Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Exp Mol Med ; 56(4): 1013-1026, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38684915

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent because it kills cancer cells while sparing normal cells. However, many cancers, including pancreatic ductal adenocarcinoma (PDAC), exhibit intrinsic or acquired resistance to TRAIL, and the molecular mechanisms underlying TRAIL resistance in cancers, particularly in PDAC, remain unclear. In this study, we demonstrated that glutamine (Gln) endows PDAC cells with resistance to TRAIL through KDM4C-mediated epigenetic regulation of cFLIP. Inhibition of glutaminolysis significantly reduced the cFLIP level, leading to TRAIL-mediated formation of death-inducing signaling complexes. Overexpression of cFLIP dramatically rescued PDAC cells from TRAIL/Gln deprivation-induced apoptosis. Alpha-Ketoglutarate (aKG) supplementation significantly reversed the decrease in the cFLIP level induced by glutaminolysis inhibition and rescued PDAC cells from TRAIL/Gln deprivation-induced apoptosis. Knockdown of glutamic-oxaloacetic transaminase 2, which facilitates the conversion of oxaloacetate and glutamate into aspartate and aKG, decreased aKG production and the cFLIP level and activated TRAIL-induced apoptosis. AKG-mediated epigenetic regulation was necessary for maintaining a high level of cFLIP. Glutaminolysis inhibition increased the abundance of H3K9me3 in the cFLIP promoter, indicating that Gln-derived aKG production is important for Jumonji-domain histone demethylase (JHDM)-mediated cFLIP regulation. The JHDM KDM4C regulated cFLIP expression by binding to its promoter, and KDM4C knockdown sensitized PDAC cells to TRAIL-induced apoptosis. The present findings suggest that Gln-derived aKG production is required for KDM4C-mediated epigenetic regulation of cFLIP, which leads to resistance to TRAIL.


Asunto(s)
Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Resistencia a Antineoplásicos , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Glutamina , Histona Demetilasas con Dominio de Jumonji , Neoplasias Pancreáticas , Ligando Inductor de Apoptosis Relacionado con TNF , Humanos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Glutamina/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Resistencia a Antineoplásicos/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Apoptosis/efectos de los fármacos , Ácidos Cetoglutáricos/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Aspartato Aminotransferasa Citoplasmática/metabolismo , Aspartato Aminotransferasa Citoplasmática/genética , Animales , Regiones Promotoras Genéticas
2.
Mol Biol Rep ; 50(8): 6627-6641, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37358765

RESUMEN

BACKGROUND: Glutamine metabolism is critical for development of hepatocellular carcinoma (HCC), which makes it a novel promising treatment target. However, clinical evidence suggested glutamine withdrawal therapy does not achieved the desired tumor suppression. Therefore, it is valuable to investigate the survival mechanisms of tumors with glutamine deprivation. METHODS: The HCC cells were cultured in glutamine-free medium or supplemented with glutamine metabolites or ferroptosis inhibitors. The parameters related to ferroptosis and the activity of GSH synthesis-related enzymes of the HCC cells were detected by corresponding kits. The expressions of glutamate oxaloacetate transaminase 1 (GOT1), c-Myc and Nrf2 were detected by western blot and qRT-PCR. The chromatin immunoprecipitation and luciferase reporter assays were performed to investigate the correlation between c-Myc and GOT1. The siRNAs of c-Myc and GOT1 were used to explore their roles in GSH (GSH) synthesis and ferroptosis in vitro and in vivo. RESULTS: Glutamine deprivation-induced ferroptosis did not completely inhibit HCC cells proliferation. Glutamine deprivation activated the expression of c-Myc, which promoted the transcription of GOT1 and Nrf2, consequently maintaining the GSH synthesis and inhibiting ferroptosis. In addition, combined inhibition of GOT1 with glutamine deprivation could result in better inhibition of HCC in vitro and in vivo. CONCLUSIONS: In our work, the results indicate that GOT1 induced by c-Myc may play an important role in combating ferroptosis due to glutamine deprivation, making it a significant target in glutamine withdrawal therapy. This study provides a theoretical foundation for the clinical targeted therapy for HCC.


Asunto(s)
Carcinoma Hepatocelular , Ferroptosis , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Aspartato Aminotransferasa Citoplasmática/metabolismo , Aspartato Aminotransferasa Citoplasmática/uso terapéutico , Neoplasias Hepáticas/metabolismo , Factor 2 Relacionado con NF-E2/genética , Glutamina/farmacología , Glutamina/metabolismo , Línea Celular Tumoral
3.
Cells ; 11(23)2022 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-36497150

RESUMEN

Recently, evidence has shown that GOT1 expression is upregulated in pancreatic cancer tissues and promotes cancer development, but the specific mechanism remains unclear. We found that GOT1 expression was upregulated in pancreatic cancer cell-derived exosomes. When PANC-1 cells were incubated with exosomes alone or transfected together with si-GOT1, we found that exosomes enhanced cell proliferation, invasion and migration, promoted ferroptosis, and si-GOT1 reversed the effects of exosomes. The results of online bioinformatics database analysis indicated that CCR2 was a potential binding protein of GOT1 and is highly expressed in pancreatic cancer tissues. PANC-1 cells were transfected with pcDNA-CCR2 or si-CCR2, and it was found that pcDNA-CCR2 enhanced cell proliferation, invasion and migration, promoted ferroptosis, and si-CCR2 had an opposite effect. Next, exosome-treated cells were transfected with si-GOT1 alone or together with pcDNA-CCR2, and we found that exosomes promoted CCR2 expression, promoted cell proliferation and invasion, and inhibited ferroptosis, the transfection of si-GOT1 abolished the effect of exosomes, and the transfection of pcDNA-CCR2 again reversed the effect of si-GOT1. Furthermore, when exosome-treated cells were transfected with si-GOT1 alone or co-incubated with Nrf2 activator NK-252, we found that si-GOT1 reversed the promoting effect of exosomes on Nrf2 and HO-1 expression, as well as its inhibitory effect on ferroptosis, but this effect was abrogated by NK-252. In vivo studies showed that knockdown of GOT1 expression inhibited tumor formation compared with tumor tissues formed upon exosome induction, which was mediated by promoting ferroptosis via suppressing the protein expression of GOT1, CCR2, Nrf2 and HO-1 in tumor tissues.


Asunto(s)
Dihidropiridinas , Exosomas , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/patología , Exosomas/metabolismo , Procesos Neoplásicos , Receptores CCR2/genética , Receptores CCR2/metabolismo , Aspartato Aminotransferasa Citoplasmática/metabolismo , Neoplasias Pancreáticas
4.
Bioorg Med Chem Lett ; 73: 128883, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35820623

RESUMEN

Glutamate oxaloacetate transaminase 1 (GOT1) plays a key role in aberrant glutamine metabolism. GOT1 suppression can arrest tumor growth and prevent the development of cancer, indicating GOT1 as a potential anticancer target. Reported GOT1 inhibitors, on the other hand, are quite restricted. Here, we developed and optimized a coupling reaction-based high-throughput screening assay for the discovery of GOT1 inhibitors. By using this screening assay, we found that the cardiovascular drug hydralazine hydrochloride inhibited GOT1 catalytic activity, with an IC50 of 26.62 ± 7.45 µM, in a non-competitive and partial-reversible manner. In addition, we determined the binding affinity of hydralazine hydrochloride to GOT1, with a Kd of 16.54 ± 8.59 µM, using a microscale thermophoresis assay. According to structure-activity relationship analysis, the inhibitory activity of hydralazine hydrochloride is mainly derived from its hydrazine group. Furthermore, it inhibits the proliferation of cancer cells MCF-7 and MDA-MB-468 with a slight inhibitory effect compared to other tested cancer cells, highlighting GOT1 as a promising therapeutic target for the treatment of breast cancer.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática , Ensayos Analíticos de Alto Rendimiento , Aspartato Aminotransferasa Citoplasmática/metabolismo , Aspartato Aminotransferasa Citoplasmática/farmacología , Línea Celular Tumoral , Proliferación Celular , Hidralazina/farmacología
5.
Acta Biochim Biophys Sin (Shanghai) ; 54(5): 625-636, 2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35593470

RESUMEN

Cartilage and subchondral bone communicate with each other through material and signal exchanges. However, direct evidence provided by experimental studies on their interactions is insufficient. In the present study, we establish a noncontact co-culture model with a transwell chamber to explore the energetic perturbations in chondrocytes influenced by osteoblasts. Our results indicate that osteoblasts induce more ATP generation in chondrocytes through an energetic shift characterized by enhanced glycolysis and impaired mitochondrial tricarboxylic acid cycle. Enhanced glycolysis is shown by an increase of secreted lactate and the upregulation of glycolytic enzymes, including glucose-6-phosphate isomerase (Gpi), liver type ATP-dependent 6-phosphofructokinase (Pfkl), fructose-bisphosphate aldolase C (Aldoc), glyceraldehyde-3-phosphate dehydrogenase (Gapdh), triosephosphate isomerase (Tpi1), and phosphoglycerate kinase 1 (Pgk1). Impaired mitochondrial tricarboxylic acid cycle is characterized by the downregulation of cytoplasmic aspartate aminotransferase (Got1) and mitochondrial citrate synthase (Cs). Osteoblasts induce the activation of Akt and P38 signaling to mediate ATP perturbations in chondrocytes. This study may deepen our understanding of the maintenance of metabolic homeostasis in the bone-cartilage unit.


Asunto(s)
Fructosa-Bifosfato Aldolasa , Glucosa-6-Fosfato Isomerasa , Glucosa-6-Fosfato Isomerasa/genética , Glucosa-6-Fosfato Isomerasa/metabolismo , Fructosa-Bifosfato Aldolasa/metabolismo , Triosa-Fosfato Isomerasa/metabolismo , Condrocitos/metabolismo , Glucosa/metabolismo , Aspartato Aminotransferasa Citoplasmática/metabolismo , Fosfoglicerato Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Citrato (si)-Sintasa/metabolismo , Glucólisis , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Fosfofructoquinasa-1/metabolismo , Osteoblastos/metabolismo , Comunicación , Lactatos , Adenosina Trifosfato/metabolismo
6.
Cell Cycle ; 21(3): 247-260, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34919012

RESUMEN

Esophageal squamous cell cancer (ESCC) is a prevalent malignant cancer with high incidence and fatality rate. Surging evidences have revealed that circular RNAs (circRNAs) act key role in ESCC tumorigenesis and progression. Therefore, the purpose of this study is to explore the role and regulatory mechanism of a novel circGOT1 in ESCC. In the present study, the transcriptional expression of circGOT1, miR-606 and GOT1, and the epithelial-mesenchymal transition (EMT) and apoptosis-related markers were examined by quantitative PCR. The protein levels of GOT1 and glycolysis-related proteins were detected by Western blotting. In addition, the glycolytic levels were determined via measuring glucose uptake, lactate production, and ATP levels. Then, the function experiments and rescue experiments were used to investigate the function and mechanism of circGOT1 in ESCC. In addition, RNA immunoprecipitation, pull-down, and luciferase activity reporter gene assays were used to analyze the circGOT1/miR-606/GOT1 axis. The xenograft mouse mode was used to determine the function of circGOT1 in vivo. Here, we identified that circGOT1 and GOT1 upregulate, whereas miR-606 was reduced in ESCC tissues and cell lines. High circGOT1 and GOT1 expression associated with poor survival and worse prognosis of ESCC patients, but miR-606 revealed opposite traits. Mechanically, circGOT1 sponged miR-606 to promote GOT1, which induced cell proliferation, migration, aerobic glycolysis, and cisplatin resistance. The tumor growth was reduced by circGOT1 inhibition in xenograft mouse. Our results indicate the oncogene role of circGOT1 in ESCC via an endogenous competition RNA (ceRNA) mechanism to promote GOT1 expression via sponging miR-606.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , MicroARNs , Animales , Aspartato Aminotransferasa Citoplasmática/genética , Aspartato Aminotransferasa Citoplasmática/metabolismo , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Cisplatino/metabolismo , Cisplatino/farmacología , Cisplatino/uso terapéutico , Células Epiteliales/metabolismo , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Glucólisis/genética , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo
7.
Nat Commun ; 12(1): 4860, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34381026

RESUMEN

Cancer metabolism is rewired to support cell survival in response to intrinsic and environmental stressors. Identification of strategies to target these adaptions is an area of active research. We previously described a cytosolic aspartate aminotransaminase (GOT1)-driven pathway in pancreatic cancer used to maintain redox balance. Here, we sought to identify metabolic dependencies following GOT1 inhibition to exploit this feature of pancreatic cancer and to provide additional insight into regulation of redox metabolism. Using pharmacological methods, we identify cysteine, glutathione, and lipid antioxidant function as metabolic vulnerabilities following GOT1 withdrawal. We demonstrate that targeting any of these pathways triggers ferroptosis, an oxidative, iron-dependent form of cell death, in GOT1 knockdown cells. Mechanistically, we reveal that GOT1 inhibition represses mitochondrial metabolism and promotes a catabolic state. Consequently, we find that this enhances labile iron availability through autophagy, which potentiates the activity of ferroptotic stimuli. Overall, our study identifies a biochemical connection between GOT1, iron regulation, and ferroptosis.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/antagonistas & inhibidores , Ferroptosis , Neoplasias Pancreáticas/metabolismo , Animales , Antioxidantes/farmacología , Aspartato Aminotransferasa Citoplasmática/genética , Aspartato Aminotransferasa Citoplasmática/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Cistina/metabolismo , Ferroptosis/efectos de los fármacos , Glutatión/biosíntesis , Humanos , Hierro/metabolismo , Ratones , Mitocondrias/metabolismo , Neoplasias Pancreáticas/patología
8.
Gastroenterology ; 161(5): 1584-1600, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34245764

RESUMEN

BACKGROUND & AIMS: SIRT5 plays pleiotropic roles via post-translational modifications, serving as a tumor suppressor, or an oncogene, in different tumors. However, the role SIRT5 plays in the initiation and progression of pancreatic ductal adenocarcinoma (PDAC) remains unknown. METHODS: Published datasets and tissue arrays with SIRT5 staining were used to investigate the clinical relevance of SIRT5 in PDAC. Furthermore, to define the role of SIRT5 in the carcinogenesis of PDAC, we generated autochthonous mouse models with conditional Sirt5 knockout. Moreover, to examine the mechanistic role of SIRT5 in PDAC carcinogenesis, SIRT5 was knocked down in PDAC cell lines and organoids, followed by metabolomics and proteomics studies. A novel SIRT5 activator was used for therapeutic studies in organoids and patient-derived xenografts. RESULTS: SIRT5 expression negatively regulated tumor cell proliferation and correlated with a favorable prognosis in patients with PDAC. Genetic ablation of Sirt5 in PDAC mouse models promoted acinar-to-ductal metaplasia, precursor lesions, and pancreatic tumorigenesis, resulting in poor survival. Mechanistically, SIRT5 loss enhanced glutamine and glutathione metabolism via acetylation-mediated activation of GOT1. A selective SIRT5 activator, MC3138, phenocopied the effects of SIRT5 overexpression and exhibited antitumor effects on human PDAC cells. MC3138 also diminished nucleotide pools, sensitizing human PDAC cell lines, organoids, and patient-derived xenografts to gemcitabine. CONCLUSIONS: Collectively, we identify SIRT5 as a key tumor suppressor in PDAC, whose loss promotes tumorigenesis through increased noncanonic use of glutamine via GOT1, and that SIRT5 activation is a novel therapeutic strategy to target PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/enzimología , Metabolismo Energético , Neoplasias Pancreáticas/enzimología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Sirtuinas/deficiencia , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Aspartato Aminotransferasa Citoplasmática/genética , Aspartato Aminotransferasa Citoplasmática/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Progresión de la Enfermedad , Metabolismo Energético/efectos de los fármacos , Activación Enzimática , Activadores de Enzimas/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Mutación , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal , Sirtuinas/genética , Carga Tumoral , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
9.
World Neurosurg ; 154: e616-e626, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34325031

RESUMEN

BACKGROUND: Glycolysis is an important metabolic manner in glioblastoma multiforme (GBM)'s rapid growth. It has been reported that glutamate-oxaloacetate transaminase 1 (GOT1) is low-expressed in GBM and patients with high-expressed GOT1 have better prognosis. However, the effect and mechanism of GOT1 on glycolysis and malignant phenotypes of GBM cells are still unclear. METHODS: The expression differences of GOT1 between GBM parenchyma and adjacent tissues were detected. The prognosis and clinical data with different levels of GOT1 were also analyzed. The glucose consumption, production of lactate and pyruvate were measured after GOT1 was knocked down or overexpressed. The effects of GOT1 on GBM cell's malignant phenotypes were analyzed by Western blot, CCK-8 assay, and flow cytometry. The relationship between GOT1 and pyruvate carboxylase (PC) was examined by immunoprecipitation and immunofluorescence. RESULTS: GOT1 was expressed little in GBM, and patients with highly expressed GOT1 had longer survival periods. Overexpressed GOT1 inhibited the glycolysis and malignant phenotypes of GBM cells. 2-DG treatment could partially reverse the enhancement of malignant phenotypes caused by knockdown of GOT1. The expression of GOT1 was positively correlated with PC. The inhibitory effect of GOT1 on glycolysis could be partially reversed by PC's knockdown. CONCLUSIONS: GOT1 could impair glycolysis by interacting with PC and further inhibit the malignant phenotypes of GBM cells.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Glucólisis , Piruvato Carboxilasa/metabolismo , Anciano , Línea Celular Tumoral , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
10.
Pharmacol Res ; 169: 105640, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33915296

RESUMEN

AIM: Brain microvascular endothelial cells (BMVECs), as the important structure of blood-brain barrier (BBB), play a vital role in ischemic stroke. Pyroptosis of different cells in the brain may aggravate cerebral ischemic injury, and PGC-1α plays a major role in pyroptosis. However, it is not known whether BMVECs undergo pyroptosis after ischemic stroke and whether PGC-1α activator Medioresinol (MDN) we discovered may be useful against pyroptosis of endothelial cells and ischemic brain injury. METHODS: For in vitro experiments, the bEnd.3 cells and BMVECs under oxygen and glucose-deprivation (OGD) were treated with or without MDN, and the LDH release, tight junction protein degradation, GSDMD-NT membrane location and pyroptosis-associated proteins were evaluated. For in vivo experiments, mice underwent transient middle cerebral artery occlusion (tMCAO) for ischemia model, and the neuroprotective effects of MDN were measured by infarct volume, the permeability of BBB and pyroptosis of BMVECs. For mechanistic study, effects of MDN on the accumulation of phenylalanine, mitochondrial reactive oxygen species (mtROS) were tested by untargeted metabolomics and MitoSOX Red probe, respectively. RESULTS: BMVECs underwent pyroptosis after ischemia. MDN dose-dependently activated PGC-1α, significantly reduced pyroptosis, mtROS and the expressions of pyroptosis-associated proteins (NLRP3, ASC, cleaved caspase-1, IL-1ß, GSDMD-NT), and increased ZO-1 and Occludin protein expressions in BMVECs. In tMCAO mice, MDN remarkably reduced brain infarct volume and the permeability of BBB, inhibited pyroptosis of BMVECs, and promoted long-term neurobehavioral functional recovery. Mechanistically, MDN promoted the interaction of PGC-1α with PPARα to increase PPARα nuclear translocation and transcription activity, further increased the expression of GOT1 and PAH, resulting in enhanced phenylalanine metabolism to reduce the ischemia-caused phenylalanine accumulation and mtROS and further ameliorate pyroptosis of BMVECs. CONCLUSION: In this study, we for the first time discovered that pyroptosis of BMVECs was involved in the pathogenesis of ischemic stroke and MDN as a novel PGC-1α activator could ameliorate the pyroptosis of endothelial cells and ischemic brain injury, which might attribute to reduction of mtROS through PPARα/GOT1 axis in BMVECs. Taken together, targeting endothelial pyroptosis by MDN may provide alternative therapeutics for brain ischemic stroke.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/metabolismo , Endotelio Vascular/efectos de los fármacos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Lignanos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , PPAR alfa/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/agonistas , Piroptosis/efectos de los fármacos , Animales , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Técnica del Anticuerpo Fluorescente , Cromatografía de Gases y Espectrometría de Masas , Células HEK293/efectos de los fármacos , Humanos , Lignanos/farmacología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Fármacos Neuroprotectores/farmacología , Ratas Sprague-Dawley
11.
Transl Res ; 230: 68-81, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33132087

RESUMEN

Glutamate oxaloacetate transaminase 1 (GOT1) enzyme plays a critical role in the cell metabolism by participating in the carbohydrate and amino acid metabolism. In ischemic stroke, we have demonstrated that recombinant GOT1 acts as a novel neuroprotective treatment against the excess of extracellular glutamate that accumulates in the brain following ischemic stroke. In this study, we investigated the inhibitory effect of GOT1 on brain metabolism and on the ischemic damage in a rat model of ischemic stroke by means of a specific antibody developed against this enzyme. Inhibition of GOT1 caused higher brain glutamate and lactate levels and this response was associated with larger ischemic lesion. This study represents the first demonstration that the inhibition of the blood GOT1 activity leads to more severe ischemic damage and poorer outcome and supports the protective role of GOT1 against ischemic insults.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/antagonistas & inhibidores , Aspartato Aminotransferasa Citoplasmática/metabolismo , Isquemia Encefálica/enzimología , Isquemia Encefálica/patología , Animales , Anticuerpos , Aspartato Aminotransferasa Citoplasmática/líquido cefalorraquídeo , Encéfalo/enzimología , Clonación Molecular , Relación Dosis-Respuesta Inmunológica , Ácido Glutámico/sangre , Células Hep G2 , Humanos , Inmunoglobulina G , Ácido Láctico/sangre , Masculino , Ratas , Ratas Sprague-Dawley
12.
Am J Med Sci ; 360(6): 711-720, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32988599

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC), featuring uncontrolled proliferation and migration of tumor cells, is one of the most serious malignancies with high morality. An increasing number of evidences have demonstrated that long noncoding RNAs (lncRNAs) are involved in the progression of multiple cancers. It has been acknowledged that lncRNA TMPO-AS1 plays an oncogenic role in diverse cancers. METHODS: Reverse transcription­quantitative polymerase chain reaction (RT-qPCR) was used to determine the expression of TMPO-AS1, miR-429 and GOT1 in HCC tissues and cell lines. Cell viability, proliferation, apoptosis, and stemness characteristics were detected by Cell Counting Kit-8 (CCK-8), colony formation, flow cytometry, sphere formation and western blot assays, separately. The relationship among TMPO-AS1, miR-429 and GOT1 was predicted by starBase database and confirmed using luciferase reporter and RNA pull-down assays. RESULTS: In this study, our findings revealed that TMPO-AS1 expression was upregulated in HCC tissues and cell lines. TMPO-AS1 aggravated HCC progression via promoting cell proliferation, stemness as well as suppressing cell apoptosis. Further, molecular mechanism exploration discovered that TMPO-AS1 functioned as a molecular sponge for miR-429 and GOT1 served as a downstream target gene of miR-429 in HCC. Furthermore, there was a negative relationship between GOT1 and miR-429 as well as a positive correlation between GOT1 and TMPO-AS1 in HCC. Rescue assays suggested that overexpression of GOT1 partially reversed the inhibitory effects of TMPO-AS1 knockdown on HCC progression. CONCLUSIONS: Taken together, these findings indicated that TMPO-AS1 acted as a tumor motivator to expedite HCC progression via targeting miR-429/GOT1 axis, which may provide a fresh treatment strategy for HCC.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/metabolismo , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroARNs/metabolismo , Proteínas Nucleares/metabolismo , ARN Largo no Codificante/metabolismo , Timopoyetinas/metabolismo , Humanos
13.
Biochem Biophys Res Commun ; 522(3): 633-638, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31787239

RESUMEN

Metabolic programs are rewired in cancer cells to support survival and tumor growth. Among these, recent studies have demonstrated that glutamate-oxaloacetate transaminase 1 (GOT1) plays key roles in maintaining redox homeostasis and proliferation of pancreatic ductal adenocarcinomas (PDA). This suggests that small molecule inhibitors of GOT1 could have utility for the treatment of PDA. However, the development of GOT1 inhibitors has been challenging, and no compound has yet demonstrated selectivity for GOT1-dependent cell metabolism or selective growth inhibition of PDA cell lines. In contrast, potent inhibitors that covalently bind to the transaminase cofactor pyridoxal-5'-phosphate (PLP), within the active site of the enzyme, have been reported for kynurenine aminotransferase (KAT) and gamma-aminobutyric acid aminotransferase (GABA-AT). Given the drug discovery successes with these transaminases, we aimed to identify PLP-dependent suicide substrate-type GOT1 inhibitors. Here, we demonstrate that PF-04859989, a known KAT2 inhibitor, has PLP-dependent inhibitory activity against GOT1 and shows selective growth inhibition of PDA cell lines.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/antagonistas & inhibidores , Carcinoma Ductal Pancreático/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Pirazoles/farmacología , Aspartato Aminotransferasa Citoplasmática/metabolismo , Carcinoma Ductal Pancreático/enzimología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Humanos , Neoplasias Pancreáticas/enzimología
14.
Bioorg Chem ; 93: 103315, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31605927

RESUMEN

Glutamic-oxaloacetic transaminase 1 (GOT1) regulates cellular metabolism through coordinating the utilization of carbohydrates and amino acids to meet nutrient requirements for sustained proliferation. As such, the GOT1 inhibitor may provide a new strategy for the treatment of various cancers. Adapalene has been approved by FDA for the treatment of acne, pimples and pustules, and it may also contribute to the adjunctive therapy for advanced stages of liver and colorectal cancers. In this work, we first examined the enzyme inhibition of over 500 compounds against GOT1 in vitro. As a result, Adapalene effectively inhibited GOT1 enzyme in a non-competitive manner. MST and DARTS assay further confirmed the high affinity between Adapalene and GOT1. Furthermore, the growth and migration of ovarian cancer ES-2 cells were obviously inhibited by the treatment of Adapalene. And it induced the apoptosis of ES-2 cells according to Western blot and Hoechst 33258 straining. In addition, molecular docking demonstrated that Adapalene coordinated in an allosteric site of GOT1 with low binding energy. Furthermore, knockdown of GOT1 in ES-2 cells decreased their anti-proliferative sensitivity to Adapalene. Together, our data strongly suggest Adapalene, as a GOT1 inhibitor, could be regarded as a potential drug candidate for ovarian cancer therapy.


Asunto(s)
Adapaleno/química , Aspartato Aminotransferasa Citoplasmática/antagonistas & inhibidores , Adapaleno/metabolismo , Adapaleno/farmacología , Sitio Alostérico , Aspartato Aminotransferasa Citoplasmática/genética , Aspartato Aminotransferasa Citoplasmática/metabolismo , Sitios de Unión , Dominio Catalítico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Cinética , Simulación del Acoplamiento Molecular , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
15.
Sci Rep ; 9(1): 6989, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061470

RESUMEN

Obesity is a progressive, chronic disease, which can be caused by long-term miscommunication between organs. It remains challenging to understand how chronic dysfunction in a particular tissue remotely impairs other organs to eventually imbalance organismal energy homeostasis. Here we introduce RNAi Pulse Induction (RiPI) mediated by short hairpin RNA (shRiPI) or double-stranded RNA (dsRiPI) to generate chronic, organ-specific gene knockdown in the adult Drosophila fat tissue. We show that organ-restricted RiPI targeting Stromal interaction molecule (Stim), an essential factor of store-operated calcium entry (SOCE), results in progressive fat accumulation in fly adipose tissue. Chronic SOCE-dependent adipose tissue dysfunction manifests in considerable changes of the fat cell transcriptome profile, and in resistance to the glucagon-like Adipokinetic hormone (Akh) signaling. Remotely, the adipose tissue dysfunction promotes hyperphagia likely via increased secretion of Akh from the neuroendocrine system. Collectively, our study presents a novel in vivo paradigm in the fly, which is widely applicable to model and functionally analyze inter-organ communication processes in chronic diseases.


Asunto(s)
Tejido Adiposo/metabolismo , Calcio/metabolismo , Proteínas de Drosophila/genética , Hiperfagia/genética , Hormonas de Insectos/genética , Obesidad/genética , Oligopéptidos/genética , Ácido Pirrolidona Carboxílico/análogos & derivados , Molécula de Interacción Estromal 1/genética , Tejido Adiposo/patología , Animales , Aspartato Aminotransferasa Citoplasmática/genética , Aspartato Aminotransferasa Citoplasmática/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Metabolismo Energético/genética , Femenino , Regulación de la Expresión Génica , Homeostasis/genética , Humanos , Hiperfagia/metabolismo , Hiperfagia/patología , Hormonas de Insectos/metabolismo , Transporte Iónico , Isoenzimas/genética , Isoenzimas/metabolismo , Metabolismo de los Lípidos/genética , Malato Deshidrogenasa/genética , Malato Deshidrogenasa/metabolismo , Masculino , Obesidad/metabolismo , Obesidad/patología , Oligopéptidos/metabolismo , Ácido Pirrolidona Carboxílico/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Molécula de Interacción Estromal 1/antagonistas & inhibidores , Molécula de Interacción Estromal 1/metabolismo
16.
Cell Rep ; 26(9): 2257-2265.e4, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30811976

RESUMEN

Cellular aspartate drives cancer cell proliferation, but signaling pathways that rewire aspartate biosynthesis to control cell growth remain largely unknown. Hypoxia-inducible factor-1α (HIF1α) can suppress tumor cell proliferation. Here, we discovered that HIF1α acts as a direct repressor of aspartate biosynthesis involving the suppression of several key aspartate-producing proteins, including cytosolic glutamic-oxaloacetic transaminase-1 (GOT1) and mitochondrial GOT2. Accordingly, HIF1α suppresses aspartate production from both glutamine oxidation as well as the glutamine reductive pathway. Strikingly, the addition of aspartate to the culture medium is sufficient to relieve HIF1α-dependent repression of tumor cell proliferation. Furthermore, these key aspartate-producing players are specifically repressed in VHL-deficient human renal carcinomas, a paradigmatic tumor type in which HIF1α acts as a tumor suppressor, highlighting the in vivo relevance of these findings. In conclusion, we show that HIF1α inhibits cytosolic and mitochondrial aspartate biosynthesis and that this mechanism is the molecular basis for HIF1α tumor suppressor activity.


Asunto(s)
Ácido Aspártico/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias/metabolismo , Proteínas Supresoras de Tumor/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Aspartato Aminotransferasa Citoplasmática/metabolismo , Aspartato Aminotransferasa Mitocondrial/metabolismo , Ácido Aspártico/farmacología , Carcinoma de Células Renales/enzimología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glutamina/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/enzimología , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/antagonistas & inhibidores , Neoplasias/patología , Oxidación-Reducción , Proteínas Supresoras de Tumor/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
17.
Biochem Biophys Res Commun ; 509(1): 241-248, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30591220

RESUMEN

MicroRNAs (miRNAs) play crucial roles in the pancreatic carcinogenesis and progression. In the present study, we found that miR-9-5p was significantly downregulated in pancreatic cancer tissues and cell lines. The expression levels of miR-9-5p were negatively correlated with tumor stage and vessel invasion. Log-rank tests demonstrated that low expression of miR-9-5p was strongly correlated with poor overall survival in pancreatic cancer patients. Moreover, overexpression of miR-9-5p remarkably inhibited pancreatic cancer cell proliferation by enhancing cell apoptosis and significantly suppressed the invasion of pancreatic cancer cells, whereas low expression of miR-9-5p exhibited the opposite effect. Bioinformatics analysis revealed that GOT1 was a potential target of miR-9-5p, and miR-9-5p inhibited the expression level of GOT1 mRNA by direct binding to its 3'-untranslated region (3'UTR). Expression of miR-9-5p was negatively correlated with GOT1 in pancreatic cancer tissues. Moreover, modulation of miR-9-5p expression could affect the glutamine metabolism and redox homeostasis in pancreatic cancer cells. Furthermore, downregulation of GOT1 counteracted the effects of miR-9-5p repression, whereas its overexpression reversed tumor inhibitory effects of miR-9-5p. Collectively, this study suggested that miR-9-5p regulates GOT1 expression in pancreatic cancer, thereby stunting proliferation, invasion, glutamine metabolism and redox homeostasis, and that miR-9-5p may serve as a prognostic or therapeutic target for pancreatic cancer.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/genética , Regulación Neoplásica de la Expresión Génica , Glutamina/metabolismo , MicroARNs/genética , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/genética , Regiones no Traducidas 3' , Aspartato Aminotransferasa Citoplasmática/metabolismo , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Regulación hacia Abajo , Humanos , Invasividad Neoplásica/patología , Oxidación-Reducción , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , ARN Mensajero/genética
18.
Biochemistry ; 57(47): 6604-6614, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30365304

RESUMEN

Pancreatic cancer cells are characterized by deregulated metabolic programs that facilitate growth and resistance to oxidative stress. Among these programs, pancreatic cancers preferentially utilize a metabolic pathway through the enzyme aspartate aminotransferase 1 [also known as glutamate oxaloacetate transaminase 1 (GOT1)] to support cellular redox homeostasis. As such, small molecule inhibitors that target GOT1 could serve as starting points for the development of new therapies for pancreatic cancer. We ran a high-throughput screen for inhibitors of GOT1 and identified a small molecule, iGOT1-01, with in vitro GOT1 inhibitor activity. Application in pancreatic cancer cells revealed metabolic and growth inhibitory activity reflecting a promiscuous inhibitory profile. We then performed an in silico docking analysis to study inhibitor-GOT1 interactions with iGOT1-01 analogues that possess improved solubility and potency properties. These results suggested that the GOT1 inhibitor competed for binding to the pyridoxal 5-phosphate (PLP) cofactor site of GOT1. To analyze how the GOT1 inhibitor bound to GOT1, a series of GOT1 mutant enzymes that abolished PLP binding were generated. Application of the mutants in X-ray crystallography and thermal shift assays again suggested but were unable to formally conclude that the GOT1 inhibitor bound to the PLP site. Mutational studies revealed the relationship between PLP binding and the thermal stability of GOT1 while highlighting the essential nature of several residues for GOT1 catalytic activity. Insight into the mode of action of GOT1 inhibitors may provide leads to the development of drugs that target redox balance in pancreatic cancer.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Inhibidores Enzimáticos/farmacología , Mutación , Neoplasias Pancreáticas/patología , Aspartato Aminotransferasa Citoplasmática/genética , Aspartato Aminotransferasa Citoplasmática/metabolismo , Sitios de Unión , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Metabolómica , Modelos Moleculares , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Conformación Proteica , Relación Estructura-Actividad , Células Tumorales Cultivadas
19.
Biochem Pharmacol ; 156: 444-450, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30222970

RESUMEN

Metformin, a commonly used agent in the treatment of type 2 diabetes, is also associated with reduced risk of cancer development and improvement in cancer survival. Although much is known about metformin, the mechanisms behind its anti-cancer properties are not fully understood. In this study we addressed the role of a mitochondrial transporter commonly upregulated in cancer cells, SLC25A10, for cell survival and metabolism in the presence of metformin. SLC25A10 is a carrier in the mitochondrial inner membrane that transports malate and succinate out of the mitochondria, in exchange of phosphate and sulfate. We show that metformin treatment results in decreased gene expression of the SLC25A10 carrier both in lung cancer A549 mock cells and A549 SLC25A10 knockdown (siSLC25A10) cells. The decrease was even more pronounced when cells were grown at low glucose concentrations. The expression levels of key enzymes in glucose metabolism showed slightly altered mean values for all genes tested in both control cells and siSLC25A10 cells upon metformin treatment. The gene expression of the metabolic regulator glutamic-oxaloacetic transaminase 1 decreased in wild type cells upon metformin treatment whereas there was a trend of increased expression in the siSLC25A10 cells upon metformin treatment. In addition, the gene expression of the cyclin-dependent kinase inhibitor 1A was markedly increased in the siSLC25A10 compared to control A549 cells, and with even larger increases in the presence of metformin and at low glucose concentration. Our data show that in siSLC25A10 cell lines, metformin significantly alters the SLC25A10 carrier at both mRNA and protein levels and can thereby affect the supply of nutrients and the metabolic state of cancer cells.


Asunto(s)
Transportadores de Ácidos Dicarboxílicos/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Glucosa/metabolismo , Metformina/farmacología , Células A549 , Aspartato Aminotransferasa Citoplasmática/genética , Aspartato Aminotransferasa Citoplasmática/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Transportadores de Ácidos Dicarboxílicos/genética , Regulación de la Expresión Génica , Humanos , Especies Reactivas de Oxígeno
20.
BMC Cancer ; 18(1): 559, 2018 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-29751795

RESUMEN

BACKGROUND: Glutamate oxaloacetate transaminase 1 (GOT1) regulates cellular metabolism through coordinating the utilization of carbohydrates and amino acids to meet nutrient requirements. KRAS mutated cancer cells were recently shown to rely on GOT1 to support long-term cell proliferation. The aim of the present study was to address the role of GOT1 in the metabolic adaption of cancer cells. METHODS: GOT1-null and knockdown cell lines were established through CRISPR/Cas9 and shRNA techniques. The growth properties, colony formation ability, autophagy and selected gene expression profiles were analysed. Glucose deprivation decreased the viability of the GOT1-null cells and rescue experiments were conducted with selected intermediates. The redox NADH/NAD+ homeostasis as well as lactate secretion were determined. GOT1 expression levels and correlation with survival rates were analysed in selected tumor databases. RESULTS: Inhibition of GOT1 sensitized the cancer cells to glucose deprivation, which was partially counteracted by oxaloacetate and phosphoenol pyruvate, metabolic intermediates downstream of GOT1. Moreover, GOT1-null cells accumulated NADH and displayed a decreased ratio of NADH/NAD+ with nutrient depletion. The relevance of GOT1 as a potential target in cancer therapy was supported by a lung adenocarcinoma RNA-seq data set as well as the GEO:GSE database of metastatic melanoma where GOT1 expression was increased. High levels of GOT1 were further linked to poor survival as analysed by the GEPIA web tool, in thyroid and breast carcinoma and in lung adenocarcinoma. CONCLUSIONS: Our study suggests an important role of GOT1 to coordinate the glycolytic and the oxidative phosphorylation pathways in KRAS mutated cancer cells. GOT1 is crucial to provide oxaloacetate at low glucose levels, likely to maintain the redox homeostasis. Our data suggest GOT1 as a possible target in cancer therapy.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/metabolismo , Reprogramación Celular , Glucosa/metabolismo , Neoplasias/metabolismo , Células A549 , Aspartato Aminotransferasa Citoplasmática/genética , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ácido Láctico/metabolismo , Neoplasias/genética , Neoplasias/mortalidad , Ácido Oxaloacético/metabolismo , Fosforilación Oxidativa , Proteínas Proto-Oncogénicas p21(ras)/genética , Tasa de Supervivencia , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA