Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 26(1): 233-242, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28011710

RESUMO

Chromogranins are pro-hormone secretory proteins released from neuroendocrine cells, with effects on control of blood pressure. We conducted a genome-wide association study for plasma catestatin, the catecholamine release inhibitory peptide derived from chromogranin A (CHGA), and other CHGA- or chromogranin B (CHGB)-related peptides, in 545 US and 1252 Australian subjects. This identified loci on chromosomes 4q35 and 5q34 affecting catestatin concentration (P = 3.40 × 10-30 for rs4253311 and 1.85 × 10-19 for rs2731672, respectively). Genes in these regions include the proteolytic enzymes kallikrein (KLKB1) and Factor XII (F12). In chromaffin cells, CHGA and KLKB1 proteins co-localized in catecholamine storage granules. In vitro, kallikrein cleaved recombinant human CHGA to catestatin, verified by mass spectrometry. The peptide identified from this digestion (CHGA360-373) selectively inhibited nicotinic cholinergic stimulated catecholamine release from chromaffin cells. A proteolytic cascade involving kallikrein and Factor XII cleaves chromogranins to active compounds both in vivo and in vitro.


Assuntos
Biomarcadores/metabolismo , Catecolaminas/metabolismo , Células Cromafins/metabolismo , Cromogranina A/sangue , Loci Gênicos/genética , Hipertensão/genética , Fragmentos de Peptídeos/sangue , Adolescente , Glândulas Suprarrenais/metabolismo , Adulto , Idoso , Animais , Austrália , Biomarcadores/análise , Células Cultivadas , Fator XII/genética , Fator XII/metabolismo , Feminino , Estudo de Associação Genômica Ampla , Humanos , Hipertensão/sangue , Calicreínas/genética , Calicreínas/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Ratos , Estados Unidos , Adulto Jovem
2.
J Neurochem ; 129(1): 48-59, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24266713

RESUMO

Chromogranin B (CHGB) is the major matrix protein in human catecholamine storage vesicles. CHGB genetic variation alters catecholamine secretion and blood pressure. Here, effective Chgb protein under-expression was achieved by siRNA in PC12 cells, resulting in ~ 48% fewer secretory granules on electron microscopy, diminished capacity for catecholamine uptake (by ~ 79%), and a ~ 73% decline in stores available for nicotinic cholinergic-stimulated secretion. In vivo, loss of Chgb in knockout mice resulted in a ~ 35% decline in chromaffin granule abundance and ~ 44% decline in granule diameter, accompanied by unregulated catecholamine release into plasma. Over-expression of CHGB was achieved by transduction of a CHGB-expressing lentivirus, resulting in ~ 127% elevation in CHGB protein, with ~ 122% greater abundance of secretory granules, but only ~ 14% increased uptake of catecholamines, and no effect on nicotinic-triggered secretion. Human CHGB protein and its proteolytic fragments inhibited nicotinic-stimulated catecholamine release by ~ 72%. One conserved-region CHGB peptide inhibited nicotinic-triggered secretion by up to ~ 41%, with partial blockade of cationic signal transduction. We conclude that bi-directional quantitative derangements in CHGB abundance result in profound changes in vesicular storage and release of catecholamines. When processed and released extra-cellularly, CHGB proteolytic fragments exert a feedback effect to inhibit catecholamine secretion, especially during nicotinic cholinergic stimulation.


Assuntos
Catecolaminas/metabolismo , Grânulos Cromafim/metabolismo , Cromogranina B/fisiologia , Líquido Extracelular/fisiologia , Líquido Intracelular/fisiologia , Sequência de Aminoácidos , Animais , Catecolaminas/genética , Grânulos Cromafim/genética , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Ratos
3.
Cell Mol Neurobiol ; 30(8): 1395-400, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21061160

RESUMO

Chromogranin A (CHGA) plays a fundamental role in the biogenesis of catecholamine secretory granules. Changes in storage and release of CHGA in clinical and experimental hypertension prompted us to study whether genetic variation at the CHGA locus might contribute to alterations in autonomic function, and hence hypertension and its target organ consequences such as hypertensive renal disease (nephrosclerosis). Systematic polymorphism discovery across the human CHGA locus revealed both common and unusual variants in both the open reading frame and such regulatory regions as the proximal promoter and 30-UTR. In chromaffin cell-transfected CHGA 30-UTR and promoter/luciferase reporter plasmids, the functional consequences of the regulatory/non-coding allelic variants were documented. Variants in both the proximal promoter and the 30-UTR displayed statistical associations with hypertension. Genetic variation in the proximal CHGA promoter predicted glomerular filtration rate in healthy twins. However, for hypertensive renal damage, both end-stage renal disease and rate of progression of earlier disease were best predicted by variants in the 30-UTR. Finally, mechanistic studies were undertaken initiated by the clue that CHGA promoter variation predicted circulating endothelin-1. In cultured endothelial cells, CHGA triggered co-release of not only the vasoconstrictor and pro-fibrotic endothelin-1, but also the pro-coagulant von Willebrand Factor and the pro-angiogenic angiopoietin-2. These findings, coupled with stimulation of endothelin-1 release from glomerular capillary endothelial cells by CHGA, suggest a plausible mechanism whereby genetic variation at the CHGA locus eventuates in alterations in human renal function. These results document the consequences of genetic variation at the CHGA locus for cardiorenal disease and suggest mechanisms whereby such variation achieves functional effects.


Assuntos
Cromogranina A/genética , Predisposição Genética para Doença , Variação Genética , Hipertensão Renal/genética , Humanos , Hipertensão Renal/fisiopatologia , Rim/fisiopatologia , Testes de Função Renal , Nefroesclerose/genética , Nefroesclerose/fisiopatologia , Fenótipo , Caracteres Sexuais
4.
Biochem Biophys Res Commun ; 386(2): 284-8, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19501042

RESUMO

Neurotoxic beta-amyloid (Abeta) peptides participate in Alzheimer's disease (AD); therefore, reduction of Abeta generated from APP may provide a therapeutic approach for AD. Gene knockout studies in transgenic mice producing human Abeta may identify targets for reducing Abeta. This study shows that knockout of the cathepsin B gene in mice expressing human wild-type APP (hAPPwt) results in substantial decreases in brain Abeta40 and Abeta42 by 67% and decreases in levels of the C-terminal beta-secretase fragment (CTFbeta) derived from APP. In contrast, knockout of cathepsin B in mice expressing hAPP with the rare Swedish (Swe) and Indiana (Ind) mutations had no effect on Abeta. The difference in reduction of Abeta in hAPPwt mice, but not in hAPPSwe/Ind mice, shows that the transgenic model can affect cathepsin B gene knockout results. Since most AD patients express hAPPwt, these data validate cathepsin B as a target for development of inhibitors to lower Abeta in AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Catepsina B/deficiência , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/genética , Animais , Catepsina B/genética , Técnicas de Inativação de Genes , Humanos , Camundongos , Camundongos Transgênicos , Mutação
5.
Science ; 364(6436)2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30975860

RESUMO

To understand the health impact of long-duration spaceflight, one identical twin astronaut was monitored before, during, and after a 1-year mission onboard the International Space Station; his twin served as a genetically matched ground control. Longitudinal assessments identified spaceflight-specific changes, including decreased body mass, telomere elongation, genome instability, carotid artery distension and increased intima-media thickness, altered ocular structure, transcriptional and metabolic changes, DNA methylation changes in immune and oxidative stress-related pathways, gastrointestinal microbiota alterations, and some cognitive decline postflight. Although average telomere length, global gene expression, and microbiome changes returned to near preflight levels within 6 months after return to Earth, increased numbers of short telomeres were observed and expression of some genes was still disrupted. These multiomic, molecular, physiological, and behavioral datasets provide a valuable roadmap of the putative health risks for future human spaceflight.


Assuntos
Adaptação Fisiológica , Astronautas , Voo Espacial , Imunidade Adaptativa , Peso Corporal , Artérias Carótidas/diagnóstico por imagem , Espessura Intima-Media Carotídea , Dano ao DNA , Metilação de DNA , Microbioma Gastrointestinal , Instabilidade Genômica , Humanos , Masculino , Homeostase do Telômero , Fatores de Tempo , Estados Unidos , United States National Aeronautics and Space Administration
6.
ACS Chem Biol ; 13(9): 2513-2521, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30085657

RESUMO

Proteases within the C1B hydrolase family are encoded by many organisms. We subjected a putative C1B-like cysteine protease secreted by the human gut commensal Parabacteroides distasonis to mass spectrometry-based substrate profiling to find preferred peptide substrates. The P. distasonis protease, which we termed Pd_dinase, has a sequential diaminopeptidase activity with strong specificity for N-terminal glycine residues. Using the substrate sequence information, we verified the importance of the P2 glycine residue with a panel of fluorogenic substrates and calculated kcat and KM for the dipeptide glycine-arginine-AMC. A potent and irreversible dipeptide inhibitor with a C-terminal acyloxymethyl ketone warhead, glycine-arginine- AOMK, was then synthesized and demonstrated that the Pd_dinase active site requires a free N-terminal amine for potent and rapid inhibition. We next determined the homohexameric Pd_dinase structure in complex with glycine-arginine- AOMK and uncovered unexpected active site features that govern the strict substrate preferences and differentiate this protease from members of the C1B and broader papain-like C1 protease families. We finally showed that Pd_dinase hydrolyzes several human antimicrobial peptides and therefore posit that this P. distasonis enzyme may be secreted into the extracellular milieu to assist in gut colonization by inactivation of host antimicrobial peptides.


Assuntos
Aminopeptidases/metabolismo , Peptídeos Catiônicos Antimicrobianos/metabolismo , Bacteroides/enzimologia , Microbioma Gastrointestinal , Glicina/metabolismo , Aminopeptidases/química , Peptídeos Catiônicos Antimicrobianos/química , Bacteroides/química , Bacteroides/metabolismo , Glicina/química , Humanos , Modelos Moleculares , Multimerização Proteica , Proteólise , Especificidade por Substrato
7.
BioDrugs ; 20(2): 105-19, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16626168

RESUMO

The nervous system represents a key area for development of novel therapeutic agents for the treatment of neurological and neurodegenerative diseases. Recent research has demonstrated the critical importance of neuroproteases for the production of specific peptide neurotransmitters and for the production of toxic peptides in major neurodegenerative diseases that include Alzheimer, Huntington, and Parkinson diseases. This review illustrates the successful criteria that have allowed identification of proteases responsible for converting protein precursors into active peptide neurotransmitters, consisting of dual cysteine protease and subtilisin-like protease pathways in neuroendocrine cells. These peptide neurotransmitters are critical regulators of neurologic conditions, including analgesia and cognition, and numerous behaviors. Importantly, protease pathways also represent prominent mechanisms in neurodegenerative diseases, especially Alzheimer, Huntington, and Parkinson diseases. Recent studies have identified secretory vesicle cathepsin B as a novel beta-secretase for production of the neurotoxic beta-amyloid (Abeta) peptide of Alzheimer disease. Moreover, inhibition of cathepsin B reduces Abeta peptide levels in brain. These neuroproteases potentially represent new drug targets that should be explored in future pharmaceutical research endeavors for drug discovery.


Assuntos
Desenho de Fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Neuropeptídeos/metabolismo , Neurotransmissores , Peptídeo Hidrolases/metabolismo , Inibidores de Proteases , Animais , Humanos , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/enzimologia , Sistema Nervoso/metabolismo , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/metabolismo , Neuropeptídeos/biossíntese , Neurotransmissores/farmacologia , Neurotransmissores/uso terapêutico , Peptídeo Hidrolases/genética , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico
8.
J Biol Chem ; 2004 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-15316004

RESUMO

This study demonstrates utilization of the novel GTG initiation codon for translation of a human mRNA transcript that encodes the serpin endopin 2B, a protease inhibitor. Molecular cloning revealed the nucleotide sequence of the human endopin 2B cDNA. Its deduced primary sequence shows high homology to bovine endopin 2A that possesses cross-class protease inhibition of elastase and papain. Notably, the human endopin 2B cDNA sequence revealed GTG as the predicted translation initiation codon; the predicted translation product of 46 kDa endopin 2B was produced by in vitro translation of 35S-endopin 2B with mammalian (rabbit) protein translation components. Importantly, bioinformatic studies demonstrated the presence of the entire human endopin 2B cDNA sequence with GTG as initiation codon within the human genome on chromosome 14. Further evidence for GTG as a functional initiation codon was illustrated by GTG-mediated in vitro translation of the heterologous protein EGFP, and by GTG-mediated expression of EGFP in mammalian PC12 cells. Mutagenesis of GTG to GTC resulted in the absence of EGFP expression in PC12 cells, indicating the function of GTG as an initiation codon. In addition, it was apparent that the GTG initiation codon produces lower levels of translated protein compared to ATG as initiation codon. Significantly, GTG-mediated translation of endopin 2B demonstrates a functional human gene product not previously predicted from initial analyses of the human genome. Further analyses based on GTG as an alternative initiation codon may predict new candidate genes of the human genome.

9.
Ann N Y Acad Sci ; 971: 426-44, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12438161

RESUMO

Endopin 1 and endopin 2 represent two novel serpin protease inhibitors localized within chromaffin granules, secretory vesicles of adrenomedullary chromaffin cells that represent a model neuroendocrine cell for synthesis and secretion of peptide neurotransmitters. This chapter describes the molecular features of the primary sequences of endopin 1 and endopin 2 that provided prediction of their distinct target protease specificities. Endopin 1 inhibits trypsin that cleaves at basic residues. In contrast, endopin 2 possesses cross-class inhibition of papain and elastase that represent cysteine and serine proteases, respectively. Cell biological studies indicate that endopin 1 and endopin 2 are localized within chromaffin granules. These results implicate endopin 1 inhibition in vivo of trypsin-like proteases in secretory vesicles, and endopin 2 inhibition of papain- or elastase-like proteases. Indeed, endopin 2 inhibits the endogenous cysteine protease PTP (prohormone thiol protease), present in chromaffin granules, that participates in the proteolytic processing of proenkephalin. These findings indicate the presence of endogenous endopin 1 and endopin 2 in secretory vesicle function.


Assuntos
Células Cromafins/metabolismo , Serpinas/metabolismo , Serpinas/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Relação Dose-Resposta a Droga , Humanos , Cinética , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
10.
Peptides ; 24(5): 717-25, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12895658

RESUMO

The regulation of cellular levels of adrenocorticotropin hormone (ACTH) in response to stimulated secretion was investigated to define the extent of cellular depletion of ACTH and subsequent increases to replenish ACTH levels in anterior pituitary cells (in primary culture). Treatment of cells with secretagogues for short-term incubation times (hours) resulted in extensive depletion of cellular ACTH. Corticotropin releasing factor (CRF) induced depletion of cellular levels of ACTH by 60-70% of control levels. The CRF-induced reduction of cellular ACTH was inhibited by the glucocorticoid dexamethasone. Phorbol myristate acetate (PMA), which stimulates protein kinase C (PKC), reduced ACTH levels by 50-60%. Forskolin, a stimulator of cAMP production, produced a moderate reduction in cellular ACTH. During prolonged incubation of cells (2 days) with these secretagogues, further reduction of ACTH levels by 70-80% was observed. However, increased cellular levels of ACTH occurred with continued treatment of cells with secretagogues, which provided nearly complete replenishment of cellular ACTH after 5 days treatment with secretagogues. Notably, the rising levels of cellular ACTH were inhibited by the aspartyl protease inhibitor acetyl-pepstatin A, and by the cysteine protease inhibitor E64d. These results demonstrate that depletion and recovery of ACTH levels are coordinately regulated, and that the increases in cellular levels of ACTH during the recovery phase involves participation of aspartyl and cysteine proteases. Thus, aspartyl and cysteine proteases may be involved in the cellular metabolism of ACTH.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Cisteína Endopeptidases/metabolismo , Adeno-Hipófise/metabolismo , Hormônio Adrenocorticotrópico/biossíntese , Animais , Colforsina/farmacologia , Hormônio Liberador da Corticotropina/farmacologia , AMP Cíclico/fisiologia , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Adeno-Hipófise/efeitos dos fármacos , Proteína Quinase C/fisiologia , Ratos , Acetato de Tetradecanoilforbol/farmacologia , Regulação para Cima
11.
Peptides ; 23(8): 1409-18, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12182941

RESUMO

The regulation of cellular levels of alpha-melanocyte stimulating factor (alpha-MSH) and beta-endorphin in response to stimulated secretion from intermediate pituitary cells in primary culture was investigated in this study. Regulation of the cell content of alpha-MSH and beta-endorphin occurred in two phases consisting of (a) initial depletion of cellular levels of these peptide hormones during short-term secretion (3 h) induced by isoproterenol, forskolin, or phorbol myristate acetate (PMA) which was followed by (b) long-term (24 h) increases in cellular levels of alpha-MSH and beta-endorphin in response to stimulated secretion induced by isoproterenol and PMA. In short-term experiments (3 h), cellular levels of alpha-MSH and beta-endorphin were reduced by 30-50% during stimulated secretion of these peptide hormones by isoproterenol (agonist for the beta-adrenergic receptor), forskolin that activates protein kinase A (PKA), and PMA that activates protein kinase C (PKC). Moreover, dopamine inhibited isoproterenol-induced depletion of cellular alpha-MSH and beta-endorphin. During long-term incubation of cells (24 h) with isoproterenol, cellular alpha-MSH and beta-endorphin were increased to twice that of controls (unstimulated cells). Treatment with PMA for 24 h also increased cellular levels of alpha-MSH and beta-endorphin. Moreover, cellular levels of alpha-MSH and beta-endorphin were decreased during long-term treatment of cells with an aspartyl protease inhibitor, pepstatin A, and with the cysteine protease inhibitor E64c. These results implicate aspartyl and cysteine proteases in the cellular production of alpha-MSH and beta-endorphin that requires proteolytic processing of their common precursor proopiomelanocortin (POMC). These findings demonstrate the parallel regulation of cellular levels of alpha-MSH and beta-endorphin during their cosecretion, which may involve aspartyl and cysteine proteases in the metabolism of these peptide hormones.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Cisteína Endopeptidases/metabolismo , Hipófise/metabolismo , alfa-MSH/metabolismo , beta-Endorfina/metabolismo , Animais , Colforsina/farmacologia , Dopamina/metabolismo , Isoproterenol/farmacologia , Masculino , Hipófise/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Acetato de Tetradecanoilforbol/farmacologia , alfa-MSH/efeitos dos fármacos , beta-Endorfina/efeitos dos fármacos
12.
Neuropeptides ; 37(3): 140-8, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12860111

RESUMO

The prohormone convertase 2 (PC2) is hypothesized to convert multiple pro-neuropeptides into active peptides that function as neurotransmitters. To examine the in vivo role of PC2 in neuropeptide production, the tissue contents of six different neuropeptides in brain and peripheral nervous tissues were examined in PC2 deficient mice. Specific neuropeptide radioimmunoassays and RP-HPLC (reverse-phase HPLC) provided evaluation of processed, active neuropeptides in brain and neuroendocrine tissues of PC2 deficient mice. Results demonstrated three features with regard to the selective roles of PC2 in determining the production of NPY, somatostatin-28, enkephalin, VIP, galanin, and CRF in neuroendocrine tissues. Firstly, PC2 deficient mice showed changes in several neuropeptides, but not all neuropeptides examined. The absence of active PC2 resulted in altered cellular levels of NPY, somatostatin-28, and (Met)enkephalin; few changes in VIP or galanin occurred in the tissues examined. CRF content was not altered in brains of PC2 deficient mice. Secondly, comparison of a single neuropeptide among different tissues of PC2 deficient mice demonstrated tissue-selective roles for PC2 in production of the neuropeptide. For example, NPY levels were decreased in ileum of PC2 deficient mice, but NPY content was not altered in hypothalamus that is abundant in NPY. In addition, (Met)enkephalin levels in hypothalamus and cortex were decreased in PC2 deficient mice, but no changes were observed in adrenal or intestine. Thirdly, a single tissue region often showed selective alterations among different neuropeptides. For example, the neuropeptide-rich hypothalamus region showed decreased (Met)enkephalin in PC2 deficient mice, but NPY, VIP, galanin, and CRF were not altered. These results demonstrate the selective role of PC2 in neuropeptide production that provides active peptide neurotransmitter or hormones for biological functions in brain and neuroendocrine systems.


Assuntos
Encéfalo/metabolismo , Neuropeptídeos/metabolismo , Sistemas Neurossecretores/metabolismo , Subtilisinas/fisiologia , Animais , Hormônio Liberador da Corticotropina/metabolismo , Encefalina Metionina/metabolismo , Galanina/metabolismo , Camundongos , Camundongos Knockout , Neuropeptídeo Y/metabolismo , Especificidade de Órgãos , Pró-Proteína Convertase 2 , Precursores de Proteínas/metabolismo , Radioimunoensaio , Somatostatina/metabolismo , Subtilisinas/deficiência , Subtilisinas/genética , Peptídeo Intestinal Vasoativo/metabolismo
13.
J Hypertens ; 30(10): 1961-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22871890

RESUMO

BACKGROUND: Cathepsin L (CTSL1) catalyzes the formation of peptides that influence blood pressure (BP). Naturally occurring genetic variation or targeted ablation of the Ctsl1 locus in mice yield cardiovascular pathology. Here, we searched for genetic variation across the human CTSL1 locus and probed its functional effects, especially in the proximal promoter. METHODS AND RESULTS: Systematic polymorphism discovery by re-sequencing across CTSL1 in 81 patients uncovered 38 genetic variants, five of which were relatively common (MAF >5%), creating a single linkage disequilibrium block in multiple biogeographic ancestries. One of these five common variants lay in a functional domain of the gene: promoter C-171A (rs3118869), which disrupts a predicted xenobiotic response element (XRE; match C>A). In transfected CTSL1 promoter/luciferase reporter plasmids, C-171A allele influenced transcription (C>A, P = 3.36E-6), and transcription was also augmented by co-exposure to the aryl hydrocarbon receptor (AHR) complex (AHR:ARNT) in the presence of their ligand dioxin (P = 6.81E-8); allele (C vs. A) and AHR:ARNT/dioxin stimulus interacted to control gene expression (interaction P = 0.033). Endogenous Ctsl1, Ahr, and Arnt transcripts were present in chromaffin cells. Promoter functional C-171A genotype also predicted hypertension (P = 1.0E-3), SBP (P = 4.0E-4), and DBP (P = 3.0E-3), in an additive pattern for diploid genotypes (A/A > C/A > C/C) in 868 patients, and the results were extended by validation analysis into an independent population sample of 986 patients. CONCLUSION: We conclude that common genetic variation in the proximal CTSL1 promoter, especially at position C-171A, is functional in cells, and alters transcription so as to explain the association of CTSL1 with BP in vivo. At the XRE, endogenous genetic variation plus exogenous aryl hydrocarbon stimulation interact to control CTSL1 gene expression. These results unveil a novel control point whereby heredity and environment can intersect to control a complex trait, and point to new transcriptional strategies for intervention into transmitter biosynthesis and its cardiovascular consequences.


Assuntos
Pressão Sanguínea/genética , Catepsina L/genética , Interação Gene-Ambiente , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Transcrição Gênica , Xenobióticos/farmacologia , Humanos
14.
J Biol Chem ; 283(12): 7745-53, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18184658

RESUMO

Elucidation of Abeta-lowering agents that inhibit processing of the wild-type (WT) beta-secretase amyloid precursor protein (APP) site, present in most Alzheimer disease (AD) patients, is a logical approach for improving memory deficit in AD. The cysteine protease inhibitors CA074Me and E64d were selected by inhibition of beta-secretase activity in regulated secretory vesicles that produce beta-amyloid (Abeta). The regulated secretory vesicle activity, represented by cathepsin B, selectively cleaves the WT beta-secretase site but not the rare Swedish mutant beta-secretase site. In vivo treatment of London APP mice, expressing the WT beta-secretase site, with these inhibitors resulted in substantial improvement in memory deficit assessed by the Morris water maze test. After inhibitor treatment, the improved memory function was accompanied by reduced amyloid plaque load, decreased Abeta40 and Abeta42, and reduced C-terminal beta-secretase fragment derived from APP by beta-secretase. However, the inhibitors had no effects on any of these parameters in mice expressing the Swedish mutant beta-secretase site of APP. The notable efficacy of these inhibitors to improve memory and reduce Abeta in an AD animal model expressing the WT beta-secretase APP site present in the majority of AD patients provides support for CA074Me and E64d inhibitors as potential AD therapeutic agents.


Assuntos
Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Catepsina B/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Leucina/análogos & derivados , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Catepsina B/genética , Bovinos , Inibidores de Cisteína Proteinase/uso terapêutico , Dipeptídeos/uso terapêutico , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Expressão Gênica , Humanos , Leucina/farmacologia , Leucina/uso terapêutico , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos
15.
J Biol Chem ; 283(17): 11807-22, 2008 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-18299326

RESUMO

Secretogranin II (SgII) belongs to the granin family of prohormones widely distributed in dense-core secretory granules (DCGs) of endocrine, neuroendocrine, and neuronal cells, including sympathoadrenal chromaffin cells. The mechanisms by which secretory proteins, and granins in particular, are sorted into the regulated secretory pathway are unsettled. We designed a strategy based on novel chimeric forms of human SgII fused to fluorescent (green fluorescent protein) or chemiluminescent (embryonic alkaline phosphatase) reporters to identify trafficking determinants mediating DCG targeting of SgII in sympathoadrenal cells. Three-dimensional deconvolution fluorescence microscopy and secretagogue-stimulated release studies demonstrate that SgII chimeras are correctly targeted to DCGs and released by exocytosis in PC12 and primary chromaffin cells. Results from a Golgi-retained mutant form of SgII suggest that sorting of SgII into DCGs depends on a saturable sorting machinery at the trans-Golgi/trans-Golgi network. Truncation analyses reveal the presence of DCG-targeting signals within both the N- and C-terminal regions of SgII, with the putative alpha-helix-containing SgII-(25-41) and SgII-(334-348) acting as sufficient, independent sorting domains. This study defines sequence features of SgII mediating vesicular targeting in sympathoadrenal cells and suggests a mechanism by which discrete domains of the molecule function in sorting, perhaps by virtue of a particular arrangement in tertiary structure and/or interaction with a specific component of the DCG membrane.


Assuntos
Regulação da Expressão Gênica , Secretogranina II/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Membrana Celular/metabolismo , Exocitose , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células PC12 , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/química , Transdução de Sinais
16.
Arch Biochem Biophys ; 461(2): 219-24, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17451636

RESUMO

The serpin endopin 2A inhibits the cysteine protease papain in cross-class inhibition. This study demonstrates the novel finding that both the non-RSL NH(2)-domain and the RSL domain with P1-P1' residues participate in endopin 2A inhibition. Production of a chimeric mutant of endopin 2A with replacement of its NH(2)-domain with that of endopin 1 resulted in less effective inhibition of papain, indicated by its lower k(ass) association rate constant compared to wild-type endopin 2A. This chimeric mutant formed complexes with papain, but at lower levels compared to that with wild-type endopin 2A. Papain degradation of a portion of the chimeric mutant suggested a role for the NH(2)-domain in regulating relative amounts of endopin 2A that enter the substrate pathway compared to the serpin inhibitory pathway. Furthermore, site-directed mutagenesis demonstrated that the RSL domain with intact P1-P1' residues was necessary for inhibition. These findings indicate that the NH(2)-domain and the RSL region both participate in endopin 2A inhibition of papain.


Assuntos
Papaína/antagonistas & inibidores , Serpinas/química , Serpinas/fisiologia , Animais , Sequência de Bases , Sítios de Ligação/genética , Bovinos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Papaína/química , Estrutura Terciária de Proteína/genética , Serpinas/genética
17.
Biol Chem ; 388(9): 979-83, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17696783

RESUMO

Beta-secretase inhibitors that lower brain beta-amyloid peptides (Abeta) are likely to be effective for treating Alzheimer's disease (AD). Irreversible epoxysuccinyl cysteine protease inhibitors are known to reduce brain Abeta and beta-secretase activity in the guinea pig model of human Abeta production. In this study, acetyl-L-leucyl-L-valyl-L-lysinal (Ac-LVK-CHO) is also shown to significantly reduce brain Abeta and beta-secretase activity and brain Abeta in the same model. Ac-LVK-CHO is structurally distinct from the epoxysuccinyl inhibitors and is a reversible cysteine protease inhibitor. The results suggest that cysteine protease inhibitors generally, and reversible cysteine protease inhibitors specifically, have potential for development as AD therapeutics.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Catepsina B/metabolismo , Catepsinas/metabolismo , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Oligopeptídeos/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Animais , Encéfalo/efeitos dos fármacos , Catepsina L , Inibidores de Cisteína Proteinase/química , Cobaias , Humanos , Oligopeptídeos/química
18.
Nat Protoc ; 2(5): 1248-53, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17546020

RESUMO

This protocol describes the primary culture of individual chromaffin cells derived by enzymatic digestion from the adrenal medulla of the bovine adrenal gland. Since the late 1970s, such cells have provided a useful model system to study neurotransmitter biosynthesis, storage and release in the catecholaminergic system. The protocol can be divided into three stages: isolation of cells (4-6 h), determination of viable cell numbers (approximately 30 min) and growth in culture (3-7 d). An alternative procedure is to perform studies in a continuous chromaffin (pheochromocytoma) cell line, such as PC12, although such transformed cells are typically less highly differentiated than primary cells. The bovine chromaffin cell procedure should yield approximately 10-20 million cells, suitable for several experiments over the subsequent 3-7 d. Typical experiments involve transmitter biosynthesis, vesicular storage, exocytotic release, stimulus coupling (signal transduction) toward secretion or transcription, or morphology, including ultrastructure. The total time, from adrenal gland harvest until functional experiments, is typically 4-8 d.


Assuntos
Glândulas Suprarrenais/citologia , Técnicas de Cultura de Células/métodos , Células Cromafins/citologia , Animais , Bovinos
19.
Biol Chem ; 387(10-11): 1429-39, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17081116

RESUMO

Proteases are required for the production of peptide neurotransmitters and toxic peptides in neurodegenerative diseases. Unique roles of the cysteine proteases cathepsin L and cathepsin B in secretory vesicles for the production of biologically active peptides have been demonstrated in recent studies. Secretory vesicle cathepsin L participates in the proteolytic conversion of proenkephalin into the active enkephalin, an opioid peptide neurotransmitter that mediates pain relief. Moreover, recent findings provide evidence that cathepsin B in regulated secretory vesicles participates in the production of toxic beta-amyloid peptides that are known to accumulate extracellularly in Alzheimer's disease brains. The neurobiological functions of cathepsins L and B demonstrate that these secretory vesicle cysteine proteases produce biologically active peptides. These results demonstrate newly identified roles for cathepsins L and B in neurosecretory vesicles in the production of biologically active peptides.


Assuntos
Catepsina B/metabolismo , Catepsinas/metabolismo , Cisteína Endopeptidases/metabolismo , Doenças Neurodegenerativas/metabolismo , Peptídeos/metabolismo , Vesículas Secretórias/metabolismo , Transdução de Sinais , Animais , Catepsina L , Humanos
20.
Cell Mol Neurobiol ; 26(4-6): 449-69, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16724274

RESUMO

1. Recent research demonstrates the critical importance of neuroproteases for the production of peptide neurotransmitters, and for the production of toxic peptides in major neurodegenerative diseases that include Alzheimer's (AD) and Huntington's diseases. This review describes the strategies utilized to identify the appropriate proteases responsible for producing active peptides for neurotransmission, with application of such approaches for defining protease mechanisms in neurodegenerative diseases. 2. Integration of multidisciplinary approaches in neurobiology, biochemistry, chemistry, proteomics, molecular biology, and genetics has been utilized for neuroprotease studies. These investigations have identified secretory vesicle cathepsin L for the production of the enkephalin opioid peptide neurotransmitter and other neuropeptides. Furthermore, new results using these strategies have identified secretory vesicle cathepsin B for the production of beta-amyloid (Abeta) in the major regulated secretory pathway that provides activity-dependent secretion of Abeta peptides, which accumulate in AD. 3. CNS neuroproteases that participate in peptide neurotransmission and in neurodegenerative diseases represent new candidate drug targets that may be explored in future research for the development of novel therapeutic agents for neurological conditions.


Assuntos
Doenças Neurodegenerativas/etiologia , Neuropeptídeos/biossíntese , Peptídeo Hidrolases/fisiologia , Transmissão Sináptica/fisiologia , Animais , Humanos , Redes e Vias Metabólicas , Modelos Biológicos , Doenças Neurodegenerativas/terapia , Processamento de Proteína Pós-Traducional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA