Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
PLoS One ; 17(1): e0262667, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35045102

RESUMO

Chronic kidney disease (CKD) is characterized by the loss of kidney function. The molecular mechanisms underlying the development and progression of CKD are still not fully understood. Among others, the urinary peptidome has been extensively studied, with several urinary peptides effectively detecting disease progression. However, their link to proteolytic events has not been made yet. This study aimed to predict the proteases involved in the generation of CKD-associated urinary excreted peptides in a well-matched (for age, sex, lack of heart disease) case-control study. The urinary peptide profiles from CKD (n = 241) and controls (n = 240) were compared and statistically analyzed. The in-silico analysis of the involved proteases was performed using Proteasix and proteases activity was predicted based on the abundance changes of the associated peptides. Predictions were cross-correlated to transcriptomics datasets by using the Nephroseq database. Information on the respective protease inhibitors was also retrieved from the MEROPS database. Totally, 303 urinary peptides were significantly associated with CKD. Among the most frequently observed were fragments of collagen types I, II and III, uromodulin, albumin and beta-2-microglobulin. Proteasix predicted 16 proteases involved in their generation. Through investigating CKD-associated transcriptomics datasets, several proteases are highlighted including members of matrix metalloproteinases (MMP7, MMP14) and serine proteases (PCSK5); laying the foundation for further studies towards elucidating their role in CKD pathophysiology.


Assuntos
Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Pró-Proteína Convertase 5/metabolismo , Idoso , Biomarcadores , Líquidos Corporais/metabolismo , Estudos de Casos e Controles , Bases de Dados Factuais , Feminino , Expressão Gênica/genética , Perfilação da Expressão Gênica , Humanos , Masculino , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/urina , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/urina , Peptídeo Hidrolases/metabolismo , Peptídeos/análise , Peptídeos/urina , Pró-Proteína Convertase 5/genética , Pró-Proteína Convertase 5/urina , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/urina , Transcriptoma/genética , Urina/química
2.
Front Endocrinol (Lausanne) ; 12: 690681, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149625

RESUMO

Fibroblast growth factor 23 (FGF23) is a hormone secreted from fully differentiated osteoblasts and osteocytes that inhibits phosphate reabsorption by kidney proximal tubules. The full-length (i.e., intact) protein mediates FGF23 endocrine functions, while endoproteolytic cleavage at a consensus cleavage sequence for the proprotein convertases (PCs) inactivates FGF23. Two PCs, furin and PC5, were shown to cleave FGF23 in vitro at RHTR179↓, but whether they are fulfilling this function in vivo is currently unknown. To address this question, we used here mice lacking either or both furin and PC5 in cell-specific manners and mice lacking the paired basic amino acid-cleaving enzyme 4 (PACE4) in all cells. Our analysis shows that furin inactivation in osteoblasts and osteocytes results in a 25% increase in circulating intact FGF23, without any significant impact on serum phosphate levels, whether mice are maintained on a normal or a low phosphate diet. Under conditions of iron deficiency, FGF23 is normally processed in control mice, but its processing is impaired in mice lacking furin in osteoblasts and osteocytes. In contrast, FGF23 is normally cleaved following erythropoietin or IL-1ß injections in mice lacking furin or both furin and PC5, and in PACE4-deficient mice. Altogether, these studies suggest that furin is only partially responsible for FGF23 cleavage under certain conditions in vivo. The processing of FGF23 may therefore involve the redundant action of multiple PCs or of other peptidases in osteoblasts, osteocytes and hematopoietic cells.


Assuntos
Fator de Crescimento de Fibroblastos 23/metabolismo , Furina/metabolismo , Osteoblastos/metabolismo , Osteócitos/metabolismo , Pró-Proteína Convertase 5/metabolismo , Animais , Medula Óssea/metabolismo , Fator de Crescimento de Fibroblastos 23/genética , Furina/genética , Deficiências de Ferro/genética , Deficiências de Ferro/metabolismo , Rim/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Pró-Proteína Convertase 5/genética
3.
Mol Med Rep ; 23(2)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33313955

RESUMO

Bone loss is a disease that is highly associated with aging. This deleterious health condition has become a public concern worldwide, and there is an urgent need to discover more novel therapeutic strategies for the development of age­associated osteoporosis. The present study aimed to explore the association between proprotein convertase subtilisin/kexin type 5 (PCSK5) and microRNA(miR)­338­3p in bone­formation and bone­loss processes. Western blotting assay and reverse transcription­quantitative PCR were employed to analyze PCSK5 and miR­338­3p expression levels in bone mesenchymal stem cells (BMSCs). Dual­luciferase reporter and RNA pull­down assays were used to determine the target. For osteoblastic differentiation verification, alkaline phosphatase activity, osteocalcin secretion detection, bone formation­related indicators (osterix, runt­related gene 2, osteopontin and bone sialoprotein), hematoxylin and eosin staining and Alizarin Red S staining were performed. The findings of the present study indicated that the expression level of PCSK5 was higher in BMSCs from young rat samples, whereas the expression level of miR­338­3p was higher in BMSCs from samples of old rats. Experimental results also revealed that unlike miR­338­3p, downregulation of PCSK5 inhibited osteoblastic differentiation and osteogenesis by inhibiting alkaline phosphatase, osteocalcin, osterix, runt­related transcription factor 2, osteopontin, bone sialoprotein and mineralized nodule formation. Overall, the results suggested that miR­338­3p could suppress age­associated osteoporosis by regulating PCSK5.


Assuntos
MicroRNAs/genética , MicroRNAs/metabolismo , Osteoporose/genética , Osteoporose/metabolismo , Pró-Proteína Convertase 5/genética , Pró-Proteína Convertase 5/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Osso e Ossos/patologia , Diferenciação Celular/genética , Células Cultivadas , Biologia Computacional , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Sialoproteína de Ligação à Integrina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteogênese/genética , Osteopontina/metabolismo , RNA Interferente Pequeno , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo
4.
Endocr Rev ; 42(3): 259-294, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-33382413

RESUMO

The kexin-like proprotein convertases perform the initial proteolytic cleavages that ultimately generate a variety of different mature peptide and proteins, ranging from brain neuropeptides to endocrine peptide hormones, to structural proteins, among others. In this review, we present a general introduction to proprotein convertase structure and biochemistry, followed by a comprehensive discussion of each member of the kexin-like subfamily of proprotein convertases. We summarize current knowledge of human proprotein convertase insufficiency syndromes, including genome-wide analyses of convertase polymorphisms, and compare these to convertase null and mutant mouse models. These mouse models have illuminated our understanding of the roles specific convertases play in human disease and have led to the identification of convertase-specific substrates; for example, the identification of procorin as a specific PACE4 substrate in the heart. We also discuss the limitations of mouse null models in interpreting human disease, such as differential precursor cleavage due to species-specific sequence differences, and the challenges presented by functional redundancy among convertases in attempting to assign specific cleavages and/or physiological roles. However, in most cases, knockout mouse models have added substantively both to our knowledge of diseases caused by human proprotein convertase insufficiency and to our appreciation of their normal physiological roles, as clearly seen in the case of the furin, proprotein convertase 1/3, and proprotein convertase 5/6 mouse models. The creation of more sophisticated mouse models with tissue- or temporally-restricted expression of specific convertases will improve our understanding of human proprotein convertase insufficiency and potentially provide support for the emerging concept of therapeutic inhibition of convertases.


Assuntos
Estudo de Associação Genômica Ampla , Pró-Proteína Convertases , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Pró-Proteína Convertase 5/metabolismo , Pró-Proteína Convertases/química , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo
5.
Dis Model Mech ; 12(6)2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31101650

RESUMO

Global inhibition of N-linked glycosylation broadly reduces glycan occupancy on glycoproteins, but identifying how this inhibition functionally impacts specific glycoproteins is challenging. This limits our understanding of pathogenesis in the congenital disorders of glycosylation (CDG). We used selective exo-enzymatic labeling of cells deficient in the two catalytic subunits of oligosaccharyltransferase - STT3A and STT3B - to monitor the presence and glycosylation status of cell surface glycoproteins. We show reduced abundance of two canonical tyrosine receptor kinases - the insulin receptor and insulin-like growth factor 1 receptor (IGF-1R) - at the cell surface in STT3A-null cells, due to decreased N-linked glycan site occupancy and proteolytic processing in combination with increased endoplasmic reticulum localization. Providing cDNA for Golgi-resident proprotein convertase subtilisin/kexin type 5a (PCSK5a) and furin cDNA to wild-type and mutant cells produced under-glycosylated forms of PCSK5a, but not furin, in cells lacking STT3A. Reduced glycosylation of PCSK5a in STT3A-null cells or cells treated with the oligosaccharyltransferase inhibitor NGI-1 corresponded with failure to rescue receptor processing, implying that alterations in the glycosylation of this convertase have functional consequences. Collectively, our findings show that STT3A-dependent inhibition of N-linked glycosylation on receptor tyrosine kinases and their convertases combines to impair receptor processing and surface localization. These results provide new insight into CDG pathogenesis and highlight how the surface abundance of some glycoproteins can be dually impacted by abnormal glycosylation.


Assuntos
Processamento de Proteína Pós-Traducional , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Animais , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Furina/metabolismo , Glicosilação , Células HEK293 , Hexosiltransferases/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Polissacarídeos/metabolismo , Pró-Proteína Convertase 5/metabolismo , Proteômica , Receptores Proteína Tirosina Quinases/metabolismo , Receptor de Insulina/metabolismo
6.
Bone ; 107: 45-55, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29126984

RESUMO

Seven proprotein convertases cleave the basic amino acid consensus sequence K/R-Xn-K/R↓ (where n=0, 2, 4 or 6 variable amino acids) to activate precursor proteins. Despite similarities in substrate specificity, basic amino acid-specific proprotein convertases have a distinct tissue distribution allowing for enzymatic actions on tissue-resident substrates. Proprotein convertase 5/6 (PC5/6) has two splice variants - soluble PC5/6A and membrane-bound PC5/6B - and is expressed during mouse development in many tissues including bone and tooth, but little is known about the substrates for PC5/6 therein. Osteopontin (OPN) is an abundant bone extracellular matrix protein with roles in mineralization, cell adhesion and cell migration, and it has putative consensus sequence sites for cleavage by PC5/6, which may modify its function in bone. Since PC5/6-knockout mouse embryos show developmental abnormalities, and reduced overall mineralization, we designed this study to determine whether OPN is a substrate of PC5/6. In silico analysis of OPN protein sequences identified four potential PC5/6 consensus cleavage sites in human OPN, and three sites - including a noncanonical sequence - in mouse OPN. Ex vivo co-transfections with human OPN revealed complete OPN cleavage reducing full-length OPN (~70kDa) to an N-terminal fragment migrating at ~50kDa and two C-terminal fragments at ~18kDa and ~16kDa. Direct cleavage of OPN by PC5/6A - the predominant isoform expressed in human osteoblast cells - was confirmed by cell-free enzyme-substrate assays and by mass spectrometry. The latter was also used to investigate potential cleavage sites. Co-transfections of PC5/6 and mouse OPN showed partial cleavage of OPN into a C-terminal OPN fragment migrating at ~30kDa and an N-terminal fragment migrating at ~29kDa. Micro-computed tomography of PC5/6-knockout embryos at E18.5 confirmed a reduction in mineralized bone, and in situ hybridization performed on cryo-sections of normal mouse bone using Pcsk5 and Opn anti-sense and control-sense cRNA probes indicated the co-localization of the expression of these genes in bone cells. This mRNA expression profile was supported by semi-quantitative RT-PCR using osteoblast primary cultures, and cultured MC3T3-E1 osteoblast and MLO-Y4 osteocyte cell lines. Immunoblotting for OPN from mouse bone extracts showed altered OPN processing in PC5/6-knockout mice compared to wildtype mice. OPN fragments migrated at ~25kDa and ~16kDa in wildtype bone and were not present in PC5/6-deficient bone. In conclusion, this study demonstrates that Pcsk5 is expressed in bone-forming cells, and that OPN is a novel substrate for PC5/6. Cleavage of OPN by PC5/6 may modify the function of OPN in bone and/or modulate other enzymatic cleavages of OPN, leading to alterations in the bone phenotype.


Assuntos
Osso e Ossos/metabolismo , Osteopontina/metabolismo , Pró-Proteína Convertase 5/metabolismo , Animais , Calcificação Fisiológica/fisiologia , Humanos , Camundongos , Camundongos Knockout , Especificidade por Substrato
7.
J Mol Histol ; 48(5-6): 379-387, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28975535

RESUMO

This study examined the expression patterns of proprotein convertase subtilisin/kexin type 5 (Pcsk5) during anorectal development in normal and anorectal malformations (ARM) rat embryos, determine the possible role of Pcsk5 in the pathogenesis of ARM. An ARM rat model was developed by the administration of ethylenethiourea gestational day 10 (GD10). Embryos were harvested by surgical excision from GD13 to GD16, and the spatiotemporal expression of Pcsk5 was evaluated, using immunohistochemistry staining, Western blotting and real time RT-PCR. Immunohistochemistry staining in normal embryos revealed that Pcsk5 was abundantly expressed on the epithelium of the cloaca (CL) on GD13. On GD14 and GD15, positive cells were noted on the urorectal septum and the thin anal membrane. However, the epithelium of the CL of ARM embryos only faintly expressed Pcsk5 from GD13 to GD15. Western blotting and real time RT-PCR showed time-dependent increase of Pcsk5 expression in the developing hindgut. Pcsk5 expression levels were lower in the ARM group from GD14 to GD16 (p ≤ 0.05). These results indicate that downregulation of Pcsk5 during cloaca development into the rectum and urethra might be related to the formation of ARMs.


Assuntos
Malformações Anorretais/genética , Feto/anormalidades , Feto/metabolismo , Pró-Proteína Convertase 5/genética , Animais , Malformações Anorretais/patologia , Western Blotting , Cloaca/embriologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/metabolismo , Imuno-Histoquímica , Pró-Proteína Convertase 5/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
8.
J Clin Invest ; 127(11): 4104-4117, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28972540

RESUMO

Osteocalcin (OCN) is an osteoblast-derived hormone that increases energy expenditure, insulin sensitivity, insulin secretion, and glucose tolerance. The cDNA sequence of OCN predicts that, like many other peptide hormones, OCN is first synthesized as a prohormone (pro-OCN). The importance of pro-OCN maturation in regulating OCN and the identity of the endopeptidase responsible for pro-OCN cleavage in osteoblasts are still unknown. Here, we show that the proprotein convertase furin is responsible for pro-OCN maturation in vitro and in vivo. Using pharmacological and genetic experiments, we also determined that furin-mediated pro-OCN cleavage occurred independently of its γ-carboxylation, a posttranslational modification that is known to hamper OCN endocrine action. However, because pro-OCN is not efficiently decarboxylated and activated during bone resorption, inactivation of furin in osteoblasts in mice resulted in decreased circulating levels of undercarboxylated OCN, impaired glucose tolerance, and reduced energy expenditure. Furthermore, we show that Furin deletion in osteoblasts reduced appetite, a function not modulated by OCN, thus suggesting that osteoblasts may secrete additional hormones that regulate different aspects of energy metabolism. Accordingly, the metabolic defects of the mice lacking furin in osteoblasts became more apparent under pair-feeding conditions. These findings identify furin as an important regulator of bone endocrine function.


Assuntos
Osso e Ossos/enzimologia , Furina/fisiologia , Osteocalcina/metabolismo , Sequência de Aminoácidos , Animais , Osso e Ossos/citologia , Células Cultivadas , Sistema Endócrino , Metabolismo Energético , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/enzimologia , Pró-Proteína Convertase 5/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteólise , Células RAW 264.7
9.
Eur J Cell Biol ; 96(5): 457-468, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28624236

RESUMO

Proprotein convertases (PCs) are involved in various physiological and pathological processes ranging from embryogenesis to carcinogenesis. Here, using the zebrafish fin regeneration model, we report induced expression of furin and PC5 but not PACE4 and PC7 during fin regeneration that is associated with increased PC activity. Stabilization of HIF by cobalt chloride (CoCl2) further increases these processes. The use of the general PC-inhibitor decanoyl-RVKR-cholromethyl ketone (CMK) inhibited control and CoCl2-induced PC activity. CoCl2 inhibits embryonic zebrafish ZF4 cell proliferation and caudal fin regeneration that is associated with the expression of the anti-proliferative genes P21, P16, PC3 and P53 in ZF4 cells and in non-regenerating stump tissues. In contrast, during fin regeneration, HIF stabilization failed to promote the expression of these anti-proliferative genes and maintained high expression of cyclin D. Further analysis revealed that CoCl2 maintained the formation of immature regenerating vasculature that was associated with amplified expression of OCT4 and various angiogenic factors reported to be PC substrates and/or downstream effectors. These findings revealed that while furin and PC5 expression/activity and their substrates/effectors are regulated during fin regeneration, HIF stabilization by CoCl2 has the potential to modulate these processes and impact on the regenerative process and vessels organization.


Assuntos
Furina/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Neovascularização Fisiológica/fisiologia , Pró-Proteína Convertase 5/metabolismo , Nadadeiras de Animais/fisiologia , Animais , Animais Geneticamente Modificados , Regeneração/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
10.
J Biol Chem ; 292(25): 10564-10573, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28468828

RESUMO

Protein C, a secretory vitamin K-dependent anticoagulant serine protease, inactivates factors Va/VIIIa. It is exclusively synthesized in liver hepatocytes as an inactive zymogen (proprotein C). In humans, thrombin cleavage of the propeptide at PR221↓ results in activated protein C (APC; residues 222-461). However, the propeptide is also cleaved by a furin-like proprotein convertase(s) (PCs) at KKRSHLKR199↓ (underlined basic residues critical for the recognition by PCs), but the order of cleavage is unknown. Herein, we present evidence that at the surface of COS-1 cells, mouse proprotein C is first cleaved by the convertases furin, PC5/6A, and PACE4. In mice, this cleavage occurs at the equivalent site, KKRKILKR198↓, and requires the presence of Arg198 at P1 and a combination of two other basic residues at either P2 (Lys197), P6 (Arg193), or P8 (Lys191) positions. Notably, the thrombin-resistant R221A mutant is still cleaved by these PCs, revealing that convertase cleavage can precede thrombin activation. This conclusion was supported by the fact that the APC-specific activity in the medium of COS-1 cells is exclusively dependent on prior cleavage by the convertases, because both R198A and R221A lack protein C activity. Primary cultures of hepatocytes derived from wild-type or hepatocyte-specific furin, PC5/6, or complete PACE4 knock-out mice suggested that the cleavage of overexpressed proprotein C is predominantly performed by furin intracellularly and by all three proprotein convertases at the cell surface. Indeed, plasma analyses of single-proprotein convertase-knock-out mice showed that loss of the convertase furin or PC5/6 in hepatocytes results in a ∼30% decrease in APC levels, with no significant contribution from PACE4. We conclude that prior convertase cleavage of protein C in hepatocytes is critical for its thrombin activation.


Assuntos
Hepatócitos/enzimologia , Fígado/enzimologia , Pró-Proteína Convertase 5/metabolismo , Proteína C/metabolismo , Substituição de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática/fisiologia , Células Hep G2 , Humanos , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Pró-Proteína Convertase 5/genética , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Proteína C/genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Trombina/genética , Trombina/metabolismo
11.
BMC Dev Biol ; 17(1): 6, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28446132

RESUMO

BACKGROUND: Loss of proprotein convertase subtilisin/kexin type 5 (Pcsk5) results in multiple developmental anomalies including cardiac malformations, caudal regression, pre-sacral mass, renal agenesis, anteroposterior patterning defects, and tracheo-oesophageal and anorectal malformations, and is a model for VACTERL/caudal regression/Currarino syndromes (VACTERL association - Vertebral anomalies, Anal atresia, Cardiac defects, Tracheoesophageal fistula and/or Esophageal atresia, Renal & Radial anomalies and Limb defects). RESULTS: Using magnetic resonance imaging (MRI), we examined heart development in mouse embryos with zygotic and cardiac specific deletion of Pcsk5. We show that conditional deletion of Pcsk5 in all epiblastic lineages recapitulates all developmental malformations except for tracheo-esophageal malformations. Using a conditional deletion strategy, we find that there is an essential and specific requirement for Pcsk5 in the cranio-cardiac mesoderm for cardiogenesis, but not for conotruncal septation or any other aspect of embryonic development. Surprisingly, deletion of Pcsk5 in cardiogenic or pharyngeal mesodermal progenitors that form later from the cranio-cardiac mesoderm does not affect heart development. Neither is Pcsk5 essential in the neural crest, which drives conotruncal septation. CONCLUSIONS: Our results suggest that Pcsk5 may have an essential and early role in the cranio-cardiac mesoderm for heart development. Alternatively, it is possible that Pcsk5 may still play a critical role in Nkx2.5-expressing cardiac progenitors, with persistence of mRNA or protein accounting for the lack of effect of deletion on heart development.


Assuntos
Coração/embriologia , Mesoderma/embriologia , Organogênese/genética , Pró-Proteína Convertase 5/genética , Pró-Proteína Convertase 5/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Camundongos , Camundongos Knockout , Mutação
12.
Mol Cell Biol ; 35(21): 3684-700, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26283733

RESUMO

The proprotein convertases (PCs) furin, PC5, PACE4, and PC7 cleave secretory proteins after basic residues, including the HIV envelope glycoprotein (gp160) and Vpr. We evaluated the abundance of PC mRNAs in postmortem brains of individuals exhibiting HIV-associated neurocognitive disorder (HAND), likely driven by neuroinflammation and neurotoxic HIV proteins (e.g., envelope and Vpr). Concomitant with increased inflammation-related gene expression (interleukin-1ß [IL-1ß]), the mRNA levels of the above PCs are significantly increased, together with those of the proteinase-activated receptor 1 (PAR1), an inflammation-associated receptor that is cleaved by thrombin at ProArg41↓ (where the down arrow indicates the cleavage location), and potentially by PCs at Arg41XXXXArg46↓. The latter motif in PAR1, but not its R46A mutant, drives its interactions with PCs. Indeed, PAR1 upregulation leads to the inhibition of membrane-bound furin, PC5B, and PC7 and inhibits gp160 processing and HIV infectivity. Additionally, a proximity ligation assay revealed that furin and PC7 interact with PAR1. Reciprocally, increased furin expression reduces the plasma membrane abundance of PAR1 by trapping it in the trans-Golgi network. Furthermore, soluble PC5A/PACE4 can target/disarm cell surface PAR1 through cleavage at Arg46↓. PACE4/PC5A decreased calcium mobilization induced by thrombin stimulation. Our data reveal a new PC-PAR1-interaction pathway, which offsets the effects of HIV-induced neuroinflammation, viral infection, and potentially the development of HAND.


Assuntos
Encéfalo/patologia , Infecções por HIV/complicações , Inflamação/complicações , Transtornos Neurocognitivos/complicações , Pró-Proteína Convertases/metabolismo , Mapas de Interação de Proteínas , Receptor PAR-1/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Linhagem Celular , Furina/genética , Regulação da Expressão Gênica , Proteína gp160 do Envelope de HIV/metabolismo , Infecções por HIV/genética , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Dados de Sequência Molecular , Transtornos Neurocognitivos/genética , Transtornos Neurocognitivos/metabolismo , Transtornos Neurocognitivos/patologia , Pró-Proteína Convertase 5/análise , Pró-Proteína Convertase 5/metabolismo , Pró-Proteína Convertases/análise , Pró-Proteína Convertases/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Receptor PAR-1/análise , Receptor PAR-1/genética , Serina Endopeptidases/análise , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Subtilisinas/análise , Subtilisinas/genética , Subtilisinas/metabolismo , Trombina/metabolismo
13.
Eur J Cell Biol ; 94(7-9): 375-83, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26095298

RESUMO

Cellular proteases are reponsible for activation of influenza virus hemagglutinin (HA) in epithelial tissues of the respiratory tract. The trans-Golgi network (TGN) is the main subcellular compartment where HA cleavage occurs during its biosynthesis. The proteolytic HA cleavage is an indispensable prerequisite for the fusion of viral with endosomal membrane and the delivery of the virus genome into the cell. Both, the structure and accessibility of the HA cleavage site determine the responsible host protease(s) for cutting. Most influenza virus strains contain a HA sequence with a single arginine at the cleavage site suitable for processing by the trypsin-like serine proteases human airway trypsin-like protease (HAT) and transmembrane protease serine 2 (TMPRSS2), albeit a minority of viruses possesses HA cleavage site motifs that are processed by other proteases. TMPRSS2-deficient mice demonstrated the relevance of TMPRSS2 for pneumotropism and pathogenicity of H1N1 and H7N9 virus infections. In contrast, H3N2 virus infections are promoted by an additional not yet identified protease. Highly pathogenic avian H5 and H7 viruses are characterized by an enlarged cleavage site loop containing a multibasic amino acid motif, where the eukaryotic subtilases furin or PC5/6 cleave. Their ubiquitous presence in the organism allows a systemic virus infection. Peptidomimetic inhibitors derived from the HA cleavage site inhibit the HA-activating proteases and thus virus propagation.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A Subtipo H1N1/patogenicidade , Subtipo H7N9 do Vírus da Influenza A/patogenicidade , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Serina Proteases/metabolismo , Animais , Furina/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/biossíntese , Humanos , Vírus da Influenza A Subtipo H3N2/patogenicidade , Influenza Humana/patologia , Influenza Humana/virologia , Camundongos , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Pró-Proteína Convertase 5/metabolismo , Mucosa Respiratória/virologia , Rede trans-Golgi/metabolismo
14.
FASEB J ; 29(9): 4011-22, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26077903

RESUMO

Embryo implantation requires a healthy embryo and a receptive endometrium (inner lining of the uterus); endometrial receptivity acquisition involves considerable epithelial surface remodeling. Dystroglycan (DG), a large cell surface glycoprotein, consists of α- and ß-subunits; ß-DG anchors within the plasma membrane whereas α-DG attaches extracellularly to ß-DG. The glycosylated central α-DG mediates adhesion, but it is obstructed by its large N terminus (α-DG-N); α-DG-N removal enables DG's adhesive function. We demonstrate here that full-length α-DG in the human endometrial epithelium is a barrier for embryo attachment and that removal of α-DG-N by proprotein convertase 5/6 (PC6; a protease critical for implantation) regulates receptivity. This was evidenced by: 1) α-DG contains a PC6-cleavage site near α-DG-N, and PC6 cleaves a peptide harboring such a site; 2) PC6 knockdown reduces α-DG-N removal from endometrial epithelial cell surface and blastocyst adhesion; 3) mutating the PC6-cleavage site prevents α-DG-N removal, causing cell surface retention of full-length α-DG and loss of adhesiveness; 4) α-DG-N is removed from endometrial tissue in vivo for receptivity and uterine fluid α-DG-N reflects tissue removal and receptivity. We thus identified α-DG-N removal as an important posttranslational control of endometrial receptivity and uterine fluid α-DG-N as a potential biomarker for receptivity in women.


Assuntos
Distroglicanas/metabolismo , Implantação do Embrião/fisiologia , Endométrio/metabolismo , Pró-Proteína Convertase 5/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Proteólise , Blastocisto/citologia , Blastocisto/metabolismo , Linhagem Celular , Distroglicanas/genética , Endométrio/citologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Epitélio/metabolismo , Feminino , Humanos , Pró-Proteína Convertase 5/genética , Estrutura Terciária de Proteína
15.
Anal Biochem ; 475: 14-21, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25554488

RESUMO

Embryo implantation requires a healthy embryo and a receptive uterus. In women, the inner lining of the uterus, the endometrium, remains in a hostile state and becomes receptive for embryo implantation for only a short period during each menstrual cycle. Determining endometrial receptivity is vital in in vitro fertilization (IVF) treatment because the timing of embryo transfer needs to be synchronized with endometrial receptivity. We have previously demonstrated that proprotein convertase 5/6A (PC6) is highly expressed in the receptive endometrium and that PC6 is critical for receptivity establishment in women. Furthermore, endometrial PC6 is secreted into the uterine fluid, and levels correlate with receptivity status. Detection of PC6 in uterine fluids, therefore, would provide a nonsurgical assessment of endometrial receptivity. However, to date no assays are available for human PC6. In this study, we produced three PC6 monoclonal antibodies (mAbs) and developed a sandwich enzyme-linked immunosorbent assay (ELISA) for PC6 detection in human uterine fluids. The PC6 mAbs were confirmed to be highly specific to PC6, and the ELISA detected PC6 in human uterine fluids with a significantly higher level during the receptive phase. This newly established PC6 ELISA provides an important tool in the development of noninvasive strategies to detect endometrial receptivity in women.


Assuntos
Anticorpos Monoclonais Murinos/química , Endométrio/enzimologia , Pró-Proteína Convertase 5/metabolismo , Animais , Implantação do Embrião/fisiologia , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Humanos , Camundongos
16.
Mol Hum Reprod ; 21(3): 262-70, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25429785

RESUMO

Establishment of endometrial receptivity is vital for successful embryo implantation. Proprotein convertase 5/6 (referred to as PC6) is up-regulated in the human endometrium specifically at the time of epithelial receptivity. PC6, a serine protease of the proprotein convertase family, plays an important role in converting precursor proteins into their active forms through specific proteolysis. The proform of platelet-derived growth factor A (pro-PDGFA) requires PC cleavage to convert to the active-PDGFA. We investigated the PC6-mediated activation of PDGFA in the human endometrium during the establishment of receptivity. Proteomic analysis identified that the pro-PDGFA was increased in the conditioned medium of HEC1A cells in which PC6 was stably knocked down by small interfering RNA (PC6-siRNA). Western blot analysis demonstrated an accumulation of the pro-PDGFA but a reduction in the active-PDGFA in PC6-siRNA cell lysates and medium compared with control. PC6 cleavage of pro-PDGFA was further confirmed in vitro by incubation of recombinant pro-PDGFA with PC6. Immunohistochemistry revealed cycle-stage-specific localization of the active-PDGFA in the human endometrium. During the non-receptive phase, the active-PDGFA was barely detectable. In contrast, it was localized specifically to the apical surface of the luminal and glandular epithelium in the receptive phase. Furthermore, the active-PDGFA was detected in uterine lavage with levels being significantly higher in the receptive than the non-receptive phase. We thus identified that the secreted PDGFA may serve as a biomarker for endometrial receptivity. This is also the first study demonstrating that the active-PDGFA localizes to the apical surface of the endometrium during receptivity.


Assuntos
Endométrio/metabolismo , Células Epiteliais/metabolismo , Período Fértil/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Pró-Proteína Convertase 5/metabolismo , Adulto , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Implantação do Embrião/fisiologia , Embrião de Mamíferos , Endométrio/citologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Período Fértil/metabolismo , Fase Folicular/genética , Fase Folicular/metabolismo , Expressão Gênica , Inativação Gênica , Humanos , Fator de Crescimento Derivado de Plaquetas/genética , Pró-Proteína Convertase 5/antagonistas & inibidores , Pró-Proteína Convertase 5/genética , Proteólise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
17.
PLoS One ; 8(12): e81380, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324690

RESUMO

Uterine proprotein convertase (PC) 6 plays a critical role in embryo implantation and is pivotal for pregnancy establishment. Inhibition of PC6 may provide a novel approach for the development of non-hormonal and female-controlled contraceptives. We investigated a class of five synthetic non-peptidic small molecule compounds that were previously reported as potent inhibitors of furin, another PC member. We examined (i) the potency of these compounds in inhibiting PC6 activity in vitro; (ii) their binding modes in the PC6 active site in silico; (iii) their efficacy in inhibiting PC6-dependent cellular processes essential for embryo implantation using human cell-based models. All five compounds showed potent inhibition of PC6 activity in vitro, and in silico docking demonstrated that these inhibitors could adopt a similar binding mode in the PC6 active site. However, when these compounds were tested for their inhibition of decidualization of primary human endometrial stromal cells, a PC6-dependent cellular process critical for embryo implantation, only one (compound 1o) showed potent inhibition. The lack of activity in the cell-based assay may reflect the inability of the compounds to penetrate the cell membrane. Because compound's lipophilicity is linked to cell penetration, a measurement of lipophilicity (logP) was calculated for each compound. Compound 1o is unique as it appears the most lipophilic among the five compounds. Compound 1o also inhibited another crucial PC6-dependent process, the attachment of human trophoblast spheroids to endometrial epithelial cells (a model for human embryo attachment). We thus identified compound 1o as a potent small molecule PC6 inhibitor with pharmaceutical potential to inhibit embryo implantation. Our findings also highlight that human cell-based functional models are vital to complement the biochemical and in silico analyses in the selection of promising drug candidates. Further investigations for compound 1o are warranted in animal models to test its utility as an implantation-inhibiting contraceptive drug.


Assuntos
Implantação do Embrião/efeitos dos fármacos , Pró-Proteína Convertase 5/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Domínio Catalítico , Linhagem Celular , Simulação por Computador , Decídua/efeitos dos fármacos , Endométrio/citologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Integrina alfaV/metabolismo , Lipídeos/química , Modelos Moleculares , Peso Molecular , Gravidez , Pró-Proteína Convertase 5/metabolismo , Inibidores de Proteases/química , Bibliotecas de Moléculas Pequenas/química , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos
18.
Am J Physiol Lung Cell Mol Physiol ; 305(2): L130-40, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23686857

RESUMO

Nitric oxide and cGMP modulate vascular smooth muscle cell (SMC) phenotype by regulating cell differentiation and proliferation. Recent studies suggest that cGMP-dependent protein kinase I (PKGI) cleavage and the nuclear translocation of a constitutively active kinase fragment, PKGIγ, are required for nuclear cGMP signaling in SMC. However, the mechanisms that control PKGI proteolysis are unknown. Inspection of the amino acid sequence of a PKGI cleavage site that yields PKGIγ and a protease database revealed a putative minimum consensus sequence for proprotein convertases (PCs). Therefore we investigated the role of PCs in regulating PKGI proteolysis. We observed that overexpression of PCs, furin and PC5, but not PC7, which are all expressed in SMC, increase PKGI cleavage in a dose-dependent manner in human embryonic kidney (HEK) 293 cells. Moreover, furin-induced proteolysis of mutant PKGI, in which alanines were substituted into the putative PC consensus sequence, was decreased in these cells. In addition, overexpression of furin increased PKGI proteolysis in LoVo cells, which is an adenocarcinoma cell line expressing defective furin without PC activity. Also, expression of α1-PDX, an engineered serpin-like PC inhibitor, reduced PC activity and decreased PKGI proteolysis in HEK293 cells. Last, treatment of low-passage rat aortic SMC with membrane-permeable PC inhibitor peptides decreased cGMP-stimulated nuclear PKGIγ translocation. These data indicate for the first time that PCs have a role in regulating PKGI proteolysis and the nuclear localization of its active cleavage product, which are important for cGMP-mediated SMC phenotype.


Assuntos
Núcleo Celular/enzimologia , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Furina/metabolismo , Pró-Proteína Convertase 5/metabolismo , Proteólise , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Tumoral , Núcleo Celular/genética , GMP Cíclico/genética , GMP Cíclico/metabolismo , Furina/genética , Células HEK293 , Humanos , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/enzimologia , Pró-Proteína Convertase 5/genética , Ratos , Subtilisinas/genética , Subtilisinas/metabolismo
19.
Neoplasia ; 14(10): 880-92, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23097623

RESUMO

We recently demonstrated that lack of Furin-processing of the N-cadherin precursor (proNCAD) in highly invasive melanoma and brain tumor cells results in the cell-surface expression of a nonadhesive protein favoring cell migration and invasion in vitro. Quantitative polymerase chain reaction analysis of malignant human brain tumor cells revealed that of all proprotein convertases (PCs) only the levels of Furin and PC5A are modulated, being inversely (Furin) or directly (PC5A) correlated with brain tumor invasive capacity. Intriguingly, the N-terminal sequence following the Furin-activated NCAD site (RQKR↓DW(161), mouse nomenclature) reveals a second putative PC-processing site (RIRSDR↓DK(189)) located in the first extracellular domain. Cleavage at this site would abolish the adhesive functions of NCAD because of the loss of the critical Trp(161). This was confirmed upon analysis of the fate of the endogenous prosegment of proNCAD in human malignant glioma cells expressing high levels of Furin and low levels of PC5A (U343) or high levels of PC5A and negligible Furin levels (U251). Cellular analyses revealed that Furin is the best activating convertase releasing an ~17-kDa prosegment, whereas PC5A is the major inactivating enzyme resulting in the secretion of an ~20-kDa product. Like expression of proNCAD at the cell surface, cleavage of the NCAD molecule at RIRSDR↓DK(189) renders the U251 cancer cells less adhesive to one another and more migratory. Our work modifies the present view on posttranslational processing and surface expression of classic cadherins and clarifies how NCAD possesses a range of adhesive potentials and plays a critical role in tumor progression.


Assuntos
Antígenos CD/metabolismo , Neoplasias Encefálicas/metabolismo , Caderinas/metabolismo , Movimento Celular , Furina/metabolismo , Glioma/metabolismo , Pró-Proteína Convertase 5/metabolismo , Antígenos CD/genética , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Caderinas/genética , Furina/antagonistas & inibidores , Furina/genética , Glioma/genética , Glioma/patologia , Células HeLa , Humanos , Técnicas Imunoenzimáticas , Pró-Proteína Convertase 5/antagonistas & inibidores , Pró-Proteína Convertase 5/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Cicatrização
20.
Hum Reprod ; 27(9): 2766-74, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22740495

RESUMO

BACKGROUND: Proprotein convertases (PCs) post-translationally activate a large number of protein precursors through limited cleavage. PC5/6 (PC6) in the human endometrium is tightly regulated during receptivity for embryo implantation. Integrins are transmembrane glycoproteins, some of which play an important role in the adhesive interactions between the trophoblast (blastocyst) and uterine epithelium at implantation. Integrins require PC cleavage for post-translational modification. We hypothesize that pro-integrin-αs in the endometrial epithelium are post-translationally cleaved by PC6 into functional subunits for the binding of blastocyst and adhesion of extracellular matrix proteins. METHODS AND RESULTS: We first used the endometrial epithelial cell line, HEC1A, into which siRNA specific to human PC6 (PC6-siRNA) or scrambled sequence (control) was stably transfected. The specific knockdown was confirmed by real-time RT-PCR. PC6-siRNA cells reduced their capacity to attach to trophoblast spheroids and bind to fibronectin compared with control. Knockdown of PC6 decreased cell surface presentation of functional integrins-α1, α2, α5, αV and αVß5. Western blot analysis demonstrated that PC6 was responsible for the post-translational cleavage of pro-integrin-α5 and integrin-αV into their heavy and light chains in HEC1A cells. We then isolated primary human endometrial epithelial cells and validated that PC6 mediated the post-translational cleavage of integrin-αs in these cells. CONCLUSIONS: This study implicates PC6 as a key regulatory protein essential for the attachment of the blastocyst to the endometrial epithelium through the processing of pro-integrin-αs. Compromised PC6 action reduces the post-translational modification of integrin-αs, thus compromising implantation.


Assuntos
Endométrio/enzimologia , Regulação Enzimológica da Expressão Gênica , Cadeias alfa de Integrinas/química , Pró-Proteína Convertase 5/metabolismo , Biópsia , Blastocisto/citologia , Cálcio/química , Adesão Celular , Células Cultivadas , Implantação do Embrião , Endométrio/citologia , Endométrio/metabolismo , Feminino , Fibronectinas/metabolismo , Glicoproteínas/metabolismo , Humanos , Cadeias alfa de Integrinas/metabolismo , Gravidez , Processamento de Proteína Pós-Traducional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA