Network


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

Hotspot


Dive into the research topics where Gregory M. Dick is active.

Publication


Featured researches published by Gregory M. Dick.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2006

Hydrogen Peroxide A Feed-Forward Dilator That Couples Myocardial Metabolism to Coronary Blood Flow

Shu Ichi Saitoh; Cuihua Zhang; Johnathan D. Tune; Barry J. Potter; Takahiko Kiyooka; Paul A. Rogers; Jarrod D. Knudson; Gregory M. Dick; Albert N. Swafford; William M. Chilian

Objective—We tested the hypothesis that hydrogen peroxide (H2O2), the dismutated product of superoxide (O2·−), couples myocardial oxygen consumption to coronary blood flow. Accordingly, we measured O2·− and H2O2 production by isolated cardiac myocytes, determined the role of mitochondrial electron transport in the production of these species, and determined the vasoactive properties of the produced H2O2. Methods and Results—The production of O2·− is coupled to oxidative metabolism because inhibition of complex I (rotenone) or III (antimycin) enhanced the production of O2·− during pacing by about 50% and 400%, respectively; whereas uncoupling oxidative phosphorylation by decreasing the protonmotive force with carbonylcyanide-p-trifluoromethoxyphenyl-hydrazone (FCCP) decreased pacing-induced O2·− production. The inhibitor of cytosolic NAD(P)H oxidase assembly, apocynin, did not affect O2·− production by pacing. Aliquots of buffer from paced myocytes produced vasodilation of isolated arterioles (peak response 67±8% percent of maximal dilation) that was significantly reduced by catalase (5±0.5%, P<0.05) or the antagonist of Kv channels, 4-aminopyridine (18±4%, P<0.05). In intact animals, tissue concentrations of H2O2 are proportionate to myocardial oxygen consumption and directly correlated to coronary blood flow. Intracoronary infusion of catalase reduced tissue levels of H2O2 by 30%, and reduced coronary flow by 26%. Intracoronary administration of 4-aminopyridine also shifted the relationship between myocardial oxygen consumption and coronary blood flow or coronary sinus po2. Conclusions—Taken together, our results demonstrate that O2·− is produced in proportion to cardiac metabolism, which leads to the production of the vasoactive reactive oxygen species, H2O2. Our results further suggest that the production of H2O2 in proportion to metabolism couples coronary blood flow to myocardial oxygen consumption.


American Journal of Physiology-heart and Circulatory Physiology | 2008

Impaired capsaicin-induced relaxation of coronary arteries in a porcine model of the metabolic syndrome

Ian N. Bratz; Gregory M. Dick; Johnathan D. Tune; Jason M. Edwards; Zachary P. Neeb; U. Deniz Dincer; Michael Sturek

Recent studies implicate channels of the transient receptor potential vanilloid family (e.g., TRPV1) in regulating vascular tone; however, little is known about these channels in the coronary circulation. Furthermore, it is unclear whether metabolic syndrome alters the function and/or expression of TRPV1. We tested the hypothesis that TRPV1 mediates coronary vasodilation through endothelium-dependent mechanisms that are impaired by the metabolic syndrome. Studies were conducted on coronary arteries from lean and obese male Ossabaw miniature swine. In lean pigs, capsaicin, a TRPV1 agonist, relaxed arteries in a dose-dependent manner (EC50 = 116 +/- 41 nM). Capsaicin-induced relaxation was blocked by the TRPV1 antagonist capsazepine, endothelial denudation, inhibition of nitric oxide synthase, and K+ channel antagonists. Capsaicin-induced relaxation was impaired in rings from pigs with metabolic syndrome (91 +/- 4% vs. 51 +/- 10% relaxation at 100 microM). TRPV1 immunoreactivity was prominent in coronary endothelial cells. TRPV1 protein expression was decreased 40 +/- 11% in obese pigs. Capsaicin (100 microM) elicited divalent cation influx that was abolished in endothelial cells from obese pigs. These data indicate that TRPV1 channels are functionally expressed in the coronary circulation and mediate endothelium-dependent vasodilation through a mechanism involving nitric oxide and K+ channels. Impaired capsaicin-induced vasodilation in the metabolic syndrome is associated with decreased expression of TRPV1 and cation influx.


American Journal of Physiology-heart and Circulatory Physiology | 2009

Impaired function of coronary BKCa channels in metabolic syndrome

Léna Borbouse; Gregory M. Dick; Shinichi Asano; Shawn B. Bender; U. Deniz Dincer; Gregory A. Payne; Zachary P. Neeb; Ian N. Bratz; Michael Sturek; Johnathan D. Tune

The role of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels in regulation of coronary microvascular function is widely appreciated, but molecular and functional changes underlying the deleterious influence of metabolic syndrome (MetS) have not been determined. Male Ossabaw miniature swine consumed for 3-6 mo a normal diet (11% kcal from fat) or an excess-calorie atherogenic diet that induces MetS (45% kcal from fat, 2% cholesterol, 20% kcal from fructose). MetS significantly impaired coronary vasodilation to the BK(Ca) opener NS-1619 in vivo (30-100 microg) and reduced the contribution of these channels to adenosine-induced microvascular vasodilation in vitro (1-100 microM). MetS reduced whole cell penitrem A (1 microM)-sensitive K(+) current and NS-1619-activated (10 microM) current in isolated coronary vascular smooth muscle cells. MetS increased the concentration of free intracellular Ca(2+) and augmented coronary vasoconstriction to the L-type Ca(2+) channel agonist BAY K 8644 (10 pM-10 nM). BK(Ca) channel alpha and beta(1) protein expression was increased in coronary arteries from MetS swine. Coronary vascular dysfunction in MetS is related to impaired BK(Ca) channel function and is accompanied by significant increases in L-type Ca(2+) channel-mediated coronary vasoconstriction.


Microcirculation | 2007

Mechanisms of Coronary Dysfunction in Obesity and Insulin Resistance

Jarrod D. Knudson; U. Deniz Dincer; Ian N. Bratz; Michael Sturek; Gregory M. Dick; Johnathan D. Tune

ABSTRACT


Experimental Biology and Medicine | 2010

Role of potassium channels in coronary vasodilation

Gregory M. Dick; Johnathan D. Tune

K+ channels in coronary arterial smooth muscle cells (CASMC) determine the resting membrane potential (E m) and serve as targets of endogenous and therapeutic vasodilators. E m in CASMC is in the voltage range for activation of L-type Ca2+ channels; therefore, when K+ channel activity changes, Ca2+ influx and arterial tone change. This is why both Ca2+ channel blockers and K+ channel openers have such profound effects on coronary blood flow; the former directly inhibits Ca2+ influx through L-type Ca2+ channels, while the latter indirectly inhibits Ca2+ influx by hyperpolarizing E m and reducing Ca2+ channel activity. K+ channels in CASMC play important roles in vasodilation to endothelial, ischemic and metabolic stimuli. The purpose of this article is to review the types of K+ channels expressed in CASMC, discuss the regulation of their activity by physiological mechanisms and examine impairments related to cardiovascular disease.


American Journal of Physiology-heart and Circulatory Physiology | 2008

Voltage-dependent K+ channels regulate the duration of reactive hyperemia in the canine coronary circulation

Gregory M. Dick; Ian N. Bratz; Léna Borbouse; Gregory A. Payne; U. Deniz Dincer; Jarrod D. Knudson; Paul A. Rogers; Johnathan D. Tune

We previously demonstrated a role for voltage-dependent K(+) (K(V)) channels in coronary vasodilation elicited by myocardial metabolism and exogenous H(2)O(2), as responses were attenuated by the K(V) channel blocker 4-aminopyridine (4-AP). Here we tested the hypothesis that K(V) channels participate in coronary reactive hyperemia and examined the role of K(V) channels in responses to nitric oxide (NO) and adenosine, two putative mediators. Reactive hyperemia (30-s occlusion) was measured in open-chest dogs before and during 4-AP treatment [intracoronary (ic), plasma concentration 0.3 mM]. 4-AP reduced baseline flow 34 +/- 5% and inhibited hyperemic volume 32 +/- 5%. Administration of 8-phenyltheophylline (8-PT; 0.3 mM ic or 5 mg/kg iv) or N(G)-nitro-L-arginine methyl ester (L-NAME; 1 mg/min ic) inhibited early and late portions of hyperemic flow, supporting roles for adenosine and NO. 4-AP further inhibited hyperemia in the presence of 8-PT or L-NAME. Adenosine-induced blood flow responses were attenuated by 4-AP (52 +/- 6% block at 9 microg/min). Dilation of arterioles to adenosine was attenuated by 0.3 mM 4-AP and 1 microM correolide, a selective K(V)1 antagonist (76 +/- 7% and 47 +/- 2% block, respectively, at 1 microM). Dilation in response to sodium nitroprusside, an NO donor, was attenuated by 4-AP in vivo (41 +/- 6% block at 10 microg/min) and by correolide in vitro (29 +/- 4% block at 1 microM). K(V) current in smooth muscle cells was inhibited by 4-AP (IC(50) 1.1 +/- 0.1 mM) and virtually eliminated by correolide. Expression of mRNA for K(V)1 family members was detected in coronary arteries. Our data indicate that K(V) channels play an important role in regulating resting coronary blood flow, determining duration of reactive hyperemia, and mediating adenosine- and NO-induced vasodilation.


British Journal of Pharmacology | 2010

Bisphenol A activates Maxi‐K (KCa1.1) channels in coronary smooth muscle

Shinichi Asano; Johnathan D. Tune; Gregory M. Dick

Background and purpose:  Bisphenol A (BPA) is used to manufacture plastics, including containers for food into which it may leach. High levels of exposure to this oestrogenic endocrine disruptor are associated with diabetes and heart disease. Oestrogen and oestrogen receptor modulators increase the activity of large conductance Ca2+/voltage‐sensitive K+ (Maxi‐K; KCa1.1) channels, but the effects of BPA on Maxi‐K channels are unknown. We tested the hypothesis that BPA activates Maxi‐K channels through a mechanism that depends upon the regulatory β1 subunit.


Microcirculation | 2010

Contribution of Adenosine A2A and A2B Receptors to Ischemic Coronary Dilation: Role of KV and KATP Channels

Zachary C. Berwick; Gregory A. Payne; Brandon Lynch; Gregory M. Dick; Michael Sturek; Johnathan D. Tune

Please cite this paper as: Berwick, Payne, Lynch, Dick, Sturek and Tune (2010). Contribution of Adenosine A2A and A2B Receptors to Ischemic Coronary Dilation: Role of KV and KATP Channels. Microcirculation17(8), 600–607.


American Journal of Physiology-heart and Circulatory Physiology | 2010

Metabolic syndrome reduces the contribution of K+ channels to ischemic coronary vasodilation

Léna Borbouse; Gregory M. Dick; Gregory A. Payne; Zachary C. Berwick; Zachary P. Neeb; Mouhamad Alloosh; Ian N. Bratz; Michael Sturek; Johnathan D. Tune

This investigation tested the hypothesis that metabolic syndrome decreases the relative contribution of specific K(+) channels to coronary reactive hyperemia. Ca(2+)-activated (BK(Ca)), voltage-activated (K(V)), and ATP-dependent (K(ATP)) K(+) channels were investigated. Studies were conducted in anesthetized miniature Ossabaw swine fed a normal maintenance diet (11% kcal from fat) or an excess calorie atherogenic diet (43% kcal from fat, 2% cholesterol, 20% kcal from fructose) for 20 wk. The latter diet induces metabolic syndrome, increasing body weight, fasting glucose, total cholesterol, and triglyceride levels. Ischemic vasodilation was determined by the coronary flow response to a 15-s occlusion before and after cumulative administration of antagonists for BK(Ca) (penitrem A; 10 microg/kg iv), K(V) (4-aminopyridine; 0.3 mg/kg iv) and K(ATP) (glibenclamide; 1 mg/kg iv) channels. Coronary reactive hyperemia was diminished by metabolic syndrome as the repayment of flow debt was reduced approximately 30% compared with lean swine. Inhibition of BK(Ca) channels had no effect on reactive hyperemia in either lean or metabolic syndrome swine. Subsequent inhibition of K(V) channels significantly reduced the repayment of flow debt ( approximately 25%) in both lean and metabolic syndrome swine. Additional blockade of K(ATP) channels further diminished ( approximately 45%) the repayment of flow debt in lean but not metabolic syndrome swine. These data indicate that the metabolic syndrome impairs coronary vasodilation in response to cardiac ischemia via reductions in the contribution of K(+) channels to reactive hyperemia.


American Journal of Physiology-heart and Circulatory Physiology | 2010

Contribution of BKCa channels to local metabolic coronary vasodilation: effects of metabolic syndrome

Léna Borbouse; Gregory M. Dick; Gregory A. Payne; Brittany D. Payne; Mark Svendsen; Zachary P. Neeb; Mouhamad Alloosh; Ian N. Bratz; Michael Sturek; Johnathan D. Tune

This investigation was designed to examine the hypothesis that impaired function of coronary microvascular large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels in metabolic syndrome (MetS) significantly attenuates the balance between myocardial oxygen delivery and metabolism at rest and during exercise-induced increases in myocardial oxygen consumption (MVo(2)). Studies were conducted in conscious, chronically instrumented Ossabaw swine fed a normal maintenance diet (11% kcal from fat) or an excess calorie atherogenic diet (43% kcal from fat, 2% cholesterol, 20% kcal from fructose) that induces many common features of MetS. Data were collected under baseline/resting conditions and during graded treadmill exercise before and after selective blockade of BK(Ca) channels with penitrem A (10 microg/kg iv). We found that the exercise-induced increases in blood pressure were significantly elevated in MetS swine. No differences in baseline cardiac function or heart rate were noted. Induction of MetS produced a parallel downward shift in the relationship between coronary venous Po(2) and MVo(2) (P < 0.001) that was accompanied by a marked release of lactate (negative lactate uptake) as MVo(2) was increased with exercise (P < 0.005). Inhibition of BK(Ca) channels with penitrem A did not significantly affect blood pressure, heart rate, or the relationship between coronary venous Po(2) and MVo(2) in lean or MetS swine. These data indicate that BK(Ca) channels are not required for local metabolic control of coronary blood flow under physiological (lean) or pathophysiological (MetS) conditions. Therefore, diminished function of BK(Ca) channels does not contribute to the impairment of myocardial oxygen-supply demand balance in MetS.

Collaboration


Dive into the Gregory M. Dick's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian N. Bratz

Northeast Ohio Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jarrod D. Knudson

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Paul A. Rogers

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ibra Fancher

West Virginia University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge