Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Acta Biomater ; 174: 281-296, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-37951519

RESUMO

RNA interference (RNAi) presents great potential against intractable liver diseases. However, the establishment of specific, efficient, and safe delivery systems targeting hepatocytes remains a great challenge. Herein, we described a promising hepatocytes-targeting system through integrating triantennary N-acetylgalactosamine (GalNAc)-engineered cell membrane with biodegradable mesoporous silica nanoparticles, which efficiently and safely delivered siRNA to hepatocytes and silenced the target PCSK9 gene expression for the treatment of non-alcoholic fatty liver disease. Having optimized the GalNAc-engineering strategy, insertion orders, and cell membrane source, we obtained the best-performing GalNAc-formulations allowing strong hepatocyte-specific internalization with reduced Kupffer cell capture, resulting in robust gene silencing and less hepatotoxicity when compared with cationic lipid-based GalNAc-formulations. Consequently, a durable reduction of lipid accumulation and damage was achieved by systemic administering siRNAs targeting PCSK9 in high-fat diet-fed mice, accompanied by displaying desirable safety profiles. Taken together, this GalNAc-engineering biomimetics represented versatile, efficient, and safe carriers for the development of hepatocyte-specific gene therapeutics, and prevention of metabolic diseases. STATEMENT OF SIGNIFICANCE: Compared to MSN@LP-GN3 (MC3-LNP), MSN@CM-GN3 exhibited strong hepatocyte targeting and Kupffer cell escaping, as well as good biocompatibility for safe and efficient siRNA delivery. Furthermore, siPCSK9 delivered by MSN@CM-GN3 reduced both serum and liver LDL-C, TG, TC levels and lipid droplets in HFD-induced mice, resulting in better performance than MSN/siPCSK9@LP-GN3 in terms of lipid-lowering effect and safety profiles. These findings indicated promising advantages of our biomimetic GN3-based systems for hepatocyte-specific gene delivery in chronic liver diseases. Our work addressed the challenges associated with the lower targeting efficiency of cell membrane-mimetic drug delivery systems and the immunogenicity of traditional GalNAc delivery systems. In conclusion, this study provided an effective and versatile approach for efficient and safe gene editing using ligand-integrated biomimetic nanoplatforms.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Pró-Proteína Convertase 9 , Camundongos , Animais , Interferência de RNA , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/farmacologia , Hepatopatia Gordurosa não Alcoólica/terapia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Biomimética , Hepatócitos/metabolismo , Fígado/metabolismo , RNA Interferente Pequeno/farmacologia , Lipídeos/farmacologia
2.
Naunyn Schmiedebergs Arch Pharmacol ; 397(3): 1727-1736, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37721554

RESUMO

Proprotein convertase subtilisin/kexin 9 (PCSK9) inhibitors have been shown to regulate lipid metabolism and reduce the risk of cardiovascular events. This study explores the effect and potential mechanism of PCSK9 inhibitors on lipid metabolism and coronary atherosclerosis. HepG2 cells were incubated with PCSK9 inhibitor. ApoE-/- mice were fed with a high fat to construct an atherosclerosis model, and then treated with PCSK9 inhibitor (8 mg/kg for 8 w). PCSK9 inhibitor downregulated microRNA (miRNA)-130a-3p expression in a dose-dependent manner. And, miR-130a-3p could bind directly to the 3' untranslated region (3'-UTR) region of LDLR to down-regulate LDLR expression in HepG2 cells, as confirmed by the luciferase reporter gene assay. In addition, miR-130a-3p overexpression significantly attenuated the promoting effect of PCSK9 inhibitor on LDLR and DiI-LDL uptake in HepG2 cells. More importantly, in vivo experiments confirmed that PCSK9 inhibitor could significantly inhibit miR-130a-3p levels and promote LDLR expression in liver tissues, thus regulating serum lipid profile and alleviating the progression of coronary atherosclerosis. PCSK9 inhibitor could moderately improve coronary atherosclerosis by regulating miR-130a-3p/LDLR axis, providing an exploitable strategy for the treatment of coronary atherosclerosis.


Assuntos
Aterosclerose , Doença da Artéria Coronariana , MicroRNAs , Camundongos , Animais , Humanos , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/farmacologia , Subtilisina/metabolismo , Subtilisina/farmacologia , Receptores de LDL/genética , Receptores de LDL/metabolismo , Camundongos Knockout para ApoE , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Pró-Proteína Convertases/farmacologia , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/metabolismo , Hepatócitos , Células Hep G2 , MicroRNAs/genética , MicroRNAs/metabolismo
3.
Int J Biol Sci ; 19(7): 2270-2288, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37151886

RESUMO

Hepatocellular carcinoma (HCC) is one of the most lethal malignant cancers across the world. It has a poor prognosis and lacks effective therapies, especially for patients with advanced-stage cancer, indicating an urgent need for new therapies and novel therapeutic targets. Here, by screening the U.S. Food and Drug Administration drug library against HCC cell lines, we identified that flubendazole, a traditional anthelmintic drug, could prominently suppress HCC cells in vivo and in vitro. RNA sequence analysis and cellular thermal shift assays showed that flubendazole reduced the expression of PCSK9 protein by direct targeting. The increased expression of PCSK9 in HCC tissues was demonstrated to be correlated with poor prognosis, and the inhibitory ability of flubendazole was selectively dependent on PCSK9 expression. PCSK9 knockdown abolished the antitumor effects of flubendazole in HCC. Mechanistically, flubendazole inhibited the Hedgehog signaling pathway induced by PCSK9, resulting in the downregulation of smoothened (SMO) and GLI Family Zinc Finger 1 (Gli1). Moreover, combining flubendazole with lenvatinib was found more effective than administering lenvatinib only for HCC treatment in vivo and in vitro. These findings reveal the therapeutic potential of flubendazole against HCC and provide clues on new repurposed drugs and targets for cancer treatment.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Pró-Proteína Convertase 9/farmacologia , Neoplasias Hepáticas/metabolismo , Reposicionamento de Medicamentos , Proliferação de Células , Linhagem Celular Tumoral , Proteínas Hedgehog/metabolismo
4.
Commun Biol ; 6(1): 452, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-37095219

RESUMO

Familial hypercholesterolemia (FH) patients suffer from excessively high levels of Low Density Lipoprotein Cholesterol (LDL-C), which can cause severe cardiovascular disease. Statins, bile acid sequestrants, PCSK9 inhibitors, and cholesterol absorption inhibitors are all inefficient at treating FH patients with homozygous LDLR gene mutations (hoFH). Drugs approved for hoFH treatment control lipoprotein production by regulating steady-state Apolipoprotein B (apoB) levels. Unfortunately, these drugs have side effects including accumulation of liver triglycerides, hepatic steatosis, and elevated liver enzyme levels. To identify safer compounds, we used an iPSC-derived hepatocyte platform to screen a structurally representative set of 10,000 small molecules from a proprietary library of 130,000 compounds. The screen revealed molecules that could reduce the secretion of apoB from cultured hepatocytes and from humanized livers in mice. These small molecules are highly effective, do not cause abnormal lipid accumulation, and share a chemical structure that is distinct from any known cholesterol lowering drug.


Assuntos
Anticolesterolemiantes , Hipercolesterolemia Familiar Homozigota , Hiperlipoproteinemia Tipo II , Células-Tronco Pluripotentes Induzidas , Humanos , Animais , Camundongos , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/farmacologia , Pró-Proteína Convertase 9/uso terapêutico , LDL-Colesterol , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Hiperlipoproteinemia Tipo II/genética , Anticolesterolemiantes/farmacologia , Apolipoproteínas B/genética , Apolipoproteínas B/farmacologia , Apolipoproteínas B/uso terapêutico , Hepatócitos
5.
J Biochem Mol Toxicol ; 37(6): e23346, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36988443

RESUMO

The protection of the blood-brain barrier (BBB) is the key direction to improving subarachnoid hemorrhage (SAH). Therefore, developing appropriate targeted drugs and therapies has become an urgent task for SAH patients. In this study, we investigated the role of dendritic cells (DCs) exosomal miR-3064-5p in repairing the BBB, providing a new basis for treating SAH. We detected the expression of miR-3064-5p in exosomes secreted by DCs (DCs-exo). An SAH rat model was constructed by intravascular perforation and characterized by HE and TUNEL-IF staining. We found that overexpression of miR-3064-5p in SAH rats suppressed iNOS expression and promoted the accumulation of tight junction proteins (Occludin, Claudin-3, ZO-1), whereas knockdown of miR-3064-5p exerted the opposite effect. Dual-LUC assay confirmed that miR-3064-5p could target and inhibit SIRT6. Knockdown of SIRT6 inhibited inflammatory cytokine (IL-6, IL-1ß, IFN-γ, and TGF-ß1) levels and apoptosis. The results of the co-IP assay showed that SIRT6 interacted with PCSK9, and knockdown of SIRT6 suppressed the expression of PCSK9. Moreover, DCs-exo reduced brain edema, upregulated miR-3064-5p and downregulated SIRT6 and PCSK9 in SAH rats. DCs-exo reduced inflammatory factors and increased tight junction proteins in SAH rats. Overexpression of miR-3064-5p enhanced the protective effect of DCs-exo, while overexpression of SIRT6 partially counteracted the effect. This study confirmed that DCs could secrete miR-3064-5p to ameliorate BBB damage after SAH. Mechanistically, miR-3064-5p alleviated BBB damage by targeting and inhibiting SIRT6/PCSk9 signaling pathway.


Assuntos
MicroRNAs , Sirtuínas , Hemorragia Subaracnóidea , Ratos , Animais , Barreira Hematoencefálica/metabolismo , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/metabolismo , Ratos Sprague-Dawley , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/farmacologia , Pró-Proteína Convertase 9/uso terapêutico , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Sirtuínas/genética , Sirtuínas/metabolismo , Células Dendríticas/metabolismo
6.
Arch Physiol Biochem ; 129(6): 1219-1228, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34270371

RESUMO

CONTEXT: The effect of Eucalyptus globulus in diabetic cardiac dysfunction and the possible mechanisms involved have not been explored. OBJECTIVE: To evaluate the effect of ethanol leaf extract of E. globulus (NEE) on the cardiac function of fructose/streptozotocin-induced diabetic rats. MATERIALS AND METHODS: Type-2 diabetes was induced in rats with 10% fructose feeding for 14 days and an intraperitoneal administration of 40 mg/kg streptozotocin. Diabetic animals were treated with NEE (100-400 mg/kg) or 5 mg/kg glibenclamide orally for 21 days. Biochemical assays, histopathological examination and analyses of PCSK9, Rho kinase and Cardiac troponin expression were performed. RESULTS: The untreated diabetic group showed decreased expression of the genes, oxidative stress, dyslipidemia, increased activities of creatine kinase MB and lactate dehydrogenase, reduced nitric oxide level, and depletion of cardiomyocytes, which were reversed in NEE treated groups. CONCLUSIONS: Eucalyptus globulus ameliorated diabetic cardiac dysfunction through increased PCSK9, Rho kinase and Cardiac troponin expression.


Assuntos
Diabetes Mellitus Experimental , Eucalyptus , Cardiopatias , Ratos , Animais , Eucalyptus/metabolismo , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/farmacologia , Estreptozocina/toxicidade , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Regulação para Cima , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/farmacologia , Estresse Oxidativo , Extratos Vegetais/farmacologia , Troponina/metabolismo , Troponina/farmacologia
7.
Cardiovasc Toxicol ; 22(12): 951-961, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36342620

RESUMO

This study investigates the effect and mechanism of proprotein convertase subtilisin/Kexin type 9 (PCSK9) on myocardial ischemia-reperfusion injury (MIRI) and provides a reference for clinical prevention and treatment of acute myocardial infarction (AMI). We established a rat model of myocardial ischemia/reperfusion (I/R) and AC16 hypoxia/reoxygenation (H/R) model. A total of 48 adult 7-week-old male Sprague-Dawley rats were randomly assigned to three groups (n = 16): control, I/R, and I/R + SiRNA. In I/R and I/R + siRNA groups, myocardial ischemia was induced via occlusion of the left anterior descending branch (LAD) of the coronary artery in rats in I/R group for 30 min and reperfused for 3 days. To assess the myocardial injury, the rats were subjected to an electrocardiogram (ECG), cardiac function tests, cardiac enzymes analysis, and 2,3,5-triphenyl tetrazolium chloride (TTC)/Evan Blue (EB) staining. Meanwhile, differences in the expression of autophagy-level proteins and Bcl-2/adenovirus E1B 19-kDa interacting protein (Bnip3) signaling-related proteins were determined by protein blotting. In vitro and in vivo experimental studies revealed that siRNA knockdown of PCSK9 reduced the expression of autophagic protein Beclin-1, light chain 3 (LC3) compared to normal control-treated cells and control-operated groups. Simultaneously, the expression of Bnip3 pathway protein was downregulated. Furthermore, the PCSK9-mediated small interfering RNA (siRNA) group injected into the left ventricular wall significantly improved cardiac function and myocardial infarct size. In ischemic/hypoxic circumstances, PCSK9 expression was dramatically increased. PCSK9 knockdown alleviated MIRI via Bnip3-mediated autophagic pathway, inhibited inflammatory response, reduced myocardial infarct size, and protected cardiac function.


Assuntos
Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Masculino , Ratos , Animais , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/farmacologia , Ratos Sprague-Dawley , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Apoptose , Autofagia , Infarto do Miocárdio/genética , Infarto do Miocárdio/prevenção & controle , Infarto do Miocárdio/metabolismo
8.
Microvasc Res ; 142: 104371, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35460665

RESUMO

OBJECTIVES: Autophagy is critical for myocardial ischemia-reperfusion (I/R) injury. However, there is still considerable debate over its protective and deleterious effects. The purpose of this study was to determine the involvement of the proprotein convertase subtilisin/Kexin type 9 (PCSK9) and its inhibitor in myocardial ischemia-reperfusion injury autophagy (MRI). METHODS: Nine groups of eighty rats were used: sham, I/R2 h, I/R4 h, I/R6 h, I/R8 h, I/R1 d, and I/R2 d. A 30-min coronary artery blockage was used to produce myocardial IR. The time required for reperfusion rose linearly with the time gradient, from 2 h to 2 days. Following the determination of the best reperfusion period, three groups were formed: sham, I/R, and I/R + P (PCSK9 inhibitor (evolocumab) 10 mg/kg diluted in 2 ml sterile injection water was administered subcutaneously 1 week and half an hour before to surgery. Each group's infarction area was determined by electrocardiography (ECG), cardiac function, and 2,3,5-triphenyltetrazolium chloride (TTC) /Evan Blue (EB) staining. To detect morphological alterations in myocardial cells in each group, hematoxylin and eosin (HE) staining was used. Meanwhile, western blotting, immunohistochemistry, and Masson trichrome staining were utilized to quantify myocardial fibrosis and PCSK9 and autophagy protein expression. RESULTS: The results indicated that PCSK9 expression levels increased significantly in MRI, as indicated by increased levels of the autophagy regulatory protein light chain 3 (LC3) and Beclin-1, which activated autophagy in cardiomyocytes, exacerbated myocardial injury, and increased the size of myocardial infarcts. Meanwhile, PCSK9 regulates mitophagy via the Bcl-2/adenovirus E1B 19-kDa interacting protein (BNIP3) pathway, which controls myocardial infarction MRI throughout. Additionally, the PCSK9 inhibitor significantly decreased autophagy, enhanced cardiac function, and reduced the extent of reperfusion injury, consequently reducing myocardial infarct size expansion. CONCLUSION: PCSK9 is upregulated in the myocardial ischemia-reperfusion injury hearts and regulates mitophagy via the BNIP3 pathway, which in turn contributes to reperfusion injury after myocardial infarction. PCSK9 inhibition protects against myocardial ischemia-reperfusion injury via suppressing autophagy.


Assuntos
Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Animais , Autofagia , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Pró-Proteína Convertase 9/farmacologia , Ratos , Ratos Sprague-Dawley
9.
Comput Intell Neurosci ; 2022: 4964262, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35378805

RESUMO

The aim of this study was to research the mechanism of proprotein convertase subtilisin-like kexin type 9 (PCSK9) inhibitor in neural protective effect on rat cerebral ischemic reperfusion injury (I/RI). The transient middle cerebral artery occlusion (tMCAO) model of rats was prepared by the suture method, and PCSK9 inhibitor was injected intraperitoneally immediately after I/R. The rats were scored for neurological deficits and the cerebral infarction volume was measured. The brain tissues were collected and western blot (WB) was used to detect the expression of PCSK9. The rat cortical neural stem cells were treated with oxygen glucose deprivation (OGD) to establish a cell model of ischemia/reperfusion. WB was used to detect the expression of PCSK9 and the apoptosis-related pathway proteins. After interfering with the expression of PCSK9 siRNA, the cell viability (cell counting kit-8 assay) and apoptosis (TUNEL staining, Annexin V/PI method) were detected, and the cell proliferation was detected by EdU staining and flow cytometry. The expression of PCSK9 in the brain tissue of the MCAO group was dramatically increased. PCSK9 inhibitor can improve neurobehavioral scores and reduce apoptosis and infarct volume. An OGD model of neural stem cells in vitro was constructed. Inhibiting PCSK9 with si-PCSK9 can increase cell viability, promote cell proliferation, and also reduce cell apoptosis. Inhibition of PCSK9 can decrease the cerebral infarct volume in rats with cerebral I/RI and improve the neural function. Mechanically, inhibition of PCSK9 can lead to the decrease of nerve cell apoptosis and promotion of cell proliferation.


Assuntos
Infarto da Artéria Cerebral Média , Pró-Proteína Convertase 9 , Animais , Apoptose , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/genética , Pró-Proteína Convertase 9/farmacologia , Ratos , Subtilisina/farmacologia
10.
Autoimmunity ; 55(4): 223-232, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35289693

RESUMO

Proprotein convertase subtilisin kexin type 9 (PCSK9) is a well-known proprotein convertase that influences foam cell formation and modulates atherosclerosis. Inclisiran is a novel chemosynthetic small interfering RNA that inhibits PCSK9 synthesis. This study aimed to explore the effect of inclisiran on oxidized low-density lipoprotein (ox-LDL)-induced foam cell formation in Raw264.7 macrophages and to investigate the underlying mechanisms. Raw264.7 cells were treated with ox-LDL to induce the formation of macrophage-derived foam cells. Oil Red O staining and high-performance liquid chromatography were performed to detect lipid accumulation and cholesterol levels. Dil-ox-LDL uptake assay, CCK-8, RT-qPCR, and Western blotting analysis were performed to examine ox-LDL uptake, cell viability, and expression of scavenger receptor-related factors. Inclisiran reduced lipid accumulation in ox-LDL-treated macrophages in a dose-dependent manner. Inclisiran significantly inhibited the levels of total cholesterol, free cholesterol, and cholesterol ester in the supernatant of Raw264.7 cells. Inclisiran reduced ox-LDL uptake and increased Raw264.7 cell viability. Meanwhile, inclisiran downregulated the expression of SR-A, LOX-1, and CD36 and upregulated SR-BI, ApoE, and ABCA1. Furthermore, inclisiran increased PPARγ activity and decreased NF-κB activity. An inhibitor of PPARγ (T0070907) reversed the beneficial effects of inclisiran on ox-LDL uptake, NF-κB inactivation, and cytokine expression. In conclusion, these data suggested that inclisiran inhibited the formation of macrophage-derived foam cells by activating the PPARγ pathway.HighlightsInclisiran reduces lipid accumulation in Raw264.7 cells;Inclisiran reduces ox-LDL uptake and increases Raw264.7 cell viability;Inclisiran inhibits foam cell formation by activating the PPARγ pathway.


Assuntos
Aterosclerose , Células Espumosas , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Colesterol/metabolismo , Colesterol/farmacologia , Células Espumosas/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Macrófagos/metabolismo , NF-kappa B/metabolismo , PPAR gama/metabolismo , PPAR gama/farmacologia , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/farmacologia , RNA Interferente Pequeno/genética
11.
Chem Biodivers ; 19(2): e202100675, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34866324

RESUMO

Hyperlipidemia (HLP) is a complex pathological condition results from lipid metabolism disorder, which is closely related to obesity, atherosclerosis and steatohepatitis. Emodin (EM), a natural anthraquinone, exhibits prominent hypolipidemic effects. However, its exact mechanism is still unclear. In this study, we successfully established hyperlipidemic zebrafish model induced by 4 % high-cholesterol diet (HCD) for 10 days and explored the anti-hyperlipidemic roles and underlying mechanisms of EM. The results indicated that EM attenuated the mortality and body mass index (BMI) of zebrafish with HLP, and ameliorated abnormal lipid levels involved in TC, TG, LDL-C and HDL-C levels. Besides, EM effectively reduced lipid accumulation in blood vessels and liver, alleviated hepatic histological damage, and inhibited vascular neutrophil inflammation. Finally, the mRNA expression of molecules related to lipid metabolism were studied by using real-time quantitative polymerase chain reaction (RT-qPCR) to investigated the underlying mechanism. Further results found that treatment with EM up-regulated AMPKα, LDLR, ABCA1 and ABCG1, and down-regulated SREBP-2, PCSK9 and HMGCR expression. In conclusion, EM showed a prominent mitigative effect on lipid metabolism disorder in zebrafish larvae with HCD-stimulated HLP, which was associated with the enhancement of LDL-C uptake and reverse cholesterol transport, and inhibition of cholesterol synthesis.


Assuntos
Emodina , Hiperlipidemias , Animais , Colesterol/metabolismo , Dieta Hiperlipídica/efeitos adversos , Emodina/farmacologia , Hiperlipidemias/tratamento farmacológico , Larva/metabolismo , Metabolismo dos Lipídeos , Fígado , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/farmacologia , Peixe-Zebra/metabolismo
12.
Arch Physiol Biochem ; 128(6): 1681-1686, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32619370

RESUMO

CONTEXT: Recent surveys have shown an association between proprotein convertase subtilisin/kexin type 9 (PCSK9) and oxidative stress. OBJECTIVE: In this investigation, the effect of evolocumab an anti-PCSK9 antibody was assessed against oxidative damage caused by hydrogen peroxide (H2O2) in human umbilical vein endothelial cells (HUVEC). MATERIAL AND METHODS: Viability of HUVEC was measured by MTT assay. Hydroperoxides and malondialdehyde (MDA) levels, and ferric reducing antioxidant power (FRAP) were detected in HUVEC that pre-treated with evolocumab and, then exposed to H2O2. RESULTS: Evolocumab significantly prevented the cytotoxicity induced by H2O2 at the concentrations of 5-100 µg/ml. Pre-treatment of HUVEC with evolocumab reduced hydroperoxides and MDA levels and also increased FRAP value in intra- and extra-cellular mediums compared with H2O2 stimulated cells at different concentration ranges. CONCLUSION: This study displayed anti-oxidative and cytoprotective activities of evolocumab against oxidative damage caused by H2O2 in endothelial cells.


Assuntos
Antioxidantes , Peróxido de Hidrogênio , Humanos , Peróxido de Hidrogênio/toxicidade , Antioxidantes/farmacologia , Estresse Oxidativo , Células Endoteliais da Veia Umbilical Humana , Malondialdeído , Subtilisinas/farmacologia , Pró-Proteína Convertase 9/farmacologia
13.
J Appl Biomed ; 20(4): 130-140, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36708718

RESUMO

Statins are primary drugs in the treatment of hyperlipidemias. This group of drugs is known for its beneficial pleiotropic effects (e.g., reduction of inflammatory state). However, a growing body of evidence suggests its diabetogenic properties. The culpable mechanism is not completely understood and might be related to the damage to pancreatic beta cells. Therefore, we conceived an in vitro study to explore the impact of atorvastatin on pancreatic islet beta cells line (1.1.E7). We evaluated the influence on viability, insulin, low-density lipoprotein (LDL) receptor, and proprotein convertase subtilisin/kexin type 9 (PCSK9) expression. A significant drop in mRNA for proinsulin and insulin expression was noted. Concurrently, a rise in LDL receptor at the protein level in cells exposed to atorvastatin was noted. Further experiments have shown that exenatide - belonging to glucagon-like peptide 1 (GLP-1) analogs that are used in a treatment of diabetes and known for its weight reducing properties - can alleviate the observed alterations. In this case, the mechanism of action of exenatide was dependent on a protein kinase A pathway. In conclusion, our results support the hypothesis that statin may have diabetogenic properties, which according to our study is related to reduced insulin expression. The concomitant use of GLP-1 receptor agonist seemed to successfully revert insulin expression.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Células Secretoras de Insulina , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/metabolismo , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/farmacologia , Exenatida/farmacologia , Exenatida/metabolismo , Secreção de Insulina , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/farmacologia , Atorvastatina/farmacologia , Atorvastatina/metabolismo , Insulina/metabolismo , Receptores de LDL/metabolismo
14.
Vasc Health Risk Manag ; 17: 527-542, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34526771

RESUMO

Lipoprotein(a) forms a subfraction of the lipid profile and is characterized by the addition of apolipprotein(a) (apo(a)) to apoB100 derived particles. Its levels are mostly genetically determined inversely related to the number of protein domain (kringle) repeats in apo(a). In epidemiological studies, it shows consistent association with cardiovascular disease (CVD) and most recently with extent of aortic stenosis. Issues with standardizing the measurement of Lp(a) are being resolved and consensus statements favor its measurement in patients at high risk of, or with family histories of CVD events. Major lipid-lowering therapies such as statin, fibrates, and ezetimibe have little effect on Lp(a) levels. Therapies such as niacin or cholesterol ester transfer protein (CETP) inhibitors lower Lp(a) as well as reducing other lipid-related risk factors but have failed to clearly reduce CVD events. Proprotein convertase subtilisin kexin-9 (PCSK9) inhibitors reduce cholesterol and Lp(a) as well as reducing CVD events. New antisense therapies specifically targeting apo(a) and hence Lp(a) have greater and more specific effects and will help clarify the extent to which intervention in Lp(a) levels will reduce CVD events.


Assuntos
Doenças Cardiovasculares/epidemiologia , Lipoproteína(a)/sangue , Pró-Proteína Convertase 9/farmacologia , Remoção de Componentes Sanguíneos , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/prevenção & controle , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases , Hiperlipidemias , Fatores de Risco
15.
Am J Cardiovasc Drugs ; 21(3): 255-265, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32929693

RESUMO

Lipoprotein(a) is a unique form of low-density lipoprotein. It is associated with a high incidence of premature atherosclerotic disease such as coronary artery disease, myocardial infarction, and stroke. Plasma levels of this lipoprotein and its activities are highly variable. This is because of a wide variability in the size of the apolipoprotein A moiety, which is determined by the number of repeats of cysteine-rich domains known as "kringles." Although the exact mechanism of lipoprotein(a)-induced atherogenicity is unknown, the lipoprotein has been found in the arterial walls of atherosclerotic plaques. It has been implicated in the formation of foam cells and lipid deposition in these plaques. Pharmacologic management of elevated levels of lipoprotein(a) with statins, fibrates, or bile acid sequestrants is ineffective. The newer and emerging lipid-lowering agents, such as the second-generation antisense oligonucleotides, cholesteryl ester transfer protein inhibitors, and proprotein convertase subtilisin/kexin type 9 inhibitors offer the most effective pharmacologic therapy.


Assuntos
Dislipidemias/tratamento farmacológico , Hipolipemiantes/uso terapêutico , Lipoproteína(a)/sangue , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Proteínas de Transferência de Ésteres de Colesterol/farmacologia , Humanos , Hipolipemiantes/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Inibidores de PCSK9 , Placa Aterosclerótica/patologia , Placa Aterosclerótica/prevenção & controle , Pró-Proteína Convertase 9/farmacologia , Fatores de Risco , Trombose/patologia , Trombose/prevenção & controle
16.
Biotechnol Lett ; 42(12): 2511-2522, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32676798

RESUMO

OBJECTIVES: To compare different approaches for the expression of an anti-PCSK9 biosimilar monoclonal antibody (mAb) in CHO cells using IRES-mediated tricistronic plasmid vectors combining different signal peptides, IRES elements and selection markers. RESULTS: Transient transfection indicated a similar level of secreted mAb 48 h post-transfection for all constructs. However, transfections carried out with circular plasmids showed a higher expression than with linearized plasmids. After two months under selection pressure, only part of the transfected pools recovered. The cultures co-transfected using two antibiotics as selection markers for double selection did not recover. Growth, metabolism and mAb production profiles of the only part of the transfected pools recovered resulting stable pools were compared and the stable pool transfected with circular L1-LC-IRES-H7-HC-IRES-NEO plasmid was chosen for further studies, due to higher cell growth and mAb production. Critical quality attributes of the protein A-purified mAb such as purity, homogeneity, binding affinity to PCSK9, and amino acid sequence were assessed confirming the success of the approach adopted in this study. CONCLUSIONS: The expression platform proposed showed to be efficient to produce a high-quality anti-PCSK9 mAb in stable CHO cell pools and provides benchmarks for fast production of different mAbs for characterization, formulation studies and pre-clinical investigation.


Assuntos
Anticorpos Monoclonais/imunologia , Medicamentos Biossimilares/farmacologia , Sítios Internos de Entrada Ribossomal/genética , Pró-Proteína Convertase 9/genética , Sequência de Aminoácidos/genética , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Células CHO , Cricetulus/genética , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Humanos , Sítios Internos de Entrada Ribossomal/efeitos dos fármacos , Plasmídeos/genética , Plasmídeos/farmacologia , Pró-Proteína Convertase 9/imunologia , Pró-Proteína Convertase 9/farmacologia , Transfecção
17.
Ann Med ; 52(5): 162-177, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32453609

RESUMO

Lipoprotein(a) (Lp(a)) was discovered more than 50 years ago, and a decade later, it was recognized as a risk factor for coronary artery disease. However, it has gained importance only in the past 10 years, with emergence of drugs that can effectively decrease its levels. Lp(a) is a low-density lipoprotein (LDL) with an added apolipoprotein(a) attached to the apolipoprotein B component via a disulphide bond. Circulating levels of Lp(a) are mainly genetically determined. Lp(a) has many functions, which include proatherosclerotic, prothrombotic and pro-inflammatory roles. Here, we review recent data on the role of Lp(a) in the atherosclerotic process, and treatment options for patients with cardiovascular diseases. Currently 'Proprotein convertase subtilisin/kexin type 9' (PCSK9) inhibitors that act through non-specific reduction of Lp(a) are the only drugs that have shown effectiveness in clinical trials, to provide reductions in cardiovascular morbidity and mortality. The effects of PCSK9 inhibitors are not purely through Lp(a) reduction, but also through LDL cholesterol reduction. Finally, we discuss new drugs on the horizon, and gene-based therapies that affect transcription and translation of apolipoprotein(a) mRNA. Clinical trials in patients with high Lp(a) and low LDL cholesterol might tell us whether Lp(a) lowering per se decreases cardiovascular morbidity and mortality.KEY MESSAGESLipoprotein(a) is an important risk factor in patients with cardiovascular diseases.Lipoprotein(a) has many functions, which include proatherosclerotic, prothrombotic and pro-inflammatory roles.Treatment options to lower lipoprotein(a) levels are currently scarce, but new drugs are on the horizon.


Assuntos
Aterosclerose/fisiopatologia , LDL-Colesterol/sangue , Lipoproteína(a)/sangue , Inibidores de PCSK9 , Anticolesterolemiantes/uso terapêutico , Aterosclerose/sangue , LDL-Colesterol/efeitos dos fármacos , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/tratamento farmacológico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Lipoproteína(a)/efeitos dos fármacos , Pró-Proteína Convertase 9/farmacologia , Medição de Risco
18.
BMC Med ; 17(1): 223, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31818299

RESUMO

BACKGROUND: Proprotein convertase subtilisin/kexin 9 (PCSK9) is an important regulator of low-density lipoprotein receptor (LDLR) and plasma levels of LDL cholesterol (LDL-C). PCSK9 inhibition is an efficient therapeutic approach for the treatment of dyslipidemia. We tested the therapeutic effect of a PCSK9 vaccine on dyslipidemia and atherosclerosis. METHODS: Lipid film hydration method was used to prepare negatively charged nanoliposomes as a vaccine delivery system. An immunogenic peptide called immunogenic fused PCSK9-tetanus (IFPT) was incorporated on the surface of nanoliposomes using DSPE-PEG-maleimide lipid (L-IFPT) and adsorbed to Alhydrogel® (L-IFPTA+). The prepared vaccine formulation (L-IFPTA+) and empty liposomes (negative control) were inoculated four times with bi-weekly intervals in C57BL/6 mice on the background of a severe atherogenic diet and poloxamer 407 (thrice weekly) injection. Antibody titers were evaluated 2 weeks after each vaccination and at the end of the study in vaccinated mice. Effects of anti-PCSK9 vaccination on plasma concentrations of PCSK9 and its interaction with LDLR were determined using ELISA. To evaluate the inflammatory response, interferon-gamma (IFN-γ)- and interleukin (IL)-10-producing splenic cells were assayed using ELISpot analysis. RESULTS: L-IFPTA+ vaccine induced a high IgG antibody response against PCSK9 peptide in the vaccinated hypercholesterolemic mice. L-IFPTA+-induced antibodies specifically targeted PCSK9 and decreased its plasma consecration by up to 58.5% (- 164.7 ± 9.6 ng/mL, p = 0.0001) compared with the control. PCSK9-LDLR binding assay showed that generated antibodies could inhibit PCSK9-LDLR interaction. The L-IFPTA+ vaccine reduced total cholesterol, LDL-C, and VLDL-C by up to 44.7%, 51.7%, and 19.2%, respectively, after the fourth vaccination booster, compared with the control group at week 8. Long-term studies of vaccinated hypercholesterolemic mice revealed that the L-IFPTA+ vaccine was able to induce a long-lasting humoral immune response against PCSK9 peptide, which was paralleled by a significant decrease of LDL-C by up to 42% over 16 weeks post-prime immunization compared to control. Splenocytes isolated from the vaccinated group showed increased IL-10-producing cells and decreased IFN-γ-producing cells when compared with control and naive mice, suggesting the immune safety of the vaccine. CONCLUSIONS: L-IFPTA+ vaccine could generate long-lasting, functional, and safe PCSK9-specific antibodies in C57BL/6 mice with severe atherosclerosis, which was accompanied by long-term therapeutic effect against hypercholesterolemia and atherosclerosis.


Assuntos
Aterosclerose/tratamento farmacológico , Pró-Proteína Convertase 9/uso terapêutico , Vacinação/métodos , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pró-Proteína Convertase 9/farmacologia
19.
Metabolism ; 94: 47-58, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30768966

RESUMO

BACKGROUND: Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a critical role in the cholesterol metabolism by negatively regulating the low-density lipoprotein receptor (LDLR). Lipid metabolic and ovarian disorders are the common clinical manifestation of polycystic ovary syndrome (PCOS). Here, we intended to elucidate the role of PCSK9 in the pathogenesis of PCOS conducted on a human population in case-control design and animal part in an interventional study. METHODS: We firstly investigated the serum levels of PCSK9 in 46 PCOS patients compared with 49 healthy women as controls, and then developed a PCOS mouse model induced by dehydroepiandrosterone (DHEA) and a high-fat diet (HFD) to determine the role of PCSK9 in abnormal lipid metabolism and ovarian dysfunction of PCOS in four groups (n = 40 per group): control, PCOS mice, PCOS plus alirocumab group, and PCOS plus vehicle group. The expression of PCSK9 in their serum, hepatic and ovarian tissues, serum lipid profiles and hormones were measured. Additionally, mRNA and protein expression levels of LDLR in hepatic and ovarian tissues, ovarian morphology and function were determined. Finally, we used freshly isolated theca-interstitial cells (TICs) and granulosa cells (GCs) from prepubertal normal mice to explore the effect of PCSK9 on LDL uptake of the cells. RESULTS: Serum PCSK9 concentrations were higher in PCOS patients than normal controls (P < 0.05). The PCOS model mice exhibited significantly increased serum levels of total cholesterol (TC), LDL-C and high-density lipoprotein-cholesterol (HDL-C; P < 0.001, P < 0.001, P = 0.0004, respectively). Moreover, the serum PCSK9 protein level was significantly increased in PCOS mice (P = 0.0002), which positively correlated with serum LDL-C (r = 0.5279, P = 0.0004) and TC (r = 0.4151, P = 0.035). In both liver and ovary of PCOS mice, PCSK9 mRNA and protein levels were significantly increased (P < 0.05), but LDLR levels were significantly decreased (P < 0.05). Furthermore, alirocumab inhibiting PCSK9 partly increased in LDLR expression in both liver and ovary in PCOS mice, also ameliorated the lipid metabolic disorders and pathological changes of ovarian morphology and function and serum reproductive hormones but not in the PCOS plus vehicle group. In vitro experiment, recombinant PCSK9 decreased LDL uptake in TICs and GCs (P < 0.001, P = 0.0011, respectively), which were partly reversed by alirocumab (P < 0.001, P = 0.012, respectively). CONCLUSION: Abnormal high expression of PCSK9 in the blood, liver and ovary may be involved in the pathogenesis of PCOS by affecting lipid metabolism and ovarian function, and the inhibition of PCSK9 may partly reverse the pathological changes of PCOS. Our research suggests a possibility of PCSK9 as a new attractive target for diagnosis and treatment of PCOS.


Assuntos
Metabolismo dos Lipídeos/efeitos dos fármacos , Ovário/efeitos dos fármacos , Síndrome do Ovário Policístico/sangue , Pró-Proteína Convertase 9/sangue , Animais , Estudos de Casos e Controles , Modelos Animais de Doenças , Feminino , Humanos , Fígado/enzimologia , Camundongos , Doenças Ovarianas/induzido quimicamente , Ovário/enzimologia , Síndrome do Ovário Policístico/etiologia , Síndrome do Ovário Policístico/metabolismo , Pró-Proteína Convertase 9/farmacologia
20.
Curr Opin Lipidol ; 29(6): 453-458, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30199407

RESUMO

PURPOSE OF REVIEW: Our primary objective is to review the most recent findings on the biology of PCSK9 and on two key aspects of PCSK9 inhibition beyond LDL control of great clinical relevance: the regulation of lipoprotein (a) circulating levels by PCSK9 inhibitors and the putative diabetogenic effects of these novel therapies. RECENT FINDINGS: The reality of two distinct extracellular and intracellular pathways by which PCSK9 decreases the abundance of the LDLR at the surface of many cell types, most importantly hepatocytes, has recently been established. In contrast, the exact mechanisms by which PCSK9 inhibitors lower the circulating levels of lipoprotein (a) remain a point of major dispute. Despite strong indications from genetic studies that PCSK9 inhibition should increase diabetes risk, no such effect has been observed in clinical trials, and in-vitro and in-vivo studies do not clarify this issue. SUMMARY: The trafficking pathways by which PCSK9 enhance LDLR degradation via the endolysosomal extracellular route or via the Golgi-lysosomal intracellular route remain to be fully elucidated. The mechanisms by which PCSK9 inhibitors reduce lipoprotein (a) also merit additional research efforts. The role of PCSK9 on glucose metabolism should likewise be studied in depth.


Assuntos
Anticolesterolemiantes/farmacologia , LDL-Colesterol/metabolismo , Pró-Proteína Convertase 9/farmacologia , Inibidores de Proteases/farmacologia , Animais , Anticolesterolemiantes/uso terapêutico , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Humanos , Pró-Proteína Convertase 9/uso terapêutico , Inibidores de Proteases/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA