Network


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

Hotspot


Dive into the research topics where Congye Li is active.

Publication


Featured researches published by Congye Li.


Bone | 2009

Du-Zhong (Eucommia ulmoides Oliv.) cortex extract prevent OVX-induced osteoporosis in rats

Rui Zhang; Zhen-Guo Liu; Congye Li; Sijun Hu; Linna Liu; Jun Wang; Qi-Bing Mei

Du-Zhong, rich in polyphenolic compounds such as lignans, phenolic acid, and flavonoids, is a kidney-tonifying herbal medicine with a long history of safe use for treatment of bone fractures and joint diseases in China. In the present study, we examined whether Du-Zhong cortex extract (DZCE) with graded doses exerted its preventive effects on estrogen deficiency-induced osteoporosis. Eighty 3-month-old female Sprague-Dawley rats were used and randomly assigned into sham-operated group (Sham) and five ovariectomy (OVX) subgroups, i.e. OVX with vehicle (OVX); OVX with 17alpha-ethinylestradiol (E(2), 25 microg/kg/day); OVX with DZCE of graded doses (100, 300, or 500 mg/kg/day). Daily oral administration of DZCE or E(2) started on week 4 after OVX for 16 weeks. Treatment with DZCE at higher doses (300 or 500 mg/kg/day) was found to be able to significantly prevent OVX-induced decrease in biomechanical quality of femur such as maximum stress and Youngs modulus. The mechanical changes were associated with the prevention of a further bone mineral density (BMD) decrease or even with some improvements in microarchitecture. DZCE dose-dependently inhibited total BMD decrease in the femur caused by OVX, which was accompanied by a significant decrease in skeletal remodeling, as was evidenced by the decreased levels of the bone turnover markers osteocalcin (OC), alkaline phosphatese (ALP), deoxypyridinoline (DPD), and urinary Ca and P excretions. muCT analysis of the femoral metaphysis showed that DZCE at the highest doses (500 mg/kg/day) significantly prevents decrease in bone volume/tissue volume (BV/TV), connect density (Conn.D), trabecula number (Tb.N) and trabecula thickness (Tb.Th), and increase in trabecula separation (Tb.Sp) and structure model index (SMI) in OVX rats. We conclude that 16 weeks of DZCE treatment improves bone biomechanical quality through modifications of BMD, and trabecular microarchitecture without hyperplastic effect on uterus, and it might be a potential alternative medicine for treatment of postmenopausal osteoporosis.


Optics Express | 2010

Experimental Cerenkov luminescence tomography of the mouse model with SPECT imaging validation

Zhenhua Hu; Jimin Liang; Weidong Yang; Weiwei Fan; Congye Li; Xiaowei Ma; Xueli Chen; Xiaopeng Ma; Xiangsi Li; Xiaochao Qu; Jing Wang; Feng Cao; Jie Tian

Optical molecular imaging resulting from Cerenkov radiation has become a motivating topic recently and will potentially open new avenues for the study of small animal imaging. Cerenkov-based optical imaging taken from living animals in vivo has been studied with two-dimensional (2D) planar geometry and three-dimensional (3D) homogeneous mouse model. In this study, we performed 3D Cerenkov-based luminescence tomography (CLT) using a heterogeneous mouse model with an implanted Na(131)I radioactive source, which provided the accurate location for the reconstructed source. Furthermore, single photon emission computed tomography (SPECT) was utilized to verify the results of 3D CLT. We reconstructed the localization and intensity of an embedded radioactive source with various concentrations, and established a quantitative relationship between the radiotracer activity and the reconstructed intensity. The results showed the ability of in vivo CLT to recover the radioactive probe distribution in the heterogeneous mouse model and the potential of a SPECT imaging validation strategy to verify the results of optical molecular tomography.


Biochimica et Biophysica Acta | 2015

A novel protective mechanism for mitochondrial aldehyde dehydrogenase (ALDH2) in type i diabetes-induced cardiac dysfunction: role of AMPK-regulated autophagy.

Yuli Guo; Wenjun Yu; Dongdong Sun; Jiaxing Wang; Congye Li; Rongqing Zhang; Sara A. Babcock; Yan Li; Min Liu; Meijuan Ma; Mingzhi Shen; Chao Zeng; Na Li; Wei He; Qian Zou; Yingmei Zhang; Haichang Wang

Mitochondrial aldehyde dehydrogenase (ALDH2) is known to offer myocardial protection against stress conditions including ischemia-reperfusion injury, alcoholism and diabetes mellitus although the precise mechanism is unclear. This study was designed to evaluate the effect of ALDH2 on diabetes-induced myocardial injury with a focus on autophagy. Wild-type FVB and ALDH2 transgenic mice were challenged with streptozotozin (STZ, 200mg/kg, i.p.) for 3months to induce experimental diabetic cardiomyopathy. Diabetes triggered cardiac remodeling and contractile dysfunction as evidenced by cardiac hypertrophy, decreased cell shortening and prolonged relengthening duration, the effects of which were mitigated by ALDH2. Lectin staining displayed that diabetes promoted cardiac hypertrophy, the effect of which was alleviated by ALDH2. Western blot analysis revealed dampened autophagy protein markers including LC3B ratio and Atg7 along with upregulated p62 following experimental diabetes, the effect of which was reconciled by ALDH2. Phosphorylation level of AMPK was decreased and its downstream signaling molecule FOXO3a was upregulated in both diabetic cardiac tissue and in H9C2 cells with high glucose exposure. All these effect were partly abolished by ALDH2 overexpression and ALDH2 agonist Alda1. High glucose challenge dampened autophagy in H9C2 cells as evidenced by enhanced p62 levels and decreased levels of Atg7 and LC3B, the effect of which was alleviated by the ALDH2 activator Alda-1. High glucose-induced cell death and apoptosis were reversed by Alda-1. The autophagy inhibitor 3-MA and the AMPK inhibitor compound C mitigated Alda-1-offered beneficial effect whereas the autophagy inducer rapamycin mimicked or exacerbated high glucose-induced cell injury. Moreover, compound C nullified Alda-1-induced protection against STZ-induced changes in autophagy and function. Our results suggested that ALDH2 protects against diabetes-induced myocardial dysfunction possibly through an AMPK -dependent regulation of autophagy. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.


Journal of Pineal Research | 2016

Melatonin facilitates adipose‐derived mesenchymal stem cells to repair the murine infarcted heart via the SIRT1 signaling pathway

Dong Han; Huang W; Xiujuan Li; Lei Gao; Tao Su; Sai Ma; Liu T; Congye Li; Chen J; Erhe Gao; Feng Cao

Mesenchymal stem cells (MSCs)‐based therapy provides a promising therapy for the ischemic heart disease (IHD). However, engrafted MSCs are subjected to acute cell death in the ischemic microenvironment, characterized by excessive inflammation and oxidative stress in the hosts infarcted myocardium. Melatonin, an indole, which is produced by many organs including pineal gland, has been shown to protect bone marrow MSCs against apoptosis although the mechanism of action remains elusive. Using a murine model of myocardial infarction (MI), this study was designed to evaluate the impact of melatonin on adipose‐derived mesenchymal stem cells (AD‐MSCs)‐based therapy for MI and the underlying mechanism involved with a focus on silent information regulator 1(SIRT1) signaling. Our results demonstrated that melatonin promoted functional survival of AD‐MSCs in infarcted heart and provoked a synergetic effect with AD‐MSCs to restore heart function. This in vivo effect of melatonin was associated with alleviated inflammation, apoptosis, and oxidative stress in infarcted heart. In vitro studies revealed that melatonin exert cytoprotective effects on AD‐MSCs against hypoxia/serum deprivation (H/SD) injury via attenuating inflammation, apoptosis, and oxidative stress. Mechanistically, melatonin enhanced SIRT1 signaling, which was accompanied with the increased expression of anti‐apoptotic protein Bcl2, and decreased the expression of Ac‐FoxO1, Ac‐p53, Ac‐NF‐ΚB, and Bax. Taken together, our findings indicated that melatonin facilitated AD‐MSCs‐based therapy in MI, possibly through promoting survival of AD‐MSCs via SIRT1 signaling. Our data support the promise of melatonin as a novel strategy to improve MSC‐based therapy for IHD, possibly through SIRT1 signaling evocation.


Biochimica et Biophysica Acta | 2017

Sirt3 deficiency exacerbates diabetic cardiac dysfunction: Role of Foxo3A-Parkin-mediated mitophagy

Wenjun Yu; Beilei Gao; Na Li; Jiaxing Wang; Cuiting Qiu; Guoyong Zhang; Min Liu; Rongqing Zhang; Congye Li; Gang Ji; Yingmei Zhang

Diabetic cardiomyopathy (DCM) is often associated with suppressed cardiac autophagy, mitochondrial structural and functional impairment. Sirtuin-3 (Sirt3) has been reported to play a crucial role in mitochondrial homeostasis and confers a protective role against the onset and development of DCM although the precise mechanism(s) remains elusive. Here we hypothesized that Sirt3 exerts cardioprotection against DCM by activating Parkin-mediated mitophagy, en route to preserved mitochondrial homeostasis and suppressed cardiomyocyte apoptosis. Adult male wild-type (WT) and Sirt3 knockout (Sirt3KO) mice were treated with streptozotocin (STZ) or vehicle for 3months prior to assessment of echocardiographic property, interstitial fibrosis, cardiomyocyte apoptosis, mitochondrial morphology, cardiac autophagy and cell signaling molecules. Our findings revealed that STZ-induced diabetes mellitus prompted cardiac dysfunction, interstitial fibrosis, cardiomyocyte apoptosis and mitochondrial injury, accompanied with suppressed autophagy and mitophagy, the effects of which were aggravated by Sirt3KO. To the contrary, Sirt3 overexpression in vitro activated autophagy and mitophagy, inhibited mitochondrial injury and cardiomyocyte apoptosis, the effects of which were attenuated by autophagy inhibition using 3-MA. Moreover, deacetylation of Foxo3A and expression of Parkin were decreased by Sirt3KO, while these effects were facilitated by Sirt3OE in diabetic and high glucose settings. Taken together, our data suggested that suppressed Sirt3-Foxo3A-Parkin signaling mediated downregulation of mitophagy may play a vital role in the development of diabetic cardiomyopathy. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure edited by Dr. Jun Ren & Yingmei Zhang.


Diabetologia | 2016

MST1 coordinately regulates autophagy and apoptosis in diabetic cardiomyopathy in mice

Mingming Zhang; Lei Zhang; Jianqiang Hu; Jie Lin; Tingting Wang; Yu Duan; Wanrong Man; Jiaxu Feng; Lei Sun; Hongbing Jia; Congye Li; Rongqing Zhang; Haichang Wang; Dongdong Sun

Aims/hypothesisDiabetic cardiomyopathy (DCM) is associated with suppressed autophagy and augmented apoptosis in the heart although the interplay between the two remains elusive. The ability of mammalian sterile 20-like kinase 1 to regulate both autophagy and apoptosis prompted us to investigate it as a possible candidate in the progression of DCM.MethodsWild-type, Mst1 (also known as Stk4) transgenic and Mst1-knockout mice were challenged with streptozotocin to induce experimental diabetes. In addition, cultured neonatal mouse cardiomyocytes were subjected to simulated diabetes to probe mechanisms.ResultsMst1 knockout alleviated while Mst1 overexpression aggravated cardiac dysfunction in diabetes. Diabetic Mst1 transgenic mice exhibited decreased LC3 expression and enhanced protein aggregation. In contrast, typical autophagosomes were observed in diabetic Mst1-knockout mice with increased LC3 expression and reduced protein aggregation. Mst1 downregulation promoted autophagic flux as demonstrated by increased LC3-II and decreased p62 expression in the presence of bafilomycin A1. Furthermore, Mst1 overexpression increased, while Mst1 knockout decreased, cardiomyocyte apoptosis both in vivo and in vitro. Co-immunoprecipitation assays showed that Mst1 overexpression promoted Beclin1 binding to B cell lymphoma 2 (Bcl-2) and induced dissociation of Bcl-2 from Bax in diabetic mice. Conversely, Mst1 knockout disrupted the Beclin1-Bcl-2 complex and enhanced the interaction between Bcl-2 and Bax.Conclusions/interpretationMst1 knockout restores autophagy and protects against apoptosis in cardiomyocytes, en route to the rescue against DCM.


Journal of Cellular and Molecular Medicine | 2016

Luteolin alleviates post-infarction cardiac dysfunction by up-regulating autophagy through Mst1 inhibition.

Jianqiang Hu; Wanrong Man; Min Shen; Mingming Zhang; Jie Lin; Tingting Wang; Yu Duan; Congye Li; Rongqing Zhang; Erhe Gao; Haichang Wang; Dongdong Sun

Myocardial infarction (MI), which is characterized by chamber dilation and LV dysfunction, is associated with substantially higher mortality. We investigated the effects and underlying mechanisms of Luteolin on post‐infarction cardiac dysfunction. Myocardial infarction was constructed by left anterior descending coronary artery ligation. In vitro, cultured neonatal cardiomyocytes subjected to simulated MI were used to probe mechanism. Luteolin significantly improved cardiac function, decreased cardiac enzyme and inflammatory cytokines release after MI. Enhanced autophagic flux as indicated by more autophagosomes puncta, less accumulation of aggresomes and P62 in the neonatal cardiomyocytes after hypoxia was observed in the Luteolin pre‐treatment group. Western blot analysis also demonstrated that Luteolin up‐regulated autophagy in the cardiomyocytes subjected to simulated MI injury. Furthermore, Luteolin increased mitochondrial membrane potential, adenosine triphosphate content, citrate synthase activity and complexes I/II/III/IV/V activities in the cardiomyocytes subjected to simulated MI injury. Interestingly, mammalian sterile 20‐like kinase 1 (Mst1) knockout abolished the protective effects of Luteolin administration. Luteolin enhances cardiac function, reduces cardiac enzyme and inflammatory markers release after MI. The protective effects of Luteolin are associated with up‐regulation of autophagy and improvement of mitochondrial biogenesis through Mst1 inhibition.


Stem Cell Research & Therapy | 2014

Inositol pyrophosphates mediate the effects of aging on bone marrow mesenchymal stem cells by inhibiting Akt signaling

Zheng Zhang; Chuanxu Zhao; Bing Liu; Dong Liang; Xing Qin; Xiujuan Li; Rongqing Zhang; Congye Li; Haichang Wang; Dongdong Sun; Feng Cao

IntroductionBone marrow-derived mesenchymal stem cells (BM-MSCs) have been proposed as an ideal autologous stem cell source for cell-based therapy for myocardial infarction (MI). However, decreased viability and impaired function of aged MSCs hampered the therapeutic efficacy of engrafted MSCs, and the underlying mechanisms remain unclarified. Here, we investigated the role of inositol phosphates 6 kinase (IP6Ks) inhibition on the therapeutic efficacy of BM-MSCs and its underlying mechanism.MethodsBM-MSCs isolated from young (8-week-old) or aged (18-month-old) donor male C57BL/6 mice, were subjected to hypoxia and serum deprivation (H/SD) injury with or without administration of inositol phosphates 6 kinase (IP6Ks) inhibitor TNP (10 μM). MSC apoptosis induced by H/SD was determined by flow cytometry and TUNEL assays. Protein expressions were evaluated by Western blot assay. Furthermore, the paracrine effects of MSCs were measured by reverse transcriptase–polymerized chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) analyses.ResultsAged BM-MSCs exhibited more Inositol pyrophosphate 7 (IP7) production, compared with young BM-MSCs. Meanwhile, the expression of phospho-Akt (Thr308) was significantly decreased in the aged MSCs, resulting in enhanced Bad activation and decreased Bax/Bcl-2 ratio. Moreover, the apoptosis in aged BM-MSCs was increased, compared with young BM-MSCs. Furthermore, TNP administration significantly inhibited IP7 production and increased the phosphorylation of Akt under both normoxic and hypoxic conditions. Meanwhile, IP6Ks inhibition reduced apoptotic index of aged MSCs, associated with decreased expressions of pro-apoptotic proteins Bax and Bad and increased anti-apoptotic protein Bcl-2. The expressions of angiogenic factors, including VEGF, bFGF, IGF-1 and HGF, were decreased in MSCs from aged mice. In addition, TNP administration enhanced the paracrine efficiency of aged BM-MSCs under normoxic and hypoxic conditions.ConclusionsThis study demonstrates for the first time that IP6Ks and IP7 play critical role in the aging related vulnerability to hypoxic injury and impaired paracrine efficiency of BM-MSCs, which is associated with impaired Akt activation.


Journal of Cellular and Molecular Medicine | 2015

Lin28a protects against cardiac ischaemia/reperfusion injury in diabetic mice through the insulin‐PI3K‐mTOR pathway

Mingming Zhang; Dongdong Sun; Shuang Li; Xietian Pan; Xiaotian Zhang; Di Zhu; Congye Li; Rongqing Zhang; Erhe Gao; Haichang Wang

The insulin‐PI3K‐mTOR pathway exhibits a variety of cardiovascular activities including protection against I/R injury. Lin28a enhanced glucose uptake and insulin‐sensitivity via insulin‐PI3K‐mTOR signalling pathway. However, the role of lin28a on experimental cardiac I/R injury in diabetic mice are not well understood. Diabetic mice underwent 30 min. of ischaemia followed by 3 hrs of reperfusion. Animals were randomized to be treated with lentivirus carrying lin28a siRNA (siLin28a) or lin28a cDNA (Lin28a) 72 hrs before coronary artery ligation. Myocardial infarct size (IS), cardiac function, cardiomyocyte apoptosis and mitochondria morphology in diabetic mice who underwent cardiac I/R injury were compared between groups. The target proteins of lin28a were examined by western blot analysis. Lin28a overexpression significantly reduced myocardial IS, improved LV ejection fraction (LVEF), decreased myocardial apoptotic index and alleviated mitochondria cristae destruction in diabetic mice underwent cardiac I/R injury. Lin28a knockdown exacerbated cardiac I/R injury as demonstrated by increased IS, decreased LVEF, increased apoptotic index and aggravated mitochondria cristae destruction. Interestingly, pre‐treatment with rapamycin abolished the beneficial effects of lin28a overexpression. Lin28a overexpression increased, while Lin28a knockdown decreased the expression of IGF1R, p‐Akt, p‐mTOR and p‐p70s6k after cardiac I/R injury in diabetic mice. Rapamycin pre‐treatment abolished the effects of increased p‐mTOR and p‐p70s6k expression exerted by lin28a overexpression. This study indicates that lin28a overexpression reduces IS, improves cardiac function, decreases cardiomyocyte apoptosis index and alleviates cardiomyocyte mitochondria impairment after cardiac I/R injury in diabetic mice. The mechanism responsible for the effects of lin28a is associated with the insulin‐PI3K‐mTOR dependent pathway.


Scientific Reports | 2016

Mst1 inhibits CMECs autophagy and participates in the development of diabetic coronary microvascular dysfunction

Jie Lin; Lei Zhang; Mingming Zhang; Jianqiang Hu; Tingting Wang; Yu Duan; Wanrong Man; Bin Wu; Jiaxu Feng; Lei Sun; Congye Li; Rongqing Zhang; Haichang Wang; Dongdong Sun

Cardiovascular complications account for a substantial proportion of morbidity and mortality in diabetic patients. Abnormalities of cardiac microvascular endothelial cells (CMECs) lead to impaired cardiac microvascular vessel integrity and subsequent cardiac dysfunction, underlining the importance of coronary microvascular dysfunction. In this study, experimental diabetes models were constructed using Mst1 transgenic, Mst1 knockout and sirt1 knockout mice. Diabetic Mst1 transgenic mice exhibited impaired cardiac microvessel integrity and decreased cardiac function. Mst1 overexpression deceased CMECs autophagy as evidenced by decreased LC3 expression and enhanced protein aggregation when subjected to high glucose culture. Mst1 knockout improved cardiac microvessel integrity and enhanced cardiac functions in diabetic mice. Mst1 knockdown up-regulated autophagy as indicated by more typical autophagosomes and increased LC3 expression in CMECs subjected to high glucose cultures. Mst1 knockdown also promoted autophagic flux in the presence of bafilomycin A1. Mst1 overexpression increased CMECs apoptosis, whereas Mst1 knockout decreased CMECs apoptosis. Sirt1 knockout abolished the effects of Mst1 overexpression in cardiac microvascular injury and cardiac dysfunction. In conclusion, Mst1 knockout preserved cardiac microvessel integrity and improved cardiac functions in diabetic mice. Mst1 decreased sirt1 activity, inhibited autophagy and enhanced apoptosis in CMECs, thus participating in the pathogenesis of diabetic coronary microvascular dysfunction.

Collaboration


Dive into the Congye Li's collaboration.

Top Co-Authors

Avatar

Feng Cao

Chinese PLA General Hospital

View shared research outputs
Top Co-Authors

Avatar

Rongqing Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Haichang Wang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

T. Gao

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiujuan Li

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Dong Han

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Dongdong Sun

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Mingming Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Sai Ma

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ling Tao

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge