Network


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

Hotspot


Dive into the research topics where Michael S. Wolin is active.

Publication


Featured researches published by Michael S. Wolin.


Circulation Research | 1995

Native Low-Density Lipoprotein Increases Endothelial Cell Nitric Oxide Synthase Generation of Superoxide Anion

Kirkwood A. Pritchard; Laura Groszek; David M. Smalley; William C. Sessa; Mingdan Wu; Patricio Villalon; Michael S. Wolin; Michael B. Stemerman

To examine mechanisms by which native low-density lipoprotein (n-LDL) perturbs endothelial cell (EC) release of superoxide anion (O2-) and nitric oxide (NO), ECs were incubated with n-LDL at 240 mg cholesterol per deciliter for 4 days with media changes every 24 hours. n-LDL increases EC release of O2- by more than fourfold and increases nitrite production by 57%. In the conditioned media from day-4 incubations, n-LDL increases total nitrogen oxides 20 times control EC (C-EC) levels. However, n-LDL did not alter EC NO synthase (eNOS) enzyme activity as measured by the [3H]citrulline assay. N omega-Nitro-L-arginine methyl ester, a specific inhibitor of eNOS activity, increases C-EC release of O2- by > 300% but decreases LDL-treated EC (LDL-EC) release by > 95%. L-Arginine inhibits the release of O2- from LDL-ECs by > 95% but did not effect C-EC release of O2-. Indomethacin and SKF 525A partially attenuate LDL-induced increases in O2- production by approximately 50% and 30%, respectively. Thus, n-LDL increases O2- and NO production, which increases the likelihood of the formation of peroxynitrite (ONOO-), a potent oxidant. n-LDL increases the levels of nitrotyrosine, a stable oxidation product of ONOO-, and tyrosine by approximately 50%. In spite of this increase in oxidative metabolism, analysis of thiobarbituric acid substances reveals that no significant changes in the oxidation of n-LDL occur during the 24-hour incubations with ECs.(ABSTRACT TRUNCATED AT 250 WORDS)


Arteriosclerosis, Thrombosis, and Vascular Biology | 2000

Interactions of Oxidants With Vascular Signaling Systems

Michael S. Wolin

Individual reactive oxygen species (ROS) and oxidation products of NO interact with vascular signaling mechanisms in ways that appear to have fundamental roles in the control of vascular physiological and pathophysiological function. The activities of ROS-producing systems (including various NADPH and NADH oxidases, xanthine oxidase, and NO synthase) in endothelium and/or vascular smooth muscle are controlled by receptor activation, oxygen tension, metabolic processes, and physiological forces associated with blood pressure and flow. This review focuses on how the chemical properties and metabolic sensing interactions of individual ROS (including superoxide anion, hydrogen peroxide, and peroxynitrite) interact with cellular regulatory systems to produce vascular responses. These species appear to often function through producing selective alterations in individual heme or thiol redox-regulated systems (including guanylate cyclase, cyclooxygenase, mitochondrial electron transport, and tyrosine phosphatases) to initiate physiological responses through signaling pathways that control phospholipases, protein kinases, ion channels, contractile proteins, and gene expression.


Circulation Research | 2002

Aging-Induced Phenotypic Changes and Oxidative Stress Impair Coronary Arteriolar Function

Anna Csiszar; Zoltan Ungvari; John G. Edwards; Pawel M. Kaminski; Michael S. Wolin; Akos Koller; Gabor Kaley

We aimed to elucidate the possible role of phenotypic alterations and oxidative stress in age-related endothelial dysfunction of coronary arterioles. Arterioles were isolated from the hearts of young adult (Y, 14 weeks) and aged (A, 80 weeks) male Sprague-Dawley rats. For videomicroscopy, pressure-induced tone of Y and A arterioles and their passive diameter did not differ significantly. In A, arterioles L-NAME (a NO synthase blocker)–sensitive flow-induced dilations were significantly impaired (Y: 41±8% versus A: 3±2%), which could be augmented by superoxide dismutase (SOD) or Tiron (but not l-arginine or the TXA2 receptor antagonist SQ29,548). For lucigenin chemiluminescence, O2·− generation was significantly greater in A than Y vessels and could be inhibited with SOD and diphenyliodonium. NADH-driven O2·− generation was also greater in A vessels. Both endothelial and smooth muscle cells of A vessels produced O2·− (shown with ethidium bromide fluorescence). For Western blotting, expression of eNOS and COX-1 was decreased in A compared with Y arterioles, whereas expressions of COX-2, Cu/Zn-SOD, Mn-SOD, xanthine oxidase, and the NAD(P)H oxidase subunits p47phox, p67phox, Mox-1, and p22phox did not differ. Aged arterioles showed an increased expression of iNOS, confined to the endothelium. Decreased eNOS mRNA and increased iNOS mRNA expression in A vessels was shown by quantitative RT-PCR. In vivo formation of peroxynitrite was evidenced by Western blotting, and immunohistochemistry showing increased 3-nitrotyrosine content in A vessels. Thus, aging induces changes in the phenotype of coronary arterioles that could contribute to the development of oxidative stress, which impairs NO-mediated dilations.


Circulation Research | 1994

Role of nitric oxide in the regulation of oxygen consumption in conscious dogs.

Weiqun Shen; Xiaobin Xu; Manuel Ochoa; Gong Zhao; Michael S. Wolin; Thomas H. Hintze

The role of nitric oxide (NO) in the regulation of O2 consumption was studied in chronically instrumented conscious dogs. A specific NO synthesis inhibitor, nitro-L-arginine (NLA, 30 mg/kg i.v.), significantly increased mean arterial pressure from 100 +/- 4 to 134 +/- 5 mm Hg (mean +/- SEM) and total peripheral resistance by 157 +/- 16% and reduced cardiac output by 47 +/- 3% and heart rate by 34 +/- 6% after 120 minutes. Changes in arterial blood gases were not observed. There were significant changes in PO2 (-14 +/- 2 mm Hg), O2 saturation (-21 +/- 2%), the percentage of hemoglobin as oxyhemoglobin (-21 +/- 2%), and O2 content (-3.0 +/- 0.9 vol%) and a significant increase in percent reduced hemoglobin (21 +/- 1%) in mixed venous blood, associated with an increase in O2 extraction (5.1 +/- 0.2 vol%) (all P < .01). O2 consumption was increased from 124 +/- 6 to 155 +/- 9 mL/min (P < .05). Methoxamine, titrated to have hemodynamic effects similar to those of NLA (eg, mean arterial pressure increased from 97 +/- 4 to 131 +/- 5 mm Hg), had much smaller effects on venous blood gases, hemoglobin, and O2 extraction (2.3 +/- 0.7 vol%) and no significant effect on O2 consumption. NLA also caused an increase in O2 consumption of 37 +/- 8% (P < .01) in quietly resting conscious dogs that had undergone pretreatment with hexamethonium and atropine, but no significant change in O2 consumption in dogs anesthetized with barbiturate.(ABSTRACT TRUNCATED AT 250 WORDS)


Circulation | 1995

Nitric Oxide An Important Signaling Mechanism Between Vascular Endothelium and Parenchymal Cells in the Regulation of Oxygen Consumption

Weiqun Shen; Thomas H. Hintze; Michael S. Wolin

BACKGROUND Nitric oxide (NO) is known to be an inhibitor of mitochondrial function. However, the physiological significance of endothelium-derived NO in the control of tissue respiration is not established. METHODS AND RESULTS Tissue O2 consumption by skeletal muscle slices of the triceps brachii of normal dogs was measured with a Clark-type O2 electrode/tissue bath system at 37 degrees C. S-Nitroso-N-acetylpenicillamine (SNAP), carbachol (CCh), or bradykinin (BK) decreased tissue O2 consumption by 12 +/- 3% to 55 +/- 8%, 15 +/- 6% to 36 +/- 11%, or 21 +/- 5% to 42 +/- 4% at doses of 10(-7) to 10(-4) mol/L, respectively. The effects of both CCh and BK but not SNAP were eliminated by nitro-L-arginine (NLA, 10(-4) mol/L), consistent with SNAP decomposing to release NO and both CCh and BK stimulating endogenous NO production from L-arginine. Oxygen consumption was also decreased by 8-bromo-cGMP. The mitochondrial uncoupler dinitrophenol blocked the effects of 8-bromo-cGMP but only slightly altered those of SNAP, indicating that the major site of action of NO is the mitochondria. In normal, chronically instrumented, resting conscious dogs, blockade of NO synthase by NLA increased mean arterial pressure by 28 +/- 2.5 mm Hg and hind limb vascular resistance by 114 +/- 12% and decreased blood flow by 39 +/- 3%. Most important, NLA also increased O2 uptake by 55 +/- 9% in hind limb skeletal muscle (P < .05), associated with decreases in PO2 and O2 saturation and an increase in reduced hemoglobin in hind limb venous blood. CONCLUSIONS Our results indicate that NO release from vascular endothelial cells appears to play an important physiological role in the regulation of tissue mitochondrial respiration in skeletal muscle and perhaps other organ systems.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2003

Increased Superoxide Production in Coronary Arteries in Hyperhomocysteinemia Role of Tumor Necrosis Factor-α, NAD(P)H Oxidase, and Inducible Nitric Oxide Synthase

Zoltan Ungvari; Anna Csiszar; John G. Edwards; Pawel M. Kaminski; Michael S. Wolin; Gabor Kaley; Akos Koller

Objective—In coronary arteries, hyperhomocysteinemia (HHcy, a known risk factor for coronary heart disease) impairs flow-induced dilations, which can be reversed by superoxide dismutase (SOD). To evidence increased O2.− generation and elucidate its source, we characterized changes in activity (lucigenin chemiluminescence, hydroethidine staining) and expression of arterial pro- and antioxidant systems (Western blotting, immunohistochemistry, cDNA microarray, reverse-transcription polymerase chain reaction) in the coronary arteries of rats by using methionine diet-induced HHcy. Methods and Results—The increased generation of O2.− by HHcy coronary arteries was inhibited by SOD, diphenyleneiodonium, apocynin, and apocynin plus amino guanidine but was unaffected by allopurinol and rotenone. Also, diphenyleneiodonium-sensitive NADPH-driven O2.− generation was increased in HHcy vessels. In HHcy arteries expression of the smooth muscle-confined NAD(P)H oxidase subunit nox1 and that of iNOS was increased. Expression of p67phox, p22phox, and p47phox subunits and that of endothelial nitric oxide synthase, Cu,Zn-SOD, Mn-SOD, extracellular SOD (mRNA), and xanthine oxidase was unchanged. Microarray analysis showed increased expression of tumor necrosis factor (TNF)-&agr; (confirmed by reverse-transcription polymerase chain reaction, Western blotting, and immunohistochemistry) that was localized in smooth muscle. In vitro incubation (18 hours) of HHcy arteries with anti-TNF-&agr; antibody decreased O2.− production, whereas incubation of control vessels with TNF-&agr; increased O2.− generation and nox1 expression. Conclusions—In coronary arteries, HHcy increases TNF-&agr; expression, which enhances oxidative stress through upregulating a nox1-based NAD(P)H oxidase and inducible nitric oxide synthase. Thus, TNF-&agr; induces a proinflammatory vascular phenotype in HHcy that potentially contributes to the development of coronary atherosclerosis.


Circulation | 2003

Potent Metalloporphyrin Peroxynitrite Decomposition Catalyst Protects Against the Development of Doxorubicin-Induced Cardiac Dysfunction

Pál Pacher; Lucas Liaudet; Péter Bai; Jon G. Mabley; Pawel M. Kaminski; László Virág; Amitabha Deb; Éva Szabó; Zoltan Ungvari; Michael S. Wolin; John T. Groves; Csaba Szabó

Background—Increased oxidative stress and dysregulation of nitric oxide have been implicated in the cardiotoxicity of doxorubicin (DOX), a commonly used antitumor agent. Peroxynitrite is a reactive oxidant produced from nitric oxide and superoxide in various forms of cardiac injury. Using a novel metalloporphyrinic peroxynitrite decomposition catalyst, FP15, and nitric oxide synthase inhibitors or knockout mice, we now delineate the pathogenetic role of peroxynitrite in rodent models of DOX-induced cardiac dysfunction. Methods and Results—Mice received a single injection of DOX (25 mg/kg IP). Five days after DOX administration, left ventricular performance was significantly depressed, and high mortality was noted. Treatment with FP15 and an inducible nitric oxide synthase inhibitor, aminoguanidine, reduced DOX-induced mortality and improved cardiac function. Genetic deletion of the inducible nitric oxide synthase gene was also accompanied by better preservation of cardiac performance. In contrast, inhibition of the endothelial isoform of nitric oxide synthase with N-nitro-l-arginine methyl ester increased DOX-induced mortality. FP15 reduced the DOX-induced increase in serum LDH and creatine kinase activities. Furthermore, FP15 prevented the DOX-induced increase in lipid peroxidation, nitrotyrosine formation, and metalloproteinase activation in the heart but not NAD(P)H-driven superoxide generation. Peroxynitrite neutralization did not interfere with the antitumor effect of DOX. FP15 also decreased ischemic injury in rats and improved cardiac function and survival of mice in a chronic model of DOX-induced cardiotoxicity. Conclusions—Thus, peroxynitrite plays a key role in the pathogenesis of DOX-induced cardiac failure. Targeting peroxynitrite formation may represent a new cardioprotective strategy after DOX exposure or in other conditions associated with peroxynitrite formation, including myocardial ischemia/reperfusion injury.


Circulation Research | 1996

Role of Endothelium-Derived Nitric Oxide in the Modulation of Canine Myocardial Mitochondrial Respiration In Vitro: Implications for the Development of Heart Failure

Yi-Wu Xie; Weiqun Shen; Gong Zhao; Xiaobin Xu; Michael S. Wolin; Thomas H. Hintze

The mechanism responsible for the regulation of cardiac function by endogenous nitric oxide (NO) remains unclear. In this investigation, O2 consumption by freshly isolated myocardial muscle segments from the left ventricular free wall of canine hearts was quantified by a Clark-type O2 electrode at 37 degrees C. S-nitroso-N-acetylpenicillamine (SNAP, 9 +/- 3% to 50 +/- 8%), bradykinin (BK, 14 +/- 3% to 30 +/- 5%), or carbachol (CCh, 15 +/- 4% to 29 +/- 4%) significantly attenuated tissue O2 consumption at doses of 10(-7) to 10(-4) mol/L (mean +/- SE, P < .05). The effects of BK and CCh, but not SNAP, were blocked by 10(-4) mol/L NG-nitro-L-arginine, consistent with both BK and CCh stimulating NO biosynthesis and with SNAP decomposing to release NO, respectively. Similar doses of 8-Br-cGMP caused a respiratory inhibition, but to a lesser extent (9 +/- 2% to 14 +/- 6%). A mitochondrial uncoupler, 2,4-dinitrophenol (at 1 mmol/L), blocked the effects of 8-Br-cGMP, but not those of SNAP, BK, or CCh, suggesting that the major site of action of NO is on mitochondrial electron transport. Myocardial muscle from dogs with pacing-induced heart failure had a basal O2 consumption rate of 251 +/- 21 nmol.min-1.g-1, which was 54% higher than the rate seen in muscle from normal healthy canine hearts. The inhibitory effects of BK and CCh on O2 consumption were not observed in failing cardiac tissue, but SNAP showed an unaltered inhibitory effect. Therefore, our results indicate that NO released from microvascular endothelium by BK, stimulation of muscarinic receptors, and perhaps flow velocity may play an important physiological role in the control of cardiac mitochondrial respiration, and the loss of this regulatory function may contribute to the development of heart failure.


Circulation Research | 1991

Inhibition of coronary artery superoxide dismutase attenuates endothelium-dependent and -independent nitrovasodilator relaxation.

Hatim A. Omar; P. D. Cherry; Michael P. Mortelliti; Theresa Burke-Wolin; Michael S. Wolin

Isolated bovine coronary arteries were treated with 10 mM diethyldithiocarbamate (DETCA) for 30 minutes to deplete the cytosolic ZnCu form of superoxide dismutase (SOD). This treatment completely inhibited the endothelium- and cGMP-dependent relaxation to acetylcholine (mediated via the endothelium-derived relaxing factor, which is thought to be nitric oxide) without significantly inhibiting endothelium-dependent relaxation to arachidonic acid (mediated by prostaglandins). DETCA treatment of endothelial cells cultured from the coronary arteries inhibited bradykinin-elicited release of endothelium-derived relaxing factor, which was detected by bioassay on an isolated rabbit aorta in the presence of extracellular SOD. DETCA also inhibited cGMP-associated relaxations to nitric oxide and to vasodilators thought to function via the generation of this mediator (nitroglycerin and nitroprusside), but cAMP-associated relaxations to isoproterenol and papaverine were not altered. The inhibitory effects of DETCA against the relaxation to nitroprusside and nitroglycerin were attenuated by severe hypoxia. DETCA treatment of isolated coronary arterial smooth muscle or cultured endothelial cells produced an increase of chemiluminescence elicited in the presence of lucigenin, a detector of superoxide anion generation. The addition of SOD markedly attenuated the effects of DETCA treatment on arterial relaxation and chemiluminescence. Therefore, control of cellular superoxide anion levels by endogenous SOD appears needed for the release of endothelium-derived relaxing factor and relaxation of vascular smooth muscle to nitrovasodilators mediated via cGMP in the bovine coronary artery, but SOD is not critical for other endothelium-dependent or cAMP-associated relaxant mechanisms.


Circulation | 2008

Reversal of Cardiac Hypertrophy and Fibrosis From Pressure Overload by Tetrahydrobiopterin: Efficacy of Recoupling Nitric Oxide Synthase as a Therapeutic Strategy

An L. Moens; Eiki Takimoto; Carlo G. Tocchetti; Khalid Chakir; Djahida Bedja; Gianfranco Cormaci; Elizabeth A. Ketner; Maulik D. Majmudar; Kathleen L. Gabrielson; Marc K. Halushka; James B. Mitchell; Shyam Biswal; Keith M. Channon; Michael S. Wolin; N J Alp; Nazareno Paolocci; Hunter C. Champion; David A. Kass

Background— Sustained pressure overload induces pathological cardiac hypertrophy and dysfunction. Oxidative stress linked to nitric oxide synthase (NOS) uncoupling may play an important role. We tested whether tetrahydrobiopterin (BH4) can recouple NOS and reverse preestablished advanced hypertrophy, fibrosis, and dysfunction. Methods and Results— C57/Bl6 mice underwent transverse aortic constriction for 4 weeks, increasing cardiac mass (190%) and diastolic dimension (144%), lowering ejection fraction (−46%), and triggering NOS uncoupling and oxidative stress. Oral BH4 was then administered for 5 more weeks of pressure overload. Without reducing loading, BH4 reversed hypertrophy and fibrosis, recoupled endothelial NOS, lowered oxidant stress, and improved chamber and myocyte function, whereas untreated hearts worsened. If BH4 was started at the onset of pressure overload, it did not suppress hypertrophy over the first week when NOS activity remained preserved even in untreated transverse aortic constriction hearts. However, BH4 stopped subsequent remodeling when NOS activity was otherwise declining. A broad antioxidant, Tempol, also reduced oxidant stress yet did not recouple NOS or reverse worsened hypertrophy/fibrosis from sustained transverse aortic constriction. Microarray analysis revealed very different gene expression profiles for both treatments. BH4 did not enhance net protein kinase G activity. Finally, transgenic mice with enhanced BH4 synthesis confined to endothelial cells were unprotected against pressure overload, indicating that exogenous BH4 targeted myocytes and fibroblasts. Conclusions— NOS recoupling by exogenous BH4 ameliorates preexisting advanced cardiac hypertrophy/fibrosis and is more effective than a less targeted antioxidant approach (Tempol). These data highlight the importance of myocyte NOS uncoupling in hypertrophic heart disease and support BH4 as a potential new approach to treat this disorder.

Collaboration


Dive into the Michael S. Wolin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sachin A. Gupte

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mansoor Ahmad

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Sharath Kandhi

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Zoltan Ungvari

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Gabor Kaley

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Dhara Patel

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Dong Sun

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Akos Koller

New York Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge