Network


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

Hotspot


Dive into the research topics where Meredith K. Owen is active.

Publication


Featured researches published by Meredith K. Owen.


Circulation | 2013

Perivascular Adipose Tissue Potentiates Contraction of Coronary Vascular Smooth Muscle Influence of Obesity

Meredith K. Owen; Frank A. Witzmann; Mikaela L. McKenney; Xianyin Lai; Zachary C. Berwick; Steven P. Moberly; Mouhamad Alloosh; Michael Sturek; Johnathan D. Tune

Background— This investigation examined the mechanisms by which coronary perivascular adipose tissue (PVAT)–derived factors influence vasomotor tone and the PVAT proteome in lean versus obese swine. Methods and Results— Coronary arteries from Ossabaw swine were isolated for isometric tension studies. We found that coronary (P=0.03) and mesenteric (P=0.04) but not subcutaneous adipose tissue augmented coronary contractions to KCl (20 mmol/L). Inhibition of CaV1.2 channels with nifedipine (0.1 µmol/L) or diltiazem (10 µmol/L) abolished this effect. Coronary PVAT increased baseline tension and potentiated constriction of isolated arteries to prostaglandin F2&agr; in proportion to the amount of PVAT present (0.1–1.0 g). These effects were elevated in tissues obtained from obese swine and were observed in intact and endothelium denuded arteries. Coronary PVAT also diminished H2O2-mediated vasodilation in lean and, to a lesser extent, in obese arteries. These effects were associated with alterations in the obese coronary PVAT proteome (detected 186 alterations) and elevated voltage-dependent increases in intracellular [Ca2+] in obese smooth muscle cells. Further studies revealed that the Rho-kinase inhibitor fasudil (1 µmol/L) significantly blunted artery contractions to KCl and PVAT in lean but not obese swine. Calpastatin (10 &mgr;mol/L) also augmented contractions to levels similar to that observed in the presence of PVAT. Conclusions— Vascular effects of PVAT vary according to anatomic location and are influenced by an obese phenotype. Augmented contractile effects of obese coronary PVAT are related to alterations in the PVAT proteome (eg, calpastatin), Rho-dependent signaling, and the functional contribution of K+ and CaV1.2 channels to smooth muscle tone.


Basic Research in Cardiology | 2013

Impaired Cardiometabolic Responses to Glucagon-Like Peptide 1 in Obesity and Type 2 Diabetes Mellitus

Steven P. Moberly; Kieren J. Mather; Zachary C. Berwick; Meredith K. Owen; Adam G. Goodwill; Eli D. Casalini; Gary D. Hutchins; Mark A. Green; Yen Ng; Robert V. Considine; Kevin M. Perry; Robin Chisholm; Johnathan D. Tune

Glucagon-like peptide 1 (GLP-1) has insulin-like effects on myocardial glucose uptake which may contribute to its beneficial effects in the setting of myocardial ischemia. Whether these effects are different in the setting of obesity or type 2 diabetes (T2DM) requires investigation. We examined the cardiometabolic actions of GLP-1 (7–36) in lean and obese/T2DM humans, and in lean and obese Ossabaw swine. GLP-1 significantly augmented myocardial glucose uptake under resting conditions in lean humans, but this effect was impaired in T2DM. This observation was confirmed and extended in swine, where GLP-1 effects to augment myocardial glucose uptake during exercise were seen in lean but not in obese swine. GLP-1 did not increase myocardial oxygen consumption or blood flow in humans or in swine. Impaired myocardial responsiveness to GLP-1 in obesity was not associated with any apparent alterations in myocardial or coronary GLP1-R expression. No evidence for GLP-1-mediated activation of cAMP/PKA or AMPK signaling in lean or obese hearts was observed. GLP-1 treatment augmented p38-MAPK activity in lean, but not obese cardiac tissue. Taken together, these data provide novel evidence indicating that the cardiometabolic effects of GLP-1 are attenuated in obesity and T2DM, via mechanisms that may involve impaired p38-MAPK signaling.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Lean and Obese Coronary Perivascular Adipose Tissue Impairs Vasodilation via Differential Inhibition of Vascular Smooth Muscle K+ Channels

Jillian N. Noblet; Meredith K. Owen; Adam G. Goodwill; Daniel J. Sassoon; Johnathan D. Tune

Objective—The effects of coronary perivascular adipose tissue (PVAT) on vasomotor tone are influenced by an obese phenotype and are distinct from other adipose tissue depots. The purpose of this investigation was to examine the effects of lean and obese coronary PVAT on end-effector mechanisms of coronary vasodilation and to identify potential factors involved. Approach and Results—Hematoxylin and eosin staining revealed similarities in coronary perivascular adipocyte size between lean and obese Ossabaw swine. Isometric tension studies of isolated coronary arteries from Ossabaw swine revealed that factors derived from lean and obese coronary PVAT attenuated vasodilation to adenosine. Lean coronary PVAT inhibited KCa and KV7, but not KATP channel-mediated dilation in lean arteries. In the absence of PVAT, vasodilation to KCa and KV7 channel activation was impaired in obese arteries relative to lean arteries. Obese PVAT had no effect on KCa or KV7 channel-mediated dilation in obese arteries. In contrast, obese PVAT inhibited KATP channel-mediated dilation in both lean and obese arteries. The differential effects of obese versus lean PVAT were not associated with changes in either coronary KV7 or KATP channel expression. Incubation with calpastatin attenuated coronary vasodilation to adenosine in lean but not in obese arteries. Conclusions—These findings indicate that lean and obese coronary PVAT attenuates vasodilation via inhibitory effects on vascular smooth muscle K+ channels and that alterations in specific factors such as calpastatin are capable of contributing to the initiation or progression of smooth muscle dysfunction in obesity.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Perivascular Adipose Tissue and Coronary Vascular Disease

Meredith K. Owen; Jillian N. Noblet; Daniel J. Sassoon; Abass M. Conteh; Adam G. Goodwill; Johnathan D. Tune

Coronary perivascular adipose tissue is a naturally occurring adipose tissue depot that normally surrounds the major coronary arteries on the surface of the heart. Although originally thought to promote vascular health and integrity, there is a growing body of evidence to support that coronary perivascular adipose tissue displays a distinct phenotype relative to other adipose depots and is capable of producing local factors with the potential to augment coronary vascular tone, inflammation, and the initiation and progression of coronary artery disease. The purpose of the present review is to outline previous findings about the cardiovascular effects of coronary perivascular adipose tissue and the potential mechanisms by which adipose-derived factors may influence coronary vascular function and the progression of atherogenesis.


Microcirculation | 2014

Contribution of Hydrogen Sulfide to the Control of Coronary Blood Flow

Eli D. Casalini; Adam G. Goodwill; Meredith K. Owen; Steven P. Moberly; Zachary C. Berwick; Johnathan D. Tune

This study examined the mechanisms by which H2S modulates coronary microvascular resistance and myocardial perfusion at rest and in response to cardiac ischemia.


Cardiovascular Research | 2014

Mechanisms underlying capsaicin effects in canine coronary artery: implications for coronary spasm

S. Christopher Hiett; Meredith K. Owen; Xingjuan Chen; Ashley M. Riley; Jillian N. Noblet; Sarah Flores; Michael Sturek; Johnathan D. Tune; Alexander G. Obukhov

AIMS The TRPV1, transient receptor potential vanilloid type 1, agonist capsaicin is considered to be beneficial for cardiovascular health because it dilates coronary arteries through an endothelial-dependent mechanism and may slow atheroma progression. However, recent reports indicate that high doses of capsaicin may constrict coronary arterioles and even provoke myocardial infarction. Thus far, the mechanisms by which TRPV1 activation modulates coronary vascular tone remain poorly understood. This investigation examined whether there is a synergistic interplay between locally acting vasoconstrictive pro-inflammatory hormones (autacoids) and capsaicin effects in the coronary circulation. METHODS AND RESULTS Experiments were performed in canine conduit coronary artery rings and isolated smooth muscle cells (CASMCs). Isometric tension measurements revealed that 1-10 μM capsaicin alone did not affect resting tension of coronary artery rings. In contrast, in endothelium-intact rings pre-contracted with a Gq/11-coupled FP/TP (prostaglandin F/thromboxane) receptor agonist, prostaglandin F2α (PGF2α; 10 μM), capsaicin first induced transient dilation that was followed by sustained contraction. In endothelium-denuded rings pre-contracted with PGF2α or thromboxane analogue U46619 (1 μM, a TP receptor agonist), capsaicin induced only sustained contraction. Blockers of the TP receptor or TRPV1 significantly inhibited capsaicin effects, but these were still observed in the presence of 50 μM nifedipine and 70 mM KCl. Capsaicin also potentiated 20 mM KCl-induced contractions. Fluorescence imaging experiments in CASMCs revealed that the Gq/11-phospholipase C (PLC)-protein kinase C (PKC) and Ca(2+)-PLC-PKC pathways are likely involved in sensitizing CASMC TRPV1 channels. CONCLUSION Capsaicin alone does not cause contractions in conduit canine coronary artery; however, pre-treatment with pro-inflammatory prostaglandin-thromboxane agonists may unmask capsaicins vasoconstrictive potential.


Cytoskeleton | 2015

Palladin expression is a conserved characteristic of the desmoplastic tumor microenvironment and contributes to altered gene expression

Austin R. Cannon; Meredith K. Owen; Michael S. Guerrero; Michael L. Kerber; Silvia M. Goicoechea; Kathryn C. Hemstreet; Brian Klazynski; Johnathan Hollyfield; Emily H. Chang; Rosa F. Hwang; Carol A. Otey; Hong Jin Kim

The stroma surrounding solid tumors contributes in complex ways to tumor progression. Cancer‐associated fibroblasts (CAFs) are the predominant cell type in the tumor stroma. Previous studies have shown that the actin‐binding protein palladin is highly expressed in the stroma of pancreas tumors, but the interpretation of these results is complicated by the fact that palladin exists as multiple isoforms. In the current study, the expression and localization of palladin isoform 4 was examined in normal specimens and adenocarcinomas of human pancreas, lung, colon, and stomach samples. Immunohistochemistry with isoform‐selective antibodies revealed that expression of palladin isoform 4 was higher in adenocarcinomas versus normal tissues, and highest in CAFs. Immunohistochemistry staining revealed that palladin was present in both the cytoplasm and the nucleus of CAFs, and this was confirmed using immunofluorescence staining and subcellular fractionation of a pancreatic CAF cell line. To investigate the functional significance of nuclear palladin, RNA Seq analysis of palladin knockdown CAFs versus control CAFs was performed, and the results showed that palladin regulates the expression of genes involved in the biosynthesis and assembly of collagen, and organization of the extracellular matrix. These results suggested that palladin isoform 4 may play a conserved role in establishing the phenotype of CAFs in multiple tumor types.


Data in Brief | 2016

Repeat cross-sectional data on the progression of the metabolic syndrome in Ossabaw miniature swine

Mikaela L. McKenney-Drake; Stacey D. Rodenbeck; Meredith K. Owen; Kyle A. Schultz; Mouhamad Alloosh; Johnathan D. Tune; Michael Sturek

Ossabaw miniature swine were fed an excess calorie, atherogenic diet for 6, 9, or 12 months. Increased body weight, hypertension, and increased plasma cholesterol and triglycerides are described in Table 1. For more detailed interpretations and conclusions about the data, see our associated research study, “Biphasic alterations in coronary smooth muscle Ca2+ regulation during coronary artery disease progression in metabolic syndrome” McKenney-Drake, et al. (2016) [1].


Circulation | 2013

Perivascular Adipose Tissue Potentiates Contraction of Coronary Vascular Smooth Muscle

Meredith K. Owen; Frank A. Witzmann; Mikaela L. McKenney; Xianyin Lai; Zachary C. Berwick; Steven P. Moberly; Mouhamad Alloosh; Michael Sturek; Johnathan D. Tune

Background— This investigation examined the mechanisms by which coronary perivascular adipose tissue (PVAT)–derived factors influence vasomotor tone and the PVAT proteome in lean versus obese swine. Methods and Results— Coronary arteries from Ossabaw swine were isolated for isometric tension studies. We found that coronary (P=0.03) and mesenteric (P=0.04) but not subcutaneous adipose tissue augmented coronary contractions to KCl (20 mmol/L). Inhibition of CaV1.2 channels with nifedipine (0.1 µmol/L) or diltiazem (10 µmol/L) abolished this effect. Coronary PVAT increased baseline tension and potentiated constriction of isolated arteries to prostaglandin F2&agr; in proportion to the amount of PVAT present (0.1–1.0 g). These effects were elevated in tissues obtained from obese swine and were observed in intact and endothelium denuded arteries. Coronary PVAT also diminished H2O2-mediated vasodilation in lean and, to a lesser extent, in obese arteries. These effects were associated with alterations in the obese coronary PVAT proteome (detected 186 alterations) and elevated voltage-dependent increases in intracellular [Ca2+] in obese smooth muscle cells. Further studies revealed that the Rho-kinase inhibitor fasudil (1 µmol/L) significantly blunted artery contractions to KCl and PVAT in lean but not obese swine. Calpastatin (10 &mgr;mol/L) also augmented contractions to levels similar to that observed in the presence of PVAT. Conclusions— Vascular effects of PVAT vary according to anatomic location and are influenced by an obese phenotype. Augmented contractile effects of obese coronary PVAT are related to alterations in the PVAT proteome (eg, calpastatin), Rho-dependent signaling, and the functional contribution of K+ and CaV1.2 channels to smooth muscle tone.


Circulation | 2013

Perivascular Adipose Tissue Potentiates Contraction of Coronary Vascular Smooth MuscleClinical Perspective: Influence of Obesity

Meredith K. Owen; Frank A. Witzmann; Mikaela L. McKenney; Xianyin Lai; Zachary C. Berwick; Steven P. Moberly; Mouhamad Alloosh; Michael Sturek; Johnathan D. Tune

Background— This investigation examined the mechanisms by which coronary perivascular adipose tissue (PVAT)–derived factors influence vasomotor tone and the PVAT proteome in lean versus obese swine. Methods and Results— Coronary arteries from Ossabaw swine were isolated for isometric tension studies. We found that coronary (P=0.03) and mesenteric (P=0.04) but not subcutaneous adipose tissue augmented coronary contractions to KCl (20 mmol/L). Inhibition of CaV1.2 channels with nifedipine (0.1 µmol/L) or diltiazem (10 µmol/L) abolished this effect. Coronary PVAT increased baseline tension and potentiated constriction of isolated arteries to prostaglandin F2&agr; in proportion to the amount of PVAT present (0.1–1.0 g). These effects were elevated in tissues obtained from obese swine and were observed in intact and endothelium denuded arteries. Coronary PVAT also diminished H2O2-mediated vasodilation in lean and, to a lesser extent, in obese arteries. These effects were associated with alterations in the obese coronary PVAT proteome (detected 186 alterations) and elevated voltage-dependent increases in intracellular [Ca2+] in obese smooth muscle cells. Further studies revealed that the Rho-kinase inhibitor fasudil (1 µmol/L) significantly blunted artery contractions to KCl and PVAT in lean but not obese swine. Calpastatin (10 &mgr;mol/L) also augmented contractions to levels similar to that observed in the presence of PVAT. Conclusions— Vascular effects of PVAT vary according to anatomic location and are influenced by an obese phenotype. Augmented contractile effects of obese coronary PVAT are related to alterations in the PVAT proteome (eg, calpastatin), Rho-dependent signaling, and the functional contribution of K+ and CaV1.2 channels to smooth muscle tone.

Collaboration


Dive into the Meredith K. Owen'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge