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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 290(30): 18609-20, 2015 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-26085104

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proproteína Convertasas/metabolismo , Proteolisis , Receptores de LDL/biosíntesis , Serina Endopeptidasas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/biosíntesis , Proteínas Adaptadoras del Transporte Vesicular/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Regulación de la Expresión Génica , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proproteína Convertasa 9 , Proproteína Convertasas/genética , Receptores de LDL/genética , Serina Endopeptidasas/genética
2.
J Biol Chem ; 287(40): 33745-55, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22875854

RESUMEN

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.


Asunto(s)
Mutación , Proproteína Convertasas/genética , Proproteína Convertasas/fisiología , Receptores de LDL/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/fisiología , Empalme Alternativo , Catálisis , Colesterol/metabolismo , LDL-Colesterol/metabolismo , Precursores Enzimáticos/química , Citometría de Flujo , Genes Dominantes , Células HEK293 , Humanos , Hipercolesterolemia/genética , Modelos Biológicos , Proproteína Convertasa 9 , Receptores de LDL/química , Transfección
3.
Nat Genet ; 34(2): 154-6, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12730697

RESUMEN

Autosomal dominant hypercholesterolemia (ADH; OMIM144400), a risk factor for coronary heart disease, is characterized by an increase in low-density lipoprotein cholesterol levels that is associated with mutations in the genes LDLR (encoding low-density lipoprotein receptor) or APOB (encoding apolipoprotein B). We mapped a third locus associated with ADH, HCHOLA3 at 1p32, and now report two mutations in the gene PCSK9 (encoding proprotein convertase subtilisin/kexin type 9) that cause ADH. PCSK9 encodes NARC-1 (neural apoptosis regulated convertase), a newly identified human subtilase that is highly expressed in the liver and contributes to cholesterol homeostasis.


Asunto(s)
Hiperlipoproteinemia Tipo II/genética , Mutación , Serina Endopeptidasas/genética , Sustitución de Aminoácidos , Cromosomas Humanos Par 1/genética , Femenino , Genes Dominantes , Ligamiento Genético , Humanos , Hiperlipoproteinemia Tipo II/enzimología , Hígado/enzimología , Masculino , Linaje , Proproteína Convertasa 9 , Proproteína Convertasas
4.
J Biol Chem ; 286(4): 2728-38, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21075846

RESUMEN

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.


Asunto(s)
Retículo Endoplásmico/enzimología , Precursores Enzimáticos/metabolismo , Subtilisinas/metabolismo , Animales , Células COS , Chlorocebus aethiops , Activación Enzimática/fisiología , Precursores Enzimáticos/genética , Furina/genética , Furina/metabolismo , Células HEK293 , Humanos , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Ratas , Subtilisinas/genética
5.
J Biol Chem ; 286(11): 9185-95, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21209099

RESUMEN

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.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Subtilisinas/metabolismo , Animales , Células COS , Chlorocebus aethiops , Factor de Crecimiento Epidérmico/genética , Receptores ErbB/genética , Células HEK293 , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Inhibidores de Proteasas/farmacología , Precursores de Proteínas/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Subtilisinas/antagonistas & inhibidores , Subtilisinas/genética
6.
J Biol Chem ; 286(26): 22785-94, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21550985

RESUMEN

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.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Furina/metabolismo , Ventrículos Cardíacos/enzimología , Miocardio/enzimología , Animales , Proteínas Morfogenéticas Óseas/genética , Células COS , Chlorocebus aethiops , Furina/antagonistas & inhibidores , Furina/genética , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Noqueados , Especificidad de Órganos , Inhibidores de Serina Proteinasa/farmacología
7.
Clin Chem ; 58(1): 183-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22065156

RESUMEN

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.


Asunto(s)
LDL-Colesterol/sangre , Fluorobencenos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Pirimidinas/uso terapéutico , Serina Endopeptidasas/sangre , Sulfonamidas/uso terapéutico , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proproteína Convertasa 9 , Proproteína Convertasas , Rosuvastatina Cálcica
8.
Br J Nutr ; 107(5): 705-11, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21787450

RESUMEN

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.


Asunto(s)
Colesterol/sangre , Dieta Mediterránea , Hipercolesterolemia/prevención & control , Síndrome Metabólico/sangre , Síndrome Metabólico/dietoterapia , Pérdida de Peso , Adolescente , Adulto , Anciano , Biomarcadores/sangre , Índice de Masa Corporal , Colesterol/metabolismo , LDL-Colesterol/sangre , LDL-Colesterol/metabolismo , Dieta Reductora , Humanos , Hipercolesterolemia/etiología , Isomerismo , Masculino , Síndrome Metabólico/metabolismo , Síndrome Metabólico/fisiopatología , Persona de Mediana Edad , Sobrepeso/fisiopatología , Fitosteroles/sangre , Proproteína Convertasa 9 , Proproteína Convertasas/sangre , Quebec , Serina Endopeptidasas/sangre , Adulto Joven
9.
J Lipid Res ; 52(3): 558-65, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21123766

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Intestinos/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Pirroles/farmacología , Adulto , Atorvastatina , Colesterol/metabolismo , Esquema de Medicación , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Duodeno/patología , Ácidos Heptanoicos/administración & dosificación , Humanos , Hiperlipidemias/patología , Mucosa Intestinal/metabolismo , Intestinos/patología , Masculino , Proteínas de Transporte de Membrana , Pirroles/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
J Biol Chem ; 285(52): 40965-78, 2010 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20937814

RESUMEN

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.


Asunto(s)
Endosomas/enzimología , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Receptores de LDL/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Endosomas/genética , Activación Enzimática/fisiología , Células HEK293 , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/enzimología , Lisosomas/genética , Mariposas Nocturnas , Péptidos/genética , Proproteína Convertasa 9 , Proproteína Convertasas , Unión Proteica/fisiología , Receptores de LDL/genética , Serina Endopeptidasas/genética
11.
Glycobiology ; 21(10): 1290-300, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21527438

RESUMEN

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.


Asunto(s)
Precursores Enzimáticos/metabolismo , Proproteína Convertasa 1/metabolismo , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Células CHO , Cricetinae , Activación Enzimática , Glicosilación , Células HEK293 , Humanos , Proopiomelanocortina/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Transporte de Proteínas , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , betaendorfina/metabolismo
12.
J Lipid Res ; 51(1): 140-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19571328

RESUMEN

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.


Asunto(s)
Monitoreo de Drogas/métodos , Ensayo de Inmunoadsorción Enzimática , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hiperlipoproteinemia Tipo II/sangre , Serina Endopeptidasas/sangre , Anticuerpos/inmunología , Azetidinas/uso terapéutico , Glucemia/análisis , Línea Celular , Colesterol/sangre , LDL-Colesterol/sangre , Estudios Transversales , Ezetimiba , Humanos , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Proproteína Convertasa 9 , Proproteína Convertasas , Serina Endopeptidasas/biosíntesis , Serina Endopeptidasas/inmunología , Triglicéridos/sangre
13.
J Clin Invest ; 111(11): 1723-32, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12782675

RESUMEN

The secretory factor VEGF-C has been directly implicated in various physiological processes during embryogenesis and human cancers. However, the importance of the conversion of its precursor proVEGF-C to mature VEGF-C in tumorigenesis, and vessel formation and the identity of the protease(s) that regulate these processes is/are not known. The intracellular processing of proVEGF-C that occurs within the dibasic motif HSIIRR(227)SL suggests the involvement of the proprotein convertases (PCs) in this process. In addition, furin and VEGF-C were found to be coordinately expressed in adult mouse tissues. Cotransfection of the furin-deficient colon carcinoma cell line LoVo with proVEGF-C and different PC members revealed that furin, PC5, and PC7 are candidate VEGF-C convertases. This finding is consistent with the in vitro digestions of an internally quenched synthetic fluorogenic peptide mimicking the cleavage site of proVEGF-C ((220)Q-VHSIIRR downward arrow SLP(230)). The processing of proVEGF-C is blocked by the inhibitory prosegments of furin, PC5, and PACE4, as well as by furin-motif variants of alpha2-macroglobulin and alpha1-antitrypsin. Subcutaneous injection of CHO cells stably expressing VEGF-C into nude mice enhanced angiogenesis and lymphangiogenesis, but not tumor growth. In contrast, expression of proVEGF-C obtained following mutation of the cleavage site (HSIIRR(227)SL to HSIISS(227)SL) inhibits angiogenesis and lymphangiogenesis as well as tumor growth. Our findings demonstrate the processing of proVEGF-C by PCs and highlight the potential use of PC inhibitors as agents for inhibiting malignancies induced by VEGF-C.


Asunto(s)
Carbamatos/metabolismo , Transformación Celular Neoplásica , Factores de Crecimiento Endotelial/metabolismo , Neoplasias/metabolismo , Oligopéptidos/metabolismo , Subtilisinas/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Western Blotting , Células CHO , Cricetinae , Relación Dosis-Respuesta a Droga , Furina , Humanos , Inmunohistoquímica , Cinética , Mutación , Biosíntesis de Péptidos , Pruebas de Precipitina , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección , Células Tumorales Cultivadas , Factor C de Crecimiento Endotelial Vascular
14.
Oncogene ; 24(46): 6925-35, 2005 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-16007151

RESUMEN

Platelet-derived growth factor-B (PDGF-B) is important for normal tissue growth and maintenance and its overexpression has been linked to several diseases, including cancer, fibrotic disease and atherosclerosis. Here, we show that synthesized as a precursor, proPDGF-B is converted to a mature form by proteolytic cleavage at two sites and its N-terminal cleavage is a prerequisite for processing at its C-terminus. The first cleavage occurs at residues RGRR81/, and the second cleavage close to residues ARPVT190, just before the C-terminal amino-acid sequence crucial for PDGF-B retention to cell surface. Cotransfection of a Furin-deficient cell line LoVo-C5 with proPDGF-B and different PC members revealed that Furin, PACE4, PC5, and PC7 are candidate proPDGF-B convertases. This finding is consistent with the in vitro digestions of a synthetic peptide mimicking the cleavage site of proPDGF-B. The processing of proPDGF-B is blocked by site-directed mutagenesis of the RGRR81/ sequence and by various PC inhibitors. Mutation of the PDGF-A and/or PDGF-B convertase sites, revealed that processing of both A and B chains is required for the formation of mature PDGF-B dimers and that the processing of the B chain controls the level of secreted and matrix-bound PDGF-BB forms. Our findings emphasize the importance of the convertase-directed processing of proPDGF-B at the RGRR81/ sequence for PDGF-B maturation and secretion.


Asunto(s)
Proproteína Convertasas/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Dimerización , Furina/metabolismo , Humanos , Hidrólisis , Inmunohistoquímica , Ratones , Datos de Secuencia Molecular , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-sis/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Cancer Res ; 63(7): 1458-63, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12670890

RESUMEN

Although altered expression of platelet-derived growth factor (PDGF)-A is a hallmark of many cancers, the importance of pro-PDGF-A conversion to PDGF-A in tumorigenesis and the cognate protease(s) is unknown. Pro-PDGF-A processing occurs at pairs of basic residues, likely involving the proprotein convertases (PCs). In the colon carcinoma cell line LoVo, we found that Furin is the most potent PDGF-A convertase. Mutation of the PC-site RRKR(86) to ARKA(86) inhibited pro-PDGF-A processing, its receptor tyrosine phosphorylation, and cell proliferation. This processing is also blocked by the PC preprosegments (pps) ppFurin, ppPC5, and ppPACE4, and by the Furin-variants of alpha2-macroglobulin and alpha1-antitrypsin. Chinese hamster ovary cells overexpressing pro-PDGF-A (ARKA(86)) failed to induce tumors in nude mice. Thus, PC-directed inhibitors might represent new agents for therapy in neoplasia induced by PDGF-A.


Asunto(s)
Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Precursores de Proteínas/metabolismo , Serina Endopeptidasas/metabolismo , Células 3T3 , Adenocarcinoma/enzimología , Adenocarcinoma/metabolismo , Animales , Sitios de Unión , Células CHO , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Cricetinae , Furina , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutagénesis Sitio-Dirigida , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/fisiología , Reacción en Cadena de la Polimerasa , Proproteína Convertasa 5 , Proproteína Convertasas , Serina Endopeptidasas/genética , Inhibidores de Serina Proteinasa/genética , Inhibidores de Serina Proteinasa/fisiología , Especificidad por Sustrato , Subtilisinas/antagonistas & inhibidores , Subtilisinas/genética , Subtilisinas/metabolismo , Transfección , Células Tumorales Cultivadas , Tirosina/metabolismo
16.
Mol Cell Biol ; 36(3): 507-21, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26598607

RESUMEN

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.


Asunto(s)
Citosol/metabolismo , Cinesinas/metabolismo , Proteínas de la Membrana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Caenorhabditis elegans , Línea Celular , Chlorocebus aethiops , Citosol/química , Drosophila melanogaster , Células HEK293 , Humanos , Cinesinas/química , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Alineación de Secuencia , Ubiquitina-Proteína Ligasas/química , Pez Cebra
17.
FEBS Lett ; 579(25): 5621-5, 2005 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-16213495

RESUMEN

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.


Asunto(s)
Retículo Endoplásmico/enzimología , Proproteína Convertasa 1/metabolismo , Red trans-Golgi/enzimología , Secuencia de Aminoácidos , Animales , Inhibidores Enzimáticos/metabolismo , Furina/genética , Furina/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Mutación , Proproteína Convertasa 1/antagonistas & inhibidores , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Vesículas Secretoras/enzimología , Subtilisinas/metabolismo
18.
Virol J ; 2: 69, 2005 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-16115318

RESUMEN

BACKGROUND: Severe acute respiratory syndrome (SARS) is caused by a newly discovered coronavirus (SARS-CoV). No effective prophylactic or post-exposure therapy is currently available. RESULTS: We report, however, that chloroquine has strong antiviral effects on SARS-CoV infection of primate cells. These inhibitory effects are observed when the cells are treated with the drug either before or after exposure to the virus, suggesting both prophylactic and therapeutic advantage. In addition to the well-known functions of chloroquine such as elevations of endosomal pH, the drug appears to interfere with terminal glycosylation of the cellular receptor, angiotensin-converting enzyme 2. This may negatively influence the virus-receptor binding and abrogate the infection, with further ramifications by the elevation of vesicular pH, resulting in the inhibition of infection and spread of SARS CoV at clinically admissible concentrations. CONCLUSION: Chloroquine is effective in preventing the spread of SARS CoV in cell culture. Favorable inhibition of virus spread was observed when the cells were either treated with chloroquine prior to or after SARS CoV infection. In addition, the indirect immunofluorescence assay described herein represents a simple and rapid method for screening SARS-CoV antiviral compounds.


Asunto(s)
Antivirales/farmacología , Cloroquina/farmacología , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Cloruro de Amonio/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente Indirecta , Glicosilación , Glicoproteínas de Membrana/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Síndrome Respiratorio Agudo Grave/metabolismo , Síndrome Respiratorio Agudo Grave/prevención & control , Síndrome Respiratorio Agudo Grave/virología , Glicoproteína de la Espiga del Coronavirus , Células Vero , Proteínas del Envoltorio Viral/metabolismo
19.
PLoS One ; 8(3): e60095, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23544125

RESUMEN

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.


Asunto(s)
Azetidinas/farmacología , Proproteína Convertasas/sangre , Serina Endopeptidasas/sangre , Simvastatina/farmacología , Regulación hacia Arriba/efectos de los fármacos , Adulto , LDL-Colesterol/sangre , Ezetimiba , Humanos , Masculino , Proproteína Convertasa 9
20.
Atherosclerosis ; 223(2): 394-400, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22683120

RESUMEN

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.


Asunto(s)
Hiperlipoproteinemia Tipo II/genética , Mutación , Proproteína Convertasas/genética , Serina Endopeptidasas/genética , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Biomarcadores/sangre , HDL-Colesterol/sangre , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Células Hep G2 , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/metabolismo , Masculino , Paris , Linaje , Fenotipo , Proproteína Convertasa 9 , Proproteína Convertasas/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Serina Endopeptidasas/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA