Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Publication year range
1.
Hum Mol Genet ; 22(18): 3624-40, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23674521

ABSTRACT

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.


Subject(s)
Chromogranin A/genetics , Chromogranin A/metabolism , Hypertension/genetics , MicroRNAs/genetics , Promoter Regions, Genetic , 3' Untranslated Regions , Adrenal Glands/metabolism , Animals , Blood Pressure/genetics , Brain Stem/metabolism , Cell Line, Tumor , Chromogranin A/blood , Chromogranin A/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genetic Linkage , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/physiopathology , Male , MicroRNAs/metabolism , PC12 Cells , Polymorphism, Genetic , Protein Structure, Secondary , Quantitative Trait Loci , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sequence Alignment , Transcription, Genetic
2.
J Biol Chem ; 287(19): 15232-41, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22393040

ABSTRACT

Proteases are required for processing precursors into active neuropeptides that function as neurotransmitters for cell-cell communication. This study demonstrates the novel function of human cathepsin V protease for producing the neuropeptides enkephalin and neuropeptide Y (NPY). Cathepsin V is a human-specific cysteine protease gene. Findings here show that expression of cathepsin V in neuroendocrine PC12 cells and human neuronal SK-N-MC cells results in production of (Met)enkephalin from proenkephalin. Gene silencing of cathepsin V by siRNA in human SK-N-MC cells results in reduction of (Met)enkephalin by more than 80%, illustrating the prominent role of cathepsin V for neuropeptide production. In vitro processing of proenkephalin by cathepsin V occurs at dibasic residue sites to generate enkephalin-containing peptides and an ∼24-kDa intermediate present in human brain. Cathepsin V is present in human brain cortex and hippocampus where enkephalin and NPY are produced and is present in purified human neuropeptide secretory vesicles. Colocalization of cathepsin V with enkephalin and NPY in secretory vesicles of human neuroblastoma cells was illustrated by confocal microscopy. Furthermore, expression of cathepsin V with proNPY results in NPY production. These findings indicate the unique function of human cathepsin V for producing enkephalin and NPY neuropeptides required for neurotransmission in health and neurological diseases.


Subject(s)
Cathepsins/metabolism , Cysteine Endopeptidases/metabolism , Enkephalins/metabolism , Neuropeptide Y/metabolism , Neurotransmitter Agents/metabolism , Aged , Amino Acid Sequence , Animals , Blotting, Western , Cathepsins/genetics , Cell Line, Tumor , Cerebral Cortex/enzymology , Chromaffin Granules/enzymology , Cysteine Endopeptidases/genetics , Enkephalins/genetics , Hippocampus/enzymology , Humans , Male , Microscopy, Confocal , Molecular Sequence Data , PC12 Cells , Protein Precursors/genetics , Protein Precursors/metabolism , RNA Interference , Rats , Transfection
3.
J Neurochem ; 127(6): 750-61, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23786442

ABSTRACT

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.


Subject(s)
Genetic Loci , Inheritance Patterns , Polymorphism, Single Nucleotide , Sarcoglycans/genetics , Sympathetic Nervous System/physiology , Adolescent , Adult , Aged , Animals , Chromogranin A/metabolism , Exocytosis , Genetic Pleiotropy , Humans , Middle Aged , Norepinephrine/blood , Norepinephrine/metabolism , PC12 Cells , Protein Transport , Quantitative Trait Loci , Quantitative Trait, Heritable , Rats , Sarcoglycans/metabolism , Young Adult
4.
J Biol Chem ; 285(13): 10030-10043, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20061385

ABSTRACT

Processes underlying the formation of dense core secretory granules (DCGs) of neuroendocrine cells are poorly understood. Here, we present evidence that DCG biogenesis is dependent on the secretory protein secretogranin (Sg) II, a member of the granin family of pro-hormone cargo of DCGs in neuroendocrine cells. Depletion of SgII expression in PC12 cells leads to a decrease in both the number and size of DCGs and impairs DCG trafficking of other regulated hormones. Expression of SgII fusion proteins in a secretory-deficient PC12 variant rescues a regulated secretory pathway. SgII-containing dense core vesicles share morphological and physical properties with bona fide DCGs, are competent for regulated exocytosis, and maintain an acidic luminal pH through the V-type H(+)-translocating ATPase. The granulogenic activity of SgII requires a pH gradient along this secretory pathway. We conclude that SgII is a critical factor for the regulation of DCG biogenesis in neuroendocrine cells, mediating the formation of functional DCGs via its pH-dependent aggregation at the trans-Golgi network.


Subject(s)
Catecholamines/metabolism , Secretogranin II/metabolism , Secretory Vesicles/metabolism , Animals , COS Cells , Chlorocebus aethiops , Chromaffin Granules/metabolism , Gene Silencing , Genetic Vectors , Hydrogen-Ion Concentration , Neuroendocrine Cells/metabolism , PC12 Cells , RNA, Small Interfering/metabolism , Rats , Recombinant Fusion Proteins/metabolism
5.
Cell Tissue Res ; 345(1): 87-102, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21597914

ABSTRACT

Pituitary adenylyl cyclase activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) augment the biosynthesis of tyrosine hydroxylase (TH). We tested whether secretin belonging to the glucagon/PACAP/VIP superfamily would increase transcription of the tyrosine hydroxylase (Th) gene and modulate catecholamine secretion. Secretin activated transcription of the endogenous Th gene and its transfected promoter (EC(50) ∼4.6 nM) in pheochromocytoma (PC12) cells. This was abolished by pre-treatment with a secretin receptor (SCTR) antagonist and by inhibition of protein kinase A (PKA), mitogen-activated protein kinase, or CREB (cAMP response element-binding protein). In agreement, secretin increased PKA activity and induced phosphorylation of CREB and binding to Th CRE, suggesting secretin signaling to transcription via a PKA-CREB pathway. Secretin stimulated catecholamine secretion (EC(50) ∼3.5 µM) from PC12 cells, but this was inhibited by pre-treatment with VIP-preferring receptor (VPAC1)/PACAP-preferring receptor (PAC1) antagonists. Secretin-evoked secretion occurred without extracellular Ca(2+) and was abolished by intracellular Ca(2+) chelation. Secretin augmented phospholipase C (PLC) activity and increased inositol-1,4,5-triphosphate (IP(3)) levels in PC12 cells; PLC-ß inhibition blocked secretin-induced catecholamine secretion, indicating the participation of intracellular Ca(2+) from a phospholipase pathway in secretion. Like PACAP, secretin evoked long-lasting catecholamine secretion, even after only a transient exposure. Thus, transcription is triggered by nanomolar concentrations of the peptide through SCTR, with signaling along the cAMP-PKA and extracellular-signal-regulated kinase 1/2 pathways and through CREB. By contrast, secretion is triggered only by micromolar concentrations of peptide through PAC1/VPAC receptors and by utilizing a PLC/intracellular Ca(2+) pathway.


Subject(s)
Catecholamines/biosynthesis , Catecholamines/metabolism , Secretin/pharmacology , Animals , Calcium/pharmacology , Calcium Channels/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/biosynthesis , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Mitogen-Activated Protein Kinases/metabolism , PC12 Cells , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Rats , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Type C Phospholipases/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
6.
J Proteome Res ; 9(10): 5065-75, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20704348

ABSTRACT

Neuropeptides are required for cell-cell communication in the regulation of physiological and pathological processes. While selected neuropeptides of known biological activities have been studied, global analyses of the endogenous profile of human peptide products derived from prohormones by proteolytic processing in vivo are largely unknown. Therefore, this study utilized the global, unbiased approach of mass spectrometry-based neuropeptidomics to define peptide profiles in secretory vesicles, isolated from human adrenal medullary pheochromocytoma of the sympathetic nervous system. The low molecular weight pool of secretory vesicle peptides was subjected to nano-LC-MS/MS with ion trap and QTOF mass spectrometry analyzed by different database search tools (InsPecT and Spectrum Mill). Peptides were generated by processing of prohormones at dibasic cleavage sites as well as at nonbasic residues. Significantly, peptide profiling provided novel insight into newly identified peptide products derived from proenkephalin, pro-NPY, proSAAS, CgA, CgB, and SCG2 prohormones. Previously unidentified intervening peptide domains of prohormones were observed, thus providing new knowledge of human neuropeptidomes generated from precursors. The global peptidomic approach of this study demonstrates the complexity of diverse neuropeptides present in human secretory vesicles for cell-cell communication.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Mass Spectrometry/methods , Neuropeptides/metabolism , Pheochromocytoma/metabolism , Protein Precursors/metabolism , Proteomics/methods , Secretory Vesicles/metabolism , Adrenal Gland Neoplasms/pathology , Adrenal Medulla/metabolism , Adrenal Medulla/pathology , Amino Acid Sequence , Cell Communication , Chromaffin Granules/metabolism , Chromatography, Liquid , Enkephalins/metabolism , Humans , Molecular Sequence Data , Peptides/metabolism , Pheochromocytoma/pathology , Secretogranin II/metabolism
7.
Cell Mol Neurobiol ; 30(8): 1391-4, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20571875

ABSTRACT

The catecholamine biosynthetic pathway consists of several enzymatic steps in series, beginning with the amino acids phenylalanine and tyrosine, and eventuating in the catecholamines norepinephrine (noradrenaline) and epinephrine (adrenaline). Since the enzyme tyrosine hydroxylase (TH; tyrosine 3-mono-oxygenase; EC 1.14.16.2; chromosome 11p15.5) is generally considered to be rate-limiting in this pathway, probed as to whether common genetic variation at the TH gene occurred, and whether such variants contributed to inter-individual alterations in autonomic function, either biochemical or physiological. We began with sequencing a tetranucleotide (TCAT) repeat in the first intron, and found that the two most common versions, (TCAT)(6) and (TCAT)(10i), predicted heritable autonomic traits in twin pairs. We then conducted systematic polymorphism discovery across the ~8 kbp locus, and discovered numerous variants, principally non-coding. The proximal promoter block contained four common variants, and its haplotypes and SNPs (especially C-824T, rs10770141) predicted catecholamine secretion, environmental stress-induced BP increments, and hypertension. Finally, we found that two of the common promoter variants, C-824T (rs10770141) and A-581G (rs10770140), were functional in that they differentially affected transcriptional activity of the isolated promoter, disrupted recognition motifs for specific transcription factor binding, altered the promoter responses to the co-transfected (exogenous) factors, and bound the endogenous factors in the chromatin fraction of the nucleus. We concluded that common variation in the proximal TH promoter is functional, giving rise to changes in autonomic function and consequently cardiovascular risk.


Subject(s)
Alleles , Autonomic Nervous System/enzymology , Autonomic Nervous System/physiopathology , Genetic Variation , Hypertension/genetics , Hypertension/physiopathology , Tyrosine 3-Monooxygenase/genetics , Blood Pressure/genetics , Humans , Hypertension/enzymology , Microsatellite Repeats/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic
8.
Biochem J ; 418(1): 81-91, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18973469

ABSTRACT

In endocrine cells, prohormones and granins are segregated in the TGN (trans-Golgi network) from constitutively secreted proteins, stored in concentrated form in dense-core secretory granules, and released in a regulated manner on specific stimulation. The mechanism of granule formation is only partially understood. Expression of regulated secretory proteins, both peptide hormone precursors and granins, had been found to be sufficient to generate structures that resemble secretory granules in the background of constitutively secreting, non-endocrine cells. To identify which segment of CgA (chromogranin A) is important to induce the formation of such granule-like structures, a series of deletion constructs fused to either GFP (green fluorescent protein) or a short epitope tag was expressed in COS-1 fibroblast cells and analysed by fluorescence and electron microscopy and pulse-chase labelling. Full-length CgA as well as deletion constructs containing the N-terminal 77 residues generated granule-like structures in the cell periphery that co-localized with co-expressed SgII (secretogranin II). These are essentially the same segments of the protein that were previously shown to be required for granule sorting in wild-type PC12 (pheochromocytoma cells) cells and for rescuing a regulated secretory pathway in A35C cells, a variant PC12 line deficient in granule formation. The results support the notion that self-aggregation is at the core of granule formation and sorting into the regulated pathway.


Subject(s)
Chromogranin A/metabolism , Endocrine Cells/metabolism , Secretory Pathway , Secretory Vesicles/metabolism , Animals , Biomarkers , Cell Line , Chlorocebus aethiops , Chromogranin A/genetics , Endocrine Cells/ultrastructure , Epitopes/immunology , Gene Deletion , Mice , Microscopy, Immunoelectron , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
J Am Soc Nephrol ; 20(7): 1623-32, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19520754

ABSTRACT

Chromogranin A (CHGA), a protein released from secretory granules of chromaffin cells and sympathetic nerves, triggers endothelin-1 release from endothelial cells. CHGA polymorphisms associate with an increased risk for ESRD, but whether altered CHGA-endothelium interactions may explain this association is unknown. Here, CHGA led to the release of endothelin-1 and Weibel-Palade body exocytosis in cultured human umbilical vein endothelial cells. In addition, CHGA triggered secretion of endothelin-1 from glomerular endothelial cells and TGF-beta1 from mesangial cells cocultured with glomerular endothelial cells. In humans, plasma CHGA correlated positively with endothelin-1 and negatively with GFR. GFR was highly heritable in twin pairs, and common promoter haplotypes of CHGA predicted GFR. In patients with progressive hypertensive renal disease, a CHGA haplotype predicted rate of GFR decline. In conclusion, these data suggest that CHGA acts through the glomerular endothelium to regulate renal function.


Subject(s)
Chromogranin A/metabolism , Endothelium/metabolism , Exocytosis/physiology , Glomerular Filtration Rate/physiology , Kidney Glomerulus/metabolism , Weibel-Palade Bodies/metabolism , Animals , Cells, Cultured , Chromogranin A/genetics , Chromogranin A/pharmacology , Chronic Disease , Coculture Techniques , Dose-Response Relationship, Drug , Endothelins/metabolism , Endothelium/cytology , Endothelium/drug effects , Humans , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Kidney Glomerulus/cytology , Kidney Glomerulus/drug effects , Mice , Polymorphism, Genetic/genetics , Risk Factors , Time Factors , Transforming Growth Factor beta1/metabolism
10.
Circulation ; 118(3): 247-57, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18591442

ABSTRACT

BACKGROUND: Chromogranin A (CHGA) triggers catecholamine secretory granule biogenesis, and its catestatin fragment inhibits catecholamine release. We approached catestatin heritability using twin pairs, coupled with genome-wide linkage, in a series of twin and sibling pairs from 2 continents. METHODS AND RESULTS: Hypertensive patients had elevated CHGA coupled with reduction in catestatin, suggesting diminished conversion of precursor to catestatin. Heritability for catestatin in twins was 44% to 60%. Six hundred fifteen nuclear families yielded 870 sib pairs for linkage, with significant logarithm of odds peaks on chromosomes 4p, 4q, and 17q. Because acidification of catecholamine secretory vesicles determines CHGA trafficking and processing to catestatin, we genotyped at positional candidate ATP6N1, bracketed by peak linkage markers on chromosome 17q, encoding a subunit of vesicular H(+)-translocating ATPase. The minor allele diminished CHGA secretion and processing to catestatin. The ATP6N1 variant also influenced blood pressure in 1178 individuals with the most extreme blood pressure values in the population. In chromaffin cells, inhibition of H(+)-ATPase diverted CHGA from regulated to constitutive secretory pathways. CONCLUSIONS: We established heritability of catestatin in twins from 2 continents. Linkage identified 3 regions contributing to catestatin, likely novel determinants of sympathochromaffin exocytosis. At 1 such positional candidate (ATP6N1), variation influenced CHGA secretion and processing to catestatin, confirming the mechanism of a novel trans-QTL for sympathochromaffin activity and blood pressure.


Subject(s)
Chromogranin A/genetics , Genetic Linkage , Genome, Human , Peptide Fragments/genetics , Quantitative Trait, Heritable , Twins/genetics , Alleles , Australia , Blood Pressure/genetics , Chromaffin Cells/enzymology , Chromaffin Cells/metabolism , Chromogranin A/blood , Chromogranin A/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 17 , Environment , Exocytosis , Female , Genetic Variation , Humans , Hypertension/genetics , Male , Middle Aged , Peptide Fragments/blood , Proton-Translocating ATPases/metabolism , Siblings , United States
11.
J Pharmacol Exp Ther ; 331(2): 419-28, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19671882

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) are combinations of subunits arranged as pentamers encircling a central cation channel. At least nine alpha and four beta subunits are expressed in the central and peripheral nervous systems; their presence in autonomic ganglia, the adrenal medulla, and central nervous system, with accompanying responses elicited by nicotinic agonists, point to their involvement in cardiovascular homeostasis. nAChRs formed by alpha3, alpha5, and beta4 subunits may regulate blood pressure (BP) by mediating release of catestatin, the endogenous nicotinic antagonist fragment of chromogranin A (CHGA) and potent inhibitor of catecholamine secretion. Genes encoding these subunits (CHRNA3, CHRNA5, and CHRNB4) are clustered on human chromosome 15q24. Because variation in this cluster may alter autonomic regulation of BP, we sequenced approximately 15 kilobase pairs in 15q24 containing their coding and 5'- and 3'-untranslated regions in 80 individuals. We identified 63 variants: 25 in coding regions of CHRNA3, CHRNA5, and CHRNB4 and 48 noncoding single-nucleotide polymorphisms (SNPs). Haplotype frequencies varied across ethnic populations. We assessed the contribution of six SNPs in the putative catestatin binding region of CHRNA3 and CHRNB4 to autonomic traits. In twins, catestatin and BP were heritable. CHRNA3 SNPs and haplotypes containing K95K (G285A) associated with circulating plasma catestatin, epinephrine levels, as well as systolic BP, suggesting altered coupling of the nAChRs to BP. Studies of chromaffin cells in vitro reveal that nicotinic agonist stimulation releases catecholamines and CHGA, a process augmented by overexpression of CHRNA3 and blocked by catestatin. These cellular events suggest a homeostatic mechanism underlying the pleiotropic actions of CHRNA3 genetic variation on autonomic function observed in twins.


Subject(s)
Autonomic Nervous System/physiology , Chromosomes, Human, Pair 15/genetics , Neurons/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/physiology , Acetylcholinesterase/blood , Alleles , Catecholamines/blood , Chromogranin A/genetics , Chromogranins/genetics , Ethnicity , Exocytosis/drug effects , Gene Frequency , Haplotypes , Humans , Linkage Disequilibrium/genetics , Models, Molecular , Multigene Family , Neurons/physiology , Peptide Fragments/genetics , Phenotype , Phylogeny , Polymorphism, Single Nucleotide/genetics , Reverse Transcriptase Polymerase Chain Reaction , United States/epidemiology
12.
Biochemistry ; 47(27): 7167-78, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18549247

ABSTRACT

Secretion of proteins and peptides from eukaryotic cells takes place by both constitutive and regulated pathways. Regulated secretion may involve interplay of proteins that are currently unknown. Recent studies suggest an important role of chromogranin A (CHGA) in the regulated secretory pathway in neuroendocrine cells, but the mechanism by which CHGA enters the regulated pathway, or even triggers the formation of the pathway, remains unclear. In this study, we used a transcriptome/proteome-wide approach, to discover binding partners for CHGA, by employing a phage display cDNA library method. Several proteins within or adjacent to the secretory pathway were initially detected as binding partners of recombinant human CHGA. We then focused on the trans-Golgi protein SCLIP (STMN3) and its stathmin paralog SCG10 (STMN2) for functional study. Co-immunoprecipitation experiments confirmed the interaction of each of these two proteins with CHGA in vitro. SCLIP and SCG10 were colocalized to the Golgi apparatus of chromaffin cells in vivo and shared localization with CHGA as it transited the Golgi. Downregulation of either SCLIP or SCG10 by synthetic siRNAs virtually abolished chromaffin cell secretion of a transfected CHGA-EAP chimera (expressing CHGA fused to an enzymatic reporter, and trafficked to the regulated pathway). SCLIP siRNA also decreased the level of secretion of endogenous CHGA and SCG2, as well as transfected human growth hormone, while SCG10 siRNA decreased the level of regulated secretion of endogenous CHGB. Moreover, a dominant negative mutant of SCG10 (Cys 22,Cys 24-->Ala 22,Ala 24) significantly blocked secretion of the transfected CHGA-EAP chimera. A decrease in the buoyant density of chromaffin granules was observed after downregulation of SCG10 by siRNA, suggesting participation of these stathmins in granule formation or maturation. We conclude that SCLIP and SCG10 interact with CHGA, share partial colocalization in the Golgi apparatus, and may be necessary for typical transmitter storage and release from chromaffin cells.


Subject(s)
Chromogranin A/metabolism , Membrane Proteins/metabolism , Neurosecretory Systems/metabolism , Stathmin/metabolism , trans-Golgi Network/metabolism , Animals , Chromaffin Cells/metabolism , Chromaffin Granules/metabolism , Chromogranin B/metabolism , Down-Regulation , Gene Silencing , Genes, Dominant , Growth Hormone/metabolism , Humans , Immunoprecipitation , Intracellular Space/metabolism , Mutant Proteins/metabolism , PC12 Cells , Protein Binding , Protein Transport , RNA, Small Interfering/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Transfection
13.
Circulation ; 115(17): 2282-91, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17438153

ABSTRACT

BACKGROUND: Endothelial dysfunction predisposes to vascular injury in association with hypertension. Endothelin (ET-1) is a potent vasoactive peptide that is synthesized and released by the vascular endothelium and is a marker of endothelial function. Chromogranin A (CHGA) regulates the storage and release of catecholamines and may have direct actions on the microvasculature. CHGA, a candidate gene for intermediate phenotypes that contribute to hypertension, shows a pattern of single nucleotide polymorphism variations that alter the expression and function of this gene both in vivo and in vitro. METHODS AND RESULTS: In a study of twins (n=238 pairs), plasma ET-1 was 58+/-5% (P<0.0001) heritable. Plasma ET-1 was both correlated and associated with chromogranin fragment levels, and the 2 were influenced by shared genetic determination (pleiotropy [rhoG]; for the CHGA precursor, rhoG=0.318+/-0.105; P=0.0032). We therefore hypothesized that variation in the CHGA gene may influence ET-1 secretion. Carriers of the CHGA promoter -988G, -462A, and -89A minor alleles showed significantly higher mean plasma ET-1 than their major allele homozygote counterparts (P=0.02, P=0.006, P=0.03, respectively). Analysis of a linkage disequilibrium block that spans these 3 single nucleotide polymorphisms showed a significant association between the GATACA haplotype and plasma ET-1 (P=0.0075). In cultured human umbilical vein endothelial cells, CHGA caused dose-dependent secretion of ET-1 over a brief (<1 hour) time course at relatively low concentrations of CHGA (10 to 100 nmol/L) with a threshold concentration (10 nmol/L) in the range found circulating in humans in vivo. CONCLUSIONS: These results suggest that common, heritable variation in expression of the human CHGA gene influences endothelial ET-1 secretion in vivo, explained by a CHGA stimulus/ET-1 secretion coupling in endothelial cells in vitro. The findings document a previously unsuspected interaction between the sympathochromaffin system and the endothelium and suggest novel genetic and cell biological approaches to the prediction, diagnosis, and mechanism of endothelial dysfunction in human disease.


Subject(s)
Chromogranin A/genetics , Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Hypertension/genetics , Hypertension/metabolism , Adult , Chromogranin A/blood , Endothelin-1/blood , Female , Genetic Variation , Haplotypes , Humans , Male , Phenotype , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Sex Factors , Sympathetic Nervous System/physiology
14.
Circulation ; 116(9): 993-1006, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17698732

ABSTRACT

BACKGROUND: Tyrosine hydroxylase (TH) is the rate-limiting enzyme in catecholamine biosynthesis. Does common genetic variation at human TH alter autonomic activity and predispose to cardiovascular disease? We undertook systematic polymorphism discovery at the TH locus and then tested variants for contributions to sympathetic function and blood pressure. METHODS AND RESULTS: We resequenced 80 ethnically diverse individuals across the TH locus. One hundred seventy-two twin pairs were evaluated for sympathetic traits, including catecholamine production, reflex control of the circulation, and environmental (cold) stress responses. To evaluate hypertension, we genotyped subjects selected from the most extreme diastolic blood pressure percentiles in the population. Human TH promoter haplotype/reporter plasmids were transfected into chromaffin cells. Forty-nine single-nucleotide polymorphisms were discovered, but coding region polymorphism did not account for common phenotypic variation. A block of linkage disequilibrium spanned 4 common variants in the proximal promoter. Catecholamine secretory traits were significantly heritable (h2), as were stress-induced blood pressure changes. In the TH promoter, significant associations were found for urinary catecholamine excretion and for blood pressure response to stress. TH promoter haplotype 2 (TGGG) showed pleiotropy, increasing both norepinephrine excretion and blood pressure during stress. Coalescent simulations suggest that TH haplotype 2 likely arose approximately 380,000 years ago. In hypertension, 2 independent case-control studies (1266 subjects with 53% women and 927 subjects with 24% women) replicated the effect of C-824T in the determination of blood pressure. CONCLUSIONS: We conclude that human catecholamine secretory traits are heritable, displaying joint genetic determination (pleiotropy) with autonomic activity and finally with blood pressure in the population. Catecholamine secretion is influenced by genetic variation in the adrenergic pathway encoding catecholamine synthesis, especially at the classically rate-limiting step, TH. The results suggest novel pathophysiological links between a key adrenergic locus, catecholamine metabolism, and blood pressure and suggest new strategies to approach the mechanism, diagnosis, and treatment of systemic hypertension.


Subject(s)
Autonomic Nervous System/physiology , Blood Pressure , Cardiovascular Diseases/genetics , Catecholamines/biosynthesis , Transcription, Genetic , Tyrosine 3-Monooxygenase/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Algorithms , Cardiovascular Diseases/epidemiology , Genetic Predisposition to Disease , Genetic Variation , Humans , Kinetics , Middle Aged , Polymorphism, Single Nucleotide , Twins, Dizygotic , Twins, Monozygotic
15.
Endocrinology ; 149(2): 749-57, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17991725

ABSTRACT

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.


Subject(s)
Chromogranin A/genetics , Chromogranin A/metabolism , Fibrinolysin/metabolism , Genetic Variation , Hypertension/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Catecholamines/metabolism , Chromogranin A/chemistry , Humans , Hypertension/epidemiology , Molecular Sequence Data , PC12 Cells , Peptide Fragments/chemistry , Point Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Risk Factors , Spectrum Analysis
16.
Endocrinology ; 148(9): 4310-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17540723

ABSTRACT

The prostanoid biosynthetic enzyme cyclooxygenase-2 (Cox-2) is up-regulated in several neuroendocrine tumors. The aim of the current study was to employ a neuroendocrine cell (PC12) model of Cox-2 overexpression to identify gene products that might be implicated in the oncogenic and/or inflammatory actions of this enzyme in the setting of neuroendocrine neoplasia. Expression array and real-time PCR analysis demonstrated that levels of the neuroendocrine marker chromogranin A (CGA) were 2- and 3.2-fold higher, respectively, in Cox-2 overexpressing cells (PCXII) vs. their control (PCMT) counterparts. Immunocytochemical and immunoblotting analyses confirmed that both intracellular and secreted levels of CGA were elevated in response to Cox-2 induction. Moreover, exogenous addition of prostaglandin E(2) (1 microm) mimicked this effect in PCMT cells, whereas treatment of PCXII cells with the Cox-2 selective inhibitor NS-398 (100 nm) reduced CGA expression levels, thereby confirming the biospecificity of this finding. Levels of neuron-specific enolase were similar in the two cell lines, suggesting that the effect of Cox-2 on CGA expression was specific and not due to a global enhancement of neuroendocrine marker expression/differentiation. Cox-2-dependent CGA up-regulation was associated with significantly increased chromaffin granule number and intracellular and secreted levels of dopamine. CGA promoter-driven reporter gene expression studies provided evidence that prostaglandin E(2)-dependent up-regulation required a proximal cAMP-responsive element (-71 to -64 bp). This study is the first to demonstrate that Cox-2 up-regulates both CGA expression and bioactivity in a neuroendocrine cell line and has major implications for the role of this polypeptide in the pathogenesis of neuroendocrine cancers in which Cox-2 is up-regulated.


Subject(s)
Chromogranin A/genetics , Chromogranin A/metabolism , Cyclooxygenase 2/genetics , Dinoprostone/pharmacology , Dinoprostone/physiology , Animals , Cyclic AMP , Gene Expression Regulation, Neoplastic/drug effects , Oligonucleotide Array Sequence Analysis , PC12 Cells , Polymerase Chain Reaction , RNA, Neoplasm/genetics , Rats , Transfection
17.
J Histochem Cytochem ; 55(5): 487-93, 2007 May.
Article in English | MEDLINE | ID: mdl-17242462

ABSTRACT

We examined whether an enhanced green fluorescent protein (EGFP)-tagged chromogranin A (CgA) gene construct could serve as a marker protein to follow the synthesis of CgA and the process of granulogenesis in non-neuroendocrine (NE) cells. We transfected a CgA-EGFP expression vector into non-NE COS-7 cells and investigated the localization of a chimeric CgA-EGFP protein using confocal laser scanning microscopy (CLSM). The fluorescent signal of CgA-EGFP was distributed granularly in the cytoplasm. An immunocytochemical study using anti-CgA antibody with a quantum dot (Qd)525 shows colocalization of fluorescent signal of chimeric CgA-EGFP and CgA-Qd525 signals in granular structures, particularly at the periphery of the cytoplasm. We interpreted granules that were immunoreactive to CgA in electron micrographs as secretory. Spectral analysis of EGFP fluorescence revealed distinct EGFP signals without CgA colocalization. This is the first report to show that a granular structure can be induced by transfecting the EGFP-tagged human CgA gene into non-NE cells. The EGFP-tagged CgA gene could be a useful tool to investigate processes of the regulatory pathway. A more precise analysis of the fluorescence signal of EGFP by combination with the Qd system or by spectral analysis with CLSM can provide insight into biological phenomena.


Subject(s)
Chromogranin A/biosynthesis , Cytoplasmic Granules/physiology , Green Fluorescent Proteins/biosynthesis , Animals , COS Cells , Chlorocebus aethiops , Chromogranin A/genetics , Cytoplasmic Granules/metabolism , Green Fluorescent Proteins/genetics , Humans , Immunoblotting , Microscopy, Confocal , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transfection
18.
Physiol Genomics ; 25(3): 470-9, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16554546

ABSTRACT

Family studies have suggested a genetic contribution to variation in blood pressure, but the genes responsible have thus far eluded identification. The use of intermediate phenotypes associated with hypertension, such as chromogranin plasma concentrations, may assist the discovery of hypertension-predisposing loci. We measured the concentrations of four chromogranin A (CHGA) and B (CHGB) peptides in 742 individuals from 235 nuclear families. The CHGA- and CHGB-derived peptides displayed significant heritability and revealed significant genetic correlations, most strikingly observed between CHGA(361-372) (catestatin) and CHGB(439-451). A 5-cM microsatellite genome scan revealed significant and suggestive evidence for linkage on several chromosomes for three of the peptides. Subsequent bivariate linkage analysis for peptides CHGA(361-372) and CHGB(439-451), which showed evidence for convergent linkage peaks on chromosomes 2, 7, and 13, resulted in increased evidence for linkage to these regions, suggesting pleiotropic effects of these three loci on multiple chromogranin traits. Because CHGA itself is on chromosome 14q32, and CHGB itself is on chromosome 20pter-p12, the pleiotropic regions on chromosomes 2, 7, and 13 must represent trans-acting quantitative trait loci coordinately affecting CHGA/CHGB biosynthesis and/or exocytotic secretion, likely by regulating efferent sympathetic outflow, a conclusion consistent with the in vitro studies presented here of the dual control of both exocytosis and transcription of these peptides by secretory stimuli in chromaffin cells. The results suggest a new approach to heritable autonomic control of circulation and the genetic basis of cardiovascular diseases such as systemic hypertension.


Subject(s)
Chromaffin Cells/metabolism , Chromogranin A/genetics , Chromogranin B/genetics , Quantitative Trait Loci , Animals , Blood Pressure/genetics , Catecholamines/metabolism , Chromaffin Cells/drug effects , Chromogranin A/chemistry , Chromogranin A/metabolism , Chromogranin B/chemistry , Chromogranin B/metabolism , Chromosome Mapping , Genes, Reporter , Genetic Linkage , Humans , Hypertension/blood , Hypertension/genetics , Hypertension/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , PC12 Cells , Peptide Fragments/blood , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Quantitative Trait, Heritable , Rats , Transfection
19.
J Clin Endocrinol Metab ; 90(9): 5414-25, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15956083

ABSTRACT

RATIONALE: The chromogranin A (CHGA) fragment pancreastatin (human CHGA250-301) impairs glucose metabolism, but the role of human pancreastatin in vivo remains unexplored. METHODS: We studied brachial arterial infusion of pancreastatin (CHGA273-301-amide at approximately 200 nm) on forearm metabolism of glucose, free fatty acids, and amino acids. Plasma pancreastatin was measured in obesity or type 2 diabetes. Systematic discovery of amino acid variation was performed, and the potency of one variant in the active carboxyl terminus (Gly297Ser) was tested. RESULTS: Pancreastatin decreased glucose uptake by approximately 48-50%; the lack of change in forearm plasma flow indicated a metabolic, rather than hemodynamic, mechanism. A control CHGA peptide (catestatin, CHGA352-372) did not affect glucose. Insulin increased glucose uptake, but pancreastatin did not antagonize this action. Pancreastatin increased spillover of free fatty acids by about 4.5- to 6.4-fold, but not spillover of amino acids. Insulin diminished spillover of both free fatty acids and amino acids, but these actions were not reversed by pancreastatin. Plasma pancreastatin was elevated approximately 3.7-fold in diabetes, but was unchanged during weight loss. Proteolytic cleavage sites for pancreastatin in vivo were documented by matrix-assisted laser desorption ionization/time of flight mass spectrometry. Three pancreastatin variants were discovered: Arg253Trp, Ala256Gly, and Gly297Ser. The Gly297Ser variant had unexpectedly increased potency to inhibit glucose uptake. CONCLUSIONS: The dysglycemic peptide pancreastatin is specifically and potently active in humans on multiple facets of intermediary metabolism, although it did not antagonize insulin. Pancreastatin is elevated in diabetes, and the variant Gly297Ser had increased potency to inhibit glucose uptake. The importance of human pancreastatin in vivo as well as its natural variants is established.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Obesity/metabolism , Pancreatic Hormones/genetics , Pancreatic Hormones/metabolism , Polymorphism, Genetic , Amino Acid Sequence , Amino Acids/metabolism , Base Sequence , Case-Control Studies , Chromogranin A , Diabetes Mellitus, Type 2/complications , Fatty Acids, Nonesterified/metabolism , Forearm , Genetic Variation , Humans , Injections, Intra-Articular , Male , Middle Aged , Molecular Sequence Data , Obesity/blood , Obesity/complications , Obesity/therapy , Pancreatic Hormones/administration & dosage , Pancreatic Hormones/pharmacology , Weight Loss
20.
Physiol Genomics ; 18(1): 119-27, 2004 Jun 17.
Article in English | MEDLINE | ID: mdl-15138309

ABSTRACT

Chromogranin B (CgB), a major member of the chromogranin/secretogranin family of catecholamine storage vesicle secretory proteins, plays both intracellular (vesiculogenic) and extracellular (prohormone) roles in the neuroendocrine system, and its biosynthesis and release are under the control of efferent sympathetic nerve traffic ("stimulus-transcription coupling"). To explore the role of heredity in control of CgB, we conducted a genome-wide linkage analysis of CgB release in 12 extended CEPH (Centre d'Etude du Polymorphisme Humain) pedigrees. Region-specific radioimmunoassays were used to measure five CgB fragments in plasma: CgB1-16, CgB312-331, CgB439-451, CgB568-577, and CgB647-657. Substantial heritability, as measured by h2r, was observed for three of the fragment concentrations, CgB312-331, CgB439-451, and CgB568-577, which yielded h2r estimates ranging from 0.378 (P = 0.002) to 0.910 (P < 0.0000001). Variance-component genome-wide linkage analysis with 654 microsatellite markers at 5 cM spacing identified a major quantitative trait locus for CgB312-331 on chromosome 11q24-q25 with a maximum multipoint LOD score of 5.84. Significant allelic associations between markers in the region and CgB levels were also observed. Although the 2-LOD confidence interval for linkage did not include the CgB locus itself, known trans-activators of the CgB gene promoter, or prohormone cleaving proteases, examination of positional candidate loci within this region yielded novel and plausible physiological candidates for further exploration. Allelic variation in this region may thus influence effects of sympathetic outflow on target organs in humans.


Subject(s)
Chromogranins/biosynthesis , Chromosomes, Human, Pair 20/genetics , Peptide Fragments/blood , Alleles , Animals , Barium/pharmacology , Catecholamines/metabolism , Chromogranin B , Chromogranins/blood , Chromogranins/genetics , Chromosomes, Human, Pair 11/genetics , Cohort Studies , Exocytosis/drug effects , Genetic Variation , Humans , Lod Score , Microsatellite Repeats , PC12 Cells/drug effects , PC12 Cells/metabolism , Parents , Pedigree , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/genetics , Protein Structure, Tertiary , Quantitative Trait Loci , Rats , Siblings
SELECTION OF CITATIONS
SEARCH DETAIL