Network


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

Hotspot


Dive into the research topics where Mingxing Wu is active.

Publication


Featured researches published by Mingxing Wu.


Current Molecular Medicine | 2014

Blockade of Jagged/Notch Pathway Abrogates Transforming Growth Factor β2-Induced Epithelial-Mesenchymal Transition in Human Retinal Pigment Epithelium Cells

X.-W. Chen; W. Xiao; X.-L. Liu; M. Zeng; L.-X. Luo; Mingxing Wu; S. Ye; Y. Liu

The epithelial-mesenchymal transition (EMT) of retinal pigment epithelium (RPE) cells plays a key role in proliferative vitreoretinopathy (PVR) and proliferative diabetic retinopathy (PDR), which lead to the loss of vision. The Jagged/Notch pathway has been reported to be essential in EMT during embryonic development, fibrotic diseases and cancer metastasis. However, the function of Jagged/Notch signaling in EMT of RPE cells is unknown. Thus, we hypothesized that a crosstalk between Notch and transforming growth factor β2 (TGF-β2) signaling could induce EMT in RPE cells, which subsequently contributes to PVR and PDR. Here, we demonstrate that Jagged-1/Notch pathway is involved in the TGF-β2-mediated EMT of human RPE cells. Blockade of Notch pathway with DAPT (a specific inhibitor of Notch receptor cleavage) and knockdown of Jagged-1 expression inhibited TGF-β2-induced EMT through regulating the expression of Snail, Slug and ZEB1. Besides the canonical Smad signaling pathway, the noncanonical PI3K/Akt and MAPK pathway also contributed to TGF-β2-induced up-regulation of Jagged-1 in RPE cells. Overexpression of Jagged-1 could mimic TGF-β2 induce EMT. Our data suggest that the Jagged-1/Notch signaling pathway plays a critical role in TGF-β2-induced EMT in human RPE cells, and may contribute to the development of PVR and PDR. Inhibition of the Jagged/Notch signaling pathway, therefore, may have therapeutic value in the prevention and treatment of PVR and PDR.


Current Molecular Medicine | 2012

The Tumor Suppressor p53 Regulates c-Maf and Prox-1 to Control Lens Differentiation

Fangyuan Liu; Xiangcheng Tang; Mi Deng; P. Chen; Weike Ji; X. Zhang; L. Gong; Zachary Woodward; Jiao Liu; L. Zhang; S. Sun; J. Liu; Kaili Wu; Mingxing Wu; X.-L. Liu; M.-B. Yu; Yizhi Liu; David Wan Cheng Li

The tumor suppressor p53 plays a key role in regulating apoptosis and cell cycle progression. In addition, p53 is implicated in control of cell differentiation in muscle, the circulatory system, ocular lens and various carcinoma tissues. However, the mechanisms by which p53 controls cell differentiation are not fully understood. Here we present evidence that p53 directly regulates c-Maf and Prox1, two important transcription factors controlling differentiation in the ocular lens. First, human and murine c-Maf and Prox1 gene promoters contain authentic p53 DNA binding sites. Second, p53 directly binds to the p53 binding sites found in the promoter regions. Third, exogenous p53 induces dose-dependent expression of the luciferase report gene driven by both c-Maf and Prox1 promoters, and p53 binds to both promoters in the ChIP assays. Fourth, in the in vitro differentiation model, knockdown of p53 significantly inhibits lens differentiation which is associated with downregulated expression of c-Maf and Prox1. Finally, in p53 knockout mice, the expression of c-Maf and Prox1 are significantly altered. Together, our results reveal that p53 regulates lens differentiation through modulation of two important transcription factors, c-Maf and Prox1, and through them p53 thus controls expression of various differentiation-related downstream crystallin genes.


International Journal of Ophthalmology | 2011

Objective evaluation of the changes in the crystalline lens during accommodation in young and presbyopic populations using Pentacam HR system.

Yao Ni; Xia-Lin Liu; Mingxing Wu; Ying Lin; Yuying Sun; Chang He; Yizhi Liu

AIM To quantify the changes in the lens profile with accommodation in different age groups. METHODS The Pentacam HR system was used to obtain the images of the anterior eye segment from 23 young and 15 presbyopic emmetropic subjects in unaccommodated (with an accommodation stimulus of 0.0D) and accommodated (with an accommodation stimulus of 5.0D for the young group and 1.0D for the presbyopic group) states. The phakic crystalline lens shape, including curvature of crystalline lens and central lens thickness (CLT), and the measurements of anterior segment length (ASL), central anterior chamber depth (CACD) were investigated. The anterior chamber volume (ACV) was also measured. RESULTS The reduction of CACD and ACV were significant in both groups after accommodation stimulus. From the profile of anterior eye segment, a significant decrease in anterior crystalline lens radii of curvature (-2.52mm) and a mean increase in CLT (0.222mm) and ASL (0.108mm) were found in the young group with an accommodation stimulus of 5.0D. However, no statistically significant changes of CLT, ASL, or crystalline lens radii of curvature were found in the presbyopic group. CONCLUSION Our data showed that the shallowing of anterior chamber during accommodation was caused by the forward bulging of the anterior lens surface, rather than by anterior shifting of lens position in either young or presbyopic subjects.


Current Molecular Medicine | 2013

p53 Directly Regulates αA- and βA3/A1-Crystallin Genes to Modulate Lens Differentiation

Weike Ji; Xiangcheng Tang; M. Yi; P.-Q. Chen; Fangyuan Liu; Xiao-Hui Hu; Weijun Hu; S.-J. Fu; J.-F. Liu; Kaili Wu; Mingxing Wu; X.-L. Liu; L.-X. Luo; S. Huang; Z.-Z. Liu; M.-B. Yu; Y. Liu; David Wan Cheng Li

It is well established that the tumor suppressor p53 plays major roles in regulating apoptosis and cell cycle progression. In addition, recent studies have demonstrated that p53 is actively involved in regulating cell differentiation in muscle, the circulatory system and various carcinoma tissues. We have recently shown that p53 also controls lens differentiation. Regarding the mechanism, we reveal that p53 directly regulates c-Maf and Prox1, two important transcription factors to control cell differentiation in the ocular lens. In the present study, we present further evidence to show that p53 can regulate lens differentiation by controlling expression of the differentiation genes coding for the lens crystallins. First, the αA and βA3/A1 gene promoters or introns all contain putative p53 binding sites. Second, gel mobility shifting assays revealed that the p53 protein in nuclear extracts from lens epithelial cells directly binds to the p53 binding sites found in these crystallin gene promoters or introns. Third, exogenous wild type p53 induces dose-dependent expression of the luciferase reporter gene driven by different crystallin gene promoters and the exogenous dominant negative mutant p53 causes dose-dependent inhibition of the same crystallin genes. Fourth, ChIP assays revealed that p53 binds to crystallin gene promoters in vivo. Finally, in the p53 knockout mouse lenses, expression levels of various crystallins were found down-regulated in comparison with those from the wild type mouse lenses. Together, our results reveal that p53 directly regulates expression of different sets of genes to control lens differentiation.


Current Molecular Medicine | 2016

Crosstalk Between the Autophagy-Lysosome Pathway and the Ubiquitin-Proteasome Pathway in Retinal Pigment Epithelial Cells.

J. Zhan; J. He; Yanhong Zhou; Mingxing Wu; Y. Liu; F. Shang; X. Zhang

BACKGROUND The accumulation of damaged or misfolded proteins in retinal pigment epithelial (RPE) cells was considered a contributing factor for RPE dysfunction in age-related macular degeneration (AMD). The ubiquitinproteasome pathway (UPP) and the autophagy-lysosome pathway (ALP) are the two major proteolytic systems for clearance of misfolded or damaged proteins. OBJECTIVE The aim is to investigate how these two systems communicate and coordinate with each other in RPE cells for eliminating intracellular misfolded and damaged proteins. METHODS Cultured ARPE-19 cells were treated with proteasome inhibitor MG132 and lysosomotropic agent chloroquine (CQ), respectively. The levels and cellular distributions of ubiquitinated proteins, LC3-I, LC3-II, LAMP1 and p62 were analyzed by Western blotting and immunofluorescence. Proteasome activity was determined using Suc-LLVY-AMC as a substrate. RESULTS The level of ubiquitinated protein aggregations was significantly increased after the treatment of MG132 in RPE cells. The levels of LC3-I, LC3-II and LAMP1 increased in MG132 treated cells. The levels of γ-tubulin and p62 also increased in MG132 treated cells, suggesting that inhibition of the UPP up-regulates autophagy-lysosome pathway. Inhibition of lysosomal activity with CQ also increased the levels of high mass ubiquitin conjugates, LC3-II and p62. In addition, proteasome activity was compromised upon prolonged lysosomal inhibition. CONCLUSIONS These data indicate that the UPP and the ALP are interrelated and that dysfunction of the ALP would also result in dysfunction of the UPP and severely compromise the capacity of eliminating misfolded and other forms of damaged proteins.


Cell Death & Differentiation | 2017

MicroRNA-26a and -26b inhibit lens fibrosis and cataract by negatively regulating Jagged-1/Notch signaling pathway

Xiaoyun Chen; Wei Xiao; Weirong Chen; Xialin Liu; Mingxing Wu; Qu Bo; Yan Luo; Shaobi Ye; Yihai Cao; Yizhi Liu

Fibrosis is a chronic process involving development and progression of multiple diseases in various organs and is responsible for almost half of all known deaths. Epithelial–mesenchymal transition (EMT) is the vital process in organ fibrosis. Lens is an elegant biological tool to investigate the fibrosis process because of its unique biological properties. Using gain- and loss-of-function assays, and different lens fibrosis models, here we demonstrated that microRNA (miR)-26a and miR-26b, members of the miR-26 family have key roles in EMT and fibrosis. They can significantly inhibit proliferation, migration, EMT of lens epithelial cells and lens fibrosis in vitro and in vivo. Interestingly, we revealed that the mechanisms of anti-EMT effects of miR-26a and -26b are via directly targeting Jagged-1 and suppressing Jagged-1/Notch signaling. Furthermore, we provided in vitro and in vivo evidence that Jagged-1/Notch signaling is activated in TGFβ2-stimulated EMT, and blockade of Notch signaling can reverse lens epithelial cells (LECs) EMT and lens fibrosis. Given the general involvement of EMT in most fibrotic diseases, cancer metastasis and recurrence, miR-26 family and Notch pathway may have therapeutic uses in treating fibrotic diseases and cancers.


PLOS ONE | 2016

Sprouty2 Suppresses Epithelial-Mesenchymal Transition of Human Lens Epithelial Cells through Blockade of Smad2 and ERK1/2 Pathways.

Xuhua Tan; Yi Zhu; Chuan Chen; Xiaoyun Chen; Yingyan Qin; Bo Qu; Lixia Luo; Haotian Lin; Mingxing Wu; Weirong Chen; Yizhi Liu

Transforming growth factor β (TGFβ)-induced epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) plays a key role in the pathogenesis of anterior subcapsular cataract (ASC) and capsule opacification. In mouse lens, Sprouty2 (Spry2) has a negative regulatory role on TGFβ signaling. However, the regulation of Spry2 during ASC development and how Spry2 modulates TGFβ signaling pathway in human LECs have not been characterized. Here, we demonstrate that Spry2 expression level is decreased in anterior capsule LECs of ASC patients. Spry2 negatively regulates TGFβ2-induced EMT and migration of LECs through inhibition of Smad2 and ERK1/2 phosphorylation. Also, blockade of Smad2 or ERK1/2 activation suppresses EMT caused by Spry2 downregulation. Collectively, our results for the first time show in human LECs that Spry2 has an inhibitory role in TGFβ signaling pathway. Our findings in human lens tissue and epithelial cells suggest that Spry2 may become a novel therapeutic target for the prevention and treatment of ASC and capsule opacification.


Scientific Reports | 2017

Improvement of Uveal and Capsular Biocompatibility of Hydrophobic Acrylic Intraocular Lens by Surface Grafting with 2-Methacryloyloxyethyl Phosphorylcholine-Methacrylic Acid Copolymer

Xuhua Tan; Jiezhao Zhan; Yi Zhu; Ji Cao; Lin Wang; Sa Liu; Yingjun Wang; Zhenzhen Liu; Yingyan Qin; Mingxing Wu; Yizhi Liu; Li Ren

Biocompatibility of intraocular lens (IOL) is critical to vision reconstruction after cataract surgery. Foldable hydrophobic acrylic IOL is vulnerable to the adhesion of extracellular matrix proteins and cells, leading to increased incidence of postoperative inflammation and capsule opacification. To increase IOL biocompatibility, we synthesized a hydrophilic copolymer P(MPC-MAA) and grafted the copolymer onto the surface of IOL through air plasma treatment. X-ray photoelectron spectroscopy, atomic force microscopy and static water contact angle were used to characterize chemical changes, topography and hydrophilicity of the IOL surface, respectively. Quartz crystal microbalance with dissipation (QCM-D) showed that P(MPC-MAA) modified IOLs were resistant to protein adsorption. Moreover, P(MPC-MAA) modification inhibited adhesion and proliferation of lens epithelial cells (LECs) in vitro. To analyze uveal and capsular biocompatibility in vivo, we implanted the P(MPC-MAA) modified IOLs into rabbits after phacoemulsification. P(MPC-MAA) modification significantly reduced postoperative inflammation and anterior capsule opacification (ACO), and did not affect posterior capsule opacification (PCO). Collectively, our study suggests that surface modification by P(MPC-MAA) can significantly improve uveal and capsular biocompatibility of hydrophobic acrylic IOL, which could potentially benefit patients with blood-aqueous barrier damage.


Journal of Ocular Pharmacology and Therapeutics | 2014

Effect of Topical Nonsteroidal Anti-Inflammatory Drugs and Nuclear Hardness on Maintenance of Mydriasis During Phacoemulsification Surgery

Chang Liu; Yuhua Liu; Shaobi Ye; Liangping Liu; Wanjun Zhang; Mingxing Wu

PURPOSE To compare the effects of topical nonsteroidal anti-inflammatory drugs on pupil dilation maintenance during phacoemulsification cataract surgery and quantify the relationships between pupil size change and nuclear hardness. METHODS This prospective randomized clinical observation study was single centered and double-masked. We studied 239 cases undergoing uneventful phacoemulsification cataract surgery in the absence of significant ocular comorbidity. Cases were randomly assigned to 1 of 6 groups receiving the following treatments: (1) diclofenac (0.1%); (2) pranoprofen (0.1%); (3) control, physiological normal saline solution; (4) diclofenac (0.1%) and epinephrine; (5) pranoprofen (0.1%) and epinephrine; (6) control, physiological normal saline and epinephrine solutions. Pupil diameter was measured at 3 intervals of cataract surgery: before the first incision, at the end of nucleus fragmentation, and at the end of cortex irrigation/aspiration. RESULTS Compared with patients who were not treated, there was a significant difference in maintaining pupil dilation throughout the operation when the patients were treated with either diclofenac or pranoprofen, P<0.001 and P<0.03, respectively. From the first incision to postnucleus fragmentation, the change in pupil size in both diclofenac and control groups was significantly associated with the hardness of the crystalline lens, P=0.001 and P=0.012, respectively. At the end of irrigation/aspiration, the change in pupil size in only the control groups was significantly associated with the hardness of the crystalline lens, P=0.022. Diclofenac treatment was most effective at inhibiting pupil miosis when the hardness of the nucleus was grade 3, P=0.009. Pupil miosis was not related to the hardness of the nucleus when the patients were treated with epinephrine. CONCLUSIONS Both diclofenac and pranoprofen treatment inhibit surgical-induced miosis. There is a negative correlation between the hardness of the crystalline lens and pupil diameter maintenance at the early stage of phacoemulsification.


Cell Death and Disease | 2017

Killing two birds with one stone: dual blockade of integrin and FGF signaling through targeting syndecan-4 in postoperative capsular opacification

Yingyan Qin; Yi Zhu; Furong Luo; Chuan Chen; Xiaoyun Chen; Mingxing Wu

The most common complication after cataract surgery is postoperative capsular opacification, which includes anterior capsular opacification (ACO) and posterior capsular opacification (PCO). Increased adhesion of lens epithelial cells (LECs) to the intraocular lens material surface promotes ACO formation, whereas proliferation and migration of LECs to the posterior capsule lead to the development of PCO. Cell adhesion is mainly mediated by the binding of integrin to extracellular matrix proteins, while cell proliferation and migration are regulated by fibroblast growth factor (FGF). Syndecan-4 (SDC-4) is a co-receptor for both integrin and FGF signaling pathways. Therefore, SDC-4 may be an ideal therapeutic target for the prevention and treatment of postoperative capsular opacification. However, how SDC-4 contributes to FGF-mediated proliferation, migration, and integrin-mediated adhesion of LECs is unclear. Here, we found that downregulation of SDC-4 inhibited FGF signaling through the blockade of ERK1/2 and PI3K/Akt/mTOR activation, thus suppressing cell proliferation and migration. In addition, downregulation of SDC-4 suppressed integrin-mediated cell adhesion through inhibiting focal adhesion kinase (FAK) phosphorylation. Moreover, SDC-4 knockout mice exhibited normal lens morphology, but had significantly reduced capsular opacification after injury. Finally, SDC-4 expression level was increased in the anterior capsule LECs of age-related cataract patients. Taken together, we for the first time characterized the key regulatory role of SDC-4 in FGF and integrin signaling in human LECs, and provided the basis for future pharmacological interventions of capsular opacification.

Collaboration


Dive into the Mingxing Wu's collaboration.

Top Co-Authors

Avatar

Yizhi Liu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Yi Zhu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Furong Luo

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Yingyan Qin

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chuan Chen

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lixia Luo

Sun Yat-sen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge