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










Base de datos
Intervalo de año de publicación
1.
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
2.
Cell Rep ; 42(9): 113013, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37632752

RESUMEN

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.


Asunto(s)
Neoplasias , Linfocitos T Citotóxicos , Humanos , Linfocitos T Citotóxicos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Glutaratos/metabolismo , Neoplasias/metabolismo , Isocitrato Deshidrogenasa
3.
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
4.
Curr Opin Biotechnol ; 83: 102976, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37515937

RESUMEN

2-hydroxyglutarate (2HG) is a biproduct of the Krebs cycle, which exists in a D- and L- enantiomer and is structurally similar to α-ketoglutarate. Both 2HG enantiomers have been described to accumulate in diverse cancer and immune cells and can influence cell fate and function. While D-2HG was originally considered as an 'oncometabolite' that aberrantly builds up in certain cancers, it is becoming clear that it also physiologically accumulates in immune cells and regulates immune function. Conversely, L-2HG is considered as an 'immunometabolite' due to its induction and regulatory function in T cells, but it can also be induced in certain cancers. Here, the authors review the effects of both 2HG enantiomers on immune cells within the tumor microenvironment.


Asunto(s)
Neoplasias , Humanos , Glutaratos , Ácidos Cetoglutáricos , Estereoisomerismo , Mutación , Microambiente Tumoral
5.
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
6.
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
7.
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
8.
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
9.
Nat Commun ; 12(1): 3686, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34140498

RESUMEN

Tumour hypoxia is associated with poor patient prognosis and therapy resistance. A unique transcriptional response is initiated by hypoxia which includes the rapid activation of numerous transcription factors in a background of reduced global transcription. Here, we show that the biological response to hypoxia includes the accumulation of R-loops and the induction of the RNA/DNA helicase SETX. In the absence of hypoxia-induced SETX, R-loop levels increase, DNA damage accumulates, and DNA replication rates decrease. Therefore, suggesting that, SETX plays a role in protecting cells from DNA damage induced during transcription in hypoxia. Importantly, we propose that the mechanism of SETX induction in hypoxia is reliant on the PERK/ATF4 arm of the unfolded protein response. These data not only highlight the unique cellular response to hypoxia, which includes both a replication stress-dependent DNA damage response and an unfolded protein response but uncover a novel link between these two distinct pathways.


Asunto(s)
Hipoxia de la Célula , Daño del ADN/genética , ADN Helicasas/metabolismo , Regulación de la Expresión Génica/genética , Enzimas Multifuncionales/metabolismo , Estructuras R-Loop/genética , ARN Helicasas/metabolismo , Respuesta de Proteína Desplegada/genética , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Inmunoprecipitación de Cromatina , ADN Helicasas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Enzimas Multifuncionales/genética , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Oxígeno/farmacología , Estructuras R-Loop/efectos de los fármacos , ARN Helicasas/genética , RNA-Seq , Respuesta de Proteína Desplegada/efectos de los fármacos , Regulación hacia Arriba , Cinostatina/farmacología , eIF-2 Quinasa/metabolismo
10.
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
11.
FEBS J ; 288(24): 7143-7161, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33410283

RESUMEN

Hypoxia has a significant impact on many physiological and pathological processes. Over the recent years, its role in modulation of epigenetic remodelling has also become clearer. In cancer, low oxygen environments and aberrant epigenomes often go hand in hand, and changes in DNA methylation are now commonly recognised as potential outcome indicators. TET (ten-eleven translocation) family enzymes are alpha-ketoglutarate-, iron- and oxygen-dependent DNA demethylases and are key players in these processes. Although TETs have historically been considered tumour suppressors, recent studies suggest that their functions in cancer might not be straightforward. Recently, inhibition of TETs has been reported to have positive impact in cancer immunotherapy and vaccination studies. This underlines the current interest in developing targeted pharmaceutical inhibitors of these enzymes. Here, we will survey the complexity of TET roles in cancer, and its hypoxic modulation, as well as highlight the potential of these enzymes as therapeutic targets.


Asunto(s)
Oxigenasas de Función Mixta/metabolismo , Neoplasias/enzimología , Oxígeno/metabolismo , Animales , Humanos , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/genética
12.
Cell Mol Life Sci ; 78(7): 3443-3465, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33247761

RESUMEN

During central nervous system (CNS) development, proper and timely induction of neurite elongation is critical for generating functional, mature neurons, and neuronal networks. Despite the wealth of information on the action of extracellular cues, little is known about the intrinsic gene regulatory factors that control this developmental decision. Here, we report the identification of Prox1, a homeobox transcription factor, as a key player in inhibiting neurite elongation. Although Prox1 promotes acquisition of early neuronal identity and is expressed in nascent post-mitotic neurons, it is heavily down-regulated in the majority of terminally differentiated neurons, indicating a regulatory role in delaying neurite outgrowth in newly formed neurons. Consistently, we show that Prox1 is sufficient to inhibit neurite extension in mouse and human neuroblastoma cell lines. More importantly, Prox1 overexpression suppresses neurite elongation in primary neuronal cultures as well as in the developing mouse brain, while Prox1 knock-down promotes neurite outgrowth. Mechanistically, RNA-Seq analysis reveals that Prox1 affects critical pathways for neuronal maturation and neurite extension. Interestingly, Prox1 strongly inhibits many components of Ca2+ signaling pathway, an important mediator of neurite extension and neuronal maturation. In accordance, Prox1 represses Ca2+ entry upon KCl-mediated depolarization and reduces CREB phosphorylation. These observations suggest that Prox1 acts as a potent suppressor of neurite outgrowth by inhibiting Ca2+ signaling pathway. This action may provide the appropriate time window for nascent neurons to find the correct position in the CNS prior to initiation of neurites and axon elongation.


Asunto(s)
Señalización del Calcio , Sistema Nervioso Central/patología , Proteínas de Homeodominio/metabolismo , Neuroblastoma/patología , Proyección Neuronal , Neuronas/patología , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Sistema Nervioso Central/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , Neuroblastoma/metabolismo , Neuronas/metabolismo , Fosforilación , Transducción de Señal , Proteínas Supresoras de Tumor/genética
13.
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
14.
FEBS J ; 285(9): 1563-1571, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29288533

RESUMEN

The term hypoxia refers to any condition where insufficient oxygen is available and therefore encompasses a range of actual oxygen concentrations. The regions of tumours adjacent to necrotic areas are at almost anoxic levels and are known to be extremely therapy resistant (radiobiological hypoxia). The biological response to radiobiological hypoxia includes the rapid accumulation of replication stress and subsequent DNA damage response, including both ATR- and ATM-mediated signalling, despite the absence of detectable DNA damage. The causes and consequences of hypoxia-induced replication stress will be discussed.


Asunto(s)
Hipoxia de la Célula/fisiología , Replicación del ADN , Animales , Proteínas de Ciclo Celular/fisiología , Daño del ADN , Reparación del ADN , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/fisiología , Desoxirribonucleótidos/metabolismo , Humanos , Neoplasias/genética , Oxígeno/farmacología , Ribonucleótido Reductasas/metabolismo , Estrés Fisiológico/genética , Microambiente Tumoral
15.
Mol Cell Oncol ; 4(5): e1335272, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29057303

RESUMEN

How tumor cells adapt and survive under hypoxia significantly impacts patient prognosis. We recently demonstrated that the oxygen-requiring ribonucleotide reductase (RNR) enzyme, which provides cells with deoxyribonucleotides, responds to limited oxygen availability by switching small subunits from RRM2 to RRM2B. This property of RNR is essential for hypoxic cell viability and therefore contributes to the most aggressive and therapy-resistant fraction of tumors.

16.
Mol Cell ; 66(2): 206-220.e9, 2017 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28416140

RESUMEN

Cells exposed to hypoxia experience replication stress but do not accumulate DNA damage, suggesting sustained DNA replication. Ribonucleotide reductase (RNR) is the only enzyme capable of de novo synthesis of deoxyribonucleotide triphosphates (dNTPs). However, oxygen is an essential cofactor for mammalian RNR (RRM1/RRM2 and RRM1/RRM2B), leading us to question the source of dNTPs in hypoxia. Here, we show that the RRM1/RRM2B enzyme is capable of retaining activity in hypoxia and therefore is favored over RRM1/RRM2 in order to preserve ongoing replication and avoid the accumulation of DNA damage. We found two distinct mechanisms by which RRM2B maintains hypoxic activity and identified responsible residues in RRM2B. The importance of RRM2B in the response to tumor hypoxia is further illustrated by correlation of its expression with a hypoxic signature in patient samples and its roles in tumor growth and radioresistance. Our data provide mechanistic insight into RNR biology, highlighting RRM2B as a hypoxic-specific, anti-cancer therapeutic target.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias del Colon/enzimología , Replicación del ADN , ADN de Neoplasias/biosíntesis , Oxígeno/metabolismo , Ribonucleótido Reductasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias del Colon/radioterapia , Daño del ADN , ADN de Neoplasias/genética , Femenino , Células HCT116 , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Interferencia de ARN , Tolerancia a Radiación , Ribonucleósido Difosfato Reductasa/metabolismo , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/genética , Factores de Tiempo , Transfección , Carga Tumoral , Hipoxia Tumoral , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Adv Exp Med Biol ; 899: 11-25, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27325259

RESUMEN

It is imperative that dividing cells maintain replication fork integrity in order to prevent DNA damage and cell death. The investigation of DNA replication is of high importance as alterations in this process can lead to genomic instability, a known causative factor of tumor development. A simple, sensitive, and informative technique which enables the study of DNA replication, is the DNA fiber assay, an adaptation of which is described in this chapter. The DNA fiber method is a powerful tool, which allows the quantitative and qualitative analysis of DNA replication at the single molecule level. The sequential pulse labeling of live cells with two thymidine analogues and the subsequent detection with specific antibodies and fluorescence imaging allows direct examination of sites of DNA synthesis. In this chapter, we describe how this assay can be performed in conditions of low oxygen levels (hypoxia)-a physiologically relevant stress that occurs in most solid tumors. Moreover, we suggest ways on how to overcome the technical problems that arise while using the hypoxic chambers.


Asunto(s)
Replicación del ADN , Coloración y Etiquetado/métodos , Hipoxia de la Célula , Línea Celular Tumoral , Humanos , Imagenología Tridimensional , Estadística como Asunto
18.
FEMS Yeast Res ; 15(8)2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26500234

RESUMEN

We have functionally characterized the four Saccharomyces cerevisiae (Sc) Jen1 homologues of Debaryomyces hansenii (Dh) by heterologous expression in S. cerevisiae. Debaryomyces hansenii cells display mediated transport for the uptake of lactate, acetate, succinate and malate. DHJEN genes expression was detected by RT-PCR in all carbon sources assayed, namely lactate, succinate, citrate, glycerol and glucose. The heterologous expression in the S. cerevisiae W303-1A jen1Δ ady2Δ strain demonstrated that the D. hansenii JEN genes encode four carboxylate transporters. DH27 gene encodes an acetate transporter (Km 0.94 ± 0.17 mM; Vmax 0.43 ± 0.03 nmol s(-1) mg(-1)), DH17 encodes a malate transporter (Km 0.27 ± 0.04 mM; Vmax 0.11 ± 0.01 nmol s(-1) mg(-1)) and both DH18 and DH24 encode succinate transporters with the following kinetic parameters, respectively, Km 0.31 ± 0.06 mM; Vmax 0.83 ± 0.04 nmol s(-1) mg(-1)and Km 0.16 ± 0.02 mM; Vmax 0.19 ± 0.02 nmol s(-1) mg(-1). Surprisingly, no lactate transporter was found, although D. hansenii presents a mediated transport for this acid. This work advanced the current knowledge on yeast carboxylate transporters by characterizing four new plasma membrane transporters in D. hansenii.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Saccharomycetales/genética , Saccharomycetales/metabolismo , Transporte Biológico , Ácidos Carboxílicos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
19.
J Clin Invest ; 125(6): 2385-98, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25961455

RESUMEN

Restoration of hypoxia-induced apoptosis in tumors harboring p53 mutations has been proposed as a potential therapeutic strategy; however, the transcriptional targets that mediate hypoxia-induced p53-dependent apoptosis remain elusive. Here, we demonstrated that hypoxia-induced p53-dependent apoptosis is reliant on the DNA-binding and transactivation domains of p53 but not on the acetylation sites K120 and K164, which, in contrast, are essential for DNA damage-induced, p53-dependent apoptosis. Evaluation of hypoxia-induced transcripts in multiple cell lines identified a group of genes that are hypoxia-inducible proapoptotic targets of p53, including inositol polyphosphate-5-phosphatase (INPP5D), pleckstrin domain-containing A3 (PHLDA3), sulfatase 2 (SULF2), B cell translocation gene 2 (BTG2), cytoplasmic FMR1-interacting protein 2 (CYFIP2), and KN motif and ankyrin repeat domains 3 (KANK3). These targets were also regulated by p53 in human cancers, including breast, brain, colorectal, kidney, bladder, and melanoma cancers. Downregulation of these hypoxia-inducible targets associated with poor prognosis, suggesting that hypoxia-induced apoptosis contributes to p53-mediated tumor suppression and treatment response. Induction of p53 targets, PHLDA3, and a specific INPP5D transcript mediated apoptosis in response to hypoxia through AKT inhibition. Moreover, pharmacological inhibition of AKT led to apoptosis in the hypoxic regions of p53-deficient tumors and consequently increased radiosensitivity. Together, these results identify mediators of hypoxia-induced p53-dependent apoptosis and suggest AKT inhibition may improve radiotherapy response in p53-deficient tumors.


Asunto(s)
Apoptosis , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tolerancia a Radiación , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Hipoxia de la Célula/genética , Línea Celular Tumoral , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Inositol Polifosfato 5-Fosfatasas , Neoplasias/genética , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Sulfatasas , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
20.
Mol Cell ; 52(5): 758-66, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24268576

RESUMEN

ATM-mediated signaling in response to DNA damage is a barrier to tumorigenesis. Here we asked whether replication stress could also contribute to ATM signaling. We demonstrate that, in the absence of DNA damage, ATM responds to replication stress in a hypoxia-induced heterochromatin-like context. In certain hypoxic conditions, replication stress occurs in the absence of detectable DNA damage. Hypoxia also induces H3K9me3, a histone modification associated with gene repression and heterochromatin. Hypoxia-induced replication stress together with increased H3K9me3 leads to ATM activation. Importantly, ATM prevents the accumulation of DNA damage in hypoxia. Most significantly, we describe a stress-specific role for ATM in maintaining DNA replication rates in a background of increased H3K9me3. Furthermore, the ATM-mediated response to oncogene-induced replication stress is enhanced in hypoxic conditions. Together, these data indicate that hypoxia plays a critical role in the activation of the DNA damage response, therefore contributing to this barrier to tumorigenesis.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Replicación del ADN/genética , Heterocromatina/genética , Animales , Hipoxia de la Célula/genética , Línea Celular , Daño del ADN , Proteínas de Unión al ADN/genética , Células HEK293 , Histonas/genética , Humanos , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA