Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
J Biol Chem ; 296: 100312, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33482195

RESUMEN

Elevated plasma triglycerides are a risk factor for coronary artery disease, which is the leading cause of death worldwide. Lipoprotein lipase (LPL) reduces triglycerides in the blood by hydrolyzing them from triglyceride-rich lipoproteins to release free fatty acids. LPL activity is regulated in a nutritionally responsive manner by macromolecular inhibitors including angiopoietin-like proteins 3 and 4 (ANGPTL3 and ANGPTL4). However, the mechanism by which ANGPTL3 inhibits LPL is unclear, in part due to challenges in obtaining pure protein for study. We used a new purification protocol for the N-terminal domain of ANGPTL3, removing a DNA contaminant, and found DNA-free ANGPTL3 showed enhanced inhibition of LPL. Structural analysis showed that ANGPTL3 formed elongated, flexible trimers and hexamers that did not interconvert. ANGPTL4 formed only elongated flexible trimers. We compared the inhibition of ANGPTL3 and ANGPTL4 using human very-low-density lipoproteins as a substrate and found both were noncompetitive inhibitors. The inhibition constants for the trimeric ANGPTL3 (7.5 ± 0.7 nM) and ANGPTL4 (3.6 ± 1.0 nM) were only 2-fold different. Heparin has previously been reported to interfere with ANGPTL3 binding to LPL, so we questioned if the negatively charged heparin was acting in a similar fashion to the DNA contaminant. We found that ANGPTL3 inhibition is abolished by binding to low-molecular-weight heparin, whereas ANGPTL4 inhibition is not. Our data show new similarities and differences in how ANGPTL3 and ANGPTL4 regulate LPL and opens new avenues of investigating the effect of heparin on LPL inhibition by ANGPTL3.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/química , Proteínas Similares a la Angiopoyetina/química , Enfermedad de la Arteria Coronaria/genética , Lipoproteína Lipasa/química , Conformación Proteica , Proteína 3 Similar a la Angiopoyetina , Proteína 4 Similar a la Angiopoyetina/genética , Proteína 4 Similar a la Angiopoyetina/ultraestructura , Proteínas Similares a la Angiopoyetina/genética , Proteínas Similares a la Angiopoyetina/ultraestructura , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/patología , Heparina/farmacología , Humanos , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/ultraestructura , Lipoproteínas VLDL/química , Lipoproteínas VLDL/genética , Unión Proteica/efectos de los fármacos , Especificidad por Sustrato , Triglicéridos/sangre
2.
Circ Res ; 127(11): 1347-1361, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-32912065

RESUMEN

RATIONALE: Single-nucleotide polymorphisms near the ILRUN (inflammation and lipid regulator with ubiquitin-associated-like and NBR1 [next to BRCA1 gene 1 protein]-like domains) gene are genome-wide significantly associated with plasma lipid traits and coronary artery disease (CAD), but the biological basis of this association is unknown. OBJECTIVE: To investigate the role of ILRUN in plasma lipid and lipoprotein metabolism. METHODS AND RESULTS: ILRUN encodes a protein that contains a ubiquitin-associated-like domain, suggesting that it may interact with ubiquitinylated proteins. We generated mice globally deficient for Ilrun and found they had significantly lower plasma cholesterol levels resulting from reduced liver lipoprotein production. Liver transcriptome analysis uncovered altered transcription of genes downstream of lipid-related transcription factors, particularly PPARα (peroxisome proliferator-activated receptor alpha), and livers from Ilrun-deficient mice had increased PPARα protein. Human ILRUN was shown to bind to ubiquitinylated proteins including PPARα, and the ubiquitin-associated-like domain of ILRUN was found to be required for its interaction with PPARα. CONCLUSIONS: These findings establish ILRUN as a novel regulator of lipid metabolism that promotes hepatic lipoprotein production. Our results also provide functional evidence that ILRUN may be the casual gene underlying the observed genetic associations with plasma lipids at 6p21 in human.


Asunto(s)
Hepatocitos/metabolismo , Lipoproteínas/sangre , Hígado/metabolismo , Animales , Glucemia/metabolismo , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , HDL-Colesterol/sangre , HDL-Colesterol/genética , Regulación de la Expresión Génica , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/genética , Células HEK293 , Humanos , Lipoproteínas/genética , Lipoproteínas VLDL/sangre , Lipoproteínas VLDL/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Unión Proteica , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Transcriptoma , Triglicéridos/sangre , Triglicéridos/genética , Ubiquitinación
3.
Curr Opin Lipidol ; 31(3): 132-139, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32332433

RESUMEN

PURPOSE OF REVIEW: To critically appraise new insights into the biology of remnant lipoproteins and their putative role in the pathophysiology of atherosclerotic cardiovascular disease, and to compare the atherogenicity of remnant particles with that of low-density lipoproteins (LDL). RECENT FINDINGS: New in-vivo stable isotope tracer studies of the kinetics of apoB48 and apoB100-containing lipoproteins in postprandial conditions have revealed that apoB48-containing very low-density lipoproteins (VLDL) accumulated markedly in hypertriglyceridemic patients. These intestinally-derived particles were cleared slowly, and represented up to 25% of circulating VLDL; as part of the remnant particle population, they may increase cardiovascular risk. Importantly, the PCSK9 inhibitor, evolocumab, was shown to reduce remnant levels (-29%) during the postprandial period in diabetic patients on statin therapy - an effect which may be additive to that of LDL-cholesterol reduction in conferring cardiovascular benefit. In recent Mendelian randomization studies, the effect of lowering triglyceride-rich lipoproteins or LDL-cholesterol translated to similar clinical benefit per unit of apoB. Finally, in randomized trials involving statin-treated patients with atherosclerotic cardiovascular disease, remnant cholesterol levels were associated with coronary atheroma progression independently of LDL-cholesterol. SUMMARY: Overall, data from observational studies in large cohorts, Mendelian randomization studies, meta-regression analyses, and post-hoc analyses of randomized trials are consistent with the contention that remnants are highly atherogenic particles and contribute to the atherosclerotic burden in an equivalent manner to that of LDL.


Asunto(s)
Apolipoproteína B-100/genética , Aterosclerosis/genética , Enfermedades Cardiovasculares/genética , Proproteína Convertasa 9/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Aterosclerosis/metabolismo , Aterosclerosis/patología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Humanos , Marcaje Isotópico , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Inhibidores de PCSK9
4.
J Biol Chem ; 294(21): 8577-8591, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30971429

RESUMEN

Enhancer of zeste homolog 2 (EZH2), an epigenetic regulator that plays a key role in cell differentiation and oncogenesis, was reported to promote adipogenic differentiation in vitro by catalyzing trimethylation of histone 3 lysine 27. However, inhibition of EZH2 induced lipid accumulation in certain cancer and hepatocyte cell lines. To address this discrepancy, we investigated the role of EZH2 in adipogenic differentiation and lipid metabolism using primary human and mouse preadipocytes and adipose-specific EZH2 knockout (KO) mice. We found that the EZH2-selective inhibitor GSK126 induced lipid accumulation in human adipocytes, without altering adipocyte differentiation marker gene expression. Moreover, adipocyte-specific EZH2 KO mice, generated by crossing EZH2 floxed mice with adiponectin-Cre mice, displayed significantly increased body weight, adipose tissue mass, and adipocyte cell size and reduced very low-density lipoprotein (VLDL) levels, as compared with littermate controls. These phenotypic alterations could not be explained by differences in feeding behavior, locomotor activity, metabolic energy expenditure, or adipose lipolysis. In addition, human adipocytes treated with either GSK126 or vehicle exhibited comparable rates of glucose-stimulated triglyceride accumulation and fatty acid uptake. Mechanistically, lipid accumulation induced by GSK126 in adipocytes was lipoprotein-dependent, and EZH2 inhibition or gene deletion promoted lipoprotein-dependent lipid uptake in vitro concomitant with up-regulated apolipoprotein E (ApoE) gene expression. Deletion of ApoE blocked the effects of GSK126 to promote lipoprotein-dependent lipid uptake in murine adipocytes. Collectively, these results indicate that EZH2 inhibition promotes lipoprotein-dependent lipid accumulation via inducing ApoE expression in adipocytes, suggesting a novel mechanism of lipid regulation by EZH2.


Asunto(s)
Adipocitos/metabolismo , Apolipoproteínas E/metabolismo , Diferenciación Celular , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Lipogénesis , Lipólisis , Adipocitos/citología , Animales , Apolipoproteínas E/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Humanos , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Ratones , Regulación hacia Arriba
5.
J Cell Physiol ; 235(9): 6246-6256, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32012270

RESUMEN

To evaluate the effects of fatty acids on endoplasmic reticulum (ER) stress, oxidative stress, and lipid damage. We treated BRL3A rat liver cells with, linoleic (LA), linolenic, oleic (OA), palmitic (PA), palmitoleic (POA), or stearic (SA) acid for 12 hr. The characteristics of cell lipid deposition, oxidative stress indexes, ER stress markers, nuclear factor κB p65 (NF-κB p65), lipid synthesis and transport regulators, and cholesterol metabolism regulators were analyzed. Endoplasmic chaperones like glucose-regulated protein 78, CCAAT-enhancer-binding protein, NF-κB p65, hydrogen peroxide, and malonaldehyde in PA- and SA-treated cells were significantly higher than in other treated cells. Deposition of fatty acids especially LA and POA were significantly increased than in other treated cells. De novo lipogenesis regulators sterol regulatory element-binding protein 1c, fatty acid synthase, and acetyl-coenzyme A carboxylase 1 (ACC1) expression were significantly increased in all fatty acid stimulation groups, and PA- and SA-treated cells showed lower p-ACC1 expression and higher scd1 expression than other fatty acid groups. Very low-density lipoprotein synthesis and apolipoprotein B100 expression in free fatty acids treated cells were significantly lower than control. PA, SA, OA, and POA had shown significantly increased cholesterol synthesis than other treated cells. PA and SA showed the lower synthesis of cytochrome P7A1 and total bile acids than other fatty acids treated cells. Excess of saturated fatty acids led to severe ER and oxidative stress. Excess unsaturated fatty acids led to increased lipid deposition in cultured hepatocytes. A balanced fatty acid intake is needed to maintain lipid homeostasis.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Ácidos Grasos/farmacología , Hígado Graso/tratamiento farmacológico , Lipogénesis/genética , Acetil-CoA Carboxilasa/genética , Animales , Células Cultivadas , Ácido Graso Sintasas/genética , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Hígado Graso/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ácido Linoleico/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/biosíntesis , Lípidos/genética , Lipogénesis/efectos de los fármacos , Lipoproteínas VLDL/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Ácido Oléico/farmacología , Estrés Oxidativo/efectos de los fármacos , Ácido Palmítico/farmacología , Ratas , Ácidos Esteáricos/farmacología , Ácido alfa-Linolénico/farmacología
6.
FASEB J ; 33(7): 8666-8687, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31002535

RESUMEN

We recently described myonectin (also known as erythroferrone) as a novel skeletal muscle-derived myokine with metabolic functions. Here, we use a genetic mouse model to determine myonectin's requirement for metabolic homeostasis. Female myonectin-deficient mice had larger gonadal fat pads and developed mild insulin resistance when fed a high-fat diet (HFD) and had reduced food intake during refeeding after an unfed period but were otherwise indistinguishable from wild-type littermates. Male mice lacking myonectin, however, had reduced physical activity when fed ad libitum and in the postprandial state but not during the unfed period. When stressed with an HFD, myonectin-knockout male mice had significantly elevated VLDL-triglyceride (TG) and strikingly impaired lipid clearance from circulation following an oral lipid load. Fat distribution between adipose and liver was also altered in myonectin-deficient male mice fed an HFD. Greater fat storage resulted in significantly enlarged adipocytes and was associated with increased postprandial lipoprotein lipase activity in adipose tissue. Parallel to this was a striking reduction in liver steatosis due to significantly reduced TG accumulation. Liver metabolite profiling revealed additional significant changes in bile acids and 1-carbon metabolism pathways. Combined, our data affirm the physiologic importance of myonectin in regulating local and systemic lipid metabolism.-Little, H. C., Rodriguez, S., Lei, X., Tan, S. Y., Stewart, A. N., Sahagun, A., Sarver, D. C., Wong, G. W. Myonectin deletion promotes adipose fat storage and reduces liver steatosis.


Asunto(s)
Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Citocinas/genética , Hígado Graso/genética , Hígado Graso/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Proteínas Musculares/genética , Adipocitos/metabolismo , Adipocitos/patología , Adiposidad/genética , Animales , Citocinas/metabolismo , Dieta Alta en Grasa , Hígado Graso/patología , Femenino , Homeostasis/genética , Insulina/genética , Insulina/metabolismo , Resistencia a la Insulina/genética , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Triglicéridos/genética , Triglicéridos/metabolismo
7.
EMBO J ; 34(3): 344-60, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25510864

RESUMEN

In mammals, glucocorticoids (GCs) and their intracellular receptor, the glucocorticoid receptor (GR), represent critical checkpoints in the endocrine control of energy homeostasis. Indeed, aberrant GC action is linked to severe metabolic stress conditions as seen in Cushing's syndrome, GC therapy and certain components of the Metabolic Syndrome, including obesity and insulin resistance. Here, we identify the hepatic induction of the mammalian conserved microRNA (miR)-379/410 genomic cluster as a key component of GC/GR-driven metabolic dysfunction. Particularly, miR-379 was up-regulated in mouse models of hyperglucocorticoidemia and obesity as well as human liver in a GC/GR-dependent manner. Hepatocyte-specific silencing of miR-379 substantially reduced circulating very-low-density lipoprotein (VLDL)-associated triglyceride (TG) levels in healthy mice and normalized aberrant lipid profiles in metabolically challenged animals, mediated through miR-379 effects on key receptors in hepatic TG re-uptake. As hepatic miR-379 levels were also correlated with GC and TG levels in human obese patients, the identification of a GC/GR-controlled miRNA cluster not only defines a novel layer of hormone-dependent metabolic control but also paves the way to alternative miRNA-based therapeutic approaches in metabolic dysfunction.


Asunto(s)
Glucocorticoides/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , MicroARNs/metabolismo , Obesidad/metabolismo , Animales , Línea Celular , Femenino , Silenciador del Gen , Glucocorticoides/genética , Humanos , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Obesos , MicroARNs/genética , Obesidad/genética , Triglicéridos/genética , Triglicéridos/metabolismo
8.
Lipids Health Dis ; 18(1): 224, 2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31856839

RESUMEN

BACKGROUND: APOC3 is important in lipid transport and metabolism with limited studies reporting genetic sequence variations in specific ethnic groups. The present study aimed to analyze the full APOC3 sequence among Kuwaiti Arabs and test the association of selected variants with lipid levels and BMI. METHODS: Variants were identified by Sanger sequencing the entire APOC3 gene in 100 Kuwaiti Arabs. Variants and their genotypes were fully characterized and used to construct haplotype blocks. Four variants (rs5128, rs2854117, rs2070668, KUAPOC3N3 g.5196 A > G) were selected for testing association with serum lipid levels and BMI in a cohort (n = 733). RESULTS: APOC3 sequence (4.3 kb) of a Kuwaiti Arab was deposited in Genbank (accession number KJ437193). Forty-two variants including 3 novels were identified including an "A" insertion at genomic positions 116,700,599-116,700,600 (promoter region) and two substitutions in intron 1 at genomic positions 116,700,819 and 116,701,159. Only three variants, (rs5128, rs2854117, and rs2070668) were analyzed for association of which rs5128 showed a trend for association with increased BMI, TG and VLDL levels that was further investigated using multivariate analysis. A significant association of rs5128 with BMI (p <  0.05) was observed following a dominant genetic model with increased risk by an OR of 4.022 (CI: 1.13-14.30). CONCLUSION: The present study is the first to report sequence analysis of APOC3 in an Arab ethnic group. This study supports the inclusion of rs5128 as a marker for assessing genetic risk to dyslipidemia and obesity and the inclusion of the novel variant g.5196 A > G for population stratification of Arabs.


Asunto(s)
Apolipoproteína C-III/genética , Índice de Masa Corporal , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Adolescente , Adulto , Anciano , Árabes/genética , Femenino , Genotipo , Humanos , Metabolismo de los Lípidos/genética , Lipoproteínas VLDL/sangre , Lipoproteínas VLDL/genética , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Triglicéridos/sangre , Triglicéridos/genética , Adulto Joven
9.
Curr Opin Lipidol ; 29(3): 218-223, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29547398

RESUMEN

PURPOSE OF REVIEW: Lysosomal acid lipase (LAL), encoded by the LIPA gene, is an essential lysosomal enzyme that hydrolyzes cholesteryl ester and triglyceride delivered to the lysosome. This review highlights the novel pathophysiological role of LAL, the functional genomic discoveries of LIPA as a risk locus for coronary heart diseases (CHD), and the clinical advance in therapies for LAL deficiency. RECENT FINDINGS: The essential role of LAL in lipid metabolism has been confirmed in human and mice with LAL deficiency. In humans, loss-of-function mutations of LIPA cause rare lysosomal disorders, Wolman disease, and cholesteryl ester storage disease, in which LAL enzyme replacement therapy has shown significant benefits in a phase 3 clinical trial. Recent studies have revealed the role of LAL-mediated lysosomal lipolysis in regulating macrophage M2 polarization, lipid mediator production, VLDL secretion, lysosomal function and autophagy, extracellular degradation of aggregated-LDL, and adipose tissue lipolysis. Genome-wide association studies and functional genomic studies have identified LIPA as a risk locus for CHD, but the causal variants and mechanisms remain to be determined. SUMMARY: Despite years of research, our understanding of LAL is incomplete. Future studies will continue to focus on the key pathophysiological functions of LAL in health and diseases including CHD.


Asunto(s)
Tejido Adiposo/metabolismo , Metabolismo de los Lípidos , Lipoproteínas VLDL/metabolismo , Lisosomas/metabolismo , Esterol Esterasa/metabolismo , Enfermedad de Wolman/metabolismo , Tejido Adiposo/patología , Animales , Humanos , Lipoproteínas VLDL/genética , Lisosomas/genética , Lisosomas/patología , Ratones , Esterol Esterasa/genética , Enfermedad de Wolman/genética , Enfermedad de Wolman/patología , Enfermedad de Wolman
10.
Curr Opin Lipidol ; 29(3): 186-193, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29708925

RESUMEN

PURPOSE OF REVIEW: Type 2 diabetes is associated with a characteristic dyslipidemia that may exacerbate cardiovascular risk. The causes of, and the effects of new antihyperglycemia medications on, this dyslipidemia, are under investigation. In an unexpected reciprocal manner, lowering LDL-cholesterol with statins slightly increases the risk of diabetes. Here we review the latest findings. RECENT FINDINGS: The inverse relationship between LDL-cholesterol and diabetes has now been confirmed by multiple lines of evidence. This includes clinical trials, genetic instruments using aggregate single nucleotide polymorphisms, as well as at least eight individual genes - HMGCR, NPC1L1, HNF4A, GCKR, APOE, PCKS9, TM6SF2, and PNPLA3 - support this inverse association. Genetic and pharmacologic evidence suggest that HDL-cholesterol may also be inversely associated with diabetes risk. Regarding the effects of diabetes on lipoproteins, new evidence suggests that insulin resistance but not diabetes per se may explain impaired secretion and clearance of VLDL-triglycerides. Weight loss, bariatric surgery, and incretin-based therapies all lower triglycerides, whereas SGLT2 inhibitors may slightly increase HDL-cholesterol and LDL-cholesterol. SUMMARY: Diabetes and lipoproteins are highly interregulated. Further research is expected to uncover new mechanisms governing the metabolism of glucose, fat, and cholesterol. This topic has important implications for treating type 2 diabetes and cardiovascular disease.


Asunto(s)
HDL-Colesterol , LDL-Colesterol , Diabetes Mellitus Tipo 2 , Dislipidemias , Lipoproteínas VLDL , Polimorfismo de Nucleótido Simple , Triglicéridos , Animales , HDL-Colesterol/genética , HDL-Colesterol/metabolismo , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Dislipidemias/genética , Dislipidemias/metabolismo , Dislipidemias/terapia , Humanos , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Triglicéridos/genética , Triglicéridos/metabolismo
11.
IUBMB Life ; 70(1): 41-49, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29281185

RESUMEN

Hepatitis C virus (HCV) has infected over 170 million people world-wide. This infection causes severe liver damage that can progress to hepatocellular carcinoma leading to death of the infected patients. Development of a cell culture model system for the study of HCV infection in the recent past has helped the researchers world-wide to understand the biology of this virus. Studies over the past decade have revealed the tricks played by the virus to sustain itself, for as long as 40 years, in the host setup without being eliminated by the immune system. Today we understand that the host organelles and different cellular proteins are affected during HCV infection. This cytoplasmic virus has all the cellular organelles at its disposal to successfully replicate, from ribosomes and intracellular membranous structures to the nucleus. It modulates these organelles at both the structural and the functional levels. The vast knowledge about the viral genome and viral proteins has also helped in the development of drugs against the virus. Despite the achieved success rate to cure the infected patients, we struggle to eliminate the cases of recurrence and the non-responders. Such cases might emerge owing to the property of the viral genome to accumulate mutations during its succeeding replication cycles which favours its survival. The current situation calls an urgent need for alternate therapeutic strategies to counter this major problem of human health. © 2017 IUBMB Life, 70(1):41-49, 2018.


Asunto(s)
Carcinoma Hepatocelular/virología , Hepacivirus/patogenicidad , Hepatitis C Crónica/virología , Hepatocitos/virología , Evasión Inmune , Neoplasias Hepáticas/virología , Antivirales/uso terapéutico , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/prevención & control , Núcleo Celular/inmunología , Núcleo Celular/virología , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/virología , Regulación de la Expresión Génica , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Hepatitis C Crónica/complicaciones , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/inmunología , Hepatocitos/inmunología , Humanos , Gotas Lipídicas/inmunología , Gotas Lipídicas/virología , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/inmunología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/prevención & control , ARN Viral/biosíntesis , ARN Viral/genética , Ribosomas/inmunología , Ribosomas/virología , Transducción de Señal , Proteínas Virales/genética , Proteínas Virales/inmunología , Replicación Viral/efectos de los fármacos
12.
J Clin Gastroenterol ; 52(5): 444-451, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28362682

RESUMEN

BACKGROUND: Recent genome-wide association studies have identified 2 genetic polymorphisms in association with nonalcoholic fatty liver disease (NAFLD): patatin-like phospholipase domain containing 3 (PNPLA3) and transmembrane 6 superfamily member 2 (TM6SF2), both of which appear to influence the production of very low density lipoprotein (VLDL). The impact of these genetic variations on lipoprotein metabolism in the setting of nonalcoholic steatohepatitis and liver fibrosis are not fully characterized. MATERIALS AND METHODS: We measured comprehensive lipoprotein profiles by nuclear magnetic resonance among 170 serially recruited patients in an NAFLD registry, and determined their relationships with PNPLA3 and TM6SF2 genotypes. RESULTS: In this cohort, 72% patients had at least 1 allele of either PNPLA3 I148M or TM6SF2 E167K, and 30% carried 2 alleles. In multivariate models adjusting for histologic features of nonalcoholic steatohepatitis and liver fibrosis, PNPLA3 I148M is associated with a decrease in VLDL particle size. Both PNPLA3 I148M and TM6SF2 E167K genotypes were associated with increases in the size of low density lipoprotein (LDL) and high density lipoprotein particles, phenotypes considered atheroprotective. When adjusted for both genotypes, NAFLD activity score, in particular the degree of hepatic steatosis was strongly associated with increases in the size of VLDL particles, the concentration of LDL, especially small LDL particles, and a decrease in the size of high density lipoprotein particles, all of which are linked with a proatherogenic phenotype. CONCLUSIONS: PNPLA3 and TM6SF2 are common genetic variants among NAFLD patients and impact lipoprotein profiles in slightly different ways. The interactions between genotypes, hepatic steatosis, and lipoprotein metabolism shed lights on the pathophysiology of NAFLD, and provide opportunities for personalized treatment in the era of emerging NAFLD therapeutics.


Asunto(s)
Lipasa/genética , Cirrosis Hepática/genética , Proteínas de la Membrana/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Adulto , Anciano , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Cirrosis Hepática/patología , Espectroscopía de Resonancia Magnética , Masculino , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Polimorfismo de Nucleótido Simple , Estudios Prospectivos
13.
Int J Mol Sci ; 19(11)2018 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-30413026

RESUMEN

Rabbits (Oryctolagus cuniculus) are one of the most widely used animal models for the study of human lipid metabolism and atherosclerosis because they are more sensitive to a cholesterol diet than other experimental animals such as rodents. Currently, two hypercholesterolemic rabbit models are frequently used for atherosclerosis studies. One is a cholesterol-fed wild-type rabbit and the other is the Watanabe heritable hyperlipidemic (WHHL) rabbit, which is genetically deficient in low density lipoprotein (LDL) receptor function. Wild-type rabbits can be easily induced to develop severe hypercholesterolemia with a cholesterol-rich diet due to the marked increase in hepatically and intestinally derived remnant lipoproteins, called ß-very low density lipoproteins (VLDL), which are rich in cholesteryl esters. WHHL rabbits are characterized by elevated plasma LDL levels on a standard chow diet, which resembles human familial hypercholesterolemia. Therefore, both rabbit models develop aortic and coronary atherosclerosis, but the elevated plasma cholesterol levels are caused by completely different mechanisms. In addition, cholesterol-fed rabbits but not WHHL rabbits exhibit different degrees of hepatosteatosis. Recently, we along with others have shown that there are many differentially expressed genes in the atherosclerotic lesions and livers of cholesterol-fed rabbits that are either significantly up- or down-regulated, compared with those in normal rabbits, including genes involved in the regulation of inflammation and lipid metabolism. Therefore, dietary cholesterol plays an important role not only in hypercholesterolemia and atherosclerosis but also in hepatosteatosis. In this review, we make an overview of the recent progress in genomic and transcriptomic analyses of hypercholesterolemic rabbits. These transcriptomic profiling data should provide novel insight into the relationship between hypercholesterolemia and atherosclerosis or hepatic dysfunction caused by dietary cholesterol.


Asunto(s)
Colesterol/genética , Genoma/genética , Hipercolesterolemia/genética , Transcriptoma/genética , Animales , Colesterol/metabolismo , Modelos Animales de Enfermedad , Genómica , Humanos , Hipercolesterolemia/metabolismo , Hipercolesterolemia/patología , Lipoproteínas VLDL/genética , Conejos , Receptores de LDL/genética
14.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(10 Pt A): 1035-1043, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28694219

RESUMEN

ATP binding cassette transporter A1 (ABCA1) is a membrane transporter that facilitates nascent HDL formation. Tangier disease subjects with complete ABCA1 deficiency have <5% of normal levels of plasma HDL, elevated triglycerides (TGs), and defective vesicular trafficking in fibroblasts and macrophages. Hepatocyte-specific ABCA1 knockout mice (HSKO) have a similar lipid phenotype with 20% of normal plasma HDL levels and a two-fold elevation of plasma TGs due to hepatic overproduction of large, triglyceride-enriched VLDL. We hypothesized that enhanced VLDL TG secretion in the absence of hepatocyte ABCA1 is due to altered intracellular trafficking of apolipoprotein B (apoB), resulting in augmented TG addition to nascent VLDL. We found that trafficking of newly synthesized apoB through the secretory pathway was delayed in ABCA1-silenced rat hepatoma cells and HSKO primary hepatocytes, relative to controls. Endoglycosidase H treatment of cellular apoB revealed a likely delay in apoB trafficking in post-ER compartments. The reduced rate of protein trafficking was also observed for an adenoviral-expressed GPI-linked fluorescent fusion protein, but not albumin, suggesting a selective delay of secretory cargoes in the absence of hepatocyte ABCA1. Our results suggest an important role for hepatic ABCA1 in regulating secretory trafficking and modulating VLDL expansion during the TG accretion phase of hepatic lipoprotein particle assembly.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Apolipoproteínas B/metabolismo , Hepatocitos/metabolismo , Lipoproteínas VLDL/metabolismo , Hígado/metabolismo , Triglicéridos/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Animales , Apolipoproteínas B/genética , Línea Celular Tumoral , Silenciador del Gen , Lipoproteínas VLDL/genética , Hígado/patología , Ratones , Ratones Noqueados , Transporte de Proteínas/fisiología , Ratas , Triglicéridos/genética
15.
J Biol Chem ; 290(52): 31003-12, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26547624

RESUMEN

Disruption of the body clock has been recognized as a risk factor for cardiovascular disease. How the circadian pacemaker interacts with the genetic factors associated with plasma lipid traits remains poorly understood. Recent genome-wide association studies have identified an expanding list of genetic variants that influence plasma cholesterol and triglyceride levels. Here we analyzed circadian regulation of lipid-associated candidate genes in the liver and identified two distinct groups exhibiting rhythmic and non-rhythmic patterns of expression during light-dark cycles. Liver-specific inactivation of Bmal1 led to elevated plasma LDL/VLDL cholesterol levels as a consequence of the disruption of the PCSK9/LDL receptor regulatory axis. Ablation of the liver clock perturbed diurnal regulation of lipid-associated genes in the liver and markedly reduced the expression of the non-rhythmically expressed gene Trib1. Adenovirus-mediated rescue of Trib1 expression lowered plasma PCSK9 levels, increased LDL receptor protein expression, and restored plasma cholesterol homeostasis in mice lacking a functional liver clock. These results illustrate an unexpected mechanism through which the biological clock regulates cholesterol homeostasis through its regulation of non-rhythmic genes in the liver.


Asunto(s)
Colesterol/metabolismo , Relojes Circadianos/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Proproteína Convertasas/biosíntesis , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de LDL/biosíntesis , Serina Endopeptidasas/biosíntesis , Animales , Colesterol/genética , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Ratones , Ratones Transgénicos , Proproteína Convertasa 9 , Proproteína Convertasas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de LDL/genética , Serina Endopeptidasas/genética
16.
Avian Pathol ; 45(4): 450-7, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26957042

RESUMEN

Salmonella enterica serovar Enteritidis (SE) is a public health concern and infected chickens serve as a reservoir that potentially transmits to humans through food. Although SE seldom causes systemic disease in chickens, virulent SE strains can colonize in intestines and lead a persistent infection of the liver. The liver is the primary organ for lipid metabolism in chickens and the site for production and assembly of main components in yolk. We performed a time-course experiment using LMH-2A cells that were infected with SE and co-incubated with ß-oestradiol to evaluate if SE infection affected lipid metabolism and subsequently changed lipoprotein formation for egg yolk. The results indicated that lipid accumulation significantly increased in infected LMH-2A cells while the viability of these cells was only slightly decreased. The mRNA expressions of lipid transportation and most lipogenetic genes including sterol regulatory element binding protein 1, acetyl-CoA carboxylase, fatty-acid synthase, long-chain-fatty-acid-CoA ligase 1, peroxisome proliferator-activated receptor-γ, and very-low-density lipoproteins (VLDLs) II were significantly up-regulated while the expression of lipogenetic-related stearoyl-CoA denaturase 1 was down-regulated. Moreover, decline in lipid transportation of hepatocytes was evidenced by the down-regulation of oestrogen receptor α which promotes VLDLy formation, an increase of intra-cellular accumulation of Apoprotein B (ApoB) protein, and a decrease of cellular excretion of VLDL protein. Conclusively, SE infection could elevate lipid synthesis and reduce lipid transportation in the chicken hepatocytes. These changes may lead excessive lipid accumulation in liver and slower lipoprotein deposition in yolk.


Asunto(s)
Pollos/microbiología , Regulación Bacteriana de la Expresión Génica , Metabolismo de los Lípidos , Salmonelosis Animal/microbiología , Salmonella enteritidis/fisiología , Animales , Transporte Biológico , Células Cultivadas , Pollos/metabolismo , Coenzima A Ligasas/genética , Reservorios de Enfermedades , Regulación hacia Abajo , Hepatocitos/metabolismo , Hepatocitos/microbiología , Lipoproteínas VLDL/genética , Hígado/metabolismo , Hígado/microbiología , Óvulo/metabolismo , Óvulo/microbiología , Receptores Activados del Proliferador del Peroxisoma/genética , Estearoil-CoA Desaturasa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Regulación hacia Arriba
17.
Genet Mol Res ; 15(2)2016 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-27323131

RESUMEN

Previous studies have shown that cytokines can affect serum lipoprotein concentrations. The aim of this study was to examine the association between IL-10 gene polymorphisms and serum lipoprotein levels of Han Chinese individuals. A total of 359 Han Chinese people were enrolled in this investigation. IL-10 -592, -819, and -1082 genotypes were established using polymerase chain reaction-restriction fragment length polymorphism analysis. An automatic biochemistry analyzer was used to determine serum concentrations of total cholesterol (TC), triglycerides (TG), low-density lipoprotein (LDL), high-density lipoprotein (HDL), and very low-density lipoprotein (VLDL) in each individual. We observed that the three IL-10 polymorphisms did not significantly differ in terms of age or age of carrier (P > 0.05), and the -592 and -819 variants did not significantly affect serum lipoprotein levels (P > 0.05). HDL concentrations were higher and TG levels were lower in carriers of the -1082 GA genotype compared to those with the AA genotype, and these differences were statistically significant (P < 0.05). However, TC, VLDL, and LDL levels were unaffected by this sequence variation (P > 0.05). Our results suggest that the polymorphism at position -1082 in the promoter region of IL-10 may affect serum HDL and TG concentrations, while other variants of this gene appear to have no relationship with serum lipoprotein levels.


Asunto(s)
Interleucina-10/genética , Lipoproteínas HDL/sangre , Lipoproteínas LDL/sangre , Lipoproteínas VLDL/sangre , Adulto , Anciano , Colesterol/sangre , Colesterol/genética , Femenino , Genotipo , Humanos , Interleucina-10/sangre , Lipoproteínas HDL/genética , Lipoproteínas LDL/genética , Lipoproteínas VLDL/genética , Masculino , Persona de Mediana Edad , Polimorfismo Genético , Triglicéridos/sangre , Triglicéridos/genética
18.
J Lipid Res ; 56(3): 612-619, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25616437

RESUMEN

We previously determined that hamster cholesteryl ester transfer protein (CETP), unlike human CETP, promotes a novel one-way transfer of TG from VLDL to HDL, causing HDL to gain lipid. We hypothesize that this nonreciprocal lipid transfer activity arises from the usually high TG/cholesteryl ester (CE) substrate preference of hamster CETP. Consistent with this, we report here that ∼25% of the total lipid transfer promoted by the human Q199A CETP mutant, which prefers TG as substrate, is nonreciprocal transfer. Other human CETP mutants with TG/CE substrate preferences higher or lower than wild-type also possess nonreciprocal lipid transfer activity. Mutants with high TG/CE substrate preference promote the nonreciprocal lipid transfer of TG from VLDL to HDL, but mutants with low TG/CE substrate preference promote the nonreciprocal lipid transfer of CE, not TG, and this lipid flow is in the reverse direction (from HDL to VLDL). Anti-CETP TP2 antibody alters the TG/CE substrate preference of CETP and also changes the extent of nonreciprocal lipid transfer, showing the potential for externally acting agents to modify the transfer properties of CETP. Overall, these data show that the lipid transfer properties of CETP can be manipulated. Function-altering pharmaceuticals may offer a novel approach to modify CETP activity and achieve specific modifications in lipoprotein metabolism.


Asunto(s)
Proteínas de Transferencia de Ésteres de Colesterol/química , Ésteres del Colesterol/química , Diseño de Fármacos , Triglicéridos/química , Sustitución de Aminoácidos , Animales , Proteínas de Transferencia de Ésteres de Colesterol/antagonistas & inhibidores , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Ésteres del Colesterol/genética , Ésteres del Colesterol/metabolismo , Cricetinae , Humanos , Lipoproteínas HDL/química , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Lipoproteínas VLDL/química , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Mutación Missense , Relación Estructura-Actividad , Especificidad por Sustrato , Triglicéridos/genética , Triglicéridos/metabolismo
19.
J Biol Chem ; 289(33): 22672-22691, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24928512

RESUMEN

Growing clinical and experimental evidence suggests that sterile inflammation contributes to alcoholic liver disease (ALD). High mobility group box-1 (HMGB1) is highly induced during liver injury; however, a link between this alarmin and ALD has not been established. Thus, the aim of this work was to determine whether HMGB1 contributes to the pathogenesis of ALD. Liver biopsies from patients with ALD showed a robust increase in HMGB1 expression and translocation, which correlated with disease stage, compared with healthy explants. Similar findings were observed in chronic ethanol-fed wild-type (WT) mice. Using primary cell culture, we validated the ability of hepatocytes from ethanol-fed mice to secrete a large amount of HMGB1. Secretion was time- and dose-dependent and responsive to prooxidants and antioxidants. Selective ablation of Hmgb1 in hepatocytes protected mice from alcohol-induced liver injury due to increased carnitine palmitoyltransferase-1, phosphorylated 5'AMP-activated protein kinase-α, and phosphorylated peroxisome proliferator-activated receptor-α expression along with elevated LDL plus VLDL export. Native and post-translationally modified HMGB1 were detected in humans and mice with ALD. In liver and serum from control mice and in serum from healthy volunteers, the lysine residues within the peptides containing nuclear localization signals (NLSs) 1 and 2 were non-acetylated, and all cysteine residues were reduced. However, in livers from ethanol-fed mice, in addition to all thiol/non-acetylated isoforms of HMGB1, we observed acetylated NLS1 and NLS2, a unique phosphorylation site in serine 35, and an increase in oxidation of HMGB1 to the disulfide isoform. In serum from ethanol-fed mice and from patients with ALD, there was disulfide-bonded hyperacetylated HMGB1, disulfide-bonded non-acetylated HMGB1, and HMGB1 phosphorylated in serine 35. Hepatocytes appeared to be a major source of these HMGB1 isoforms. Thus, hepatocyte HMGB1 participates in the pathogenesis of ALD and undergoes post-translational modifications (PTMs) that could condition its toxic effects.


Asunto(s)
Proteína HMGB1/metabolismo , Hepatocitos/metabolismo , Hepatopatías Alcohólicas/mortalidad , Hígado/metabolismo , Procesamiento Proteico-Postraduccional , Acetilación , Animales , Antioxidantes/farmacología , Células Cultivadas , Femenino , Proteína HMGB1/genética , Hepatocitos/patología , Humanos , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Hígado/patología , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/patología , Masculino , Ratones , Ratones Noqueados , Oxidantes/farmacología , Fosforilación/genética , Cultivo Primario de Células
20.
J Biol Chem ; 289(3): 1688-703, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24293365

RESUMEN

Obesity is associated with adipose tissue remodeling, characterized by adipocyte hypertrophy and macrophage infiltration. Previously, we have shown that very low density lipoprotein receptor (VLDLR) is virtually absent in preadipocytes but is strongly induced during adipogenesis and actively participates in adipocyte hypertrophy. In this study, we investigated the role of VLDLR in adipose tissue inflammation and adipocyte-macrophage interactions in wild type and VLDLR-deficient mice fed a high fat diet. The results show that VLDLR deficiency reduced high fat diet-induced inflammation and endoplasmic reticulum (ER) stress in adipose tissue in conjunction with reduced macrophage infiltration, especially those expressing pro-inflammatory markers. In adipocyte culture, VLDLR deficiency prevented adipocyte hypertrophy and strongly reduced VLDL-induced ER stress and inflammation. Likewise, cultures of primary peritoneal macrophages show that VLDLR deficiency reduced lipid accumulation and inflammation but did not alter chemotactic response of macrophages to adipocyte signals. Moreover, VLDLR deficiency tempered the synergistic inflammatory interactions between adipocytes and macrophages in a co-culture system. Collectively, these results show that VLDLR contributes to adipose tissue inflammation and mediates VLDL-induced lipid accumulation and induction of inflammation and ER stress in adipocytes and macrophages.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Regulación de la Expresión Génica , Macrófagos Peritoneales/metabolismo , Paniculitis/metabolismo , Receptores de LDL/biosíntesis , Adipocitos/patología , Tejido Adiposo/patología , Animales , Comunicación Celular/efectos de los fármacos , Comunicación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Noqueados , Paniculitis/inducido químicamente , Paniculitis/genética , Paniculitis/patología , Receptores de LDL/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda