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











Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Gen Subj ; 1867(12): 130492, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37871770

RESUMEN

BACKGROUND: The mitochondrial pyruvate carrier (MPC) is a protein complex composed of two subunits, MPC1 and MPC2. This carrier is at the interface between glycolysis and mitochondrial metabolism and plays an essential role in hepatic glucose production. METHODS: Here we describe an in vitro screen for small molecule inhibitors of the MPC using a strain of Lactococcus lactis that has been engineered to co-express the two subunits of the human MPC and is able to import exogenous 14C-pyruvate. We then tested the top candidates for potential antidiabetic effects through the repression of gluconeogenesis. RESULTS: By screening the Prestwick compound library of 1'200 drugs approved by the Food and Drug Administration for inhibitors of pyruvate uptake, twelve hit molecules were identified. In a secondary screen, the most potent inhibitors were found to inhibit pyruvate-driven oxygen consumption in mouse C2C12 muscle cells. Assessment of gluconeogenesis showed that Zaprinast, as well as the established MPC inhibitor UK5099, inhibited in vitro and in vivo hepatic glucose production. However, when tested acutely in mice without the administration of gluconeogenic substrates, MPC inhibitors raised blood glucose levels, pointing to liver-independent effects. Furthermore, chronic treatment with Zaprinast failed to correct hyperglycemia in both lean and obese diabetic mouse models. CONCLUSIONS: New MPC inhibitors have been identified, showing inhibitory effects on hepatic glucose production. GENERAL SIGNIFICANCE: For potential antidiabetic applications, MPC inhibitors should target the liver without undesired inhibition of mitochondrial pyruvate metabolism in the skeletal muscles or pancreatic beta-cells in order to avoid dual effects on glycemia.


Asunto(s)
Diabetes Mellitus , Glucosa , Estados Unidos , Humanos , Ratones , Animales , Glucosa/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/farmacología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Hígado/metabolismo , Diabetes Mellitus/metabolismo , Hipoglucemiantes/farmacología , Piruvatos/metabolismo , Piruvatos/farmacología
2.
J Bone Joint Surg Am ; 105(5): 369-379, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36728458

RESUMEN

BACKGROUND: High-intensity interval training (HIIT) reportedly improves bone metabolism and increases bone mineral density (BMD). The purpose of the present study was to investigate whether lactate mediates the beneficial effects of exercise on BMD, bone microarchitecture, and biomechanical properties in an established osteoporotic animal model. In addition, we hypothesized that lactate-induced bone augmentation is achieved through enhanced osteoblast differentiation and mineralization. METHODS: A total of 50 female C57BL/6 mice were randomly allocated into 5 groups: the nonovariectomized group, the ovariectomized group (OVX), the HIIT group (OVX + HIIT), the HIIT with lactate transporter inhibition group (OVX + HIIT + INH), and the lactate subcutaneous injection group (OVX + LAC). After 7 weeks of intervention, bone mass, bone strength, and bone formation/resorption processes were evaluated via microcomputed tomography (micro-CT), biomechanical testing, histological analysis, and serum biochemical assays; in vitro studies were performed to explore the bone anabolic effect of lactate at the cellular level. RESULTS: Micro-CT revealed significantly increased BMD in both the OVX + HIIT group (mean difference, 41.03 mg hydroxyapatite [HA]/cm 3 [95% CI, 2.51 to 79.54 mg HA/cm 3 ]; p = 0.029) and the OVX + LAC group (mean difference, 40.40 mg HA/cm 3 [95% CI, 4.08 to 76.71 mg HA/cm 3 ]; p = 0.031) compared with the OVX group. Biomechanical testing demonstrated significantly improved mechanical properties in those 2 groups. However, the beneficial effects of exercise on bone microstructure and biomechanics were largely abolished by blocking the lactate transporter. Notably, histological and biochemical results indicated that increased bone formation was responsible for the bone augmentation effects of HIIT and lactate. Cell culture studies showed a marked increase in the expression of osteoblastic markers with lactate treatment, which could be eliminated by blocking the lactate transporter. CONCLUSIONS: Lactate may have mediated the bone anabolic effect of HIIT in osteoporotic mice, which may have resulted from enhanced osteoblast differentiation and mineralization. CLINICAL RELEVANCE: Lactate may mediate the bone anabolic effect of HIIT and serve as a potential inexpensive therapeutic strategy for bone augmentation.


Asunto(s)
Anabolizantes , Resorción Ósea , Entrenamiento de Intervalos de Alta Intensidad , Femenino , Ratones , Animales , Humanos , Osteogénesis , Anabolizantes/metabolismo , Anabolizantes/farmacología , Microtomografía por Rayos X , Ácido Láctico/metabolismo , Ácido Láctico/farmacología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/farmacología , Ratones Endogámicos C57BL , Densidad Ósea , Diferenciación Celular , Osteoblastos , Ovariectomía
3.
Hepatology ; 78(6): 1800-1815, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36651176

RESUMEN

BACKGROUND AND AIMS: NAFLD has become a major metabolic disease worldwide. A few studies have reported the potential relationship between mitochondrial pyruvate carrier 1 (MPC1) and inflammation, fibrosis, and insulin sensitivity in obese or NASH mouse models. However, the impact of MPC1 on NAFLD-related liver lipid metabolism and its role in the NAFLD progression require further investigation. APPROACH AND RESULTS: MPC1 expression was measured in liver tissues from normal controls and patients with NAFLD. We characterized the metabolic phenotypes and expression of genes involved in hepatic lipid accumulation in MPC1 systemic heterozygous knockout (MPC1 +/- ) mice. Hepatic protein lactylation was detected using Tandem Mass Tags proteomics and verified by the overexpression of lactylation mutants in cells. Finally, the effect of MPC1 inhibition on liver inflammation was examined in mice and AML-12 cells. Here, we found that MPC1 expression was positively correlated to liver lipid deposition in patients with NAFLD. MPC1 +/- mice fed with high-fat diet had reduced hepatic lipid accumulation but no change in the expression of lipid synthesis-related genes. MPC1 knockout affected the lactylation of several proteins, especially fatty acid synthase, through the regulation of lactate levels in hepatocytes. Lactylation at the K673 site of fatty acid synthase inhibited fatty acid synthase activity, which mediated the downregulation of liver lipid accumulation by MPC1. Moreover, although MPC1 knockout caused lactate accumulation, inflammation level was controlled because of mitochondrial protection and macrophage polarization. CONCLUSIONS: In NAFLD, MPC1 levels are positively correlated with hepatic lipid deposition; the enhanced lactylation at fatty acid synthase K673 site may be a downstream mechanism.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/farmacología , Hígado/metabolismo , Metabolismo de los Lípidos/genética , Inflamación/metabolismo , Lactatos/metabolismo , Lactatos/farmacología , Lípidos , Ratones Endogámicos C57BL , Dieta Alta en Grasa
4.
Nutr Metab Cardiovasc Dis ; 32(10): 2439-2449, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36096978

RESUMEN

BACKGROUND AND AIMS: Betaine supplementation has been shown to enhance hepatic lipid metabolism in obese mice and improve exercise performance in healthy populations. We examined effects of betaine supplementation, alone or in combination with treadmill exercise, on the metabolic consequences of high fat diet (HFD)-induced obesity in mice. METHODS AND RESULTS: Male C57BL/6 J mice were fed chow or HFD. After 15 weeks, HFD mice were split into: HFD, HFD with betaine (1.5% w/v), HFD with treadmill exercise, and HFD with both betaine and exercise (15 m/min for 45min, 6 days/week; n = 12/group) for 10 weeks. Compared to HFD mice, body weight was significantly reduced in exercise and exercise-betaine mice, but not in mice given betaine alone. Similarly, adiposity was reduced by exercise but not by betaine alone. HFD-induced glucose intolerance was slightly improved by exercise, but not with betaine alone. Significantly greater benefits were observed in exercise-betaine mice, compared to exercise alone, such that GTT-outcomes were similar to controls. This was associated with reduced insulin levels during ipGTT, suggesting enhanced insulin sensitivity. Modest benefits were observed in fatty acid metabolism genes in skeletal muscle, whilst limited effects were observed in the liver. HFD-induced increases in hepatic Mpc1 (mitochondrial pyruvate carrier 1) were normalized by all treatments, suggesting potential links to altered glucose metabolism. CONCLUSIONS: Our data show that drinking 1.5% betaine was sufficient to augment metabolic benefits of exercise in obese mice. These processes appear to be facilitated by altered glucose metabolism, with limited effects on hepatic lipid metabolism.


Asunto(s)
Resistencia a la Insulina , Insulinas , Animales , Betaína/metabolismo , Betaína/farmacología , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos/metabolismo , Glucosa , Insulinas/metabolismo , Insulinas/farmacología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/farmacología , Obesidad/metabolismo
5.
Cancer Biol Med ; 19(9)2022 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-35972052

RESUMEN

OBJECTIVE: This study aimed to evaluate the effects of mitochondrial pyruvate carrier (MPC) blockade on the sensitivity of detection and radiotherapy of prostate cancer (PCa). METHODS: We investigated glycolysis reprogramming and MPC changes in patients with PCa by using metabolic profiling, RNA-Seq, and tissue microarrays. Transient blockade of pyruvate influx into mitochondria was observed in cellular studies to detect its different effects on prostate carcinoma cells and benign prostate cells. Xenograft mouse models were injected with an MPC inhibitor to evaluate the sensitivity of 18F-fluorodeoxyglucose positron emission tomography with computed tomography and radiotherapy of PCa. Furthermore, the molecular mechanism of this different effect of transient blockage towards benign prostate cells and prostate cancer cells was studied in vitro. RESULTS: MPC was elevated in PCa tissue compared with benign prostate tissue, but decreased during cancer progression. The transient blockade increased PCa cell proliferation while decreasing benign prostate cell proliferation, thus increasing the sensitivity of PCa cells to 18F-PET/CT (SUVavg, P = 0.016; SUVmax, P = 0.03) and radiotherapy (P < 0.01). This differential effect of MPC on PCa and benign prostate cells was dependent on regulation by a VDAC1-MPC-mitochondrial homeostasis-glycolysis pathway. CONCLUSIONS: Blockade of pyruvate influx into mitochondria increased glycolysis levels in PCa but not in non-carcinoma prostate tissue. This transient blockage sensitized PCa to both detection and radiotherapy, thus indicating that glycolytic potential is a novel mechanism underlying PCa progression. The change in the mitochondrial pyruvate influx caused by transient MPC blockade provides a critical target for PCa diagnosis and treatment.


Asunto(s)
Neoplasias de la Próstata , Ácido Pirúvico , Animales , Modelos Animales de Enfermedad , Fluorodesoxiglucosa F18/metabolismo , Fluorodesoxiglucosa F18/farmacología , Glucólisis , Humanos , Masculino , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/farmacología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/farmacología , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Ácido Pirúvico/metabolismo , Ácido Pirúvico/farmacología
6.
Br J Cancer ; 122(6): 895-903, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31937921

RESUMEN

BACKGROUND: Monocarboxylate transporter 1 (MCT1) is a regulator of cell metabolism and a therapeutic target for cancer treatment. Understanding the changes in tumour function accompanying MCT1 inhibition will better characterise the anti-tumour effects of MCT1 inhibitors, potentially enabling the identification of pharmacodynamic biomarkers for the clinical development of these agents. METHODS: We assessed the impact of the MCT1 inhibitor AZD3965 on tumour metabolism and immune cell infiltration as key determinants of tumour biological function in the MCT1-dependent Raji B cell lymphoma model. RESULTS: Treatment of Raji xenograft-bearing severe combined immunodeficiency mice with AZD3965 led to inhibition of tumour growth paralleled with a decrease in tumour choline, as detected by non-invasive in vivo proton nuclear magnetic resonance spectroscopy. This effect was attributed to inhibition of phosphocholine de novo synthesis following decreased choline kinase α protein and messenger RNA expression that correlated with the AZD3965-induced build-up in intracellular lactate. These changes were concomitant with increased tumour immune cell infiltration involving dendritic and natural killer cells. CONCLUSIONS: Our data provide new insights into the metabolic and cellular changes that occur in the tumour microenvironment following MCT1 blockade, which may contribute to the anti-tumour activity of AZD3965 and could have potential as pharmacodynamic biomarkers of MCT1 inhibition.


Asunto(s)
Metabolismo de los Lípidos/efectos de los fármacos , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Transportadores de Ácidos Monocarboxílicos/uso terapéutico , Pirimidinonas/uso terapéutico , Tiofenos/uso terapéutico , Animales , Técnicas de Cultivo de Célula , Línea Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Transportadores de Ácidos Monocarboxílicos/farmacología , Pirimidinonas/farmacología , Tiofenos/farmacología
7.
Vet Comp Oncol ; 18(3): 324-341, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31661586

RESUMEN

Monocarboxylate transporters (MCTs) support tumour growth by regulating the transport of metabolites in the tumour microenvironment. High MCT1 or MCT4 expression is correlated with poor outcomes in human patients with head and neck squamous cell carcinoma (HNSCC). Recently, drugs targeting these transporters have been developed and may prove to be an effective treatment strategy for HNSCC. Feline oral squamous cell carcinoma (OSCC) is an aggressive and treatment-resistant malignancy resembling advanced or recurrent HNSCC. The goals of this study were to investigate the effects of a previously characterized dual MCT1 and MCT4 inhibitor, MD-1, in OSCC as a novel treatment approach for feline oral cancer. We also sought to determine the potential of feline OSCC as a large animal model for the further development of MCT inhibitors to treat human HNSCC. In vitro, MD-1 reduced the viability of feline OSCC and human HNSCC cell lines, altered glycolytic and mitochondrial metabolism and synergized with platinum-based chemotherapies. While MD-1 treatment increased lactate concentrations in an HNSCC cell line, the inhibitor failed to alter lactate levels in feline OSCC cells, suggesting an MCT-independent activity. In vivo, MD-1 significantly inhibited tumour growth in a subcutaneous xenograft model and prolonged overall survival in an orthotopic model of feline OSCC. Our results show that MD-1 may be an effective therapy for the treatment of feline oral cancer. Our findings also support the further investigation of feline OSCC as a large animal model to inform the development of MCT inhibitors and future clinical studies in human HNSCC.


Asunto(s)
Enfermedades de los Gatos/tratamiento farmacológico , Proteínas Mitocondriales/farmacología , Transportadores de Ácidos Monocarboxílicos/farmacología , Neoplasias de la Boca/veterinaria , Carcinoma de Células Escamosas de Cabeza y Cuello/veterinaria , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/farmacología , Animales , Gatos , Línea Celular Tumoral , Humanos , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/genética , Transportadores de Ácidos Monocarboxílicos/genética , Neoplasias de la Boca/tratamiento farmacológico , Proteínas Musculares/genética , Proteínas Musculares/farmacología , Análisis de Secuencia de ARN , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico
8.
Nat Rev Cancer ; 17(10): 577-593, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28912578

RESUMEN

The high metabolic demand of cancer cells leads to an accumulation of H+ ions in the tumour microenvironment. The disorganized tumour vasculature prevents an efficient wash-out of H+ ions released into the extracellular medium but also favours the development of tumour hypoxic regions associated with a shift towards glycolytic metabolism. Under hypoxia, the final balance of glycolysis, including breakdown of generated ATP, is the production of lactate and a stoichiometric amount of H+ ions. Another major source of H+ ions results from hydration of CO2 produced in the more oxidative tumour areas. All of these events occur at high rates in tumours to fulfil bioenergetic and biosynthetic needs. This Review summarizes the current understanding of how H+-generating metabolic processes segregate within tumours according to the distance from blood vessels and inversely how ambient acidosis influences tumour metabolism, reducing glycolysis while promoting mitochondrial activity. The Review also presents novel insights supporting the participation of acidosis in cancer progression via stimulation of autophagy and immunosuppression. Finally, recent advances in the different therapeutic modalities aiming to either block pH-regulatory systems or exploit acidosis will be discussed.


Asunto(s)
Acidosis/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Microambiente Tumoral , Acidosis/genética , Animales , Autofagia , Anhidrasas Carbónicas/farmacología , Respiración de la Célula , Progresión de la Enfermedad , Glucólisis , Homeostasis/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Tolerancia Inmunológica , Transportadores de Ácidos Monocarboxílicos/farmacología , Neoplasias/fisiopatología , Inhibidores de la Bomba de Protones/farmacología , Linfocitos T , Microambiente Tumoral/inmunología
10.
Biol Pharm Bull ; 28(12): 2235-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16327156

RESUMEN

Telmisartan is the most recently marketed angiotensin II type 1 receptor antagonist. Drug-drug interactions involving transporters can directly affect the therapeutic safety and efficacy of many important drugs. In clinical practice, telmisartan is coadministered with many kinds of drugs. However, little is known about the contribution of transporters to the intestinal transport of telmisartan. The aim of this study was to determine the transport mechanism of telmisartan across intestinal epithelial cells. In the presence of an inwardly directed proton gradient, the apical-to-basal transport of telmisartan was greater than basal-to-apical transport. Thus, we focused on the uptake mechanism of telmisartan across brush-border membranes. The uptake of telmisartan by Caco-2 cells was shown to be energy- and proton-dependent. Although some monocarboxylates inhibited the uptake of telmisartan, L-lactic acid, which is a typical substrate of the monocarboxylate transporter (MCT) 1-MCT4, did not affect the uptake of telmisartan. Preloading of acetic acid enhanced the uptake of telmisartan, showing a trans-stimulation effect. These results suggest that the carrier-mediated transport system is involved in the uptake of telmisartan by Caco-2 cells and that the apical-localized transport system is similar to MCTs, but not MCT1-MCT4. It is possible that telmisartan reduce the absorption of coadministered drugs by sharing the MCTs. Since MCTs have an important role in the intestinal absorption of pharmacologically active compounds, it is important to be aware of the potential of telmisartan-drug interactions involving MCTs and to act in order to prevent undesirable and harmful consequences.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bencimidazoles/farmacología , Benzoatos/farmacología , Células CACO-2/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/antagonistas & inhibidores , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bencimidazoles/antagonistas & inhibidores , Bencimidazoles/metabolismo , Benzoatos/antagonistas & inhibidores , Benzoatos/metabolismo , Transporte Biológico/efectos de los fármacos , Células CACO-2/efectos de los fármacos , Células CACO-2/patología , Relación Dosis-Respuesta a Droga , Epitelio/patología , Humanos , Concentración de Iones de Hidrógeno , Transportadores de Ácidos Monocarboxílicos/farmacología , Telmisartán
11.
Am J Physiol Gastrointest Liver Physiol ; 288(6): G1118-26, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15691871

RESUMEN

Butyrate, a short-chain fatty acid, is the major energy fuel for the colonocytes. We have previously reported that monocarboxylate transporter isoform 1 (MCT1) mediates uptake of butyrate by human colonic Caco-2 cells. To better understand the mechanisms of MCT1 expression and regulation in the human intestine, we examined the activity and regulation of MCT1 promoter in Caco-2 cells. The transcription initiation site in the MCT1 promoter was identified as a guanine nucleotide 281 bp upstream from the translation initiation site and is surrounded by a guanine-cytosine-rich area. The promoter was found to be highly active when transfected into Caco-2 cells, and its activity decreased with deletions at its 5'-end. Gel mobility shift experiments showed binding of the transcription factors upstream stimulatory factor (USF)1 and 2 to the site -114 to -119 of the MCT1 promoter. With the use of site-directed mutagenesis and promoter activity in Caco-2 cells, the USF proteins appeared to have a repressor role on the MCT1 promoter, which was further confirmed by cotransfecting expression vectors encoding USF1 and 2 in Caco-2 cells and determining endogenous MCT1 expression in USF2 overexpressed cells. The two potential SP1 binding sites found in the same region of the promoter were found not to be involved in its regulation.


Asunto(s)
Proteínas de Unión al ADN/farmacología , Perfilación de la Expresión Génica , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Simportadores/biosíntesis , Factores de Transcripción/farmacología , Secuencia de Bases , Butiratos/metabolismo , Células CACO-2 , Secuencias Hélice-Asa-Hélice , Humanos , Leucina Zippers , Datos de Secuencia Molecular , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/farmacología , Regiones Promotoras Genéticas , Simportadores/genética , Simportadores/farmacología , Transfección , Factores Estimuladores hacia 5'
12.
J Neurosci ; 24(27): 6202-8, 2004 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-15240812

RESUMEN

Increasing evidence suggests that glutamate activates the generation of lactate from glucose in astrocytes; this lactate is shuttled to neurons that use it as a preferential energy source. We explore this multicellular "lactate shuttle" with a novel dual-cell, dual-gene therapy approach and determine the neuroprotective potential of enhancing this shuttle. Viral vector-driven overexpression of a glucose transporter in glia enhanced glucose uptake, lactate efflux, and the glial capacity to protect neurons from excitotoxicity. In parallel, overexpression of a lactate transporter in neurons enhanced lactate uptake and neuronal resistance to excitotoxicity. Finally, overexpression of both transgenes in the respective cell types provided more protection than either therapy alone, demonstrating that a dual-cell, dual-gene therapy approach gives greater neuroprotection than the conventional single-cell, single-gene strategy.


Asunto(s)
Metabolismo Energético/genética , Terapia Genética/métodos , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neurotoxinas/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Citoprotección/efectos de los fármacos , Citoprotección/genética , Sinergismo Farmacológico , Metabolismo Energético/efectos de los fármacos , Estudios de Factibilidad , Glucosa/metabolismo , Glucosa/farmacocinética , Transportador de Glucosa de Tipo 1 , Ácido Láctico/metabolismo , Ácido Láctico/farmacocinética , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/farmacología , Proteínas de Transporte de Monosacáridos/biosíntesis , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/farmacología , Neuroglía/metabolismo , Plásmidos/genética , Plásmidos/farmacología , Ratas , Ratas Sprague-Dawley , Transfección/métodos , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA