Network


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

Hotspot


Dive into the research topics where Willa A. Hsueh is active.

Publication


Featured researches published by Willa A. Hsueh.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Synthetic LXR ligand inhibits the development of atherosclerosis in mice

Sean B. Joseph; Elaine McKilligin; Liming Pei; Michael A. Watson; Alan R. Collins; Bryan A. Laffitte; Mingyi Chen; Grace Noh; Joanne Goodman; Graham N. Hagger; Jonathan Tran; Tim K. Tippin; Xuping Wang; Aldons J. Lusis; Willa A. Hsueh; Ronald E. Law; Jon L. Collins; Timothy M. Willson; Peter Tontonoz

The nuclear receptors LXRα and LXRβ have been implicated in the control of cholesterol and fatty acid metabolism in multiple cell types. Activation of these receptors stimulates cholesterol efflux in macrophages, promotes bile acid synthesis in liver, and inhibits intestinal cholesterol absorption, actions that would collectively be expected to reduce atherosclerotic risk. However, synthetic LXR ligands have also been shown to induce lipogenesis and hypertriglyceridemia in mice, raising questions as to the net effects of these compounds on the development of cardiovascular disease. We demonstrate here that the nonsteroidal LXR agonist GW3965 has potent antiatherogenic activity in two different murine models. In LDLR−/− mice, GW3965 reduced lesion area by 53% in males and 34% in females. A similar reduction of 47% was observed in male apoE−/− mice. Long-term (12-week) treatment with LXR agonist had differential effects on plasma lipid profiles in LDLR−/− and apoE−/− mice. GW3965 induced expression of ATP-binding cassettes A1 and G1 in modified low-density lipoprotein-loaded macrophages in vitro as well as in the aortas of hyperlipidemic mice, suggesting that direct actions of LXR ligands on vascular gene expression are likely to contribute to their antiatherogenic effects. These observations provide direct evidence for an atheroprotective effect of LXR agonists and support their further evaluation as potential modulators of human cardiovascular disease.


Journal of Clinical Investigation | 1996

Troglitazone inhibits vascular smooth muscle cell growth and intimal hyperplasia.

Ronald E. Law; Woerner P. Meehan; Xiao-Ping Xi; Kristof Graf; Daniel A. Wuthrich; William D. Coats; David P. Faxon; Willa A. Hsueh

Vascular smooth muscle cell (VSMC) proliferation and migration are responses to arterial injury that are highly important to the processes of restenosis and atherosclerosis. In the arterial balloon injury model in the rat, platelet-derived growth factor (PDGF) and basic fibroblast growth factor (bFGF) are induced in the vessel wall and regulate these VSMC activities. Novel insulin sensitizing agents, thiazolidinediones, have been demonstrated to inhibit insulin and epidermal growth factor-induced growth of VSMCs. We hypothesized that these agents might also inhibit the effect of PDGF and bFGF on cultured VSMCs and intimal hyperplasia in vivo. Troglitazone (1 microM), a member of the thiazolidinedione class, produced a near complete inhibition of both bFGF-induced DNA synthesis as measured by bromodeoxyuridine incorporation (6.5+/-3.9 vs. 17.6+/-4.3% cells labeled, P < 0.05) and c-fos induction. This effect was associated with an inhibition (by 73+/-4%, P < 0.01) by troglitazone of the transactivation of the serum response element, which regulates c-fos expression. Inhibition of c-fos induction by troglitazone appeared to occur via a blockade of the MAP kinase pathway at a point downstream of MAP kinase activation by MAP kinase kinase. At this dose, troglitazone also inhibited PDGF-BB-directed migration of VSMC (by 70+/-6%, P < 0.01). These in vitro effects were operative in vivo. Quantitative image analysis revealed that troglitazone-treated rats had 62% (P < 0.001) less neointima/media area ratio 14 d after balloon injury of the aorta compared with injured rats that received no troglitazone. These results suggest troglitazone is a potent inhibitor of VSMC proliferation and migration and, thus, may be a useful agent to prevent restenosis and possibly atherosclerosis.


Circulation | 2000

Expression and Function of PPARγ in Rat and Human Vascular Smooth Muscle Cells

Ronald E. Law; Stephan Goetze; Xiao-Ping Xi; Simon K. Jackson; Yasuko Kawano; Linda L. Demer; Michael C. Fishbein; Woerner P. Meehan; Willa A. Hsueh

Background—Peroxisome proliferator–activated receptor-γ (PPARγ) is activated by fatty acids, eicosanoids, and insulin-sensitizing thiazolidinediones (TZDs). The TZD troglitazone (TRO) inhibits vascular smooth muscle cell (VSMC) proliferation and migration in vitro and in postinjury intimal hyperplasia. Methods and Results—Rat and human VSMCs express mRNA and nuclear receptors for PPARγ1. Three PPARγ ligands, the TZDs TRO and rosiglitazone and the prostanoid 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), all inhibited VSMC proliferation and migration. PPARγ is upregulated in rat neointima at 7 days and 14 days after balloon injury and is also present in early human atheroma and precursor lesions. Conclusions—Pharmacological activation of PPARγ expressed in VSMCs inhibits their proliferation and migration, potentially limiting restenosis and atherosclerosis. These receptors are upregulated during vascular injury.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2001

Troglitazone Inhibits Formation of Early Atherosclerotic Lesions in Diabetic and Nondiabetic Low Density Lipoprotein Receptor–Deficient Mice

Alan R. Collins; Woerner P. Meehan; Ulrich Kintscher; Simon M. Jackson; Shu Wakino; Grace Noh; Wulf Palinski; Willa A. Hsueh; Ronald E. Law

Abstract —Peroxisome proliferator–activated receptor-&ggr; (PPAR&ggr;) is a ligand-activated nuclear receptor expressed in all of the major cell types found in atherosclerotic lesions: monocytes/macrophages, endothelial cells, and smooth muscle cells. In vitro, PPAR&ggr; ligands inhibit cell proliferation and migration, 2 processes critical for vascular lesion formation. In contrast to these putative antiatherogenic activities, PPAR&ggr; has been shown in vitro to upregulate the CD36 scavenger receptor, which could promote foam cell formation. Thus, it is unclear what impact PPAR&ggr; activation will have on the development and progression of atherosclerosis. This issue is important because thiazolidinediones, which are ligands for PPAR&ggr;, have recently been approved for the treatment of type 2 diabetes, a state of accelerated atherosclerosis. We report herein that the PPAR&ggr; ligand, troglitazone, inhibited lesion formation in male low density lipoprotein receptor–deficient mice fed either a high-fat diet, which also induces type 2 diabetes, or a high-fructose diet. Troglitazone decreased the accumulation of macrophages in intimal xanthomas, consistent with our in vitro observation that troglitazone and another thiazolidinedione, rosiglitazone, inhibited monocyte chemoattractant protein-1–directed transendothelial migration of monocytes. Although troglitazone had some beneficial effects on metabolic risk factors (in particular, a reduction of insulin levels in the diabetic model), none of the systemic cardiovascular risk factors was consistently improved in either model. These observations suggest that the inhibition of early atherosclerotic lesion formation by troglitazone may result, at least in part, from direct effects of PPAR&ggr; activation in the artery wall.


Annals of Internal Medicine | 1998

Metabolic Effects of Troglitazone Monotherapy in Type 2 Diabetes Mellitus: A Randomized, Double-Blind, Placebo-Controlled Trial

David G. Maggs; Thomas A. Buchanan; Charles F. Burant; Gary W. Cline; Barry Gumbiner; Willa A. Hsueh; Silvio E. Inzucchi; David E. Kelley; John J. Nolan; Jerrold M. Olefsky; Kenneth S. Polonsky; David Silver; Thomas R. Valiquett; Gerald I. Shulman

Type 2 diabetes mellitus is characterized by two major pathophysiologic defects: insulin resistance and impaired capacity to secrete insulin [1, 2]. A major component of insulin resistance exists in peripheral tissues, where insulins ability to stimulate glucose uptake from the circulation is blunted. During the past three decades, treatment of hyper-glycemia in patients with type 2 diabetes mellitus who do not respond to such behavioral modifications as diet and exercise has focused on improving the relative insulin deficiency through therapy with sulfonylurea drugs to stimulate endogenous insulin secretion or through administration of insulin itself. Two additional drugs have recently become available: metformin, which seems to exert much of its glucose-lowering effect by suppressing hepatic glucose production [3], and acarbose, which changes the pattern of glucose absorption from the gastrointestinal tract [4]. Thus, no pharmacologic intervention for type 2 diabetes mellitus has had a major effect on improving insulin resistance in peripheral tissues. New compounds, the thiazolidinediones, have recently been developed as glucose-lowering agents. Early studies showed that the glucose-lowering effect of thiazolidinediones was evident in animal models of type 2 diabetes mellitus but not those of type 1 diabetes mellitus [5, 6], suggesting that some endogenous insulin secretion is needed for these agents to act. Troglitazone has been shown to decrease levels of not only plasma glucose and glycosylated hemoglobin [7-13] but also insulin and C-peptide. These observations, coupled with direct measures of whole-body insulin sensitivity in a small number of patients with type 2 diabetes mellitus [7], suggest that troglitazone exerts its major glucose-lowering effect by ameliorating insulin resistance. However, it is not clear whether troglitazone exerts its major insulin-sensitizing effect predominantly in the liver or in peripheral tissues. We studied this issue using detailed metabolic measurements in a large group of patients with type 2 diabetes mellitus. Methods This multicenter study was conducted at six sites: University of Chicago, Chicago, Illinois; University of Southern California, Los Angeles, California; University of Rochester, Rochester, New York; University of Pittsburgh, Pittsburgh, Pennsylvania; University of California, San Diego, San Diego, California; and Yale University, New Haven, Connecticut. Sample size was projected on the basis of study design, major end points, and standard power analysis. Each center enrolled patients while adhering to a common protocol with the same inclusion and exclusion criteria. At each center, patients gave written informed consent to participate in the study, which was approved by the respective university human investigation committees. All patients were studied in a 6-month, randomized, placebo-controlled, double-blind protocol. Patients were randomly assigned to treatment according to a blocked randomization code (block size, five) that was generated by a central computer. In each center, study personnel (executors of treatment assignment) and patients were blinded to the treatment code. Patients were consecutively assigned to treatments; equal numbers of troglitazone or matching placebo tablets were dispensed in a double-blind fashion. Patients Patients had to have type 2 diabetes mellitus according to the criteria of the National Diabetes Data Group [14], HbA1c levels above the upper limit of normal, and fasting C-peptide levels of 0.49 nmol/L or greater. Therapy with oral antidiabetic medication was discontinued before randomization. Patients were excluded if they had clinically symptomatic heart disease, had had a vascular occlusive event in the previous 3 months, had had cancer in the past 5 years, had a serum creatinine level greater than 176.8 mol/L, or had serum amino-transferase levels above the upper limit of normal. Study Design After medical screening, a 2-week wash-out period was allowed for discontinuation of therapy with oral antidiabetic medication in patients who were taking such medication. Metabolic studies were done before patients were randomly assigned to one of five treatment groups: 100, 200, 400, or 600 mg of troglitazone daily or placebo. At 6 months, follow-up metabolic studies were repeated 24 hours after patients received the last troglitazone or placebo tablet. At baseline and 6 months, patients were hospitalized and fasted overnight before a meal tolerance test (day 1) and a euglycemic-hyperinsulinemic clamp procedure (day 2) [15]. During the study, patients were prescribed a diet designed to maintain baseline body weight. Dietary assessment at the time of enrollment determined the patients caloric needs [16]. The prescribed diet consisted of 50% carbohydrates, 34% fat (ratio of saturated fat to polyunsaturated fat, 1:4) and 16% protein. Patients were seen at monthly outpatient visits between the baseline and 6-month metabolic studies so that their clinical condition could be monitored. Meal Tolerance Test At approximately 7:00 a.m., patients were placed on bed rest and an intravenous catheter was inserted into an antecubital vein for blood sampling. A small volume of normal saline (0.9%) was infused to maintain patency. At approximately 8:00 a.m., patients ingested a liquid formula meal (Sustacal-HC [Mead Johnson & Co., Evansville, Indiana], which contained 33% of total daily caloric requirements); this was followed 4 hours later by an identical meal. Fasting blood samples were drawn, and additional samples were obtained every hour thereafter for 8 hours. Samples were processed immediately and stored at 80C for measurement of serum levels of glucose, insulin, free fatty acids, and triglycerides and plasma levels of C-peptide. Fasting blood was also drawn for measurement of HbA1c. After completing the test, patients received an evening meal according to their prescribed diet. They then fasted until the end of the euglycemic-hyperinsulinemic clamp procedure the following day. The intravenous line was left in situ for the clamp procedure. Euglycemic-Hyperinsulinemic Clamp Procedure At 6:00 a.m., a 4-hour primed (corrected for ambient fasting plasma glucose level), continuous (2 mg/m2 body surface area per minute) infusion of [6,6- 2H]-glucose (di-deuterated glucose) isotope into the antecubital vein began. During the third hour of infusion, a retrograde cannula was inserted into a contralateral hand vein. The hand was warmed for sampling of arterialized venous blood. A small volume of normal saline (0.9%) was infused through the sampling cannula to maintain patency. Blood samples were drawn at 10-minute intervals during the final 40 minutes of the fourth hour for measurement of plasma glucose and insulin levels and glucose isotope enrichment. After 4 hours of isotope infusion, a two-step priming dose of insulin was administered (480 mU/m2 per minute followed by 240 mU/m2 per minute; each lasted 5 minutes); this was followed by a continuous infusion of insulin (120 mU/m2 per minute) that lasted 300 minutes (total, 5 hours). The plasma glucose level was allowed to decrease to 5.5 mmol/L; exogenous glucose (dextrose, 20 g/100 mL of water enriched to approximately 2.5% with di-deuterated glucose) was then infused to maintain the plasma glucose level, measured every 5 minutes, at 5.5 mmol/L. The basal isotope infusion was stopped when the exogenous glucose infusion began. Patients also received a continuous infusion of potassium (KCl and KPo 4), 0.105 mmol/L per minute, during the insulin infusion to maintain the serum potassium level between 3.5 and 4.5 mmol/L. During the final hour of the clamp procedure, blood samples were drawn every 10 minutes for measurement of plasma insulin levels and steady-state glucose isotope enrichment. For comparison with diabetic patients, eight persons without diabetes (mean age SD, 46 6 years; mean fasting plasma glucose level, 5.3 0.2 mmol/L; mean body mass index, 29 3 kg/m2) were also studied on one occasion under basal and clamped conditions after an identical hyperinsulinemic clamp protocol. Substrate and Hormone Measurements Serum and plasma samples were shipped frozen to Corning Nichols Institute for chemical analysis and to Yale University for measurement of isotope enrichment. Serum total triglyceride levels (Boehringer Mannheim Diagnostics, Indianapolis, Indiana) and plasma free fatty acid levels (NEFA C-test, Wako Chemicals, Richmond, Virginia) were determined enzymatically; interassay coefficients of variation were 2% and 3.6%, and intraassay coefficients of variation were 1.6% and 1%, respectively. Insulin and C-peptide levels were measured by radioimmunoassay (Corning Nichols Institute); the interassay coefficients of variation were 12.3% and 12.0%, and the intraassay coefficients of variation were 7.4% and 6.5%, respectively. Levels of HbA1c were measured by high-performance liquid chromatography using BioRad (Hercules, California) equipment (Corning Nichols Institute), with a normal reference range of 0.045 to 0.059. At each center, plasma glucose levels were measured at the bedside by using a Beckman glucose analyzer (Fullerton, California). Glucose Isotope Data Gas chromatography mass spectrometer analysis of enrichment of di-deuterated glucose in plasma and infusates was done at one center (Yale Stable Isotope Core Facility, New Haven, Connecticut) by using the penta-acetate derivative of glucose [17]. Calculations Basal hepatic glucose production was calculated as follows: Basal hepatic glucose production = (f/sa) x ([enrichmentinf/enrichmentplasma] 1), where f = basal [6,6- 2H] glucose infusion rate (mg/min), sa = body surface area (m2), enrichmentinf = [6,6- 2H] glucose infusate enrichment (%), and enrichmentplasma = steady-state basal plasma [6,6- 2H] glucose enrichment (%). The term enrichment refers to the fraction of isotope of glucose to naturally occurring (native) glucose,


Journal of Clinical Investigation | 2003

Angiotensin II–accelerated atherosclerosis and aneurysm formation is attenuated in osteopontin-deficient mice

Dennis Bruemmer; Alan R. Collins; Grace Noh; Wei Wang; Mary C. Territo; Sarah Arias-Magallona; Michael C. Fishbein; Florian Blaschke; Ulrich Kintscher; Kristof Graf; Ronald E. Law; Willa A. Hsueh

Osteopontin (OPN) is expressed in atherosclerotic lesions, particularly in diabetic patients. To determine the role of OPN in atherogenesis, ApoE-/-OPN+/+, ApoE-/-OPN+/-, and ApoE-/-OPN-/- mice were infused with Ang II, inducing vascular OPN expression and accelerating atherosclerosis. Compared with ApoE-/-OPN+/+ mice, ApoE-/-OPN+/- and ApoE-/-OPN-/- mice developed less Ang II-accelerated atherosclerosis. ApoE-/- mice transplanted with bone marrow derived from ApoE-/-OPN-/- mice had less Ang II-induced atherosclerosis compared with animals receiving ApoE-/-OPN+/+ cells. Aortae from Ang II-infused ApoE-/-OPN-/- mice expressed less CD68, C-C-chemokine receptor 2, and VCAM-1. In response to intraperitoneal thioglycollate, recruitment of leukocytes in OPN-/- mice was impaired, and OPN-/- leukocytes exhibited decreased basal and MCP-1-directed migration. Furthermore, macrophage viability in atherosclerotic lesions from Ang II-infused ApoE-/-OPN-/- mice was decreased. Finally, Ang II-induced abdominal aortic aneurysm formation in ApoE-/-OPN-/- mice was reduced and associated with decreased MMP-2 and MMP-9 activity. These data suggest an important role for leukocyte-derived OPN in mediating Ang II-accelerated atherosclerosis and aneurysm formation.


American Journal of Cardiology | 2003

Role of endothelial dysfunction in insulin resistance

Willa A. Hsueh; Manuel J. Quiñones

The endothelium regulates vascular tone through the release of vasodilating and vasoconstricting substances. The most important of these vasodilating substances is nitric oxide (NO), which is also vascular protective and inhibits inflammation, oxidation, vascular smooth muscle cell proliferation, and migration. Damage to the endothelium causes endothelial dysfunction with impaired release of NO and loss of its antiatherogenic protection. Traditional risk factors for coronary artery disease, including diabetes, hypercholesterolemia, hypertension, and low levels of high-density lipoprotein cholesterol, are associated with endothelial dysfunction and thus promote the atherogenic process. More recently, insulin resistance in the absence of overt diabetes or the metabolic syndrome has been associated with endothelial dysfunction. This association provides evidence that the atherosclerotic process may actually begin earlier in the spectrum of insulin resistance, ultimately resulting in a progression of the metabolic syndrome to prediabetes and then to type 2 diabetes. Aggressive treatment of dyslipidemia and hypertension, even before the onset of type 2 diabetes, would appear prudent in decreasing the progression of the atherosclerotic process. The thiazolidinediones are peroxisome proliferator-activated receptor-gamma agonists that improve glucose and lipid metabolism. These agents have recently been shown to improve endothelial function in the early stages of insulin resistance. Results from ongoing trials with thiazolidinediones will reveal whether they will also reduce cardiovascular end points.


Circulation | 2000

Angiotensin II Has Multiple Profibrotic Effects in Human Cardiac Fibroblasts

Hiroaki Kawano; Yung S. Do; Yasuko Kawano; Vaughn A. Starnes; Mark L. Barr; Ronald E. Law; Willa A. Hsueh

BACKGROUND Angiotensin II (Ang II) is implicated in cardiac remodeling and is increasingly recognized for its profibrotic activity. METHODS AND RESULTS Because little is known about the direct cellular effects of Ang II on human cardiac fibroblasts, we isolated fibroblasts from ventricles of explanted human hearts and added Ang II (100 nmol/L) to determine its role in growth, extracellular matrix accumulation, and adhesion. To assess which Ang II receptor is involved, Ang II was added in the presence of irbesartan (10 micromol/L), a specific AT(1) receptor antagonist; PD 123319 (10 micromol/L), a specific AT(2) receptor antagonist, or divalinil (100 nmol/L), an AT(4) receptor inhibitor. In human ventricles (n=13), message levels of atrial natriuretic peptide and AT(1) receptor were inversely correlated, which suggests a decrease in AT(1) receptor expression with progressive heart failure. Northern analysis and fluorescence-activated cell sorting demonstrated AT(1) receptor in cultured human cardiac fibroblasts. Ang II increased mitogen-activated protein kinase activity and in DNA synthesis (5-fold, P<0.01) stimulated a 2-fold increase in transforming growth factor-beta(1) (P<0.05) mRNA levels at 2 hours and a 2-fold increase in laminin (P<0.05) and fibronectin (P<0.05) mRNA levels at 24 hours. Ang II also enhanced plasminogen activator inhibitor-1 expression, which inhibits metalloproteinases that degrade the extracellular matrix. All of these effects were inhibited by irbesartan but not PD 123319 or divalinil. In addition, Ang II increased cardiac fibroblast attachment to collagens I and III, which was associated with an increase in focal adhesion kinase activity. CONCLUSIONS Activation of the AT(1) receptor in human heart promotes fibrosis. Ang II plays a novel role in stimulation of plasminogen activator inhibitor-1 expression and adhesion of cardiac fibroblasts to collagen.


Journal of Clinical Investigation | 1995

Blood pressure lowering by pioglitazone. Evidence for a direct vascular effect.

Thomas A. Buchanan; William P. Meehan; Young Y. Jeng; Dong Yang; Timothy M. Chan; Jerry L. Nadler; Steven Scott; Robert K. Rude; Willa A. Hsueh

To examine potential mechanisms for the blood pressure-lowering action of the thiazolidinedione compound, pioglitazone (PIO), we studied the effects of the drug on blood pressure and insulin action in vivo and on vascular tissue in vitro. In vivo, PIO lowered blood pressure in fructose-fed and chow-fed rats to an extent that could not be explained by alterations in fasting plasma insulin or free magnesium concentrations or by alterations in whole-body insulin sensitivity. In vitro, PIO caused significant blunting of the contractile responses of aortic rings to NE, arginine vasopressin (AVP), and potassium chloride; the blunting of responses to NE was maintained after removal of the endothelium. To assess the potential importance of extracellular calcium to the vasodepressor effect of PIO, we measured contractile responses to NE in the absence of calcium, and then after acute restoration of calcium in the presence of NE. PIO had no effect on the contractile response in the absence of calcium. By contrast, PIO blunted by 42% the contractile response that occurred when the extracellular calcium supply was acutely restored in the presence of NE, suggesting that the blunting was mediated by blockade of calcium uptake by vascular smooth muscle. Such an effect was confirmed in cultured a7r5 vascular smooth muscle cells, which exhibited a brisk increase in intracellular calcium in response to AVP that was blocked by PIO in a dose-dependent fashion. Our data indicate that PIO has a direct vascular effect that appears to be mediated at least in part by inhibition of agonist-mediated calcium uptake by vascular smooth muscle. The direct vascular effect may contribute to the blood pressure-lowering actions of PIO in vivo, because that effect could not be explained by alterations in whole-body insulin sensitivity.


Circulation | 2005

Coronary Circulatory Dysfunction in Insulin Resistance, Impaired Glucose Tolerance, and Type 2 Diabetes Mellitus

John O. Prior; Manuel J. Quiñones; Miguel Hernandez-Pampaloni; Alvaro D. Facta; Thomas H. Schindler; James Sayre; Willa A. Hsueh; Heinrich R. Schelbert

Background—Abnormal coronary endothelial reactivity has been demonstrated in diabetes and is associated with an increased rate of cardiovascular events. Our objectives were to investigate the presence of functional coronary circulatory abnormalities over the full spectrum of insulin resistance and to determine whether these would differ in severity with more advanced states of insulin resistance. Methods and Results—Myocardial blood flow (MBF) was measured with positron emission tomography and 13N-ammonia to characterize coronary circulatory function in states of insulin resistance without carbohydrate intolerance (IR), impaired glucose tolerance (IGT), and normotensive and hypertensive type 2 diabetes mellitus (DM) compared with insulin-sensitive (IS) individuals. Indices of coronary function were total vasodilator capacity (mostly vascular smooth muscle–mediated) during pharmacological vasodilation and the nitric oxide–mediated, endothelium-dependent vasomotion in response to cold pressor testing. Total vasodilator capacity was similar in normoglycemic individuals (IS, IR, and IGT), whereas it was significantly decreased in normotensive (−17%) and hypertensive (−34%) DM patients. Compared with IS, endothelium-dependent coronary vasomotion was significantly diminished in IR (−56%), as well as in IGT and normotensive and hypertensive diabetic patients (−85%, −91%, and −120%, respectively). Conclusions—Progressively worsening functional coronary circulatory abnormalities of nitric oxide–mediated, endothelium-dependent vasomotion occur with increasing severity of insulin-resistance and carbohydrate intolerance. Attenuated total vasodilator capacity accompanies the more clinically evident metabolic abnormalities in diabetes.

Collaboration


Dive into the Willa A. Hsueh's collaboration.

Top Co-Authors

Avatar

Ronald E. Law

University of California

View shared research outputs
Top Co-Authors

Avatar

Jerome I. Rotter

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yung S. Do

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kristof Graf

Humboldt University of Berlin

View shared research outputs
Top Co-Authors

Avatar

Eckart Fleck

Humboldt State University

View shared research outputs
Top Co-Authors

Avatar

Mark O. Goodarzi

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiuqing Guo

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge