Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Mol Cell Biol ; 36(3): 507-21, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26598607

RESUMO

In humans and in mice, mutations in the Ostm1 gene cause the most severe form of osteopetrosis, a major bone disease, and neuronal degeneration, both of which are associated with early death. To gain insight into Ostm1 function, we first investigated by sequence and biochemical analysis an immature 34-kDa type I transmembrane Ostm1 protein with a unique cytosolic tail. Mature Ostm1 is posttranslationally processed and highly N-glycosylated and has an apparent mass of ∼60 kDa. Analysis the subcellular localization of Ostm1 showed that it is within the endoplasmic reticulum, trans-Golgi network, and endosomes/lysosomes. By a wide protein screen under physiologic conditions, several novel cytosolic Ostm1 partners were identified and validated, for which a direct interaction with the kinesin 5B heavy chains was demonstrated. These results determined that Ostm1 is part of a cytosolic scaffolding multiprotein complex, imparting an adaptor function to Ostm1. Moreover, we uncovered a role for the Ostm1/KIF5B complex in intracellular trafficking and dispersion of cargos from the endoplasmic reticulum to late endosomal/lysosomal subcellular compartments. These Ostm1 molecular and cellular functions could elucidate all of the pathophysiologic mechanisms underlying the wide phenotypic spectrum of Ostm1-deficient mice.


Assuntos
Citosol/metabolismo , Cinesinas/metabolismo , Proteínas de Membrana/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Caenorhabditis elegans , Linhagem Celular , Chlorocebus aethiops , Citosol/química , Drosophila melanogaster , Células HEK293 , Humanos , Cinesinas/química , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional , Transporte Proteico , Alinhamento de Sequência , Ubiquitina-Proteína Ligases/química , Peixe-Zebra
2.
J Biol Chem ; 290(30): 18609-20, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26085104

RESUMO

Amyloid precursor-like protein 2 (APLP2) and sortilin were reported to individually bind the proprotein convertase subtilisin/kexin type 9 (PCSK9) and regulate its activity on the low-density lipoprotein receptor (LDLR). The data presented herein demonstrate that mRNA knockdowns of APLP2, sortilin, or both in the human hepatocyte cell lines HepG2 and Huh7 do not affect the ability of extracellular PCSK9 to enhance the degradation of the LDLR. Furthermore, mice deficient in APLP2 or sortilin do not exhibit significant changes in liver LDLR or plasma total cholesterol levels. Moreover, cellular overexpression of one or both proteins does not alter PCSK9 secretion, or its activity on the LDLR. We conclude that PCSK9 enhances the degradation of the LDLR independently of either APLP2 or sortilin both ex vivo and in mice. Interestingly, when co-expressed with PCSK9, both APLP2 and sortilin were targeted for lysosomal degradation. Using chemiluminescence proximity and co-immunoprecipitation assays, as well as biosynthetic analysis, we discovered that sortilin binds and stabilizes APLP2, and hence could regulate its intracellular functions on other targets.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Pró-Proteína Convertases/metabolismo , Proteólise , Receptores de LDL/biossíntese , Serina Endopeptidases/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/biossíntese , Proteínas Adaptadoras de Transporte Vesicular/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Regulação da Expressão Gênica , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/genética , Receptores de LDL/genética , Serina Endopeptidases/genética
3.
PLoS One ; 8(3): e60095, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23544125

RESUMO

BACKGROUND: Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted inhibitor of the low-density lipoprotein (LDL) receptor and an important regulator of LDL metabolism. Elevated PCSK9 levels have been associated with cardiovascular risk. The purpose of this study was to investigate how ezetimibe and simvastatin, alone and in combination, affect PCSK9 circulating concentrations. METHODS: A single center, randomized, open-label parallel 3-group study in healthy men (mean age 32±9 years, body mass index 25.7±3.2 kg/m(2)) was performed. Each group of 24 subjects was treated for 14 days with either simvastatin 40 mg/d, ezetimibe 10 mg/d, or with both drugs. Multivariate analysis was used to investigate parameters influencing the change in PCSK9 concentrations under treatment. RESULTS: The baseline plasma PCSK9 concentrations in the total cohort were 52±20 ng/mL with no statistically significant differences between the groups. They were increased by 68±85% by simvastatin (P = 0.0014), by 10±38% by ezetimibe (P = 0.51) and by 67±91% by simvastatin plus ezetimibe (P = 0.0013). The increase in PCSK9 was inversely correlated with baseline PCSK9 concentrations (Spearman's R = -0.47, P<0.0001) and with the percent change in LDL cholesterol concentrations (Spearman's R = -0.30, P<0.01). In multivariate analyses, only baseline PCSK9 concentrations (ß = -1.68, t = -4.04, P<0.0001), percent change in LDL cholesterol from baseline (ß = 1.94, t = 2.52, P = 0.014), and treatment with simvastatin (P = 0.016), but not ezetimibe (P = 0.42), significantly influenced changes in PCSK9 levels. Parameters without effect on PCSK9 concentration changes were age, body mass index, body composition, thyroid function, kidney function, glucose metabolism parameters, adipokines, markers of cholesterol synthesis and absorption, and molecular markers of cholesterol metabolism. CONCLUSIONS: Ezetimibe does not increase circulating PCSK9 concentrations while simvastatin does. When added to simvastatin, ezetimibe does not cause an incremental increase in PCSK9 concentrations. Changes in PCSK9 concentrations are tightly regulated and mainly influenced by baseline PCSK9 levels and changes in LDL cholesterol. TRIAL REGISTRATION: ClinicalTrials.gov NCT00317993.


Assuntos
Azetidinas/farmacologia , Pró-Proteína Convertases/sangue , Serina Endopeptidases/sangue , Sinvastatina/farmacologia , Regulação para Cima/efeitos dos fármacos , Adulto , LDL-Colesterol/sangue , Ezetimiba , Humanos , Masculino , Pró-Proteína Convertase 9
4.
J Biol Chem ; 287(40): 33745-55, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22875854

RESUMO

The proprotein convertase PCSK9 is a major target in the treatment of hypercholesterolemia because of its ability bind the LDL receptor (LDLR) and enhance its degradation in endosomes/lysosomes. In the endoplasmic reticulum, the zymogen pro-PCSK9 is first autocatalytically cleaved at its internal Gln(152)↓, resulting in a secreted enzymatically inactive complex of PCSK9 with its inhibitory prosegment (prosegment·PCSK9), which is the active form of PCSK9 on the LDLR. We mutagenized the P1 cleavage site Gln(152) into all other residues except Cys and analyzed the expression and secretion of the resulting mutants. The data demonstrated the following. 1) The only P1 residues recognized by PCSK9 are Gln > Met > Ala > Ser > Thr ≈ Asn, revealing an unsuspected specificity. 2) All other mutations led to the formation of an unprocessed zymogen that acted as a dominant negative retaining the native protein in the endoplasmic reticulum. Analysis of a large panoply of known natural and artificial point mutants revealed that this general dominant negative observation applies to all PCSK9 mutations that result in the inability of the protein to exit the endoplasmic reticulum. Such a tight quality control property of the endoplasmic reticulum may lead to the development of specific PCSK9 small molecule inhibitors that block its autocatalytic processing. Finally, inspired by the most active gain-of-function mutant, D374Y, we evaluated the LDLR degradation activity of 18 Asp(374) variants of PCSK9. All Asp(374) mutations resulted in similar gain-of-function activity on the LDLR except that D374E was as active as native PCSK9, D374G was relatively less active, and D374N and D374P were completely inactive.


Assuntos
Mutação , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/fisiologia , Receptores de LDL/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/fisiologia , Processamento Alternativo , Catálise , Colesterol/metabolismo , LDL-Colesterol/metabolismo , Precursores Enzimáticos/química , Citometria de Fluxo , Genes Dominantes , Células HEK293 , Humanos , Hipercolesterolemia/genética , Modelos Biológicos , Pró-Proteína Convertase 9 , Receptores de LDL/química , Transfecção
5.
Atherosclerosis ; 223(2): 394-400, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22683120

RESUMO

BACKGROUND: The identification of mutations in PCSK9 (proprotein convertase subtilisin kexin9) in autosomal dominant hypercholesterolemia (ADH), has revealed the existence of a new player in cholesterol homeostasis. PCSK9 has been shown to enhance the degradation of the LDL receptor (LDLR) at the cell surface. Gain-of-function mutations of PCSK9 induce ADH and are very rare, but their identification is crucial in studying PCSK9's role in hypercholesterolemia, its detailed trafficking pathway and its impact on the LDLR. METHODS: In order to identify new mutations and understand the exact mechanisms of action of mutated PCSK9, PCSK9 was sequenced in 75 ADH patients with no mutations in the LDLR or APOB genes. Functional analyses in cell culture were conducted and the impact of novel PCSK9 mutations on the quantitative and qualitative features of lipoprotein particles and on the HDL-mediated cellular cholesterol efflux was studied. RESULTS: Among these 75 ADH probands with no mutations in the LDLR or APOB genes, four gain-of-function mutations of PCSK9 were identified, of which two were novel: the p.Leu108Arg and the p.Asp35Tyr substitutions. In vitro studies of their consequences on the activity of PCSK9 on cell surface levels of LDLR showed that the p.Leu108Arg mutation clearly results in a gain-of-function, while the p.Asp35Tyr mutation created a novel Tyr-sulfation site, which may enhance the intracellular activity of PCSK9. CONCLUSION: These data further contribute to the characterization of PCSK9 mutations and to better understanding of the impact on cholesterol metabolism of this new therapeutic target.


Assuntos
Hiperlipoproteinemia Tipo II/genética , Mutação , Pró-Proteína Convertases/genética , Serina Endopeptidases/genética , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Biomarcadores/sangue , HDL-Colesterol/sangue , Análise Mutacional de DNA , Feminino , Predisposição Genética para Doença , Células HEK293 , Células Hep G2 , Humanos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/metabolismo , Masculino , Paris , Linhagem , Fenótipo , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Transfecção
6.
Clin Chem ; 58(1): 183-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22065156

RESUMO

BACKGROUND: Although statin therapy is known to increase concentrations of PCSK9, whether this effect is related to the magnitude of LDL reduction is uncertain. This study was undertaken to understand the extent of this effect and examine the relationship between PCSK9 and LDL cholesterol (LDL-C) reduction. METHODS: We measured plasma PCSK9 concentrations by ELISA at baseline and at 1 year in 500 men and 500 women participating in the Justification for Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) trial that randomly allocated participants to rosuvastatin 20 mg daily or placebo. We also evaluated rs11591147, a single nucleotide polymorphism known to have an impact on plasma PCSK9 concentrations. RESULTS: At baseline, median (interquartile range) PCSK9 concentrations were higher in women [73 (62-90)] ng/mL than in men [69 (57-81) ng/mL] (P<0.005). During 1 year, there was no change in PCSK9 concentrations in the placebo arm, suggesting stability in time. In contrast, the rosuvastatin increased PCSK9 by 35% in women [101 (82-117) ng/mL] and 28% in men [89 (71-109) ng/mL] (P<0.0001). Among those allocated to rosuvastatin, greater reductions in LDL-C were associated with greater increases in PCSK9 on both absolute and relative scales (r=-0.15, P<0.0005). Furthermore PCSK9 (rs11591147) did not alter the magnitude of LDL-C reduction associated with rosuvastatin use. CONCLUSIONS: In this randomized trial, rosuvastatin increased plasma concentration of PCSK9 in proportion to the magnitude of LDL-C reduction; the LDL-C response to statin could not be inferred by PCSK9 concentrations.


Assuntos
LDL-Colesterol/sangue , Fluorbenzenos/uso terapêutico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Pirimidinas/uso terapêutico , Serina Endopeptidases/sangue , Sulfonamidas/uso terapêutico , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Rosuvastatina Cálcica
7.
Br J Nutr ; 107(5): 705-11, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21787450

RESUMO

The mechanisms implicated in the LDL-cholesterol (LDL-C)-lowering effects of the Mediterranean-type diet (MedDiet) are unknown. The present study assessed the impact of the MedDiet consumed under controlled feeding conditions, with and without weight loss, on surrogate markers of cholesterol absorption, synthesis and clearance using plasma phytosterols, lathosterol and proprotein convertase subtilisin/kexin-9 (PCSK9) concentrations, respectively, in men with the metabolic syndrome. The subjects' diet (n 19, 24-62 years) was first standardised to a baseline North American control diet (5 weeks) followed by a MedDiet (5 weeks), both under weight-maintaining isoenergetic feeding conditions. The participants then underwent a 20-week free-living energy restriction period (10 (sd 3) % reduction in body weight, P < 0·01), followed by the consumption of the MedDiet (5 weeks) under controlled isoenergetic feeding conditions. The LDL-C-lowering effect of the MedDiet in the absence of weight loss ( - 9·9 %) was accompanied by significant reductions in plasma PCSK9 concentrations ( - 11·7 %, P < 0·01) and in the phytosterol:cholesterol ratio ( - 9·7 %, P < 0·01) compared with the control diet. The addition of weight loss to the MedDiet had no further impact on plasma LDL-C concentrations and on these surrogate markers of LDL clearance and cholesterol absorption. The present results suggest that the MedDiet reduces plasma LDL-C concentrations primarily by increasing LDL clearance and reducing cholesterol absorption, with no synergistic effect of body weight loss in this process.


Assuntos
Colesterol/sangue , Dieta Mediterrânea , Hipercolesterolemia/prevenção & controle , Síndrome Metabólica/sangue , Síndrome Metabólica/dietoterapia , Redução de Peso , Adolescente , Adulto , Idoso , Biomarcadores/sangue , Índice de Massa Corporal , Colesterol/metabolismo , LDL-Colesterol/sangue , LDL-Colesterol/metabolismo , Dieta Redutora , Humanos , Hipercolesterolemia/etiologia , Isomerismo , Masculino , Síndrome Metabólica/metabolismo , Síndrome Metabólica/fisiopatologia , Pessoa de Meia-Idade , Sobrepeso/fisiopatologia , Fitosteróis/sangue , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/sangue , Quebeque , Serina Endopeptidases/sangue , Adulto Jovem
8.
J Biol Chem ; 286(26): 22785-94, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21550985

RESUMO

Bone morphogenetic protein 10 (BMP10) is a member of the TGF-ß superfamily and plays a critical role in heart development. In the postnatal heart, BMP10 is restricted to the right atrium. The inactive pro-BMP10 (∼60 kDa) is processed into active BMP10 (∼14 kDa) by an unknown protease. Proteolytic cleavage occurs at the RIRR(316)↓ site (human), suggesting the involvement of proprotein convertase(s) (PCs). In vitro digestion of a 12-mer peptide encompassing the predicted cleavage site with furin, PACE4, PC5/6, and PC7, showed that furin cleaves the best, whereas PC7 is inactive on this peptide. Ex vivo studies in COS-1 cells, a cell line lacking PC5/6, revealed efficient processing of pro-BMP10 by endogenous PCs other than PC5/6. The lack of processing of overexpressed pro-BMP10 in the furin- and PACE4-deficient cell line, CHO-FD11, and in furin-deficient LoVo cells, was restored by stable (CHO-FD11/Fur cells) or transient (LoVo cells) expression of furin. Use of cell-permeable and cell surface inhibitors suggested that endogenous PCs process pro-BMP10 mostly intracellularly, but also at the cell surface. Ex vivo experiments in mouse primary hepatocytes (wild type, PC5/6 knock-out, and furin knock-out) corroborated the above findings that pro-BMP10 is a substrate for endogenous furin. Western blot analyses of heart right atria extracts from wild type and PACE4 knock-out adult mice showed no significant difference in the processing of pro-BMP10, implying no in vivo role of PACE4. Overall, our in vitro, ex vivo, and in vivo data suggest that furin is the major convertase responsible for the generation of BMP10.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Furina/metabolismo , Ventrículos do Coração/enzimologia , Miocárdio/enzimologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Células COS , Chlorocebus aethiops , Furina/antagonistas & inibidores , Furina/genética , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Inibidores de Serina Proteinase/farmacologia
9.
Glycobiology ; 21(10): 1290-300, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21527438

RESUMO

The limited proteolysis of proteins by the proprotein convertases (PCs) is a common means of producing bioactive proteins or peptides. The PCs are associated with numerous human pathologies and their activity can be reduced through the use of specific inhibitors. Here, we demonstrate an alternative approach to inhibiting PCs by altering their N-glycosylation. Through site-directed mutagenesis, we show that the convertase PC1/3 contains two N-glycans, only one of which is critical for its prosegment cleavage. The exact structure of PC1/3 N-glycans does not significantly affect its zymogen activation within endocrine cells, but glycosylation of Asn(146) is critical. Processing of the PC1/3's substrate proopiomelanocortin (POMC) was used in a cell-based assay to screen a collection of 45 compounds structurally related to known glycosidase inhibitors. Two 5-thiomannose-containing disaccharide derivatives were discovered to block PC1/3 and POMC processing into the analgesic peptide ß-endorphin. These compounds also reduced the zymogen activation of the convertase subtilisin kexin isozyme-1 (SKI-1), blocked the processing of its substrate the sterol regulatory element-binding protein SREBP-2 and altered its glycosylation. Thus, modification of PC glycosylation may also be a means of blocking their activity, an effect which, in the case of SKI-1, may be of possible therapeutic use since SREBP-2 regulates sterol levels including cholesterol biosynthesis and its metabolism.


Assuntos
Precursores Enzimáticos/metabolismo , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Animais , Células CHO , Cricetinae , Ativação Enzimática , Glicosilação , Células HEK293 , Humanos , Pró-Opiomelanocortina/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Transporte Proteico , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , beta-Endorfina/metabolismo
10.
J Biol Chem ; 286(11): 9185-95, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21209099

RESUMO

Although the processing profile of the membrane-bound epidermal growth factor precursor (pro-EGF) is tissue-specific, it has not been investigated at the cellular level nor have the cognate proteinases been defined. Among the proprotein convertases (PCs), only the membrane-bound PC7, the most ancient and conserved basic amino acid-specific PC family member, induces the processing of pro-EGF into an ∼115-kDa transmembrane form (EGF-115) at an unusual VHPR(290)↓A motif. Because site-directed mutagenesis revealed that Arg(290) is not critical, the generation of EGF-115 by PC7 is likely indirect. This was confirmed by testing a wide range of protease inhibitors, which revealed that the production of EGF-115 is most probably achieved via the activation by PC7 of a latent serine and/or cysteine protease(s). EGF-115 is more abundant at the cell surface than pro-EGF and is associated with a stronger EGF receptor (EGFR) activation, as evidenced by higher levels of phosphorylated ERK1/2. This suggests that the generation of EGF-115 represents a regulatory mechanism of juxtacrine EGFR activation. Thus, PC7 is distinct from the other PCs in its ability to enhance the activation of the cell surface EGFR.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Subtilisinas/metabolismo , Animais , Células COS , Chlorocebus aethiops , Fator de Crescimento Epidérmico/genética , Receptores ErbB/genética , Células HEK293 , Humanos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Inibidores de Proteases/farmacologia , Precursores de Proteínas/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Subtilisinas/antagonistas & inibidores , Subtilisinas/genética
11.
J Lipid Res ; 52(3): 558-65, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21123766

RESUMO

Inhibition of cholesterol synthesis by 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CoAR) inhibitors has been associated with an increase in intestinal cholesterol absorption. This study examined how HMG-CoAR inhibition by atorvastatin modulates expression of key genes involved in intestinal cholesterol metabolism. A crossover study was conducted in which 22 hyperlipidemic men received atorvastatin, 40 mg/day, or placebo, each for 12 weeks. Gene expression was assessed by real-time PCR using duodenal biopsy samples obtained at the end of each phase of treatment. Treatment with atorvastatin was associated with a 76% reduction in lathosterol and significant increases in sitosterol (70%). Atorvastatin significantly increased intestinal mRNA levels of HMG-CoAR (59%), LDL receptor (LDLR) (52%), PCSK9 (187%), SREBP-2 (44%), and HNF-4α (13%). Furthermore, atorvastatin significantly increased intestinal mRNA levels of NPC1L1 by 19% and decreased mRNA levels of both ABCG5 and ABCG8 by 14%. Positive correlations were observed between changes in SREBP-2 and HNF-4α expression and concurrent changes in the intestinal mRNA levels of HMG-CoAR, LDLR, and NPC1L1. These results indicate that HMG-CoAR inhibition with atorvastatin stimulates the intestinal expression of NPC1L1, LDLR, and PCSK9; increases cholesterol absorption; and reduces expression of ABCG5/8; these effects are most likely mediated by upregulation of the transcription factors SREBP-2 and HNF-4α.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Ácidos Heptanoicos/farmacologia , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Intestinos/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Pirróis/farmacologia , Adulto , Atorvastatina , Colesterol/metabolismo , Esquema de Medicação , Duodeno/efeitos dos fármacos , Duodeno/metabolismo , Duodeno/patologia , Ácidos Heptanoicos/administração & dosagem , Humanos , Hiperlipidemias/patologia , Mucosa Intestinal/metabolismo , Intestinos/patologia , Masculino , Proteínas de Membrana Transportadoras , Pirróis/administração & dosagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
12.
J Biol Chem ; 286(4): 2728-38, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21075846

RESUMO

The zymogen activation mechanism and physiological functions of the most ancient and highly conserved basic amino acid-specific proprotein convertase 7 (PC7) are not known. Herein, we characterized the biosynthesis, subcellular localization, and trafficking of the membrane-bound full-length rat and human PC7. The prosegment of PC7 is primarily secreted alone as a non-inhibitory protein via the conventional, Golgi-dependent, secretory pathway. Mature PC7 is partially sulfated and thus reaches the cell surface via the conventional route. However, a fraction of PC7 reaches the cell surface through a brefeldin A- and COPII-independent unconventional secretory pathway. The latter trafficking may explain the rapid (<10 min) transit of a fraction of PC7 from the ER to the cell surface. Electron microscopy further confirmed the localization of PC7 to the cell surface of HEK293 cells. Within the cytosolic tail, only two cysteines (Cys(699) and Cys(704)) are palmitoylated, but this modification does not affect the choice of trafficking pathway. Swapping the transmembrane-cytosolic tail (TMCT) sequences of the convertases Furin and PC7 revealed that PC7(TMCT-Furin) is much more sulfated and hence traffics more efficiently through the conventional secretory pathway. In contrast, the Furin(TMCT-PC7) is no longer sulfated and thus reaches the cell surface by the unconventional pathway. Because trafficking of PC7(CT-Furin) and Furin(CT-PC7) resemble their wild type counterparts, we deduce that the transmembrane domain of PC7 regulates the sorting of PC7 toward the unconventional secretory pathway. In conclusion, PC7 is distinct from other proprotein convertases in its zymogen activation, subcellular localization, and trafficking.


Assuntos
Retículo Endoplasmático/enzimologia , Precursores Enzimáticos/metabolismo , Subtilisinas/metabolismo , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática/fisiologia , Precursores Enzimáticos/genética , Furina/genética , Furina/metabolismo , Células HEK293 , Humanos , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Ratos , Subtilisinas/genética
13.
J Biol Chem ; 285(52): 40965-78, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20937814

RESUMO

PCSK9, a target for the treatment of dyslipidemia, enhances the degradation of the LDL receptor (LDLR) in endosomes/lysosomes, up-regulating LDL-cholesterol levels. Whereas the targeting and degradation of the PCSK9-LDLR complex are under scrutiny, the roles of the N- and C-terminal domains of PCSK9 are unknown. Although autocatalytic zymogen processing of PCSK9 occurs at Gln(152)↓, here we show that human PCSK9 can be further cleaved in its N-terminal prosegment at Arg(46)↓ by an endogenous enzyme of insect High Five cells and by a cellular mammalian protease, yielding an ∼4-fold enhanced activity. Removal of the prosegment acidic stretch resulted in ∼3-fold higher binding to LDLR in vitro, in ≥4-fold increased activity on cellular LDLR, and faster cellular internalization in endosome/lysosome-like compartments. Finally, swapping the acidic stretch of PCSK9 with a similar one found in the glycosylphosphatidylinositol-anchored heparin-binding protein 1 does not impair PCSK9 autoprocessing, secretion, or activity and confirmed that the acidic stretch acts as an inhibitor of PCSK9 function. We also show that upon short exposure to pH values 6.5 to 5.5, an ∼2.5-fold increase in PCSK9 activity on total and cell surface LDLR occurs, and PCSK9 undergoes a second cleavage at Arg(248), generating a two-chain PCSK9-ΔN(248). At pH values below 5.5, PCSK9 dissociates from its prosegment and loses its activity. This pH-dependent activation of PCSK9 represents a novel pathway to further activate PCSK9 in acidic endosomes. These data enhance our understanding of the functional role of the acidic prosegment and on the effect of pH in the regulation of PCSK9 activity.


Assuntos
Endossomos/enzimologia , Peptídeos/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Animais , Endossomos/genética , Ativação Enzimática/fisiologia , Células HEK293 , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Lisossomos/enzimologia , Lisossomos/genética , Mariposas , Peptídeos/genética , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Ligação Proteica/fisiologia , Receptores de LDL/genética , Serina Endopeptidases/genética
14.
J Lipid Res ; 51(1): 140-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19571328

RESUMO

The proprotein convertase subtilisin kexin-9 (PCSK9) circulates in plasma as mature and furin-cleaved forms. A polyclonal antibody against human PCSK9 was used to develop an ELISA that measures total plasma PCSK9 rather than only the mature form. A cross-sectional study evaluated plasma levels in normal (n = 254) and hypercholesterolemic (n = 200) subjects treated or untreated with statins or statin plus ezetimibe. In controls, mean plasma PCSK9 (89.5 +/- 31.9 ng/ml) correlated positively with age, total cholesterol, LDL-cholesterol (LDL-C), triglycerides, and fasting glucose. Sequencing PCSK9 from individuals at the extremes of the normal PCSK9 distribution identified a new loss-of-function R434W variant associated with lower levels of circulating PCSK9 and LDL-C. In hypercholesterolemic subjects, PCSK9 levels were higher than in controls (99.3 +/- 31.7 ng/ml, P < 0.04) and increased in proportion to the statin dose, combined or not with ezetimibe. In treated patients (n = 139), those with familial hypercholesterolemia (FH; due to LDL receptor gene mutations) had higher PCSK9 values than non-FH (147.01 +/- 42.5 vs. 127.2 +/- 40.8 ng/ml, P < 0.005), but LDL-C reduction correlated positively with achieved plasma PCSK9 levels to a similar extent in both subsets (r = 0.316, P < 0.02 in FH and r = 0.275, P < 0.009 in non-FH). The detection of circulating PCSK9 in both FH and non-FH subjects means that this assay could be used to monitor response to therapy in a wide range of patients.


Assuntos
Monitoramento de Medicamentos/métodos , Ensaio de Imunoadsorção Enzimática , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hiperlipoproteinemia Tipo II/sangue , Serina Endopeptidases/sangue , Anticorpos/imunologia , Azetidinas/uso terapêutico , Glicemia/análise , Linhagem Celular , Colesterol/sangue , LDL-Colesterol/sangue , Estudos Transversais , Ezetimiba , Humanos , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Serina Endopeptidases/biossíntese , Serina Endopeptidases/imunologia , Triglicerídeos/sangue
15.
J Biol Chem ; 283(32): 21899-908, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18559340

RESUMO

Chikungunya virus (CHIKV) is a mosquito-transmitted Alphavirus that causes in humans an acute infection characterized by polyarthralgia, fever, myalgia, and headache. Since 2005 this virus has been responsible for an epidemic outbreak of unprecedented magnitude. By analogy with other alphaviruses, it is thought that cellular proteases are able to process the viral precursor protein E3E2 to produce the receptor-binding E2 protein that associates as a heterodimer with E1. Destabilization of the heterodimer by exposure to low pH allows viral fusion and infection. We show that among a large panel of proprotein convertases, membranous furin but also PC5B can process E3E2 from African CHIKV strains at the HRQRR(64) / ST site, whereas a CHIKV strain of Asian origin is cleaved at RRQRR(64) / SI by membranous and soluble furin, PC5A, PC5B, and PACE4 but not by PC7 or SKI-1. Using fluorogenic model peptides and recombinant convertases, we observed that the Asian strain E3E2 model peptide is cleaved most efficiently by furin and PC5A. This cleavage was also observed in CHIKV-infected cells and could be blocked by furin inhibitor decanoyl-RVKR-chloromethyl ketone. This inhibitor was compared with chloroquine for its ability to inhibit CHIKV spreading in myoblast cell cultures, a cell-type previously described as a natural target of this virus. Our results demonstrate the role of furin-like proteases in the processing of CHIKV particles and point out new approaches to inhibit this infection.


Assuntos
Infecções por Alphavirus/enzimologia , Infecções por Alphavirus/virologia , Vírus Chikungunya/fisiologia , Furina/antagonistas & inibidores , Furina/metabolismo , Mioblastos/virologia , Proteínas do Envelope Viral/metabolismo , Infecções por Alphavirus/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Antimaláricos/farmacologia , Linhagem Celular , Vírus Chikungunya/efeitos dos fármacos , Cloroquina/farmacologia , Humanos , Mioblastos/citologia , Pró-Proteína Convertases/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
16.
J Biol Chem ; 283(4): 2363-72, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18039658

RESUMO

The proprotein convertase PCSK9 gene is the third locus implicated in familial hypercholesterolemia, emphasizing its role in cardiovascular diseases. Loss of function mutations and gene disruption of PCSK9 resulted in a higher clearance of plasma low density lipoprotein cholesterol, likely due to a reduced degradation of the liver low density lipoprotein receptor (LDLR). In this study, we show that two of the closest family members to LDLR are also PCSK9 targets. These include the very low density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2) implicated in neuronal development and lipid metabolism. Our results show that wild type PCSK9 and more so its natural gain of function mutant D374Y can efficiently degrade the LDLR, VLDLR, and ApoER2 either following cellular co-expression or re-internalization of secreted human PCSK9. Such PCSK9-induced degradation does not require its catalytic activity. Membrane-bound PCSK9 chimeras enhanced the intracellular targeting of PCSK9 to late endosomes/lysosomes and resulted in a much more efficient degradation of the three receptors. We also demonstrate that the activity of PCSK9 and its binding affinity on VLDLR and ApoER2 does not depend on the presence of LDLR. Finally, in situ hybridization show close localization of PCSK9 mRNA expression to that of VLDLR in mouse postnatal day 1 cerebellum. Thus, this study demonstrates a more general effect of PCSK9 on the degradation of the LDLR family that emphasizes its major role in cholesterol and lipid homeostasis as well as brain development.


Assuntos
Hipercolesterolemia/metabolismo , Metabolismo dos Lipídeos/fisiologia , Receptores de LDL/metabolismo , Receptores de Lipoproteínas/metabolismo , Serina Endopeptidases/metabolismo , Substituição de Aminoácidos , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Células CHO , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Cricetinae , Cricetulus , Homeostase/fisiologia , Humanos , Hipercolesterolemia/complicações , Hipercolesterolemia/genética , Proteínas Relacionadas a Receptor de LDL , Fígado/metabolismo , Camundongos , Mutação de Sentido Incorreto , Células NIH 3T3 , Neurônios/metabolismo , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Transporte Proteico/fisiologia , Receptores de LDL/genética , Receptores de Lipoproteínas/genética , Serina Endopeptidases/genética
17.
J Biol Chem ; 282(37): 27402-27413, 2007 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-17623657

RESUMO

Subtilisin kexin isozyme-1 (SKI-1) represents the first mammalian member of secretory subtilisin-like processing enzymes that cleaves after nonbasic residues. It is synthesized as an inactive precursor that undergoes three sequential autocatalytic processing steps of its N-terminal prosegment and an ectodomain shedding at a site near the transmembrane domain. The various cellular functions of SKI-1 emphasize the need to understand the sites of its activation and shedding. We have previously shown that SKI-1 undergoes autocatalytic shedding at the sequence KHQKLL(953) downward arrow, resulting in a membrane-bound stump called St-1 (amino acids 954-1052). However, little is known about the cellular localization of SKI-1 or its shed forms. In the present study, we have further identified a smaller C-terminal fragment St-2 generated closer to the transmembrane domain. By sequencing and mass spectrometric analysis, the start site and the molecular mass of St-2 were determined. Site-directed mutagenesis revealed the critical amino acid involved in this novel process. Mutation of Met(990) to M990A, M990I, and M990L failed to generate St-2, suggesting an internal alternate translation event at Met(990), as confirmed by an in vitro transcription/translation assay. Confocal microscopy defined the subcellular localization of SKI-1 and its fragments. The data show that most of membrane-bound SKI-1 and its stumps St-1 and St-2 localize to the Golgi and can enter the endosomal/lysosomal compartments but do not sort to the cell surface. Deletion studies showed that the transmembrane domain of SKI-1 determines its trafficking. Finally, rSt-1 and rSt-2 seem to affect the processing of ATF6 by SKI-1, but cellular stress does not regulate the production of St-2.


Assuntos
Pró-Proteína Convertases/genética , Biossíntese de Proteínas , Serina Endopeptidases/genética , Fator 6 Ativador da Transcrição/biossíntese , Sequência de Aminoácidos , Células Cultivadas , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Pró-Proteína Convertases/análise , Pró-Proteína Convertases/química , Serina Endopeptidases/análise , Serina Endopeptidases/química
18.
Traffic ; 8(6): 718-32, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17461796

RESUMO

Mutations in the proprotein convertase PCSK9 gene are associated with autosomal dominant familial hyper- or hypocholesterolemia. These phenotypes are caused by a gain or loss of function of proprotein convertase subtilisin kexin 9 (PCSK9) to elicit the degradation of the low-density lipoprotein receptor (LDLR) protein. Herein, we asked whether the subcellular localization of wild-type PCSK9 or mutants of PCSK9 and the LDLR would provide insight into the mechanism of PCSK9-dependent LDLR degradation. We show that the LDLR is the dominant partner in regulating the cellular trafficking of PCSK9. In cells lacking the LDLR, PCSK9 localized in the endoplasmic reticulum (ER). In cells expressing the LDLR, PCSK9 sorted to post-ER compartments (i.e. endosomes in cell lines and Golgi apparatus in primary hepatocytes), where it colocalized with the LDLR. In cell lines, PCSK9 also colocalized with the LDLR at the cell surface, requiring the presence of the C-terminal Cys/His-rich domain of PCSK9. We provide evidence that PCSK9 promotes the degradation of the LDLR by an endocytic mechanism, as small interfering RNA-mediated knockdown of the clathrin heavy chain reduced the functional activity of PCSK9. We also compared the subcellular localization of natural mutants of PCSK9 with that of the wild-type enzyme in human hepatic (HuH7) cells. Whereas the mutants associated with hypercholesterolemia (S127R, F216L and R218S) localized to endosomes/lysosomes, those associated with hypocholesterolemia did not reach this compartment. We conclude that the sorting of PCSK9 to the cell surface and endosomes is required for PCSK9 to fully promote LDLR degradation and that retention in the ER prevents this activity. Mutations that affect this transport can lead to hyper- or hypocholesterolemia.


Assuntos
Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Hipercolesterolemia/metabolismo , Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Complexo de Golgi/metabolismo , Humanos , Hipercolesterolemia/enzimologia , Hipercolesterolemia/genética , Lisossomos/metabolismo , Mutação , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Estrutura Terciária de Proteína , Transporte Proteico , RNA Interferente Pequeno/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/genética
19.
J Biol Chem ; 281(41): 30561-72, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16912035

RESUMO

PCSK9 is the ninth member of the proprotein convertase (PC) family. Some of its natural mutations have been genetically associated with the development of a dominant form of familial hyper- or hypocholesterolemia. The exact mechanism of action of PCSK9 is not clear, although it is known to enhance the intracellular degradation of the low density lipoprotein (LDL) receptor in acidic compartments, likely the endosomes/lysosomes. We analyzed the post-translational modifications of PCSK9 and show that it is sulfated within its prosegment at Tyr38. We also examined the susceptibility of PCSK9 to proteolytic cleavage by the other members of the PC family. The data show that the natural gain-of-function mutations R218S, F216L, and D374Y associated with hypercholesterolemia result in total or partial loss of furin/PC5/6A processing at the motif RFHR218 downward arrow. In contrast, the loss-of-function mutations A443T and C679X lead either to the lack of trans-Golgi network/recycling endosome localization and an enhanced susceptibility to furin cleavage (A443T) or to the inability of PCSK9 to exit the endoplasmic reticulum (C679X). Furthermore, we report the presence of both native and furin-like cleaved forms of PCSK9 in circulating human plasma. Thus, we propose that PCSK9 levels are finely regulated by the basic amino acid convertases furin and PC5/6A. The latter may reduce the lifetime of this proteinase and its ability to degrade the cell-surface LDL receptor, thereby regulating the levels of circulating LDL cholesterol.


Assuntos
Furina/química , Mutação , Pró-Proteína Convertase 5/química , Processamento de Proteína Pós-Traducional , Serina Endopeptidases/genética , Serina Endopeptidases/fisiologia , Sequência de Aminoácidos , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Feminino , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Dados de Sequência Molecular , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Serina Endopeptidases/metabolismo , Rede trans-Golgi/metabolismo
20.
FEBS Lett ; 579(25): 5621-5, 2005 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-16213495

RESUMO

Processing of precursor proteins by the proprotein convertases is thought to occur mainly in the trans-Golgi network or post-Golgi compartments. Such cleavage is inhibited by the prosegment of the convertases. During our studies of the use of the inhibitory prosegment of PC1, we noticed that a construct containing the prosegment fused to the C-terminal secretory granule sorting domain was cleaved in the endoplasmic reticulum (ER) at a pair of basic residues, best recognized by furin and PC7. This was further confirmed when this construct was fused at the C-terminus with a KDEL ER-retention signal. This suggests that the convertases could cleave some substrates within the ER, possibly by displacing the inhibitory prosegment associated with them.


Assuntos
Retículo Endoplasmático/enzimologia , Pró-Proteína Convertase 1/metabolismo , Rede trans-Golgi/enzimologia , Sequência de Aminoácidos , Animais , Inibidores Enzimáticos/metabolismo , Furina/genética , Furina/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Mutação , Pró-Proteína Convertase 1/antagonistas & inibidores , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Vesículas Secretórias/enzimologia , Subtilisinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...