Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 414
Filtrar
1.
J Lipid Res ; 62: 100130, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34606887

RESUMO

In 2003, the sequences of mammalian proprotein convertase subtilisin/kexin type 9 (PCSK9) were reported. Radiolabeling pulse-chase analyses demonstrated that PCSK9 was synthesized as a precursor (proPCSK9) that undergoes autocatalytic cleavage in the endoplasmic reticulum into PCSK9, which is then secreted as an inactive enzyme in complex with its inhibitory prodomain. Its high mRNA expression in liver hepatocytes and its gene localization on chromosome 1p32, a third locus associated with familial hypercholesterolemia, other than LDLR or APOB, led us to identify three patient families expressing the PCSK9 variants S127R or F216L. Although Pcsk9 and Ldlr were downregulated in mice that were fed a cholesterol-rich diet, PCSK9 overexpression led to the degradation of the LDLR. This led to the demonstration that gain-of-function and loss-of-function variations in PCSK9 modulate its bioactivity, whereby PCSK9 binds the LDLR in a nonenzymatic fashion to induce its degradation in endosomes/lysosomes. PCSK9 was also shown to play major roles in targeting other receptors for degradation, thereby regulating various processes, including hypercholesterolemia and associated atherosclerosis, vascular inflammation, viral infections, and immune checkpoint regulation in cancer. Injectable PCSK9 monoclonal antibody or siRNA is currently used in clinics worldwide to treat hypercholesterolemia and could be combined with current therapies in cancer/metastasis. In this review, we present the critical information that led to the discovery of PCSK9 and its implication in LDL-C metabolism. We further analyze the underlying functional mechanism(s) in the regulation of LDL-C, as well as the evolving novel roles of PCSK9 in both health and disease states.


Assuntos
Hipercolesterolemia/enzimologia , Neoplasias/enzimologia , Pró-Proteína Convertase 9/metabolismo , Viroses/enzimologia , Animais , Humanos
2.
J Cell Physiol ; 236(4): 2333-2351, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32875580

RESUMO

Proprotein convertase subtilisin/kexin type-9 (PCSK9), a member of the proprotein convertase family, is an important drug target because of its crucial role in lipid metabolism. Emerging evidence suggests a direct role of localized PCSK9 in the pathogenesis of vascular diseases. With this in our consideration, we reviewed PCSK9 physiology with respect to recent development and major studies (clinical and experimental) on PCSK9 functionality in vascular disease. PCSK9 upregulates low-density lipoprotein (LDL)-cholesterol levels by binding to the LDL-receptor (LDLR) and facilitating its lysosomal degradation. PCSK9 gain-of-function mutations have been confirmed as a novel genetic mechanism for familial hypercholesterolemia. Elevated serum PCSK9 levels in patients with vascular diseases may contribute to coronary artery disease, atherosclerosis, cerebrovascular diseases, vasculitis, aortic diseases, and arterial aging pathogenesis. Experimental models of atherosclerosis, arterial aneurysm, and coronary or carotid artery ligation also support PCSK9 contribution to inflammatory response and disease progression, through LDLR-dependent or -independent mechanisms. More recently, several clinical trials have confirmed that anti-PCSK9 monoclonal antibodies can reduce systemic LDL levels, total nonfatal cardiovascular events, and all-cause mortality. Interaction of PCSK9 with other receptor proteins (LDLR-related proteins, cluster of differentiation family members, epithelial Na+ channels, and sortilin) may underlie its roles in vascular disease. Improved understanding of PCSK9 roles and molecular mechanisms in various vascular diseases will facilitate advances in lipid-lowering therapy and disease prevention.


Assuntos
Artérias/enzimologia , Hipercolesterolemia/enzimologia , Pró-Proteína Convertase 9/metabolismo , Doenças Vasculares/enzimologia , Animais , Anticolesterolemiantes/uso terapêutico , Artérias/efeitos dos fármacos , Artérias/patologia , Regulação Enzimológica da Expressão Gênica , Humanos , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/genética , Hipercolesterolemia/patologia , Mutação , Inibidores de PCSK9 , Pró-Proteína Convertase 9/genética , Inibidores de Serina Proteinase/uso terapêutico , Transdução de Sinais , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/genética , Doenças Vasculares/patologia
3.
Toxicol Appl Pharmacol ; 412: 115388, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33383043

RESUMO

Phytosterol diosgenin (DG) exhibits cholesterol-lowering properties. Few studies focused on the underlying mechanism of DG attenuation of hypercholesterolemia by promoting cholesterol metabolism. To investigate the roles of SRB1/CES-1/CYP7A1/FXR pathways in accelerating cholesterol elimination and alleviating hypercholesterolemia, a rat model of hypercholesterolemia was induced by providing a high-fat diet (HFD). Experimental rat models were randomly divided into a normal control (Con) group, HFD group, low-dose DG (LDG) group (150 mg/kg/d), high-dose DG (HDG) group (300 mg/kg) and Simvastatin (Sim) group (4 mg/kg/d). Body weights, serum and hepatic lipid parameters of rats were tested. The expression levels of scavenger receptor class B type I (SRB1), carboxylesterase-1 (CES-1), cholesterol7α- hydroxylase (CYP7A1), and farnesoid X receptor (FXR) were determined. The results showed that DG reduced weight and lowered lipid levels in HFD-fed rats. Pathological morphology analyses revealed that DG notably improved hepatic steatosis and intestinal structure. Further studies showed the increased hepatic SRB1, CES-1, CYP7A1 and inhibited FXR-mediated signaling in DG-fed rats, which contributing to the decrease of hepatic cholesterol. DG also increased intestinal SRB1 and CES-1, inhibiting cholesterol absorption and promoting RCT. The expression levels of these receptors in the HDG group were higher than LDG and Sim groups. These data suggested that DG accelerated reverse cholesterol transport (RCT) and enhanced cholesterol elimination via SRB1/CES-1/CYP7A1/FXR pathway, and DG might be a new candidate for the alleviation of hypercholesterolemia.


Assuntos
Anticolesterolemiantes/farmacologia , Hidrolases de Éster Carboxílico/metabolismo , Colesterol 7-alfa-Hidroxilase/metabolismo , Colesterol/sangue , Diosgenina/farmacologia , Hipercolesterolemia/prevenção & controle , Fígado/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Depuradores Classe B/metabolismo , Animais , Biomarcadores/sangue , Dieta Hiperlipídica , Modelos Animais de Doenças , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hipercolesterolemia/sangue , Hipercolesterolemia/enzimologia , Hipercolesterolemia/patologia , Fígado/enzimologia , Fígado/patologia , Masculino , Ratos Sprague-Dawley , Transdução de Sinais , Sinvastatina/farmacologia
4.
Pharmacol Res ; 161: 105205, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32998069

RESUMO

The purpose of this study was to investigate the role of piceatannol (PT) in statin (rosuvastatin and simvastatin) resistance and tolerance and its association with PCSK9 expression via its p300 inhibitory (p300i) activity. An in vitro study was performed using HepG2 cells that were exposed to statins (rosuvastatin or simvastatin) with or without PT in delipidated serum (DLPS) medium. In the statin exposed conditions, PCSK9 expression was reduced following PT treatment when compared to HepG2 cells w/o PT treatment. Furthermore, no significant difference was observed in the expression of the transcription factors SREBP2 and HNF1α, which regulate PCSK9 expression. This resulted in low density lipoprotein receptor (LDLR) stabilization and reduced cellular cholesterol levels. This indicates that PT epigenetically controls statin-induced PCSK9 expression. Interestingly, PT attenuated p300 histone acetyltransferase (HAT) activity. Moreover, simulation of PT-p300 binding suggested that PT inhibits p300 as PT could be docked in the p300 HAT domain. Furthermore, inhibition of p300 HAT activity using C-646, a selective p300 inhibitor, or through an siRNA system effectively reduced PCSK9 induction upon statin exposure in HepG2 cells. The chromatin immunoprecipitation (ChIP) assays revealed that PT blocked the recruitment of p300 to the PCSK9 promoter region. In summary, PT attenuated statin-induced PCSK9 expression by inhibiting p300 HAT activity. Finally, co-administration of simvastatin and PT for 10 weeks further reduced plasma low-density lipoprotein-cholesterol (LDL-C) levels and stabilized the hepatic LDLR protein level compared with those resulting from single treatment of simvastatin in a high-fat diet-induced hypercholesterolemia mouse model. Our findings indicate that PT is a new nutraceutical candidate to reduce the statin resistance and tolerance that occurs in patients with hypercholesterolemia.


Assuntos
Hepatócitos/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hipercolesterolemia/tratamento farmacológico , Pró-Proteína Convertase 9/metabolismo , Rosuvastatina Cálcica/farmacologia , Sinvastatina/farmacologia , Estilbenos/farmacologia , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Animais , LDL-Colesterol/sangue , Modelos Animais de Doenças , Regulação para Baixo , Resistência a Medicamentos , Células Hep G2 , Hepatócitos/enzimologia , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/enzimologia , Hipercolesterolemia/genética , Masculino , Camundongos Endogâmicos C57BL , Pró-Proteína Convertase 9/genética , Estabilidade Proteica , Receptores de LDL/metabolismo , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
5.
Molecules ; 25(17)2020 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-32859023

RESUMO

This review provides an updated atomic-level perspective regarding the enzyme 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoAR), linking the more recent data on this enzyme with a structure/function interpretation. This enzyme catalyzes one of the most important steps in cholesterol biosynthesis and is regarded as one of the most important drug targets in the treatment of hypercholesterolemia. Taking this into consideration, we review in the present article several aspects of this enzyme, including its structure and biochemistry, its catalytic mechanism and different reported and proposed approaches for inhibiting this enzyme, including the commercially available statins or the possibility of using dimerization inhibitors.


Assuntos
Acil Coenzima A/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases , Hidroximetilglutaril-CoA Redutases NAD-Dependentes , Hipercolesterolemia , Multimerização Proteica/efeitos dos fármacos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/química , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hidroximetilglutaril-CoA Redutases NAD-Dependentes/química , Hidroximetilglutaril-CoA Redutases NAD-Dependentes/metabolismo , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/enzimologia
7.
Expert Opin Pharmacother ; 21(16): 1971-1974, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32749892

RESUMO

INTRODUCTION: If statins are unsuccessful at achieving the LDL cholesterol level goal in subjects with hypercholesterolemia, non-statin therapy should be added to reduce cardiovascular morbidity and mortality. The first inhibitors of proprotein convertase substilisin-kexin type 9 (PCSK9) were human monoclonal antibodies and these reduced LDL cholesterol and cardiovascular events. Inclisiran is a small interfering RNA molecule (siRNAs) directed against PCSK9. AREAS COVERED: This key paper evaluation focuses on Phase 3 trials that assess inclisiran in the treatment of hypercholesterolemia and heterozygous familial hypercholesterolemia. EXPERT OPINION: To date, the findings with inclisiran have been very promising as it causes large decreases in LDL cholesterol with few adverse effects. However, there are some limitations to its widespread use. Firstly, cardiovascular outcomes trials have not been completed, so we do not know how inclisiran compares to the PCSK9 monoclonal antibodies, which, seem to me, to only have a modest effect on cardiovascular outcomes. Secondly, a major problem with the PCSK9 monoclonal antibodies is that they are expensive, and their use is often discontinued or not pursued, which can leave the subjects intended for treatment at high cardiovascular risk. At present, it is not clear whether similar problems around cost will apply to inclisiran.


Assuntos
Anticolesterolemiantes/uso terapêutico , Hipercolesterolemia/tratamento farmacológico , Inibidores de PCSK9 , RNA Interferente Pequeno/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Anticolesterolemiantes/administração & dosagem , Anticolesterolemiantes/economia , LDL-Colesterol/sangue , Ensaios Clínicos como Assunto , Esquema de Medicação , Custos de Medicamentos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipercolesterolemia/sangue , Hipercolesterolemia/enzimologia , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Hiperlipoproteinemia Tipo II/enzimologia , Adesão à Medicação , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/economia
8.
Cardiovasc Drugs Ther ; 34(4): 475-485, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32415571

RESUMO

BACKGROUND: Proprotein convertase subtilisin/kexin 9 (PCSK9) serves a key regulatory function in the metabolism of low-density lipoprotein (LDL)-cholesterol (LDL-C) through interaction with the LDL receptor (LDLR) followed by its destruction that results in the elevation of the plasma levels of LDL-C. The aims of the present study were to separate and select a number of single-stranded DNA (ssDNA) aptamers against PCSK9 from a library pool (n > 1012) followed by their characterization. METHODS: The aptamers obtained from the DNA-PCSK9 complexes which presented the highest affinity against PCSK9 were separated and selected using capillary electrophoresis evolution of ligands by exponential enrichment (CE-SELEX). The selected aptamers were amplified and cloned into a T/A vector. The plasmids from the positive clones were extracted and sequenced. The Mfold web server was used to predict the secondary structure of the aptamers. RESULTS: Following three rounds of CE-SELEX, the identified anti-PCSK9 ssDNA aptamers, namely aptamer 1 (AP-1) and aptamer 2 (AP-2), presented half maximal inhibitory concentrations of 325 and 327 nM, lowest dissociation constants of 294 and 323 nM, and most negative Gibbs free energy values of - 9.17 and - 8.28 kcal/mol, respectively. CONCLUSION: The results indicated that the selected aptamers (AP-1 and AP-2) induced potent inhibitory effects against PCSK9. Further in vivo studies demand to find out AP-1 and AP-2 aptamers as suitable candidates, instead of antibodies, for using in therapeutic purposes in patients with hypercholesterolemia and cardiovascular disease.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , DNA de Cadeia Simples/farmacologia , Biblioteca Gênica , Hipercolesterolemia/tratamento farmacológico , Inibidores de PCSK9 , Técnica de Seleção de Aptâmeros , Anticolesterolemiantes , Aptâmeros de Nucleotídeos/genética , Aptâmeros de Nucleotídeos/metabolismo , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Humanos , Hipercolesterolemia/enzimologia , Hipercolesterolemia/genética , Terapia de Alvo Molecular , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo
9.
Nutr Metab Cardiovasc Dis ; 30(7): 1137-1146, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32456947

RESUMO

BACKGROUND AND AIMS: The increased risk of cardiovascular disease under hypercholesterolemia is due to associations between oxidized low-density lipoprotein (ox-LDL) and lipoprotein-associated phospholipase A2 (Lp-PLA2) and between ox-LDL and coagulant profiles. We investigated the impact of different ox-LDL levels on coagulation time and plasma metabolomes in subjects with borderline hypercholesterolemia. METHODS AND RESULTS: One hundred thirty-one subjects with borderline hypercholesterolemia (serum cholesterol ≥200 mg/dL) were divided into low ox-LDL (n = 66) and high ox-LDL (n = 65) groups. After adjusting for confounding factors, the high ox-LDL group exhibited a significantly decreased activated partial thromboplastin time (aPTT) and prothrombin time (PT) and increased Lp-PLA2 activity. Compared to the low ox-LDL group, the high ox-LDL group exhibited significantly increased intensities of 17 lysophosphatidylcholines (lysoPCs) and 7 lysophosphatidylethanolamines (lysoPEs). Ox-LDL was inversely correlated with aPTT and PT and positively correlated with Lp-PLA2 activity. Positive correlations were also found among ox-LDL, Lp-PLA2 activity, lysoPCs, and lysoPEs. LysoPCs and lysoPEs were inversely correlated with PT and aPTT. The identified plasma metabolites, including amino acids, fatty acid amides, acylcarnitines, and lysophospholipids, were significantly upregulated in the high ox-LDL group. CONCLUSION: High ox-LDL levels may be involved in the development of a procoagulant state in subjects with borderline hypercholesterolemia by increasing Lp-PLA2 activity and lysoPC and lysoPE levels.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/sangue , Fatores de Coagulação Sanguínea/análise , Coagulação Sanguínea , Hipercolesterolemia/sangue , Lipoproteínas LDL/sangue , Lisofosfatidilcolinas/sangue , Lisofosfolipídeos/sangue , Biomarcadores/sangue , Feminino , Humanos , Hipercolesterolemia/diagnóstico , Hipercolesterolemia/enzimologia , Masculino , Metabolômica , Pessoa de Meia-Idade , Tempo de Tromboplastina Parcial , Tempo de Protrombina , Regulação para Cima
10.
EBioMedicine ; 55: 102755, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32335374

RESUMO

BACKGROUND: Vorolanib (X-82, CM082) is a multi-target tyrosine kinase inhibitor. This study aimed to evaluate the tolerability, safety, pharmacokinetics and antitumor activities of vorolanib plus everolimus (an inhibitor of mammalian target of rapamycin). METHODS: Patients had histologically or cytologically confirmed advanced RCC and failed with standard therapy were eligible for this study. Dose-escalated combinations of vorolanib (100, 150 or 200 mg once daily) with everolimus (5 mg once daily) were administered on 28-day cycles until disease progression or unacceptable toxicity using a conventional 3 + 3 dose-escalation design. FINDINGS: 22 patients (100 mg n = 4, 150 mg n = 3, 200 mg n = 15) were enrolled. Only one patient experienced dose-limiting toxicity (DLT, grade 4 thrombocytopenia) in the vorolanib 200 mg combination cohort, and the maximum tolerated dose (MTD) was not reached. The most common treatment-related adverse events were proteinuria (100%), leukopenia (77%), hypercholesterolaemia (77%), increased low-density lipoprotein (68%), hypertriglyceridaemia (64%), hyperglycaemia (59%), and fatigue (55%). Most treatment-related adverse events were grade 1 to 2, with grade 3 or higher toxicities mostly seen in the 200 mg cohort. Single dosing of vorolanib demonstrated dose-proportional increases in the Cmax and AUC, and observed short t1/2z ranging from 4.74±1.44 to 12.89±7.49 h. The pharmacokinetic parameters for everolimus were similar among all cohorts. Of 19 evaluable patients, the ORR and DCR was 32% (n = 6, 95% CI, 13-57%) and 100% (95% CI, 82-100%), respectively. INTERPRETATION: Combination therapy of vorolanib 200 mg plus everolimus 5 mg once daily is potentially effective with potential activity. Further evaluation of the combination in advanced RCC patients is ongoing (NCT03095040). FUNDING: Betta Pharmaceutical Co., Ltd., Hangzhou, China.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Carcinoma de Células Renais/tratamento farmacológico , Everolimo/administração & dosagem , Indóis/administração & dosagem , Neoplasias Renais/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Pirróis/administração & dosagem , Pirrolidinas/administração & dosagem , Adolescente , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carcinoma de Células Renais/enzimologia , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Progressão da Doença , Esquema de Medicação , Everolimo/efeitos adversos , Feminino , Humanos , Hipercolesterolemia/enzimologia , Hipercolesterolemia/etiologia , Hipercolesterolemia/mortalidade , Hiperglicemia/enzimologia , Hiperglicemia/etiologia , Hiperglicemia/mortalidade , Indóis/efeitos adversos , Neoplasias Renais/enzimologia , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Leucopenia/enzimologia , Leucopenia/etiologia , Leucopenia/mortalidade , Masculino , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/efeitos adversos , Proteinúria/enzimologia , Proteinúria/etiologia , Proteinúria/mortalidade , Pirróis/efeitos adversos , Pirrolidinas/efeitos adversos , Análise de Sobrevida
11.
Nutr Metab Cardiovasc Dis ; 30(2): 282-291, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-31653513

RESUMO

BACKGROUND AND AIMS: In the association between hypercholesterolemia (HC) and thrombotic risk platelet hyper-reactivity plays an important role. The inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) to reduce plasma LDL-cholesterol merges as effective therapeutic strategy to prevent cardiovascular (CV) events. Aim of this study was to verify whether a treatment up to 12 months with the monoclonal antibodies (mAbs) anti-PCSK9 influences platelet function in primary HC. METHODS AND RESULTS: In patients affected by primary HC (n = 24), all on background of statin and 17 on acetyl salicylic acid (ASA), platelet function parameters were evaluated at baseline up to 12 months of treatment with the mAb anti-PCSK9 alirocumab or evolocumab. From baseline, the treatment with anti-PCSK9 mAbs: i) in ASA HC patients, significantly decreased platelet aggregation detected in platelet-rich plasma by light transmission aggregometry and in whole blood Platelet Function Analyzer-100 assay; ii) in all HC patients, significantly decreased platelet membrane expression of CD62P and plasma levels of the in vivo platelet activation markers soluble CD40 Ligand, Platelet Factor-4, and soluble P-Selectin. Furthermore, CD62P expression, and sP-Selectin, PF-4, sCD40L levels significantly correlated with serum PCSK9. CONCLUSION: Besides markedly lowering LDL-c levels, our results suggest that HC patients benefit from anti-PCSK9 mAb treatment also for reducing platelet reactivity and increasing platelet sensitivity to the inhibitory effects of aspirin. These effects on platelets could play a role in the reduction of CV event incidence in patients treated with PCSK9 inhibitors.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticolesterolemiantes/uso terapêutico , Plaquetas/efeitos dos fármacos , Hipercolesterolemia/tratamento farmacológico , Inibidores de PCSK9 , Inibidores da Agregação Plaquetária/uso terapêutico , Agregação Plaquetária/efeitos dos fármacos , Inibidores de Serina Proteinase/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticolesterolemiantes/efeitos adversos , Biomarcadores/sangue , Plaquetas/metabolismo , Ligante de CD40/sangue , LDL-Colesterol/sangue , Feminino , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/diagnóstico , Hipercolesterolemia/enzimologia , Itália , Masculino , Pessoa de Meia-Idade , Selectina-P/sangue , Inibidores da Agregação Plaquetária/efeitos adversos , Fator Plaquetário 4/sangue , Pró-Proteína Convertase 9/sangue , Estudos Prospectivos , Inibidores de Serina Proteinase/efeitos adversos , Fatores de Tempo , Resultado do Tratamento
12.
J Am Heart Assoc ; 9(1): e014358, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31870234

RESUMO

Background Defects in the renal fatty acid ß-oxidation pathway have been implicated in the development of renal fibrosis. Our group has developed a therapeutic vaccine targeting PCSK9 (proprotein convertase subtilisin/kexin type 9), named PCSK9Qß-003. In this study, we investigated the potential effectiveness of the PCSK9Qß-003 vaccine on hypercholesterolemia with renal fibrosis. Methods and Results The low-density lipoprotein receptor+/- male mice fed with a high-cholesterol (1%) Western diet were randomly assigned into 4 groups: the sham group (or the control group), the phosphate-buffered saline group, the Qß virus-like particles group and the PCSK9Qß-003 vaccine group. Mice of the PCSK9Qß-003 group were injected with the PCSK9Qß-003 vaccine (100 µg/time) every 2 or 4 weeks. The mice were administered with either unilateral ureteral obstruction for 2 weeks or N-nitro-l-arginine methyl ester (50 mg/kg per day) for 6 weeks to establish a renal fibrosis model. Compared with the other 3 groups, the PCSK9Qß-003 vaccine obviously decreased total cholesterol and low-density lipoprotein cholesterol in low-density lipoprotein receptor+/- mice with hypercholesterolemia. Compared with the phosphate-buffered saline and Qß virus-like particles groups, the PCSK9Qß-003 vaccine improved hepatic steatosis and renal function. Histology analysis showed that the PCSK9Qß-003 vaccine significantly ameliorated renal lipid accumulation and renal fibrosis. Moreover, the PCSK9Qß-003 vaccine obviously upregulated the expression of low-density lipoprotein receptor, very-low-density lipoprotein receptor, sterol-regulatory element binding protein 2, and fatty acid ß-oxidation-related factors, and ameliorated renal fibrosis-related molecules both in the unilateral ureteral obstruction and N-nitro-l-arginine methyl ester models. Conclusions This study suggested that the PCSK9Qß-003 vaccine improved renal lipid accumulation and renal fibrosis by regulating fatty acid ß-oxidation, which may provide a promising method for treating hypercholesterolemia with renal fibrosis.


Assuntos
Ácidos Graxos/metabolismo , Hipercolesterolemia/terapia , Nefropatias/prevenção & controle , Rim/efeitos dos fármacos , Pró-Proteína Convertase 9/metabolismo , Vacinas de Subunidades Antigênicas/farmacologia , Animais , Modelos Animais de Doenças , Fígado Gorduroso/enzimologia , Fígado Gorduroso/etiologia , Fígado Gorduroso/prevenção & controle , Fibrose , Hipercolesterolemia/complicações , Hipercolesterolemia/enzimologia , Rim/metabolismo , Rim/patologia , Nefropatias/enzimologia , Nefropatias/etiologia , Nefropatias/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Receptores de LDL/deficiência , Receptores de LDL/genética , Obstrução Ureteral/complicações
13.
Molecules ; 24(22)2019 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-31731717

RESUMO

Statins are the most popular therapeutic drugs to lower plasma low density lipoprotein cholesterol (LDL-C) synthesis by competitively inhibiting hydroxyl-3-methyl-glutaryl-CoA (HMG-CoA) reductase and up-regulating the hepatic low density lipoprotein receptor (LDLR). However, the concomitant up-regulation of proprotein convertase subtilisin/kexin type 9 (PCSK9) by statin attenuates its cholesterol lowering efficacy. Lunasin, a soybean derived 43-amino acid polypeptide, has been previously shown to functionally enhance LDL uptake via down-regulating PCSK9 and up-regulating LDLR in hepatocytes and mice. Herein, we investigated the LDL-C lowering efficacy of simvastatin combined with lunasin. In HepG2 cells, after co-treatment with 1 µM simvastatin and 5 µM lunasin for 24 h, the up-regulation of PCSK9 by simvastatin was effectively counteracted by lunasin via down-regulating hepatocyte nuclear factor 1α (HNF-1α), and the functional LDL uptake was additively enhanced. Additionally, after combined therapy with simvastatin and lunasin for four weeks, ApoE-/- mice had significantly lower PCSK9 and higher LDLR levels in hepatic tissues and remarkably reduced plasma concentrations of total cholesterol (TC) and LDL-C, as compared to each monotherapy. Conclusively, lunasin significantly improved the LDL-C lowering efficacy of simvastatin by counteracting simvastatin induced elevation of PCSK9 in hepatocytes and ApoE-/- mice. Simvastatin combined with lunasin could be a novel regimen for hypercholesterolemia treatment.


Assuntos
LDL-Colesterol/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hepatócitos/enzimologia , Pró-Proteína Convertase 9/biossíntese , Sinvastatina/farmacologia , Proteínas de Soja/farmacologia , Animais , LDL-Colesterol/genética , Hepatócitos/patologia , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/enzimologia , Hipercolesterolemia/metabolismo , Camundongos , Camundongos Knockout para ApoE , Pró-Proteína Convertase 9/genética
14.
J Med Food ; 22(11): 1110-1117, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31613687

RESUMO

This study was carried out to investigate the effects of policosanol on high-fat and high-cholesterol diet-induced hypercholesterolemic rats to provide strong evidence in support of its hypocholesterolemic effect. The hypercholesterolemic rats showed elevations in liver weight, total triglycerides, total cholesterol, and low-density lipoprotein (LDL) cholesterol in serum; however, policosanol supplementation reduced these markers significantly. In addition, we found that policosanol supplementation stimulated an increase in fecal cholesterol and bile acid contents and deactivated 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase by AMP-activated protein kinase (AMPK) phosphorylation during high-fat and high-cholesterol-containing diet-induced development of hypercholesterolemia. Policosanol supplementation decreased ApoB levels and increased LDL-receptor expression, but it did not affect the hepatic ACAT2 level in livers from hypercholesterolemic rats. Moreover, supplementation with policosanol significantly decreased aortic wall thickness and levels of P-selectin and soluble vascular cell adhesion molecule (sVCAM-1) in serum. In conclusion, we suggest that policosanol supplementation induces antihypercholesterolemia by inhibiting cholesterol biosynthesis, LDL cholesterol uptake, and cholesterol excretion.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Anticolesterolemiantes/farmacologia , Colesterol/biossíntese , Álcoois Graxos/farmacologia , Hipercolesterolemia/tratamento farmacológico , Animais , Aorta , Apolipoproteína B-100/metabolismo , HDL-Colesterol/sangue , Dieta Hiperlipídica , Hidroximetilglutaril-CoA Redutases/metabolismo , Hipercolesterolemia/enzimologia , Fígado/metabolismo , Masculino , Selectina-P/sangue , Ratos , Ratos Sprague-Dawley , Receptores de LDL/metabolismo , Esterol O-Aciltransferase/metabolismo , Triglicerídeos/sangue , Molécula 1 de Adesão de Célula Vascular/sangue , Esterol O-Aciltransferase 2
15.
Toxicology ; 418: 11-21, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30825513

RESUMO

Clinical and animal studies have indicated that hypercholesterolemia has intrauterine developmental origin. Our previous studies showed that prenatal caffeine exposure (PCE) increased the serum total cholesterol (TCH) levels in adult offspring rats. This study investigates the intrauterine programming mechanism of PCE male offspring rats susceptible to adult hypercholesterolemia. Pregnant Wistar rats were intragastrically administered caffeine (30, 60, and 120 mg/kg∙d) from gestational days (GD) 9 to 20. Male offspring were sacrificed under anesthesia at GD20 and postnatal week (PW) 12, and the serum and liver were collected. The effects of caffeine (0-100 µM, 24 h) on the expression of cholesterol synthesis related genes and their epigenetic mechanisms were confirmed in L02 cells. The results showed that PCE induced higher levels of serum TCH, LDL-C and higher ratios of TCH/HDL-C and LDL-C/HDL-C. Furthermore, the high levels of histone acetylation (via H3K14ac and H3K27ac) and the expression of genes (Srebf2, Hmgcr, Hmgcs1) were responsible for cholesterol synthesis. The results of PCE offspring in utero and the data in vitro exhibited similar changes, and accompanied by the reduced expression of adenosine A2A receptor (A2AR), cyclic adenosine monophosphate (cAMP), sirtuin1 and protein kinase A (PKA). These changes could be reversed by A2AR agonist (CGS-21680), cAMP agonist (forskolin) and sirtuin1 agonist (resveratrol). Therefore, our results confirmed that caffeine could enhance histone acetylation and expression levels of genes responsible for cholesterol synthesis via inhibiting the A2AR/cAMP/PKA pathway and down-regulating sirtuin1, which continued throughout adulthood and elevated hepatic cholesterol synthesis and hypercholesterolemia in the male offspring rats.


Assuntos
Cafeína/toxicidade , Colesterol/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Hipercolesterolemia/induzido quimicamente , Fígado/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Receptor A2A de Adenosina/metabolismo , Sirtuína 1/metabolismo , Acetilação , Fatores Etários , Animais , Linhagem Celular , Colesterol/sangue , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Feminino , Regulação Enzimológica da Expressão Gênica , Idade Gestacional , Histonas/metabolismo , Hipercolesterolemia/sangue , Hipercolesterolemia/enzimologia , Hipercolesterolemia/genética , Fígado/enzimologia , Masculino , Exposição Materna/efeitos adversos , Gravidez , Ratos Wistar , Sistemas do Segundo Mensageiro
16.
Cardiovasc Res ; 115(3): 510-518, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30629143

RESUMO

Since the discovery of the role of proprotein convertase subtilisin kexin 9 (PCSK9) in the regulation of low-density lipoprotein cholesterol (LDL-C) in 2003, a paradigm shift in the treatment of hypercholesterolaemia has occurred. The PCSK9 secreted into the circulation is a major downregulator of the low-density lipoprotein receptor (LDLR) protein, as it chaperones it to endosomes/lysosomes for degradation. Humans with loss-of-function of PCSK9 exhibit exceedingly low levels of LDL-C and are protected from atherosclerosis. As a consequence, innovative strategies to modulate the levels of PCSK9 have been developed. Since 2015 inhibitory monoclonal antibodies (evolocumab and alirocumab) are commercially available. When subcutaneously injected every 2-4 weeks, they trigger a ∼60% LDL-C lowering and a 15% reduction in the risk of cardiovascular events. Another promising approach consists of a liver-targetable specific PCSK9 siRNA which results in ∼50-60% LDL-C lowering that lasts up to 6 months (Phases II-III clinical trials). Other strategies under consideration include: (i) antibodies targeting the C-terminal domain of PCSK9, thereby inhibiting the trafficking of PCSK9-LDLR to lysosomes; (ii) small molecules that either prevent PCSK9 binding to the LDLR, its trafficking to lysosomes or its secretion from cells; (iii) complete silencing of PCSK9 by CRISPR-Cas9 strategies; (iv) PCSK9 vaccines that inhibit the activity of circulating PCSK9. Time will tell whether other strategies can be as potent and safe as monoclonal antibodies to lower LDL-C levels.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticolesterolemiantes/uso terapêutico , Colesterol/sangue , Terapia Genética/métodos , Hipercolesterolemia/terapia , Pró-Proteína Convertase 9 , Inibidores de Serina Proteinase/uso terapêutico , Vacinas/uso terapêutico , Animais , Anticorpos Monoclonais/efeitos adversos , Anticolesterolemiantes/efeitos adversos , Biomarcadores/sangue , Inativação Gênica , Marcação de Genes , Terapia Genética/efeitos adversos , Humanos , Hipercolesterolemia/enzimologia , Hipercolesterolemia/genética , Hipercolesterolemia/imunologia , Inibidores de PCSK9 , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/imunologia , Pró-Proteína Convertase 9/metabolismo , Receptores de LDL/antagonistas & inibidores , Receptores de LDL/metabolismo , Inibidores de Serina Proteinase/efeitos adversos , Resultado do Tratamento , Vacinas/efeitos adversos
17.
Nat Biotechnol ; 36(8): 717-725, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29985478

RESUMO

Clinical translation of in vivo genome editing to treat human genetic diseases requires thorough preclinical studies in relevant animal models to assess safety and efficacy. A promising approach to treat hypercholesterolemia is inactivating the secreted protein PCSK9, an antagonist of the LDL receptor. Here we show that single infusions in six non-human primates of adeno-associated virus vector expressing an engineered meganuclease targeting PCSK9 results in dose-dependent disruption of PCSK9 in liver, as well as a stable reduction in circulating PCSK9 and serum cholesterol. Animals experienced transient, asymptomatic elevations of serum transaminases owing to the formation of T cells against the transgene product. Vector DNA and meganuclease expression declined rapidly, leaving stable populations of genome-edited hepatocytes. A second-generation PCSK9-specific meganuclease showed reduced off-target cleavage. These studies demonstrate efficient, physiologically relevant in vivo editing in non-human primates, and highlight safety considerations for clinical translation.


Assuntos
Colesterol/sangue , Desoxirribonucleases/metabolismo , Fígado/enzimologia , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Animais , Dependovirus/genética , Edição de Genes , Vetores Genéticos , Células HEK293 , Hepatócitos/metabolismo , Humanos , Hipercolesterolemia/enzimologia , Hipercolesterolemia/terapia , Células-Tronco Pluripotentes Induzidas/metabolismo , Macaca mulatta , Masculino , Camundongos , Camundongos Knockout , Receptores de LDL/antagonistas & inibidores
18.
Atherosclerosis ; 276: 28-38, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30025252

RESUMO

BACKGROUND AND AIMS: Induction of low-density lipoprotein receptor (LDLR) plays a significant role in reduction of plasma LDL-cholesterol (LDL-C) levels. Therefore, strategies that enhance the protein level of LDLR provide an attractive therapeutic target for the treatment of hypercholesterolemia. With this aim in mind, we concentrated our effort on studying the role of AKT kinase in regulation of LDLR levels and proceeded to examine the effect of MK-2206, an allosteric and highly selective AKT inhibitor, on LDLR expression. METHODS: Cultured human hepatoma cells were used to examine the effect of MK-2206 on the proteolytic processing of sterol regulatory element-binding protein-2 (SREBP-2), the expression of LDLR and cellular internalization of LDL. We also examined the effect of MK-2206 on LDLR levels in primary human hepatocytes. RESULTS: MK-2206 induced the proteolytic processing of SREBP-2, upregulated LDLR expression and stimulated LDL uptake. In contrast to statins, induction of LDLR levels by MK-2206 did not rely on 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) inhibition. As a result, cotreatment of cells with MK-2206 and mevastatin potentiated the impact of mevastatin on LDLR. Importantly, MK-2206 stimulated the expression of LDLR by primary human hepatocytes. CONCLUSIONS: MK-2206 is a novel LDLR-inducing agent that, either alone or in combination with statins, exerts a stimulating effect on cellular LDL uptake.


Assuntos
Anticolesterolemiantes/farmacologia , LDL-Colesterol/metabolismo , Hepatócitos/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/farmacologia , Hipercolesterolemia/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptores de LDL/metabolismo , Transporte Biológico , Células HeLa , Células Hep G2 , Hepatócitos/enzimologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hipercolesterolemia/enzimologia , Lovastatina/análogos & derivados , Lovastatina/farmacologia , Proteólise , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de LDL/genética , Transdução de Sinais/efeitos dos fármacos , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 38(8): 1723-1737, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29930001

RESUMO

Objective- Recent cohort studies have shown that nonalcoholic fatty liver disease (NAFLD), and especially nonalcoholic steatohepatitis (NASH), associate with atherosclerosis and cardiovascular disease, independently of conventional cardiometabolic risk factors. However, the mechanisms underlying the pathophysiological link between NAFLD/NASH and cardiovascular disease still remain unclear. Our previous studies have identified STK25 (serine/threonine protein kinase 25) as a critical determinant in ectopic lipid storage, meta-inflammation, and progression of NAFLD/NASH. The aim of this study was to assess whether STK25 is also one of the mediators in the complex molecular network controlling the cardiovascular disease risk. Approach and Results- Atherosclerosis was induced in Stk25 knockout and transgenic mice, and their wild-type littermates, by gene transfer of gain-of-function mutant of PCSK9 (proprotein convertase subtilisin/kexin type 9), which induces the downregulation of hepatic LDLR (low-density lipoprotein receptor), combined with an atherogenic western-type diet. We found that Stk25-/- mice displayed reduced atherosclerosis lesion area as well as decreased lipid accumulation, macrophage infiltration, collagen formation, and oxidative stress in aortic lesions compared with wild-type littermates, independently from alterations in dyslipidemia. Reciprocally, Stk25 transgenic mice presented aggravated plaque formation and maturation compared with wild-type littermates despite similar levels of fasting plasma cholesterol. We also found that STK25 protein was expressed in all layers of the aorta, suggesting a possible direct role in cardiovascular disease. Conclusions- This study provides the first evidence that STK25 plays a critical role in regulation of cardiovascular disease risk and suggests that pharmacological inhibition of STK25 function may provide new possibilities for prevention/treatment of atherosclerosis.


Assuntos
Aorta/enzimologia , Doenças da Aorta/enzimologia , Aterosclerose/enzimologia , Hipercolesterolemia/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Placa Aterosclerótica , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Dieta Hiperlipídica , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Transferência de Genes , Hipercolesterolemia/genética , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais
20.
J Cardiovasc Pharmacol Ther ; 23(4): 301-308, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29649884

RESUMO

In addition to monoclonal antibodies against proprotein convertase subtilisin-kexin type 9 (PCSK9), vaccines against PCSK9 and smaller molecule inhibitors as well as RNA inhibitors of PCSK9 production have been created. The monoclonal antibodies against PCSK9 and the PCSK9 RNA inhibitors can reduce low-density lipoproteins (LDLs) by over 50%, non-high-density lipoprotein (HDL) cholesterol and triglycerides, and increasing HDL. Although effective in several homozygous familial hypercholesterolemia patient types, PCSK9 inhibitors does not work in all patient types. Outcome trials show no effects on mortality but do show reductions in atherosclerotic events such as myocardial infarctions, strokes, and need for coronary revascularization. PCSK9 inhibitors have a very attractive safety profile with no significant elevations in measures of muscle or liver damage. The current and more advanced experimental agents all require subcutaneous dosing, and injection site reactions are among the most common adverse events. Therapy for the Food and Drug Administration (FDA) approved agents is markedly expensive, and this is the primary barrier to utilization. However, it is possible to identify patients with a number needed to treat to prevent an atherosclerotic event low enough to render it cost-effective and one such factor is whether or not you require a 50% reduction in LDL in order to achieve your LDL goal.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticolesterolemiantes/uso terapêutico , Doenças Cardiovasculares/prevenção & controle , LDL-Colesterol/sangue , Hipercolesterolemia/tratamento farmacológico , Inibidores de PCSK9 , Inibidores de Serina Proteinase/uso terapêutico , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais Humanizados , Anticolesterolemiantes/efeitos adversos , Biomarcadores/sangue , Doenças Cardiovasculares/diagnóstico , Regulação para Baixo , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/diagnóstico , Hipercolesterolemia/enzimologia , Pró-Proteína Convertase 9/imunologia , Fatores de Risco , Inibidores de Serina Proteinase/efeitos adversos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...