Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 43(19): 3567-3581, 2023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-36977578

RESUMO

Metachromatic leukodystrophy (MLD) is a rare, inherited, demyelinating lysosomal storage disorder caused by mutations in the arylsulfatase-A gene (ARSA). In patients, levels of functional ARSA enzyme are diminished and lead to deleterious accumulation of sulfatides. Herein, we demonstrate that intravenous administration of HSC15/ARSA restored the endogenous murine biodistribution of the corresponding enzyme, and overexpression of ARSA corrected disease biomarkers and ameliorated motor deficits in Arsa KO mice of either sex. In treated Arsa KO mice, when compared with intravenously administered AAV9/ARSA, significant increases in brain ARSA activity, transcript levels, and vector genomes were observed with HSC15/ARSA Durability of transgene expression was established in neonate and adult mice out to 12 and 52 weeks, respectively. Levels and correlation between changes in biomarkers and ARSA activity required to achieve functional motor benefit was also defined. Finally, we demonstrated blood-nerve, blood-spinal and blood-brain barrier crossing as well as the presence of circulating ARSA enzyme activity in the serum of healthy nonhuman primates of either sex. Together, these findings support the use of intravenous delivery of HSC15/ARSA-mediated gene therapy for the treatment of MLD.SIGNIFICANCE STATEMENT Herein, we describe the method of gene therapy adeno-associated virus (AAV) capsid and route of administration selection leading to an efficacious gene therapy in a mouse model of metachromatic leukodystrophy. We demonstrate the therapeutic outcome of a new naturally derived clade F AAV capsid (AAVHSC15) in a disease model and the importance of triangulating multiple end points to increase the translation into higher species via ARSA enzyme activity and biodistribution profile (with a focus on the CNS) with that of a key clinically relevant biomarker.


Assuntos
Arilsulfatases , Terapia Genética , Leucodistrofia Metacromática , Animais , Camundongos , Macaca fascicularis , Arilsulfatases/genética , Camundongos Knockout , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/fisiopatologia , Leucodistrofia Metacromática/terapia , Modelos Animais de Doenças , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Encéfalo/enzimologia , Transtornos Motores/genética , Transtornos Motores/terapia , Administração Intravenosa , Biomarcadores/análise , Barreira Hematoencefálica , Masculino , Feminino , Humanos
2.
Mol Ther Methods Clin Dev ; 26: 224-238, 2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-35859693

RESUMO

Adeno-associated viruses derived from human hematopoietic stem cells (AAVHSCs) are naturally occurring AAVs. Fifteen AAVHSCs have demonstrated broad biodistribution while displaying differences in transduction. We examine the structure-function relationships of these natural amino acid variations on cellular binding. We demonstrate that AAVHSC16 is the only AAVHSC that does not preferentially bind to terminal galactose. AAVHSC16 contains two unique amino acids, 501I and 706C, compared with other AAVHSCs. Through mutagenesis, we determined that residue 501 contributes to the lack of galactose binding. Structural analysis revealed that residue 501 is in proximity to the galactose binding pocket, hence confirming its functional role in galactose binding. Biodistribution analysis of AAVHSC16 indicated significantly less liver tropism in mice and non-human primates compared with other clade F members, likely associated with overall binding differences observed in vitro. AAVHSC16 maintained robust tropism to other key tissues in the peripheral and central nervous systems after intravenous injection, including to the brain, heart, and gastrocnemius. Importantly, AAVHSC16 did not induce elevated liver enzyme levels in non-human primates after intravenous injection at high doses. The unique glycan binding and tropism of AAVHSC16 makes this naturally occurring capsid an attractive candidate for therapies requiring less liver tropism while maintaining broad biodistribution.

3.
Mol Ther Methods Clin Dev ; 17: 568-580, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32258219

RESUMO

Phenylketonuria is an inborn error of metabolism caused by loss of function of the liver-expressed enzyme phenylalanine hydroxylase and is characterized by elevated systemic phenylalanine levels that are neurotoxic. Current therapies do not address the underlying genetic disease or restore the natural metabolic pathway resulting in the conversion of phenylalanine to tyrosine. A family of hepatotropic clade F adeno-associated viruses (AAVs) was isolated from human CD34+ hematopoietic stem cells (HSCs) and one (AAVHSC15) was utilized to deliver a vector to correct the phenylketonuria phenotype in Pahenu2 mice. The AAVHSC15 vector containing a codon-optimized form of the human phenylalanine hydroxylase cDNA was administered as a single intravenous dose to Pahenu2 mice maintained on a phenylalanine-containing normal chow diet. Optimization of the transgene resulted in a vector that produced a sustained reduction in serum phenylalanine and normalized tyrosine levels for the lifespan of Pahenu2 mice. Brain levels of phenylalanine and the downstream serotonin metabolite 5-hydroxyindoleacetic acid were restored. In addition, the coat color of treated mice darkened following treatment, indicating restoration of the phenylalanine metabolic pathway. Taken together, these data support the potential of an AAVHSC15-based gene therapy as an investigational therapeutic for phenylketonuria patients.

4.
Proc Natl Acad Sci U S A ; 116(5): 1723-1732, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30559189

RESUMO

Lipoprotein lipase (LPL) is responsible for the intravascular processing of triglyceride-rich lipoproteins. The LPL within capillaries is bound to GPIHBP1, an endothelial cell protein with a three-fingered LU domain and an N-terminal intrinsically disordered acidic domain. Loss-of-function mutations in LPL or GPIHBP1 cause severe hypertriglyceridemia (chylomicronemia), but structures for LPL and GPIHBP1 have remained elusive. Inspired by our recent discovery that GPIHBP1's acidic domain preserves LPL structure and activity, we crystallized an LPL-GPIHBP1 complex and solved its structure. GPIHBP1's LU domain binds to LPL's C-terminal domain, largely by hydrophobic interactions. Analysis of electrostatic surfaces revealed that LPL contains a large basic patch spanning its N- and C-terminal domains. GPIHBP1's acidic domain was not defined in the electron density map but was positioned to interact with LPL's large basic patch, providing a likely explanation for how GPIHBP1 stabilizes LPL. The LPL-GPIHBP1 structure provides insights into mutations causing chylomicronemia.


Assuntos
Lipase Lipoproteica/metabolismo , Plasma/metabolismo , Receptores de Lipoproteínas/metabolismo , Triglicerídeos/sangue , Triglicerídeos/metabolismo , Animais , Células CHO , Capilares/metabolismo , Linhagem Celular , Cricetulus , Cristalografia por Raios X/métodos , Células Endoteliais/metabolismo , Humanos , Hidrólise , Hipertrigliceridemia/metabolismo
5.
J Invest Dermatol ; 135(12): 3060-3067, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26203639

RESUMO

Recessive dystrophic epidermolysis bullosa (RDEB) is an inherited disorder characterized by skin fragility, blistering, and multiple skin wounds with no currently approved or consistently effective treatment. It is due to mutations in the gene encoding type VII collagen (C7). Using recombinant human C7 (rhC7) purified from human dermal fibroblasts (FB-rhC7), we showed previously that intravenously injected rhC7 distributed to engrafted RDEB skin, incorporated into its dermal-epidermal junction (DEJ), and reversed the RDEB disease phenotype. Human dermal fibroblasts, however, are not used for commercial production of therapeutic proteins. Therefore, we generated rhC7 from Chinese hamster ovary (CHO) cells. The CHO-derived recombinant type VII collagen (CHO-rhC7), similar to FB-rhC7, was secreted as a correctly folded, disulfide-bonded, helical trimer resistant to protease degradation. CHO-rhC7 bound to fibronectin and promoted human keratinocyte migration in vitro. A single dose of CHO-rhC7, administered intravenously into new-born C7-null RDEB mice, incorporated into the DEJ of multiple skin sites, tongue and esophagus, restored anchoring fibrils, improved dermal-epidermal adherence, and increased the animals' life span. Furthermore, no circulating or tissue-bound anti-C7 antibodies were observed in the mice. These data demonstrate the efficacy of CHO-rhC7 in a preclinical murine model of RDEB.


Assuntos
Colágeno Tipo VII/uso terapêutico , Epidermólise Bolhosa Distrófica/tratamento farmacológico , Animais , Animais Recém-Nascidos , Células CHO , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo VII/administração & dosagem , Colágeno Tipo VII/química , Colágeno Tipo VII/imunologia , Cricetulus , Humanos , Injeções Intravenosas , Fenótipo , Proteínas Recombinantes/uso terapêutico
6.
J Biol Chem ; 290(25): 15496-15511, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25947382

RESUMO

Studies in human populations have shown a significant correlation between procollagen C-endopeptidase enhancer protein 2 (PCPE2) single nucleotide polymorphisms and plasma HDL cholesterol concentrations. PCPE2, a 52-kDa glycoprotein located in the extracellular matrix, enhances the cleavage of C-terminal procollagen by bone morphogenetic protein 1 (BMP1). Our studies here focused on investigating the basis for the elevated concentration of enlarged plasma HDL in PCPE2-deficient mice to determine whether they protected against diet-induced atherosclerosis. PCPE2-deficient mice were crossed with LDL receptor-deficient mice to obtain LDLr(-/-), PCPE2(-/-) mice, which had elevated HDL levels compared with LDLr(-/-) mice with similar LDL concentrations. We found that LDLr(-/-), PCPE2(-/-) mice had significantly more neutral lipid and CD68+ infiltration in the aortic root than LDLr(-/-) mice. Surprisingly, in light of their elevated HDL levels, the extent of aortic lipid deposition in LDLr(-/-), PCPE2(-/-) mice was similar to that reported for LDLr(-/-), apoA-I(-/-) mice, which lack any apoA-I/HDL. Furthermore, LDLr(-/-), PCPE2(-/-) mice had reduced HDL apoA-I fractional clearance and macrophage to fecal reverse cholesterol transport rates compared with LDLr(-/-) mice, despite a 2-fold increase in liver SR-BI expression. PCPE2 was shown to enhance SR-BI function by increasing the rate of HDL-associated cholesteryl ester uptake, possibly by optimizing SR-BI localization and/or conformation. We conclude that PCPE2 is atheroprotective and an important component of the reverse cholesterol transport HDL system.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Ésteres do Colesterol/metabolismo , Glicoproteínas/metabolismo , Lipoproteínas HDL/metabolismo , Receptores Depuradores Classe B/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Aorta/metabolismo , Aorta/patologia , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Aterosclerose/induzido quimicamente , Aterosclerose/genética , Aterosclerose/patologia , Transporte Biológico Ativo/genética , Células CHO , Ésteres do Colesterol/genética , Cricetulus , Glicoproteínas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipoproteínas HDL/genética , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Receptores de LDL/genética , Receptores de LDL/metabolismo , Receptores Depuradores Classe B/genética
7.
Xenobiotica ; 43(11): 963-72, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23631744

RESUMO

1. 5-(N-(4-((4-ethylbenzyl)thio)phenyl)sulfamoyl)-2-methyl benzoic acid (CP-778875), an agonist of the peroxisome proliferator-activated receptor alpha, has been evaluated in the clinic to treat dyslipidemia and type 2 diabetes mellitus. Herein, we investigate the effect of CP-778875 on the pharmacokinetics of atorvastatin acid and its metabolites in humans. 2. The study incorporated a fixed-sequence design conducted in two groups. Group A was designed to estimate the effects of multiple doses of CP-778875 on the single dose pharmacokinetics of atorvastatin. Subjects in group A (n = 26) received atorvastatin (40 mg) on days 1 and 9 and CP-778875 (1.0 mg QD) on days 5-12. Group B was designed to examine the effects of multiple doses of atorvastatin on the single dose pharmacokinetics of CP-778875. Subjects in group B (n = 29) received CP-778875 (0.3 mg) on days 1 and 9 and atorvastatin (40 mg QD) on days 5-12. 3. Mean maximum serum concentration (Cmax) and area under the curve of atorvastatin were increased by 45% and 20%, respectively, upon co-administration with CP-778875. Statistically significant increases in the systemic exposure of ortho- and para-hydroxyatorvastatin were also observed upon concomitant dosing with CP-778875. CP-778875 pharmacokinetics, however, were not impacted upon concomitant dosing with atorvastatin. 4. Inhibition of organic anion transporting polypeptide 1B1 by CP-778875 (IC50 = 2.14 ± 0.40 µM) could be the dominant cause of the pharmacokinetic interaction as CP-778875 did not exhibit significant inhibition of cytochrome P450 3A4/3A5, multidrug resistant protein 1 or breast cancer resistant protein, which are also involved in the hepatobiliary disposition of atorvastatin.


Assuntos
Benzoatos/farmacologia , Ácido Benzoico/farmacologia , Ácidos Heptanoicos/farmacologia , PPAR alfa/agonistas , Pirróis/farmacologia , Sulfonamidas/farmacologia , Animais , Atorvastatina , Benzoatos/química , Ácido Benzoico/química , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Interações Medicamentosas , Células HEK293 , Ácidos Heptanoicos/sangue , Ácidos Heptanoicos/farmacocinética , Humanos , Hidroxilação/efeitos dos fármacos , Células Madin Darby de Rim Canino , Proteínas de Membrana Transportadoras/metabolismo , Oxirredução/efeitos dos fármacos , Pirróis/sangue , Pirróis/farmacocinética , Sulfonamidas/química , Fatores de Tempo
8.
J Lipid Res ; 53(8): 1459-71, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22628619

RESUMO

Dysregulation of ceramide synthesis has been associated with metabolic disorders such as atherosclerosis and diabetes. We examined the changes in lipid homeostasis and gene expression in Huh7 hepatocytes when the synthesis of ceramide is perturbed by knocking down serine pal mitoyltransferase subunits 1, 2, and 3 (SPTLC123) or dihydroceramide desaturase 1 (DEGS1). Although knocking down all SPTLC subunits is necessary to reduce total ceramides significantly, depleting DEGS1 is sufficient to produce a similar outcome. Lipidomic analysis of distribution and speciation of multiple lipid classes indicates an increase in phospholipids in SPTLC123-silenced cells, whereas DEGS1 depletion leads to the accumulation of sphingolipid intermediates, free fatty acids, and diacylglycerol. When cer amide synthesis is disrupted, the transcriptional profiles indicate inhibition in biosynthetic processes, downregulation of genes involved in general endomembrane trafficking, and upregulation of endocytosis and endosomal recycling. SPTLC123 silencing strongly affects the expression of genes involved with lipid metabolism. Changes in amino acid, sugar, and nucleotide metabolism, as well as vesicle trafficking between organelles, are more prominent in DEGS1-silenced cells. These studies are the first to provide a direct and comprehensive understanding at the lipidomic and transcriptomic levels of how Huh7 hepatocytes respond to changes in the inhibition of ceramide synthesis.


Assuntos
Ceramidas/biossíntese , Ceramidas/metabolismo , Inativação Gênica , Homeostase/genética , Oxirredutases/genética , Serina C-Palmitoiltransferase/genética , Transcriptoma/genética , Linhagem Celular , Linhagem Celular Tumoral , Regulação Enzimológica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Oxirredutases/deficiência , Serina C-Palmitoiltransferase/deficiência , Transcrição Gênica/genética
9.
Toxicol Pathol ; 40(3): 435-47, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22301950

RESUMO

Weak peroxisome proliferator-activated receptor (PPAR) α agonists (fibrates) are used to treat dyslipidemia. This study compared the effects of the potent and selective PPARα agonist CP-778875 on peroxisomal ß-oxidation and cardiac and/or skeletal muscle injury with those of the weak PPARα agonist fenofibrate. We hypothesized that these muscle effects are mediated through the PPARα receptor, leading to increased ß-oxidation and consequent oxidative stress. CP-778875 (5 or 500 mg/kg) and fenofibrate (600 or 2,000→1,200 mg/kg, dose lowered because of intolerance) were administered to rats for six weeks. Standard end points, serum troponin I, heart and skeletal muscle ß-oxidation of palmitoyl-CoA, and acyl co-oxidase (AOX) mRNA were assessed. Both compounds dose-dependently increased the incidence and/or severity of cardiomyocyte degeneration and necrosis, heart weight, troponin I, and skeletal muscle degeneration. Mean heart ß-oxidation (3.4- to 5.1-fold control) and AOX mRNA (2.4- to 3.2-fold control) were increased with CP-778875 500 mg/kg and both doses of fenofibrate. ß-Oxidation of skeletal muscle was not affected by either compound; however, a significant increase in AOX mRNA (1.6- to 2.1-fold control) was observed with CP-778875 500 mg/kg and both doses of fenofibrate. Taken together, these findings were consistent with PPARα agonism and support the link between increased cardiac and skeletal muscle ß-oxidation and resultant muscle injury in the rat.


Assuntos
Fenofibrato/toxicidade , Coração/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , PPAR alfa/agonistas , Animais , Análise Química do Sangue , Peso Corporal , Relação Dose-Resposta a Droga , Feminino , Fenofibrato/farmacocinética , Fígado/química , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Masculino , Proteínas Musculares/metabolismo , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Doenças Musculares/induzido quimicamente , Doenças Musculares/metabolismo , Miocárdio/química , Miocárdio/metabolismo , Miocárdio/patologia , NAD/metabolismo , Peroxissomos/metabolismo , Ratos , Ratos Sprague-Dawley , Testes de Toxicidade , Troponina I/sangue , Troponina I/metabolismo
10.
Endocrinology ; 153(2): 721-31, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22186415

RESUMO

Very low-density lipoproteins (VLDL) are a class of large lipoprotein synthesized in the liver. The key function of VLDL, in vivo, is to carry triglyceride from the liver to adipose tissue. As a steroidogenic organ, the adrenal gland mainly uses lipoproteins as sources of cholesterol. Although VLDL receptors have been detected in the human adrenal, the function of VLDL in the adrenal gland remains unknown. Herein, we used primary cultures of human and bovine adrenal cells and the adrenocortical cell line H295R as models to determine the effects of VLDL on adrenal steroidogenesis. Our studies revealed that VLDL significantly increased aldosterone synthesis in all of the models tested. This increase was largely due to VLDL's stimulation of the expression of steroidogenic acute regulatory (StAR) protein and aldosterone synthase (CYP11B2). VLDL increased CYP11B2 mRNA expression in a concentration-dependent manner. Effects of VLDL on CYP11B2 transcript levels were not additive with angiotensin II or potassium but were additive with the cAMP pathway agonists ACTH and forskolin. Nifedipine completely inhibited the effects of VLDL on CYP11B2 mRNA, suggesting that calcium is the main signal transduction pathway used by VLDL in adrenal cells. Indeed, VLDL increased cytosolic free calcium levels. An in vivo study conducted in sucrose-fed rats showed a positive correlation between elevated triglyceride (VLDL) levels in plasma and CYP11B2 expression in the adrenal. In conclusion, we have shown that VLDL can stimulate aldosterone synthesis in adrenocortical cells by increasing StAR and CYP11B2 expression, an event likely mediated by a calcium-initiated signaling cascade.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/metabolismo , Aldosterona/biossíntese , Lipoproteínas VLDL/farmacologia , Córtex Suprarrenal/citologia , Animais , Cálcio/metabolismo , Bovinos , Linhagem Celular , Células Cultivadas , Colforsina/farmacologia , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Masculino , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Zona Glomerulosa/citologia , Zona Glomerulosa/efeitos dos fármacos , Zona Glomerulosa/metabolismo
11.
Am J Cardiol ; 108(3): 360-6, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21757044

RESUMO

Preß-1 high-density lipoprotein (HDL) plays a key role in reverse cholesterol transport by promoting cholesterol efflux. Our aims were (1) to test previous associations between preß-1 HDL and coronary heart disease (CHD) and (2) to investigate whether preß-1 HDL levels also are associated with risk of myocardial infarction (MI). Plasma preß-1 HDL was measured by an ultrafiltration-isotope dilution technique in 1,255 subjects recruited from the University of California-San Francisco Lipid and Cardiovascular Clinics and collaborating cardiologists. Preß-1 HDL was significantly and positively associated with CHD and MI even after adjustment for established risk factors. Inclusion of preß-1 HDL in a multivariable model for CHD led to a modest improvement in reclassification of subjects (net reclassification index 0.15, p = 0.01; integrated discrimination improvement 0.003, p = 0.2). In contrast, incorporation of preß-1 HDL into a risk model of MI alone significantly improved reclassification of subjects (net reclassification index 0.21, p = 0.008; integrated discrimination improvement 0.01, p = 0.02), suggesting that preß-1 HDL has more discriminatory power for MI than for CHD in our study population. In conclusion, these results confirm previous associations between preß-1 HDL and CHD in a large well-characterized clinical cohort. Also, this is the first study in which preß-1 HDL was identified as a novel and independent predictor of MI above and beyond traditional CHD risk factors.


Assuntos
Doença das Coronárias/sangue , Lipoproteínas de Alta Densidade Pré-beta/sangue , Infarto do Miocárdio/sangue , Adulto , Idoso , Estudos de Coortes , Doença das Coronárias/diagnóstico , Doença das Coronárias/epidemiologia , Feminino , Humanos , Funções Verossimilhança , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/epidemiologia , Valor Preditivo dos Testes , Fatores de Risco
12.
J Lipid Res ; 52(11): 1974-83, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21771977

RESUMO

Given the increased prevalence of cardiovascular disease in the world, the search for genetic variations that impact risk factors associated with the development of this disease continues. Multiple genetic association studies demonstrate that procollagen C-proteinase enhancer 2 (PCPE2) modulates HDL levels. Recent studies revealed an unexpected role for this protein in the proteolytic processing of pro-apolipoprotein (apo) A-I by enhancing the cleavage of the hexapeptide extension present at the N-terminus of apoA-I. To investigate the role of the PCPE2 protein in an in vivo model, PCPE2-deficient (PCPE2 KO) mice were examined, and a detailed characterization of plasma lipid profiles, apoA-I, HDL speciation, and function was done. Results of isoelectric focusing (IEF) electrophoresis together with the identification of the amino terminal peptides DEPQSQWDK and WHVWQQDEPQSQWDVK, representing mature apoA-I and pro-apoA-I, respectively, in serum from PCPE2 KO mice confirmed that PCPE2 has a role in apoA-I maturation. Lipid profiles showed a marked increase in plasma apoA-I and HDL-cholesterol (HDL-C) levels in PCPE2 KO mice compared with wild-type littermates, regardless of gender or diet. Changes in HDL particle size and electrophoretic mobility observed in PCPE2 KO mice suggest that the presence of pro-apoA-I impairs the maturation of HDL. ABCA1-dependent cholesterol efflux is defective in PCPE2 KO mice, suggesting that the functionality of HDL is altered.


Assuntos
Apolipoproteína A-I/sangue , Glicoproteínas/deficiência , Glicoproteínas/genética , Lipoproteínas HDL/sangue , Sequência de Aminoácidos , Animais , Apolipoproteína A-I/química , Colesterol/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Lipoproteínas HDL/química , Lipoproteínas HDL/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Tamanho da Partícula
13.
Endocrinology ; 152(3): 751-63, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21239432

RESUMO

Adrenal aldosterone production is regulated by physiological agonists at the level of early and late rate-limiting steps. Numerous studies have focused on the role of lipoproteins including high-density lipoprotein (HDL) as cholesterol providers in this process; however, recent research suggests that HDL can also act as a signaling molecule. Herein, we used the human H295R adrenocortical cell model to study the effects of HDL on adrenal aldosterone production and CYP11B2 expression. HDL, especially HDL2, stimulated aldosterone synthesis by increasing expression of CYP11B2. HDL treatment increased CYP11B2 mRNA in both a concentration- and time-dependent manner, with a maximal 19-fold increase (24 h, 250 µg/ml of HDL). Effects of HDL on CYP11B2 were not additive with natural agonists including angiotensin II or K(+). HDL effects were likely mediated by a calcium signaling cascade, because a calcium channel blocker and a calmodulin kinase inhibitor abolished the CYP11B2-stimulating effects. Of the two subfractions of HDL, HDL2 was more potent than HDL3 in stimulating aldosterone and CYP11B2. Further studies are needed to identify the active components of HDL, which regulate aldosterone production.


Assuntos
Córtex Suprarrenal/metabolismo , Aldosterona/metabolismo , HDL-Colesterol/farmacologia , Citocromo P-450 CYP11B2/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Córtex Suprarrenal/citologia , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Linhagem Celular , Citocromo P-450 CYP11B2/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo
14.
J Pharmacol Exp Ther ; 333(3): 844-53, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20190014

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are involved in the regulation of lipid and glucose metabolism. PPARgamma agonists improve insulin sensitivity and hyperglycemia and are effective in treating type 2 diabetes mellitus (T2DM), whereas PPARalpha agonists are used to treat dyslipidemia and atherosclerosis. The goal here was to examine the efficacy of a selective PPARalpha agonist {(S)-3-[3-(1-carboxy-1-methyl-ethoxy)-phenyl]-piperidine-1-carboxylic acid 4-trifluoromethyl-benzyl ester; CP-900691} on lipid, glycemic, and inflammation indices in 14 cynomolgus monkeys with spontaneous T2DM maintained on daily insulin therapy. Monkeys were dosed orally with either vehicle (n = 7) or CP-900691 (3 mg/kg, n = 7) daily for 6 weeks. CP-900691 treatment increased plasma high-density lipoprotein cholesterol (HDLC) (33 +/- 3 to 60 +/- 4 mg/dL, p < 0.001) and apolipoprotein A1 (96 +/- 5 to 157 +/- 5 mg/dL, p < 0.001), reduced plasma triglycerides (547 +/- 102 to 356 +/- 90 mg/dL, p < 0.01), and apolipoprotein B (62 +/- 3 to 45 +/- 3 mg/dL, p < 0.01), improved the lipoprotein index (HDL to non-HDLC ratio; 0.28 +/- 0.06 to 0.79 +/- 0.16, p < 0.001), decreased body weight (p < 0.01) and C-reactive protein (CRP) (1700 +/- 382 to 304 +/- 102 ng/ml, p < 0.01), and increased adiponectin (1697 +/- 542 to 4242 +/- 1070 ng/ml, p < 0.001) compared with baseline. CP-900691 treatment reduced exogenous insulin requirements by approximately 25% (p < 0.04) while lowering plasma fructosamine from 2.87 +/- 0.09 to 2.22 +/- 0.17 mM (p < 0.05), indicative of improved glycemic control. There were no changes in any of the aforementioned parameters in the vehicle group. Because low HDLC and high triglycerides are well established risk factors for cardiovascular disease, the marked improvements in these parameters, and in glycemic control, body weight, and CRP, suggest that CP-900691 may be of benefit in diabetic and obese or hyperlipidemic populations.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Hipolipemiantes , Lipídeos/sangue , Lipoproteínas/sangue , PPAR alfa/agonistas , Piperidinas/farmacologia , Propionatos/farmacologia , Adiponectina/sangue , Animais , Área Sob a Curva , Proteína C-Reativa/metabolismo , Diabetes Mellitus Tipo 2/genética , Relação Dose-Resposta a Droga , Teste de Tolerância a Glucose , Resistência à Insulina , Macaca fascicularis , Redução de Peso/efeitos dos fármacos
15.
J Lipid Res ; 51(5): 967-74, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19965592

RESUMO

The CETP inhibitor, torcetrapib, was prematurely terminated from phase 3 clinical trials due to an increase in cardiovascular and noncardiovascular mortality. Because nearly half of the latter deaths involved patients with infection, we have tested torcetrapib and other CETPIs to see if they interfere with lipopolysaccharide binding protein (LBP) or bactericidal/permeability increasing protein (BPI). No effect of these potent CETPIs on LPS binding to either protein was detected. Purified CETP itself bound weakly to LPS with a Kd >or= 25 microM compared with 0.8 and 0.5 nM for LBP and BPI, respectively, and this binding was not blocked by torcetrapib. In whole blood, LPS induced tumor necrosis factor-alpha normally in the presence of torcetrapib. Furthermore, LPS had no effect on CETP activity. We conclude that the sepsis-related mortality of the ILLUMINATE trial was unlikely due to a direct effect of torcetrapib on LBP or BPI function, nor to inhibition of an interaction of CETP with LPS. Instead, we speculate that the negative outcome seen for patients with infections might be related to the changes in plasma lipoprotein composition and metabolism, or alternatively to the known off-target effects of torcetrapib, such as aldosterone elevation, which may have aggravated the effects of sepsis.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Infecções/imunologia , Quinolinas/farmacologia , Proteínas de Fase Aguda/imunologia , Proteínas de Fase Aguda/metabolismo , Peptídeos Catiônicos Antimicrobianos/imunologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas Sanguíneas/imunologia , Proteínas Sanguíneas/metabolismo , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Humanos , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Ligação Proteica/efeitos dos fármacos , Ressonância de Plasmônio de Superfície
16.
J Lipid Res ; 50(7): 1330-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19237735

RESUMO

Given the increased prevalence of cardiovascular disease in the world, the search for genetic variations controlling the levels of risk factors associated with the development of the disease continues. Multiple genetic association studies suggest the involvement of procollagen C-proteinase enhancer-2 (PCPE2) in modulating HDL-C levels. Therefore biochemical and mechanistic studies were undertaken to determine whether there might be a basis for a role of PCPE2 in HDL biogenesis. Our studies indicate that PCPE2 accelerates the proteolytic processing of pro-apolipoprotein (apo) AI by enhancing the cleavage of the hexapeptide extension present at the N terminus of apoAI. Surface Plasmon Resonance and immunoprecipitation studies indicate that PCPE2 interacts with BMP-1 and pro-apoAI to form a ternary pro-apoAI/BMP-1/PCPE2 complex. The most favorable interaction among these proteins begins with the association of BMP-1 to pro-apoAI followed by the binding of PCPE2 which further stabilizes the complex. PCPE2 resides, along with apoAI, on the HDL fraction of lipoproteins in human plasma supporting a relationship between HDL and PCPE2. Taken together, the findings from our studies identify a new player in the regulation of apoAI post-translational processing and open a new avenue to the study of mechanisms involved in the regulation of apoAI synthesis, HDL levels, and potentially, cardiovascular disease.


Assuntos
Apolipoproteína A-I/metabolismo , Proteína Morfogenética Óssea 1/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Precursores de Proteínas/metabolismo , Animais , Apolipoproteína A-I/genética , Proteína Morfogenética Óssea 1/genética , Células CHO , Linhagem Celular Tumoral , HDL-Colesterol/sangue , Cricetinae , Cricetulus , Proteínas da Matriz Extracelular/genética , Glicoproteínas/genética , Humanos , Polimorfismo Genético , Precursores de Proteínas/genética
17.
Mol Pharmacol ; 75(2): 296-306, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18971326

RESUMO

The nuclear receptor peroxisome proliferator-activated receptor alpha (PPARalpha) is recognized as the primary target of the fibrate class of hypolipidemic drugs and mediates lipid lowering in part by activating a transcriptional cascade that induces genes involved in the catabolism of lipids. We report here the characterization of three novel PPARalpha agonists with therapeutic potential for treating dyslipidemia. These structurally related compounds display potent and selective binding to human PPARalpha and support robust recruitment of coactivator peptides in vitro. These compounds markedly potentiate chimeric transcription systems in cell-based assays and strikingly lower serum triglycerides in vivo. The transcription networks induced by these selective PPARalpha agonists were assessed by transcriptional profiling of mouse liver after short- and long-term treatment. The induction of several known PPARalpha target genes involved with fatty acid metabolism were observed, reflecting the expected pharmacology associated with PPARalpha activation. We also noted the down-regulation of a number of genes related to immune cell function, the acute phase response, and glucose metabolism, suggesting that these compounds may have anti-inflammatory action in the mammalian liver. Whereas these compounds are efficacious in acute preclinical models, extended safety studies and further clinical testing will be required before the full therapeutic promise of a selective PPARalpha agonist is realized.


Assuntos
Metabolismo dos Lipídeos/fisiologia , PPAR alfa/agonistas , Piperidinas/farmacologia , Animais , Perfilação da Expressão Gênica , Humanos , Hipolipemiantes/farmacologia , Metabolismo dos Lipídeos/genética , Fígado , Camundongos , Camundongos Transgênicos , PPAR alfa/genética , PPAR alfa/metabolismo , Piperidinas/uso terapêutico
18.
J Lipid Res ; 50(3): 546-555, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18845619

RESUMO

Lectin-like oxidized LDL (ox-LDL) receptor-1 (LOX-1) is a type-II transmembrane protein that belongs to the C-type lectin family of molecules. LOX-1 acts as a cell surface endocytosis receptor and mediates the recognition and internalization of ox-LDL by vascular endothelial cells. Internalization of ox-LDL by LOX-1 results in a number of pro-atherogenic cellular responses implicated in the development and progression of atherosclerosis. In an effort to elucidate the functional domains responsible for the binding of ox-LDL to the receptor, a series of site-directed mutants were designed using computer modeling and X-ray crystallography to study the functional role of the hydrophobic tunnel present in the LOX-1 receptor. The isoleucine residue (I(149)) sitting at the gate of the channel was replaced by phenylalanine, tyrosine, or glutamic acid to occlude the channel opening and restrict the docking of ligands to test its functional role in the binding of ox-LDL. The synthesis, intracellular processing, and cellular distribution of all mutants were identical to those of wild type, whereas there was a marked decrease in the ability of the mutants to bind ox-LDL. These studies suggest that the central hydrophobic tunnel that extends through the entire LOX-1 molecule is a key functional domain of the receptor and is critical for the recognition of modified LDL.


Assuntos
Lipoproteínas LDL/metabolismo , Receptores Depuradores Classe E/química , Receptores Depuradores Classe E/metabolismo , Substituição de Aminoácidos , Animais , Sítios de Ligação/genética , Células CHO , Clonagem Molecular , Cricetinae , Cricetulus , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Receptores Depuradores Classe E/genética , Transfecção
19.
Am J Cardiol ; 102(4): 434-9, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18678301

RESUMO

The weak peroxisome proliferator activated receptor-alpha (PPAR-alpha) agonists gemfibrozil and fenofibrate achieve only small increases in high-density lipoprotein (HDL) cholesterol. CP-778,875 is a more potent PPAR-alpha agonist developed to produce greater HDL cholesterol increases. This randomized, multicenter, double-blinded, placebo-controlled study evaluated the efficacy and safety of CP-778,875 in subjects with mixed dyslipidemia and type 2 diabetes. Eight-six subjects with low HDL cholesterol (< or =45 mg/dl for men and < or =55 mg/dl for women) and increased triglycerides (150 to 500 mg/dl) who had coexisting type 2 diabetes were randomized. Subjects received CP-778,875 doses of 0.5, 2, or 6 mg/day or placebo for 6 weeks. Any other lipid-altering therapy was stopped at screening. The primary end point was percent change in HDL cholesterol from baseline. The 2-mg/day dose of CP-778,875 significantly increased HDL cholesterol by 14%. The 2-mg dose also increased concentrations of apolipoprotein (apo) A-I, HDL(2) cholesterol, and HDL(3) cholesterol by 13%, 12%, and 19%, respectively. An unusual dose-response pattern was observed in that at 6 mg/day CP-778,875 only increased HDL cholesterol by 3% and decreased HDL(2) cholesterol by 24%. Fasting triglyceride levels were significantly decreased to a similar extent (26%) by all 3 doses of CP-778,875. CP-778,875 significantly increased homocysteine levels. There was no significant relation between change in homocysteine and change in apoA-I or HDL cholesterol. No subjects developed myopathy. In conclusion, CP-778,875 2 mg/day significantly increased HDL cholesterol, significantly lowered fasting triglycerides, and increased apoA-I and HDL subfractions. The clinical relevance of the increase in homocysteine levels is unknown.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Dislipidemias/tratamento farmacológico , Fenofibrato/uso terapêutico , Genfibrozila/uso terapêutico , Hipolipemiantes/uso terapêutico , PPAR alfa/agonistas , Adulto , Idoso , Apolipoproteína A-I/efeitos dos fármacos , Apolipoproteína B-100/efeitos dos fármacos , Proteína C-Reativa/efeitos dos fármacos , HDL-Colesterol/efeitos dos fármacos , Diabetes Mellitus Tipo 2/fisiopatologia , Método Duplo-Cego , Dislipidemias/fisiopatologia , Feminino , Fenofibrato/efeitos adversos , Genfibrozila/efeitos adversos , Homocisteína/efeitos dos fármacos , Humanos , Hipolipemiantes/efeitos adversos , Masculino , Pessoa de Meia-Idade
20.
Am J Pathol ; 169(3): 750-60, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936252

RESUMO

Peroxisomal proliferator-activated receptor (PPAR)-alpha is a ligand-activated transcriptional factor that regulates genes involved in lipid metabolism and energy homeostasis. PPAR-alpha activators, including fibrates, have been used to treat dyslipidemia for several decades. In contrast to their known effects on lipids, the pharmacological consequences of PPAR-alpha activation on cardiac metabolism and function are not well understood. Therefore, we evaluated the role that PPAR-alpha receptors play in the heart. Our studies demonstrate that activation of PPAR-alpha receptors using a selective PPAR-alpha ligand results in cardiomyocyte necrosis in mice. Studies in PPAR-alpha-deficient mice demonstrated that cardiomyocyte necrosis is a consequence of the activation of PPAR-alpha receptors. Cardiac fatty acyl-CoA oxidase mRNA levels increased at doses in which cardiac damage was observed and temporally preceded cardiomyocyte degeneration, suggesting that peroxisomal beta-oxidation correlates with the appearance of microscopic injury and cardiac injury biomarkers. Increased myocardial oxidative stress was evident in mice treated with the PPAR-alpha agonists coinciding with increased peroxisomal biomarkers of fatty acid oxidation. These findings suggest that activation of PPAR-alpha leads to increased cardiac fatty acid oxidation and subsequent accumulation of oxidative stress intermediates resulting in cardiomyocyte necrosis.


Assuntos
Cardiomiopatias/metabolismo , Metabolismo dos Lipídeos , Miócitos Cardíacos/metabolismo , PPAR alfa/metabolismo , Acil-CoA Oxidase/biossíntese , Acil-CoA Oxidase/genética , Animais , Anticolesterolemiantes/farmacologia , Biomarcadores/metabolismo , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/genética , Cardiomiopatias/patologia , Ácido Clofíbrico/farmacologia , Ácidos Graxos/genética , Ácidos Graxos/metabolismo , Traumatismos Cardíacos/induzido quimicamente , Traumatismos Cardíacos/genética , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Necrose/genética , Necrose/metabolismo , Necrose/patologia , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , PPAR alfa/agonistas , PPAR alfa/deficiência , Peroxissomos/metabolismo , Peroxissomos/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...