Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nature ; 540(7632): 236-241, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27798602

RESUMEN

R-2-hydroxyglutarate accumulates to millimolar levels in cancer cells with gain-of-function isocitrate dehydrogenase 1/2 mutations. These levels of R-2-hydroxyglutarate affect 2-oxoglutarate-dependent dioxygenases. Both metabolite enantiomers, R- and S-2-hydroxyglutarate, are detectible in healthy individuals, yet their physiological function remains elusive. Here we show that 2-hydroxyglutarate accumulates in mouse CD8+ T cells in response to T-cell receptor triggering, and accumulates to millimolar levels in physiological oxygen conditions through a hypoxia-inducible factor 1-alpha (HIF-1α)-dependent mechanism. S-2-hydroxyglutarate predominates over R-2-hydroxyglutarate in activated T cells, and we demonstrate alterations in markers of CD8+ T-cell differentiation in response to this metabolite. Modulation of histone and DNA demethylation, as well as HIF-1α stability, mediate these effects. S-2-hydroxyglutarate treatment greatly enhances the in vivo proliferation, persistence and anti-tumour capacity of adoptively transferred CD8+ T cells. Thus, S-2-hydroxyglutarate acts as an immunometabolite that links environmental context, through a metabolic-epigenetic axis, to immune fate and function.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Glutaratos/farmacología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , ADN/química , ADN/metabolismo , Metilación de ADN/efectos de los fármacos , Dioxigenasas/metabolismo , Glutaratos/inmunología , Glutaratos/metabolismo , Histonas/metabolismo , Homeostasis/efectos de los fármacos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ácidos Cetoglutáricos/metabolismo , Activación de Linfocitos , Lisina/metabolismo , Ratones , Oxígeno/metabolismo , Estabilidad Proteica , Receptores de Antígenos de Linfocitos T/inmunología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
2.
J Immunol ; 194(3): 1080-9, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25539815

RESUMEN

Ag receptors used for cancer immunotherapy are often directed against tumor-associated Ags also expressed in normal tissues. Targeting of such Ags can result in unwanted autoimmune attack of normal tissues or induction of tolerance in therapeutic T cells. We used a murine model to study the phenotype and function of T cells redirected against the murine double minute protein 2 (MDM2), a tumor-associated Ag that shows low expression in many normal tissues. Transfer of MDM2-TCR-engineered T cells into bone marrow chimeric mice revealed that Ag recognition in hematopoietic tissues maintained T cell function, whereas presentation of MDM2 in nonhematopoietic tissues caused reduced effector function. TCR-engineered CD8(+) T cells underwent rapid turnover, downmodulated CD8 expression, and lost cytotoxic function. We found that MDM2-TCR-engineered CD4(+) T cells provided help and restored cytotoxic function of CD8(+) T cells bearing the same TCR. Although the introduction of the CD8 coreceptor enhanced the ability of CD4(+) T cells to recognize MDM2 in vitro, the improved self-antigen recognition abolished their ability to provide helper function in vivo. The data indicate that the same class I-restricted TCR responsible for Ag recognition and tolerance induction in CD8(+) T cells can, in the absence of the CD8 coreceptor, elicit CD4 T cell help and partially reverse tolerance. Thus MHC class I-restricted CD4(+) T cells may enhance the efficacy of therapeutic TCR-engineered CD8(+) T cells and can be readily generated with the same TCR.


Asunto(s)
Antígenos de Neoplasias/inmunología , Autoantígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Tolerancia Inmunológica , Receptores de Antígenos de Linfocitos T/genética , Traslado Adoptivo , Animales , Comunicación Celular , Citotoxicidad Inmunológica , Expresión Génica , Inmunofenotipificación , Ratones , Ratones Transgénicos , Fenotipo , Proteínas Proto-Oncogénicas c-mdm2/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción Genética
3.
Haematologica ; 101(4): 482-90, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26802053

RESUMEN

Due to the lack of specificity for tumor antigens, allogeneic T-cell therapy is associated with graft-versus-host disease. Enhancing the anti-tumor specificity while reducing the graft-versus-host disease risk of allogeneic T cells has remained a research focus. In this study, we demonstrate that the introduction of 'dominant' T-cell receptors into primary murine T cells can suppress the expression of endogenous T-cell receptors in a large proportion of the gene-modified T cells. Adoptive transfer of allogeneic T cells expressing a 'dominant' T-cell receptor significantly reduced the graft-versus-host toxicity in recipient mice. Using two bone marrow transplant models, enhanced anti-tumor activity was observed in the presence of reduced graft-versus-host disease. However, although transfer of T-cell receptor gene-modified allogeneic T cells resulted in the elimination of antigen-positive tumor cells and improved the survival of treated mice, it was associated with accumulation of T cells expressing endogenous T-cell receptors and the development of delayed graft-versus-host disease. The in-vivo deletion of the engineered T cells, mediated by endogenous mouse mammary tumor virus MTV8 and MTV9, abolished graft-versus-host disease while retaining significant anti-tumor activity of adoptively transferred T cells. Together, this study shows that the in-vitro selection of allogeneic T cells expressing high levels of a 'dominant' T-cell receptor can lower acute graft-versus-host disease and enhance anti-tumor activity of adoptive cell therapy, while the in-vivo outgrowth of T cells expressing endogenous T-cell receptors remains a risk factor for the delayed onset of graft-versus-host disease.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Inmunoterapia Adoptiva/métodos , Virus del Tumor Mamario del Ratón/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/trasplante , Animales , Trasplante de Médula Ósea/métodos , Línea Celular Tumoral , Femenino , Expresión Génica , Genes Dominantes , Vectores Genéticos/inmunología , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Depleción Linfocítica/métodos , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Receptores de Antígenos de Linfocitos T/genética , Análisis de Supervivencia , Linfocitos T/citología , Linfocitos T/inmunología , Transgenes , Trasplante Homólogo , Irradiación Corporal Total
4.
Front Immunol ; 15: 1293723, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690263

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Ratones Noqueados , Animales , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Activación de Linfocitos/inmunología , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Ratones Endogámicos C57BL
5.
Front Immunol ; 14: 1101433, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36923405

RESUMEN

Introduction: CD8+ T cells infiltrate virtually every tissue to find and destroy infected or mutated cells. They often traverse varying oxygen levels and nutrient-deprived microenvironments. High glycolytic activity in local tissues can result in significant exposure of cytotoxic T cells to the lactate metabolite. Lactate has been known to act as an immunosuppressor, at least in part due to its association with tissue acidosis. Methods: To dissect the role of the lactate anion, independently of pH, we performed phenotypical and metabolic assays, high-throughput RNA sequencing, and mass spectrometry, on primary cultures of murine or human CD8+ T cells exposed to high doses of pH-neutral sodium lactate. Results: The lactate anion is well tolerated by CD8+ T cells in pH neutral conditions. We describe how lactate is taken up by activated CD8+ T cells and can displace glucose as a carbon source. Activation in the presence of sodium lactate significantly alters the CD8+ T cell transcriptome, including the expression key effector differentiation markers such as granzyme B and interferon-gamma. Discussion: Our studies reveal novel metabolic features of lactate utilization by activated CD8+ T cells, and highlight the importance of lactate in shaping the differentiation and activity of cytotoxic T cells.


Asunto(s)
Ácido Láctico , Transcriptoma , Ratones , Humanos , Animales , Ácido Láctico/metabolismo , Lactato de Sodio/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T Citotóxicos/metabolismo
6.
Cancer Immunol Res ; 11(3): 351-363, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36574610

RESUMEN

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.


Asunto(s)
Neoplasias , Óxido Nítrico , Animales , Ratones , Humanos , Óxido Nítrico Sintasa de Tipo II/genética , Ratones Noqueados , Neoplasias/genética , Linfocitos T CD8-positivos
7.
Elife ; 122023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37166103

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos , Oxígeno , Ratones , Humanos , Animales , Oxígeno/metabolismo , Transducción de Señal , Activación de Linfocitos , Traslado Adoptivo
8.
Nat Metab ; 5(10): 1747-1764, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37605057

RESUMEN

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.


Asunto(s)
Fenómenos Bioquímicos , Linfocitos T CD8-positivos , Linfocitos T CD8-positivos/metabolismo , Glutaratos/metabolismo
9.
Eur J Immunol ; 41(9): 2530-4, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21952812

RESUMEN

"Faster, better, more" is the conventional benchmark used to define responses of memory T cells when compared with their naïve counterparts. In this issue of the European Journal of Immunology, Mark and Warren Shlomchik and colleagues [Eur. J. Immunol. 2011. 41: 2782-2792] make the intriguing observation that murine memory CD4(+) T-cell populations enriched for alloreactive precursors are fully capable of rejecting allogeneic skin grafts but yet are incapable of inducing significant graft-versus-host disease. These observations add to the emerging concept that memory CD4(+) T-cell development is more nuanced and complex than predicted by conventional models. In particular, the data suggest that it may be just as important to consider what naïve or effector cells have "lost" in their transition to memory.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas , Complicaciones Posoperatorias , Subgrupos de Linfocitos T/metabolismo , Animales , Humanos
10.
Front Immunol ; 13: 837669, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35251031

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Vitamina B 6 , Ácido Aminooxiacético/farmacología , Animales , Linfocitos T CD8-positivos/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Fosfoproteínas Fosfatasas , Proteómica , Fosfato de Piridoxal/antagonistas & inhibidores , Vitamina B 6/metabolismo
11.
Muscle Nerve ; 44(3): 366-70, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21996796

RESUMEN

INTRODUCTION: C2C12 myoblasts undergo in vitro myogenesis to form protein-rich multinucleated myotubes. Determining the fraction of total nuclei incorporated into myotubes is a commonly used method to quantify the extent of differentiation, but it is labor-intensive and susceptible to operator bias. METHODS: We have developed a simple method to quantify myotube formation using micrographs of Jenner-Giemsa-stained C2C12 cultures. Because myotubes are darkly stained by Jenner-Giemsa dyes, the extent of myotube formation correlates with an increase in pixels attributed to the darkest tones. Thus, image histograms were obtained from photographs using ImageJ software, and the sum of the darkest tones was used as a measure of myotube density. RESULTS: Measurements of myotube density mirrored those of fusion index during C2C12 differentiation and after treatment with prostaglandin D(2) , an inhibitor of C2C12 myogenesis. CONCLUSIONS: We propose this inexpensive, quick, and unbiased method to quantify C2C12 differentiation as a complement of the fusion index analysis.


Asunto(s)
Diferenciación Celular/fisiología , Procesamiento de Imagen Asistido por Computador/métodos , Microscopía/métodos , Desarrollo de Músculos/fisiología , Mioblastos Esqueléticos/citología , Animales , Línea Celular , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Mioblastos Esqueléticos/fisiología , Programas Informáticos
12.
Front Immunol ; 12: 633586, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054802

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Glicina/análogos & derivados , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isoquinolinas/farmacología , Macrófagos/efectos de los fármacos , Oxígeno/metabolismo , Inmunidad Adaptativa , Animales , Linfocitos T CD8-positivos/fisiología , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Técnicas de Cocultivo , Glicina/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Ratones , Ratones Transgénicos , Oxígeno/farmacología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
13.
Cancer Immunol Res ; 9(4): 401-414, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33602720

RESUMEN

Adoptive transfer of antitumor cytotoxic T cells is an emerging form of cancer immunotherapy. A key challenge to expanding the utility of adoptive cell therapies is how to enhance the survival and function of the transferred T cells. Immune-cell survival requires adaptation to different microenvironments and particularly to the hypoxic milieu of solid tumors. The hypoxia-inducible factor (HIF) transcription factors are an essential aspect of this adaptation. In this study, we undertook experiments to define structural determinants of HIF that potentiate antitumor efficacy in cytotoxic T cells. We first created retroviral vectors to deliver ectopic expression of HIF1α and HIF2α in mouse CD8+ T cells, together or individually and with or without sensitivity to the oxygen-dependent HIFα inhibitors Von Hippel-Lindau and factor-inhibiting HIF (FIH). HIF2α, but not HIF1α, drove broad transcriptional changes in CD8+ T cells, resulting in increased cytotoxic differentiation and cytolytic function against tumor targets. A specific mutation replacing the hydroxyl group-acceptor site for FIH in HIF2α gave rise to the most effective antitumor T cells after adoptive transfer in vivo In addition, codelivering an FIH-insensitive form of HIF2α with an anti-CD19 chimeric antigen receptor greatly enhanced cytolytic function of human CD8+ T cells against lymphoma cells both in vitro and in a xenograft adoptive transfer model. These experiments point to a means to increase the antitumor efficacy of therapeutic CD8+ T cells via ectopic expression of the HIF transcription factor.See related Spotlight on p. 364.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Linfocitos T CD8-positivos/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Hipoxia/inmunología , Inmunoterapia Adoptiva , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Línea Celular Tumoral , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción , Microambiente Tumoral
14.
Acta Physiol (Oxf) ; 229(2): e13461, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32129933

RESUMEN

AIM: High-altitude pulmonary oedema (HAPE) is a non-cardiogenic pulmonary oedema that can occur during rapid ascent to a high-altitude environment. Classically, HAPE has been described as a condition resulting from a combination of pulmonary vasoconstriction and hypertension. Inflammation has been described as important in HAPE, although as a side effect of pulmonary oedema rather than as a causative factor. In this study, we aim to understand the role of hypoxic response in myeloid cells and its involvement in pathogenesis of HAPE. METHODS: We have generated a conditional deletion in mice of the von Hippel-Lindau factor (VHL) in myeloid cells to determine the effect of a deregulated hypoxic response in pulmonary oedema. RESULTS: The deletion of VHL in pulmonary myeloid cells gave rise to pulmonary oedema, increased pulmonary vascular permeability and reduced performance during exertion. These changes were accompanied by reduced stroke volume in the left ventricle. CONCLUSION: In this model, we show that a deregulated myeloid cell hypoxic response can trigger some of the most important symptoms of HAPE, and thus mice with a deletion of VHL in the myeloid lineage can function as a model of HAPE.


Asunto(s)
Mal de Altura , Altitud , Hipoxia , Células Mieloides , Edema Pulmonar , Animales , Modelos Animales de Enfermedad , Femenino , Hipertensión Pulmonar , Masculino , Ratones
15.
Blood Adv ; 4(18): 4483-4493, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32941648

RESUMEN

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.


Asunto(s)
Receptores de Antígenos de Linfocitos T , Receptores Quiméricos de Antígenos , Animales , Linfocitos T CD8-positivos , Glutaratos , Humanos , Inmunoterapia Adoptiva , Ratones , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética
16.
Elife ; 92020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-33095157

RESUMEN

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.


Asunto(s)
Carcinogénesis , Condicionamiento Físico Animal , Linfocitos T Citotóxicos/fisiología , Animales , Línea Celular Tumoral , Femenino , Ratones
17.
Mol Cancer ; 8: 121, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-20003443

RESUMEN

BACKGROUND: Over recent years, enzymes of the aldo-keto reductase (AKR) 1C subfamily have been implicated in the progression of prostate, breast, endometrial and leukemic cancers. This is due to the ability of AKR1C enzymes to modify androgens, estrogens, progesterone and prostaglandins (PGs) in a tissue-specific manner, regulating the activity of nuclear receptors and other downstream effects. Evidence supporting a role for AKR1C enzymes in cancer derives mostly from studies with isolated primary cells from patients or immortalized cell lines. Mice are ideal organisms for in vivo studies, using knock-out or over-expression strains. However, the functional conservation of AKR1C enzymes between human and mice has yet to be described. RESULTS: In this study, we have characterized and compared the four human (AKR1C1,-1C2, -1C3 and -1C4) and the eight murine (AKR1C6, -1C12, -1C13, -1C14, -1C18, -1C19, -1C20 and -1C21) isoforms in their phylogeny, substrate preference and tissue distribution. We have found divergent evolution between human and murine AKR1C enzymes that was reflected by differing substrate preference. Murine enzymes did not perform the 11beta-ketoreduction of prostaglandin (PG) D2, an activity specific to human AKR1C3 and important in promoting leukemic cell survival. Instead, murine AKR1C6 was able to perform the 9-ketoreduction of PGE2, an activity absent amongst human isoforms. Nevertheless, reduction of the key steroids androstenedione, 5alpha-dihydrotestosterone, progesterone and estrone was found in murine isoforms. However, unlike humans, no AKR1C isoforms were detected in murine prostate, testes, uterus and haemopoietic progenitors. CONCLUSIONS: This study exposes significant lack of phylogenetic and functional homology between human and murine AKR1C enzymes. Therefore, we conclude that mice are not suitable to model the role of AKR1C in human cancers and leukemia.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Modelos Biológicos , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Aldehído Reductasa , Aldo-Ceto Reductasas , Animales , Humanos , Ratones , Filogenia , Prostaglandinas/metabolismo , Especificidad de la Especie , Especificidad por Sustrato
18.
Oncoimmunology ; 8(3): 1542917, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30723575

RESUMEN

We explored whether engineering of T cell specificity and effector function improves immunotherapy of solid tumors. Although IL-12 can enhance cancer immunity, a strategy of safe IL-12 delivery without toxicity is currently lacking. We engineered T cells to express IL-12 controlled by the NFAT promoter responsive to TCR stimulation, or by the Tet-On promoter responsive to doxycycline. In vivo, NFAT-engineered T cells caused lethal toxicity, while Tet-engineered T cells were safe in the absence of doxycycline. Combining gene transfer of the melanoma-specific TRP2-TCR with Tet-IL-12 engineering revealed that temporal induction of IL-12 was essential to inhibit the growth of B16F10 melanoma tumors. Induced IL-12 increased the number of tumor-infiltrating T cells and also prevented the down-modulation of the TRP2-TCR and the associated up-regulation of the PD1 marker that was observed in the absence of IL-12. In addition, temporal induction of IL-12 expression also increased the number of plasmacytoid DC in the tumor micro-environment. We show that repeated induction of IL-12 can be used to enhance control of tumor growth without encountering systemic toxicity. The observation that TCR engineering combined with Tet-regulated IL-12 expression can achieve tumor immunity without the side effects that are usually associated with the in vivo use of IL-12 warrants translation of this concept into the clinic.

19.
J Muscle Res Cell Motil ; 29(6-8): 163-7, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19052883

RESUMEN

Muscle regeneration is a tightly orchestrated process where activated satellite cells (myoblasts) respond to external stimuli in order to proliferate, differentiate and fuse to damaged myofibers. Simultaneously, the injured tissue undergoes an inflammatory response and communication between leukocytes and the spectrum of differentiated and undifferentiated muscle cells is essential for proper healing. This communication is mediated by cytokines, growth factors and prostaglandins and dissecting the role of these signaling molecules might be the key to positively manipulate muscle regeneration in the future. This review will focus on the roles of prostaglandins and will consider the potential cost of using non-steroidal inflammatory drugs as popular treatment of muscle injury.


Asunto(s)
Músculo Esquelético/fisiología , Prostaglandinas/fisiología , Regeneración/fisiología , Animales , Humanos , Músculo Esquelético/citología
20.
J Clin Invest ; 128(5): 2010-2024, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29485974

RESUMEN

A key predictor for the success of gene-modified T cell therapies for cancer is the persistence of transferred cells in the patient. The propensity of less differentiated memory T cells to expand and survive efficiently has therefore made them attractive candidates for clinical application. We hypothesized that redirecting T cells to specialized niches in the BM that support memory differentiation would confer increased therapeutic efficacy. We show that overexpression of chemokine receptor CXCR4 in CD8+ T cells (TCXCR4) enhanced their migration toward vascular-associated CXCL12+ cells in the BM and increased their local engraftment. Increased access of TCXCR4 to the BM microenvironment induced IL-15-dependent homeostatic expansion and promoted the differentiation of memory precursor-like cells with low expression of programmed death-1, resistance to apoptosis, and a heightened capacity to generate polyfunctional cytokine-producing effector cells. Following transfer to lymphoma-bearing mice, TCXCR4 showed a greater capacity for effector expansion and better tumor protection, the latter being independent of changes in trafficking to the tumor bed or local out-competition of regulatory T cells. Thus, redirected homing of T cells to the BM confers increased memory differentiation and antitumor immunity, suggesting an innovative solution to increase the persistence and functions of therapeutic T cells.


Asunto(s)
Médula Ósea/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Memoria Inmunológica , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Animales , Médula Ósea/patología , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Humanos , Interleucina-15/genética , Interleucina-15/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Linfocitos T Reguladores/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA