Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 302(6): L555-68, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22207590

ABSTRACT

Chronic hypoxia causes pulmonary hypertension with vascular remodeling, increase in vascular tone, and altered reactivity to agonists. These changes involve alterations in multiple Ca(2+) pathways in pulmonary arterial smooth muscle cells (PASMCs). We have previously shown that vanilloid (TRPV)- and melastatin-related transient receptor potential (TRPM) channels are expressed in pulmonary arteries (PAs). Here we found that TRPV4 was the only member of the TRPV and TRPM subfamilies upregulated in PAs of chronic hypoxic rats. The increase in TRPV4 expression occurred within 1 day of hypoxia exposure, indicative of an early hypoxic response. TRPV4 in PASMCs were found to be mechanosensitive. Osmo-mechanical stress imposed by hypotonic solution activated Ca(2+) transients; they were inhibited by TRPV4 specific short interfering RNA, the TRPV blocker ruthenium red, and the cytochrome P450 epoxygenase inhibitor N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide. Consistent with TRPV4 upregulation, the Ca(2+) response induced by the TRPV4 agonist 4α-phorbol 12,13-didecanoate and hypotonicity was potentiated in hypoxic PASMCs. Moreover, a significant myogenic tone, sensitive to ruthenium red, was observed in pressurized endothelium denuded small PAs of hypoxic but not normoxic rats. The elevated basal intracellular Ca(2+) concentration in hypoxic PASMCs was also reduced by ruthenium red. In extension of these results, the development of pulmonary hypertension, right heart hypertrophy, and vascular remodeling was significantly delayed and suppressed in hypoxic trpv4(-/-) mice. These results suggest the novel concept that TRPV4 serves as a signal pathway crucial for the development of hypoxia-induced pulmonary hypertension. Its upregulation may provide a pathogenic feed-forward mechanism that promotes pulmonary hypertension via facilitated Ca(2+) influx, subsequently enhanced myogenic tone and vascular remodeling.


Subject(s)
Hypertension, Pulmonary/physiopathology , Hypotonic Solutions/metabolism , Hypoxia/physiopathology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/genetics , Hypoxia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/physiopathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats , Rats, Wistar , Up-Regulation
2.
Am J Physiol Cell Physiol ; 302(1): C77-87, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21940663

ABSTRACT

Pulmonary hypertension (PH) is associated with profound vascular remodeling and alterations in Ca(2+) homeostasis in pulmonary arterial smooth muscle cells (PASMCs). Previous studies show that canonical transient receptor potential (TRPC) genes are upregulated and store-operated Ca(2+) entry (SOCE) is augmented in PASMCs of chronic hypoxic rats and patients of pulmonary arterial hypertension (PAH). Here we further examine the involvement of TRPC and SOCE in PH with a widely used rat model of monocrotaline (MCT)-induced PAH. Rats developed severe PAH, right ventricular hypertrophy, and significant increase in store-operated TRPC1 and TRPC4 mRNA and protein in endothelium-denuded pulmonary arteries (PAs) 3 wk after MCT injection. Contraction of PA and Ca(2+) influx in PASMC evoked by store depletion using cyclopiazonic acid (CPA) were enhanced dramatically, consistent with augmented SOCE in the MCT-treated group. The time course of increase in CPA-induced contraction corresponded to that of TRPC1 expression. Endothelin-1 (ET-1)-induced vasoconstriction was also potentiated in PAs of MCT-treated rats. The response was partially inhibited by SOCE blockers, including Gd(3+), La(3+), and SKF-96365, as well as the general TRPC inhibitor BTP-2, suggesting that TRPC-dependent SOCE was involved. Moreover, the ET-1-induced contraction and Ca(2+) response in the MCT group were more susceptible to the inhibition caused by the various SOCE blockers. Hence, our study shows that MCT-induced PAH is associated with increased TRPC expression and SOCE, which are involved in the enhanced vascular reactivity to ET-1, and support the hypothesis that TRPC-dependent SOCE is an important pathway for the development of PH.


Subject(s)
Calcium Signaling/physiology , Hypertension, Pulmonary/metabolism , Monocrotaline/administration & dosage , Pulmonary Artery/metabolism , TRPC Cation Channels/biosynthesis , Animals , Calcium/metabolism , Calcium/physiology , Cells, Cultured , Hypertension, Pulmonary/chemically induced , Male , Organ Culture Techniques , Pulmonary Artery/drug effects , Rats , Rats, Sprague-Dawley , TRPC Cation Channels/physiology
3.
J Vasc Res ; 48(6): 525-37, 2011.
Article in English | MEDLINE | ID: mdl-21829038

ABSTRACT

BACKGROUND: Pulmonary hypertension is associated with vascular remodeling and increased extracellular matrix (ECM) deposition. While the contribution of ECM in vascular remodeling is well documented, the roles played by their receptors, integrins, in pulmonary hypertension have received little attention. Here we characterized the changes of integrin expression in endothelium-denuded pulmonary arteries (PAs) and aorta of chronic hypoxia as well as monocrotaline-treated rats. METHODS AND RESULTS: Immunoblot showed increased α(1)-, α(8)- and α(v)-integrins, and decreased α(5)-integrin levels in PAs of both models. ß(1)- and ß(3)-integrins were reduced in PAs of chronic hypoxia and monocrotaline-treated rats, respectively. Integrin expression in aorta was minimally affected. Differential expression of α(1)- and α(5)-integrins induced by chronic hypoxia was further examined. Immunostaining showed that they were expressed on the surface of PA smooth muscle cells (PASMCs), and their distribution was unaltered by chronic hypoxia. Phosphorylation of focal adhesion kinase was augmented in PAs of chronic hypoxia rats, and in chronic hypoxia PASMCs cultured on the α(1)-ligand collagen IV. Moreover, α(1)-integrin binding hexapeptide GRGDTP elicited an enhanced Ca(2+) response, whereas the response to α(5)-integrin binding peptide GRGDNP was reduced in CH-PASMCs. CONCLUSION: Integrins in PASMCs are differentially regulated in pulmonary hypertension, and the dynamic integrin-ECM interactions may contribute to the vascular remodeling accompanying disease progression.


Subject(s)
Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Integrins/metabolism , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Animals , Calcium Signaling/physiology , Chronic Disease , Disease Models, Animal , Focal Adhesion Kinase 1/metabolism , Hypertension, Pulmonary/chemically induced , Male , Monocrotaline/pharmacology , Phosphorylation/physiology , Rats , Rats, Wistar
4.
Am J Physiol Lung Cell Mol Physiol ; 292(6): L1598-608, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17369291

ABSTRACT

Reactive oxygen species (ROS) generated from NADPH oxidases and mitochondria have been implicated as key messengers for pulmonary vasoconstriction and vascular remodeling induced by agonists and hypoxia. Since Ca(2+) mobilization is essential for vasoconstriction and cell proliferation, we sought to characterize the Ca(2+) response and to delineate the Ca(2+) pathways activated by hydrogen peroxide (H(2)O(2)) in rat intralobar pulmonary arterial smooth muscle cells (PASMCs). Exogenous application of 10 microM to 1 mM H(2)O(2) elicited concentration-dependent increase in intracellular Ca(2+) concentration in PASMCs, with an initial rise followed by a plateau or slow secondary increase. The initial phase was related to intracellular release. It was attenuated by the inositol trisphosphate (IP(3)) receptor antagonist 2-aminoethyl diphenylborate, ryanodine, or thapsigargin, but was unaffected by the removal of Ca(2+) in external solution. The secondary phase was dependent on extracellular Ca(2+) influx. It was unaffected by the voltage-gated Ca(2+) channel blocker nifedipine or the nonselective cation channel blockers SKF-96365 and La(3+), but inhibited concentration dependently by millimolar Ni(2+), and potentiated by the Na(+)/Ca(2+) exchange inhibitor KB-R 7943. H(2)O(2) did not alter the rate of Mn(2+) quenching of fura 2, suggesting store- and receptor-operated Ca(2+) channels were not involved. By contrast, H(2)O(2) elicited a sustained inward current carried by Na(+) at -70 mV, and the current was inhibited by Ni(2+). These results suggest that H(2)O(2) mobilizes intracellular Ca(2+) through multiple pathways, including the IP(3)- and ryanodine receptor-gated Ca(2+) stores, and Ni(2+)-sensitive cation channels. Activation of these Ca(2+) pathways may play important roles in ROS signaling in PASMCs.


Subject(s)
Calcium/metabolism , Hydrogen Peroxide/pharmacology , Muscle, Smooth, Vascular/physiology , Oxidants/pharmacology , Pulmonary Artery/physiology , Animals , Cells, Cultured , Fluorescent Dyes , Fura-2 , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Manganese/pharmacokinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques , Pulmonary Artery/cytology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sodium/metabolism , Sodium-Calcium Exchanger/physiology
5.
J Biol Chem ; 281(11): 7205-13, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16410241

ABSTRACT

Receptor activity-modifying proteins (RAMPs) enable calcitonin receptor-like receptor (CRLR) to function as a calcitonin gene-related peptide receptor (CRLR/RAMP1) or an adrenomedullin (AM) receptor (CRLR/RAMP2 or -3). Here we investigated the functions of the cytoplasmic C-terminal tails (C-tails) of human RAMP1, -2, and -3 (hRAMP1, -2, and -3) by cotransfecting their C-terminal deletion or progressive truncation mutants into HEK-293 cells stably expressing hCRLR. Deletion of the C-tail from hRAMP1 had little effect on the surface expression, function, or intracellular trafficking of the mutant heterodimers. By contrast, deletion of the C-tail from hRAMP2 disrupted transport of hCRLR to the cell surface, resulting in significant reductions in (125)I-hAM binding and evoked cAMP accumulation. The transfection efficiency for the hRAMP2 mutant was comparable with that for wild-type hRAMP2; moreover, immunocytochemical analysis showed that the mutant hRAMP2 remained within the endoplasmic reticulum. FACS analysis revealed that deleting the C-tail from hRAMP3 markedly enhances AM-evoked internalization of the mutant heterodimers, although there was no change in agonist affinity. Truncating the C-tails by removing the six C-terminal amino acids of hRAMP2 and -3 or exchanging their C-tails with one another had no effect on surface expression, agonist affinity, or internalization of hCRLR, which suggests that the highly conserved Ser-Lys sequence within hRAMP C-tails is involved in cellular trafficking of the two AM receptors. Notably, deleting the respective C-tails from hRAMPs had no effect on lysosomal sorting of hCRLR. Thus, the respective C-tails of hRAMP2 and -3 differentially affect hCRLR surface delivery and internalization.


Subject(s)
Calcitonin Gene-Related Peptide/chemistry , Cytoplasm/metabolism , Receptors, Peptide/chemistry , Amino Acid Sequence , Blotting, Western , Cell Line , Cell Membrane/metabolism , Cell Proliferation , Cell Separation , Cyclic AMP/metabolism , Cycloheximide/pharmacology , DNA/chemistry , DNA, Complementary/metabolism , Dimerization , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , Flow Cytometry , Gene Deletion , Green Fluorescent Proteins/chemistry , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/chemistry , Lysosomes/chemistry , Lysosomes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Protein 2 , Receptor Activity-Modifying Proteins , Receptors, Adrenomedullin , Recombinant Fusion Proteins/chemistry , Serine/chemistry , Time Factors , Transfection
6.
FEBS Lett ; 579(22): 4997-5001, 2005 Sep 12.
Article in English | MEDLINE | ID: mdl-16115629

ABSTRACT

We developed a transgenic (Tg) rat model that overexpresses human proadrenomedullin N-terminal 20 peptide (PAMP) only and then compared the effects of unilateral nephrectomy followed by a high salt diet for five weeks in Tg and wild-type rats. We found that systolic blood pressure was significantly lower in Tg UNX rats and cardiac hypertrophy and myocardial fibrosis was also attenuated in Tg rats. Evaluation of gene expression showed suppression of cardiac local renin-angiotensin system (RAS) in Tg rat. These results suggest that in addition to reducing blood pressure, PAMP suppresses cardiac hypertrophy through negative regulation of the local cardiac RAS.


Subject(s)
Blood Pressure/physiology , Cardiomegaly/pathology , Hypertension/metabolism , Myocardium/pathology , Peptide Fragments/metabolism , Protein Precursors/metabolism , Proteins/metabolism , Adrenomedullin , Animals , Animals, Genetically Modified , Cardiomegaly/metabolism , Diet , Fibrosis/pathology , Humans , Kidney/cytology , Kidney/pathology , Myocardium/cytology , Myocardium/metabolism , Peptide Fragments/genetics , Protein Precursors/genetics , Proteins/genetics , Rats , Renin-Angiotensin System/physiology , Salts/administration & dosage , Transgenes
7.
Biochem Biophys Res Commun ; 332(3): 866-72, 2005 Jul 08.
Article in English | MEDLINE | ID: mdl-15913562

ABSTRACT

We have investigated the antioxidant effect of adrenomedullin (AM) on endothelial function in the Dahl salt-sensitive (DS) rat hypertension model. Dahl salt-resistant (DR) and DS rats were fed an 8% NaCl diet. In addition, the DS rats were subcutaneously infused with either saline or recombinant human AM for 4 weeks. Although systolic blood pressures measured weekly in AM- and saline-infused rats did not significantly differ, aortic O2*- levels were significantly (P<0.01) higher in the latter. Likewise, both endothelial nitric oxide synthase (eNOS) mRNA and protein were significantly higher in saline-infused DS rats. Infusion of AM reduced both O2*- and eNOS expression to levels comparable to those seen in DR rats. AM infusion also upregulated the gene expression of guanosine-5'-triphosphate cyclohydrolase I and downregulated the expression of p22(phox), suggesting that AM increased the NOS coupling and bioavailability of NO. AM possesses significant antioxidant properties that improve endothelial function.


Subject(s)
Antioxidants/pharmacology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Peptides/pharmacology , Adrenomedullin , Animals , Blood Pressure/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Humans , In Vitro Techniques , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Peptides/blood , Peptides/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Dahl , Recombinant Proteins/blood , Recombinant Proteins/pharmacology , Superoxides/metabolism , Vasodilation/drug effects
8.
Cardiovasc Res ; 65(4): 921-9, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15721873

ABSTRACT

OBJECTIVE: The extracellular matrix (ECM) determines the structural integrity of the heart and vasculature, participating in cardiovascular remodeling. We previously reported that adrenomedullin (AM) inhibited cellular proliferation and protein synthesis of cardiac fibroblasts; however, the precise mechanisms of AM actions as an antifibrotic factor remain unknown. The purpose of this study was to examine the biological actions of AM against the profibrotic factor angiotensin II (Ang II) in coronary adventitia. METHODS AND RESULTS: Rats with hypertension induced by Ang II infusion were administered 0.06 mug/kg/min recombinant human AM subcutaneously for 14 days. The AM infusion significantly (p<0.05) reduced the Ang II-induced increase of coronary adventitial fibroblasts expressing Ki-67 and alpha-smooth muscle actin (alpha-SMA) in the left ventricle, by 65%, and 62%, respectively, without affecting systolic blood pressure, left ventricle/body weight, or cross-sectional area of myocardial fibers. Collagen deposition of coronary arteries was reduced by the AM infusion (-24%, p<0.01), and these effects of AM were accompanied by significant reductions in gene expression of type 1 collagen (-49%, p<0.05) and transforming growth factor-beta1 (TGF-beta1) (-55%, p<0.01). In cultured cardiac fibroblasts, 10(-7) mol/L AM exerted an inhibitory effect on TGF-beta1-induced alpha-SMA expression (p<0.01) that was mimicked by 8-bromo-cAMP and attenuated by the protein kinase A inhibitor H-89. CONCLUSION: AM decreased Ang II-induced collagen deposition surrounding the coronary arteries, inhibiting myofibroblast differentiation and expressions of ECM-related genes in rats. The present findings further support the biological action of AM as an antifibrotic factor in vascular remodeling.


Subject(s)
Angiotensin II/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Hypertension/pathology , Peptides/pharmacology , Actins/metabolism , Adrenomedullin , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Cardiotonic Agents/blood , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Size/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Connective Tissue/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Heart Ventricles/pathology , Humans , Hypertension/chemically induced , Hypertension/metabolism , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peptides/blood , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Ventricular Remodeling/drug effects
9.
Biochem Biophys Res Commun ; 325(1): 80-4, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15522203

ABSTRACT

BACKGROUND: The delicate balance of the extracellular matrix (ECM) determines the stiffness of the vascular wall, and adventitial fibroblasts are involved in ECM formation by synthesizing and degrading matrix proteins. In the present study, we examined the effect of the bioactive peptide adrenomedullin (AM) on activity and expression of matrix metalloproteinases (MMPs) in cultured aortic adventitial fibroblasts. METHODS AND RESULTS: In cultured adventitial fibroblasts isolated from aorta of adult Wistar rats, 10(-6)mol/L angiotensin II (Ang II) significantly (p<0.05) down-regulated MMP-2 activity as determined by in vitro gelatin zymography. In contrast, 10(-7)mol/L synthetic rat AM significantly (p<0.05) stimulated zymographic MMP-2 activity by 23%, increasing intracellular cAMP, and AM abolished the action of Ang II, augmenting the MMP-2 activity. Similarly, Ang II down-regulated MMP-2 protein expression assessed by Western blotting, whereas AM increased it. Furthermore, 8-bromo-cAMP, an analogue of cAMP, mimicked the effect of AM, and H-89, an inhibitor for protein kinase A (PKA), significantly decreased the basal and AM-induced MMP-2 activity. CONCLUSION: This study provides a new insight into the biological action of AM and its intracellular signaling system of cAMP/PKA stimulating the matrix degrading enzyme MMP-2, suggesting an important role for this molecule in modulating ECM deposition in the adventitial layer.


Subject(s)
Aorta/anatomy & histology , Connective Tissue/anatomy & histology , Fibroblasts/drug effects , Matrix Metalloproteinase 2/metabolism , Peptides/pharmacology , Vasodilator Agents/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adrenomedullin , Angiotensin II/metabolism , Animals , Cells, Cultured , Connective Tissue/metabolism , Cyclic AMP/metabolism , Enzyme Activation , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Isoquinolines/pharmacology , Male , Peptides/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Sulfonamides/pharmacology , Vasodilator Agents/metabolism
10.
Peptides ; 25(11): 2003-12, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15501534

ABSTRACT

Three receptor activity modifying proteins (RAMPs) chaperone calcitonin-like receptor (CLR) to the cell surface. RAMP2 enables CLR to form an adrenomedullin (AM)-specific receptor that is sensitive to AM-(22-52) (AM(1) receptor). RAMP3 enables CLR to form an AM receptor sensitive to both calcitonin gene-related peptide (CGRP)-(8-37) and AM-(22-52) (AM(2) receptor), though rat and mouse AM(2) receptors show a clear preference for CGRP alpha-(8-37) over AM-(22-52). RAMP1 enables CRL to form the CGRP-(8-37)-sensitive CGRP(1) receptor, which can also be activated by higher concentrations of AM. Here we review the available information on the pharmacological features and possible pathophysiological roles of the aforementioned AM receptors.


Subject(s)
Receptors, Calcitonin/metabolism , Receptors, Peptide/metabolism , Animals , Forecasting , Humans , Protein Binding , RNA, Messenger/metabolism , Receptors, Adrenomedullin
11.
Peptides ; 25(7): 1115-21, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15245870

ABSTRACT

We examined the effects of tumor necrosis factor (TNF)-alpha on the expression and functionality of adrenomedullin (AM) receptors in cultured human coronary artery smooth muscle cells. Analysis of real-time quantitative polymerase chain reactions showed that these cells abundantly express two AM receptors comprised of calcitonin receptor-like receptor (CRLR) and receptor activity-modifying protein 1 (RAMP1) or RAMP2. TNF-alpha induced time- and dose-dependent decreases in the expression of CRLR and RAMP1/2 mRNAs, thereby diminishing AM-evoked cAMP production. The suppression of these three mRNAs was unaffected by inhibiting NOS, protein kinase G, protein kinase A, superoxide formation or NF-kappaB activation.


Subject(s)
Coronary Vessels/cytology , Down-Regulation/drug effects , Myocytes, Smooth Muscle/drug effects , Receptors, Peptide/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Calcitonin Receptor-Like Protein , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Protein 2 , Receptor Activity-Modifying Proteins , Receptors, Adrenomedullin , Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism , Receptors, Peptide/genetics
12.
Biochem Biophys Res Commun ; 314(4): 1057-63, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14751240

ABSTRACT

We examined the effects of recombinant human C-reactive protein (rhCRP) on atherosclerosis-related factors in cultured human coronary artery endothelial and smooth muscle cells (HCAECs and HCASMCs). After removing endotoxin from commercial rhCRP preparations using the appropriate column, the purified (P)-rhCRP retained the ability to Ca(2+)-dependently bind to phosphorylcholine, but did not augment the secretion of interleukin-6 and MCP-1 from HCAECs, as non-purified (NP)-rhCRP did. By contrast, P-rhCRP elicited 2- to 3-fold increases in the secretion of both hormones from HCASMCs, though the effect was smaller than that obtained with NP-rhCRP. Production of PAI-1 and endothelin-1 was little affected by either rhCRP preparation in either cell type. In addition, P-rhCRP dose-dependently diminished adrenomedullin release from both cell types, but did not affect adrenomedullin receptor expression or function. Our findings highlight the importance of removing endotoxin from commercial rCRP preparations and show that hCRP elicits atherogenic responses from HCASMCs, but not HCAECs.


Subject(s)
C-Reactive Protein/drug effects , Coronary Vessels/metabolism , Muscle, Smooth/drug effects , Peptides/physiology , Adrenomedullin , Cells, Cultured , Coronary Vessels/cytology , Humans , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Receptors, Adrenomedullin , Receptors, Peptide/metabolism , Recombinant Proteins/pharmacology
13.
Mol Pharmacol ; 65(1): 207-13, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14722252

ABSTRACT

Coexpression of receptor activity-modifying proteins (RAMPs) with calcitonin receptor 2 (CTR2) or calcitonin receptor-like receptor (CRLR) leads to the formation of four functional heterodimeric receptors for human calcitonin gene-related peptide (hCGRP). In this study, we transfected hCGRP receptors into human embryonic kidney 293 cells and examined their pharmacological profiles using three dominant-negative (DN) RAMP mutants and various hCGRPalpha analogs. Fluorescence-activated cell-sorting analysis revealed that their cotransfection with CTR2 induced cell surface expression of all three RAMPs, and the three CTR2/RAMP heterodimers mediated equivalent levels of cAMP production in response to hCGRPalpha that were approximately 50-fold greater than were seen with CTR2 alone. By contrast, [Tyr0]hCGRPalpha binding and signaling were markedly weaker with CTR2/RAMP2 or -3 than with CTR2/RAMP1 or CRLR/RAMP1; likewise, 125I-[His10]hCGRPalpha bound most potently to CTR2/RAMP1. When CTR2 was coexpressed with DN RAMP1 or -2, hCGRPalpha-evoked responses were similar to those seen with CTR2 alone, despite the expression of both CTR2 and DN RAMP at the cell surface. But coexpression of DN RAMP3 with CTR2 significantly diminished hCGRPalpha signaling compared with that seen with CTR2 alone, indicating that DN RAMP3 is able to function as a negative regulator of CTR2 function. Competition experiments showed the relative agonist sensitivity of the four receptors to be hCGRPalpha > [Tyr0]hCGRPalpha > [Cys(Et)2,7]hCGRPalpha > [Cys(ACM)2,7]hCGRPalpha. Of the linear analogs, [Cys(ACM)2,7]hCGRPalpha (ACM, acetylmethoxy) enhanced cAMP formation only via CTR2/RAMP1, whereas [Cys(Et2,7)]hCGRPalpha acted via CRLR/RAMP1 and somewhat less potently via CTR2/RAMP1. Thus, among the three CGRP8-37-insensitive receptors, CTR2/RAMP1 is most sensitive to the two linear analogs, suggesting that it could be classified as a CGRP2 receptor. Moreover, the combined use of iodinated CGRPalpha analogs may be useful for defining the CGRP1 receptor.


Subject(s)
Membrane Proteins/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , Cell Line , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Peptides/pharmacology , Radioligand Assay , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Protein 2 , Receptor Activity-Modifying Protein 3 , Receptor Activity-Modifying Proteins , Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism , Receptors, Calcitonin Gene-Related Peptide/genetics
14.
Hypertension ; 42(3): 369-72, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12913064

ABSTRACT

The vasodilator peptide adrenomedullin (AM) elicits diuresis and natriuresis and inhibits aldosterone secretion. The aim of this study was to better understand the role of AM in maintaining water and electrolyte balance during chronic salt loading. Male Wistar rats were divided into a high salt (HS) group that received a diet containing 8% sodium chloride (NaCl) and a normal salt group that received a diet containing 0.4% NaCl. Plasma AM concentrations as well as expression of AM mRNA in the adrenal gland and kidney were then measured after 3, 7, 14, and 28 days. After 28 days, sodium and water excretion were significantly higher in HS rats than in control, although blood pressure and fluid volume were not significantly affected. Moreover, although plasma AM remained unchanged for up to 14 days, it was increased 2.5-fold in HS rats after 28 days on a high salt diet, and there were corresponding 3-fold and 1.5-fold increases in the levels of AM mRNA in the adrenal gland and kidney, respectively. At the same time, expression of calcitonin receptor-like receptor mRNA was significantly upregulated in both kidney and adrenal gland, as was expression of receptor activity-modify protein 1 (RAMP1) and RAMP2 mRNA in the adrenals and expression of RAMP3 in kidneys. Taken together, these results suggest that AM plays a role in the regulation of water and electrolyte balance in animals chronically ingesting high levels of salt.


Subject(s)
Adrenal Glands/drug effects , Kidney/drug effects , Peptides/genetics , Receptors, Peptide/genetics , Sodium Chloride, Dietary/administration & dosage , Adrenal Glands/metabolism , Adrenomedullin , Aldosterone/blood , Animals , Blood Pressure/drug effects , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins , Kidney/metabolism , Male , Membrane Proteins/genetics , Peptides/blood , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Protein 2 , Receptor Activity-Modifying Protein 3 , Receptor Activity-Modifying Proteins , Receptors, Adrenomedullin , Receptors, Peptide/metabolism , Time Factors , Water-Electrolyte Balance/drug effects
15.
Regul Pept ; 113(1-3): 109-14, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12686469

ABSTRACT

Human adrenomedullin (hAM) is an endogenous peptide that has potent vasodilator activity. Mature AM is biosynthesized from its intermediate form, glycine-extended AM (AM-gly), by carboxy-terminal amidation. AM-gly is generally considered to be biologically inactive but is a major molecular form in human and rat plasma. The present study demonstrated that recombinant human AM-gly (hAM-gly) elicits potent vasodilator effect on isolated rat aorta. In aortic rings, hAM-gly produced dose-dependent (0.1-100 nM) relaxation in phenylephrine-precontracted strips (pD(2) 8.4+/-0.5). The vasorelaxant potency of hAM-gly was comparable to that of hAM (pD(2) 8.6+/-0.2) but hAM-gly took a significantly (P<0.01) longer time to reach the maximal relaxation compared with hAM (T(max) 23+/-4 vs. 5+/-2 min). Vasorelaxant responses to hAM-gly were abolished by endothelial removal. N(omega)-nitro-L-arginine (L-NNA) and AM(22-52) significantly (P<0.01) reduced the vasodilator effect of hAM-gly. Furthermore, 4-phenyl-3-butenoic acid (PBA), an alpha-amidation enzyme inhibitor, significantly (P<0.05) inhibited the vasorelaxant responses to hAM-gly without any effect on the hAM-induced relaxation, suggesting the possible process of amidation in the rat aorta. We further clarified that the aorta has the ability to convert exogenous hAM-gly to mature hAM and the conversion is inhibited by PBA. These results suggest that the circulating AM-gly may play a role in regulating vascular tone and increased plasma AM-gly may be involved in the pathophysiology of cardiovascular diseases.


Subject(s)
Aorta/drug effects , Glycine/chemistry , Peptides/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Adrenomedullin , Amides/chemistry , Animals , Aorta/physiology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Humans , In Vitro Techniques , Male , Nitroarginine/pharmacology , Peptides/chemistry , Rats , Rats, Wistar , Recombinant Proteins/pharmacology
16.
J Biol Chem ; 278(25): 22623-30, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12684503

ABSTRACT

When co-expressed with receptor activity-modifying protein (RAMP) 1, calcitonin receptor-like receptor (CRLR) can function as a receptor for both calcitonin gene-related peptide (CGRP) and adrenomedullin (AM). To investigate the structural determinants of ligand binding specificity, we examined the extracellular domain of human (h) RAMP1 using various deletion mutants. Co-expression of the hRAMP1 mutants with hCRLR in HEK-293 cells revealed that deletion of residues 91-94, 96-100, or 101-103 blocked [125I]CGRP binding and completely abolished intracellular cAMP accumulation normally elicited by CGRP or AM. On the other hand, the deletion of residues 78-80 or 88-90 significantly attenuated only AM-evoked responses. In all of these cases, the receptor heterodimers were fully expressed at the cell surface. Substituting alanine for residues 91-103 one at a time had little effect on CGRP-induced responses, indicating that although this segment is essential for high affinity agonist binding to the receptors, none of the residues directly interacts with either CGRP or AM. This finding suggests that RAMPs probably determine ligand specificity by contributing to the structure of the ligand-binding pocket or by allosteric modulation of the conformation of the receptor. Interestingly, the L94A mutant up-regulated surface expression of the receptor heterodimer to a greater degree than wild-type hRAMP1, thereby increasing CGRP binding and signaling. L94A also significantly increased cell surface expression of the hRAMP1 deletion mutant D101-103 when co-transfected with hCRLR, and expression of a L94A/D101-103 double mutant markedly attenuated the activity of endogenous RAMP1 in HEK-293T cells.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Peptides/metabolism , Adrenomedullin , Amino Acid Sequence , Binding Sites , Cell Line , Flow Cytometry , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Mutagenesis , RNA, Messenger/genetics , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Proteins , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...