangerous and widespread diseases. The strongly recommended (core) dog vaccines are against Canine Parvovirus type 2 (CPV-2), Canine Distemper Virus (CDV), and Canine Adenovirus (CAdV-1), but vaccination protocols should be tailored to dog lifestyles. Vaccination guidelines suggest vaccinating adult dogs no more frequently than every 3 years using modified live (attenuated) vaccines (MLV), thus obtaining a long-lasting (sometimes throughout life) specific protection in many but not all animals. The aim of this study was to determine the actual levels of seroprotection against CPV-2, CDV and CAdV-1 in a cohort of Italian dogs by using the in-practice test VacciCheck. A total of 1,027 dogs (951 vaccinated and 76 unvaccinated) were analyzed for Protective Antibody Titers (PATs) against CPV-2, CDV, and CAdV-1. Differences related to sex, age, breed size, health status, and time elapsed since last vaccination were evaluated. Half of the entire canine cohort (50.6%) had PATs for all three viruses (68.5% considering only vaccinated dogs). In particular, 90.8% of dogs were protected against CPV-2, 68.6% against CDV, and 79.8% against CAdV-1. Most dogs remained protected for 3 years after vaccination or longer. Revaccination on a 3-year basis can then be recommended for core MLV vaccines without altering individual's seroprotection or even herd immunity.

6.
Cancer Immunol Res ; 11(3): 351-363, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36574610

RESUMO

Nitric oxide (NO) is a signaling molecule produced by NO synthases (NOS1-3) to control processes such as neurotransmission, vascular permeability, and immune function. Although myeloid cell-derived NO has been shown to suppress T-cell responses, the role of NO synthesis in T cells themselves is not well understood. Here, we showed that significant amounts of NO were synthesized in human and murine CD8+ T cells following activation. Tumor growth was significantly accelerated in a T cell-specific, Nos2-null mouse model. Genetic deletion of Nos2 expression in murine T cells altered effector differentiation, reduced tumor infiltration, and inhibited recall responses and adoptive cell transfer function. These data show that endogenous NO production plays a critical role in T cell-mediated tumor immunity.


Assuntos
Neoplasias , Óxido Nítrico , Animais , Camundongos , Humanos , Óxido Nítrico Sintase Tipo II/genética , Camundongos Knockout , Neoplasias/genética , Linfócitos T CD8-Positivos
7.
Front Immunol ; 13: 837669, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35251031

RESUMO

Targeting T cell metabolism is an established method of immunomodulation. Following activation, T cells engage distinct metabolic programs leading to the uptake and processing of nutrients that determine cell proliferation and differentiation. Redirection of T cell fate by modulation of these metabolic programs has been shown to boost or suppress immune responses in vitro and in vivo. Using publicly available T cell transcriptomic and proteomic datasets we identified vitamin B6-dependent transaminases as key metabolic enzymes driving T cell activation and differentiation. Inhibition of vitamin B6 metabolism using the pyridoxal 5'-phosphate (PLP) inhibitor, aminoxyacetic acid (AOA), suppresses CD8+ T cell proliferation and effector differentiation in a dose-dependent manner. We show that pyridoxal phosphate phosphatase (PDXP), a negative regulator of intracellular vitamin B6 levels, is under the control of the hypoxia-inducible transcription factor (HIF1), a central driver of T cell metabolism. Furthermore, by adoptive transfer of CD8 T cells into a C57BL/6 mouse melanoma model, we demonstrate the requirement for vitamin B6-dependent enzyme activity in mediating effective anti-tumor responses. Our findings show that vitamin B6 metabolism is required for CD8+ T cell proliferation and effector differentiation in vitro and in vivo. Targeting vitamin B6 metabolism may therefore serve as an immunodulatory strategy to improve anti-tumor immunotherapy.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Vitamina B 6 , Ácido Amino-Oxiacético/farmacologia , Animais , Linfócitos T CD8-Positivos/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Fosfoproteínas Fosfatases , Proteômica , Fosfato de Piridoxal/antagonistas & inibidores , Vitamina B 6/metabolismo
8.
Front Immunol ; 12: 633586, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054802

RESUMO

Myeloid cell interactions with cells of the adaptive immune system are an essential aspect of immunity. A key aspect of that interrelationship is its modulation by the microenvironment. Oxygen is known to influence myelosuppression of T cell activation in part via the Hypoxia inducible (HIF) transcription factors. A number of drugs that act on the HIF pathway are currently in clinical use and it is important to evaluate how they act on immune cell function as part of a better understanding of how they will influence patient outcomes. We show here that increased activation of the HIF pathway, either through deletion of the negative regulator of HIF, the von Hippel-Lindau (VHL) gene, in myeloid cells, or through pharmacological inhibitors of VHL-mediated degradation of HIF, potently suppresses T cell proliferation in myeloid cell/T cell culture. These data demonstrate that both pharmacological and genetic activation of HIF in myeloid cells can suppress adaptive cell immune response.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glicina/análogos & derivados , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isoquinolinas/farmacologia , Macrófagos/efeitos dos fármacos , Oxigênio/metabolismo , Imunidade Adaptativa , Animais , Linfócitos T CD8-Positivos/fisiologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Técnicas de Cocultura , Glicina/farmacologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Camundongos , Camundongos Transgênicos , Oxigênio/farmacologia , Proteína Supressora de Tumor Von Hippel-Lindau/genética
9.
Elife ; 92020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-33095157

RESUMO

Exercise has a wide range of systemic effects. In animal models, repeated exertion reduces malignant tumor progression, and clinically, exercise can improve outcome for cancer patients. The etiology of the effects of exercise on tumor progression are unclear, as are the cellular actors involved. We show here that in mice, exercise-induced reduction in tumor growth is dependent on CD8+ T cells, and that metabolites produced in skeletal muscle and excreted into plasma at high levels during exertion in both mice and humans enhance the effector profile of CD8+ T-cells. We found that activated murine CD8+ T cells alter their central carbon metabolism in response to exertion in vivo, and that immune cells from trained mice are more potent antitumor effector cells when transferred into tumor-bearing untrained animals. These data demonstrate that CD8+ T cells are metabolically altered by exercise in a manner that acts to improve their antitumoral efficacy.


Exercise affects almost all tissues in the body, and scientists have found that being physically active can reduce the risk of several types of cancer as well as improving outcomes for cancer patients. However, it is still unknown how exercise exerts its protective effects. One of the hallmarks of cancer is the ability of cancer cells to evade detection by the immune system, which can in some cases stop the body from eliminating tumor cells. Rundqvist et al. used mice to investigate how exercise helps the immune system act against tumor progression. They found that when mice exercised, tumor growth was reduced, and this decrease in growth depended on the levels of a specific type of immune cell, the CD8+ T cell, circulating in the blood. Additionally, Rundqvist et al. found that CD8+ T cells were made more effective by molecules that muscles released into the blood during exercise. Isolating immune cells after intense exercise showed that these super-effective CD8+ T cells alter how they use molecules for energy production after exertion. Next, immune cells from mice that had exercised frequently were transferred into mice that had not exercised, where they were more effective against tumor cells than the immune cells from untrained mice. These results demonstrate that CD8+ T cells are altered by exercise to improve their effectiveness against tumors. The ability of T cells to identify and eliminate cancer cells is essential to avoid tumor growth, and is one of the foundations of current immune therapy treatments. Exercise could improve the outcome of these treatments by increasing the activation of the immune system, making tumor-fighting cells more effective.


Assuntos
Carcinogênese , Condicionamento Físico Animal , Linfócitos T Citotóxicos/fisiologia , Animais , Linhagem Celular Tumoral , Feminino , Camundongos
10.
Blood Adv ; 4(18): 4483-4493, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32941648

RESUMO

Cancer immunotherapy is advancing rapidly and gene-modified T cells expressing chimeric antigen receptors (CARs) show particular promise. A challenge of CAR-T cell therapy is that the ex vivo-generated CAR-T cells become exhausted during expansion in culture, and do not persist when transferred back to patients. It has become clear that naive and memory CD8 T cells perform better than the total CD8 T-cell populations in CAR-T immunotherapy because of better expansion, antitumor activity, and persistence, which are necessary features for therapeutic success and prevention of disease relapse. However, memory CAR-T cells are rarely used in the clinic due to generation challenges. We previously reported that mouse CD8 T cells cultured with the S enantiomer of the immunometabolite 2-hydroxyglutarate (S-2HG) exhibit enhanced antitumor activity. Here, we show that clinical-grade human donor CAR-T cells can be generated from naive precursors after culture with S-2HG. S-2HG-treated CAR-T cells establish long-term memory cells in vivo and show superior antitumor responses when compared with CAR-T cells generated with standard clinical protocols. This study provides the basis for a phase 1 clinical trial evaluating the activity of S-2HG-treated CD19-CAR-T cells in patients with B-cell malignancies.


Assuntos
Receptores de Antígenos de Linfócitos T , Receptores de Antígenos Quiméricos , Animais , Linfócitos T CD8-Positivos , Glutaratos , Humanos , Imunoterapia Adotiva , Camundongos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética
11.
Cell Death Dis ; 10(11): 857, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31719530

RESUMO

Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by dopaminergic neuronal loss that initiates in the substantia nigra pars compacta and by the formation of intracellular inclusions mainly constituted by aberrant α-synuclein (α-syn) deposits known as Lewy bodies. Most cases of PD are sporadic, but about 10% are familial, among them those caused by mutations in SNCA gene have an autosomal dominant transmission. SNCA encodes α-syn, a small 140-amino acids protein that, under physiological conditions, is mainly localized at the presynaptic terminals. It is prevalently cytosolic, but its presence has been reported in the nucleus, in the mitochondria and, more recently, in the mitochondria-associated ER membranes (MAMs). Whether different cellular localizations may reflect specific α-syn activities is presently unclear and its action at mitochondrial level is still a matter of debate. Mounting evidence supports a role for α-syn in several mitochondria-derived activities, among which maintenance of mitochondrial morphology and modulation of complex I and ATP synthase activity. α-syn has been proposed to localize at the outer membrane (OMM), in the intermembrane space (IMS), at the inner membrane (IMM) and in the mitochondrial matrix, but a clear and comparative analysis of the sub-mitochondrial localization of WT and mutant α-syn is missing. Furthermore, the reasons for this spread sub-mitochondrial localization under physiological and pathological circumstances remain elusive. In this context, we decided to selectively monitor the sub-mitochondrial distribution of the WT and PD-related α-syn mutants A53T and A30P by taking advantage from a bimolecular fluorescence complementation (BiFC) approach. We also investigated whether cell stress could trigger α-syn translocation within the different mitochondrial sub-compartments and whether PD-related mutations could impinge on it. Interestingly, the artificial targeting of α-syn WT (but not of the mutants) to the mitochondrial matrix impacts on ATP production, suggesting a potential role within this compartment.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Mitocôndrias/genética , Doença de Parkinson/metabolismo , alfa-Sinucleína/genética , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/genética , Citosol/metabolismo , Citosol/patologia , Dopamina/genética , Dopamina/metabolismo , Neurônios Dopaminérgicos/patologia , Expressão Gênica/genética , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , ATPases Mitocondriais Próton-Translocadoras/genética , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Proteínas Mutantes/genética , Doença de Parkinson/patologia , Parte Compacta da Substância Negra/metabolismo , Parte Compacta da Substância Negra/patologia , Terminações Pré-Sinápticas/metabolismo
12.
Sci Transl Med ; 9(416)2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29141887

RESUMO

The immunosuppressive activity of mesenchymal stromal cells (MSCs) is well documented. However, the therapeutic benefit is completely unpredictable, thus raising concerns about MSC efficacy. One of the affecting factors is the unresolved conundrum that, despite being immunosuppressive, MSCs are undetectable after administration. Therefore, understanding the fate of infused MSCs could help predict clinical responses. Using a murine model of graft-versus-host disease (GvHD), we demonstrate that MSCs are actively induced to undergo perforin-dependent apoptosis by recipient cytotoxic cells and that this process is essential to initiate MSC-induced immunosuppression. When examining patients with GvHD who received MSCs, we found a striking parallel, whereby only those with high cytotoxic activity against MSCs responded to MSC infusion, whereas those with low activity did not. The need for recipient cytotoxic cell activity could be replaced by the infusion of apoptotic MSCs generated ex vivo. After infusion, recipient phagocytes engulf apoptotic MSCs and produce indoleamine 2,3-dioxygenase, which is ultimately necessary for effecting immunosuppression. Therefore, we propose the innovative concept that patients should be stratified for MSC treatment according to their ability to kill MSCs or that all patients could be treated with ex vivo apoptotic MSCs.


Assuntos
Apoptose/fisiologia , Células-Tronco Mesenquimais/citologia , Animais , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/fisiopatologia , Humanos , Imunomodulação/genética , Imunomodulação/fisiologia , Terapia de Imunossupressão/métodos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA