Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Mol Cell ; 81(18): 3820-3832.e7, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34233158

RESUMEN

A metabolic imbalance between lipid synthesis and degradation can lead to hepatic lipid accumulation, a characteristic of patients with non-alcoholic fatty liver disease (NAFLD). Here, we report that high-fat-diet-induced sterol regulatory element-binding protein (SREBP)-1c, a key transcription factor that regulates lipid biosynthesis, impairs autophagic lipid catabolism via altered H2S signaling. SREBP-1c reduced cystathionine gamma-lyase (CSE) via miR-216a, which in turn decreased hepatic H2S levels and sulfhydration-dependent activation of Unc-51-like autophagy-activating kinase 1 (ULK1). Furthermore, Cys951Ser mutation of ULK1 decreased autolysosome formation and promoted hepatic lipid accumulation in mice, suggesting that the loss of ULK1 sulfhydration was directly associated with the pathogenesis of NAFLD. Moreover, silencing of CSE in SREBP-1c knockout mice increased liver triglycerides, confirming the connection between CSE, autophagy, and SREBP-1c. Overall, our results uncover a 2-fold mechanism for SREBP-1c-driven hepatic lipid accumulation through reciprocal activation and inhibition of hepatic lipid biosynthesis and degradation, respectively.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Hígado Graso/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Autofagia , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/fisiología , Línea Celular Tumoral , Dieta Alta en Grasa/efectos adversos , Hígado Graso/fisiopatología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Lipogénesis , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transducción de Señal/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Triglicéridos/metabolismo
2.
J Endocrinol ; 250(2): 49-65, 2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34060475

RESUMEN

Hyperandrogenemia (HA) is a hallmark of polycystic ovary syndrome (PCOS) and is an integral element of non-alcoholic fatty liver disease (NALFD) in females. Administering low-dose dihydrotestosterone (DHT) induced a normal weight PCOS-like female mouse model displaying NAFLD. The molecular mechanism of HA-induced NAFLD has not been fully determined. We hypothesized that DHT would regulate hepatic lipid metabolism via increased SREBP1 expression leading to NAFLD. We extracted liver from control and low-dose DHT female mice; and performed histological and biochemical lipid profiles, Western blot, immunoprecipitation, chromatin immunoprecipitation, and real-time quantitative PCR analyses. DHT lowered the 65 kD form of cytosolic SREBP1 in the liver compared to controls. However, DHT did not alter the levels of SREBP2 in the liver. DHT mice displayed increased SCAP protein expression and SCAP-SREBP1 binding compared to controls. DHT mice exhibited increased AR binding to intron-8 of SCAP leading to increased SCAP mRNA compared to controls. FAS mRNA and protein expression was increased in the liver of DHT mice compared to controls. p-ACC levels were unaltered in the liver. Other lipid metabolism pathways were examined in the liver, but no changes were observed. Our findings support evidence that DHT increased de novo lipogenic proteins resulting in increased hepatic lipid content via regulation of SREBP1 in the liver. We show that in the presence of DHT, the SCAP-SREBP1 interaction was elevated leading to increased nuclear SREBP1 resulting in increased de novo lipogenesis. We propose that the mechanism of action may be increased AR binding to an ARE in SCAP intron-8.


Asunto(s)
Dihidrotestosterona/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Síndrome del Ovario Poliquístico/metabolismo , Animales , Composición Corporal , Peso Corporal , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/inducido químicamente , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Transcripción Genética/efectos de los fármacos
3.
Toxicol Lett ; 348: 50-58, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34052307

RESUMEN

Antimony is a common environmental contaminant that causes biological toxicity in exposed populations worldwide. Previous studies have revealed that antimony promotes prostate cancer growth by stabilizing the c-Myc protein and mimicking androgen activity. However, the role of lncRNAs in the regulation of antimony-induced carcinogenesis remains unknown, and the precise mechanisms need to be explored. In the present study, we found that chronic exposure to antimony promoted cell growth and lipid metabolic disequilibrium in prostate cancer. Mechanistically, we identified a long noncoding RNA molecule, PCA3, that was substantially upregulated in LNCaP cells in response to long-term antimony exposure. Functional studies indicated that abnormal PCA3 expression modulated antimony-induced proliferation and cellular triglyceride and cholesterol levels. In addition, PCA3 levels were found to be inversely correlated with MIR-132-3 P levels by acting as a decoy for MIR-132-3P. Besides, SREBP1 directly interacted with MIR-132-3 P to increase cell growth and disrupt lipid metabolism by targeting its 3'UTR regions. Taken together, our results revealed that lncRNA PCA3 promotes antimony-induced lipid metabolic disorder in prostate cancer by targeting MIR-132-3 P/SREBP1 signaling.


Asunto(s)
Antígenos de Neoplasias/fisiología , Antimonio/toxicidad , Metabolismo de los Lípidos/efectos de los fármacos , MicroARNs/fisiología , Neoplasias de la Próstata/metabolismo , ARN Largo no Codificante/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/fisiología
4.
Br J Nutr ; 122(11): 1201-1211, 2019 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-31782376

RESUMEN

Disturbances in lipid metabolism are at the core of several health issues facing modern society, including fatty liver and obesity. The sterol regulatory element-binding protein 1 (SREBP-1) is one important transcription factor regulating lipid metabolism, but the relevant mechanism still remains unknown. The present study determined the transcriptional regulation of SREBP-1 and its target genes (including acetyl-CoA carboxylase α (accα), fatty acid synthase (fas) and stearoyl-CoA desaturase 1 (scd1)) in a freshwater teleost, grass carp Ctenopharyngodon idella. We cloned and characterised the 1988 bp, 2043 bp, 1632 bp and 1889 bp sequences of srebp-1, accα, scd1 and fas promoters, respectively. A cluster of putative binding sites of transcription factors, such as specific protein, yin yang 1, nuclear factor Y, sterol response elements (SRE) and enhancer box (E-box) element, were predicted on their promoter regions. Overexpression of nSREBP-1 reduced srebp-1 promoter activity, increased scd1 and fas promoter activity but did not influence accα promoter activity. The site-mutation and electrophoretic mobility shift assay analysis indicated that srebp-1, fas and scd1 promoters, but not accα promoter, possessed SRE. In Ctenopharyngodon idella kidney (CIK) cells of grass carp, nSREBP-1 overexpression significantly reduced srebp-1 mRNA expression and up-regulated miR-29 mRNA expression. The 3'UTR of srebp-1 possessed the potential miR-29 binding site and miR-29 up-regulated the luciferase activity of srebp-1 3'UTR and srebp-1 mRNA expression, implying a self-activating loop of SREBP-1 and miR-29 in grass carp. Based on the above-mentioned results, we found two novel transcriptional mechanisms for SREBP-1 in grass carp: (1) the auto-regulation sited on the SREBP-1 promoter regions was suppressive and (2) there was a self-activating loop of SREBP-1 and miR-29.


Asunto(s)
Carpas/metabolismo , Lipogénesis/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Acetil-CoA Carboxilasa/genética , Animales , Carpas/genética , Células Cultivadas , Clonación Molecular , Ácido Graso Sintasas/genética , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Riñón/química , Riñón/metabolismo , Lipogénesis/genética , MicroARNs/genética , MicroARNs/fisiología , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN/veterinaria , Estearoil-CoA Desaturasa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Transcripción Genética/fisiología , Transfección
5.
Mol Nutr Food Res ; 63(23): e1900773, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31482642

RESUMEN

SCOPE: Considerable evidence supports the view that high-fructose intake is associated with increased and early incidence of obesity and dyslipidemia. However, knowledge on physiopathological alterations introduced by fructose overconsumption is lacking. Therefore, an integrated omics analysis is carried out to investigate the consequences of short-term fructose overfeeding (SFO) and identify the underlying molecular mechanisms. METHODS AND RESULTS: SFO of rats demonstrates obvious histopathological hepatic lipid accumulation and significant elevation in adiposity, total cholesterol, and fasting plasma glucose levels. Integrated omics analysis demonstrates that SFO disturbed metabolic homeostasis and initiated metabolic stress. Hepatic lipogenesis pathways are also negatively impacted by SFO. Analysis of molecular networks generated by ingenuity pathway analysis (IPA) implicates involvement of the extracellular signal regulated kinase (ERK) signaling pathway in SFO and its consequences. Moreover, it is identified that an inherent negative feedback regulation of hepatic sterol regulatory element binding protein 1 (SREBP1) plays an active role in regulating hepatic de novo lipogenesis. CONCLUSION: The findings indicate that SFO disturbs metabolic homeostasis and that endogenous small molecules positively mediate SFO-induced metabolic adaption. The results also underline that an inherent regulatory mechanism of resilience occurs in response to fructose overconsumption, suggesting that efforts to maintain resilience can be a promising target to prevent and treat metabolic disorder-like conditions.


Asunto(s)
Fructosa/administración & dosificación , Resiliencia Psicológica , Estrés Fisiológico , Animales , Metabolismo Energético , Perfilación de la Expresión Génica , Metabolismo de los Lípidos , Hígado/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Metabolómica , Ratas , Ratas Sprague-Dawley , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología
6.
Sci Rep ; 9(1): 10369, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31316083

RESUMEN

de novo fatty acid biosynthesis (DNFA) is a hallmark adaptation of many cancers that supports survival, proliferation, and metastasis. Here we elucidate previously unexplored aspects of transcription regulation and clinical relevance of DNFA in cancers. We show that elevated expression of DNFA genes is characteristic of many tumor types and correlates with poor prognosis, especially in melanomas. Elevated DNFA gene expression depends on the SREBP1 transcription factor in multiple melanoma cell lines. SREBP1 predominantly binds to the transcription start sites of DNFA genes, regulating their expression by recruiting RNA polymerase II to promoters for productive transcription elongation. We find that SREBP1-regulated DNFA represents a survival trait in melanoma cells, regardless of proliferative state and oncogenic mutation status. Indeed, malignant melanoma cells exhibit elevated DNFA gene expression after the BRAF/MEK signaling pathway is blocked (e.g. by BRAF inhibitors), and DNFA expression remains higher in melanoma cells resistant to vemurafenib treatment than in untreated cells. Accordingly, DNFA pathway inhibition, whether by direct targeting of SREBP1 with antisense oligonucleotides, or through combinatorial effects of multiple DNFA enzyme inhibitors, exerts potent cytotoxic effects on both BRAFi-sensitive and -resistant melanoma cells. Altogether, these results implicate SREBP1 and DNFA enzymes as enticing therapeutic targets in melanomas.


Asunto(s)
Ácidos Grasos/biosíntesis , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Proteínas de Neoplasias/fisiología , Neoplasias Cutáneas/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Línea Celular Tumoral , Supervivencia Celular , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indazoles/farmacología , Estimación de Kaplan-Meier , Melanoma/genética , Melanoma/mortalidad , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/genética , Piperazinas/farmacología , Pronóstico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Transcripción Genética , Vemurafenib/farmacología
7.
FASEB J ; 33(9): 10089-10103, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31199678

RESUMEN

Sorafenib is a multikinase inhibitor that is effective in treating advanced liver cancer. Although its mechanism of action through several established cancer-related protein kinase targets is well-characterized, sorafenib induces variable responses among human tumors, and the cause for this variation is yet unknown. To investigate the underlying mechanisms, we applied mass spectrometry-based proteomic analysis to Huh7.5 human liver cancer cells and found that sorafenib significantly affected the expression of the key lipogenic enzymes, especially stearoyl coenzyme A desaturase 1 (SCD1), in these cells. Given that SCD1 catalyzes the most crucial and rate-limiting step in the synthesis of monounsaturated fatty acids (FAs), we performed a lipidomic analysis, which showed a dramatically altered lipid profile in sorafenib-treated cells. Detection and analysis of free FAs showed that the levels of monounsaturated FAs, including oleate, were significantly decreased in those cells treated by sorafenib. Addition of oleate protected liver cancer cells from sorafenib-induced death and alleviated the abnormalities of mitochondrial morphology and function caused by the drug. Treatment with sorafenib suppressed ATP production, resulting in AMPK activation via phosphorylation. Further secondary effects included reduction of the levels of sterol regulatory element-binding protein 1 (SREBP1) and the phosphorylation of mammalian target of rapamycin (mTOR) in liver cancer cells. These effects were partly abolished in the presence of compound C (an AMPK inhibitor) and ATP and adenosine, and SREBP1c overexpression also could be resistant to the effects of sorafenib, suggesting that the sorafenib-induced reduction in cell viability was mediated by the ATP-AMPK-mTOR-SREBP1 signaling pathway. Taken together, our results suggest that sorafenib's anticancer activity in liver cancer cells is based on the inhibition of ATP production, SCD1 expression, and monounsaturated FA synthesis. In addition, the decreased monounsaturated FA synthesis further triggered the more serious reduction of ATP production in sorafenib-treated cells. To our knowledge, this is the first evidence that sorafenib disrupts lipogenesis and triggers liver cancer cell death by targeting SCD1 through the ATP-AMPK-mTOR-SREBP1 pathway.-Liu, G., Kuang, S., Cao, R., Wang, J., Peng, Q., Sun, C. Sorafenib kills liver cancer cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids via the ATP-AMPK-mTOR- SREBP1 signaling pathway.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Sorafenib/farmacología , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/fisiología , Animales , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Lipidómica , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Ácido Oléico/farmacología , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes/metabolismo , Sorafenib/uso terapéutico , Estearoil-CoA Desaturasa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Serina-Treonina Quinasas TOR/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Clin Endocrinol Metab ; 104(10): 4783-4792, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31150065

RESUMEN

CONTEXT: LH receptor (LHR) expression has been shown to be regulated posttranscriptionally by LHR mRNA binding protein (LRBP) in rodent and human ovaries. LRBP was characterized as mevalonate kinase. The gene that encodes mevalonate kinase is a member of a family of genes that encode enzymes involved in lipid synthesis and are regulated by the transcription factor sterol regulatory element binding proteins (SREBPs). OBJECTIVE: The current study examined the regulation of LHR mRNA expression in human granulosa-lutein cells in response to alterations in cholesterol metabolism. DESIGN: Using atorvastatin, an inhibitor of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase to inhibit cholesterol biosynthesis, we examined its effect on LHR mRNA expression. The effect of atorvastatin on SREBP and mRNA expression as well as LHR mRNA binding protein expression was examined. Finally, the effect of atorvastatin on human chorionic gonadotropin (hCG)-stimulated progesterone production and the expression of key steroidogenic enzymes was also examined. RESULTS: Statin treatment reduced LHR mRNA expression by increasing the levels of SREBP1a and SREBP2, leading to an increase in LRBP. RNA gel shift assay showed that increased binding of LHR mRNA to LRBP occurred in response to atorvastatin, leading to LHR mRNA degradation. The granulosa-lutein cells pretreated with atorvastatin also showed decreased responsiveness to hCG by decreasing the mRNA and protein expression of steroidogenic enzymes. Atorvastatin also attenuated LH/hCG-induced progesterone production. CONCLUSION: These results imply that LHR mRNA expression by the human granulosa-lutein cells is regulated by cholesterol, through a mechanism involving SREBP and SREBP cleavage activating protein serving as the cholesterol sensor.


Asunto(s)
Células Lúteas/metabolismo , Receptores de HL/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/fisiología , Atorvastatina/farmacología , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Lúteas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/fisiología
9.
Environ Toxicol Pharmacol ; 69: 104-111, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31004931

RESUMEN

Mono-2-ethylhexyl phthalate (MEHP) is a major bioactive metabolite in the widely used industrial plasticizer diethylhexyl phthalate (DEHP) that has been found to be toxic to the liver. The aim of this study is to determine whether MEHP exposure can change the expression of fatty acid metabolism-related genes in HepG2 cells, which might be related to non-alcoholic fatty liver disease (NAFLD). The results revealed that exposure to MEHP promoted lipid accumulation in HepG2 cells. The levels of intracellular triglycerides in the hepatocytes increased after exposure to 0.8-100 µM MEHP for 24 h and 48 h. The genetic expressions of SREBP-1c, ChREBP, ACC1, FASN, and SCD significantly increased at 6 h after exposure to MEHP. At 24 h, the expression of the SREBP-1c and ChREBP genes remained increased, while the expression of the FASN and SCD genes decreased. At 48 h, the expression of SREBP-1c, ChREBP, ACC1, FASN, and SCD decreased. Furthermore, the levels of proteins including ACC1, FASN, SCD, and ChREBP (except SREBP-1c) increased at 24 h. These findings suggest that MEHP exposure can promote fatty acid synthesis in hepatocytes by regulating the expression of relevant genes and proteins, contributing to NAFLD.


Asunto(s)
Dietilhexil Ftalato/análogos & derivados , Metabolismo de los Lípidos/efectos de los fármacos , Acetil-CoA Carboxilasa/fisiología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Dietilhexil Ftalato/toxicidad , Acido Graso Sintasa Tipo I/fisiología , Células Hep G2 , Humanos , Estearoil-CoA Desaturasa/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Triglicéridos/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 39(3): 373-386, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30700132

RESUMEN

Objective- APOA5 variants are strongly associated with hypertriglyceridemia, as well as increased risks of cardiovascular disease and acute pancreatitis. Hypertriglyceridemia in apo AV dysfunction often aggravates by environmental factors such as high-carbohydrate diets or aging. To date, the molecular mechanisms by which these environmental factors induce hypertriglyceridemia are poorly defined, leaving the high-risk hypertriglyceridemia condition undertreated. Previously, we reported that LXR (liver X receptor)-SREBP (sterol regulatory element-binding protein)-1c pathway regulates large-VLDL (very low-density lipoprotein) production induced by LXR agonist. However, the pathophysiological relevance of the finding remains unknown. Approach and Results- Here, we reconstitute the environment-induced hypertriglyceridemia phenotype of human APOA5 deficiency in Apoa5-/- mice and delineate the role of SREBP-1c in vivo by generating Apoa5-/- ;Srebp-1c-/- mice. The Apoa5-/- mice, which showed moderate hypertriglyceridemia on a chow diet, developed severe hypertriglyceridemia on high-carbohydrate feeding or aging as seen in patients with human apo AV deficiency. These responses were nearly completely abolished in the Apoa5-/- ;Srebp-1c-/- mice. Further mechanistic studies revealed that in response to these environmental factors, SREBP-1c was activated to increase triglyceride synthesis and to permit the incorporation of triglyceride into abnormally large-VLDL particles, which require apo AV for efficient clearance. Conclusions- Severe hypertriglyceridemia develops only when genetic factors (apo AV deficiency) and environmental effects (SREBP-1c activation) coexist. We demonstrate that the regulated production of large-sized VLDL particles via SREBP-1c determines plasma triglyceride levels in apo AV deficiency. Our findings explain the long-standing enigma of the late-onset hypertriglyceridemia phenotype of apo AV deficiency and suggest a new approach to treat hypertriglyceridemia by targeting genes that mediate environmental effects.


Asunto(s)
Apolipoproteína A-V/deficiencia , Hipertrigliceridemia/sangre , Lipoproteínas VLDL/biosíntesis , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Envejecimiento/metabolismo , Alimentación Animal/efectos adversos , Animales , Apolipoproteína A-V/genética , Apolipoproteínas/sangre , Quilomicrones/metabolismo , Femenino , Fructosa/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Interacción Gen-Ambiente , Humanos , Hidrocarburos Fluorados/farmacología , Hipertrigliceridemia/inducido químicamente , Hipertrigliceridemia/genética , Lípidos/sangre , Receptores X del Hígado/agonistas , Receptores X del Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Aceite de Oliva/toxicidad , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/deficiencia , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Sulfonamidas/farmacología
11.
Nutrients ; 10(10)2018 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-30274245

RESUMEN

De novo lipogenesis (DNL) is a complex and highly regulated process in which carbohydrates from circulation are converted into fatty acids that are then used for synthesizing either triglycerides or other lipid molecules. Dysregulation of DNL contributes to human diseases such as obesity, type 2 diabetes, and cardiovascular diseases. Thus, the lipogenic pathway may provide a new therapeutic opportunity for combating various pathological conditions that are associated with dysregulated lipid metabolism. Hepatic DNL has been well documented, but lipogenesis in adipocytes and its contribution to energy homeostasis and insulin sensitivity are less studied. Recent reports have gained significant insights into the signaling pathways that regulate lipogenic transcription factors and the role of DNL in adipose tissues. In this review, we will update the current knowledge of DNL in white and brown adipose tissues with the focus on transcriptional, post-translational, and central regulation of DNL. We will also summarize the recent findings of adipocyte DNL as a source of some signaling molecules that critically regulate energy metabolism.


Asunto(s)
Tejido Adiposo/metabolismo , Homeostasis , Lipogénesis/fisiología , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Metabolismo Energético , Regulación de la Expresión Génica , Humanos , Resistencia a la Insulina , Receptores X del Hígado , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Transcripción Genética
12.
Arch Gynecol Obstet ; 298(5): 961-969, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30206735

RESUMEN

PURPOSE: The prevalence of endometrial cancer (EC) is increasing worldwide. Progestin therapy is effective for both early stage EC patients who require preserving fertility and advanced or recurrent patients. Progestin resistance resulting from downregulation of progesterone receptor (PR) remains a major problem, and its mechanism is currently unclear. It was demonstrated that Sirtuin 1 (SIRT1), forkhead transcription factor 1 (FoxO1) and sterol regulatory element binding protein-1 (SREBP-1) may act as a pathway and play crucial roles in the development of EC in our previous studies. In the present study, we investigated the effect on the development of progestin resistance and the relationship with PR of SIRT1/FoxO1/SREBP-1. METHODS: A progestin-resistant Ishikawa cell line was established in the stimulation and selection of medroxyprogesterone acetate (MPA), and the resistance was analyzed by MTT assay, flow cytometry, and Transwell invasion assay. qRT-PCR and western blotting were conducted to detect the expression of SIRT1, FoxO1, SREBP-1 and PR. SIRT1 knockdown progestin-resistant cells were established by lentiviral transduction. RESULTS: The new progestin-resistant cell line presented sufficient resistance to MPA in aspects of proliferation, distribution of cell cycle and apoptosis compared with original Ishikawa cells. Besides, the invasion capability of progestin-resistant cells was observably increased. In both protein and mRNA levels, SIRT1 and SREBP-1 were upregulated in progestin-resistant cells, while PR and FoxO1 were downregulated. SIRT1 was knocked down by lentivirus transfection in progestin-resistant cells, resulting in upregulation of PR, FoxO1 and downregulation of SREBP-1, thereby SIRT1 knockdown cells were more sensitive to MPA compared with progestin-resistant cells. CONCLUSION: SIRT1/FoxO1/SREBP-1 act as a pathway targeting PR and involve in the development of progestin resistance in Ishikawa cells.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Neoplasias Endometriales/tratamiento farmacológico , Proteína Forkhead Box O1/fisiología , Progestinas/uso terapéutico , Sirtuina 1/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Proteína Forkhead Box O1/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Acetato de Medroxiprogesterona/uso terapéutico , Progestinas/farmacología , ARN Mensajero/análisis , Receptores de Progesterona/genética , Sirtuina 1/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
13.
PLoS One ; 13(5): e0196704, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29723221

RESUMEN

Sterol Regulatory Element Binding Protein-1 (SREBP-1) is a conserved transcription factor of the basic helix-loop-helix leucine zipper family (bHLH-Zip) that plays a central role in regulating expression of genes of carbohydrate and fatty acid metabolism in the liver. SREBP-1 activity is essential for the control of insulin-induced anabolic processes during the fed state. In addition, SREBP-1 regulates expression of key molecules in the insulin signaling pathway, including insulin receptor substrate 2 (IRS2) and a subunit of the phosphatidylinositol 3-kinase (PI3K) complex, PIK3R3, suggesting that feedback mechanisms exist between SREBP-1 and this pathway. Nevertheless, the overall contribution of SREBP-1 activity to maintain insulin signal transduction is unknown. Furthermore, Akt is a known activator of mTORC1, a sensor of energy availability that plays a fundamental role in metabolism, cellular growth and survival. We have silenced SREBP-1 and explored the impact on insulin signaling and mTOR in mice under fed, fasted and refed conditions. No alterations in circulating levels of insulin were observed. The studies revealed that depletion of SREBP-1 had no impact on IRS1Y612, AktS473, and downstream effectors GSK3αS21 and FoxO1S256 during the fed state. Nevertheless, reduced levels of these molecules were observed under fasting conditions. These effects were not associated with changes in phosphorylation of mTOR. Overall, our data indicate that the contribution of SREBP-1 to maintain insulin signal transduction in liver is modest.


Asunto(s)
Insulina/fisiología , Hígado/metabolismo , Transducción de Señal/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Animales , Glucemia/análisis , Células Cultivadas , Metabolismo Energético/genética , Ayuno/metabolismo , Proteína Forkhead Box O1/biosíntesis , Proteína Forkhead Box O1/genética , Vectores Genéticos , Glucoquinasa/metabolismo , Gluconeogénesis/genética , Glucógeno Sintasa Quinasa 3/biosíntesis , Glucógeno Sintasa Quinasa 3/genética , Hepatocitos/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Periodo Posprandial/fisiología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Serina-Treonina Quinasas TOR/metabolismo
14.
Transl Res ; 196: 17-30, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29421523

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in the general population. Its severity ranges from simple steatosis to cirrhosis. C-C chemokine ligand type 5 or RANTES (Regulated upon Activation, Normal T-cell Expressed, and Secreted) plays an important role in the progression of hepatic inflammation and fibrosis. Our objective was to examine the preventive and therapeutic effects of maraviroc (MVC), a C-C chemokine receptor 5 antagonist, on liver pathology in an NAFLD mouse model. A total of 60 male C57BL/6 mice were randomly assigned to 1 of 4 groups: (1) high-fat diet (HFD) group or control group, (2) preventive group (HFD group plus MVC in drinking water since the beginning of the study), (3) early-therapeutic group (HFD group plus MVC in drinking starting at week 24 of the study), and (4) late-therapeutic group (HFD group plus MVC in drinking water starting at week 36 of the study). All mice were sacrificed at week 48. The hepatic triglyceride concentration in the HFD group was significantly higher than that in the groups treated with MVC at any time. Gene expression associated with lipogenesis (diacylglycerol acyltransferase 2 and proliferator-activated receptor-γ), insulin resistance (insulin receptor substrate-2), and ß-oxidation (carnitine palmitoyltransferase 1A and acyl-CoA oxidase) was significantly reduced in all the groups treated with MVC. In summary, the beneficial effect of MVC on hepatic steatosis is maintained throughout the study.


Asunto(s)
Antagonistas de los Receptores CCR5/uso terapéutico , Ciclohexanos/uso terapéutico , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Triazoles/uso terapéutico , Triglicéridos/metabolismo , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Resistencia a la Insulina , Lipogénesis/efectos de los fármacos , Masculino , Maraviroc , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , PPAR gamma/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología
15.
PLoS One ; 13(2): e0192447, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29389988

RESUMEN

SCOPE: Conjugated linoleic acids are linoleic acid isomers found in the diet that can also be produced through bacterial metabolism of polyunsaturated fatty acids. Our objective was to evaluate the contribution of fatty acid metabolites produced from polyunsaturated fatty acids by the gut microbiota in vivo to regulation of hepatic lipid metabolism and steatosis. METHODS AND RESULTS: In mice with depleted n-3 polyunsaturated fatty acids, we observed an accumulation of trans-11,trans-13 CLA and cis-9,cis-11 conjugated linoleic acids in the liver tissue that were associated with an increased triglyceride content and expression of lipogenic genes. We used an in vitro model to evaluate the impact of these two conjugated linoleic acids on hepatic lipid metabolism. In HepG2 cells, we observed that only trans-11,trans-13 conjugated linoleic acids recapitulated triglyceride accumulation and increased lipogenic gene expression, which is a phenomenon that may implicate the nuclear factors sterol regulatory element binding protein 1c (SREBP-1c) and carbohydrate-responsive element-binding protein (ChREBP). CONCLUSION: The trans-11,trans-13 conjugated linoleic acids can stimulate hepatic lipogenesis, which supports the conclusion that gut microbiota and related metabolites should be considered in the treatment of non-alcoholic liver disease.


Asunto(s)
Hígado Graso/inducido químicamente , Ácidos Linoleicos Conjugados/farmacología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Factores de Transcripción/fisiología
16.
Mol Nutr Food Res ; 62(6): e1700737, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29380937

RESUMEN

SCOPE: To investigate the effect of sulforaphane (SFN) on the abnormal lipid metabolism and underlying mechanisms. METHODS AND RESULTS: Models with abnormal lipid metabolism are established both in rats and human hepatocytes. Hepatic steatosis is detected by hematoxylin and eosin and oil red O staining. The structure of endoplasmic reticulum is visualized by transmission electron microscopy. The expressions of X-box binding protein 1 (XBP1), protein kinase-like ER kinase (PERK), sterol regulatory element binding protein-1c (SREBP1c), and lipogenic enzymes are determined by real-time PCR and western blot analysis. SFN lowers the content of triglyceride and cholesterol. SFN alleviates the swelling of ER and decreases the perimeter of ER. SFN significantly decreases the expressions of acetyl CoA carboxylase 1 (ACC1), stearoyl-CoA desaturase 1 (SCD1), and fatty acid synthase. SFN inhibits SREBP1c by blocking the PERK. Meanwhile, SFN suppresses ACC1 and SCD1 via blocking the formation of splicing-type XBP1. The key roles of XBP1 and SREBP1c in SFN-reduced lipid droplets are confirmed by a timed sequence of measurement according to time points. CONCLUSION: SFN improves abnormal lipid metabolism via both ER-stress-dependent and -independent pathways.


Asunto(s)
Acetil-CoA Carboxilasa/fisiología , Estrés del Retículo Endoplásmico/fisiología , Ácido Graso Sintasas/fisiología , Isotiocianatos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Estearoil-CoA Desaturasa/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Proteína 1 de Unión a la X-Box/fisiología , Animales , Células Cultivadas , Humanos , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Masculino , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Sulfóxidos
17.
J Agric Food Chem ; 66(5): 1197-1205, 2018 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-29323924

RESUMEN

Akt serine/threonine kinase acts as a central mediator in the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, regulating a series of biological processes. In lipid metabolism, Akt activation regulates a series of gene expressions, including genes related to intracellular fatty acid synthesis. However, the regulatory mechanisms of Akt in dairy goat mammary lipid metabolism have not been elaborated. In this study, the coding sequences of goat Akt1 gene were cloned and analyzed. Gene expression of Akt1 in different lactation stages was also investigated. For in vitro studies, a eukaryotic expression vector of Akt1 was constructed and transfected to goat mammary epithelial cells (GMECs), and specific inhibitors of Akt/mammalian target of rapamycin (mTOR) signaling were applied to GMECs. Results showed that Akt1 protein was highly conserved, and its mRNA was highly expressed in midlactation. In vitro studies indicated that Akt1 phosphorylation activated mTOR and subsequently enhanced sterol regulatory element binding protein 1 (SREBP1), thus increasing intracellular triacylglycerol content. Inhibition of Akt/mTOR signaling down-regulated the gene expression of lipogenic genes. Overall, Akt1 plays an important role in regulating de novo fatty acid synthesis in goat mammary epithelial cells, and this process probably is through the mTOR/SREBP1 axis.


Asunto(s)
Ácidos Grasos/biosíntesis , Cabras , Glándulas Mamarias Animales/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Epitelio/metabolismo , Regulación de la Expresión Génica/fisiología , Lipogénesis/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/fisiología
18.
Mol Nutr Food Res ; 62(5)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29210166

RESUMEN

SCOPE: This study aims to characterize the effect of fenugreek (Trigonella foenum-graecum) seed and its phytoceutical trigonelline in antimetabolic inflammation and ameliorating overproduction of very low density lipoprotein (VLDL) in insulin resistance. METHODS AND RESULTS: Two groups of genetic hyperlipidemic mice generated by depletion of cAMP responsive element binding protein H (CREBH) are fed either a chow containing 2% fenugreek seed or vehicle for 7 weeks. Q-RT-PCR and immunoblotting analysis demonstrated that fenugreek seed containing diet inhibits hepatic SREBP-1c activation and the subsequent de novo lipogenesis by enhancing expression of insulin-inducible gene-1 (Insig-1) and gene-2 (Insig-2). mRNA expression of PPARα and its target genes that are involved in fatty acid ß-oxidation are also upregulated in the fenugreek seed fed-mice which is accompanied by significantly reduced hepatic lipid accumulation and VLDL secretion, improved endoplasmic reticulum (ER) stress, and ameliorated metabolic inflammation. These actions enhance insulin sensitivity and improve hyperlipidemia. In vitro, treating a rat hepatoma cell line, McA-RH7777 (McA), with trigonelline is able to recapitulate the results observed in vivo. CONCLUSIONS: This study unveils a novel mechanism of fenugreek seed and trigonelline in countering hepatic VLDL overproduction and insulin resistance by enhancing the Insig signaling pathways and ameliorating metabolic inflammatory stress in the liver.


Asunto(s)
Alcaloides/farmacología , Resistencia a la Insulina , Lipoproteínas VLDL/antagonistas & inhibidores , Proteínas de la Membrana/fisiología , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Lipoproteínas VLDL/biosíntesis , Ratones , Ratas , Transducción de Señal/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Trigonella , Células Tumorales Cultivadas
19.
J Dairy Res ; 84(3): 283-288, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28831970

RESUMEN

This research communication describes the profile of gene expression related to the synthesis of yak milk as determined via quantitative reverse transcription polymerase chain reaction (RT-qPCR). Significant up-regulation during lactation were observed in genes related to fatty acid (FA) uptake from blood (LPL, CD36), intracellular FA transport (FABP3), intracellular FA activation of long- and short-chain FAs (ACSS1, ACSS2, ACSL1), de novo synthesis (ACACA), desaturation (SCD), triacyglycerol (TAG) synthesis (AGPAT6, GPAM, LPIN1), lipid droplet formation (PLIN2, BTN1A1, XDH), ketone body utilisation (BDH1, OXCT1), and transcription regulation (THRSP, PPARGC1A). In particular, intracellular de novo FA synthesis (ACSS2, ACACA, and FABP3) and TAG synthesis (GPAM, AGPAT6, and LPIN1), whose regulation might be orchestrated as part of the gene network under the control of SERBF1 in the milk fat synthesis process, were more activated compared to levels in dairy cows. However, the genes involved in lipid droplet formation (PLIN2, XDH, and BTN1A1) were expressed at lower levels compared to those in dairy cows, where these genes are mainly controlled by the PPARG regulator.


Asunto(s)
Bovinos/metabolismo , Expresión Génica , Lactancia/fisiología , Lípidos/biosíntesis , Glándulas Mamarias Animales/metabolismo , Leche/química , Adaptación Fisiológica , Altitud , Animales , Bovinos/genética , China , Ácidos Grasos/biosíntesis , Ácidos Grasos/metabolismo , Femenino , Regulación de la Expresión Génica , Lípidos/genética , Lipogénesis/genética , PPAR gamma/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Especificidad de la Especie , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Transcriptoma/fisiología , Triglicéridos
20.
Eur Neuropsychopharmacol ; 27(6): 589-598, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27492885

RESUMEN

Schizophrenia is a serious psychotic disorder, with disabling symptoms and markedly reduced life expectancy. The onset is usually in late adolescence or early adulthood, which in time overlaps with the maturation of the brain including the myelination process. Interestingly, there seems to be a link between myelin abnormalities and schizophrenia. The oligodendrocyte-derived myelin membranes in the CNS are highly enriched for lipids (cholesterol, phospholipids and glycosphingolipids), thereby pointing at lipid homeostasis as a relevant target for studying the genetics and pathophysiology of schizophrenia. The biosynthesis of fatty acids and cholesterol is regulated by the sterol regulatory element binding protein (SREBP) transcription factors SREBP1 and SREBP2, which are encoded by the SREBF1 and SREBF2 genes on chromosome 17p11.2 and 22q13.2, respectively. Here we review the evidence for the involvement of SREBF1 and SREBF2 as genetic risk factors in schizophrenia and discuss the role of myelination and SREBP-mediated lipid biosynthesis in the etiology, pathophysiology and drug treatment of schizophrenia.


Asunto(s)
Antipsicóticos/uso terapéutico , Lipogénesis/fisiología , Esquizofrenia/genética , Esquizofrenia/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Proteína 2 de Unión a Elementos Reguladores de Esteroles/fisiología , Antipsicóticos/farmacología , Humanos , Lipogénesis/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA