Network


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

Hotspot


Dive into the research topics where Xinyu Zhou is active.

Publication


Featured researches published by Xinyu Zhou.


Acta Pharmacologica Sinica | 2015

Ginsenoside Rd promotes neurogenesis in rat brain after transient focal cerebral ischemia via activation of PI3K/Akt pathway

Xin-yu Liu; Xinyu Zhou; Jincai Hou; Hua Zhu; Zhong Wang; Jianxun Liu; Yongqiu Zheng

Aim:To investigate the effects of ginsenoside Rd (Rd) on neurogenesis in rat brain after ischemia/reperfusion injury (IRI).Methods:Male SD rats were subjected to transient middle cerebral artery occlusion (MCAO) followed by reperfusion. The rats were injected with Rd (1, 2.5, and 5 mg·kg−1·d−1, ip) from d 1 to d 3 after MCAO, and with BrdU (50 mg·kg−1·d−1, ip) from d 3 to d 6, then sacrificed on 7 d. The infarct size and neurological scores were assessed. Neurogenesis in the brains was detected by BrdU, DCX, Nestin, and GFAP immunohistochemistry staining. PC12 cells subjected to OGD/reperfusion were used as an in vitro model of brain ischemia. VEGF and BDNF levels were assessed with ELISA, and Akt and ERK phosphorylation was measured using Western blotting.Results:Rd administration dose-dependently decreased the infarct size and neurological scores in the rats with IRI. The high dose of Rd 5 (mg·kg−1·d−1) significantly increased Akt phosphorylation in ipsilateral hemisphere, and markedly increased the number of BrdU/DCX and Nestin/GFAP double-positive cells in ischemic area, which was partially blocked by co-administration of the PI3 kinase inhibitor LY294002. Treatment with Rd (25, 50, and 100 μmol/L) during reperfusion significantly increased the expression of VEGF and BDNF in PC12 cells with IRI. Furthermore, treatment with Rd dose-dependently increased the phosphorylation of Akt and ERK, and significantly decreased PC12 cell apoptosis, which were blocked by co-application of LY294002.Conclusion:Rd not only attenuates ischemia/reperfusion injury in rat brain, but also promotes neurogenesis via increasing VEGF and BDNF expression and activating the PI3K/Akt and ERK1/2 pathways.


Nature Communications | 2014

Treatment of acute lung injury by targeting MG53-mediated cell membrane repair

Yanlin Jia; Ken Chen; Peihui Lin; Gissela Lieber; Miyuki Nishi; Rosalie Yan; Zhen Wang; Yonggang Yao; Yu Li; Bryan A. Whitson; Pu Duann; Haichang Li; Xinyu Zhou; H. Zhu; Hiroshi Takeshima; John C. Hunter; Robbie L. McLeod; Noah Weisleder; Chunyu Zeng; Jianjie Ma

Injury to lung epithelial cells has a role in multiple lung diseases. We previously identified mitsugumin 53 (MG53) as a component of the cell membrane repair machinery in striated muscle cells. Here we show that MG53 also has a physiological role in the lung and may be used as a treatment in animal models of acute lung injury. Mice lacking MG53 show increased susceptibility to ischemia-reperfusion and over-ventilation induced injury to the lung when compared with wild type mice. Extracellular application of recombinant human MG53 (rhMG53) protein protects cultured lung epithelial cells against anoxia/reoxygenation-induced injuries. Intravenous delivery or inhalation of rhMG53 reduces symptoms in rodent models of acute lung injury and emphysema. Repetitive administration of rhMG53 improves pulmonary structure associated with chronic lung injury in mice. Our data indicate a physiological function for MG53 in the lung and suggest that targeting membrane repair may be an effective means for treatment or prevention of lung diseases.


Science Translational Medicine | 2015

MG53-mediated cell membrane repair protects against acute kidney injury

Pu Duann; Haichang Li; Peihui Lin; Tao Tan; Zhen Wang; Ken Chen; Xinyu Zhou; Kristyn Gumpper; H. Zhu; Thomas Ludwig; Peter J. Mohler; Brad H. Rovin; William T. Abraham; Chunyu Zeng; Jianjie Ma

Recombinant MG53 translocates to sites of injury in the proximal tubule of the kidney and protects mice from acute kidney injury induced by ischemia or drugs. A molecular bandage for kidney injury MG53 is a protein that is primarily expressed in muscles and helps protect muscle cells from damage. Now, Duann et al. have shown that MG53 performs a similar function in the kidney as well. The authors evaluated the role of MG53 in mouse models of kidney injury induced by ischemia and reperfusion, as well as by cisplatin, a highly nephrotoxic chemotherapy drug. In each case, recombinant MG53 could be given intravenously, and the authors found that it bound to the sites of injury on kidney cells and protected them from further damage and death. MG53 treatment did not interfere with the effectiveness of cisplatin against cancer cells, suggesting that MG53 may be useful for protecting patients’ kidneys during chemotherapy. Injury to the renal proximal tubular epithelium (PTE) represents the underlying consequence of acute kidney injury (AKI) after exposure to various stressors, including nephrotoxins and ischemia/reperfusion (I/R). Although the kidney has the ability to repair itself after mild injury, insufficient repair of PTE cells may trigger inflammatory and fibrotic responses, leading to chronic renal failure. We report that MG53, a member of the TRIM family of proteins, participates in repair of injured PTE cells and protects against the development of AKI. We show that MG53 translocates to acute injury sites on PTE cells and forms a repair patch. Ablation of MG53 leads to defective membrane repair. MG53-deficient mice develop pronounced tubulointerstitial injury and increased susceptibility to I/R-induced AKI compared to wild-type mice. Recombinant human MG53 (rhMG53) protein can target injury sites on PTE cells to facilitate repair after I/R injury or nephrotoxin exposure. Moreover, in animal studies, intravenous delivery of rhMG53 ameliorates cisplatin-induced AKI without affecting the tumor suppressor efficacy of cisplatin. These findings identify MG53 as a vital component of reno-protection, and targeting MG53-mediated repair of PTE cells represents a potential approach to prevention and treatment of AKI.


Circulation Research | 2014

Trimeric Intracellular Cation Channels and Sarcoplasmic/Endoplasmic Reticulum Calcium Homeostasis

Xinyu Zhou; Peihui Lin; Daiju Yamazaki; Ki Ho Park; Shinji Komazaki; S. R. Wayne Chen; Hiroshi Takeshima; Jianjie Ma

Trimeric intracellular cation channels (TRIC) represents a novel class of trimeric intracellular cation channels. Two TRIC isoforms have been identified in both the human and the mouse genomes: TRIC-A, a subtype predominantly expressed in the sarcoplasmic reticulum (SR) of muscle cells, and TRIC-B, a ubiquitous subtype expressed in the endoplasmic reticulum (ER) of all tissues. Genetic ablation of either TRIC-A or TRIC-B leads to compromised K+ permeation and Ca2+ release across the SR/ER membrane, supporting the hypothesis that TRIC channels provide a counter balancing K+ flux that reduces SR/ER membrane depolarization for maintenance of the electrochemical gradient that drives SR/ER Ca2+ release. TRIC-A and TRIC-B seem to have differential functions in Ca2+ signaling in excitable and nonexcitable cells. Tric-a−/− mice display defective Ca2+ sparks and spontaneous transient outward currents in arterial smooth muscle and develop hypertension, in addition to skeletal muscle dysfunction. Knockout of TRIC-B results in abnormal IP3 receptor–mediated Ca2+ release in airway epithelial cells, respiratory defects, and neonatal lethality. Double knockout mice lacking both TRIC-A and TRIC-B show embryonic lethality as a result of cardiac arrest. Such an aggravated lethality indicates that TRIC-A and TRIC-B share complementary physiological functions in Ca2+ signaling in embryonic cardiomyocytes. Tric-a−/− and Tric-b+/− mice are viable and susceptible to stress-induced heart failure. Recent evidence suggests that TRIC-A directly modulates the function of the cardiac ryanodine receptor 2 Ca2+ release channel, which in turn controls store-overload–induced Ca2+ release from the SR. Thus, the TRIC channels, in addition to providing a countercurrent for SR/ER Ca2+ release, may also function as accessory proteins that directly modulate the ryanodine receptor/IP3 receptor channel functions.


Scientific Reports | 2018

An Injectable Oxygen Release System to Augment Cell Survival and Promote Cardiac Repair Following Myocardial Infarction

Zhaobo Fan; Zhaobin Xu; Hong Niu; Ning Gao; Ya Guan; Chao Li; Yu Dang; Xiaoyu Cui; Xuanyou Liu Liu; Yunyan Duan; Haichang Li; Xinyu Zhou; Peihui Lin; Jianjie Ma; Jianjun Guan

Oxygen deficiency after myocardial infarction (MI) leads to massive cardiac cell death. Protection of cardiac cells and promotion of cardiac repair are key therapeutic goals. These goals may be achieved by re-introducing oxygen into the infarcted area. Yet current systemic oxygen delivery approaches cannot efficiently diffuse oxygen into the infarcted area that has extremely low blood flow. In this work, we developed a new oxygen delivery system that can be delivered specifically to the infarcted tissue, and continuously release oxygen to protect the cardiac cells. The system was based on a thermosensitive, injectable and fast gelation hydrogel, and oxygen releasing microspheres. The fast gelation hydrogel was used to increase microsphere retention in the heart tissue. The system was able to continuously release oxygen for 4 weeks. The released oxygen significantly increased survival of cardiac cells under the hypoxic condition (1% O2) mimicking that of the infarcted hearts. It also reduced myofibroblast formation under hypoxic condition (1% O2). After implanting into infarcted hearts for 4 weeks, the released oxygen significantly augmented cell survival, decreased macrophage density, reduced collagen deposition and myofibroblast density, and stimulated tissue angiogenesis, leading to a significant increase in cardiac function.


Biomacromolecules | 2017

Sustained Release of a Peptide-based Matrix Metalloproteinase-2 Inhibitor to Attenuate Adverse Cardiac Remodeling and Improve Cardiac Function Following Myocardial Infarction

Zhaobo Fan; Minghuan Fu; Zhaobin Xu; Bo Zhang; Zhihong Li; Haichang Li; Xinyu Zhou; Xuanyou Liu; Yunyan Duan; Peihui Lin; Pu Duann; Xiaoyun Xie; Jianjie Ma; Zhenguo Liu; Jianjun Guan

Following myocardial infarction (MI), degradation of extracellular matrix (ECM) by upregulated matrix metalloproteinases (MMPs) especially MMP-2 decreases tissue mechanical properties, leading to cardiac function deterioration. Attenuation of cardiac ECM degradation at the early stage of MI has the potential to preserve tissue mechanical properties, resulting in cardiac function increase. Yet the strategy for efficiently preventing cardiac ECM degradation remains to be established. Current preclinical approaches have shown limited efficacy because of low drug dosage allocated to the heart tissue, dose-limiting side effects, and cardiac fibrosis. To address these limitations, we have developed a MMP-2 inhibitor delivery system that can be specifically delivered into infarcted hearts at early stage of MI to efficiently prevent MMP-2-mediated ECM degradation. The system was based on an injectable, degradable, fast gelation, and thermosensitive hydrogel, and a MMP-2 specific inhibitor, peptide CTTHWGFTLC (CTT). The use of fast gelation hydrogel allowed to completely retain CTT in the heart tissue. The system was able to release low molecular weight CTT over 4 weeks possibly due to the strong hydrogen bonding between the hydrogel and CTT. The release kinetics was modulated by amount of CTT loaded into the hydrogel, and using chondroitin sulfate and heparin that can interact with CTT and the hydrogel. Both glycosaminoglycans augmented CTT release, while heparin more greatly accelerated the release. After it was injected into the infarcted hearts for 4 weeks, the released CTT efficiently prevented cardiac ECM degradation as it not only increased tissue thickness but also preserved collagen composition similar to that in the normal heart tissue. In addition, the delivery system significantly improved cardiac function. Importantly, the delivery system did not induce cardiac fibrosis. These results demonstrate that the developed MMP-2 inhibitor delivery system has potential to efficiently reduce adverse myocardial remodeling and improve cardiac function.


Oncotarget | 2016

MG53 permeates through blood-brain barrier to protect ischemic brain injury

Yonggang Yao; Bo Zhang; H. Zhu; Haichang Li; Yu Han; Ken Chen; Zhen Wang; Jing Zeng; Yukai Liu; Xinquan Wang; Yu Li; Duofen He; Peihui Lin; Xinyu Zhou; Ki Ho Park; Zehua Bian; Zhishui Chen; Nianqiao Gong; Tao Tan; Jingsong Zhou; Meng Zhang; Jianjie Ma; Chunyu Zeng

Ischemic injury to neurons represents the underlying cause of stroke to the brain. Our previous studies identified MG53 as an essential component of the cell membrane repair machinery. Here we show that the recombinant human (rh)MG53 protein facilitates repair of ischemia-reperfusion (IR) injury to the brain. MG53 rapidly moves to acute injury sites on neuronal cells to form a membrane repair patch. IR-induced brain injury increases permeability of the blood-brain-barrier, providing access of MG53 from blood circulation to target the injured brain tissues. Exogenous rhMG53 protein can protect cultured neurons against hypoxia/reoxygenation-induced damages. Transgenic mice with increased levels of MG53 in the bloodstream are resistant to IR-induced brain injury. Intravenous administration of rhMG53, either prior to or after ischemia, can effectively alleviate brain injuries in rats. rhMG53-mediated neuroprotection involves suppression of apoptotic neuronal cell death, as well as activation of the pro-survival RISK signaling pathway. Our data indicate a physiological function for MG53 in the brain and suggest that targeting membrane repair or RISK signaling may be an effective means to treat ischemic brain injury.


PLOS ONE | 2015

Effect of Metabolic Syndrome on Mitsugumin 53 Expression and Function

Hanley Ma; Jason Liu; Zehua Bian; Yuqi Cui; Xinyu Zhou; Xuefeng Zhou; Bo Zhang; T.M. Ayodele Adesanya; Frank Yi; Ki Ho Park; Tao Tan; Zhishui Chen; H. Zhu

Metabolic syndrome is a cluster of risk factors, such as obesity, insulin resistance, and hyperlipidemia that increases the individual’s likelihood of developing cardiovascular diseases. Patients inflicted with metabolic disorders also suffer from tissue repair defect. Mitsugumin 53 (MG53) is a protein essential to cellular membrane repair. It facilitates the nucleation of intracellular vesicles to sites of membrane disruption to create repair patches, contributing to the regenerative capacity of skeletal and cardiac muscle tissues upon injury. Since individuals suffering from metabolic syndrome possess tissue regeneration deficiency and MG53 plays a crucial role in restoring membrane integrity, we studied MG53 activity in mice models exhibiting metabolic disorders induced by a 6 month high-fat diet (HFD) feeding. Western blotting showed that MG53 expression is not altered within the skeletal and cardiac muscles of mice with metabolic syndrome. Rather, we found that MG53 levels in blood circulation were actually reduced. This data directly contradicts findings presented by Song et. al that indict MG53 as a causative factor for metabolic syndrome (Nature 494, 375-379). The diminished MG53 serum level observed may contribute to the inadequate tissue repair aptitude exhibited by diabetic patients. Furthermore, immunohistochemical analyses reveal that skeletal muscle fibers of mice with metabolic disorders experience localization of subcellular MG53 around mitochondria. This clustering may represent an adaptive response to oxidative stress resulting from HFD feeding and may implicate MG53 as a guardian to protect damaged mitochondria. Therapeutic approaches that elevate MG53 expression in serum circulation may be a novel method to treat the degenerative tissue repair function of diabetic patients.


Journal of Visualized Experiments | 2014

Assessment of Calcium Sparks in Intact Skeletal Muscle Fibers

Ki Ho Park; Noah Weisleder; Jingsong Zhou; Kristyn Gumpper; Xinyu Zhou; Pu Duann; Jianjie Ma; Peihui Lin


Biophysical Journal | 2014

The Therapeutic Role of Recombinant Human MG53 Protein in Wound Healing

Haichang Li; Pu Duann; Zhaobo Fan; Li Zhao; Peihui Lin; Mingzhai Sun; Gejing De; Xinyu Zhou; Jianjun Guan; Jianjie Ma

Collaboration


Dive into the Xinyu Zhou's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

H. Zhu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Tao Tan

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pu Duann

Ohio State University

View shared research outputs
Top Co-Authors

Avatar

Bo Zhang

Ohio State University

View shared research outputs
Top Co-Authors

Avatar

Jingsong Zhou

Rush University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge