Network


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

Hotspot


Dive into the research topics where Kenneth E. Chapman is active.

Publication


Featured researches published by Kenneth E. Chapman.


Circulation Research | 2008

IP3 Constricts Cerebral Arteries via IP3 Receptor–Mediated TRPC3 Channel Activation and Independently of Sarcoplasmic Reticulum Ca2+ Release

Qi Xi; Adebowale Adebiyi; Guiling Zhao; Kenneth E. Chapman; Christopher M. Waters; Aviv Hassid; Jonathan H. Jaggar

Vasoconstrictors that bind to phospholipase C–coupled receptors elevate inositol-1,4,5-trisphosphate (IP3). IP3 is generally considered to elevate intracellular Ca2+ concentration ([Ca2+]i) in arterial myocytes and induce vasoconstriction via a single mechanism: by activating sarcoplasmic reticulum (SR)-localized IP3 receptors, leading to intracellular Ca2+ release. We show that IP3 also stimulates vasoconstriction via a SR Ca2+ release–independent mechanism. In isolated cerebral artery myocytes and arteries in which SR Ca2+ was depleted to abolish Ca2+ release (measured using D1ER, a fluorescence resonance energy transfer–based SR Ca2+ indicator), IP3 activated 15 pS sarcolemmal cation channels, generated a whole-cell cation current (ICat) caused by Na+ influx, induced membrane depolarization, elevated [Ca2+]i, and stimulated vasoconstriction. The IP3-induced ICat and [Ca2+]i elevation were attenuated by cation channel (Gd3+, 2-APB) and IP3 receptor (xestospongin C, heparin, 2-APB) blockers. TRPC3 (canonical transient receptor potential 3) channel knockdown with short hairpin RNA and diltiazem and nimodipine, voltage-dependent Ca2+ channel blockers, reduced the SR Ca2+ release–independent, IP3-induced [Ca2+]i elevation and vasoconstriction. In pressurized arteries, SR Ca2+ depletion did not alter IP3-induced constriction at 20 mm Hg but reduced IP3-induced constriction by ≈39% at 60 mm Hg. [Ca2+]i elevations and constrictions induced by endothelin-1, a phospholipase C–coupled receptor agonist, were both attenuated by TRPC3 knockdown and xestospongin C in SR Ca2+-depleted arteries. In summary, we describe a novel mechanism of IP3-induced vasoconstriction that does not occur as a result of SR Ca2+ release but because of IP3 receptor–dependent ICat activation that requires TRPC3 channels. The resulting membrane depolarization activates voltage-dependent Ca2+ channels, leading to a myocyte [Ca2+]i elevation, and vasoconstriction.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2008

Localized elasticity measured in epithelial cells migrating at a wound edge using atomic force microscopy

Ajay A. Wagh; Esra Roan; Kenneth E. Chapman; Leena P. Desai; David A. Rendon; Eugene C. Eckstein; Christopher M. Waters

Restoration of lung homeostasis following injury requires efficient wound healing by the epithelium. The mechanisms of lung epithelial wound healing include cell spreading and migration into the wounded area and later cell proliferation. We hypothesized that mechanical properties of cells vary near the wound edge, and this may provide cues to direct cell migration. To investigate this hypothesis, we measured variations in the stiffness of migrating human bronchial epithelial cells (16HBE cells) approximately 2 h after applying a scratch wound. We used atomic force microscopy (AFM) in contact mode to measure the cell stiffness in 1.5-microm square regions at different locations relative to the wound edge. In regions far from the wound edge (>2.75 mm), there was substantial variation in the elastic modulus in specific cellular regions, but the median values measured from multiple fields were consistently lower than 5 kPa. At the wound edge, cell stiffness was significantly lower within the first 5 microm but increased significantly between 10 and 15 microm before decreasing again below the median values away from the wound edge. When cells were infected with an adenovirus expressing a dominant negative form of RhoA, cell stiffness was significantly decreased compared with cells infected with a control adenovirus. In addition, expression of dominant negative RhoA abrogated the peak increase in stiffness near the wound edge. These results suggest that cells near the wound edge undergo localized changes in cellular stiffness that may provide signals for cell spreading and migration.


Journal of Cellular Physiology | 2002

Continuous exposure of airway epithelial cells to hydrogen peroxide: Protection by KGF

Kenneth E. Chapman; Christopher M. Waters; William M. Miller

Reactive oxygen species (ROS) increase permeability in the airway epithelium. Extended periods of oxidant exposure may be experienced by those suffering from chronic inflammation of the lungs, receiving supplemental oxygen, or living in areas with high levels of air pollution. We studied the effects of long‐term, continuous exposure to hydrogen peroxide (H2O2) on the trans‐epithelial electrical resistance (TER) across cultured monolayers of a transformed cell line of human bronchial epithelial cells, 16HBE14o− (16HBE). A TER perfusion system was employed to continuously monitor the TER without disturbing the tissue model. The TER decreased in a dose‐dependent manner with increasing concentrations of H2O2 (0.1, 0.5, and 1.0 mM), regardless of pre‐incubation conditions. Cell cultures pre‐treated with 50 ng/ml keratinocyte growth factor (KGF) showed a significant delay in oxidant‐induced TER decreases caused by 0.1 mM H2O2. Exposure to 0.1 mM H2O2 for 350 min led to disruption of tight junction proteins, ZO‐1 and occludin, but KGF treatment prevented this damage. The recovery of epithelial barrier function after exposure to oxidants was also studied. Tissue models exposed to 0.5 mM H2O2 for 25 min showed complete recovery of TER after 20 h, independent of culture pre‐treatment. In contrast, KGF pre‐incubation enhanced the recovery of 16HBE cultures exposed for 50 min to 0.5 mM H2O2.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2005

Cyclic mechanical strain increases reactive oxygen species production in pulmonary epithelial cells

Kenneth E. Chapman; Scott E. Sinclair; Daming Zhuang; Aviv Hassid; Leena P. Desai; Christopher M. Waters


Molecular Biology of the Cell | 2004

Regulation of Cell Motility by Tyrosine Phosphorylated Villin

Alok Tomar; Yaohong Wang; Narendra Kumar; Sudeep P. George; Bogdan Ceacareanu; Aviv Hassid; Kenneth E. Chapman; Ashish M. Aryal; Christopher M. Waters; Seema Khurana


American Journal of Physiology-lung Cellular and Molecular Physiology | 2007

High tidal volume mechanical ventilation with hyperoxia alters alveolar type II cell adhesion

Leena P. Desai; Scott E. Sinclair; Kenneth E. Chapman; Aviv Hassid; Christopher M. Waters


American Journal of Physiology-heart and Circulatory Physiology | 2004

Nitric oxide attenuates insulin- or IGF-I-stimulated aortic smooth muscle cell motility by decreasing H2O2 levels: essential role of cGMP

Darning Zhuang; Alice-Corina Ceacareanu; Yi Lin; Bogdan Ceacareanu; Madhulika Dixit; Kenneth E. Chapman; Christopher M. Waters; Gadiparthi N. Rao; Aviv Hassid


American Journal of Physiology-cell Physiology | 2006

Nitric oxide attenuates IGF-I-induced aortic smooth muscle cell motility by decreasing Rac1 activity: essential role of PTP-PEST and p130cas

Alice Corina Ceacareanu; Bogdan Ceacareanu; Daming Zhuang; Yingzi Chang; Ramesh M. Ray; Leena P. Desai; Kenneth E. Chapman; Christopher M. Waters; Aviv Hassid


Archive | 2015

cultureproperties of alveolar epithelial cells in

Mark A. Schroeder; Rolf D. Hubmayr; Christopher M. Waters; Ajay A. Wagh; Esra Roan; Kenneth E. Chapman; Leena P. Desai; David A. Rendon; C Eugene; Evren U. Azeloglu; Jahar Bhattacharya; Kevin D. Costa


Archive | 2013

role of PTP-PEST and p130cas muscle cell motility by decreasing Rac1 activity: essential Nitric oxide attenuates IGF-I-induced aortic smooth

M. Ray; Leena P. Desai; Kenneth E. Chapman; Christopher M. Waters; Aviv Hassid; Alice-Corina Ceacareanu; Bogdan Ceacareanu; Daming Zhuang; Yingzi Chang

Collaboration


Dive into the Kenneth E. Chapman's collaboration.

Top Co-Authors

Avatar

Christopher M. Waters

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Aviv Hassid

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Leena P. Desai

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Bogdan Ceacareanu

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Alice-Corina Ceacareanu

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Daming Zhuang

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Scott E. Sinclair

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Adebowale Adebiyi

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Ajay A. Wagh

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Darning Zhuang

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge