Network


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

Hotspot


Dive into the research topics where Miensheng Chu is active.

Publication


Featured researches published by Miensheng Chu.


Journal of Molecular and Cellular Cardiology | 2011

Nogo-A knockdown inhibits hypoxia/reoxygenation-induced activation of mitochondrial-dependent apoptosis in cardiomyocytes

Jason Sarkey; Miensheng Chu; M. McShane; E. Bovo; Y. Ait Mou; Aleksey V. Zima; P.P. de Tombe; Gwendolyn L. Kartje; Jody L. Martin

Programmed cell death of cardiomyocytes following myocardial ischemia increases biomechanical stress on the remaining myocardium, leading to myocardial dysfunction that may result in congestive heart failure or sudden death. Nogo-A is well characterized as a potent inhibitor of axonal regeneration and plasticity in the central nervous system, however, the role of Nogo-A in non-nervous tissues is essentially unknown. In this study, Nogo-A expression was shown to be significantly increased in cardiac tissue from patients with dilated cardiomyopathy and from patients who have experienced an ischemic event. Nogo-A expression was clearly associated with cardiomyocytes in culture and was localized predominantly in the endoplasmic reticulum. In agreement with the findings from human tissue, Nogo-A expression was significantly increased in cultured neonatal rat cardiomyocytes subjected to hypoxia/reoxygenation. Knockdown of Nogo-A in cardiomyocytes markedly attenuated hypoxia/reoxygenation-induced apoptosis, as indicated by the significant reduction of DNA fragmentation, phosphatidylserine translocation, and caspase-3 cleavage, by a mechanism involving the preservation of mitochondrial membrane potential, the inhibition of ROS accumulation, and the improvement of intracellular calcium regulation. Together, these data demonstrate that knockdown of Nogo-A may serve as a novel therapeutic strategy to prevent the loss of cardiomyocytes following ischemic/hypoxic injury.


Cardiovascular Research | 2011

Serine 910 Phosphorylation of Focal Adhesion Kinase is Critical for Sarcomere Reorganization In Cardiomyocyte Hypertrophy

Miensheng Chu; Rekha Iyengar; Yevgeniya E. Koshman; Taehoon Kim; Brenda Russell; Jody L. Martin; Alain Heroux; Seth L. Robia; Allen M. Samarel

AIMS Tyrosine-phosphorylated focal adhesion kinase (FAK) is required for the hypertrophic response of cardiomyocytes to growth factors and mechanical load, but the role of FAK serine phosphorylation in this process is unknown. The aims of the present study were to characterize FAK serine phosphorylation in cultured neonatal rat ventricular myocytes (NRVM), analyse its functional significance during hypertrophic signalling, and examine its potential role in the pathogenesis of human dilated cardiomyopathy (DCM). METHODS AND RESULTS Endothelin-1 (ET-1) and other hypertrophic factors induced a time- and dose-dependent increase in FAK-S910 phosphorylation. ET-1-induced FAK-S910 phosphorylation required ET(A)R-dependent activation of PKCδ and Src via parallel Raf-1 → MEK1/2 → ERK1/2 and MEK5 → ERK5 signalling pathways. Replication-deficient adenoviruses expressing wild-type (WT) FAK and a non-phosphorylatable, S910A-FAK mutant were then used to examine the functional significance of FAK-S910 phosphorylation. Unlike WT-FAK, S910A-FAK increased the half-life of GFP-tagged paxillin within costameres (as determined by total internal reflection fluorescence microscopy and fluorescence recovery after photobleaching) and increased the steady-state FAK-paxillin interaction (as determined by co-immunoprecipitation and western blotting). These alterations resulted in reduced NRVM sarcomere reorganization and cell spreading. Finally, we found that FAK was serine-phosphorylated at multiple sites in non-failing, human left ventricular tissue. FAK-S910 phosphorylation and ERK5 expression were dramatically reduced in patients undergoing heart transplantation for end-stage DCM. CONCLUSION FAK undergoes S910 phosphorylation via PKCδ and Src-dependent pathways that are important for cell spreading and sarcomere reorganization. Reduced FAK-S910 phosphorylation may contribute to sarcomere disorganization in DCM.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Focal Adhesion Kinase–Related Nonkinase Inhibits Vascular Smooth Muscle Cell Invasion by Focal Adhesion Targeting, Tyrosine 168 Phosphorylation, and Competition for p130 Cas Binding

Yevgeniya E. Koshman; Miensheng Chu; Steven J. Engman; Taehoon Kim; Rekha Iyengar; Seth L. Robia; Allen M. Samarel

Objective—Focal adhesion kinase–related nonkinase (FRNK), the C-terminal domain of focal adhesion kinase (FAK), is a tyrosine-phosphorylated, vascular smooth muscle cell (VSMC)–specific inhibitor of cell migration. FRNK inhibits both FAK and proline-rich tyrosine kinase 2 (PYK2) in cultured VSMCs, and both kinases may be involved in VSMC invasion during vascular remodeling. Methods and Results—Adenovirally mediated gene transfer of green fluorescent protein–tagged, wild-type (wt) FRNK into balloon-injured rat carotid arteries confirmed that FRNK overexpression inhibited both FAK and PYK2 phosphorylation and downstream signaling in vivo. To identify which kinase was involved in regulating VSMC invasion, adenovirally mediated expression of specific short hairpin RNAs was used to knock down FAK versus PYK2 in cultured VSMCs, but only FAK short hairpin RNA was effective in reducing VSMC invasion. The role of FRNK tyrosine phosphorylation was then examined using adenoviruses expressing nonphosphorylatable (Tyr168Phe-, Tyr232Phe-, and Tyr168,232Phe-) green fluorescent protein–FRNK mutants. wtFRNK and all FRNK mutants localized to FAs, but only Tyr168 phosphorylation was required for FRNK to inhibit invasion. Preventing Tyr168 phosphorylation also increased FRNK-paxillin interaction, as determined by coimmunoprecipitation, total internal reflection fluorescence microscopy, and fluorescence recovery after photobleaching. Furthermore, wtFRNK competed with FAK for binding to p130Cas (a critically important regulator of cell migration) and prevented its phosphorylation. However, Tyr168Phe-FRNK was unable to bind p130Cas. Conclusion—We propose a 3-stage mechanism for FRNK inhibition: focal adhesion targeting, Tyr168 phosphorylation, and competition with FAK for p130Cas binding and phosphorylation, which are all required for FRNK to inhibit VSMC invasion.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

FRNK Inhibition of Focal Adhesion Kinase–Dependent Signaling and Migration in Vascular Smooth Muscle Cells

Yevgeniya E. Koshman; Taehoon Kim; Miensheng Chu; Steven J. Engman; Rekha Iyengar; Seth L. Robia; Allen M. Samarel

Objective—To examine whether interference with FRNK targeting to focal adhesions (FAs) affects its inhibitory activity and tyrosine phosphorylation. Methods and Results—Focal adhesion kinase and its autonomously expressed C-terminal inhibitor, focal adhesion kinase-related nonkinase (FRNK), regulate vascular smooth muscle cell (VSMC) signaling and migration. FRNK-paxillin binding was reduced by a point mutation in its FA targeting domain (L341S-FRNK). Green fluorescent protein-tagged wild type and L341S-FRNK were then adenovirally expressed in VSMCs. L341S-FRNK targeted to VSMC FAs, despite previous studies in other cell types. L341S-FRNK affected FA binding kinetics (assessed by total internal reflection fluorescnece [TIRF] microscopy and fluorescence recovery after photobleaching [FRAP]) and reduced its steady-state paxillin interaction (determined by coimmunoprecipitation). Both wt-FRNK and L341S-FRNK lowered basal and angiotensin II-stimulated focal adhesion kinase, paxillin, and extracellular signal-regulated kinase 1/2 phosphorylation. However, the degree of inhibition was significantly reduced by L341S-FRNK. L341S-FRNK also demonstrated significantly greater migratory activity compared with wt-FRNK-expressing VSMCs. Angiotensin II-induced Y168 phosphorylation was Src dependent, as evident by a significant reduction in Y168 phosphorylation by the Src family kinase inhibitor PP2 is 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2). Surprisingly, Y168 phosphorylation was unaffected by its targeting. Furthermore, Y232 phosphorylation increased approximately 3-fold in L341S-FRNK, which was less sensitive to PP2. Conclusion—FRNK inhibition of VSMC migration requires both FA targeting and Y168 phosphorylation by Src family kinases. FRNK-Y232 phosphorylation occurs outside of FAs, probably by a PP2-insensitive kinase.


Journal of Molecular and Cellular Cardiology | 2014

Cardiomyocyte-specific expression of CRNK, the C-terminal domain of PYK2, maintains ventricular function and slows ventricular remodeling in a mouse model of dilated cardiomyopathy.

Yevgeniya E. Koshman; Miensheng Chu; Taehoon Kim; Olivia Kalmanson; Mariam Farjah; Mohit Kumar; William Lewis; David L. Geenen; Pieter P. de Tombe; Paul H. Goldspink; R. John Solaro; Allen M. Samarel

Up-regulation and activation of PYK2, a member of the FAK family of protein tyrosine kinases, is involved in the pathogenesis of left ventricular (LV) remodeling and heart failure (HF). PYK2 activation can be prevented by CRNK, the C-terminal domain of PYK2. We previously demonstrated that adenoviral-mediated CRNK gene transfer improved survival and LV function, and slowed LV remodeling in a rat model of coronary artery ligation-induced HF. We now interrogate whether cardiomyocyte-specific, transgenic CRNK expression prevents LV remodeling and HF in a mouse model of dilated cardiomyopathy (DCM) caused by constitutively active Protein Kinase Cε (caPKCε). Transgenic (TG; FVB/N background) mice were engineered to express rat CRNK under control of the α-myosin heavy chain promoter, and crossed with FVB/N mice with cardiomyocyte-specific expression of caPKCε to create double TG mice. LV structure, function, and gene expression were evaluated in all 4 groups (nonTG FVB/N; caPKCε(+/-); CRNK(+/-); and caPKCε×CRNK (PXC) double TG mice) at 1, 3, 6, 9 and 12mo of age. CRNK expression followed a Mendelian distribution, and CRNK mice developed and survived normally through 12mo. Cardiac structure, function and selected gene expression of CRNK mice were similar to nonTG littermates. CRNK had no effect on caPKCε expression and vice versa. PYK2 was up-regulated ~6-fold in caPKCε mice, who developed a non-hypertrophic, progressive DCM with reduced systolic (Contractility Index=151±5 vs. 90±4s(-1)) and diastolic (Tau=7.5±0.5 vs. 14.7±1.3ms) function, and LV dilatation (LV Remodeling Index (LVRI)=4.2±0.1 vs. 6.0±0.3 for FVB/N vs. caPKCε mice, respectively; P<0.05 for each at 12mo). In double TG PXC mice, CRNK expression significantly prolonged survival, improved contractile function (Contractile Index=115±8s(-1); Tau=9.5±1.0ms), and reduced LV remodeling (LVRI=4.9±0.1). Cardiomyocyte-specific expression of CRNK improves contractile function and slows LV remodeling in a mouse model of DCM.


Archive | 2012

Mechanotransduction in Cardiac Hypertrophy and Ischemia

Miensheng Chu; Yevgeniya E. Koshman; Allen M. Samarel

Mechanotransduction is the process by which load-bearing cells sense physical forces, transduce the forces into biochemical signals, and generate adaptive or maladaptive responses that lead to alterations in cell structure and function. Mechanotransduction in the heart not only affects the beat-to-beat regulation of cardiac performance, but also profoundly affects the growth, differentiation, and survival of the cellular components that comprise the human myocardium. Understanding the molecular basis for mechanotransduction is, therefore, important to our overall understanding of growth regulation and function during cardiac hypertrophy and ischemia. Cardiomyocytes rely on several intracellular components to sense mechanical load, and convert mechanical stimuli into biochemical events that affect cellular structure and function. These sensors include protein components within the myofilaments and Z-discs, integrins and other membrane-associated proteins that link the extracellular matrix to the cytoskeleton, and stretch-activated ion channels. A complex signaling web then transmits signals from mechanosensors to the nucleus and other organelles. Ultimately, it is hoped that new pharmacological and molecular genetic approaches targeted to specific components of the mechanotransduction machinery will be developed to translate this wealth of basic knowledge into therapeutic strategies designed to reduce cardiac hypertrophy, further protect the ischemic myocardium, and prevent its transition to heart failure.


Journal of Cardiac Failure | 2013

Regulation of Connective Tissue Growth Factor Gene Expression and Fibrosis in Human Heart Failure

Yevgeniya E. Koshman; Nilamkumar Patel; Miensheng Chu; Rekha Iyengar; Taehoon Kim; Çağatay Erşahin; William Lewis; Alain Heroux; Allen M. Samarel


Archive | 2013

Basic Science and Experimental Study Regulation of Connective Tissue Growth Factor Gene Expression and Fibrosis in Human Heart Failure

Yevgeniya E. Koshman; Nilamkumar Patel; Miensheng Chu; Rekha Iyengar; Taehoon Kim; Çağatay Erşahin; William Lewis; Alain Heroux; Allen M. Samarel


The FASEB Journal | 2011

S910 phosphorylation of focal adhesion kinase (FAK) regulates cell spreading and Z-body formation in hypertrophying cardiomyocytes

Miensheng Chu; Rekha Iyengar; Taehoon Kim; Yevgeniya E. Koshman; Seth L. Robia; Allen M. Samarel


Biophysical Journal | 2011

Erratum: (Biophysical Journal)

Jason Sarkey; Miensheng Chu; M. McShane; Elisa Bovo; Y. Ait Mou; Aleksey V. Zima; P. P. De Tombe; Gwendolyn L. Kartje; Jody L. Martin

Collaboration


Dive into the Miensheng Chu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Taehoon Kim

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Rekha Iyengar

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Seth L. Robia

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Alain Heroux

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jody L. Martin

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aleksey V. Zima

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge