Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34006633

RESUMO

Neovascularization is a key feature of ischemic retinal diseases and the wet form of age-related macular degeneration (AMD), all leading causes of severe vision loss. Vascular endothelial growth factor (VEGF) inhibitors have transformed the treatment of these disorders. Millions of patients have been treated with these drugs worldwide. However, in real-life clinical settings, many patients do not experience the same degree of benefit observed in clinical trials, in part because they receive fewer anti-VEGF injections. Therefore, there is an urgent need to discover and identify novel long-acting VEGF inhibitors. We hypothesized that binding to heparan-sulfate proteoglycans (HSPG) in the vitreous, and possibly other ocular structures, may be a strategy to promote intraocular retention, ultimately leading to a reduced burden of intravitreal injections. We designed a series of VEGF receptor 1 variants and identified some with strong heparin-binding characteristics and ability to bind to vitreous matrix. Our data indicate that some of our variants have longer duration and greater efficacy in animal models of intraocular neovascularization than current standard of care. Our study represents a systematic attempt to exploit the functional diversity associated with heparin affinity of a VEGF receptor.


Assuntos
Neovascularização de Coroide/tratamento farmacológico , Proteoglicanas de Heparan Sulfato/farmacologia , Degeneração Macular/tratamento farmacológico , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Inibidores da Angiogênese/química , Animais , Proliferação de Células/efeitos dos fármacos , Neovascularização de Coroide/genética , Neovascularização de Coroide/patologia , Cristalografia por Raios X , Células Endoteliais/efeitos dos fármacos , Olho/efeitos dos fármacos , Olho/patologia , Proteoglicanas de Heparan Sulfato/genética , Proteoglicanas de Heparan Sulfato/imunologia , Heparina/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/farmacologia , Fragmentos Fc das Imunoglobulinas/ultraestrutura , Injeções Intravítreas , Degeneração Macular/genética , Degeneração Macular/patologia , Camundongos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/ultraestrutura , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Corpo Vítreo/efeitos dos fármacos
2.
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
3.
BMC Med Genet ; 17: 21, 2016 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-26969407

RESUMO

BACKGROUND: Plasma coagulation Factor XIIa (Hageman factor; encoded by F12) and kallikrein (KAL or Fletcher factor; encoded by KLKB1) are proteases of the kallikerin-kinin system involved in converting the inactive circulating prorenin to renin. Renin is a key enzyme in the formation of angiotensin II, which regulates blood pressure, fluid and electrolyte balance and is a biomarker for cardiovascular, metabolic and renal function. The renin-angiotensin system is implicated in extinction learning in posttraumatic stress disorder. METHODS & RESULTS: Active plasma renin was measured from two independent cohorts- civilian twins and siblings, as well as U.S. Marines, for a total of 1,180 subjects. Genotyping these subjects revealed that the carriers of the minor alleles at the two loci- F12 and KLKB1 had a significant association with reduced levels of active plasma renin. Meta-analyses confirmed the association across cohorts. In vitro studies verified digestion of human recombinant pro-renin by kallikrein (KAL) to generate active renin. Subsequently, the active renin was able to digest the synthetic substrate angiotensinogen to angiotensin-I. Examination of mouse juxtaglomerular cell line and mouse kidney sections showed co-localization of KAL with renin. Expression of either REN or KLKB1 was regulated in cell line and rodent models of hypertension in response to oxidative stress, interleukin or arterial blood pressure changes. CONCLUSIONS: The functional variants of KLKB1 (rs3733402) and F12 (rs1801020) disrupted the cascade of enzymatic events, resulting in diminished formation of active renin. Using genetic, cellular and molecular approaches we found that conversion of zymogen prorenin to renin was influenced by these polymorphisms. The study suggests that the variant version of protease factor XIIa due to the amino acid substitution had reduced ability to activate prekallikrein to KAL. As a result KAL has reduced efficacy in converting prorenin to renin and this step of the pathway leading to activation of renin affords a potential therapeutic target.


Assuntos
Fator XIIa/genética , Calicreínas/genética , Polimorfismo de Nucleotídeo Único , Sistema Renina-Angiotensina/genética , Renina/sangue , Adolescente , Adulto , Idoso , Alelos , Angiotensina I/sangue , Angiotensinogênio/sangue , Animais , Pressão Sanguínea , Proteínas de Ciclo Celular , Linhagem Celular , Regulação da Expressão Gênica , Loci Gênicos , Estudo de Associação Genômica Ampla , Técnicas de Genotipagem , Humanos , Hipertensão/genética , Sistema Justaglomerular/citologia , Calicreínas/sangue , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Pré-Calicreína/metabolismo , Renina/genética , Serina Endopeptidases/metabolismo , Transferases , Adulto Jovem
4.
Hum Mol Genet ; 22(18): 3624-40, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23674521

RESUMO

Hypertension is a common hereditary syndrome with unclear pathogenesis. Chromogranin A (Chga), which catalyzes formation and cargo storage of regulated secretory granules in neuroendocrine cells, contributes to blood pressure homeostasis centrally and peripherally. Elevated Chga occurs in spontaneously hypertensive rat (SHR) adrenal glands and plasma, but central expression is unexplored. In this report, we measured SHR and Wistar-Kyoto rat (control) Chga expression in central and peripheral nervous systems, and found Chga protein to be decreased in the SHR brainstem, yet increased in the adrenal and the plasma. By re-sequencing, we systematically identified five promoter, two coding and one 3'-untranslated region (3'-UTR) polymorphism at the SHR (versus WKY or BN) Chga locus. Using HXB/BXH recombinant inbred (RI) strain linkage and correlations, we demonstrated genetic determination of Chga expression in SHR, including a cis-quantitative trait loci (QTLs) (i.e. at the Chga locus), and such expression influenced biochemical determinants of blood pressure, including a cascade of catecholamine biosynthetic enzymes, catecholamines themselves and steroids. Luciferase reporter assays demonstrated that the 3'-UTR polymorphism (which disrupts a microRNA miR-22 motif) and promoter polymorphisms altered gene expression consistent with the decline in SHR central Chga expression. Coding region polymorphisms did not account for changes in Chga expression or function. Thus, we hypothesized that the 3'-UTR and promoter mutations lead to dysregulation (diminution) of Chga in brainstem cardiovascular control nuclei, ultimately contributing to the pathogenesis of hypertension in SHR. Accordingly, we demonstrated that in vivo administration of miR-22 antagomir to SHR causes substantial (∼18 mmHg) reductions in blood pressure, opening a novel therapeutic avenue for hypertension.


Assuntos
Cromogranina A/genética , Cromogranina A/metabolismo , Hipertensão/genética , MicroRNAs/genética , Regiões Promotoras Genéticas , Regiões 3' não Traduzidas , Glândulas Suprarrenais/metabolismo , Animais , Pressão Sanguínea/genética , Tronco Encefálico/metabolismo , Linhagem Celular Tumoral , Cromogranina A/sangue , Cromogranina A/química , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Ligação Genética , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , MicroRNAs/metabolismo , Células PC12 , Polimorfismo Genético , Estrutura Secundária de Proteína , Locos de Características Quantitativas , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Alinhamento de Sequência , Transcrição Gênica
5.
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
6.
J Biol Chem ; 287(27): 23141-51, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22535963

RESUMO

Chromogranin A knock-out (Chga-KO) mice display increased adiposity despite high levels of circulating catecholamines and leptin. Consistent with diet-induced obese mice, desensitization of leptin receptors caused by hyperleptinemia is believed to contribute to the obese phenotype of these KO mice. In contrast, obesity in ob/ob mice is caused by leptin deficiency. To characterize the metabolic phenotype, Chga-KO mice were treated with the CHGA-derived peptide catestatin (CST) that is deficient in these mice. CST treatment reduced fat depot size and increased lipolysis and fatty acid oxidation. In liver, CST enhanced oxidation of fatty acids as well as their assimilation into lipids, effects that are attributable to the up-regulation of genes promoting fatty acid oxidation (Cpt1α, Pparα, Acox, and Ucp2) and incorporation into lipids (Gpat and CD36). CST did not affect basal or isoproterenol-stimulated cAMP production in adipocytes but inhibited phospholipase C activation by the α-adrenergic receptor (AR) agonist phenylephrine, suggesting inhibition of α-AR signaling by CST. Indeed, CST mimicked the lipolytic effect of the α-AR blocker phentolamine on adipocytes. Moreover, CST reversed the hyperleptinemia of Chga-KO mice and improved leptin signaling as determined by phosphorylation of AMPK and Stat3. CST also improved peripheral leptin sensitivity in diet-induced obese mice. In ob/ob mice, CST enhanced leptin-induced signaling in adipose tissue. In conclusion, our results implicate CST in a novel pathway that promotes lipolysis and fatty acid oxidation by blocking α-AR signaling as well as by enhancing leptin receptor signaling.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Fármacos Antiobesidade/farmacologia , Cromogranina A/farmacologia , Leptina/metabolismo , Obesidade/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Receptores Adrenérgicos alfa/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Fármacos Antiobesidade/metabolismo , Catecolaminas/metabolismo , Cromogranina A/genética , Cromogranina A/metabolismo , Ácidos Graxos/sangue , Ácidos Graxos/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Lipólise/efeitos dos fármacos , Lipólise/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/genética , Obesidade/metabolismo , Fragmentos de Peptídeos/metabolismo , Cultura Primária de Células , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
7.
J Neurochem ; 127(6): 750-61, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23786442

RESUMO

The Syrian Cardiomyopathic Hamster (BIO-14.6/53.58 strains) model of cardiac failure, resulting from naturally occurring deletion at the SGCD (delta-sarcoglycan) locus, displays widespread disturbances in catecholamine metabolism. Rare Mendelian myopathy disorders of human SGCD occur, although common naturally occurring SGCD genetic variation has not been evaluated for effects on human norepinephrine (NE) secretion. This study investigated the effect of SGCD genetic variation on control of NE secretion in healthy twin pairs. Genetic associations profiled SNPs across the SGCD locus. Trait heritability (h(2)) and genetic covariance (pleiotropy; shared h(2)) were evaluated. Sympathochromaffin exocytosis in vivo was probed in plasma by both catecholamines and Chromogranin B (CHGB). Plasma NE is substantially heritable (p = 3.19E-16, at 65.2 ± 5.0% of trait variance), sharing significant (p < 0.05) genetic determination with circulating and urinary catecholamines, CHGB, eGFR, and several cardio-metabolic traits. Participants with higher pNE showed significant (p < 0.05) differences in several traits, including increased BP and hypertension risk factors. Peak SGCD variant rs1835919 predicted elevated systemic vascular compliance, without changes in specifically myocardial traits. We used a chimeric-regulated secretory pathway photoprotein (CHGA-EAP) to evaluate the effect of SGCD on the exocytotic pathway in transfected PC12 cells; in transfected cells, expression of SGCD augmented CHGA trafficking into the exocytotic regulated secretory pathway. Thus, our investigation determined human NE secretion to be a highly heritable trait, influenced by common genetic variation within the SGCD locus. Circulating NE aggregates with BP and hypertension risk factors. In addition, coordinate NE and CHGB elevation by rs1835919 implicates exocytosis as the mechanism of release.


Assuntos
Loci Gênicos , Padrões de Herança , Polimorfismo de Nucleotídeo Único , Sarcoglicanas/genética , Sistema Nervoso Simpático/fisiologia , Adolescente , Adulto , Idoso , Animais , Cromogranina A/metabolismo , Exocitose , Pleiotropia Genética , Humanos , Pessoa de Meia-Idade , Norepinefrina/sangue , Norepinefrina/metabolismo , Células PC12 , Transporte Proteico , Locos de Características Quantitativas , Característica Quantitativa Herdável , Ratos , Sarcoglicanas/metabolismo , Adulto Jovem
8.
Bioorg Med Chem ; 21(18): 5855-69, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23916146

RESUMO

The endogenous catecholamine release-inhibitory peptide catestatin (CST) regulates events leading to hypertension and cardiovascular disease. Earlier we studied the structure of CST by NMR, molecular modeling, and amino acid scanning mutagenesis. That structure has now been exploited for elucidation of interface pharmacophores that mediate binding of CST to its target, with consequent secretory inhibition. Designed pharmacophore models allowed screening of 3D structural domains. Selected compounds were tested on both cultured catecholaminergic cells and an in vivo model of hypertension; in each case, the candidates showed substantial mimicry of native CST actions, with preserved or enhanced potency and specificity. The approach and compounds have thus enabled rational design of novel drug candidates for treatment of hypertension or autonomic dysfunction.


Assuntos
Anti-Hipertensivos/química , Catecolaminas/metabolismo , Cromogranina A/química , Fragmentos de Peptídeos/química , Sequência de Aminoácidos , Animais , Anti-Hipertensivos/farmacologia , Anti-Hipertensivos/uso terapêutico , Cálcio/metabolismo , Cromogranina A/farmacologia , Cromogranina A/uso terapêutico , Desenho de Fármacos , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Modelos Moleculares , Células PC12 , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Ratos
9.
Neuropsychopharmacology ; 47(13): 2252-2260, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35347268

RESUMO

Identification of biomarkers for psychiatric disorders remains very challenging due to substantial symptom heterogeneity and diagnostic comorbidity, limiting the ability to map symptoms to underlying neurobiology. Dimensional symptom clusters, such as anhedonia, hyperarousal, etc., are complex and arise due to interactions of a multitude of complex biological relationships. The primary aim of the current investigation was to use multi-set canonical correlation analysis (mCCA) to derive biomarkers (biochemical, physiological) linked to dimensional symptoms across the anxiety and depressive spectrum. Active-duty service members (N = 2,592) completed standardized depression, anxiety and posttraumatic stress questionnaires and several psychophysiological and biochemical assays. Using this approach, we identified two phenotype associations between distinct physiological and biological phenotypes. One was characterized by symptoms of dysphoric arousal (anhedonia, anxiety, hypervigilance) which was associated with low blood pressure and startle reactivity. This finding is in line with previous studies suggesting blunted physiological reactivity is associated with subpopulations endorsing anxiety with comorbid depressive features. A second phenotype of anxious fatigue (high anxiety and reexperiencing/avoidance symptoms coupled with fatigue) was associated with elevated blood levels of norepinephrine and the inflammatory marker C-reactive protein in conjunction with high blood pressure. This second phenotype may describe populations in which inflammation and high sympathetic outflow might contribute to anxious fatigue. Overall, these findings support the growing consensus that distinct neuropsychiatric symptom patterns are associated with differential physiological and blood-based biological profiles and highlight the potential of mCCA to reveal important psychiatric symptom biomarkers from several psychophysiological and biochemical measures.


Assuntos
Anedonia , Transtornos de Estresse Pós-Traumáticos , Humanos , Transtornos de Estresse Pós-Traumáticos/psicologia , Ansiedade/psicologia , Biomarcadores , Fadiga , Depressão/psicologia
10.
EMBO Mol Med ; 14(1): e14511, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34779136

RESUMO

In the course of our studies aiming to discover vascular bed-specific endothelial cell (EC) mitogens, we identified leukemia inhibitory factor (LIF) as a mitogen for bovine choroidal EC (BCE), although LIF has been mainly characterized as an EC growth inhibitor and an anti-angiogenic molecule. LIF stimulated growth of BCE while it inhibited, as previously reported, bovine aortic EC (BAE) growth. The JAK-STAT3 pathway mediated LIF actions in both BCE and BAE cells, but a caspase-independent proapoptotic signal mediated by cathepsins was triggered in BAE but not in BCE. LIF administration directly promoted activation of STAT3 and increased blood vessel density in mouse eyes. LIF also had protective effects on the choriocapillaris in a model of oxidative retinal injury. Analysis of available single-cell transcriptomic datasets shows strong expression of the specific LIF receptor in mouse and human choroidal EC. Our data suggest that LIF administration may be an innovative approach to prevent atrophy associated with AMD, through protection of the choriocapillaris.


Assuntos
Atrofia Geográfica , Fator Inibidor de Leucemia , Mitógenos , Animais , Corioide/irrigação sanguínea , Corioide/metabolismo , Células Endoteliais/metabolismo , Atrofia Geográfica/metabolismo , Janus Quinases/metabolismo , Fator Inibidor de Leucemia/metabolismo , Fator Inibidor de Leucemia/farmacologia , Camundongos , Mitógenos/metabolismo , Mitógenos/farmacologia , Fator de Transcrição STAT3/metabolismo
11.
Curr Hypertens Rep ; 13(1): 36-45, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21104344

RESUMO

Hypertension is a complex trait with deranged autonomic control of the circulation. The sympathoadrenal system exerts minute-to-minute control over cardiac output and vascular tone. Catecholamine storage vesicles (or chromaffin granules) of the adrenal medulla contain remarkably high concentrations of chromogranins/secretogranins (or "granins"), catecholamines, neuropeptide Y, adenosine triphosphate (ATP), and Ca(2+). Within secretory granules, granins are co-stored with catecholamine neurotransmitters and co-released upon stimulation of the regulated secretory pathway. The principal granin family members, chromogranin A (CHGA), chromogranin B (CHGB), and secretogranin II (SCG2), may have evolved from shared ancestral exons by gene duplication. This article reviews human genetic variation at loci encoding the major granins and probes the effects of such polymorphisms on blood pressure, using twin pairs to probe heritability and individuals with the most extreme blood pressure values in the population to study hypertension.


Assuntos
Catecolaminas/metabolismo , Cromogranina A/genética , Cromogranina B/genética , Hipertensão/genética , Polimorfismo Genético/genética , Secretogranina II/genética , Análise de Variância , Catecolaminas/genética , Distribuição de Qui-Quadrado , Cromograninas/genética , Cromograninas/metabolismo , Intervalos de Confiança , Progressão da Doença , Feminino , Variação Genética , Genótipo , Humanos , Hipertensão/metabolismo , Hipertensão/patologia , Masculino , Razão de Chances , Fatores de Risco
12.
BMC Nephrol ; 12: 27, 2011 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-21679467

RESUMO

BACKGROUND: Renal kallikrein (KLK1) synthesis and urinary excretion are reportedly diminished during AKI (acute kidney injury) in animal models, and provision of kallikrein abrogates renal injury in this setting, but data in human AKI is limited. Therefore we first examined KLK1 renal excretion in human AKI, and then probed potential endocrine and epigenetic mechanisms for its alterations. METHODS: KLK1 enzymatic activity excretion was evaluated in urine from patients with established or incipient AKI, versus healthy/non-hospital as well as ICU controls. Endocrine control of KLK1 excretion was then probed by catecholamine and aldosterone measurements in established AKI versus healthy controls. To examine epigenetic control of KLK1 synthesis, we tested blood and urine DNA for changes in promoter CpG methylation of the KLK1 gene, as well as LINE-1 elements, by bisulfite sequencing. RESULTS: Patients with early/incipient AKI displayed a modest reduction of KLK1 excretion, but unexpectedly, established AKI displayed substantially elevated urine KLK1 excretion, ~11-fold higher than healthy controls, and ~3-fold greater than ICU controls. We then probed potential mechanisms of the change. Established AKI patients had lower SBP, higher heart rate, and higher epinephrine excretion than healthy controls, though aldosterone excretion was not different. Promoter KLK1 CpG methylation was higher in blood than urine DNA, while KLK1 methylation in blood DNA was significantly higher in established AKI than healthy controls, though KLK1 methylation in urine tended to be higher in AKI, directionally consistent with earlier/incipient but not later/established changes in KLK1 excretion in AKI. On multivariate ANOVA, AKI displayed coordinate changes in KLK1 excretion and promoter methylation, though directionally opposite to expectation. Control (LINE-1 repetitive element) methylation in blood and urine DNA was similar between AKI and controls. CONCLUSIONS: Unexpectedly, increased KLK1 excretion in AKI patients was found; this increase is likely to be due in part to increments in adrenergic tone during BP depression. Epigenetic changes at KLK1 may also play a role in early changes of KLK1 expression and thus AKI susceptibility or recovery.


Assuntos
Injúria Renal Aguda/enzimologia , Injúria Renal Aguda/genética , Epigênese Genética , Regulação Enzimológica da Expressão Gênica , Calicreínas/genética , Calicreínas/urina , Adulto , Idoso , Sequência de Bases , Biomarcadores/urina , Estudos de Coortes , Epigênese Genética/genética , Feminino , Seguimentos , Humanos , Unidades de Terapia Intensiva , Calicreínas/biossíntese , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Estudos Prospectivos , Regulação para Cima/genética
13.
Physiol Genomics ; 41(1): 91-101, 2010 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-20009010

RESUMO

Chromogranin A (CHGA) plays a catalytic role in formation of catecholamine storage vesicles and also serves as precursor to the peptide fragment catestatin, a catecholamine secretory inhibitor whose expression is diminished in the hypertensive individuals. We previously reported the hypertensive, hyperadrenergic phenotype of Chga-/- knockout (KO) mice and rescue by the human ortholog. In the present study, we compare two humanized CHGA mouse models. Into the Chga null background, by bacterial artificial chromosome transgenesis human CHGA transgene has been introduced. Both lines have the complete approximately 12 kbp CHGA gene integrated stably in the genome but have substantial differences in CHGA expression, as well as consequent sympathochromaffin biochemistry and physiology. A mouse model with longer-insert HumCHGA31 displays integration encompassing not only CHGA but also long human flanking sequences. This is in contrast to mouse model HumCHGA19 with limited flanking human sequence co-integrated. As a consequence, HumCHGA19 mice have normal though diminished pattern of spatial expression of CHGA, and 14-fold lower circulating CHGA, with failure to rescue KO phenotypes to normalcy. In the longer-insert HumCHGA31 mice, catecholamine secretion, exaggerated responses to environmental stress, and hypertension were all alleviated. Promoter regions of the transgenes in both HumCHGA19 and HumCHGA31 display minimal CpG methylation, weighing against differential "position effects" of integration, and thus suggesting that lack of cis elements required for optimal CHGA expression occurs in HumCHGA19 mice. Such "humanized" CHGA mouse models may be useful in probing the physiological consequences of variation in CHGA expression found in humans, with consequences for susceptibility to hypertension and cardiovascular disease.


Assuntos
Cromogranina A/genética , Cromossomos Artificiais Bacterianos/genética , Cromossomos Humanos Par 14/genética , Glândulas Suprarrenais/patologia , Animais , Sequência de Bases , Pressão Sanguínea , Catecolaminas/metabolismo , Cromogranina A/deficiência , Cromogranina A/metabolismo , Dosagem de Genes , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Mutagênese Insercional/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Restrição Física , Estresse Fisiológico , Transgenes/genética
14.
J Biol Chem ; 284(42): 28498-509, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19706599

RESUMO

Chromogranin A (CHGA/Chga), a proprotein, widely distributed in endocrine and neuroendocrine tissues (not expressed in muscle, liver, and adipose tissues), generates at least four bioactive peptides. One of those peptides, pancreastatin (PST), has been reported to interfere with insulin action. We generated a Chga knock-out (KO) mouse by the targeted deletion of the Chga gene in neuroendocrine tissues. KO mice displayed hypertension, higher plasma catecholamine, and adipokine levels and lower IL-6 and lipid levels compared with wild type mice. Liver glycogen content was elevated, but the nitric oxide (NO) level was diminished. Glucose, insulin, and pyruvate tolerance tests and hyperinsulinemic-euglycemic clamp studies established increased insulin sensitivity in liver but decreased glucose disposal in muscle. Despite higher catecholamine and ketone body levels and muscle insulin resistance, KO mice maintained euglycemia due to increased liver insulin sensitivity. Suppressed mRNA abundance of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase (G6Pase) in KO mice further support this conclusion. PST administration in KO mice stimulated phosphoenolpyruvate carboxykinase and G6Pase mRNA abundance and raised the blood glucose level. In liver cells transfected with G6Pase promoter, PST caused transcriptional activation in a protein kinase C (PKC)- and NO synthase-dependent manner. Thus, PST action may be mediated by suppressing IRS1/2-phosphatidylinositol 3-kinase-Akt-FOXO-1 signaling and insulin-induced maturation of SREBP1c by PKC and a high level of NO. The combined effects of conventional PKC and endothelial NO synthase activation by PST can suppress insulin signaling. The rise in blood PST level with age and in diabetes suggests that PST is a negative regulator of insulin sensitivity and glucose homeostasis.


Assuntos
Cromogranina A/genética , Cromogranina A/fisiologia , Glucose/metabolismo , Insulina/metabolismo , Hormônios Pancreáticos/fisiologia , Adipócitos/metabolismo , Animais , Composição Corporal , Homeostase , Lipídeos/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/química , Hormônios Pancreáticos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais
15.
Cell Tissue Res ; 342(3): 353-61, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21052719

RESUMO

In the heart, the secretory granules containing the atrial natriuretic peptides (ANP) and B-type myocardial natriuretic peptide (BNP) provide the basis for the endocrine function of this organ. We sought to determine whether atrial and myocardial secretory granules contain chromogranin/secretogranin proteins including chromogranin A (CHGA/Chga), chromogranin B (CHGB/Chgb) and secretogranin II (SCG2/Scg2). Deconvolution microscopy on immunolabeled proteins revealed the presence of Chga, Chgb, and Scg2 in murine cardiac secretory granules. The presence of low plasma catestatin (CST: mChga(364-384)) in older mice indicates diminished processing of Chga to CST with advancement of age, which is comparable to that found in humans. We have previously shown that CST (hCHGA(352-372)) exerts potent cardio-suppressive effects on frog and rat heart, but the source of CST for such action has remained elusive. In the present study, we found CST-related peptides in cardiomyocytes and in heart, which establishes an autocrine/paracrine function of CST in cardiac tissue. We conclude that cardiac secretory granules contain Chga, Chgb and Scg2 and that Chga is processed to CST in murine heart.


Assuntos
Cromogranina A/metabolismo , Miócitos Cardíacos/metabolismo , Fragmentos de Peptídeos/metabolismo , Secretogranina II/metabolismo , Vesículas Secretórias/metabolismo , Fatores Etários , Animais , Células Cultivadas , Cromogranina A/sangue , Cromogranina B/sangue , Eletroforese em Gel de Poliacrilamida , Imunofluorescência , Humanos , Immunoblotting , Camundongos , Miócitos Cardíacos/ultraestrutura , Secretogranina II/sangue
16.
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
17.
J Hypertens ; 38(3): 456-466, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31714338

RESUMO

BACKGROUND: Chromogranin A (CHGA) is an index granin protein critical for biogenesis and exocytotic release of catecholamine storage granules. It is elevated in plasma of patients with sympathetic over-activity and kidney dysfunction. Several CHGA polymorphisms are associated with hypertensive kidney disease. Previously, we unraveled the molecular mechanism by which CHGA expression is regulated in African Americans carrying a genetic variation associated with hypertensive chronic kidney disease (CKD). METHOD: Experimental CKD mouse model were created by 5/6th nephrectomy (Npx) using wild-type and Chga-/- knockout mouse strains to delineate the role of CHGA in CKD. RESULT: Wild-type-Npx mice expressing Chga developed exacerbated azotemia and fibrosis as compared with their knockout-Npx counterparts. Gene expression profiling revealed downregulation of mitochondrial respiratory complexes genes consistent with maladaptive mitochondria in wild-type-Npx mice, contrasted to knockout-Npx. In healthy individuals, an inverse relationship between circulating CHGA levels and glomerular function was observed. In vitro, mesangial cells treated with CHGA-triggered nitric oxide release by a signaling mechanism involving scavenger receptor SR-A. The CHGA-treated and untreated mesangial cells displayed differential expression of cytokine, chemokine, complement, acute phase inflammatory and apoptotic pathway genes. Thus, build-up of plasma CHGA because of kidney injury served as an insult to the mesangial cells resulting in expression of genes promoting inflammation, fibrosis, and progression of CKD. CONCLUSION: These findings improve understanding of the role of elevated CHGA in the progression of CKD and reveal novel pathways that could be exploited for therapeutic strategies in hypertensive kidney disease.


Assuntos
Cromogranina A , Hipertensão Renal , Nefrite , Animais , Cromogranina A/genética , Cromogranina A/metabolismo , Hipertensão Renal/genética , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Camundongos , Camundongos Knockout , Nefrite/genética , Nefrite/metabolismo , Nefrite/patologia
19.
Sci Rep ; 9(1): 5055, 2019 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-30911067

RESUMO

The intra-renal dopamine (DA) system is highly expressed in the proximal tubule and contributes to Na+ and blood pressure homeostasis, as well as to the development of nephropathy. In the kidney, the enzyme DOPA Decarboxylase (DDC) originating from the circulation. We used a twin/family study design, followed by polymorphism association analysis at DDC locus to elucidate heritable influences on renal DA production. Dense single nucleotide polymorphism (SNP) genotyping across the DDC locus on chromosome 7p12 was analyzed by re-sequencing guided by trait-associated genetic markers to discover the responsible genetic variation. We also characterized kinetics of the expressed DDC mutant enzyme. Systematic polymorphism screening across the 15-Exon DDC locus revealed a single coding variant in Exon-14 that was associated with DA excretion and multiple other renal traits indicating pleiotropy. When expressed and characterized in eukaryotic cells, the 462Gln variant displayed lower Vmax (maximal rate of product formation by an enzyme) (21.3 versus 44.9 nmol/min/mg) and lower Km (substrate concentration at which half-maximal product formation is achieved by an enzyme.)(36.2 versus 46.8 µM) than the wild-type (Arg462) allele. The highly heritable DA excretion trait is substantially influenced by a previously uncharacterized common coding variant (Arg462Gln) at the DDC gene that affects multiple renal tubular and glomerular traits, and predicts accelerated functional decline in chronic kidney disease.

20.
Endocrinology ; 149(2): 749-57, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17991725

RESUMO

The plasma level of chromogranin A (CgA) is elevated in genetic hypertension. Conversely, the plasma level of the CgA peptide catestatin is diminished in individuals with established hypertension and those with a genetic risk of this disease. Resequencing of the human CHGA gene identified three naturally occurring variants of catestatin (Gly364Ser, Pro370Leu, and Arg374Gln) that exhibit different potencies in inhibiting catecholamine secretion. Here, we have examined whether there is any differential processing of the three CHGA variants to catestatin by the endoproteolytic enzyme plasmin. Plasmin digestion of the purified CgA proteins generated a stable biologically active 14-amino acid peptide (human CgA(360-373)) from the wild-type, Gly364Ser, and Arg374Gln proteins despite the disruption of the dibasic site (Arg(373)Arg(374)) in the Arg374Gln variant. Unexpectedly, the action of plasmin in generating the catestatin peptide from the Pro370Leu protein was less efficient. The efficiency of cleavage at the dibasic Arg(373) downward arrowArg(374) site in synthetic human CgA(360-380) was 3- to 4-fold less in Pro370Leu CgA, compared with the wild type. Circular dichroism of the synthetic CgA(352-372) suggested a difference in the amount of alpha-helix and beta-sheet between the wild-type and Pro370Leu CgA peptides. Because the Pro(370) residue is in the P4 position, the local secondary structure in the vicinity of the cleavage site may enforce the specificity or accessibility to plasmin. The less efficient proteolytic processing of the Pro370Leu protein by plasmin, coupled with the strong association of this variant with ethnicity, suggests that the Pro370Leu CHGA gene variant may contribute to the differential prevalence of cardiovascular disease across ethnic groups.


Assuntos
Cromogranina A/genética , Cromogranina A/metabolismo , Fibrinolisina/metabolismo , Variação Genética , Hipertensão/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Catecolaminas/metabolismo , Cromogranina A/química , Humanos , Hipertensão/epidemiologia , Dados de Sequência Molecular , Células PC12 , Fragmentos de Peptídeos/química , Mutação Puntual , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Risco , Análise Espectral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA