Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(8): 687-695, 2024 Aug.
Artículo en Chino | MEDLINE | ID: mdl-39215666

RESUMEN

Objective To investigate the mechanism of the basement membrane proteoglycan lumican (LUM) in cisplatin resistance in ovarian cancer and to preliminarily explore its effect on type 1 T helper (Th1) cell differentiation. Methods Differentially expressed genes (DEGs) between cisplatin-resistant and cisplatin-sensitive ovarian cancer cell lines were screened using the public gene expression database (GEO). The expression levels of these genes were detected by RT-qPCR. LUM expression in human ovarian cancer cells was knocked down using small interfering RNA (siRNA), and the knockdown efficiency was verified by Western blotting. Flow cytometry was used to detect the effects of LUM knockdown on the cell cycle and apoptosis of cisplatin-treated ovarian cancer cell lines. Potential target proteins of LUM were screened through the PPI network, and their interactions were validated by molecular docking. The TIMER database was used to screen the effects of LUM on cytokine secretion in ovarian cancer cell lines, and the results were validated by ELISA and RT-qPCR. Flow cytometry was performed to analyze the regulatory effect of LUM on the differentiation of CD4+ T cells. Results GEO data showed that LUM was significantly upregulated in cisplatin-resistant cell lines, and its expression level was correlated with patient prognosis. LUM expression level was higher in that of cisplatin-resistant ovarian cancer cell lines, and cisplatin treatment promoted LUM expression. Knockdown of LUM increased cisplatin-induced apoptosis and cell cycle arrest in ovarian cancer cells, enhancing drug sensitivity. Target gene screening suggested that LUM might regulate cisplatin sensitivity in ovarian cancer cells through interaction with Src homology region phosphatase 2(SHP2). Additionally, TIMER database analysis suggested that high LUM expression inhibited Th1 cell differentiation. Knockdown of LUM in cisplatin-resistant cell lines promoted Th1 cell differentiation by regulating the secretion of interferon γ(IFN-γ) and interleukin 12(IL-12) cytokines, thereby influencing the tumoricidal activity of immune cells. Conclusion LUM is upregulated in cisplatin-resistant ovarian cancer cells and reduces cisplatin sensitivity in ovarian cancer cells by regulating the SHP2-related signaling pathway. LUM also promotes tumor resistance by inhibiting Th1 cell differentiation through the regulation of cytokine secretion by ovarian cancer cells, making it a potential target for ovarian cancer treatment.


Asunto(s)
Diferenciación Celular , Cisplatino , Resistencia a Antineoplásicos , Neoplasias Ováricas , Células TH1 , Humanos , Cisplatino/farmacología , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Resistencia a Antineoplásicos/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Células TH1/inmunología , Células TH1/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Antineoplásicos/farmacología
2.
Blood ; 137(7): 945-958, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33254233

RESUMEN

Isocitrate dehydrogenase (IDH) mutations are common genetic alterations in myeloid disorders, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Epigenetic changes, including abnormal histone and DNA methylation, have been implicated in the pathogenic build-up of hematopoietic progenitors, but it is still unclear whether and how IDH mutations themselves affect hematopoiesis. Here, we show that IDH1-mutant mice develop myeloid dysplasia in that these animals exhibit anemia, ineffective erythropoiesis, and increased immature progenitors and erythroblasts. In erythroid cells of these mice, D-2-hydroxyglutarate, an aberrant metabolite produced by the mutant IDH1 enzyme, inhibits oxoglutarate dehydrogenase activity and diminishes succinyl-coenzyme A (CoA) production. This succinyl-CoA deficiency attenuates heme biosynthesis in IDH1-mutant hematopoietic cells, thus blocking erythroid differentiation at the late erythroblast stage and the erythroid commitment of hematopoietic stem cells, while the exogenous succinyl-CoA or 5-ALA rescues erythropoiesis in IDH1-mutant erythroid cells. Heme deficiency also impairs heme oxygenase-1 expression, which reduces levels of important heme catabolites such as biliverdin and bilirubin. These deficits result in accumulation of excessive reactive oxygen species that induce the cell death of IDH1-mutant erythroid cells. Our results clearly show the essential role of IDH1 in normal erythropoiesis and describe how its mutation leads to myeloid disorders. These data thus have important implications for the devising of new treatments for IDH-mutant tumors.


Asunto(s)
Eritropoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Hemo/biosíntesis , Isocitrato Deshidrogenasa/genética , Mutación Missense , Mutación Puntual , Preleucemia/genética , Acilcoenzima A/biosíntesis , Acilcoenzima A/deficiencia , Anemia/genética , Animales , Médula Ósea/patología , Eritroblastos/metabolismo , Técnicas de Sustitución del Gen , Glutaratos/metabolismo , Hemo/deficiencia , Hemo-Oxigenasa 1/metabolismo , Isocitrato Deshidrogenasa/fisiología , Complejo Cetoglutarato Deshidrogenasa/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Mielopoyesis/genética , Preleucemia/metabolismo , Preleucemia/patología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Esplenomegalia/etiología , Trombocitopenia/genética
3.
Oncogene ; 39(40): 6340-6353, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32855525

RESUMEN

Isocitrate dehydrogenase (IDH) mutation is the most important initiating event in gliomagenesis, and the increasing evidence shows that IDH mutation is associated with the metabolic reprogramming in the tumor. Dysregulated cholesterol metabolism is a hallmark of tumor cells, but the cholesterol homeostasis in IDH-mutated glioma is still unknown. In this study, we found that astrocyte-specific mutant IDH1(R132H) knockin reduced the cholesterol contents and damaged the structure of myelin in mouse brains. In U87 and U251 cells, the expression of mutant IDH1 consistently reduced the cholesterol levels. Furthermore, we found that IDH1 mutation enhanced the production of 24(S)-hydroxycholesterol (24-OHC), which is not only the metabolite of cholesterol elimination, but also functions as an endogenous ligand for the liver X receptors (LXRs). In IDH1-mutant glioma cells, the elevated 24-OHC activated LXRs, which consequently accelerated the low-density lipoprotein receptor (LDLR) degradation by upregulating the inducible degrader of the LDLR (IDOL). The reduced LDLR expressions in IDH1-mutant glioma cells abated the uptakes of low-density lipoprotein (LDL) to decrease the cholesterol influx. In addition, the activated LXRs also promoted the cholesterol efflux by elevating the ATP-binding cassette transporter A1 (ABCA1), ABCG1, and apolipoprotein E (ApoE) in both IDH1-mutant astrocytes and glioma cells. As a feedback, the reduced cholesterol levels stimulated the cholesterol biosynthesis, which made IDH1-mutated glioma cells more sensitive to atorvastatin, an inhibitor of 3-hydroxy-3-methylglutaryl-CoA reductase. The altered cholesterol homeostasis regulated by mutant IDH provides a pivotal therapeutical strategy for the IDH-mutated gliomas.


Asunto(s)
Neoplasias Encefálicas/genética , Encéfalo/patología , Glioma/genética , Hidroxicolesteroles/metabolismo , Isocitrato Deshidrogenasa/genética , Animales , Astrocitos/metabolismo , Atorvastatina/uso terapéutico , Encéfalo/citología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Técnicas de Sustitución del Gen , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Isocitrato Deshidrogenasa/metabolismo , Ratones , Ratones Transgénicos , Mutación , Vaina de Mielina/patología , Cultivo Primario de Células
4.
Transl Oncol ; 13(4): 100758, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32208352

RESUMEN

Isocitrate dehydrogenase (IDH) mutations occur frequently in lower-grade gliomas, which result in genome-wide epigenetic alterations. The wild-type IDH1 is reported to participate in lipid biosynthesis and amino acid metabolism, but its role in tumorigenesis is still unclear. In this study, the expressions of IDH1 and podoplanin (Pdpn) were determined in IDH-mutated and IDH-wild-type gliomas, and their relationships in glioma were further analyzed. In addition, the regulation of wild-type IDH1 and mutant IDH1 on Pdpn expression was investigated by luciferase assays and promoter methylation analysis. Our study showed that Pdpn was almost undetectable in IDH-mutated glioma but strongly expressed in higher-grade IDH-wild-type glioma. Pdpn overexpression promoted the migration of glioma cells but had little effect on cell growth. Moreover, Pdpn expression was positively correlated with the increased wild-type IDH1 levels in IDH-wild-type glioma. Consistently, the wild-type IDH1 greatly promoted the transcription and expression of Pdpn, but the mutant IDH1 and D-2-hydroxyglutarate significantly suppressed Pdpn expression in glioma cells. Besides, our results revealed that the methylation of CpG islands in the Pdpn promoter was opposingly regulated by wild-type and mutant IDH1 in glioma. Collectively, our results indicated that wild-type and mutant IDH1 opposingly controlled the Pdpn expression in glioma by regulating its promoter methylation, which provides a basis for understanding the relationship between wild-type and mutant IDH1 in epigenetic regulation and tumorigenesis.

5.
J Cell Biochem ; 120(9): 16075-16087, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31119787

RESUMEN

Hepatic lipid accumulation is the most common pathological characteristic of alcoholic liver disease (ALD). In mammalian cells, excess neutral lipids are stored in lipid droplets (LDs). As a member of perilipin family proteins, Plin3 was recently found to regulate the LD biogenesis. However, the roles and mechanism of Plin3 in ALD progression remain unclear. Herein, we found that alcohol stimulated Plin3 expression in both mouse livers and cultured AML12 mouse hepatic cells, which was accompanied by excess LD accumulation in hepatocytes. The elevations of Plin3 in alcohol-treated hepatocytes paralleled with the levels of both PPARα and γ, and the protein degradation of Plin3 was also reduced after alcohol exposure. Moreover, Plin3 knockdown increased cellular sensitivity to alcohol-induced apoptosis, endoplasmic reticulum (ER) stress, and inflammatory cytokines release, including TNF-α, IL-1, and IL-6ß. Notably, alcohol exacerbated triglycerides (TG) accumulation in the ER and caused ER dilation in Plin3-knockdown AML12 cells. Finally, we observed that Plin3 interacted with dynein subunit Dync1i1 and mediated the colocalization of LDs and microtubules, while high concentration of alcohol disrupted microtubules and caused dispersion of excess small LDs in cytoplasm. Summarily, Plin3 promotes lipid export from the ER and reduces ER lipotoxic stress, thereby, protecting against alcoholic liver injury. Moreover, Plin3 could be an adapter protein mediating LD transport by microtubules. This study explored the roles of Plin3 in alcohol-induced hepatic injury, suggesting Plin3 as a potential target for the prevention of ALD progression.


Asunto(s)
Retículo Endoplásmico/metabolismo , Etanol/efectos adversos , Hepatocitos/citología , Perilipina-3/metabolismo , Animales , Línea Celular , Dineínas Citoplasmáticas/metabolismo , Estrés del Retículo Endoplásmico , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Gotas Lipídicas/metabolismo , Ratones , Modelos Biológicos , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Perilipina-3/genética , Triglicéridos/metabolismo
6.
Lipids Health Dis ; 18(1): 87, 2019 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-30954078

RESUMEN

BACKGROUND: The homeostasis of lipid droplets (LDs) plays a crucial role in maintaining the physical metabolic processes in cells, and is regulated by many LD-associated proteins, including perilipin 5 (Plin5) in liver. As the putative sites of hepatitis C virus (HCV) virion assembly, LDs are vital to viral infection. In addition, the hepatic LD metabolism can be disturbed by non-structural HCV proteins, such as NS5A, but the details are still inexplicit. METHODS: HCV NS5A was overexpressed in the livers and hepatocytes of wild-type and Plin5-null mice. BODIPY 493/503 and oil red O staining were used to detect the lipid content in mouse livers and hepatocytes. The levels of lipids, lipid peroxidation and inflammation biomarkers were further determined. Immunofluorescence assay and co-immunoprecipitation assay were performed to investigate the relationship of Plin5 and NS5A. RESULTS: One week after adenovirus injection, livers expressing NS5A showed more inflammatory cell aggregation and more severe hepatic injuries in Plin5-null mice than in control mice, which was consistent with the increased serum levels of IL-2 and TNF-α (P < 0.05) observed in Plin5-null mice. Moreover, Plin5 deficiency in the liver and hepatocytes aggravated the elevation of MDA and 4-HNE levels induced by NS5A expression (P < 0.01). The triglyceride (TG) content was increased approximately 25% by NS5A expression in the wild-type liver and hepatocytes but was unchanged in the Plin5-null liver and hepatocytes. More importantly, Plin5 deficiency in the liver and hepatocytes exacerbated the elevation of non-esterified fatty acids (NEFAs) stimulated by NS5A expression (P < 0.05 and 0.01 respectively). Using triacsin C to block acyl-CoA biosynthesis, we found that Plin5 deficiency aggravated the NS5A-induced lipolysis of TG. In contrast, Plin5 overexpression in HepG2 cells ameliorated the NS5A-induced lipolysis and lipotoxic injuries. Immunofluorescent staining demonstrated that NS5A expression stimulated the targeting of Plin5 to the surface of the LDs in hepatocytes without altering the protein levels of Plin5. By co-IP, we found that the N-terminal domain (aa 32-128) of Plin5 was pivotal for its binding with NS5A. CONCLUSIONS: Our data highlight a protective role of Plin5 against hepatic lipotoxic injuries induced by HCV NS5A, which is helpful for understanding the steatosis and injuries in liver during HCV infection.


Asunto(s)
Hígado Graso/genética , Hepatitis C/genética , Hígado/metabolismo , Perilipina-5/genética , Proteínas no Estructurales Virales/genética , Acilcoenzima A/antagonistas & inhibidores , Acilcoenzima A/biosíntesis , Adenoviridae/genética , Animales , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Hígado Graso/terapia , Regulación Viral de la Expresión Génica/genética , Hepacivirus/genética , Hepacivirus/patogenicidad , Hepatitis C/metabolismo , Hepatitis C/patología , Hepatitis C/virología , Hepatocitos/metabolismo , Hepatocitos/patología , Hepatocitos/virología , Humanos , Gotas Lipídicas/metabolismo , Gotas Lipídicas/patología , Metabolismo de los Lípidos/genética , Lipólisis/genética , Hígado/lesiones , Hígado/patología , Hígado/virología , Ratones , Triazenos/administración & dosificación , Triglicéridos/genética , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/genética
7.
J Neurooncol ; 143(2): 187-196, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31004262

RESUMEN

INTRODUCTION: Tumour-associated angiogenesis is associated with the malignancy and poor prognosis of glioma. Isocitrate dehydrogenase (IDH) mutations are present in the majority of lower-grade (WHO grade II and III) and secondary glioblastomas, but their roles in tumour angiogenesis remain unclear. METHODS: Using magnetic resonance imaging (MRI), the cerebral blood flow (CBF) of IDH-mutated glioma was measured and compared with the IDH-wildtype glioma. The densities of microvessels in IDH-mutated and wildtype astrocytoma and glioblastoma were assessed by immunohistochemical (IHC) staining with CD34, and the pericytes were labelled with α-smooth muscle antigen (α-SMA), neural-glial antigen 2 (NG2) and PDGF receptor-ß (PDGFR-ß), respectively. Furthermore, glia-specific mutant IDH1 knock-in mice were generated to evaluate the roles of mutant IDH1 on brain vascular architectures. The transcriptions of the angiogenesis-related genes were assessed in TCGA datasets, including ANGPT1, PDGFB and VEGFA. The expressions of these genes were further determined by western blot in U87-MG cells expressing a mutant IDH1 or treated with 2-HG. RESULTS: The MRI results indicated that CBF was reduced in the IDH-mutated gliomas. The IHC staining showed that the pericyte coverages of microvessels were significantly decreased, but the microvessel densities (MVDs) were only slightly decreased in IDH-mutated glioma. The mutant IDH1 knock-in also impeded the pericyte coverage of brain microvessels in mice. Moreover, the TCGA database showed the mRNA levels of angiogenesis factors, including ANGPT1, PDGFB and VEGFA, were downregulated, and their promoters were also highly hyper-methylated in IDH-mutated gliomas. In addition, both mutant IDH1 and D-2-HG could downregulate the expression of these genes in U87-MG cells. CONCLUSIONS: Our results suggested that IDH mutations could reduce the pericyte coverage of microvessels in astrocytic tumours by inhibiting the expression of angiogenesis factors. As vascular pericytes play an essential role in maintaining functional blood vessels to support tumour growth, our findings imply a potential avenue of therapeutic strategy for IDH-mutated gliomas.


Asunto(s)
Astrocitoma/patología , Isocitrato Deshidrogenasa/genética , Microvasos/patología , Mutación , Neovascularización Patológica , Pericitos/patología , Animales , Astrocitoma/genética , Astrocitoma/metabolismo , Circulación Cerebrovascular , Humanos , Isocitrato Deshidrogenasa/fisiología , Ratones , Microvasos/metabolismo , Pericitos/metabolismo , Células Tumorales Cultivadas
8.
Mol Med Rep ; 19(4): 2509-2518, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30720071

RESUMEN

Increasing evidence has indicated that mutations of isocitrate dehydrogenase 1/2 (IDH1/2) contribute to the metabolic reprogramming of cancer cells; however their functions in lipid metabolism remain unknown. In the present study, the parental and IDH1 (R132H/+) mutant HCT116 cells were treated with various concentrations of oleic acid (OA) or palmitic acid (PA) in the presence or absence of glucose. The results demonstrated that mutation of IDH1 exacerbated the effects of OA and PA on cell viability and apoptosis, and consistently elevated the production of reactive oxygen species in HCT116 cells, particularly in the absence of glucose. Furthermore, mutation of IDH1 inhibited the rate of fatty acid oxidation (FAO), but elevated the glucose consumption in HCT116 cells. The results of immunoblotting and reverse transcription­quantitative polymerase chain reaction (RT­qPCR) indicated that the expression of glucose transporter 1 was upregulated, whereas that of carnitine palmitoyl transferase 1 was downregulated in IDH1 mutant HCT116 cells. Although mitochondrial DNA quantification demonstrated that mutation of IDH1 had no effect on the quantity of mitochondria, immunoblotting and RT­qPCR revealed that mutation of IDH1 in HCT116 cells significantly downregulated the expression of cytochrome c (CYCS) and CYCS oxidase IV, two important components in mitochondrial respiratory chain. These results indicated that mutation of IDH1 aggravated the fatty acid­induced oxidative stress in HCT116 cells, by suppressing FAO and disrupting the mitochondrial respiratory chain. The results of the present study may provide novel insight into therapeutic strategies for the treatment of cancer types with IDH mutation.


Asunto(s)
Transporte de Electrón/efectos de los fármacos , Ácidos Grasos/farmacología , Isocitrato Deshidrogenasa/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Glucosa/metabolismo , Células HCT116 , Humanos
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(12): 1512-1519, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28919478

RESUMEN

Statins have been proven to be effective in treating non-alcoholic fatty liver disease (NAFLD). Recently, it was reported that statins decreased the hepatic expression of perilipin 5 (Plin5), a lipid droplet (LD)-associated protein, which plays critical roles in regulating lipid accumulation and lipolysis in liver. However, the function and regulation mechanism of Plin5 have not yet been well-established in NAFLD treatment with statins. In this study, we observed that atorvastatin moderately reduced the expression of Plin5 in livers without changing the protein level of Plin5 in the hepatic LD fraction of mice fed with high-fat diet (HFD). Intriguingly, atorvastatin stimulated the PKA-mediated phosphorylation of Plin5 and reduced the triglyceride (TG) accumulation in hepatocytes with overexpression of wide type (Plin5-WT) compared to serine-155 mutant Plin5 (Plin5-S155A). Moreover, PKA-stimulated FA release of purified LDs carrying Plin5-WT but not Plin5-S155A. Glucagon, a PKA activator, stimulated the phosphorylation of Plin5-WT and inhibited its interaction with CGI-58. The results indicated that atorvastatin promoted lipolysis and reduced TG accumulation in the liver by increasing PKA-mediated phosphorylation of Plin5. This new mechanism of lipid-lowering effects of atorvastatin might provide a new strategy for NAFLD treatment.


Asunto(s)
Atorvastatina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipólisis/efectos de los fármacos , Hígado/metabolismo , Proteínas Musculares/metabolismo , Triglicéridos/metabolismo , Sustitución de Aminoácidos , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Grasas de la Dieta/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Lipólisis/genética , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Mutación Missense , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Triglicéridos/genética
10.
Sci Rep ; 7: 42574, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28218306

RESUMEN

Myocardial ischaemia-reperfusion (I/R) injury is a complex pathophysiological process. Current research has suggested that energy metabolism disorders, of which the abnormal consumption of fatty acids is closely related, compose the main pathological basis for myocardial I/R injury. Lipid droplets (LD) are critical regulators of lipid metabolism by LD-associated proteins. Among the lipid droplet proteins, the perilipin family members regulate lipolysis and lipogenesis through different mechanisms. Plin5, an important perilipin protein, promotes LD generation and lowers fatty acid oxidation, thus protecting the myocardium from lipotoxicity. This study investigated the protective effects of Plin5 in I/R myocardium. Our results indicated that Plin5 deficiency exacerbated the myocardial infarct area, aggravated left ventricular systolic dysfunction, reduced lipid storage, and elevated free fatty acids. Plin5-deficient myocardium exhibited severely damaged mitochondria, elevated reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and decreased superoxide dismutase (SOD) activity. Furthermore, the decreased phosphorylation of PI3K/Akt in Plin5-null cardiomyocytes might contribute to I/R injury aggravation. In conclusion, Plin5, a new regulator of myocardial lipid metabolism, decreases free fatty acid peroxidation by inhibiting the lipolysis of intracellular lipid droplets, thus providing cardioprotection against I/R injury and shedding new light on therapeutic solutions for I/R diseases.


Asunto(s)
Gotas Lipídicas/metabolismo , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Estrés Oxidativo , Perilipina-5/genética , Animales , Metabolismo de los Lípidos , Lipólisis , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Oxidación-Reducción , Perilipina-5/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Disfunción Ventricular/genética
11.
Proc Natl Acad Sci U S A ; 114(2): 292-297, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28011762

RESUMEN

Although the enzymatic activity of isocitrate dehydrogenase 1 (IDH1) was defined decades ago, its functions in vivo are not yet fully understood. Cytosolic IDH1 converts isocitrate to α-ketoglutarate (α-KG), a key metabolite regulating nitrogen homeostasis in catabolic pathways. It was thought that IDH1 might enhance lipid biosynthesis in liver or adipose tissue by generating NADPH, but we show here that lipid contents are relatively unchanged in both IDH1-null mouse liver and IDH1-deficient HepG2 cells generated using the CRISPR-Cas9 system. Instead, we found that IDH1 is critical for liver amino acid (AA) utilization. Body weights of IDH1-null mice fed a high-protein diet (HPD) were abnormally low. After prolonged fasting, IDH1-null mice exhibited decreased blood glucose but elevated blood alanine and glycine compared with wild-type (WT) controls. Similarly, in IDH1-deficient HepG2 cells, glucose consumption was increased, but alanine utilization and levels of intracellular α-KG and glutamate were reduced. In IDH1-deficient primary hepatocytes, gluconeogenesis as well as production of ammonia and urea were decreased. In IDH1-deficient whole livers, expression levels of genes involved in AA metabolism were reduced, whereas those involved in gluconeogenesis were up-regulated. Thus, IDH1 is critical for AA utilization in vivo and its deficiency attenuates gluconeogenesis primarily by impairing α-KG-dependent transamination of glucogenic AAs such as alanine.


Asunto(s)
Aminoácidos/metabolismo , Isocitrato Deshidrogenasa/deficiencia , Hígado/metabolismo , Animales , Glucemia/metabolismo , Línea Celular Tumoral , Ayuno/metabolismo , Gluconeogénesis , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Isocitrato Deshidrogenasa/metabolismo , Ácidos Cetoglutáricos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regulación hacia Arriba/fisiología
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(2): 168-72, 2016 Feb.
Artículo en Chino | MEDLINE | ID: mdl-26927374

RESUMEN

OBJECTIVE: To investigate the impact of metformin (Met) on THP-1 macrophage-derived foam cell formation induced by lipopolysaccharide (LPS) and observe the changes of lipid droplets (LDs) and LDs-associated proteins. METHODS: THP-1 cells were induced to differentiate into macrophages by 100 ng/mL phorbol 12-myristate 13-acetate (PMA) for 48 hours, and then the macrophages were further induced to generate foam cells by 50 µg/mL oxidized low-density lipoprotein (ox-LDL) and 1 µg/mL LPS. During this process, these foam cells were treated with 0, 100, 200 µmol/L Met. Under the fluorescence microscope, the effect of Met on foam cell formation was evaluated by Oil red O staining and the number and morphology of LDs were observed by BODIPY493/503 staining. Intracellular triglyceride (TG) were extracted and measured by TG quantitative kits. The expressions of adipose differentiation-related protein (ADRP) and tail-interacting protein of 47 kDa (TIP47) were detected by Western blotting. RESULTS: Compared with the untreated group, the LDs in foam cells were reduced significantly and the size became smaller after treated with 100 or 200 µmol/L Met. What's more, the quantitative data showed that the intracellular TG content decreased markedly in a dose-dependent manner, and the TG content decreased about 25% in foam cells treated with 200 µmol/L Met. Western blotting showed that Met reduced the expression of ADRP, but not TIP47 in the THP-1 macrophage-derived foam cells. CONCLUSION: Met could inhibit THP-1-derived foam cell formation induced by LPS, reduce intracellular lipid accumulation, and down-regulate the expression of ADRP.


Asunto(s)
Células Espumosas/efectos de los fármacos , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Metformina/farmacología , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Células Espumosas/metabolismo , Humanos , Hipoglucemiantes/farmacología , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Lipoproteínas LDL/farmacología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Perilipina-2 , Perilipina-3 , Acetato de Tetradecanoilforbol/farmacología , Triglicéridos/metabolismo , Proteínas de Transporte Vesicular/metabolismo
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(3): 360-3, 2016 Mar.
Artículo en Chino | MEDLINE | ID: mdl-26927556

RESUMEN

OBJECTIVE: To explore the correlations of the expression of mutant isocitrate dehydrogenase (IDH1) (R132H) protein with angiogenesis and cell proliferation in glioma. METHODS: We performed polymerase chain reaction-based IDH gene mutation screening in 385 glioma samples, and the subcellular localization and expression levels of IDH1 (R132H) was examined by immunohistochemistry (IHC). Ki-67 labeling index was introduced to determine the proliferation of glioma cells, and the microvessel density was measured through CD34 staining. Statistical analyses were performed to show the correlations of IDH1 mutation with cell proliferation and microvessel density. RESULTS: The mutant rates of IDH1 were about 50%-60% in grade II-III gliomas and secondary glioblastomas, which were significantly higher than those in pilocytic astrocytoma (grade I) and primary glioblastoma (grade IV). Moreover, the level of IDH1 (R132H) protein was positively correlated with Ki-67 labeling index and microvessel density. CONCLUSION: IDH mutation was common in grade II-III glioma and secondary glioblastoma, and the mutant IDH1 (R132H) might play a critical role in the cell proliferation and angiogenesis of glioma.


Asunto(s)
Proliferación Celular/genética , Glioma/genética , Isocitrato Deshidrogenasa/genética , Mutación Missense , Neovascularización Patológica/genética , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Antígenos CD34/metabolismo , Secuencia de Bases , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Glioma/irrigación sanguínea , Glioma/metabolismo , Humanos , Inmunohistoquímica , Isocitrato Deshidrogenasa/biosíntesis , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Neovascularización Patológica/metabolismo , Adulto Joven
14.
Mol Vis ; 17: 768-78, 2011 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-21527991

RESUMEN

PURPOSE: To explore the expression of the lens crystallins (αA- and ßB1-crystallin) in Xenopus laevis embryonic lens development and regeneration and to analyze the order of different crystallins generated in the regenerating lens. METHODS: Real Time-PCR, Immunofluorescence, and 2D-PAGE were used to analyze the expressions of αA-crystallin and ßB1-crystallin, and related factors during embryonic lens development and regeneration in Xenopus laevis. RESULTS: αA-crystallin and ßB1-crystallin were first detected at stage 29/30 during normal development, and the two crystallins were simultaneously detected in regeneration. During embryonic lens development, the relative expression level of the ßB1-crystallin gene was higher than that of the αA-crystallin gene. In the process of the lens regeneration, however, the relative expression level of the ßB1-crystallin gene was lower than that of the αA-crystallin gene. Throughout embryonic lens development, the two crystallin transcripts showed the same variation trends, and similar occurrence did in the regeneration process. Crystallins showed different localization and distribution during the ontogeny and regeneration, especially in the lens fiber region. 2D-electrophores revealed the patterns of the sequential synthesis of crystallins, with regard to the different classes and apparent variations of some auxiliary regulatory factors. CONCLUSIONS: The ontogeny and localization of the crystallins during embryonic lens development and regeneration indicated a different development program, although they have identical origins, the ectoderm. The expression level of crystallin transcripts displayed a consistent variation tendency, but the presence of appreciable differences was still exposed. In addition to stably producing the crystallins of different classes in accordance with established procedure, these auxiliary factors may perform the function, to some extent, because of significant changes in their expression throughout the process of lens regeneration.


Asunto(s)
Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Cristalino/metabolismo , Proteínas Recombinantes/metabolismo , Xenopus laevis/metabolismo , Cadena A de alfa-Cristalina/metabolismo , Cadena B de beta-Cristalina/metabolismo , Animales , Clonación Molecular , Ectodermo/metabolismo , Electroforesis en Gel Bidimensional , Embrión no Mamífero/metabolismo , Escherichia coli , Hibridación in Situ , Cristalino/embriología , Reacción en Cadena de la Polimerasa , Proteómica , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Proteínas Recombinantes/genética , Regeneración/genética , Xenopus laevis/embriología , Xenopus laevis/genética , Cadena A de alfa-Cristalina/genética , Cadena B de beta-Cristalina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA