Network


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

Hotspot


Dive into the research topics where Michael E. Mendelsohn is active.

Publication


Featured researches published by Michael E. Mendelsohn.


Journal of Clinical Investigation | 1990

Impaired vasodilation of forearm resistance vessels in hypercholesterolemic humans.

Mark A. Creager; John P. Cooke; Michael E. Mendelsohn; Shelly J. Gallagher; Sharon M. Coleman; Joseph Loscalzo; Victor J. Dzau

The effect of hypercholesterolemia on vascular function was studied in humans. To eliminate the potential confounding effects of atherosclerosis, vascular reactivity was measured in the forearm resistance vessels of 11 normal subjects (serum LDL cholesterol = 111 +/- 7 mg/dl) and 13 patients with hypercholesterolemia (serum LDL cholesterol = 211 +/- 19 mg/dl, P less than 0.05). Each subject received intrabrachial artery infusions of methacholine, which releases endothelium-derived relaxant factor, and nitroprusside which directly stimulates guanylate cyclase in vascular smooth muscle. Maximal vasodilatory potential was determined during reactive hyperemia. Vasoconstrictive responsiveness was examined during intra-arterial phenylephrine infusion. Forearm blood flow was determined by venous occlusion plethysmography. Basal forearm blood flow in normal and hypercholesterolemic subjects was comparable. Similarly, reactive hyperemic blood flow did not differ between the two groups. In contrast, the maximal forearm blood flow response to methacholine in hypercholesterolemic subjects was less than that observed in normal subjects. In addition, the forearm blood flow response to nitroprusside was less in hypercholesterolemic subjects. There was no difference in the forearm vasoconstrictive response to phenylephrine in the two groups. Thus, the vasodilator responses to methacholine and nitroprusside were blunted in patients with hypercholesterolemia. We conclude that in humans with hypercholesterolemia, there is a decreased effect of nitrovasodilators, including endothelium-derived relaxing factor, on the vascular smooth muscle of resistance vessels.


Journal of Clinical Investigation | 1999

Estrogen receptor a mediates the nongenomic activation of endothelial nitric oxide synthase by estrogen

Zhong Chen; Ivan S. Yuhanna; Zoya Galcheva-Gargova; Richard H. Karas; Michael E. Mendelsohn; Philip W. Shaul

Estrogen is an important vasoprotective molecule that causes the rapid dilation of blood vessels by activating endothelial nitric oxide synthase (eNOS) through an unknown mechanism. In studies of intact ovine endothelial cells, 17beta-estradiol (E2) caused acute (five-minute) activation of eNOS that was unaffected by actinomycin D but was fully inhibited by concomitant acute treatment with specific estrogen receptor (ER) antagonists. Overexpression of the known transcription factor ERalpha led to marked enhancement of the acute response to E2, and this was blocked by ER antagonists, was specific to E2, and required the ERalpha hormone-binding domain. In addition, the acute response of eNOS to E2 was reconstituted in COS-7 cells cotransfected with wild-type ERalpha and eNOS, but not by transfection with eNOS alone. Furthermore, the inhibition of tyrosine kinases or mitogen-activated protein (MAP) kinase kinase prevented the activation of eNOS by E2, and E2 caused rapid ER-dependent activation of MAP kinase. These findings demonstrate that the short-term effects of estrogen central to cardiovascular physiology are mediated by ERalpha functioning in a novel, nongenomic manner to activate eNOS via MAP kinase-dependent mechanisms.


Nature Medicine | 2001

High-density lipoprotein binding to scavenger receptor-BI activates endothelial nitric oxide synthase

Ivan S. Yuhanna; Yan Zhu; Blair E. Cox; Lisa Hahner; Sherri Osborne-Lawrence; Ping Lu; Yves L. Marcel; Richard G. W. Anderson; Michael E. Mendelsohn; Helen H. Hobbs; Philip W. Shaul

Atherosclerosis is the primary cause of cardiovascular disease, and the risk for atherosclerosis is inversely proportional to circulating levels of high-density lipoprotein (HDL) cholesterol. However, the mechanisms by which HDL is atheroprotective are complex and not well understood. Here we show that HDL stimulates endothelial nitric oxide synthase (eNOS) in cultured endothelial cells. In contrast, eNOS is not activated by purified forms of the major HDL apolipoproteins ApoA-I and ApoA-II or by low-density lipoprotein. Heterologous expression experiments in Chinese hamster ovary cells reveal that scavenger receptor-BI (SR-BI) mediates the effects of HDL on the enzyme. HDL activation of eNOS is demonstrable in isolated endothelial-cell caveolae where SR-BI and eNOS are colocalized, and the response in isolated plasma membranes is blocked by antibodies to ApoA-I and SR-BI, but not by antibody to ApoA-II. HDL also enhances endothelium- and nitric-oxide–dependent relaxation in aortae from wild-type mice, but not in aortae from homozygous null SR-BI knockout mice. Thus, HDL activates eNOS via SR-BI through a process that requires ApoA-I binding. The resulting increase in nitric-oxide production might be critical to the atheroprotective properties of HDL and ApoA-I.


Circulation | 1994

Human vascular smooth muscle cells contain functional estrogen receptor.

Richard H. Karas; B L Patterson; Michael E. Mendelsohn

BACKGROUND The decreased incidence of coronary artery disease observed in postmenopausal women given estrogen (E2) replacement demonstrates an atheroprotective effect of E2 that is generally believed to be mediated by indirect, E2-induced changes in cardiovascular risk factor profiles. We hypothesized that the atheroprotective effect of E2 may be in part mediated by a direct effect of E2 on vascular smooth muscle cells (VSMCs). Therefore, a series of experiments was performed to determine whether human VSMCs contain a competent E2 receptor, a ligand-activated transcription factor known to mediate E2-induced effects in nonvascular cells. METHODS AND RESULTS Ribonuclease protection assays, with a probe derived from the human E2 receptor, were used to demonstrate E2-receptor mRNA in human saphenous vein VSMCs. To show that VSMCs contain E2-receptor protein as well as message, immunoblotting and immunofluorescence studies with a monoclonal anti-E2-receptor antibody were performed, and E2-receptor protein was detected by both methods. Transient transfection assays using a specific E2-responsive reporter system were used next to determine whether the VSMC E2 receptor is capable of E2-induced transcriptional transactivation. Initial studies using mammary artery-derived VSMCs resulted in a 2.4-fold increase in reporter activity in response to 10(-7) mol/L E2. Subsequent studies using saphenous vein VSMCs demonstrated increasing levels of reporter activation as the concentration of E2 was increased from 10(-9) mol/L (1.3-fold increase; SEM, 0.07; P = .05, n = 3) to 10(-7) mol/L (1.6-fold increase; SEM, 0.04; P = .002, n = 6). The specificity of the E2-induced transactivation of the reporter gene was shown by dose-dependent inhibition of transactivation by the pure E2 antagonist ICI 164,384 and by enhancement of the transactivation by simultaneous overexpression of the E2 receptor. CONCLUSIONS We have demonstrated for the first time that human VSMCs express E2-receptor mRNA and protein and that the E2 receptor in VSMCs is capable of estrogen-dependent gene activation. These data suggest a mechanism by which estrogen may directly alter VSMC function.


Nature Medicine | 2003

Regulator of G-protein signaling-2 mediates vascular smooth muscle relaxation and blood pressure

Mary Tang; Guang Wang; Ping Lu; Richard H. Karas; Mark Aronovitz; Scott P. Heximer; Kevin M. Kaltenbronn; Kendall J. Blumer; David P. Siderovski; Yan Zhu; Michael E. Mendelsohn

Nitric oxide (NO) inhibits vascular contraction by activating cGMP-dependent protein kinase I-α (PKGI-α), which causes dephosphorylation of myosin light chain (MLC) and vascular smooth muscle relaxation. Here we show that PKGI-α attenuates signaling by the thrombin receptor protease-activated receptor-1 (PAR-1) through direct activation of regulator of G-protein signaling-2 (RGS-2). NO donors and cGMP cause cGMP-mediated inhibition of PAR-1 and membrane localization of RGS-2. PKGI-α binds directly to and phosphorylates RGS-2, which significantly increases GTPase activity of Gq, terminating PAR-1 signaling. Disruption of the RGS-2–PKGI-α interaction reverses inhibition of PAR-1 signaling by nitrovasodilators and cGMP. Rgs2−/− mice develop marked hypertension, and their blood vessels show enhanced contraction and decreased cGMP-mediated relaxation. Thus, PKGI-α binds to, phosphorylates and activates RGS-2, attenuating receptor-mediated vascular contraction. Our study shows that RGS-2 is required for normal vascular function and blood pressure and is a new drug development target for hypertension.


Circulation Research | 2002

Estrogen Receptor-α Mediates the Protective Effects of Estrogen Against Vascular Injury

Gary Pare; Andrée Krust; Richard H. Karas; Sonia Dupont; Mark Aronovitz; Pierre Chambon; Michael E. Mendelsohn

Blood vessel cells express the 2 known estrogen receptors, &agr; and &bgr; (ER&agr;, ER&bgr;), which are thought to mediate estrogen inhibition of vascular injury and atherosclerosis, but the relative role of ER&agr; and ER&bgr; in these events is controversial. Estrogen inhibits the vascular injury response to the same extent in ovariectomized female wild-type mice and in the original single gene knockout mice for ER&agr; (ER&agr;KOChapel Hill [ER&agr;KOCH]) and ER&bgr; (ER&bgr;KOChapel Hill [ER&bgr;KOCH]). In double gene knockout mice generated by crossing these animals (ER&agr;,&bgr;KOCH), estrogen no longer inhibits medial thickening after vascular injury, but still inhibits vascular smooth muscle cell proliferation and increases uterine weight. The partial retention of estrogen responsiveness in ER&agr;,&bgr;KOCH mice could be due either to the presence of a novel, unidentified estrogen receptor or to functional expression of an estrogen receptor-&agr; splice variant in the parental ER&agr;KOCH mice. To distinguish between these possibilities, we studied recently generated mice fully null for estrogen receptor &agr; (ER&agr;KOStrasbourg [ER&agr;KOSt]) and examined the effect of estrogen on the response to vascular injury. In the present study, we show that after vascular injury in ovariectomized ER&agr;KOSt mice, estrogen has no detectable effect on any measure of vascular injury, including medial area, proteoglycan deposition, or smooth muscle cell proliferation. These data demonstrate that estrogen receptor-&agr; mediates the protective effects of estrogen on the response to vascular injury.


Circulation Research | 1998

Increased Expression of Estrogen Receptor-β mRNA in Male Blood Vessels After Vascular Injury

Volkhard Lindner; Sung K. Kim; Richard H. Karas; George Kuiper; Jan-Åke Gustafsson; Michael E. Mendelsohn

Estrogen exerts direct effects on vascular endothelial and smooth muscle cells that are important for vascular protection. Estrogen receptor-alpha (ERalpha) is expressed in vascular cells from males and females and may mediate some of the effects of estrogen on vascular tissue. However, we recently found that estrogen is able to protect against vascular injury in ovariectomized female ERalpha knockout mice. These mice express the newly described estrogen receptor-beta (ERbeta) in their aortas, suggesting that ERbeta may also mediate some of the direct effects of estrogen on the vasculature. In this study, the level of expression of ERalpha and ERbeta mRNA in male rat aortas was examined before and after vascular injury using en face (Häutchen) preparations and in situ hybridization. Little or no change in ERalpha expression was observed after vascular injury in either vascular endothelial or smooth muscle cells at any time point. In contrast, ERbeta mRNA was found to be expressed markedly after balloon injury. In endothelial cells, ERbeta was increased by 2 days after injury, and high levels of expression were maintained at 8 and 14 days. Furthermore, ERbeta expression was high in luminal smooth muscle cells at 8 and 14 days after injury and had decreased to low levels by 28 days after injury. These data demonstrate the presence of ERbeta in male vascular tissues and the induction of ERbeta mRNA expression after vascular injury, supporting a role for ERbeta in the direct vascular effects of estrogen.


Current Opinion in Cardiology | 1994

Estrogen and the blood vessel wall.

Michael E. Mendelsohn; Richard H. Karas

This article reviews historical studies and recent advances regarding the direct effects of estrogen on the blood vessel wall. It is organized into two sections that summarize effects of estrogen on vasomotor tone and on vascular cell growth and atherogenesis, based on two recognized actions of estrogen on the vasculature: a rapid vasodilatory effect, and an atheroprotective effect involving inhibition of smooth-muscle cell proliferation. These effects are likely mediated by different mechanisms. The rapid vasodilatory effects of estrogen are probably nongenomic, whereas the antiproliferative effects of estrogen are likely due to estrogen receptor-dependent alterations in gene expression. Overlap between these two mechanisms also exists, in that genes regulating the production of two important vasodilators synthesized by the vessel wall (prostacyclin and nitric oxide) can be up-regulated by estrogen. Potential molecular mechanisms by which estrogen exerts its effects are discussed, and future directions in this rapidly evolving area of research are considered.


Circulation Research | 2005

Angiotensin II and Aldosterone Regulate Gene Transcription Via Functional Mineralocortocoid Receptors in Human Coronary Artery Smooth Muscle Cells

Iris Z. Jaffe; Michael E. Mendelsohn

Inhibition or blockade of the angiotensin-aldosterone system consistently decreases ischemic cardiovascular events in clinical trials. The steroid hormone aldosterone acts by binding to the mineralocorticoid receptor (MR), a ligand activated transcription factor that is a member of the nuclear hormone receptor superfamily. MR binds and is activated by aldosterone and cortisol with equal affinity, but MR activation by cortisol is diminished in tissues that express the cortisol-inactivating enzyme 11-&bgr;-hydroxysteroid-dehydrogenase-2 (11&bgr;HSD2). Although previous studies support that the vasculature is a target tissue of aldosterone, MR-mediated gene expression in vascular cells has not been demonstrated or systematically explored. We investigated whether functional MR and 11&bgr;HSD2 are expressed in human blood vessels. Human coronary and aortic vascular smooth muscle cells (VSMCs) express mRNA and protein for both MR and 11&bgr;HSD2. The endogenous VSMC MR mediates aldosterone-dependent gene expression, which is blocked by the competitive MR antagonist spironolactone. Inhibition of 11&bgr;HSD2 in coronary artery VSMCs enhances gene transactivation by cortisol, supporting that the VSMC 11&bgr;HSD2 is functional. Angiotensin II also activates MR-mediated gene transcription in coronary artery VSMCs. Angiotensin II activation of MR-mediated gene expression is inhibited by both the AT1 receptor blocker losartan and by spironolactone, but not by aldosterone synthase inhibition. Microarray and quantitative RT-PCR experiments show that aldosterone activates expression of endogenous human coronary VSMC genes, including several involved in vascular fibrosis, inflammation, and calcification. These data support a new MR-dependent mechanism by which aldosterone and angiotensin II influence ischemic cardiovascular events, and suggest that ACE inhibitors and MR antagonists may decrease clinical ischemic events by inhibiting MR-dependent gene expression in vascular cells.


Circulation Research | 2004

17β-Estradiol Reduces Cardiomyocyte Apoptosis In Vivo and In Vitro via Activation of Phospho-Inositide-3 Kinase/Akt Signaling

Richard D. Patten; Isaac Pourati; Mark Aronovitz; Jason Baur; Flore Celestin; Xin Chen; Ashour Michael; Syed Haq; Simone Nuedling; Christian Grohé; Thomas Force; Michael E. Mendelsohn; Richard H. Karas

Female gender and estrogen-replacement therapy in postmenopausal women are associated with improved heart failure survival, and physiological replacement of 17&bgr;-estradiol (E2) reduces infarct size and cardiomyocyte apoptosis in animal models of myocardial infarction (MI). Here, we characterize the molecular mechanisms of E2 effects on cardiomyocyte survival in vivo and in vitro. Ovariectomized female mice were treated with placebo or physiological E2 replacement, followed by coronary artery ligation (placebo-MI or E2-MI) or sham operation (sham) and hearts were harvested 6, 24, and 72 hours later. After MI, E2 replacement significantly increased activation of the prosurvival kinase, Akt, and decreased cardiomyocyte apoptosis assessed by terminal deoxynucleotidyltransferase dUTP nick-end labeling (TUNEL) staining and caspase 3 activation. In vitro, E2 at 1 or 10 nmol/L caused a rapid 2.7-fold increase in Akt phosphorylation and a decrease in apoptosis as measured by TUNEL staining, caspase 3 activation, and DNA laddering in cultured neonatal rat cardiomyocytes. The E2-mediated reduction in apoptosis was reversed by an estrogen receptor (ER) antagonist, ICI 182,780, and by phospho-inositide-3 kinase inhibitors, LY294002 and Wortmannin. Overexpression of a dominant negative-Akt construct also blocked E2-mediated reduction in cardiomyocyte apoptosis. These data show that E2 reduces cardiomyocyte apoptosis in vivo and in vitro by ER- and phospho-inositide-3 kinase–Akt–dependent pathways and support the relevance of these pathways in the observed estrogen-mediated reduction in myocardial injury.

Collaboration


Dive into the Michael E. Mendelsohn's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David E. Housman

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Kass

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Amanda M. Shearman

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Philip W. Shaul

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yan Zhu

Tianjin University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Inga Peter

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge