Network


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

Hotspot


Dive into the research topics where Mengcheng Shen is active.

Publication


Featured researches published by Mengcheng Shen.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Divergent roles of matrix metalloproteinase 2 in pathogenesis of thoracic aortic aneurysm.

Mengcheng Shen; Jiwon Lee; Ratnadeep Basu; Siva S.V.P. Sakamuri; Xiuhua Wang; Dong Fan; Zamaneh Kassiri

Objective— Aortic aneurysm, focal dilation of the aorta, results from impaired integrity of aortic extracellular matrix (ECM). Matrix metalloproteinases (MMPs) are traditionally known as ECM-degrading enzymes. MMP2 has been associated with aneurysm in patients and in animal models. We investigated the role of MMP2 in thoracic aortic aneurysm using 2 models of aortic remodeling and aneurysm. Approach and Results— Male 10-week-old MMP2-deficient (MMP2−/−) and wild-type mice received angiotensin II (Ang II, 1.5 mg/kg/day) or saline (Alzet pump) for 4 weeks. Although both genotypes exhibited dilation of the ascending aorta after Ang II infusion, MMP2−/− mice showed more severe dilation of the thoracic aorta and thoracic aortic aneurysm. The Ang II–induced increase in elastin and collagen (mRNA and protein) was markedly suppressed in MMP2−/− thoracic aorta and smooth muscle cells, whereas only mRNA levels were reduced in MMP2−/−-Ang II abdominal aorta. Consistent with the absence of MMP2, proteolytic activities were lower in MMP2−/−-Ang II compared with wild-type-Ang II thoracic and abdominal aorta. MMP2-deficiency suppressed the activation of latent transforming growth factor-&bgr; and the Smad2/3 pathway in vivo and in vitro. Intriguingly, MMP2−/− mice were protected against CaCl2-induced thoracic aortic aneurysm, which triggered ECM degradation but not synthesis. Conclusions— This study reveals the dual role of MMP2 in ECM degradation, as well as ECM synthesis. Moreover, the greater susceptibility of the thoracic aorta to impaired ECM synthesis, compared with vulnerability of the abdominal aorta to aberrant ECM degradation, provides an insight into the regional susceptibility of the aorta to aneurysm development.


Circulation-heart Failure | 2014

Myocardial Recovery From Ischemia–Reperfusion Is Compromised in the Absence of Tissue Inhibitor of Metalloproteinase 4

Abhijit Takawale; Dong Fan; Ratnadeep Basu; Mengcheng Shen; Nirmal Parajuli; Wang Wang; Xiuhua Wang; Gavin Y. Oudit; Zamaneh Kassiri

Background—Myocardial reperfusion after ischemia (I/R), although an effective approach in rescuing the ischemic myocardium, can itself trigger several adverse effects including aberrant remodeling of the myocardium and its extracellular matrix. Tissue inhibitor of metalloproteinases (TIMPs) protect the extracellular matrix against excess degradation by matrix metalloproteinases (MMPs). TIMP4 levels are reduced in myocardial infarction; however, its causal role in progression of post-I/R injury has not been explored. Methods and Results—In vivo I/R (20-minute ischemia, 1-week reperfusion) resulted in more severe systolic and diastolic dysfunction in TIMP4−/− mice with enhanced inflammation, oxidative stress (1 day post-I/R), hypertrophy, and interstitial fibrosis (1 week). After an initial increase in TIMP4 (1 day post-I/R), TIMP4 mRNA and protein decreased in the ischemic myocardium from wild-type mice by 1 week post-I/R and in tissue samples from patients with myocardial infarction, which correlated with enhanced activity of membrane-bound MMP, membrane-type 1 MMP. By 4 weeks post-I/R, wild-type mice showed no cardiac dysfunction, elevated TIMP4 levels (to baseline), and normalized membrane-type 1 MMP activity. TIMP4-deficient mice, however, showed exacerbated diastolic dysfunction, sustained elevation of membrane-type 1 MMP activity, and worsened myocardial hypertrophy and fibrosis. Ex vivo I/R (20- or 30-minute ischemia, 45-minute reperfusion) resulted in comparable cardiac dysfunction in wild-type and TIMP4−/− mice. Conclusions—TIMP4 is essential for recovery from myocardial I/R in vivo, primarily because of its membrane-type 1 MMP inhibitory function. TIMP4 deficiency does not increase susceptibility to ex vivo I/R injury. Replenishment of myocardial TIMP4 could serve as an effective therapy in post-I/R recovery for patients with reduced TIMP4.


Journal of Molecular and Cellular Cardiology | 2016

ADAMs family and relatives in cardiovascular physiology and pathology

Pu Zhang; Mengcheng Shen; Carlos Fernandez-Patron; Zamaneh Kassiri

A disintegrin and metalloproteinases (ADAMs) are a family of membrane-bound proteases. ADAM-TSs (ADAMs with thrombospondin domains) are a close relative of ADAMs that are present in soluble form in the extracellular space. Dysregulated production or function of these enzymes has been associated with pathologies such as cancer, asthma, Alzheimers and cardiovascular diseases. ADAMs contribute to angiogenesis, hypertrophy and apoptosis in a stimulus- and cell type-dependent manner. Among the ADAMs identified so far (34 in mouse, 21 in human), ADAMs 8, 9, 10, 12, 17 and 19 have been shown to be involved in cardiovascular development or cardiomyopathies; and among the 19 ADAM-TSs, ADAM-TS1, 5, 7 and 9 are important in development of the cardiovascular system, while ADAM-TS13 can contribute to vascular disorders. Meanwhile, there remain a number of ADAMs and ADAM-TSs whose function in the cardiovascular system has not been yet explored. The current knowledge about the role of ADAMs and ADAM-TSs in the cardiovascular pathologies is still quite limited. The most detailed studies have been performed in other cell types (e.g. cancer cells) and organs (nervous system) which can provide valuable insight into the potential functions of ADAMs and ADAM-TSs, their mechanism of action and therapeutic potentials in cardiomyopathies. Here, we review what is currently known about the structure and function of ADAMs and ADAM-TSs, and their roles in development, physiology and pathology of the cardiovascular system.


Hypertension | 2016

A Disintegrin and Metalloprotease-17 Regulates Pressure Overload–Induced Myocardial Hypertrophy and Dysfunction Through Proteolytic Processing of Integrin β1

Dong Fan; Abhijit Takawale; Mengcheng Shen; Victor Samokhvalov; Ratnadeep Basu; Vaibhav B. Patel; Xiuhua Wang; Carlos Fernandez-Patron; John M. Seubert; Gavin Y. Oudit; Zamaneh Kassiri

A disintegrin and metalloprotease-17 (ADAM17) belongs to a family of transmembrane enzymes, and it can mediate ectodomain shedding of several membrane-bound molecules. ADAM17 levels are elevated in patients with hypertrophic and dilated cardiomyopathy; however, its direct role in hypertrophic cardiomyopathy is unknown. Cardiomyocyte-specific ADAM17 knockdown mice (ADAM17flox/flox/&agr;MHC-Cre; ADAM17f/f/Cre) and littermates with intact ADAM17 levels (ADAM17f/f) were subjected to cardiac pressure–overload by transverse aortic constriction. Cardiac function/architecture was assessed by echocardiography at 2 and 5 weeks post transverse aortic constriction. ADAM17 knockdown enhanced myocardial hypertrophy, fibrosis, more severe left ventricular dilation, and systolic dysfunction at 5 weeks post transverse aortic constriction. Pressure overload–induced upregulation of integrin &bgr;1 was much greater with ADAM17 knockdown, concomitant with the greater activation of the focal adhesion kinase pathway, suggesting that integrin &bgr;1 could be a substrate for ADAM17. ADAM17 knockdown did not alter other cardiomyocyte integrins, integrin &agr;5 or &agr;7, and HB-EGF (heparin-bound epidermal growth factor), another potential substrate for ADAM17, remained unaltered after pressure overload. ADAM17-mediated cleavage of integrin &bgr;1 was confirmed by an in vitro assay. Intriguingly, ADAM17 knockdown did not affect the myocardial hypertrophy induced by a subpressor dose of angiotensin II, which occurs independent from the integrin &bgr;1–mediated pathway. ADAM17-knockdown enhanced the hypertrophic response to cyclic mechanical stretching in neonatal rat cardiomyocytes. This study reports a novel cardioprotective function for ADAM17 in pressure overload cardiomyopathy, where loss of ADAM17 promotes hypertrophy by reducing the cleavage of cardiac integrin &bgr;1, a novel substrate for ADAM17. This function of ADAM17 is selective for pressure overload–induced myocardial hypertrophy and dysfunction, and not agonist-induced hypertrophy.


Circulation-heart Failure | 2015

Cardiomyocyte A Disintegrin And Metalloproteinase 17 (ADAM17) Is Essential in Post–Myocardial Infarction Repair by Regulating Angiogenesis

Dong Fan; Abhijit Takawale; Mengcheng Shen; Wang Wang; Xiuhua Wang; Ratnadeep Basu; Gavin Y. Oudit; Zamaneh Kassiri

Background—A disintegrin and metalloproteinase 17 (ADAM17) is a membrane-bound enzyme that mediates shedding of many membrane-bound molecules, thereby regulating multiple cellular responses. We investigated the role of cardiomyocyte ADAM17 in myocardial infarction (MI). Methods and Results—Cardiomyocyte-specific ADAM17 knockdown mice (ADAM17flox/flox/&agr;-MHC-Cre; f/f/Cre) and parallel controls (ADAM17flox/flox; f/f) were subjected to MI by ligation of the left anterior descending artery. Post MI, f/f/Cre mice showed compromised survival, higher rates of cardiac rupture, more severe left ventricular dilation, and suppressed ejection fraction compared with parallel f/f-MI mice. Ex vivo ischemic injury (isolated hearts) resulted in comparable recovery in both genotypes. Myocardial vascular density (fluorescent-labeled lectin perfusion and CD31 immunofluorescence staining) was significantly lower in the infarct areas of f/f/Cre-MI compared with f/f-MI mice. Activation of vascular endothelial growth factor receptor 2 (VEGFR2), its mRNA, and total protein levels were reduced in infarcted myocardium in ADAM17 knockdown mice. Transcriptional regulation of VEGFR2 by ADAM17 was confirmed in cocultured cardiomyocyte–fibroblast as ischemia-induced VEGFR2 expression was blocked by ADAM17-siRNA. Meanwhile, ADAM17-siRNA did not alter VEGFA bioavailability in the conditioned media. ADAM17 knockdown mice (f/f/Cre-MI) exhibited reduced nuclear factor-&kgr;B activation (DNA binding) in the infarcted myocardium, which could underlie the suppressed VEGFR2 expression in these hearts. Post MI, inflammatory response was not altered by ADAM17 downregulation. Conclusions—This study highlights the key role of cardiomyocyte ADAM17 in post-MI recovery by regulating VEGFR2 transcription and angiogenesis, thereby limiting left ventricular dilation and dysfunction. Therefore, ADAM17 upregulation, within the physiological range, could provide protective effects in ischemic cardiomyopathy.


Circulation-heart Failure | 2015

Cardiomyocyte ADAM17 Is Essential in Post-Myocardial Infarction Repair by Regulating Angiogenesis

Dong Fan; Abhijit Takawale; Mengcheng Shen; Wang Wang; Xiuhua Wang; Ratnadeep Basu; Gavin Y. Oudit; Zamaneh Kassiri

Background—A disintegrin and metalloproteinase 17 (ADAM17) is a membrane-bound enzyme that mediates shedding of many membrane-bound molecules, thereby regulating multiple cellular responses. We investigated the role of cardiomyocyte ADAM17 in myocardial infarction (MI). Methods and Results—Cardiomyocyte-specific ADAM17 knockdown mice (ADAM17flox/flox/&agr;-MHC-Cre; f/f/Cre) and parallel controls (ADAM17flox/flox; f/f) were subjected to MI by ligation of the left anterior descending artery. Post MI, f/f/Cre mice showed compromised survival, higher rates of cardiac rupture, more severe left ventricular dilation, and suppressed ejection fraction compared with parallel f/f-MI mice. Ex vivo ischemic injury (isolated hearts) resulted in comparable recovery in both genotypes. Myocardial vascular density (fluorescent-labeled lectin perfusion and CD31 immunofluorescence staining) was significantly lower in the infarct areas of f/f/Cre-MI compared with f/f-MI mice. Activation of vascular endothelial growth factor receptor 2 (VEGFR2), its mRNA, and total protein levels were reduced in infarcted myocardium in ADAM17 knockdown mice. Transcriptional regulation of VEGFR2 by ADAM17 was confirmed in cocultured cardiomyocyte–fibroblast as ischemia-induced VEGFR2 expression was blocked by ADAM17-siRNA. Meanwhile, ADAM17-siRNA did not alter VEGFA bioavailability in the conditioned media. ADAM17 knockdown mice (f/f/Cre-MI) exhibited reduced nuclear factor-&kgr;B activation (DNA binding) in the infarcted myocardium, which could underlie the suppressed VEGFR2 expression in these hearts. Post MI, inflammatory response was not altered by ADAM17 downregulation. Conclusions—This study highlights the key role of cardiomyocyte ADAM17 in post-MI recovery by regulating VEGFR2 transcription and angiogenesis, thereby limiting left ventricular dilation and dysfunction. Therefore, ADAM17 upregulation, within the physiological range, could provide protective effects in ischemic cardiomyopathy.


Journal of Molecular and Cellular Cardiology | 2017

Loss of smooth muscle cell disintegrin and metalloproteinase 17 transiently suppresses angiotensin II-induced hypertension and end-organ damage

Mengcheng Shen; Jude S. Morton; Sandra T. Davidge; Zamaneh Kassiri

Hypertension is associated with hypertrophy and hyperplasia of smooth muscle cells (SMCs). Disintegrin and metalloproteinase 17 (ADAM17) is a membrane-bound enzyme reported to mediate SMC hypertrophy through activation of epidermal growth factor receptor (EGFR). We investigated the role of ADAM17 in Ang II-induced hypertension and end-organ damage. VSMC was isolated from mice with intact ADAM17 expression (Adam17f/f) or lacking ADAM17 in the SMC (Adam17f/f/CreSm22). Human VSMCs were isolated from the aorta of donors, and ADAM17 deletion was achieved by siRNA transfection. Ang II suppressed proliferation and migration of Adam17-deficient SMCs, but did not affect apoptosis (mouse and human), this was associated with reduced activation of EGFR and Erk1/2 signaling. Adam17f/f/CreSm22 and littermate Adam17f/f mice received saline or Ang II (Alzet pumps, 1.5mg/kg/d; 2 or 4weeks). Daily blood pressure measurement in conscious mice (telemetry) showed suppressed hypertension in Adam17f/f/CreSm22 mice during the first week of Ang II infusion, but by the second week, it become comparable to that in Adam17f/f mice. EGFR activation remained suppressed in Adam17f/f/CreSm22-Ang II arteries. Ex vivo vascular function and compliance assessed in mesenteric arteries were comparable between genotypes. Consistent with the transient protection against Ang II-induced hypertension, Adam17f/f/CreSm22 mice exhibited significantly lower cardiac hypertrophy and fibrosis, and renal fibrosis at 2weeks post-Ang II, however this protection was abolished by the fourth week of Ang II infusion. In conclusion, while Adam17-deficiency suppresses Ang II-induced SMC remodeling in vitro, in vivo Adam17-deficiency provides only a transient protective effect against Ang II-mediated hypertension and end-organ damage.


American Journal of Physiology-heart and Circulatory Physiology | 2018

TIMP3 deficiency exacerbates iron-overload mediated cardiomyopathy and liver disease

Pavel Zhabyeyev; Subhash K. Das; Ratnadeep Basu; Mengcheng Shen; Vaibhav B. Patel; Zamaneh Kassiri; Gavin Y. Oudit

Chronic iron overload results in heart and liver diseases and is a common cause of morbidity and mortality in patients with genetic hemochromatosis and secondary iron overload. We investigated the role of tissue inhibitor of metalloproteinase 3 (TIMP3) in iron overload-mediated tissue injury by subjecting male mice lacking Timp3 ( Timp3-/-) and wild-type (WT) mice to 12 wk of chronic iron overload. Whereas WT mice with iron overload developed diastolic dysfunction, iron-overloaded Timp3-/- mice showed worsened cardiac dysfunction coupled with systolic dysfunction. In the heart, loss of Timp3 was associated with increased myocardial fibrosis, greater Timp1, matrix metalloproteinase ( Mmp) 2, and Mmp9 expression, increased active MMP-2 levels, and gelatinase activity. Iron overload in Timp3-/- mice showed twofold higher iron accumulation in the liver compared with WT mice because of constituently lower levels of ferroportin. Loss of Timp3 enhanced the hepatic inflammatory response to iron overload, leading to greater neutrophil and macrophage infiltration and increased hepatic fibrosis. Expression of inflammation-related MMPs (MMP-12 and MMP-13) and inflammatory cytokines (IL-1β and monocyte chemoattractant protein-1) was elevated to a greater extent in iron-overloaded Timp3-/- livers. Gelatin zymography demonstrated equivalent increases in MMP-2 and MMP-9 levels in WT and Timp3-/- iron-overloaded livers. Loss of Timp3 enhanced the susceptibility to iron overload-mediated heart and liver injury, suggesting that Timp3 is a key protective molecule against iron-mediated pathology. NEW & NOTEWORTHY In mice, loss of tissue inhibitor of metalloproteinase 3 ( Timp3) was associated with systolic and diastolic dysfunctions, twofold higher hepatic iron accumulation (attributable to constituently lower levels of ferroportin), and increased hepatic inflammation. Loss of Timp3 enhanced the susceptibility to iron overload-mediated injury, suggesting that Timp3 plays a key protective role against iron-mediated pathology.


Circulation Research | 2018

Cell-Specific Functions of ADAM17 Regulate the Progression of Thoracic Aortic Aneurysm

Mengcheng Shen; Mei Hu; Paul W.M. Fedak; Gavin Y. Oudit; Zamaneh Kassiri

Rationale: ADAM17 (a disintegrin and metalloproteinase-17) is a membrane-bound enzyme that regulates bioavailability of multiple transmembrane proteins by proteolytic processing. ADAM17 has been linked to several pathologies, but its role in thoracic aortic aneurysm (TAA) has not been determined. Objective: The objective of this study was to explore the cell-specific functions of vascular ADAM17 in the pathogenesis and progression of TAA. Methods and Results: In aneurysmal thoracic aorta from patients, ADAM17 was increased in tunica media and intima. To determine the function of ADAM17 in the major cells types within these regions, we generated mice lacking ADAM17 in smooth muscle cells (SMC; Adam17 f/f /Sm22 Cre/+ ) or endothelial cells (Adam17 f/f /Tie2 Cre/+ ). ADAM17 deficiency in either cell type was sufficient to suppress TAA dilation markedly and adverse remodeling in males and females (in vivo) although through different mechanisms. ADAM17 deficiency in SMCs prevented the contractile-to-synthetic phenotypic switching in these cells after TAA induction, preventing perivascular fibrosis, inflammation, and adverse aortic remodeling. Loss of ADAM17 in endothelial cells protected the integrity of the intimal barrier by preserving the adherens junction (vascular endothelial-cadherin) and tight junctions (junctional adhesion molecule-A and claudin). In vitro studies on primary mouse thoracic SMCs and human primary aortic SMCs and endothelial cells (±ADAM17 small interfering RNA) confirmed the cell-specific functions of ADAM17 and demonstrated the cross-species validity of these findings. To determine the impact of ADAM17 inhibition in treating TAA, we used an ADAM17-selective inhibitor (PF-548) before or 3 days after TAA induction. In both cases, ADAM17 inhibition prevented progression of aneurysmal growth. Conclusions: We have identified distinct cell-specific functions of ADAM17 in TAA progression, promoting pathological remodeling of SMC and impairing integrity of the intimal endothelial cell barrier. The dual impact of ADAM17 deficiency (or inhibition) in protecting 2 major cell types in the aortic wall highlights the unique position of this proteinase as a critical treatment target for TAA.


Archive | 2015

Remodelling of the Cardiac Extracellular Matrix: Role of Collagen Degradation and Accumulation in Pathogenesis of Heart Failure

Abhijit Takawale; Mengcheng Shen; Dong Fan; Zamaneh Kassiri

The extracellular matrix (ECM) serves a number of functions in every tissue including the myocardium. While the function of ECM as a structural scaffold is well established, it has become increasingly recognized that it plays a number of additional functions including providing a reservoir for growth factors and cytokines allowing their rapid release and activation in response to environmental cues. In addition, components of the ECM are critical in the interstitial transport of numerous molecules and drugs. Therefore, impaired integrity of the ECM would influence multiple aspects of an organ’s structural and function. In the myocardium, the primary component of the ECM network structure is the fibrillar collagens I and III. Multiple steps and various enzymes are involved from collagen mRNA synthesis to collagen fibre formation. Alterations in each step can impact collagen fibre production resulting in an uncoupling between collagen mRNA and protein levels. In this chapter, we will provide an overview of the mechanisms involved in myocardial fibrosis, the disease-dependent nature and consequence of different types of fibrosis, clinical biomarkers of collagen turnover, and potential therapeutic approaches in managing myocardial fibrosis.

Collaboration


Dive into the Mengcheng Shen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dong Fan

University of Alberta

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wang Wang

University of Alberta

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge