Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
1.
Int Immunopharmacol ; 128: 111537, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38232538

RESUMO

Respiratory failure caused by severe acute lung injury (ALI) is the main cause of mortality in patients with COVID-19.This study aimed to investigate the effects and underlying biological mechanism of Apolipoprotein C3 (ApoC3) in ALI. To establish an in vivo model, C57BL/6 mice were exposed by lipopolysaccharide (LPS). For the in vitro model, murine bone marrow-derived macrophages (BMDMs) or RAW264.7 cells were stimulated with LPS + adenosine triphosphate (ATP). Serum levels of ApoC3 were found to be upregulated in patients with COVID-19 or pneumonia-induced ALI. Inhibition of ApoC3 reduced lung injury in an ALI model, while overexpression of ApoC3 promoted lung injury. ApoC3 induced mitochondrial damage-mediated pyroptosis in ALI through the activation of the NOD-like receptorprotein 3 (NLRP3) inflammasome. ApoC3 recombinant protein significantly increased SCIMP expression in the lung tissue of mice models with ALI. ApoC3 also facilitated the interaction between the SLP adapter and CSK-interacting membrane protein (SCIMP) protein and Spleen tyrosine kinase (SYK) protein in the ALI model. Moreover, ApoC3 accelerated calcium-dependent reactive oxygen species (ROS) production in the ALI model. The effects of ApoC3 on pyroptosis were mitigated by the use of a pyroptosis inhibitor or an ROS inhibitor in the ALI model. Furthermore, ApoC3 activated the expression of SYK, which in turn induced NLRP3 inflammasome-regulated pyroptosis in the ALI model. METTL3 was found to mediate the m6A mRNA expression of ApoC3. Overall, our study highlights the crucial role of ApoC3 in promoting macrophage pyroptosis in ALI through calcium-dependent ROS production and NLRP3 inflammasome activation via the SCIMP-SYK pathway, providing a potential therapeutic strategy for ALI and other inflammatory diseases.


Assuntos
Lesão Pulmonar Aguda , COVID-19 , Metiltransferases , Animais , Humanos , Camundongos , Lesão Pulmonar Aguda/tratamento farmacológico , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cálcio/metabolismo , Inflamassomos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose , Espécies Reativas de Oxigênio/metabolismo , Quinase Syk/metabolismo , Apolipoproteínas C/metabolismo
2.
Atherosclerosis ; 380: 117214, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37573768

RESUMO

BACKGROUND AND AIMS: Higher truncated-to-native proteoform ratios of apolipoproteins (apo) C-I (C-I'/C-I) and C-II (C-II'/C-II) are associated with less atherogenic lipid profiles. We examined prospective relationships of C-I'/C-II and C-II'/C-II with coronary heart disease (CHD) and coronary artery calcium (CAC). METHODS: ApoC-I and apoC-II proteoforms were measured by mass spectrometry immunoassay in 5790 MESA baseline plasma samples. CHD events (myocardial infarction, resuscitated cardiac arrest, fatal CHD, n = 434) were evaluated for up to 17 years. CAC was measured 1-4 times over 10 years for incident CAC (if baseline CAC = 0), and changes (follow-up adjusted for baseline) in CAC score and density (if baseline CAC>0). RESULTS: C-II'/C-II was inversely associated with CHD (n = 434 events) after adjusting for non-lipid cardiovascular risk factors (Hazard ratio: 0.89 [95% CI: 0.81-0.98] per SD), however, the association was attenuated after further adjustment for HDL levels (0.93 [0.83-1.03]). There was no association between C-I'/C-I and CHD (0.98 [0.88-1.08]). C-II'/C-II was positively associated with changes in CAC score (3.4% [95%CI: 0.6, 6.3]) and density (6.3% [0.3, 4.2]), while C-I'/C-I was inversely associated with incident CAC (Risk ratio: 0.89 [95% CI: 0.81, 0.98]) in fully adjusted models that included plasma lipids. Total apoC-I and apoC-II concentrations were not associated with CHD, incident CAC or change in CAC score. CONCLUSIONS: Increased apoC-II truncation was associated with reduced CHD, possibly explained by differences in lipid metabolism. Increased apoC-I and apoC-II truncations were also associated with less CAC progression and/or development of denser coronary plaques.


Assuntos
Aterosclerose , Doença da Artéria Coronariana , Humanos , Doença da Artéria Coronariana/complicações , Cálcio/metabolismo , Estudos Prospectivos , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/metabolismo , Aterosclerose/metabolismo , Cálcio da Dieta , Apolipoproteínas C/metabolismo , Fatores de Risco , Medição de Risco
3.
Nutr Metab Cardiovasc Dis ; 32(2): 295-308, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34895805

RESUMO

AIMS: Recently, Apolipoprotein CIII (Apo-CIII) has gained remarkable attention since its overexpression has been strongly correlated to cardiovascular disease (CVD) occurrence. The aim of this review was to summarize the latest findings of Apo-CIII as a CVDs and diabetes risk factor, as well as the plausible mechanisms involved in the development of these pathologies, with particular emphasis on current clinical and dietetic therapies. DATA SYNTHESIS: Apo-CIII is a small protein (∼8.8 kDa) that, among other functions, inhibits lipoprotein lipase, a key enzyme in lipid metabolism. Apo-CIII plays a fundamental role in the physiopathology of atherosclerosis, type-1, and type-2 diabetes. Apo-CIII has become a potential clinical target to tackle these multifactorial diseases. Dietetic (omega-3 fatty acids, stanols, polyphenols, lycopene) and non-dietetic (fibrates, statins, and antisense oligonucleotides) therapies have shown promising results to regulate Apo-CIII and triglyceride levels. However, more information from clinical trials is required to validate it as a new target for atherosclerosis and diabetes types 1 and 2. CONCLUSIONS: There are still several pathways involving Apo-CIII regulation that might be affected by bioactive compounds that need further research. The mechanisms that trigger metabolic responses following bioactive compounds consumption are mainly related to higher LPL expression and PPARα activation, although the complete pathways are yet to be elucidated.


Assuntos
Dietética , Inibidores de Hidroximetilglutaril-CoA Redutases , Apolipoproteína C-III/genética , Apolipoproteínas C/metabolismo , Humanos , Triglicerídeos
4.
Nat Commun ; 10(1): 3310, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31346172

RESUMO

Alzheimer's disease (AD) is a leading cause of mortality in the elderly. While the coding change of APOE-ε4 is a key risk factor for late-onset AD and has been believed to be the only risk factor in the APOE locus, it does not fully explain the risk effect conferred by the locus. Here, we report the identification of AD causal variants in PVRL2 and APOC1 regions in proximity to APOE and define common risk haplotypes independent of APOE-ε4 coding change. These risk haplotypes are associated with changes of AD-related endophenotypes including cognitive performance, and altered expression of APOE and its nearby genes in the human brain and blood. High-throughput genome-wide chromosome conformation capture analysis further supports the roles of these risk haplotypes in modulating chromatin states and gene expression in the brain. Our findings provide compelling evidence for additional risk factors in the APOE locus that contribute to AD pathogenesis.


Assuntos
Doença de Alzheimer/genética , Apolipoproteínas E/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/psicologia , Apolipoproteínas C/genética , Apolipoproteínas C/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Estudos de Casos e Controles , Cognição , Feminino , Expressão Gênica , Predisposição Genética para Doença , Variação Genética , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Nectinas/genética , Nectinas/metabolismo , Polimorfismo de Nucleotídeo Único
5.
J Cell Biochem ; 120(10): 18246-18265, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31211449

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is among the most common and lethal malignancies worldwide. Apolipoproteins (APOs) have been reported increasingly for their relationships with tumors. We aim at exploring the potential relationships of apolipoprotein A (APOA) and apolipoprotein C (APOC) family members with HCC. METHODS: A data set, containing 212 hepatitis B virus-related HCC patients, was used for analysis. The diagnostic and prognostic ability of APOA and APOC family genes was figured out. Risk score models and nomograms were developed for the HCC prognosis prediction. Moreover, molecular mechanism exploration were identified biological processes and metabolic pathways of these genes involved in. Validation analysis was carried out using online website. RESULTS: APOA1, APOC1, APOC3, and APOC4 showed robust diagnosis significance (all P < 0.05). APOA4, APOC3, and APOC4 were associated with the overall survival (OS) while APOA4 and APOC4 were linked to recurrence-free survival (RFS, all P ≤ 0.05). Risk score models and nomograms had the advantage of predicting OS and RFS for HCC. Molecular mechanism exploration indicated that these genes were involved in the steroid metabolic process, the PPAR signaling pathway, and fatty acid metabolism. Besides that, validation analysis revealed that APOC1 and APOC4 had an association with OS; and APOC3 was associated with OS and RFS (all P ≤ 0.05). CONCLUSIONS: APOA1, APOC1, APOC3, and APOC4 are likely to be potential diagnostic biomarkers and APOC3 and APOC4 are likely to be potential prognostic biomarkers for hepatitis B virus-related HCC. They may be involved in the steroid metabolic process, PPAR signaling pathway, and fatty acid metabolism.


Assuntos
Apolipoproteínas A/genética , Apolipoproteínas C/genética , Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Hepatite/complicações , Neoplasias Hepáticas/genética , RNA Mensageiro/genética , Apolipoproteínas A/metabolismo , Apolipoproteínas C/metabolismo , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/metabolismo , Feminino , Perfilação da Expressão Gênica/métodos , Ontologia Genética , Redes Reguladoras de Genes , Hepatite/virologia , Vírus da Hepatite B/fisiologia , Humanos , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/metabolismo , Masculino , Pessoa de Meia-Idade , Nomogramas , Prognóstico , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo
6.
Trends Pharmacol Sci ; 39(8): 733-747, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29970260

RESUMO

Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of morbidity and mortality in developed nations. Therapeutic modulation of dyslipidemia by inhibiting 3'-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase is standard practice throughout the world. However, based on findings from Mendelian studies and genetic sequencing in prospective longitudinal cohorts from around the world, novel therapeutic targets regulating lipid and lipoprotein metabolism, such as apoprotein C3, angiopoietin-like proteins 3 and 4, and lipoprotein(a), have been identified. These targets may provide additional avenues to prevent and treat atherosclerotic disease. We therefore review these novel molecular targets by addressing available Mendelian and observational data, therapeutic agents in development, and early outcomes results.


Assuntos
Aterosclerose/tratamento farmacológico , Dislipidemias/tratamento farmacológico , Acil Coenzima A/antagonistas & inibidores , Acil Coenzima A/genética , Proteínas Semelhantes a Angiopoietina/genética , Proteínas Semelhantes a Angiopoietina/metabolismo , Animais , Apolipoproteínas C/genética , Apolipoproteínas C/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Dislipidemias/genética , Dislipidemias/metabolismo , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Terapia de Alvo Molecular , Estudos Prospectivos
7.
J Pediatr ; 183: 170-177.e1, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28139241

RESUMO

OBJECTIVE: To describe the clinical, biochemical, and genetic features of patients with congenital disorders of glycosylation (CDG) identified in Spain during the last 20 years. STUDY DESIGN: Patients were selected among those presenting with multisystem disease of unknown etiology. The isoforms of transferrin and of ApoC3 and dolichols were analyzed in serum; phosphomannomutase and mannosephosphate isomerase activities were measured in fibroblasts. Conventional or massive parallel sequencing (customized panel or Illumina Clinical-Exome Sequencing TruSight One Gene Panel) was used to identify genes and mutations. RESULTS: Ninety-seven patients were diagnosed with 18 different CDG. Eighty-nine patients had a type 1 transferrin profile; 8 patients had a type 2 transferrin profile, with 6 of them showing an alteration in the ApoC3 isoform profile. A total of 75% of the patients had PMM2-CDG presenting with a heterogeneous mutational spectrum. The remaining patients showed mutations in any of the following genes: MPI, PGM1, GFPT1, SRD5A3, DOLK, DPGAT1, ALG1, ALG6, RFT1, SSR4, B4GALT1, DPM1, COG6, COG7, COG8, ATP6V0A2, and CCDC115. CONCLUSION: Based on literature and on this population-based study of CDG, a comprehensive scheme including reported clinical signs of CDG is offered, which will hopefully reduce the timeframe from clinical suspicion to genetic confirmation. The different defects of CDG identified in Spain have contributed to expand the knowledge of CDG worldwide. A predominance of PMM2 deficiency was detected, with 5 novel PMM2 mutations being described.


Assuntos
Acetiltransferases/metabolismo , Apolipoproteínas C/metabolismo , Defeitos Congênitos da Glicosilação/diagnóstico , Defeitos Congênitos da Glicosilação/epidemiologia , Acetiltransferases/genética , Apolipoproteínas C/genética , Estudos de Coortes , Bases de Dados Factuais , Feminino , Marcadores Genéticos , Predisposição Genética para Doença , Testes Genéticos/métodos , Humanos , Incidência , Recém-Nascido , Masculino , Mutação , Estudos Retrospectivos , Medição de Risco , Espanha/epidemiologia
8.
Eur Arch Psychiatry Clin Neurosci ; 267(3): 199-212, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27549216

RESUMO

Proteomic analyses facilitate the interpretation of molecular biomarker probes which are very helpful in diagnosing schizophrenia (SZ). In the current study, we attempt to test whether potential differences in plasma protein expressions in SZ and bipolar disorder (BD) are associated with cognitive deficits and their underlying brain structures. Forty-two plasma proteins of 29 SZ patients, 25 BD patients and 93 non-clinical controls were quantified and analysed using multiple reaction monitoring-based triple quadrupole mass spectrometry approach. We also computed group comparisons of protein expressions between patients and controls, and between SZ and BD patients, as well. Potential associations of protein levels with cognitive functioning (psychomotor speed, executive functioning, crystallised intelligence) as well as underlying brain volume in the hippocampus were explored, using bivariate correlation analyses. The main finding of this study was that apolipoprotein expression differed between patients and controls and that these alterations in both disease groups were putatively related to cognitive impairments as well as to hippocampus volumes. However, none of the protein level differences were related to clinical symptom severity. In summary, altered apolipoprotein expression in BD and SZ was linked to cognitive decline and underlying morphological changes in both disorders. Our results suggest that the detection of molecular patterns in association with cognitive performance and its underlying brain morphology is of great importance for understanding of the pathological mechanisms of SZ and BD, as well as for supporting the diagnosis and treatment of both disorders.


Assuntos
Apolipoproteínas C/metabolismo , Transtorno Bipolar/complicações , Transtorno Bipolar/patologia , Transtornos Cognitivos/etiologia , Hipocampo/metabolismo , Esquizofrenia/complicações , Esquizofrenia/patologia , Adulto , Proteínas Sanguíneas/metabolismo , Feminino , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Testes Neuropsicológicos , Escalas de Graduação Psiquiátrica , Estatística como Assunto
9.
Orv Hetil ; 157(19): 746-52, 2016 May 08.
Artigo em Húngaro | MEDLINE | ID: mdl-27133274

RESUMO

The incidence and the public health importance of diabetes mellitus are growing continuously. Despite the improvement observed in recent years, the leading cause of morbidity and mortality of diabetics are cardiovascular diseases. The diagnosis of diabetes mellitus constitutes such a high risk as the known presence of vascular disease. Diabetic dyslipidaemia is characterised by high fasting and postprandial triglyceride levels, low HDL level, and slightly elevated LDL-cholesterol with domination of atherogenic small dense LDL. These are not independent components of the atherogenic dyslipidaemia, but are closely linked to each other. Beside the known harmful effects of low HDL and small dense LDL, recent findings confirmed the atherogenicity of the triglyceride-rich lipoproteins and their remnants. It has been shown that the key of this process is the overproduction and delayed clearance of triglyceride-rich lipoproteins in the liver. In this metabolism the lipoprotein lipase has a determining role; its function is accelerated by ApoA5 and attenuated by ApoC3. The null mutations of the ApoC3 results in a reduced risk of myocardial infarction, the loss-of-function mutation of ApoA5 was associated with a 60% elevation of triglyceride level and 2.2-times increased risk of myocardial infarction. In case of diabetes mellitus, insulin resistance, obesity, metabolic syndrome and chronic kidney disease the non-HDL-cholesterol is a better marker of the risk than the LDL-cholesterol. Its value can be calculated by subtraction of HDL-cholesterol from total cholesterol. Target values of non-HDL-cholesterol can be obtained by adding 0.8 mmol/L to the LDL-cholesterol targets (this means 3.3 mmol/L in high, and 2.6 mmol/L in very high risk patients). The drugs of first choice in the treatment of diabetic dyslipidaemia are statins. Nevertheless, it is known that even if statin therapy is optimal (treated to target), a considerable residual (lipid) risk remains. For its reduction treatment of low HDL-cholesterol and high triglyceride levels is obvious by the administration of fibrates. In addition to statin therapy, fenofibrate can be recommended.


Assuntos
Aterosclerose/etiologia , Diabetes Mellitus Tipo 2/complicações , Dislipidemias/tratamento farmacológico , Dislipidemias/etiologia , Hipolipemiantes/uso terapêutico , Lipase Lipoproteica/metabolismo , Triglicerídeos/metabolismo , Apolipoproteína A-V , Apolipoproteínas A/metabolismo , Apolipoproteínas C/metabolismo , Aterosclerose/sangue , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Doenças Cardiovasculares/etiologia , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Complicações do Diabetes/etiologia , Complicações do Diabetes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/sangue , Fenofibrato/uso terapêutico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Resistência à Insulina , Lipoproteínas/sangue , Síndrome Metabólica/complicações , Obesidade/complicações , Valor Preditivo dos Testes , Insuficiência Renal Crônica/complicações , Fatores de Risco , Triglicerídeos/sangue
10.
Chem Phys Lipids ; 195: 63-70, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26756862

RESUMO

Lipoprotein lipase (LPL)-mediated triacylglycerol (TAG) hydrolysis in very low density lipoprotein (VLDL) is accompanied by the release of surface material containing phospholipids (PL), free cholesterol (FC) and apolipoproteins, E (apoE) and Cs (apoCII, apoCIII). The released molecules are accepted by high density lipoprotein (HDL), and new HDL-sized apoE-containing particles are also generated. A decrease in the number of HDL particles or abnormalities in their structure is associated with unfavourable changes in the features of VLDL remnants. Phosphatidylcholine liposomes (PC-L) can also act as acceptors of surface material components released from lipoproteins. Thus, the aim of this study was to assess the impact of liposomes on compositional changes of VLDL during its LPL-mediated lipolysis. VLDL isolated from human sera was incubated with LPL (LPL:VLDLTAG; 24 µg/ml:90 mg/dl) and/or PC-L (VLDLPL:PC-LPL; 1:30 weight ratio). After incubation (2h, 37 °C) VLDL was separated from other reaction products, and VLDL lipid and apolipoprotein content were analysed. Newly generated HDL-sized apoE-containing lipoproteins were separated by two-dimensional non-denaturing gradient gel electrophoresis (2D-PAGGE). The reaction of VLDL with PC-L in the presence or absence of LPL significantly affected the VLDL composition. The ratio of core (TAG+cholesteryl ester) to surface (PL+FC) lipids in VLDL decreased 1.8-fold with PC-L, 1.2-fold with LPL and 3-fold with PC-L+LPL. The reaction with PC-L and PC-L+LPL caused a 3.7-fold and 3.2-fold decrease of apoCs/apoE average weight ratio, respectively. Compositional changes in VLDL under the influence of PC-L were accompanied by an increase in the efficiency of VLDL lipolysis and the generation of apoE-containing HDL-sized particles, heterogeneous in size (from ∼ 9 to ∼ 18.8 nm) and mobility (γ and preß). We conclude that PL-rich particles, similarly to HDL, promote the release of surface material components from VLDL during LPL-mediated lipolysis and positively influence VLDL features which can facilitate VLDL metabolism. Such PC-L activity may impact on its antiatherogenic properties.


Assuntos
Lipase Lipoproteica/metabolismo , Lipossomos/metabolismo , Fosfatidilcolinas/química , Apolipoproteínas C/metabolismo , Apolipoproteínas E/metabolismo , Eletroforese em Gel Bidimensional , Humanos , Lipólise , Lipoproteínas VLDL/química , Lipoproteínas VLDL/metabolismo , Lipossomos/química
11.
Biochim Biophys Acta ; 1831(4): 737-46, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23321267

RESUMO

In human high-density lipoprotein (HDL) represents the major cholesterol carrying lipoprotein class in cord blood, while cholesterol is mainly carried by low-density lipoprotein in maternal serum. Additionally, to carrying cholesterol, HDL also associates with a range of proteins as cargo. We tested the hypothesis that fetal HDL carries proteins qualitatively and quantitatively different from maternal HDL. These differences then contribute to distinct HDL functionality in both circulations. Shotgun proteomics and biochemical analyses were used to assess composition/function of fetal and maternal HDL isolated from uncomplicated human pregnancies at term of gestation. The pattern of analyzed proteins that were statistically elevated in fetal HDL (apoE, proteins involved in coagulation, transport processes) suggests a particle characteristic for the light HDL2 sub-fraction. In contrast, proteins that were enriched in maternal HDL (apoL, apoF, PON1, apoD, apoCs) have been described almost exclusively in the dense HDL3 fraction and relevant to its anti-oxidative function and role in innate immunity. Strikingly, PON1 mass and activity were 5-fold lower (p<0.01) in the fetus, which was accompanied by attenuation of anti-oxidant capacity of fetal HDL. Despite almost equal quantity of CETP in maternal and fetal HDL, its enzymatic activity was 55% lower (p<0.001) in the fetal circulation, whereas LCAT activity was not altered. These findings indicate that maternally derived HDL differs from fetal HDL with respect to its proteome, size and function. Absence of apoA-1, apoL and PON1 on fetal HDL is associated with decreased anti-oxidative properties together with deficiency in innate immunity collectively indicating distinct HDLs in fetuses.


Assuntos
Feto/metabolismo , Lipoproteínas HDL/química , Lipoproteínas HDL/metabolismo , Apolipoproteínas/metabolismo , Apolipoproteínas C/metabolismo , Apolipoproteínas D/metabolismo , Apolipoproteínas E/metabolismo , Arildialquilfosfatase/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Feminino , Humanos , Gravidez
12.
J Biol Chem ; 287(15): 12578-88, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22362781

RESUMO

Hepatic fat accumulation resulting from increased de novo fatty acid synthesis leads to hepatic steatosis and hepatic insulin resistance. We have shown previously that acetyl-CoA carboxylase 2 (Acc2(-/-)) mutant mice, when fed a high-fat (HF) or high-fat, high-carbohydrate (HFHC) diet, are protected against diet-induced obesity and maintained whole body and hepatic insulin sensitivity. To determine the effect of an ACC2 deletion on hepatic fat metabolism, we studied the regulation of the enzymes involved in the lipogenic pathway under Western HFHC dietary and de novo lipogenic conditions. After completing the HFHC regimen, Acc2(-/-) mutant mice were found to have lower body weight, smaller epididymal fat pads, lower blood levels of nonesterified fatty acids and triglycerides, and higher hepatic cholesterol than wild-type mice. Significant up-regulation of lipogenic enzymes and an elevation in hepatic peroxisome proliferator-activated receptor-γ (PPAR-γ) protein were found in Acc2(-/-) mutant mice under de novo lipogenic conditions. The increase in lipogenic enzyme levels was accompanied by up-regulation of the transcription factors, sterol regulatory element-binding proteins 1 and 2, and carbohydrate response element-binding protein. In contrast, hepatic levels of the PPAR-γ and PPAR-α proteins were significantly lower in the Acc2(-/-) mutant mice fed an HFHC diet. When compared with wild-type mice fed the same diet, Acc2(-/-) mutant mice exhibited a similar level of AKT but with a significant increase in pAKT. Hence, deleting ACC2 ameliorates the metabolic syndrome and protects against fatty liver despite increased de novo lipogenesis and dietary conditions known to induce obesity and diabetes.


Assuntos
Acetil-CoA Carboxilase/genética , Dieta Hiperlipídica/efeitos adversos , Carboidratos da Dieta/efeitos adversos , Fígado Gorduroso/enzimologia , Lipogênese , Acetil-CoA Carboxilase/metabolismo , Adiposidade , Animais , Apolipoproteínas C/metabolismo , Glicemia , Peso Corporal , Jejum , Ácido Graxo Sintases/metabolismo , Fígado Gorduroso/sangue , Fígado Gorduroso/etiologia , Regulação da Expressão Gênica , Lipídeos/sangue , Fígado/enzimologia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Tamanho do Órgão , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
13.
Eksp Klin Gastroenterol ; (7): 6-11, 2010.
Artigo em Russo | MEDLINE | ID: mdl-21033077

RESUMO

UNLABELLED: The aim of the work was to study the clinical and genetic factors at children with obese that predispose to the development of MS, and the development of algorithm for generating risk of MS. MATERIALS AND METHODS: Two comparable age and sex groups of children--148 children with obesity and 46--with normal body weight. We assessed anthropometric indices, blood pressure (BP), lipid profile, carbohydrate metabolism, the level of uric acid. 83 children with obesity were genotyped for polymorphisms: I/D gene ACE, G-75A ApoA1, S19W ApoA5, Sstl ApoC3, E2/E3/E4 ApoE and W/R ADRB3. RESULTS: 98,0% of children had abdominal obesity. In 35,8% was identified high blood pressure. In 47,4% was diagnosed hypo-alpha cholesterolemia and/or hypertriglyceridemia (HTG). In 21,0% of children was identified hyperglycemia. 25,7%were suffered from hyperuricemia. Among the genotyped children 57,0% of homo-and heterozygous carriers of D allele ACE gene had high blood pressure. More than half of the holders of 19W-allele ApoA5 (68,5%),--75A-allele of ApoA1 (56,0%), 52-allele of the gene ApoC3 (53,0%), E4-ApoE gene (85,7%), in the heterozygous state had metabolic TG and/or HDL. In 60,3% of the carriers W/W genotype of ADRB3 gene revealed a combination of hyperglycemia with hyperinsulinemia and/or TG. CONCLUSION: As a result of, aiming aimed at early detection of the major manifestations of MS clinical and genetic study was revealed stable combination of constitutional, metabolic and molecular-genetic factors. Based on these data was developed algorithm for forming groups at risk of MS and individual tactics to prevent and/or therapy.


Assuntos
Síndrome Metabólica/genética , Obesidade Abdominal/genética , Polimorfismo Genético , Adolescente , Alelos , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Apolipoproteína A-V , Apolipoproteínas A/genética , Apolipoproteínas A/metabolismo , Apolipoproteínas C/genética , Apolipoproteínas C/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Criança , Feminino , Genótipo , Humanos , Masculino , Síndrome Metabólica/metabolismo , Síndrome Metabólica/fisiopatologia , Obesidade Abdominal/metabolismo , Obesidade Abdominal/fisiopatologia , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo
14.
J Lipid Res ; 51(6): 1524-34, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20097930

RESUMO

We have shown that expression of apolipoprotein (apo) C-III promotes VLDL secretion from transfected McA-RH7777 cells under lipid-rich conditions. To determine structural elements within apoC-III that confer to this function, we contrasted wild-type apoC-III with a mutant Ala23Thr originally identified in hypotriglyceridemia subjects. Although synthesis of [(3)H]glycerol-labeled TAG was comparable between cells expressing wild-type apoC-III (C3wt cells) or Ala23Thr mutant (C3AT cells), secretion of [(3)H]TAG from C3AT cells was markedly decreased. The lowered [(3)H]TAG secretion was associated with an inability of C3AT cells to assemble VLDL(1). Moreover, [(3)H]TAG within the microsomal lumen in C3AT cells was 60% higher than that in C3wt cells, yet the activity of microsomal triglyceride-transfer protein in C3AT cells was not elevated. The accumulated [(3)H]TAG in C3AT microsomal lumen was mainly associated with lumenal IDL/LDL-like lipoproteins. Phenotypically, this [(3)H]TAG fractionation profiling resembled what was observed in cells treated with brefeldin A, which at low dose specifically blocked the second-step VLDL(1) maturation. Furthermore, lumenal [(35)S]Ala23Thr protein accumulated in IDL/LDL fractions and was absent in VLDL fractions in C3AT cells. These results suggest that the presence of Ala23Thr protein in lumenal IDL/LDL particles might prevent effective fusion between lipid droplets and VLDL precursors. Thus, the current study reveals an important structural element residing within the N-terminal region of apoC-III that governs the second step VLDL(1) maturation.


Assuntos
Apolipoproteínas C/genética , Apolipoproteínas C/metabolismo , Transtornos do Metabolismo dos Lipídeos/genética , Mutação de Sentido Incorreto , Triglicerídeos/metabolismo , Alanina/genética , Apolipoproteínas C/química , Brefeldina A/farmacologia , Proteínas de Transporte/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Lipoproteínas IDL/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/metabolismo , Microssomos/metabolismo , Modelos Moleculares , Ligação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Treonina/genética
15.
J Lipid Res ; 50(9): 1842-51, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19417222

RESUMO

Plasma cholesteryl ester transfer protein (CETP) activity is high in rabbits, intermediate in humans, and nondetectable in rodents. Human apolipoprotein CI (apoCI) was found to be a potent inhibitor of CETP. The aim of this study was to compare the ability of rabbit and human apoCI to modulate the interaction of CETP with HDLs and to evaluate to which extent apoCI contributes to plasma cholesteryl ester transfer rate in normolipidemic humans and rabbits. Rabbit apoCI gene was cloned and sequenced, rabbit and human apoCI were purified to homogeneity, and their ability to modify the surface charge properties and the CETP inhibitory potential of HDL were compared. It is demonstrated that unlike human apoCI, rabbit apoCI does not modulate cholesteryl ester transfer rate in total plasma. Whereas both human and rabbit apoCI readily associate with HDL, only human apoCI was found to modify the electrostatic charge of HDL. In humans, both CETP and apoCI at normal, physiological levels contribute significantly to the plasma cholesteryl ester transfer rate. In contrast, CETP is the sole major determinant of cholesteryl ester transfer in normolipidemic rabbit plasma as a result of the inability of rabbit apoCI to change HDL electronegativity.


Assuntos
Apolipoproteínas C/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/sangue , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Sequência de Aminoácidos , Animais , Apolipoproteínas C/química , Apolipoproteínas C/genética , Clonagem Molecular , Feminino , Humanos , Lipase Lipoproteica/metabolismo , Lipoproteínas HDL/metabolismo , Masculino , Pessoa de Meia-Idade , Coelhos , Análise de Sequência de DNA
16.
Exp Cell Res ; 315(7): 1200-11, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19353766

RESUMO

Hepatocyte nuclear factor 4alpha (HNF4alpha) is a critical transcription factor for pancreas and liver development and functions in islet beta cells to maintain glucose homeostasis. Mutations in the human HNF4A gene lead to maturity onset diabetes of the young (MODY1) and polymorphisms are associated with increased risk for type 2 diabetes mellitus (T2DM). Expression of six HNF4alpha variants, three each from two developmentally regulated promoters, has been firmly established. We have now detected a new set of HNF4alpha variants designated HNF4alpha10-12 expressed from distal promoter P2. These variants, generated by inclusion of previously undetected exon 1E (human=222 nt, rodent=136 nt) following exon 1D have an altered N-terminus but identical remaining reading frame. HNF4alpha10-alpha12 are expressed in pancreatic islets (and liver) and exhibit transactivation potentials similar to the corresponding alpha7-alpha9 isoforms. DNA-binding analyses implied much higher protein levels of HNF4alpha10-alpha12 in liver than expected from the RT-PCR data. Our results provide evidence for a more complex expression pattern of HNF4alpha than previously appreciated. We recommend inclusion of exon 1E and nearby DNA sequences in screening for HNF4alpha mutations and polymorphisms in genetic analyses of MODY1 and T2DM.


Assuntos
Diabetes Mellitus Tipo 2/genética , Éxons , Fator 4 Nuclear de Hepatócito , Mutagênese Insercional , Polimorfismo Genético , Regiões Promotoras Genéticas , Isoformas de Proteínas , Animais , Apolipoproteínas C/genética , Apolipoproteínas C/metabolismo , Sequência de Bases , Linhagem Celular , Diabetes Mellitus Tipo 2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alinhamento de Sequência
17.
J Hepatol ; 49(5): 787-98, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18809223

RESUMO

BACKGROUND/AIMS: We previously reported that hepatitis C virus (HCV) core protein up regulated transcription of apolipoprotein C-IV (ApoC-IV, 10.7-fold increase), a member of the apolipoprotein family implicated in liver steatosis. Here, we identified host transcription factors regulating the ApoC-IV gene expression. METHODS: Transcriptional regulators were identified by DNA affinity purification and steatosis was detected by oil red O staining and triglyceride assay. RESULTS: We defined a 163-bp ApoC-IV promoter as a core protein responsive element, and identified Ku antigen complex (Ku70 and Ku80) as well as nuclear receptors PPARgamma/RXRalpha as key regulators of ApoC-IV gene expression. Both Ku70 overexpression and PPARgamma agonist significantly increased ApoC-IV promoter activity; conversely, Ku70 silencing or mutation of PPARgamma binding site diminished the ApoC-IV promoter activity. Interestingly, transient transfection of ApoC-IV cDNA into a human hepatoma cell line was able to trigger moderate lipid accumulation. In agreement with this in vitro study, ApoC-IV transcript level was increased in HCV infected livers which correlated with triglyceride accumulation. CONCLUSIONS: ApoC-IV overexpression may perturb lipid metabolism leading to lipid accumulation. HCV core protein may modulate ApoC-IV expression through Ku antigen and PPARgamma/RXRalpha complex.


Assuntos
Apolipoproteínas C/genética , DNA Helicases/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , PPAR gama/metabolismo , Apolipoproteínas C/metabolismo , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Hepacivirus/genética , Hepatite C/genética , Hepatite C/metabolismo , Hepatite C/virologia , Humanos , Autoantígeno Ku , Metabolismo dos Lipídeos , Modelos Biológicos , Dados de Sequência Molecular , Complexos Multiproteicos , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Receptor X Retinoide alfa/metabolismo , Ativação Transcricional , Proteínas do Core Viral/genética
18.
Circulation ; 116(19): 2173-81, 2007 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-17967778

RESUMO

BACKGROUND: Lipopolysaccharide (LPS), which is released from gram-negative bacteria on multiplication or lysis, aggravates atherosclerosis in humans and rodents by inducing inflammation via toll-like receptors. Because apolipoprotein C-I (apoCI) enhances the LPS-induced inflammatory response in macrophages in vitro and in mice, we investigated the effect of endogenous apoCI expression on LPS-induced atherosclerosis in mice. METHODS AND RESULTS: Twelve-week-old apoe-/- apoc1-/- and apoe-/- apoc1+/+ mice received weekly intraperitoneal injections of LPS (50 microg) or vehicle for a period of 10 weeks, and atherosclerosis development was assessed in the aortic root. LPS administration did not affect atherosclerotic lesion area in apoe-/- apoc1-/- mice but increased it in apoe-/- apoc1+/+ mice. In fact, apoCI expression increased the LPS-induced atherosclerotic lesion area by 60% (P<0.05), concomitant with an increase in LPS-induced plasma levels of fibrinogen and E-selectin. This indicated that apoCI increased the LPS-induced inflammatory state, both systemically (ie, fibrinogen) and at the level of the vessel wall (ie, E-selectin). In addition, both macrophage-derived apoCI and HDL-associated apoCI increased the LPS-induced tumor necrosis factor-alpha response by macrophages in vitro. CONCLUSIONS: We conclude that apoCI is crucially involved in LPS-induced atherosclerosis in apoe-/- mice, which mainly relates to an increased inflammatory response toward LPS. We anticipate that apoCI plasma levels contribute to accelerated atherosclerosis development in individuals who have chronic infection.


Assuntos
Apolipoproteínas C/genética , Apolipoproteínas C/metabolismo , Aterosclerose/genética , Aterosclerose/imunologia , Animais , Aterosclerose/patologia , Biomarcadores/sangue , Células Cultivadas , HDL-Colesterol/sangue , Selectina E/sangue , Feminino , Fibrinogênio/metabolismo , Hipercolesterolemia/genética , Hipercolesterolemia/imunologia , Hipercolesterolemia/patologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Monócitos/patologia , Linfócitos T/patologia , Fator de Necrose Tumoral alfa/metabolismo , Vasculite/genética , Vasculite/imunologia , Vasculite/patologia
19.
Biochem J ; 398(3): 439-50, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16771709

RESUMO

HNF-4 (hepatocyte nuclear factor 4) is a key regulator of liver-specific gene expression in mammals. We have shown previously that the activity of the human APOC3 (apolipoprotein C-III) promoter is positively regulated by the anti-inflammatory cytokine TGFbeta (transforming growth factor beta) and its effectors Smad3 (similar to mothers against decapentaplegic 3) and Smad4 proteins via physical and functional interactions between Smads and HNF-4. We now show that the pro-inflammatory cytokine TNFalpha (tumour necrosis factor alpha) antagonizes TGFbeta for the regulation of APOC3 gene expression in hepatocytes. TNFalpha was a strong inhibitor of the activity of apolipoprotein promoters that harbour HNF-4 binding sites and this inhibition required HNF-4. Using specific inhibitors of TNFalpha-induced signalling pathways, it was shown that inhibition of the APOC3 promoter by TNFalpha involved NF-kappaB (nuclear factor kappaB). Latent membrane protein 1 of the Epstein-Barr virus, which is an established potent activator of NF-kappaB as well as wild-type forms of various NF-kappaB signalling mediators, also inhibited strongly the APOC3 promoter and the transactivation function of HNF-4. TNFalpha had no effect on the stability or the nuclear localization of HNF-4 in HepG2 cells, but inhibited the binding of HNF-4 to the proximal APOC3 HRE (hormone response element). Using the yeast-transactivator-GAL4 system, we showed that both AF-1 and AF-2 (activation functions 1 and 2) of HNF-4 are inhibited by TNFalpha and that this inhibition was abolished by overexpression of different HNF-4 co-activators, including PGC-1 (peroxisome-proliferator-activated-receptor-gamma co-activator 1), CBP [CREB (cAMP-response-element-binding protein) binding protein] and SRC3 (steroid receptor co-activator 3). In summary, our findings indicate that TNFalpha, or other factors that trigger an NF-kappaB response in hepatic cells, inhibit the transcriptional activity of the APOC3 and other HNF-4-dependent promoters and that this inhibition could be accounted for by a decrease in DNA binding and the down-regulation of the transactivation potential of the AF-1 and AF-2 domains of HNF-4.


Assuntos
Fator 4 Nuclear de Hepatócito/antagonistas & inibidores , Fator 4 Nuclear de Hepatócito/metabolismo , NF-kappa B/metabolismo , Transcrição Gênica/fisiologia , Animais , Apolipoproteína C-III , Apolipoproteínas C/genética , Apolipoproteínas C/metabolismo , Linhagem Celular , Chlorocebus aethiops , Fator 4 Nuclear de Hepatócito/genética , Humanos , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Ativação Transcricional , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
20.
Nutr Metab Cardiovasc Dis ; 16(3): 215-21, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16580589

RESUMO

BACKGROUND AND AIM: ApoE and apoC-III are important components of lipoprotein metabolism. While the function of both apoproteins is relatively well understood, little is known about the in vivo metabolism of these proteins, partly because of the lack of a standardized method to isolate these apoproteins in large sample numbers. METHODS AND RESULTS: We developed a new reverse phase HPLC method (acetonitril/phosphate gradient; Aquapore RP-300, 7 microm, 220 x 4.6 mm) to isolate a number of different apoproteins, including apoC-III and apoE from VLDL. This method was then used in a study which aimed at determining VLDL-apoE-3 and VLDL-apoC-III metabolism. In addition VLDL-apoB and LDL-apoB metabolism was determined. Endogenous labeling with d(3)-leucine, mass spectrometry and multicompartmental modeling was used in 6 normolipidemic healthy male subjects. Tracer/tracee ratios of free plasma leucine, VLDL-apoE, -apoC-III, -apoB, and LDL-apoB leucine were determined over 60 h following a bolus of d(3)-leucine (5 mg kg(-1)). In all subjects sufficient apoC-III could be isolated by reverse phase HPLC to derive metabolic parameters, while apoE metabolic parameters could only be determined if apoE plasma concentration was 0.75 mg dl(-1) or higher. Compared to VLDL-apoB (FCR 10.4 +/- 3.3 d(-1), production 17.8 +/- 4.5 mg kg(-1) d(-1)), VLDL-apoE-3 (FCR 1.03 +/- 0.11 d(-1), production 0.50 +/- 0.29 mg kg(-1) d(-1)) and VLDL-apoC-III (FCR 1.67 +/- 1.22 d(-1), production 0.44 +/- 0.24 mg kg(-1) d(-1)) parameters were much lower. This indicates that apoE-3 and apoC-III recirculate in plasma and that only a small fraction of apoE and apoC-III on VLDL is newly synthesized. CONCLUSIONS: We conclude that HPLC methodology can be used to isolate VLDL-apoC-III and apoE for metabolic studies and that the metabolic fate of apoC-III and apoE is different from that of apoB because both apoproteins recycle through the VLDL fraction.


Assuntos
Apolipoproteínas B/metabolismo , Apolipoproteínas C/metabolismo , Apolipoproteínas E/metabolismo , VLDL-Colesterol/química , Cromatografia Líquida de Alta Pressão/métodos , Lipoproteínas/metabolismo , Adulto , Apolipoproteína C-III , Apolipoproteínas B/sangue , Apolipoproteínas C/sangue , Apolipoproteínas E/sangue , Área Sob a Curva , VLDL-Colesterol/análise , VLDL-Colesterol/metabolismo , Humanos , Leucina/metabolismo , Lipoproteínas VLDL/análise , Lipoproteínas VLDL/química , Lipoproteínas VLDL/metabolismo , Masculino , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA