Network


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

Hotspot


Dive into the research topics where Scott C. Supowit is active.

Publication


Featured researches published by Scott C. Supowit.


American Journal of Physiology-heart and Circulatory Physiology | 2008

Deletion of the mouse α-calcitonin gene-related peptide gene increases the vulnerability of the heart to ischemia-reperfusion injury

Ruiping Huang; Amrita Karve; Ibrahim Shah; Mark C. Bowers; Donald J. DiPette; Scott C. Supowit; George S. Abela

Calcitonin gene-related peptide (CGRP), a potent vasodilator released from capsaicin-sensitive C-fiber and Adelta-fiber sensory nerves, has been suggested to play a beneficial role in myocardial ischemia-reperfusion (I/R) injury. Because most previous studies showing a cardioprotective role of CGRP employed pharmacological experiments, the purpose of this study was to utilize a genetic approach by using mice with a targeted deletion of the alpha-CGRP gene to determine whether this neuropeptide had a modulatory function on the severity of I/R injury. To accomplish this goal, isolated, perfused hearts from alpha-CGRP knockout (KO) and wild-type (WT) mice were subjected to 30 min of ischemia followed by 5, 15, and 30 min of reperfusion. Cardiac functional parameters, including coronary flow rates, left ventricular developed pressure, maximum rates of pressure development, and left ventricular end-diastolic pressure, were measured before and after I/R injury, as were levels of creatine kinase, to assess myocardial damage, and malonaldehyde, to assess oxidative stress. Following I/R injury, cardiac performance was significantly reduced in the hearts from the alpha-CGRP KO mice compared with their WT counterparts. The marked reduction in myocardial function in the alpha-CGRP KO hearts compared with WT hearts after I/R injury was associated with a significant elevation in creatine kinase release into the perfusates and malonaldehyde production in the cardiac tissue. Therefore, these data indicate that, in this in vitro setting, deletion of alpha-CGRP makes the heart more vulnerable to I/R injury, possibly due, at least in part, to increased oxidative stress.


Cardiovascular Research | 2011

Substance P induces adverse myocardial remodelling via a mechanism involving cardiac mast cells

Giselle C. Meléndez; Jianping Li; Brittany A. Law; Joseph S. Janicki; Scott C. Supowit; Scott P. Levick

AIMSnSubstance P and neurokinin A (NKA) are sensory nerve neuropeptides encoded by the TAC1 gene. Substance P is a mast cell secretagogue and mast cells are known to play a role in adverse myocardial remodelling. Therefore, we wondered whether substance P and/or NKA modulates myocardial remodelling via a mast cell-mediated mechanism.nnnMETHODS AND RESULTSnVolume overload was induced by aortocaval fistula in TAC1(-/-) mice and their respective wild types. Left ventricular internal diameter of wild-type (WT) fistulas increased by 31.9%; this was prevented in TAC1(-/-) mice (4.2%). Matrix metalloproteinase (MMP) activity was significantly increased in WT fistula mice and was prevented in TAC1(-/-) mice. Myocardial collagen volume fraction was decreased in WT fistula mice; this collagen degradation was not observed in the TAC1(-/-) group. There were no significant differences between any groups in tumour necrosis factor (TNF)-α or cell death. Cardiac mast cells were isolated from rat hearts and stimulated with substance P or NKA. We found that these cells degranulated only to substance P, via the neurokinin-1 receptor. To determine the effect of substance P on mast cells in vivo, volume overload was created in Sprague-Dawley rats treated with the NK-1 receptor antagonist L732138 (5 mg/kg/day) for a period of 3 days. L732138 prevented: (i) increases in cardiac mast cell density; (ii) increased myocardial TNF-α; and (iii) collagen degradation.nnnCONCLUSIONSnOur studies suggest that substance P may be important in mediating adverse myocardial remodelling secondary to volume overload by activating cardiac mast cells, leading to increased TNF-α and MMP activation with subsequent degradation of the extracellular matrix.


Food Science and Nutrition | 2014

Protective effect of resveratrol against pressure overload-induced heart failure

Prakash K. Gupta; Donald J. DiPette; Scott C. Supowit

Transverse aortic constriction (TAC)-induced pressure overload (PO) causes adverse cardiac remodeling and dysfunction that progresses to heart failure (HF). The purpose of this study was to determine whether the potent antioxidant, resveratrol, significantly attenuates PO-induced HF in wild-type mice. Male C57BL6 mice were subjected to either sham or TAC surgery. One group of TAC mice was given daily resveratrol treatment. Echocardiographic, biometric, and immunohistological analyses were performed on the three groups of mice. All echocardiographic parameters demonstrated significantly greater adverse cardiac remodeling and dysfunction in the TAC compared to the sham mice. Increases in the ratios of heart weight (HW)/body weight (BW) and lung weight (LW)/BW and a sharp decline in the percentage of ejection fraction and fractional shortening were found in TAC relative to sham mice. Likewise, the TAC protocol increased markers of oxidative stress, cardiac hypertrophy, inflammation, fibrosis, hypoxia, and apoptosis. These pathological changes were significantly attenuated by resveratrol treatment. Resveratrol treatment significantly attenuates the adverse cardiac remodeling and dysfunction produced by the TAC protocol in C57/BL6 mice and this activity is mediated, at least in part, by the inhibition of oxidative stress and inflammation indicating a therapeutic potential of resveratrol in HF.


Regulatory Peptides | 2013

Alpha-calcitonin gene-related peptide is protective against pressure overload-induced heart failure.

Jianping Li; Scott P. Levick; Donald J. DiPette; Joseph S. Janicki; Scott C. Supowit

The sensory neuropeptide, α-calcitonin gene-related peptide (α-CGRP) is protective against hypertension-induced heart damage and cardiac ischemia/reperfusion injury. To determine whether this neuropeptide is also cardioprotective in heart failure, this study examined whether the absence of α-CGRP exacerbated the adverse cardiac remodeling, dysfunction and mortality in pressure overload heart failure induced by transverse aortic constriction (TAC). Male α-CGRP knockout (KO) and wild type (WT) mice had TAC or sham surgery at day 0 and were studied on days 3, 14, 21, and 28. The survival rate of TAC α-CGRP KO mice was lower than the TAC WT mice over the duration of the protocol. Left ventricular α-CGRP content in TAC WT mice was higher at days 3, 14, and 21 than sham WT mice. Echocardiography demonstrated greater adverse cardiac remodeling and dysfunction in the TAC α-CGRP KO compared to the TAC WT mice. The lung/body weight ratios and left ventricular masses were higher in TAC α-CGRP KO compared to the TAC WT mice. While there was increased cardiac fibrosis in the TAC WT mice compared to shams, the TAC α-CGRP KO mice had markedly increased fibrosis above that of the TAC WT mice. TAC WT mice had greater cardiac inflammation, cell death, and adaptive angiogenesis compared to sham mice. Importantly, the TAC α-CGRP KO mice had greater inflammation, cell death, and attenuation of angiogenesis compared to TAC WT hearts. Thus, α-CGRP plays a significant protective role in TAC-induced heart failure which may be mediated by decreased inflammation, cell death, and fibrosis.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

Knockout of the neurokinin-1 receptor reduces cholangiocyte proliferation in bile duct-ligated mice

Shannon Glaser; Eugenio Gaudio; Anastasia Renzi; Romina Mancinelli; Yoshiyuki Ueno; Julie Venter; Mellanie White; Shelley Kopriva; Valorie L. Chiasson; Sharon DeMorrow; Heather Francis; Fanyin Meng; Marco Marzioni; Antonio Franchitto; Domenico Alvaro; Scott C. Supowit; Donald J. DiPette; Paolo Onori; Gianfranco Alpini

In bile duct-ligated (BDL) rats, cholangiocyte proliferation is regulated by neuroendocrine factors such as α-calcitonin gene-related peptide (α-CGRP). There is no evidence that the sensory neuropeptide substance P (SP) regulates cholangiocyte hyperplasia. Wild-type (WT, (+/+)) and NK-1 receptor (NK-1R) knockout (NK-1R(-/-)) mice underwent sham or BDL for 1 wk. Then we evaluated 1) NK-1R expression, transaminases, and bilirubin serum levels; 2) necrosis, hepatocyte apoptosis and steatosis, and the number of cholangiocytes positive by CK-19 and terminal deoxynucleotidyl transferase biotin-dUTP nick-end labeling in liver sections; 3) mRNA expression for collagen 1α and α-smooth muscle (α-SMA) actin in total liver samples; and 4) PCNA expression and PKA phosphorylation in cholangiocytes. In cholangiocyte lines, we determined the effects of SP on cAMP and D-myo-inositol 1,4,5-trisphosphate levels, proliferation, and PKA phosphorylation. Cholangiocytes express NK-1R with expression being upregulated following BDL. In normal NK-1R(-/-) mice, there was higher hepatocyte apoptosis and scattered hepatocyte steatosis compared with controls. In NK-1R (-)/(-) BDL mice, there was a decrease in serum transaminases and bilirubin levels and the number of CK-19-positive cholangiocytes and enhanced biliary apoptosis compared with controls. In total liver samples, the expression of collagen 1α and α-SMA increased in BDL compared with normal mice and decreased in BDL NK-1R(-/-) compared with BDL mice. In cholangiocytes from BDL NK-1R (-)/(-) mice there was decreased PCNA expression and PKA phosphorylation. In vitro, SP increased cAMP levels, proliferation, and PKA phosphorylation of cholangiocytes. Targeting of NK-1R may be important in the inhibition of biliary hyperplasia in cholangiopathies.


American Journal of Physiology-renal Physiology | 2013

Renal protective effects of α-calcitonin gene-related peptide in deoxycorticosterone-salt hypertension

Jianping Li; Kevin A. Carnevale; Donald J. DiPette; Scott C. Supowit

Deoxycorticosterone salt (DOC-salt) hypertension-induced renal damage is enhanced in α-calcitonin gene-related peptide (α-CGRP) knockout (KO) compared with wild-type (WT) mice. However, since the α-CGRP KO mice have a 15-20 mmHg higher baseline mean arterial pressure (MAP) than WT mice, they also have a higher MAP than WT mice throughout the course of DOC-salt hypertension. To determine the mechanism by which the absence of α-CGRP enhances hypertension-induced renal damage, DOC-salt hypertension was induced in telemetry probe implanted α-CGRP KO and WT mice. To equalize the blood pressure (BP) to that of DOC-salt WT mice, an additional group of DOC-salt α-CGRP KO mice was given 0.025% hydralazine to drink. The DOC-salt protocol increased the final MAP in α-CGRP KO mice to 155 ± 6 mmHg and in WT mice to 140 ± 5 mmHg. The MAP of the hydralazine-treated DOC-salt α-CGRP KO mice was 139 ± 6 mmHg. Urinary excretion of microalbumin and isoprostane, a marker for oxidative stress, was increased, and creatinine clearance was decreased in DOC-salt α-CGRP KO compared with DOC-salt WT mice. Equalization of the MAP in DOC-salt α-CGRP KO to that of DOC-salt WT mice did not significantly improve these parameters. Renal macrophage infiltration; desmin, a marker of podocyte damage; and the inflammatory cytokines TNF-α and IFN-γ and the chemokines monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1α (MIP-1α) were increased in DOC-salt α-CGRP KO mice and were not reduced by hydralazine treatment. However, BP equalization did improve the renal histopathological damage, as determined by light microscopy. Therefore, in DOC-salt hypertension in mice, the mechanism(s) of the renal protective effects of α-CGRP are both BP independent and BP dependent.


Regulatory Peptides | 2011

Bradykinin and prostaglandin E1 regulate calcitonin gene-related peptide expression in cultured rat sensory neurons

Scott C. Supowit; Huawei Zhao; Khurshed A. Katki; Prakash C. Gupta; Donald J. DiPette

Primary cultures of adult rat dorsal root ganglia (DRG) sensory neurons were used to determine whether bradykinin and prostaglandins E₁ (PGE₁), E₂ (PGE₂) or I₂ (PGI₂) stimulate long-term calcitonin gene-related peptide (CGRP) mRNA accumulation and peptide release. Treatment (24 h) of neurons with either bradykinin or PGE₁, significantly increased CGRP mRNA content and iCGRP release. However, PGE₂ or PGI₂ was without effect. Exposure of the cultured neurons to increasing concentrations of bradykinin or PGE₁ demonstrated that the stimulation of CGRP expression was concentration-dependent, while time-course studies showed that maximal levels of CGRP mRNA accumulation and peptide release were maintained for at least 48 h. Treatment of the neuronal cultures with a bradykinin B₂ receptor antagonist significantly inhibited the bradykinin-induced increase in CGRP expression and release. In addition, preincubation of neuronal cultures with the cyclooxygenase inhibitor indomethacin did not alter the PGE₁-mediated stimulation of CGRP but blocked completely the bradykinin-induced increase in CGRP production. Therefore, these data indicate that bradykinin and PGE₁ can regulate the synthesis and release of CGRP in DRG neurons and that the stimulatory effects of bradykinin on CGRP are mediated by a cyclooxygenase product(s). Thus, these findings suggest a direct relationship between chronic alterations in bradykinin/prostaglandin production that may arise from pathophysiological causes and long-term changes in CGRP expression.


American Journal of Physiology-heart and Circulatory Physiology | 2005

Calcitonin gene-related peptide and substance P contribute to reduced blood pressure in sympathectomized rats

Scott C. Supowit; R. T. Ethridge; Huawei Zhao; Khurshed A. Katki; Donald J. DiPette


Hypertension | 2014

Abstract 418: Substance P and Mast Cell Interaction Exacerbates Pressure Overload-Induced Cardiac Remodeling

Scott C. Supowit; Jianping Li; Donald J. DiPette


Hypertension | 2013

Abstract 100: Mechanisms That Mediate the Cardioprotective Activities of Resveratrol in Pressure Overload-Induced Heart Failure

Scott C. Supowit; Prakash C. Gupta; Donald J. DiPette

Collaboration


Dive into the Scott C. Supowit's collaboration.

Top Co-Authors

Avatar

Donald J. DiPette

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jianping Li

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph S. Janicki

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Scott P. Levick

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Antonio Franchitto

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Paolo Onori

Sapienza University of Rome

View shared research outputs
Researchain Logo
Decentralizing Knowledge