Network


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

Hotspot


Dive into the research topics where Kathryn M. Gauthier is active.

Publication


Featured researches published by Kathryn M. Gauthier.


Circulation Research | 2002

14,15-Epoxyeicosa-5(Z)-Enoic Acid. A Selective Epoxyeicosatrienoic Acid Antagonist That Inhibits Endothelium-Dependent Hyperpolarization and Relaxation in Coronary Arteries

Kathryn M. Gauthier; Christina Deeter; U. Murali Krishna; Y.Krishna Reddy; Muralidhar Bondlela; John R. Falck; William B. Campbell

Endothelium-dependent hyperpolarization and relaxation of vascular smooth muscle are mediated by endothelium-derived hyperpolarizing factors (EDHFs). EDHF candidates include cytochrome P-450 metabolites of arachidonic acid, K+, hydrogen peroxide, or electrical coupling through gap junctions. In bovine coronary arteries, epoxyeicosatrienoic acids (EETs) appear to function as EDHFs. A 14,15-EET analogue, 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE) was synthesized and identified as an EET-specific antagonist. In bovine coronary arterial rings preconstricted with U46619, 14,15-EET, 11,12-EET, 8,9-EET, and 5,6-EET induced concentration-related relaxations. Preincubation of the arterial rings with 14,15-EEZE (10 &mgr;mol/L) inhibited the relaxations to 14,15-EET, 11,12-EET, 8,9-EET, and 5,6-EET but was most effective in inhibiting 14,15-EET–induced relaxations. 14,15-EEZE also inhibited indomethacin-resistant relaxations to methacholine and arachidonic acid and indomethacin-resistant and l-nitroarginine-resistant relaxations to bradykinin. It did not alter relaxation responses to sodium nitroprusside, iloprost, or the K+ channel activators (NS1619 and bimakalim). Additionally, in small bovine coronary arteries pretreated with indomethacin and l-nitroarginine and preconstricted with U46619, 14,15-EEZE (3 &mgr;mol/L) inhibited bradykinin (10 nmol/L)–induced smooth muscle hyperpolarizations and relaxations. In rat renal microsomes, 14,15-EEZE (10 &mgr;mol/L) did not decrease EET synthesis and did not alter 20-hydroxyeicosatetraenoic acid synthesis. This analogue acts as an EET antagonist by inhibiting the following: (1) EET-induced relaxations, (2) the EDHF component of methacholine-induced, bradykinin-induced, and arachidonic acid–induced relaxations, and (3) the smooth muscle hyperpolarization response to bradykinin. Thus, a distinct molecular structure is required for EET activity, and alteration of this structure modifies agonist and antagonist activity. These findings support a role of EETs as EDHFs.


American Journal of Physiology-heart and Circulatory Physiology | 2008

Effects of the selective EET antagonist, 14,15-EEZE, on cardioprotection produced by exogenous or endogenous EETs in the canine heart

Garrett J. Gross; Kathryn M. Gauthier; Jeannine Moore; John R. Falck; Bruce D. Hammock; William B. Campbell; Kasem Nithipatikom

Previously, we demonstrated (17) that 11,12- and 14,15-epoxyeicosatrienoic acids (EETs) produce marked reductions in myocardial infarct size. Although it is assumed that this cardioprotective effect of the EETs is due to a specific interaction with a membrane-bound receptor, no evidence has indicated that novel EET antagonists selectively block the EET actions in dogs. Our goals were to investigate the effects of 11,12- and 14,15-EET, the soluble epoxide hydrolase inhibitor, 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA), and the putative selective EET antagonist, 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE), on infarct size of barbital anesthetized dogs subjected to 60 min of coronary artery occlusion and 3 h of reperfusion. Furthermore, the effect of 14,15-EEZE on the cardioprotective actions of the selective mitochondrial ATP-sensitive potassium channel opener diazoxide was investigated. Both 11,12- and 14,15-EET markedly reduced infarct size [expressed as a percentage of the area at risk (IS/AAR)] from 21.8 +/- 1.6% (vehicle) to 8.7 +/- 2.2 and 9.4 +/- 1.3%, respectively. Similarly, AUDA significantly reduced IS/AAR from 21.8 +/- 1.6 to 14.4 +/- 1.2% (low dose) and 9.4 +/- 1.8% (high dose), respectively. Interestingly, the combination of the low dose of AUDA with 14,15-EET reduced IS/AAR to 5.8 +/- 1.6% (P < 0.05), further than either drug alone. Diazoxide also reduced IS/AAR significantly (10.2 +/- 1.9%). In contrast, 14,15-EEZE had no effect on IS/AAR by itself (21.0 +/- 3.6%), but completely abolished the effect of 11,12-EET (17.8 +/- 1.4%) and 14,15-EET (19.2 +/- 2.4%) and AUDA (19.3 +/- 1.6%), but not that of diazoxide (10.4 +/- 1.4%). These results suggest that activation of the EET pathway, acting on a putative receptor, by exogenous EETs or indirectly by blocking EET metabolism, produced marked cardioprotection, and the combination of these two approaches resulted in a synergistic effect. These data also suggest that 14,15-EEZE is not blocking the mitochondrial ATP-sensitive potassium channel as a mechanism for antagonizing the cardioprotective effects of the EETs.


Current Opinion in Nephrology and Hypertension | 2002

What is new in endothelium-derived hyperpolarizing factors?

William B. Campbell; Kathryn M. Gauthier

The chemical identification and functional characterization of endothelium-derived hyperpolarizing factors varies depending on vascular size, vascular bed and species. Three major candidates are the epoxyeicosatrienoic acids, cytochrome P450 metabolites of arachidonic acid, potassium ion and hydrogen peroxide. Additionally, electrical coupling through myoendothelial gap junctions serves to conduct electrical changes from the endothelium to the smooth muscle and may mediate or propagate hyperpolarization. Endothelium-derived hyperpolarizing factors are important mediators of vascular relaxation most specifically in resistance sized arteries where they regulate tissue blood flow. The release of the factors is modulated by a number of influences including agonist stimulation, shear stress, estrogen and disease. This article reviews the latest studies concerning the characterization of endothelium-derived hyperpolarizing factors, the mechanisms of factor release and alterations of the factors.


Hypertension | 2005

14,15-Epoxyeicosatrienoic Acid Represents a Transferable Endothelium-Dependent Relaxing Factor in Bovine Coronary Arteries

Kathryn M. Gauthier; Erik M. Edwards; John R. Falck; Dendi S. Reddy; William B. Campbell

Bradykinin causes arterial relaxation and hyperpolarization, which is mediated by a transferable endothelium-derived hyperpolarizing factor (EDHF). In coronary arteries, epoxyeicosatrienoic acids (EETs) are involved in the EDHF response. However, the role of EETs as transferable mediators of EDHF-dependent relaxation remains poorly defined. Two small bovine coronary arteries were cannulated and perfused in tandem in the presence of the nitric oxide synthase inhibitor, nitro-l-arginine (30 &mgr;mol/L), and the cyclooxygenase inhibitor, indomethacin (10 &mgr;mol/L). Luminal perfusate from donor arteries with intact endothelium perfused endothelium-denuded detector arteries. Detector arteries were constricted with U46619 and diameters were monitored. Bradykinin (10 nmol/L) added to detector arteries did not induce dilation (5±2%), whereas bradykinin addition to donor arteries dilated detector arteries by 26.5±7% (P<0.05). These dilations were blocked by donor artery endothelium removal and detector artery treatment with the EET-selective antagonist, 14,15-epoxyeicosa-5(Z)-monoenoic acid (14,15-EEZE; 10 &mgr;mol/L, −5±6%) but not 14,15-EEZE treatment of donor arteries (20±5%). 14,15-EET (0.1 to 10 &mgr;mol/L) added to detector arteries induced maximal dilations of 82±5% that were inhibited 50% by detector artery treatment with 14,15-EEZE (32±12%) but not donor artery treatment with 14,15-EEZE. Liquid chromatography–electrospray ionization mass spectrometry analysis verified the presence of 14,15-EET in the perfusate from an endothelium-intact but not denuded artery. These results show that bradykinin stimulates donor artery 14,15-EET release that dilates detector arteries. 14,15-EEZE blocked the donor artery, endothelium-dependent, bradykinin-induced relaxations, and attenuated relaxations to 14,15-EET. These results suggest that EETs are transferable EDHFs in coronary arteries.


Advances in pharmacology (San Diego) | 2010

Vascular Pharmacology of Epoxyeicosatrienoic Acids

Sandra L. Pfister; Kathryn M. Gauthier; William B. Campbell

Epoxyeicosatrienoic acids (EETs) are cytochrome P450 metabolites of arachidonic acid that are produced by the vascular endothelium in responses to various stimuli such as the agonists acetylcholine (ACH) or bradykinin or by shear stress which activates phospholipase A(2) to release arachidonic acid. EETs are important regulators of vascular tone and homeostasis. In the modulation of vascular tone, EETs function as endothelium-derived hyperpolarizing factors (EDHFs). In models of vascular inflammation, EETs attenuate inflammatory signaling pathways in both the endothelium and vascular smooth muscle. Likewise, EETs regulate blood vessel formation or angiogenesis by mechanisms that are still not completely understood. Soluble epoxide hydrolase (sEH) converts EETs to dihydroxyeicosatrienoic acids (DHETs) and this metabolism limits many of the biological actions of EETs. The recent development of inhibitors of sEH provides an emerging target for pharmacological manipulation of EETs. Additionally, EETs may initiate their biological effects by interacting with a cell surface protein that is a G protein-coupled receptor (GPCR). Since GPCRs represent a common target of most drugs, further characterization of the EET receptor and synthesis of specific EET agonists and antagonist can be used to exploit many of the beneficial effects of EETs in vascular diseases, such as hypertension and atherosclerosis. This review will focus on the current understanding of the contribution of EETs to the regulation of vascular tone, inflammation, and angiogenesis. Furthermore, the therapeutic potential of targeting the EET pathway in vascular disease will be highlighted.


The Journal of Physiology | 2002

Freshly isolated bovine coronary endothelial cells do not express the BKCa channel gene

Kathryn M. Gauthier; Caiqiong Liu; Aleksandra Popovic; Sulayma Albarwani; Nancy J. Rusch

Recent reports have suggested that different types of Ca2+‐activated K+ channels may be selectively expressed either in the vascular endothelial cells (ECs) or smooth muscle cells (SMCs) of a single artery. In this study, we directly compared mRNA, protein and functional expression of the high‐conductance Ca2+‐activated K+ (BKCa) channel between freshly isolated ECs and SMCs from bovine coronary arteries. Fresh ECs and SMCs were enzymatically isolated, and their separation verified by immunofluorescent detection of α‐actin and platelet/endothelium cell adhesion molecule (PECAM) proteins, respectively. Subsequently, studies using a sequence‐specific antibody directed against the pore‐forming α‐subunit of the BKCa channel only detected its expression in the SMCs, whereas PECAM‐positive ECs were devoid of the α‐subunit protein. Additionally, multicell RT‐PCR performed using cDNA derived from either SMCs or ECs only detected mRNA encoding the BKCaα‐subunit in the SMCs. Finally, whole‐cell recordings of outward K+ current detected a prominent iberiotoxin‐sensitive BKCa current in SMCs that was absent in ECs, and the BKCa channel opener NS 1619 only enhanced K+ current in the SMCs. Thus, bovine coronary SMCs densely express BKCa channels whereas adjacent ECs in the same artery appear to lack the expression of the BKCa channel gene. These findings indicate a cell‐specific distribution of Ca2+‐activated K+ channels in SMCs and ECs from a single arterial site.


American Journal of Physiology-heart and Circulatory Physiology | 2009

Role of arachidonic acid lipoxygenase metabolites in the regulation of vascular tone

Yuttana Chawengsub; Kathryn M. Gauthier; William B. Campbell

Stimulation of vascular endothelial cells with agonists such as acetylcholine (ACh) or bradykinin or with shear stress activates phospholipases and releases arachidonic acid (AA). AA is metabolized by cyclooxygenases, cytochrome P-450s, and lipoxygenases (LOs) to vasoactive products. In some arteries, a substantial component of the vasodilator response is dependent on LO metabolites of AA. Nitric oxide (NO)- and prostaglandin (PG)-independent vasodilatory responses to ACh and AA are reduced by inhibitors of LO and by antisense oligonucleotides specifically against 15-LO-1. Vasoactive 15-LO metabolites derived from the vascular endothelium include 15-hydroxy-11,12-epoxyeicosatrienoic acid (15-H-11,12-HEETA) that is hydrolyzed by soluble epoxide hydrolase to 11,12,15-trihydroxyeicosatrienoic acid (11,12,15-THETA). HEETA and THETA are endothelium-derived hyperpolarizing factors that induce vascular relaxations by activation of smooth muscle apamin-sensitive, calcium-activated, small-conductance K(+) channels causing hyperpolarization. In other arteries, the 12-LO metabolite 12-hydroxyeicosatetraenoic acid is synthesized by the vascular endothelium and relaxes smooth muscle by large-conductance, calcium-activated K(+) channel activation. Thus formation of vasodilator eicosanoids derived from LO pathways contributes to the regulation of vascular tone, local blood flow, and blood pressure.


Hypertension | 2004

Apamin-Sensitive K+ Currents Mediate Arachidonic Acid-Induced Relaxations of Rabbit Aorta

Kathryn M. Gauthier; Nancy Spitzbarth; Erik M. Edwards; William B. Campbell

Abstract—Arachidonic acid induces an endothelium-dependent relaxation of the rabbit aorta that is blocked by lipoxygenase inhibitors. The cellular vasodilatory mechanisms activated by arachidonic acid metabolites remain undefined. In rabbit thoracic aortic rings pretreated with indomethacin (10 &mgr;mol/L) and contracted with phenylephrine, arachidonic acid (0.1 to 100 &mgr;mol/L) induced concentration-dependent relaxations. Maximal relaxations averaged 45±3% and were inhibited by increasing extracellular K+ (30 mmol/L, 15±5%; P <0.001) or incubation with apamin (100 nmol/L, 26±7%; P <0.05) but not incubation with charybdotoxin (100 nmol/L, 41±5%). In aortic strips with an intact endothelium that were treated with phenylephrine, arachidonic acid (10 &mgr;mol/L) increased the membrane potential from −28.7±1.3 to −37.8±3.0 mV (P <0.01). Preincubation with apamin did not alter basal membrane potential but inhibited arachidonic acid-induced hyperpolarization (−31.5±1.5 mV). Incubation of rabbit aortic segments with apamin or charybdotoxin did not alter [14C]arachidonic acid metabolism. Whole-cell outward K+ currents from isolated rabbit aortic smooth muscle cells averaged 43.0±4.8 pA/pF at 60 mV and were significantly decreased to 35.7±4.2 pA/pF by apamin (P <0.001). Subsequent addition of charybdotoxin further decreased maximal currents to 14.4±2.3 pA/pF. Addition of 11,12,15-trihydroxyeicosatrienoic acid increased the outward whole-cell K+ current. In inside-out patches of aortic smooth muscle, apamin inhibited the calcium activation (100 to 300 nmol/L; P <0.001) of a small-conductance K+ channel (≈24 pS). These results suggest that arachidonic acid induces endothelium-dependent hyperpolarization and relaxation of rabbit aorta through activation of smooth muscle, apamin-sensitive K+ currents.


Hypertension | 2003

14,15-Epoxyeicosa-5(Z)-Enoic-mSI: A 14,15- and 5,6-EET Antagonist in Bovine Coronary Arteries

Kathryn M. Gauthier; Setti G. Jagadeesh; John R. Falck; William B. Campbell

Abstract—Endothelium-dependent hyperpolarizations and relaxation of vascular smooth muscle induced by acetylcholine and bradykinin are mediated by endothelium-derived hyperpolarizing factors (EDHFs). In bovine coronary arteries, arachidonic acid metabolites, epoxyeicosatrienoic acids (EETs), function as EDHFs. The 14,15-EET analog 14,15-epoxyeicosa-5(Z)-enoic-methylsulfonylimide (14,15-EEZE-mSI) was synthesized and tested for agonist and antagonist activity. In U46619-preconstricted bovine coronary arterial rings, 14,15-, 11,12-, 8,9-, and 5,6-EET induced maximal concentration-related relaxation averaging 75% to 87% at 10 &mgr;mol/L, whereas, 14,15-EEZE-mSI induced maximal relaxation averaging only 7%. 14,15-EEZE-mSI (10 &mgr;mol/L) preincubation inhibited relaxation to 14,15- and 5,6- EET but not 11,12- or 8,9- EET. 14,15-EEZE-mSI also inhibited indomethacin-resistant relaxation to arachidonic acid and indomethacin-resistant and l-nitroarginine-resistant relaxation to bradykinin and methacholine. It did not alter the relaxation to sodium nitroprusside, iloprost, or the K+ channel openers bimakalim or NS1619. In cell-attached patches of isolated bovine coronary arterial smooth muscle cells, 14,15-EEZE-mSI (100 nmol/L) blocked the 14,15-EET–induced (100 nmol/L) activation of large-conductance, calcium-activated K+ channels. Mass spectrometric analysis of rat renal cortical microsomes incubated with arachidonic acid showed that 14,15-EEZE-mSI (10 &mgr;mol/L) increased EET concentrations while decreasing the concentrations of the corresponding dihydroxyeicosatrienoic acids. Therefore, 14,15-EEZE-mSI inhibits relaxation to 5,6- and 14,15- EET and the K+ channel activation by 14,15-EET. It also inhibits the EDHF component of bradykinin-induced, methacholine-induced, and arachidonic acid–induced relaxation. These results suggest that 14,15- or 5,6 -EET act as an EDHF in bovine coronary arteries.


Journal of Cardiovascular Pharmacology | 2007

Roles of epoxyeicosatrienoic acids in vascular regulation and cardiac preconditioning.

Kathryn M. Gauthier; Wenqi Yang; Garrett J. Gross; William B. Campbell

Continuing investigations of the roles of cytochrome P450 (CYP) arachidonic acid epoxygenase metabolites in the regulation of cardiovascular physiology and pathophysiology have revealed their complex and diverse biological effects. Often these metabolites demonstrate protective properties that are revealed during cardiovascular disease. In this regard, the epoxyeicosatrienoic acids (EETs) are an emerging target for pharmacological manipulation aimed at enhancing their cardiac and vascular protective mechanisms. This review will focus on the role of EETs in the regulation of vascular tone, with emphasis on the coronary circulation, their role in limiting platelet aggregation, vascular inflammation and EET contribution to preconditioning of the ischemic myocardium. Production and metabolism of EETs as well as their specific cellular signaling mechanisms are discussed.

Collaboration


Dive into the Kathryn M. Gauthier's collaboration.

Top Co-Authors

Avatar

William B. Campbell

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

John R. Falck

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

David X. Zhang

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Kasem Nithipatikom

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Yuttana Chawengsub

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Tamas Kriska

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Cody Cepura

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Nitin T. Aggarwal

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Sandra L. Pfister

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge