Network


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

Hotspot


Dive into the research topics where Wendy Baur is active.

Publication


Featured researches published by Wendy Baur.


Circulation Research | 2008

Functional Mineralocorticoid Receptors in Human Vascular Endothelial Cells Regulate Intercellular Adhesion Molecule-1 Expression and Promote Leukocyte Adhesion

Massimiliano Caprio; Brenna G. Newfell; Andrea la Sala; Wendy Baur; Andrea Fabbri; Giuseppe Rosano; Michael E. Mendelsohn; Iris Z. Jaffe

In clinical trials, aldosterone antagonists decrease cardiovascular mortality and ischemia by unknown mechanisms. The steroid hormone aldosterone acts by binding to the mineralocorticoid receptor (MR), a ligand-activated transcription factor. In humans, aldosterone causes MR-dependent endothelial cell (EC) dysfunction and in animal models, aldosterone increases vascular macrophage infiltration and atherosclerosis. MR antagonists inhibit these effects without changing blood pressure, suggesting a direct role for vascular MR in EC function and atherosclerosis. Whether human vascular ECs express functional MR is not known. Here, we show that human coronary artery and aortic ECs express MR mRNA and protein and that EC MR mediates aldosterone-dependent gene transcription. Human ECs also express the enzyme 11-&bgr;-hydroxysteroid dehydrogenase-2 (11&bgr;HSD2), and inhibition of 11&bgr;HSD2 in aortic ECs enhances gene transactivation by cortisol, supporting that EC 11&bgr;HSD2 is functional. Furthermore, aldosterone stimulates transcription of the proatherogenic leukocyte–EC adhesion molecule intercellular adhesion molecule (ICAM)1 gene and protein expression on human coronary artery ECs, an effect inhibited by the MR antagonist spironolactone and by MR knock down with small interfering RNA. Cell adhesion assays demonstrate that aldosterone promotes leukocyte–EC adhesion, an effect that is inhibited by spironolactone and ICAM1 blocking antibody, supporting that aldosterone induction of EC ICAM1 surface expression via MR mediates leukocyte–EC adhesion. These data show that aldosterone activates endogenous EC MR and proatherogenic gene expression in clinically important human ECs. These studies describe a novel mechanism by which aldosterone may influence ischemic cardiovascular events and support a new explanation for the decrease in ischemic events in patients treated with aldosterone antagonists.


Circulation Research | 2009

ROCK Isoform Regulation of Myosin Phosphatase and Contractility in Vascular Smooth Muscle Cells

Yuepeng Wang; Xiaoyu Rayne Zheng; Nadeene Riddick; Meredith Bryden; Wendy Baur; Xin Zhang; Howard K. Surks

Abnormal vascular smooth muscle cell (VSMC) contraction plays an important role in vascular diseases. The RhoA/ROCK signaling pathway is now well recognized to mediate vascular smooth muscle contraction in response to vasoconstrictors by inhibiting myosin phosphatase (MLCP) activity and increasing myosin light chain phosphorylation. Two ROCK isoforms, ROCK1 and ROCK2, are expressed in many tissues, yet the isoform-specific roles of ROCK1 and ROCK2 in vascular smooth muscle and the mechanism of ROCK-mediated regulation of MLCP are not well understood. In this study, ROCK2, but not ROCK1, bound directly to the myosin binding subunit of MLCP, yet both ROCK isoforms regulated MLCP and myosin light chain phosphorylation. Despite that both ROCK1 and ROCK2 regulated MLCP, the ROCK isoforms had distinct and opposing effects on VSMC morphology and ROCK2, but not ROCK1, had a predominant role in VSMC contractility. These data support that although the ROCK isoforms both regulate MLCP and myosin light chain phosphorylation through different mechanisms, they have distinct roles in VSMC function.


Proceedings of the National Academy of Sciences of the United States of America | 2008

High blood pressure arising from a defect in vascular function

Simon K. Michael; Howard K. Surks; Yuepeng Wang; Yan Zhu; Robert M. Blanton; Michelle Jamnongjit; Mark Aronovitz; Wendy Baur; Ken-ichi Ohtani; Michael K. Wilkerson; Adrian D. Bonev; Mark T. Nelson; Richard H. Karas; Michael E. Mendelsohn

Hypertension, a major cardiovascular risk factor and cause of mortality worldwide, is thought to arise from primary renal abnormalities. However, the etiology of most cases of hypertension remains unexplained. Vascular tone, an important determinant of blood pressure, is regulated by nitric oxide, which causes vascular relaxation by increasing intracellular cGMP and activating cGMP-dependent protein kinase I (PKGI). Here we show that mice with a selective mutation in the N-terminal protein interaction domain of PKGIα display inherited vascular smooth muscle cell abnormalities of contraction, abnormal relaxation of large and resistance blood vessels, and increased systemic blood pressure. Renal function studies and responses to changes in dietary sodium in the PKGIα mutant mice are normal. These data reveal that PKGIα is required for normal VSMC physiology and support the idea that high blood pressure can arise from a primary abnormality of vascular smooth muscle cell contractile regulation, suggesting a new approach to the diagnosis and therapy of hypertension and cardiovascular diseases.


Circulation | 2012

Rapid Estrogen Receptor Signaling is Essential for the Protective Effects of Estrogen Against Vascular Injury

Sophie J. Bernelot Moens; Gavin R. Schnitzler; Moriah Nickerson; Huiming Guo; Kazutaka Ueda; Qing Lu; Mark Aronovitz; Heather Nickerson; Wendy Baur; Ulla Hansen; Lakshmanan K. Iyer; Richard H. Karas

Background— Clinical trial and epidemiological data support that the cardiovascular effects of estrogen are complex, including a mixture of both potentially beneficial and harmful effects. In animal models, estrogen protects females from vascular injury and inhibits atherosclerosis. These effects are mediated by estrogen receptors (ERs), which, when bound to estrogen, can bind to DNA to directly regulate transcription. ERs can also activate several cellular kinases by inducing a rapid nonnuclear signaling cascade. However, the biological significance of this rapid signaling pathway has been unclear. Methods and Results— In the present study, we develop a novel transgenic mouse in which rapid signaling is blocked by overexpression of a peptide that prevents ERs from interacting with the scaffold protein striatin (the disrupting peptide mouse). Microarray analysis of ex vivo treated mouse aortas demonstrates that rapid ER signaling plays an important role in estrogen-mediated gene regulatory responses. Disruption of ER-striatin interactions also eliminates the ability of estrogen to stimulate cultured endothelial cell migration and to inhibit cultured vascular smooth muscle cell growth. The importance of these findings is underscored by in vivo experiments demonstrating loss of estrogen-mediated protection against vascular injury in the disrupting peptide mouse after carotid artery wire injury. Conclusions— Taken together, these results support the concept that rapid, nonnuclear ER signaling contributes to the transcriptional regulatory functions of ER and is essential for many of the vasoprotective effects of estrogen. These findings also identify the rapid ER signaling pathway as a potential target for the development of novel therapeutic agents.


Journal of Clinical Investigation | 1998

Growth factor activation of the estrogen receptor in vascular cells occurs via a mitogen-activated protein kinase-independent pathway.

Richard H. Karas; Elizabeth Ann Gauer; Hallie E. Bieber; Wendy Baur; Michael E. Mendelsohn

The classical estrogen receptor ERalpha mediates many of the known cardiovascular effects of estrogen and is expressed in male and female vascular cells. Estrogen-independent activation of ERalpha is known to occur in cells from reproductive tissues, but has not been investigated previously in vascular cells. In this study, transient transfection assays in human saphenous vein smooth muscle cells (HSVSMC) and pulmonary vein endothelial cells (PVEC) demonstrated ERalpha-dependent activation of estrogen response element-based, and vascular endothelial growth factor-based reporter plasmids by both estrogen-deficient FBS (ED-FBS) and EGF. In nonvascular cells, ERalpha-mediated gene expression can be activated via mitogen-activated protein (MAP) kinase- induced phosphorylation of serine 118 of ERalpha. However, in vascular cells, we found that pharmacologic inhibition of MAP kinase did not alter EGF-mediated ERalpha activation. In addition, a mutant ER containing an alanine-for-serine substitution at position 118 was activated to the same degree as the wild-type receptor by ED-FBS and EGF in both HSVSMC and PVEC. Furthermore, constitutively active MAP kinase kinase (MAPKK) activated ERalpha in Cos1 cells as expected, but MAPKK inhibited ER activation in PVEC. We conclude that growth factors also stimulate ERalpha-mediated gene expression in vascular cells, but find that this occurs via a MAP kinase-independent pathway distinct from that reported previously in nonvascular cells.


FEBS Letters | 2002

MAP kinase mediates growth factor‐induced nuclear translocation of estrogen receptor α

Qing Lu; Heather Ebling; Jens Mittler; Wendy Baur; Richard H. Karas

In addition to mediating the classical transcriptional effects of estrogen, estrogen receptors (ERs) are now known to regulate gene expression in the absence of estrogen by ligand‐independent activation pathways, and to mediate the rapid, non‐genomic effects of estrogen as well. ERs have been shown to associate with the cell membrane, and recent studies demonstrate that this subpopulation of membrane‐associated ER mediates the rapid effects of estrogen. To date, however, little is known regarding the pathways that regulate the distribution of the ER between the nuclear and membrane fractions. In the current study, we demonstrate membrane localization of transiently transfected ERα in human vascular smooth muscle cells, and translocation of ERα from the membrane to the nucleus in response to both estrogen‐dependent and estrogen‐independent stimulation. Mutational analyses identified serine 118 as the critical residue regulating nuclear localization following estrogen‐independent stimulation, but not following estrogen stimulation. Induction of nuclear localization of ERα by estrogen‐independent, but not estrogen‐dependent stimulation was blocked by both pharmacologic and genetic inhibition of mitogen‐activated protein (MAP) kinase activation. Furthermore, constitutive activation of MAP kinase resulted in nuclear translocation of ERα. These overexpression studies support that MAP kinase‐mediated phosphorylation of ERα induces nuclear localization of the ER in response to estrogen‐independent, but not estrogen‐dependent stimulation, demonstrating stimulus‐specific molecular pathways regulate the nuclear localization of the ER. These findings identify a previously unrecognized pathway that regulates the intracellular localization of the ER, and represent the first demonstration that the distribution of the ER between membrane and nuclear compartments is regulated by physiologic stimuli.


FEBS Letters | 1995

Human vascular smooth muscle cells express an estrogen receptor isoform.

Richard H. Karas; Wendy Baur; Martin van Eickles; Michael E. Mendelsohn

In women, estrogen (E2) exerts a clinically relevant anti‐atherogenic effect. The atheroprotective effects of E2 are mediated both by E2‐induced changes in systemic factors and by direct effects of E2 on the blood vessel wall. In studies to characterize E2 signaling pathways in vascular smooth muscle cells (VSMC), we recently demonstrated that human VSMC express a functional estrogen receptor [1]. In the present study, we applied a reverse transcription/PCR‐based strategy to identify isoforms of the E2 receptor in human VSMC. We now report that in addition to the classical E2 receptor, human VSMC derived from both mammary artery and saphenous vein express an estrogen receptor isoform containing an in‐frame deletion of Exon 4 (ERΔ4). RNase protection assays confirm the presence of ERΔ4 message in VSMC and demonstrate it is nearly as abundant as the classical E2 receptor. Transient transfection experiments in VSMC and HeLa cells demonstrate that, in contrast to the classical 67 kDa nuclear‐localized E2 receptor, ERΔ4: (a) is a 55 kDa protein that is widely distributed throughout the cell; (b) does not transactivate an E2 response element‐driven reporter plasmid in response to E2; and (c) does not modulate transactivation of the ERE‐reporter by the classical (wild type) estrogen receptor. Thus, human VSMC express an E2 receptor isoform that does not appear to alter gene transcription. The presence of a novel isoform of the E2 receptor may have important implications for studies of E2‐mediated signaling in VSMC.


Journal of Clinical Investigation | 2001

A complex role for the progesterone receptor in the response to vascular injury

Richard H. Karas; Martin van Eickels; John P. Lydon; Sean P. Roddy; Moon Kwoun; Mark Aronovitz; Wendy Baur; Orla M. Conneely; Bert W. O’Malley; Michael E. Mendelsohn

Clinical studies of hormone replacement therapy to prevent cardiovascular diseases have heightened interest in the cardiovascular effects of progestins. However, the role of the progesterone receptor (PR) in vascular biology has not been studied in vivo. We studied ovariectomized female PR knockout (PRKO) mice and their wild-type (WT) littermates using the mouse carotid artery injury model. Placebo-treated PRKO mice showed significantly greater vascular medial hypertrophy and vascular smooth muscle cell (VSMC) proliferation in response to vascular injury than did WT mice. Progesterone had no significant effect in the PRKO mice, but worsened the response to injury in WT mice. VSMCs cultured from PRKO mouse aortae were markedly hyperproliferative, and their growth was not affected by progesterone. In contrast to the in vivo findings, progesterone inhibited proliferation of WT-derived VSMCs. Furthermore, reintroduction of PR into PRKO-derived VSMCs using adenoviral methods restored progesterone-mediated inhibition of proliferation to these cells. This effect was reversed by the PR antagonist, RU 486. Thus, the effects of PR and progesterone differ markedly between cultured VSMCs and intact blood vessels. These data demonstrate a direct role for the PR in regulating the response to vascular injury and VSMC proliferation.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Estrogen Receptor–Mediated Regulation of MicroRNA Inhibits Proliferation of Vascular Smooth Muscle Cells

Jin Zhao; Gregory A. Imbrie; Wendy Baur; Lakshmanan K. Iyer; Mark Aronovitz; Tanya Kershaw; Greta M. Haselmann; Qing Lu; Richard H. Karas

Objective—Estradiol (E2) regulates gene transcription by activating estrogen receptor-&agr; and estrogen receptor-&bgr;. Many of the genes regulated by E2 via estrogen receptors are repressed, yet the molecular mechanisms that mediate E2-induced gene repression are currently unknown. We hypothesized that E2, acting through estrogen receptors, regulates expression of microRNAs (miRs) leading to repression of expression of specific target genes. Methods and Results—Here, we report that E2 significantly upregulates the expression of 26 miRs and downregulates the expression of 6 miRs in mouse aorta. E2-mediated upregulation of one of these miRs, miR-203, was chosen for further study. In cultured vascular smooth muscle cells (VSMC), E2-mediated upregulation of miR-203 is mediated by estrogen receptor-&agr; (but not estrogen receptor-&bgr;) via transcriptional upregulation of the primary miR. We demonstrate that the transcription factors Zeb-1 and AP-1 play critical roles in mediating E2-induced upregulation of miR-203 transcription. We show further that miR-203 mediates E2-induced repression of Abl1, and p63 protein abundance in VSMC. Finally, knocking-down miR-203 abolishes E2-mediated inhibition of VSMC proliferation, and overexpression of miR-203 inhibits cultured VSMC proliferation, but not vascular endothelial cell proliferation. Conclusion—Our findings demonstrate that E2 regulates expression of miRs in the vasculature and support the estrogen receptors-dependent induction of miRs as a mechanism for E2-mediated gene repression. Furthermore, our findings demonstrate that miR-203 contributes to E2-induced inhibition of VSMC proliferation and highlight the potential of miR-203 as a therapeutic agent in the treatment of proliferative cardiovascular diseases.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Rapid Estrogen Receptor Signaling Mediates Estrogen-Induced Inhibition of Vascular Smooth Muscle Cell Proliferation

Kazutaka Ueda; Qing Lu; Wendy Baur; Mark Aronovitz; Richard H. Karas

Objective—The proliferation of vascular smooth muscle cells (VSMCs) plays a crucial role in vascular diseases, such as atherosclerosis and restenosis, after percutaneous coronary intervention. Many studies have shown that estrogen inhibits VSMC proliferation in response to vascular injury in the mouse carotid injury model. However, the mechanisms that mediate these effects remain unclear. Here, we investigated the mechanisms by which estrogen inhibits VSMC proliferation. Approach and Results—We established a novel transgenic mouse line, referred to as the disrupting peptide mice, in which rapid estrogen receptor (ER)–mediated signaling is abolished by overexpression of a peptide that prevents the ER from forming a signaling complex necessary for rapid signaling. Carotid artery VSMCs from disrupting peptide mice or littermate wild-type female mice were obtained by the explant method. In VSMCs derived from wild-type mice, estrogen significantly inhibited VSMC proliferation. Phosphorylation levels of Akt and extracellular regulated kinase induced by platelet derived growth factor were significantly inhibited by estrogen pretreatment. Estrogen enhanced complex formation between ER&agr; and protein phosphatase 2A (PP2), and enhanced PP2A activity. The blockade of PP2A activity abolished the estrogen-induced antiproliferative effect on VSMCs. In contrast, none of these effects of estrogen observed in the wild-type VSMCs were observed in VSMCs derived from disrupting peptide mice. These results support that rapid, non-nuclear ER signaling is required for estrogen-induced inhibition of VSMC proliferation, and further that PP2A activation by estrogen mediates estrogen-induced antiproliferative effects. Conclusions—These findings support that PP2A activation via rapid, non-nuclear ER signaling may be a novel target for therapeutic approaches to inhibit VSMC proliferation, which plays a central role in atherosclerosis and restenosis.

Collaboration


Dive into the Wendy Baur's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qing Lu

Tufts Medical Center

View shared research outputs
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
Researchain Logo
Decentralizing Knowledge