Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Perfusion ; 37(6): 575-581, 2022 09.
Article in English | MEDLINE | ID: mdl-33878977

ABSTRACT

BACKGROUND: We analyzed the use of Extracorporeal Membranous Oxygenation (ECMO) in acute care surgery patients at our Level-1 trauma center. We hypothesized that this patient population has improved ECMO outcomes. METHODS: This was a retrospective analysis of emergency general surgery and trauma patients placed on ECMO between the periods of October 2013 and February 2020. There were 10 surgical and 12 trauma patients studied, who eventually required ECMO support. ECMO support and ECMO type/modality were analyzed with injury and survival prognostic scores examined. MAIN RESULTS: Overall, 16 of the 22 patients survived to hospital discharge, for a survival rate of 73%. Mean age was 34.18 years. Mean hospital length of stay was 23.4 days with mean days on ECMO equal to 7.5. The net negative fluid balance was 5.36 L. CONCLUSIONS: The survival of our ECMO cohort is notably higher than previously cited studies. Our group demonstrated decreased length of time on ECMO, decreased length of stay in the hospital, and similar rates of complications compared to prior reports. ECMO is a useful modality in acute care surgical patients and should be considered in these patient populations. Our focus on net negative fluid balance for ECMO patients demonstrates improved survival. ECMO should be considered early in surgical patients and early in advanced trauma life support.


Subject(s)
Extracorporeal Membrane Oxygenation , Adult , Critical Care , Humans , Patient Discharge , Retrospective Studies , Survival Rate , Treatment Outcome
2.
J Cardiothorac Surg ; 16(1): 264, 2021 Sep 19.
Article in English | MEDLINE | ID: mdl-34538270

ABSTRACT

BACKGROUND: The study purpose is to examine survival prognostic and extracorporeal membrane oxygenation (ECMO) application outcomes at our tertiary care center. METHODS: This is a retrospective analysis, January 2014 to September 2019. We analyzed 60 patients who underwent cardiac surgery and required peri-operative ECMO. All inpatients with demographic and intervention data was examined. 52 patients (86.6%) had refractory cardiogenic shock, 7 patients (11.6%) had pulmonary insufficiency, and 1 patient (1.6%) had hemorrhagic shock, all patients required either venous-arterial (VA) (n = 53, 88.3%), venous-venous (VV) (n = 5, 8.3%) or venous-arterial-venous (VAV) (n = 2, 3.3%) ECMO for hemodynamic support. ECMO parameters were analyzed and common postoperative complications were examined in the setting of survival with comorbidities. RESULTS: In-hospital mortality was 60.7% (n = 37). Patients who survived were younger (52 ± 3.3 vs 66 ± 1.5, p < 0.001) with longer hospital stays (35 ± 4.0 vs 20 ± 1.5, p < 0.03). Survivors required fewer blood products (13 ± 2.3 vs 25 ± 2.3, p = 0.02) with a net negative fluid balance (- 3.5 ± 1.6 vs 3.4 ± 1.6, p = 0.01). Cardiac re-operations worsened survival. CONCLUSION: ECMO is a viable rescue strategy for cardiac surgery patients with a 40% survival to discharge rate. Careful attention to volume management and blood transfusion are important markers for potential survival.


Subject(s)
Cardiac Surgical Procedures , Extracorporeal Membrane Oxygenation , Cardiac Surgical Procedures/adverse effects , Hospital Mortality , Humans , Retrospective Studies , Shock, Cardiogenic/etiology , Shock, Cardiogenic/therapy , Treatment Outcome
3.
Physiol Rep ; 5(23)2017 Dec.
Article in English | MEDLINE | ID: mdl-29192067

ABSTRACT

The objective of this study is to test the hypothesis that increased muscle mass has positive effects on cardiovascular function. Specifically, we tested the hypothesis that increases in lean body mass caused by deletion of myostatin improves cardiac performance and vascular function. Echocardiography was used to quantify left ventricular function at baseline and after acute administration of propranolol and isoproterenol to assess ß-adrenergic reactivity. Additionally, resistance vessels in several beds were removed, cannulated, pressurized to 60 mmHg and reactivity to vasoactive stimuli was assessed. Hemodynamics were measured using in vivo radiotelemetry. Myostatin deletion results in increased fractional shortening at baseline. Additionally, arterioles in the coronary and muscular microcirculations are more sensitive to endothelial-dependent dilation while nonmuscular beds or the aorta were unaffected. ß-adrenergic dilation was increased in both coronary and conduit arteries, suggesting a systemic effect of increased muscle mass on vascular function. Overall hemodynamics and physical characteristics (heart weight and size) remained unchanged. Myostatin deletion mimics in part the effects of exercise on cardiovascular function. It significantly increases lean muscle mass and results in muscle-specific increases in endothelium-dependent vasodilation. This suggests that increases in muscle mass may serve as a buffer against pathological states that specifically target cardiac function (heart failure), the ß-adrenergic system (age), and nitric oxide bio-availability (atherosclerosis). Taken together, pharmacological inhibition of the myostatin pathway could prove an excellent mechanism by which the benefits of exercise can be conferred in patients that are unable to exercise.


Subject(s)
Coronary Vessels/metabolism , Heart/physiology , Microvessels/metabolism , Myostatin/genetics , Vasodilation , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Blood Pressure , Coronary Vessels/drug effects , Coronary Vessels/physiology , Gene Deletion , Heart Rate , Isoproterenol/pharmacology , Male , Mice , Mice, Inbred ICR , Microvessels/drug effects , Microvessels/physiology , Muscle, Skeletal/growth & development , Propranolol/pharmacology , Ventricular Function
4.
Physiol Rep ; 3(12)2015 Dec.
Article in English | MEDLINE | ID: mdl-26660551

ABSTRACT

Heterozygous endothelial nitric oxide synthase (eNOS) deficiency is associated with normal endothelium-dependent responses, however, little is known regarding the mechanisms that maintain or impair endothelial function with heterozygous eNOS deficiency. The goals of this study were to (1) determine mechanism(s) which serve to maintain normal endothelial function in the absence of a single eNOS gene; and (2) to determine whether heterozygous eNOS deficiency predisposes blood vessels to endothelial dysfunction in response to a high-fat diet (HFD). Responses of carotid arteries were examined in wild-type (eNOS(+/+)) and heterozygous eNOS-deficient (eNOS(+/-)) treated with either vehicle (saline), N(G)-nitro-L-arginine (L-NNA, 100 µmol/L), an inhibitor of nitric oxide synthase, or 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, 1 µmol/L), an inhibitor of soluble guanylyl cyclase (sGC), and in eNOS(+/+) and eNOS(+/-) mice fed a control (10%) or a 45% HFD (kcal from fat). Responses to acetylcholine (ACh) were similar in vehicle-treated arteries from eNOS(+/+) and eNOS(+/-) mice, and were equally inhibited by L-NNA and ODQ. Phosphorylation of eNOS Ser1176, a site associated with increased eNOS activity, was significantly greater in eNOS(+/-) mice most likely as a compensatory response for the loss of a single eNOS gene. In contrast, responses to ACh were markedly impaired in carotid arteries from eNOS(+/-), but not eNOS(+/+), mice fed a HFD. Vascular superoxide levels as well as plasma levels of the pro-inflammatory cytokine interleukin-6 (IL-6) were selectively increased in HFD-fed eNOS(+/-) mice. In reconstitution experiments, IL-6 produced concentration-dependent impairment of endothelial responses as well as greater increases in NADPH-stimulated superoxide levels in arteries from eNOS(+/-) mice fed a control diet compared to eNOS(+/+) mice. Our findings of increased Ser1176-phosphorylation reveal a mechanism by which NOS- and sGC-dependent endothelial function can be maintained with heterozygous eNOS deficiency. In addition, heterozygous eNOS deficiency predisposes blood vessels to developing endothelial dysfunction in response to a HFD. The impairment produced by a HFD in eNOS(+/-) mice appears to be mediated by IL-6-induced increases in vascular superoxide. These findings serve as an important example of eNOS haploinsufficiency, one that may contribute to the development of carotid artery disease in obese humans.

5.
PLoS One ; 8(10): e78626, 2013.
Article in English | MEDLINE | ID: mdl-24205282

ABSTRACT

Recent studies have shown that circadian clock disruption is associated with pathological remodeling in the arterial structure and vascular stiffness. Moreover, chronic circadian disruption is associated with dysfunction in endothelial responses and signaling. Reactive oxygen species have emerged as key regulators in vascular pathology. Previously, we have demonstrated that circadian clock dysfunction exacerbates superoxide production through eNOS uncoupling. To date, the impact of circadian clock mutation on vascular NADPH oxidase expression and function is not known. The goal in the current study was to determine if the circadian clock controls vascular Nox4 expression and hydrogen peroxide formation in arteries, particularly in endothelial and vascular smooth muscle cells. In aorta, there was an increase in hydrogen peroxide and Nox4 expression in mice with a dysfunctional circadian rhythm (Bmal1-KO mice). In addition, the Nox4 gene promoter is activated by the core circadian transcription factors. Lastly, in synchronized cultured human endothelial cells, Nox4 gene expression exhibited rhythmic oscillations. These data reveal that the circadian clock plays an important role in the control of Nox4 and disruption of the clock leads to subsequent production of reaction oxygen species.


Subject(s)
Aorta/metabolism , Circadian Clocks , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Animals , Aorta/physiology , Gene Expression Regulation , Gene Knockout Techniques , Humans , Hydrogen Peroxide/metabolism , Mice , NADPH Oxidase 4 , NADPH Oxidases/genetics , Promoter Regions, Genetic/genetics
6.
Circ Res ; 111(9): 1157-65, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22912383

ABSTRACT

RATIONALE: Disruption of the circadian clock in mice produces vascular dysfunction as evidenced by impairments in endothelium-dependent signaling, vasomotion, and blood vessel remodeling. Although the altered function of endothelial NO synthase and the overproduction of reactive oxygen species are central to dysfunction of the endothelium, to date, the impact of the circadian clock on endothelial NO synthase coupling and vascular reactive oxygen species production is not known. OBJECTIVE: The goals of the present study were to determine whether deletion of a critical component of the circadian clock, Bmal1, can influence endothelial NO synthase coupling and reactive oxygen species levels in arteries from Bmal1-knockout (KO) mice. METHODS AND RESULTS: Endothelial function was reduced in aortae from Bmal1-KO mice and improved by scavenging reactive oxygen species with polyethylene glycol-superoxide dismutase and nonselectively inhibiting cyclooxygenase isoforms with indomethacin. Aortae from Bmal1-KO mice exhibited enhanced superoxide levels as determined by electron paramagnetic resonance spectroscopy and dihydroethidium fluorescence, an elevation that was abrogated by administration of nitro-l-arginine methyl ester. High-performance liquid chromatography analysis revealed a reduction in tetrahydrobiopterin and an increase in dihydrobiopterin levels in the lung and aorta of Bmal1-KO mice, whereas supplementation with tetrahydrobiopterin improved endothelial function in the circadian clock KO mice. Furthermore, levels of tetrahydrobiopterin, dihydrobiopterin, and the key enzymes that regulate biopterin bioavailability, GTP cyclohydrolase and dihydrofolate reductase exhibited a circadian expression pattern. CONCLUSIONS: Having an established influence in the metabolic control of glucose and lipids, herein, we describe a novel role for the circadian clock in metabolism of biopterins, with a significant impact in the vasculature, to regulate coupling of endothelial NO synthase, production of superoxide, and maintenance of endothelial function.


Subject(s)
ARNTL Transcription Factors/deficiency , Aorta/metabolism , Arteries/metabolism , Circadian Clocks/physiology , Nitric Oxide Synthase Type III/metabolism , Superoxides/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Aorta/cytology , Arteries/cytology , Biopterins/analogs & derivatives , Biopterins/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , GTP Cyclohydrolase/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Reactive Oxygen Species/metabolism , Tetrahydrofolate Dehydrogenase/metabolism
7.
Am J Physiol Heart Circ Physiol ; 302(1): H159-66, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22058149

ABSTRACT

Arginase can cause vascular dysfunction by competing with nitric oxide synthase for l-arginine and by increasing cell proliferation and collagen formation, which promote vascular fibrosis/stiffening. We have shown that increased arginase expression/activity contribute to vascular endothelial cell (EC) dysfunction. Here, we examined the roles of the two arginase isoforms, arginase I and II (AI and AII, respectively), in this process. Experiments were performed using streptozotocin-induced diabetic mice: wild-type (WT) mice and knockout mice lacking the AII isoform alone (AI(+/+)AII(-/-)) or in combination with partial deletion of AI (AI(+/-)AII (-/-)). EC-dependent vasorelaxation of aortic rings and arterial fibrosis and stiffness were assessed in relation to arginase activity and expression. Diabetes reduced mean EC-dependent vasorelaxation markedly in diabetic WT and AI(+/+)AII(-/-) aortas (53% and 44% vs. controls, respectively) compared with a 27% decrease in AI(+/-)AII (-/-) vessels. Coronary fibrosis was also increased in diabetic WT and AI(+/+)AII(-/-) mice (1.9- and 1.7-fold vs. controls, respectively) but was not altered in AI(+/-)AII (-/-) diabetic mice. Carotid stiffness was increased by 142% in WT diabetic mice compared with 51% in AI(+/+)AII(-/-) mice and 19% in AI(+/-)AII (-/-) mice. In diabetic WT and AI(+/+)AII(-/-) mice, aortic arginase activity and AI expression were significantly increased compared with control mice, but neither parameter was altered in AI(+/-)AII (-/-) mice. In summary, AI(+/-)AII (-/-) mice exhibit better EC-dependent vasodilation and less vascular stiffness and coronary fibrosis compared with diabetic WT and AI(+/+)AII(-/-) mice. These data indicate a major involvement of AI in diabetes-induced vascular dysfunction.


Subject(s)
Arginase/metabolism , Arteries/enzymology , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/etiology , Vasodilation , Animals , Aorta/enzymology , Aorta/physiopathology , Arginase/genetics , Arteries/drug effects , Arteries/pathology , Arteries/physiopathology , Carotid Arteries/enzymology , Carotid Arteries/physiopathology , Compliance , Coronary Vessels/enzymology , Coronary Vessels/physiopathology , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/enzymology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Dose-Response Relationship, Drug , Fibrosis , Hydrogen Peroxide/metabolism , Hydroxyproline/metabolism , Lipid Peroxidation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Superoxides/metabolism , Vasoconstriction , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
8.
Arterioscler Thromb Vasc Biol ; 30(12): 2535-43, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20829506

ABSTRACT

OBJECTIVE: To determine if elasticity in blood vessels is compromised in circadian clock-mutant mice (Bmal1-knockout [KO] and Per-triple KO) and if matrix metalloproteinases (MMPs) might confer these changes in compliance. METHODS AND RESULTS: High-resolution ultrasonography in vivo revealed impaired remodeling and increased pulse-wave velocity in the arteries of Bmal1-KO and Per-triple KO mice. In addition, compliance of remodeled arteries and naïve pressurized arterioles ex vivo from Bmal1-KO and Per-triple KO mice was reduced, consistent with stiffening of the vascular bed. The observed vascular stiffness was coincident with dysregulation of MMP-2 and MMP-9 in Bmal1-KO mice. Furthermore, inhibition of MMPs improved indexes of pathological remodeling in wild-type mice, but the effect was abolished in Bmal1-KO mice. CONCLUSIONS: Circadian clock dysfunction contributes to hardening of arteries, which may involve impaired control of the extracellular matrix composition.


Subject(s)
Carotid Arteries/enzymology , Carotid Artery Diseases/enzymology , Circadian Clocks/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Animals , Blood Pressure , Carotid Arteries/diagnostic imaging , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Carotid Artery Diseases/diagnostic imaging , Carotid Artery Diseases/genetics , Carotid Artery Diseases/physiopathology , Cells, Cultured , Elasticity , Endothelial Cells/enzymology , Extracellular Matrix/metabolism , Matrix Metalloproteinase Inhibitors , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Myocytes, Smooth Muscle/enzymology , Period Circadian Proteins/deficiency , Period Circadian Proteins/genetics , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Protease Inhibitors/pharmacology , Thiophenes/pharmacology , Time Factors , Ultrasonography, Doppler, Pulsed
9.
Circ Res ; 105(10): 1013-22, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19797171

ABSTRACT

RATIONALE: Obesity is a risk factor for cardiovascular dysfunction, yet the underlying factors driving this impaired function remain poorly understood. Insulin resistance is a common pathology in obese patients and has been shown to impair vascular function. Whether insulin resistance or obesity, itself, is causal remains unclear. OBJECTIVE: The present study tested the hypothesis that insulin resistance is the underlying mediator for impaired NO-mediated dilation in obesity by genetic deletion of the insulin-desensitizing enzyme protein tyrosine phosphatase (PTP)1B in db/db mice. METHODS AND RESULTS: The db/db mouse is morbidly obese, insulin-resistant, and has tissue-specific elevation in PTP1B expression compared to lean controls. In db/db mice, PTP1B deletion improved glucose clearance, dyslipidemia, and insulin receptor signaling in muscle and fat. Hepatic insulin signaling in db/db mice was not improved by deletion of PTP1B, indicating specific amelioration of peripheral insulin resistance. Additionally, obese mice demonstrate an impaired endothelium dependent and independent vasodilation to acetylcholine and sodium nitroprusside, respectively. This impairment, which correlated with increased superoxide in the db/db mice, was corrected by superoxide scavenging. Increased superoxide production was associated with increased expression of NAD(P)H oxidase 1 and its molecular regulators, Noxo1 and Noxa1. CONCLUSIONS: Deletion of PTP1B improved both endothelium dependent and independent NO-mediated dilation and reduced superoxide generation in db/db mice. PTP1B deletion did not affect any vascular function in lean mice. Taken together, these data reveal a role for peripheral insulin resistance as the mediator of vascular dysfunction in obesity.


Subject(s)
Endothelium, Vascular/enzymology , Gene Deletion , Gene Expression Regulation, Enzymologic , Insulin Resistance , Leptin/metabolism , Obesity/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Acetylcholine/pharmacology , Adaptor Proteins, Signal Transducing , Adipose Tissue/enzymology , Animals , Dyslipidemias/enzymology , Dyslipidemias/genetics , Glucose/genetics , Glucose/metabolism , Leptin/genetics , Mice , Mice, Inbred BALB C , Mice, Obese , Muscles/enzymology , NADH, NADPH Oxidoreductases/biosynthesis , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Obesity/genetics , Oxidation-Reduction/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Proteins/genetics , Proteins/metabolism , Superoxides/metabolism , Vasodilation/drug effects , Vasodilation/genetics , Vasodilator Agents/pharmacology
10.
Basic Res Cardiol ; 104(6): 707-17, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19548058

ABSTRACT

Impairment of vascular growth is a hallmark of diabetic complications, but the progression and mechanisms are poorly understood. To determine whether obesity and early diabetes impair endothelium-dependent vasodilatation and vascular response to ischemia, microvascular function as well as angiogenic responses to ischemia were assessed in young (C57) and 6-month-old lean mice (old C57), in obese (db-C57) mice, and in mice suffering an early (db-KsJ) and sustained type 2 diabetes (old db-KsJ). Glycemia gradually increased from the db-C57 to the old db-KsJ. Early and established type II diabetes significantly reduced the level of insulin that was significantly increased in obese mice. Endothelial function was assessed in isolated resistance arteries while the angiogenic response induced by unilateral hindlimb ischemia was analyzed, after 28 days, with a laser Doppler flowmeter and angiography. Aging (-21%), obesity (-45%), as well as early (-58%) and sustained type II diabetes (-69%) induced a progressive impairment of the endothelium-dependent relaxation of the gracilis artery. Laser Doppler measurements demonstrated that only early and sustained type II diabetes impaired skin blood flow recovery. Vascular collateralization was reduced with aging and severely impaired in older db-KsJ mice, the two strains of mice in which ischemia reduced eNOS expression. These results demonstrate that endothelial dysfunction induced by obesity is insufficient to alter the angiogenic response to ischemia. Furthermore, the development of frank type II diabetes or increasing age is required to impair the vascular response to hindlimb ischemia. We conclude that additional risk factors or severe endothelial dysfunction may be requisite to impede the angiogenic response to ischemia.


Subject(s)
Diabetes Mellitus, Experimental/complications , Endothelial Cells/pathology , Insulin Resistance/physiology , Ischemia/complications , Obesity/complications , Vasodilation/physiology , Aging/physiology , Angiography , Animals , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Hindlimb/blood supply , Ischemia/metabolism , Ischemia/physiopathology , Laser-Doppler Flowmetry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/biosynthesis , Obesity/metabolism , Obesity/physiopathology , Regional Blood Flow , Skin/blood supply
11.
Microcirculation ; 16(5): 414-23, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19382001

ABSTRACT

Obesity and diabetes are major risk factors for the development of vascular disease in the lower limbs. Previous studies have demonstrated reduced nitric oxide (NO)-mediated vasodilation, increased adrenergic constriction, and inward, atrophic remodeling in the limb circulation of obese Zucker rats, but the component of the "metabolic syndrome" driving these changes is unclear. Because insulin resistance precedes the state of frank diabetes, the current study hypothesized that insulin resistance independent of obesity induced by fructose feeding would impair microvascular function in the skeletal muscle circulation in lean Zucker rats (LZR). A 66% fructose diet impaired glucose tolerance and induced moderate insulin resistance with no changes in whole-body hemodynamics of anesthetized rats (FF-LZR), compared to control LZR. NO-mediated vasodilation of isolated gracilis arteries, assessed in vitro with acetylcholine and sodium nitroprusside, was reduced approximately 20% in FF-LZR vs. LZR. NO-independent cGMP-mediated vasodilation was unimpaired. Pretreatment of isolated vessels with the superoxide scavenger, tempol, improved responses to both vasodilators. Reactivity to adrenergic stimulation was unaltered in FF-LZR vs. LZR, although constriction to endothelin was increased. Structural and passive mechanical characteristics of isolated gracilis arteries were similar in both LZR and FF-LZR. Taken together, these findings indicate that moderate insulin resistance is sufficient to impair endothelial function in an oxidant-dependent manner in the rat hindlimb circulation. Other aspects of skeletal muscle vascular function documented in obese models, specifically adrenergic tone and inward remodeling, must reflect either severe insulin resistance or other aspects of obesity. The factors accounting for nonendothelial vasculopathies remain unknown.


Subject(s)
Arteries/metabolism , Endothelium, Vascular/metabolism , Fructose/pharmacology , Insulin Resistance , Muscle, Skeletal/blood supply , Sweetening Agents/pharmacology , Acetylcholine/pharmacology , Animals , Cyclic GMP , Diet , Endothelium, Vascular/pathology , Hindlimb/blood supply , Male , Muscle, Skeletal/metabolism , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Obesity/metabolism , Oxidants/metabolism , Rats , Rats, Zucker , Vasoconstriction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...