Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stephen A. Douglas is active.

Publication


Featured researches published by Stephen A. Douglas.


The Lancet | 2002

Congestive heart failure and expression of myocardial urotensin II

Stephen A. Douglas; Lara Tayara; Eliot H. Ohlstein; Nadine Halawa; Adel Giaid

BACKGROUND Human urotensin II has several cardiovascular actions, including potent vasoactive, and cardiac inotropic and hypertropic properties. Our aim was to ascertain degree of expression of urotensin II and its receptor GPR14 (now known as UT receptor) in the myocardium of patients with congestive heart failure (CHF). METHODS We obtained specimens of myocardium from the hearts of 19 patients with end-stage CHF (12 ischaemic heart disease, seven dilated cardiomyopathy), five patients with early-stage CHF, and eight healthy controls. We used immunohistochemistry, in-situ hybridisation, reverse transcriptase-PCR (RT-PCR), and fluorescein isothiocyanate (FITC)-conjugated urotensin II to ascertain degree of myocardial expression of urotensin II and binding urotensin receptor. FINDINGS Our results showed strong expression of urotensin II in the cardiomyocytes, and to a lesser extent in the vascular smooth muscle cells, endothelial cells, and inflammatory cells of patients with end-stage CHF. There was significantly less urotensin II expression in the myocardium of patients with early-stage CHF (p<0.0001). Also, there was little to no urotensin II expression in the myocardium of healthy controls. Myocardial expression of urotensin II correlated significantly with left ventricular end-diastolic dimension (p=0.0092), and inversely with ejection fraction (p=0.0002). RT-PCR showed increased concentrations of urotensin II and presence of urotensin receptor mRNA in the myocardium of patients with CHF. The confocal microscopy results showed a significant increase in the binding sites for urotensin in the myocardium of patients with end-stage CHF (p<0.0001). INTERPRETATION Our findings suggest a possible role for urotensin II in the cardiac dysfunction and remodelling characteristic of CHF.


Trends in Cardiovascular Medicine | 2000

Human urotensin-II, the most potent mammalian vasoconstrictor identified to date, as a therapeutic target for the management of cardiovascular disease.

Stephen A. Douglas; Eliot H. Ohlstein

The novel cyclic undecapeptide human urotensin-II (hU-II) and its high-affinity G-protein-coupled receptor, GPR14, are both expressed within the human cardiovasculature (vascular smooth muscle, endothelium, myocardium, coronary atheroma, etc.) and may, therefore, contribute to the (patho)physiological regulation of cardiovascular homeostasis in humans. Indeed, hU-II is an efficacious, sustained spasmogen of mammalian isolated blood vessels including those from rats, rabbits, dogs, pigs, non-human primates and humans (where it is one to two orders of magnitude more potent than endothelin(ET)-1). In vivo, hU-II markedly alters systemic hemodynamics in the anesthetized primate (increase cardiac contractility [dP/dt], increase stroke volume, decrease total peripheral resistance) ultimately resulting in fatal cardiovascular collapse. As such, the development of selective hU-II receptor antagonists may be of utility in the management of cardiovascular disorders characterized by aberrant vasoconstriction, myocardial dysfunction and/or cardiac remodeling (e.g., myocardial infarction, congestive heart failure).


British Journal of Pharmacology | 2000

Contractile responses to human urotensin-II in rat and human pulmonary arteries: effect of endothelial factors and chronic hypoxia in the rat.

Margaret R. MacLean; D. Alexander; Alison Stirrat; Marie Gallagher; Stephen A. Douglas; Eliot H. Ohlstein; Ian Morecroft; K Polland

Responses to human urotensin‐II (hU‐II) were investigated in human and rat pulmonary arteries. Rat pulmonary arteries: hU‐II was a potent vasoconstrictor of main pulmonary arteries (2–3 mm i.d.) (pEC50, 8.55±0.08, n=21) and was ∼4 fold more potent than endothelin‐1 [ET‐1] (P<0.01), although its Emax was considerably less (∼2.5 fold, P<0.001). The potency of hU‐II increased 2.5 fold with endothelium removal (P<0.05) and after raising vascular tone with ET‐1 (P<0.01). Emax was enhanced ∼1.5 fold in the presence of Nω‐nitro‐L‐arginine methylester (L‐NAME, 100 μM, P<0.01) and ∼2 fold in vessels from pulmonary hypertensive rats exposed to 2 weeks chronic hypoxia (P<0.05). hU‐II did not constrict smaller pulmonary arteries. Human pulmonary arteries (∼250 μm i.d.): in the presence of L‐NAME, 3 out of 10 vessels contracted to hU‐II and this contraction was highly variable. hU‐II is, therefore, a potent vasoconstrictor of rat main pulmonary arteries and this response is increased by endothelial factors, vascular tone and onset of pulmonary hypertension. Inhibition of nitric oxide synthase uncovers contractile responses to hU‐II in human pulmonary arteries.


British Journal of Pharmacology | 2002

Molecular and pharmacological characterization of genes encoding urotensin-II peptides and their cognate G-protein-coupled receptors from the mouse and monkey

Nabil Elshourbagy; Stephen A. Douglas; Usman Shabon; Stephen M. Harrison; Graham Duddy; Jan L. Sechler; Zhaohui Ao; Beverly E. Maleeff; Diane Naselsky; Jyoti Disa; Nambi Aiyar

Urotensin‐II (U‐II) and its receptor (UT) represent novel therapeutic targets for management of a variety of cardiovascular diseases. To test such hypothesis, it will be necessary to develop experimental animal models for the manipulation of U‐II/UT receptor system. The goal of this study was to clone mouse and primate preproU‐II and UT for pharmacological profiling. Monkey and mouse preproU‐II genes were identified to encode 123 and 125 amino acids. Monkey and mouse UT receptors were 389, and 386 amino acids, respectively. Genomic organization of mouse genes showed that the preproU‐II has four exons, while the UT receptor has one exon. Although initially viewed by many exclusively as cardiovascular targets, the present study demonstrates expression of mouse and monkey U‐II/UT receptor mRNA in extra‐vascular tissue including lung, pancreas, skeletal muscle, kidney and liver. Ligand binding studies showed that [125I]h U‐II bound to a single sites to the cloned receptors in a saturable/high affinity manner (Kd 654±154 and 214±65 pM and Bmax of 1011±125 and 497±68 fmol mg−1 for mouse and monkey UT receptors, respectively). Competition binding analysis demonstrated equipotent, high affinity binding of numerous mammalian, amphibian and piscine U‐II isopeptides to these receptors (Ki=0.8 – 3 nM). Fluorescein isothiocyanate (FITC) labelled U‐II, bound specifically to HEK‐293 cells expressing mouse or monkey UT receptor, confirming cell surface expression of recombinant UT receptor. Exposure of these cells to human U‐II resulted in an increase in intracellular [Ca2+] concentrations (EC50 3.2±0.8 and 1.1±0.3 nM for mouse and monkey UT receptors, respectively) and inositol phosphate (Ip) formation (EC50 7.2±1.8 and 0.9±0.2 nM for mouse and monkey UT receptors, respectively) consistent with the primary signalling pathway for UT receptor involving phospholipase C activation.


International Journal for Parasitology | 2001

The role of endothelin in the pathogenesis of Chagas' disease.

Stefka B. Petkova; Huan Huang; Stephen M. Factor; Richard G. Pestell; Boumediene Bouzahzah; Linda A. Jelicks; Louis M. Weiss; Stephen A. Douglas; Murray Wittner; Herbert B. Tanowitz

Infection with Trypanosoma cruzi causes a generalised vasculitis of several vascular beds. This vasculopathy is manifested by vasospasm, reduced blood flow, focal ischaemia, platelet thrombi, increased platelet aggregation and elevated plasma levels of thromboxane A(2) and endothelin-1. In the myocardium of infected mice, myonecrosis and a vasculitis of the aorta, coronary artery, smaller myocardial vessels and the endocardial endothelium are observed. Immunohistochemistry studies employing anti-endothelin-1 antibody revealed increased expression of endothelin-1, most intense in the endocardial and vascular endothelium. Elevated levels of mRNA for prepro endothelin-1, endothelin converting enzyme and endothelin-1 were observed in the infected myocardium. When T. cruzi-infected mice were treated with phosphoramidon, an inhibitor of endothelin converting enzyme, there was a decrease in heart size and severity of pathology. Mitogen-activated protein kinases and the transcription factor activator-protein-1 regulate the expression of endothelin-1. Therefore, we examined the activation of mitogen-activated protein kinases in the myocardium by T. cruzi. Western blot demonstrated an extracellular signal regulated kinase. In addition, the activator-protein-1 DNA binding activity, as determined by electrophoretic mobility shift assay, was increased. Increased expression of cyclins A and cyclin D1 was observed in the myocardium, and immunohistochemistry studies revealed that interstitial cells and vascular and endocardial endothelial cells stained intensely with antibodies to these cyclins. These data demonstrate that T. cruzi infection of the myocardium activates extracellular signal regulated kinase, activator-protein-1, endothelin-1, and cyclins. The activation of these pathways is likely to contribute to the pathogenesis of chagasic heart disease. These experimental observations suggest that the vasculature plays a role in the pathogenesis of chagasic cardiomyopathy. Additionally, the identification of these pathways provides possible targets for therapeutic interventions to ameliorate or prevent the development of cardiomyopathy during T. cruzi infection.


Molecular and Cellular Biochemistry | 2007

Lysophosphatidylcholine induces inflammatory activation of human coronary artery smooth muscle cells

Nambi Aiyar; Jyoti Disa; Zhaohui Ao; Haisong Ju; Sandhya S. Nerurkar; Robert N. Willette; Colin H. Macphee; Douglas G. Johns; Stephen A. Douglas

Lysophosphatidylcholine (LPC) is the major bioactive lipid component of oxidized LDL, thought to be responsible for many of the inflammatory effects of oxidized LDL described in both inflammatory and endothelial cells. Inflammation-induced transformation of vascular smooth muscle cells from a contractile phenotype to a proliferative/secretory phenotype is a hallmark of the vascular remodeling that is characteristic of atherogenesis; however, the role of LPC in this process has not been fully described. The present study tested the hypothesis that LPC is an inflammatory stimulus in coronary artery smooth muscle cells (CASMCs). In cultured human CASMCs, LPC stimulated time- and concentration-dependent release of arachidonic acid that was sensitive to phospholipase A2 and C inhibition. LPC stimulated the release of arachidonic acid metabolites leukotriene-B4 and 6-keto-prostaglandin F1α, within the same time course. LPC was also found to stimulate basic fibroblast growth factor release as well as stimulating the release of the cytokines GM-CSF, IL-6, and IL-8. Optimal stimulation of these signals was obtained via palmitic acid-substituted LPC species. Stimulation of arachidonic acid, inflammatory cytokines and growth factor release, implies that LPC might play a multifactorial role in the progression of atherosclerosis, by affecting inflammatory processes.


Peptides | 2005

Expression of urotensin-II in human coronary atherosclerosis.

Ghada S. Hassan; Stephen A. Douglas; Eliot H. Ohlstein; Adel Giaid

The vasoactive peptide urotensin-II (U-II) is best known for its ability to regulate peripheral vascular and cardiac contractile function in vivo, and recent in vitro studies have suggested a role for the peptide in the control of vascular remodeling by inducing smooth muscle proliferation and fibroblast-mediated collagen deposition. Therefore, U-II may play a role in the etiology of atherosclerosis. In the present study we sought to determine the expression of U-II in coronary arteries from patients with coronary atherosclerosis and from normal control subjects, using immunohistochemistry and in situ hybridization. In normal coronary arteries, there was little expression of U-II in all types of cells. In contrast, in patients with coronary atherosclerosis, endothelial expression of U-II was significantly increased in all diseased segments (P<0.05). Greater expression of U-II was noted in endothelial cells of lesions with subendothelial inflammation or fibrofatty lesion compared with that of endothelial cells underlined by dense fibrosis or minimal intimal thickening. Myointimal cells and foam cells also expressed U-II. In most diseased segments, medial smooth muscle cells exhibited moderate expression of U-II. These findings demonstrate upregulation of U-II in endothelial, myointimal and medial smooth muscle cells of atherosclerotic human coronary arteries, and suggest a possible role for U-II in the pathogenesis of coronary atherosclerosis.


British Journal of Pharmacology | 2003

Deletion of the UT receptor gene results in the selective loss of urotensin-II contractile activity in aortae isolated from UT receptor knockout mice

David J. Behm; Stephen M. Harrison; Zhaohui Ao; Kristeen Maniscalco; Susan Pickering; Evelyn Grau; Tina N. Woods; Robert W. Coatney; Christopher P. Doe; Robert N. Willette; Douglas G. Johns; Stephen A. Douglas

Urotensin‐II (U‐II) is among the most potent mammalian vasoconstrictors identified and may play a role in the aetiology of essential hypertension. Currently, only one mouse U‐II receptor (UT) gene has been cloned. It is postulated that this protein is solely responsible for mediating U‐II‐induced vasoconstriction. This hypothesis has been investigated in the present study, which assessed basal haemodynamics and vascular reactivity to hU‐II in wild‐type (UT(+/+)) and UT receptor knockout (UT(−/−)) mice. Basal left ventricular end‐diastolic and end‐systolic volumes/pressures, stroke volumes, mean arterial blood pressures, heart rates, cardiac outputs and ejection fractions in UT(+/+) mice and in UT(−/−) mice were similar. Relative to UT(+/+) mouse isolated thoracic aorta, where hU‐II was a potent spasmogen (pEC50=8.26±0.08) that evoked relatively little vasoconstriction (17±2% 60 mM KCl), vessels isolated from UT(−/−) mice did not respond to hU‐II. However, in contrast, the superior mesenteric artery isolated from both the genotypes did not contract in the presence of hU‐II. Reactivity to unrelated vasoconstrictors (phenylephrine, endothelin‐1, KCl) and endothelium‐dependent/independent vasodilator agents (carbachol, sodium nitroprusside) was similar in the aorta and superior mesenteric arteries isolated from both the genotypes. The present study is the first to directly link hU‐II‐induced vasoconstriction with the UT receptor. Deletion of the UT receptor gene results in loss of hU‐II contractile action with no ‘nonspecific’ alterations in vascular reactivity. However, as might be predicted based on the limited contractile efficacy recorded in vitro, the contribution that hU‐II and its receptor make to basal systemic haemodynamics appears to be negligible in this species.


European Journal of Pharmacology | 1992

Regional vasodilation to endothelin-1 is mediated by a non-ETA receptor subtype in the anaesthetized rat: effect of BQ-123 on systemic haemodynamic responses

Stephen A. Douglas; John D. Elliott; Eliot H. Ohlstein

The sustained infusion of 75 nmol/kg per min BQ-123 selectively attenuates the systemic pressor responses to i.v. bolus endothelin-1 in the anaesthetized rat; the initial systemic vasodepression is only attenuated when a 10-fold higher dose of this ETA-selective antagonist is infused. These doses of BQ-123 do not antagonize the haemodynamic actions of angiotensin II or calcitonin gene-related peptide nor do they alter basal systemic haemodynamics. The secondary increase in carotid vascular resistance associated with the systemic pressor actions of endothelin-1 is selectively attenuated by the bolus i.v. administration of BQ-123 (1.6 mumol/kg); the maximum degree of the vasorelaxation that precedes this more sustained constriction is unaltered. Thus, the initial carotid vasodilation observed to endothelin-1, and the associated systemic vasodepression, is mediated by a non-ETA receptor subtype. Furthermore, subtle differences might exist between the receptors mediating mesenteric and carotid vasoconstriction since only the latter is sensitive to BQ-123.


Peptides | 2006

Urotensin-II receptor blockade with SB-611812 attenuates cardiac remodeling in experimental ischemic heart disease

Nicolas Bousette; Julia Pottinger; Wisam Ramli; Eliot H. Ohlstein; Dashyant Dhanak; Stephen A. Douglas; Adel Giaid

It is now well established that urotensin-II (UII) levels are increased in several cardiovascular diseases. We previously demonstrated that UII and the UII receptor (UT) protein levels are significantly increased in the hearts of both humans and rats with congestive heart failure (CHF). We have also recently demonstrated that UII blockade, with a selective UII antagonist, improves heart function in a rat model of ischemic CHF. Here, we evaluated the attenuation of cardiac remodeling associated with UII antagonism in the same rat model of ischemic CHF. Animals were administered a specific UT receptor antagonist, SB-611812 (30 mg/kg/day, gavage), or vehicle 30 min prior to coronary artery ligation followed by daily treatment for 8 weeks. Myocardial interstitial fibrosis was analyzed by Massons trichrome and picrosirius red staining. RT-PCR analysis was utilized for mRNA expression studies. We used Western blotting to assess levels of collagen types I and III. Mitogenic activity of UII on cultured neonatal cardiac fibroblasts was also evaluated. Following coronary ligation, SB-611812 significantly attenuated both myocardial and endocardial interstitial fibrosis, and reduced collagen type I:III ratio (P<0.01). UII induced proliferation of cardiac fibroblasts and this mitogenic effect was significantly inhibited with 1 microM of SB-611218 (P<0.05). We demonstrate here that selective blockade of UT reduces diastolic dysfunction by decreasing myocardial fibrosis post-coronary ligation in vivo, and inhibits UII-mediated fibroblast proliferation in vitro.

Collaboration


Dive into the Stephen A. Douglas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Jin

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge