Network


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

Hotspot


Dive into the research topics where Yu-Hua Chen is active.

Publication


Featured researches published by Yu-Hua Chen.


Microvascular Research | 2011

Inactivation of EphA2 promotes tight junction formation and impairs angiogenesis in brain endothelial cells.

Na Zhou; Wei-Dong Zhao; Dong-Xin Liu; Yue Liang; Wen-Gang Fang; Bo Li; Yu-Hua Chen

Eph receptor tyrosine kinases and ephrin ligands participate in the regulation of a wide variety of biological processes, such as axon guidance, synaptic plasticity, angiogenesis, and tumorigenesis. The role of Eph receptors and ephrin ligands in brain endothelial cells remains unknown. Here, we examined the expression profile of EphA receptors and ephrin-A ligands in human brain microvascular endothelial cell line (HBMEC). Our results showed that multiple EphA receptors and ephrin-A ligands are expressed in HBMEC. We found that the phosphorylation of EphA2, but not other EphA receptors, was significantly increased in HBMEC treated with recombinant ephrin-A1/Fc. Meanwhile, elevated EphA2 phosphorylation was accompanied by disassembly of tight junctions in HBMEC. Furthermore, EphA2 RNAi in HBMEC could promote tight junction formation and prevent the ephrin-A1-induced tight junction disruption. Also, when a kinase-inactive mutant of EphA2 (EphA2-K646M) was expressed in HBMEC, the tight junction was enhanced and the ephrin-A1-induced tight junction disruption was blocked. In addition, EphA2 RNAi and expression of EphA2-K646M in HBMEC inhibited in vitro cell migration and angiogenesis of HBMEC. These data indicated an important role of EphA2 in regulating both tight junction formation and angiogenesis in brain endothelial cells.


Brain Research | 2013

Involvement of PI3K and ROCK signaling pathways in migration of bone marrow-derived mesenchymal stem cells through human brain microvascular endothelial cell monolayers

Mei-Na Lin; De-Shu Shang; Wei Sun; Bo Li; Xin Xu; Wen-Gang Fang; Wei-Dong Zhao; Liu Cao; Yu-Hua Chen

Bone marrow-derived mesenchymal stem cells (MSC) represent an important and easily available source of stem cells for potential therapeutic use in neurological diseases. The entry of circulating cells into the central nervous system by intravenous administration requires, firstly, the passage of the cells across the blood-brain barrier (BBB). However, little is known of the details of MSC transmigration across the BBB. In the present study, we employed an in vitro BBB model constructed using a human brain microvascular endothelial cell monolayer to study the mechanism underlying MSC transendothelial migration. Transmigration assays, transendothelial electrical resistance (TEER) and horseradish peroxidase (HRP) flux assays showed that MSC could transmigrate through human brain microvascular endothelial cell monolayers by a paracellular pathway. Cell fractionation and immunofluorescence assays confirmed the disruption of tight junctions. Inhibition assays showed that a Rho-kinase (ROCK) inhibitor (Y27632) effectively promoted MSC transendothelial migration; conversely, a PI3K inhibitor (LY294002) blocked MSC transendothelial migration. Interestingly, adenovirus-mediated interference with ROCK in MSC significantly increased MSC transendothelial migration, and overexpression of a PI3K dominant negative mutant in MSC cells could block transendothelial migration. Our findings provide clear evidence that the PI3K and ROCK pathways are involved in MSC migration through human brain microvascular endothelial cell monolayers. The information yielded by this study may be helpful in constructing gene-modified mesenchymal stem cells that are able to penetrate the BBB effectively for cell therapy.


PLOS ONE | 2013

CXCL1 contributes to β-amyloid-induced transendothelial migration of monocytes in Alzheimer's disease.

Ke Zhang; Li Tian; Li Liu; Yu Feng; Yanbin Dong; Bo Li; De-Shu Shang; Wen-Gang Fang; Yun-Peng Cao; Yu-Hua Chen

Background Bone marrow-derived microglia that originates in part from hematopoietic cells, and more particularly from monocytes preferentially attach to amyloid deposition in brains of Alzheimer’s disease (AD). However, the mechanism of monocytes recruited into the amyloid plaques with an accelerated process in AD is unclear. Methodology/Principal Findings Here we reported that monocytes from AD patients express significantly higher chemokine (C-X-C motif) ligand 1 (CXCL1) compared to age-matched controls. AD patient’s monocytes or CXCL1-overexpressing THP-1 cells had enhanced ability of β-amyloid (Aβ)-induced transendothelial migration and Aβ-induced transendothelial migration for AD patient’s monocytes or CXCL1-overexpressing THP-1 cells was almost abrogated by anti-CXCL1 antibody. Furthermore, monocytes derived from a transgenic mouse model of AD also expressed significantly higher CXCL1. CD11b+CD45hi population of cells that were recruited from the peripheral blood were markedly bolcked in APP mouse brain by anti-CXCL1 antibody. Accordingly, in response to Aβ, human brain microvascular endothelial cells (HBMEC) significantly up-regulated CXC chemokine receptor 2 (CXCR2) expression, which was the only identified receptor for CXCL1. In addition, a high level expression of CXCR2 in HBMEC significantly promoted the CXCL1-overexpressing THP-1 cells transendothelial migration, which could be was abrogated by anti-CXCR2 antibody. Further examination of possible mechanisms found that CXCL1-overexpressing THP-1 cells induced transendothelial electrical resistance decrease, horseradish peroxidase flux increase, ZO-1 discontinuous and occludin re-distribution from insoluble to soluble fraction through interacting with CXCR2. ROCK inhibitor, Y27632, could block CXCL1-overexpressing THP-1 cells transendothelial migration, whereas other inhibitors had no effects. Conclusions/Significance The present data indicate that monocytes derived from AD patients overexpressing CXCL1, which is a determinant for Aβ-induced transendothelial migration. CXCL1 expressed by monocytes and CXCR2 on HBMEC is involved in monocytes migrating from blood to brain in AD patients.


FEBS Letters | 2010

Involvement of Src tyrosine kinase in Escherichia coli invasion of human brain microvascular endothelial cells

Wei Liu; Wei-Dong Zhao; Jin-Chun Yan; Zhi-Yuan Ren; Wen-Gang Fang; Li Zhu; De-Shu Shang; Yu-Hua Chen

MINT‐7296149: F‐actin (uniprotkb:P60709) and Src‐DN (uniprotkb:P12931) colocalize (MI:0403) by fluorescence microscopy (MI:0416)


Anatomical Record-advances in Integrative Anatomy and Evolutionary Biology | 2014

Ephrin-A3 and Ephrin-A4 Contribute to Microglia-Induced Angiogenesis in Brain Endothelial Cells

Ying Li; Dong-Xin Liu; Mei-Yang Li; Xiao-Xue Qin; Wen-Gang Fang; Wei-Dong Zhao; Yu-Hua Chen

The association of microglia with brain vasculature during development and the reduced brain vascular complexity in microglia‐deficient mice suggest the role of microglia in cerebrovascular angiogenesis. However, the underlying molecular mechanism remains unclear. Here, using an in vitro angiogenesis model, we found the culture supernatant of BV2 microglial cells significantly enhanced capillary‐like tube formation and migration of brain microvascular endothelial cells (BMECs). The expression of angiogenic factors, ephrin‐A3 and ephrin‐A4, were specifically upregulated in BMECs exposed to BV2‐derived culture supernatant. Knockdown of ephrin‐A3 and ephrin‐A4 in BMECs by siRNA significantly attenuated the enhanced angiogenesis and migration of BMECs induced by BV2 supernatant. Our further results indicated that the ability of BV2 supernatant to promote endothelial angiogenesis was caused by the soluble tumor necrosis factor α (TNF‐α) released from BV2 microglial cells. Moreover, the upregulations of ephrin‐A3 and ephrin‐A4 in BMECs in response to BV2 supernatant were effectively abolished by neutralization antibody against TNF‐α and TNF receptor 1, respectively. The present study provides evidence that microglia upregulates endothelial ephrin‐A3 and ephrin‐A4 to facilitate in vitro angiogenesis of brain endothelial cells, which is mediated by microglia‐released TNF‐α. Anat Rec, 297:1908–1918, 2014.


Anatomical Record-advances in Integrative Anatomy and Evolutionary Biology | 2014

VEGF Increases Paracellular Permeability in Brain Endothelial Cells via Upregulation of EphA2

Zi-Wei Miao; Yanbin Dong; Wen-Gang Fang; De-Shu Shang; Dong-Xin Liu; Ke Zhang; Bo Li; Yu-Hua Chen

Neurological disorders are associated with an increase in the permeability of human brain microvascular endothelial cells (HBMEC). Our previous findings have indicated that EphA2 could increase the permeability of HBMEC. Recent evidence has linked EphA2 and vascular endothelial growth factor (VEGF) to abnormalities in the vascular response. However, it is unclear whether EphA2 is involved in the VEGF‐induced changes in the permeability of HBMEC. Here, changes in permeability were determined by measuring transendothelial electrical resistance (TEER) and the flux of FITC‐dextran. We found that knockdown of EphA2 in HBMEC abolished the VEGF‐induced reduction in TEER and increase in flux of fluorescent dextran. Moreover, VEGF‐induced redistribution of ZO‐1 and the recruitment of detergent‐soluble occludin and claudin‐5 were also prevented. Further results showed that VEGF increased EphA2 expression in a time‐ and dose‐dependent manner, which was inhibited by a neutralizing antibody against VEGFR2 or SU1498. VEGF‐induced EphA2 expression was suppressed in the brain endothelium following treatments with the PI3K inhibitor LY294002, Akt inhibitor or transfection with the dominant‐negative PI3K mutants (Δp110). Similar results were obtained when ERK1/2 activation was inhibited by PD98059 or ERK1/2 siRNA transfection. Our data suggest that VEGF upregulates the expression of EphA2 in HBMEC through binding to VEGFR2 and subsequently activating the intracellular PI3K/Akt and ERK1/2 signaling pathways, which contribute to an increase in paracellular permeability. These data reveal a novel role for VEGF as a regulator of EphA2 expression in the brain endothelial cells and provide insights into the molecular mechanisms of VEGF‐mediated changes in paracellular permeability. Anat Rec, 297:964–972, 2014.


Journal of Alzheimer's Disease | 2014

Decreased expression of cathepsin D in monocytes is related to the defective degradation of amyloid-β in Alzheimer's disease.

Li Tian; Ke Zhang; Zhi-Ying Tian; Tao Wang; De-Shu Shang; Bo Li; Dong-Xin Liu; Wen-Gang Fang; Zhan-You Wang; Yu-Hua Chen

Alzheimers disease (AD) is a progressive neurodegenerative dementia characterized by pathological senile plaques composed of amyloid-β (Aβ) in the cerebral cortex and hippocampus. Bone marrow-derived monocytes of patients with AD migrate across the blood-brain barrier into the brain, but are defective at clearing Aβ in the neuritic plaques. However, the underlying mechanisms remain unclear. Here, in patients with AD, we found that cathepsin D, a major lysosomal aspartic protease, was underexpressed in monocytes, resulting in the defective degradation of Aβ by monocytes/macrophages. Further, downregulation of cathepsin D in THP-1 cells significantly reduced the clearance of amyloid plaques in the brain sections of AβPP/PS1 mice. The clearance ability was recovered by the overexpression of cathepsin D in AD monocytes. These results suggest that decreased expression of cathepsin D in the peripheral monocytes is a potential signature of AD, and that this decreased expression is involved in Aβ degradation and AD pathogenesis.


Biochemical and Biophysical Research Communications | 2012

cPLA2α-mediated actin rearrangements downstream of the Akt signaling is required for Cronobacter sakazakii invasion into brain endothelial cells.

Dong-Xin Liu; Wei-Dong Zhao; Wen-Gang Fang; Yu-Hua Chen

Cronobacter sakazakii (C. sakazakii) is an opportunistic pathogen that causes sepsis and meningitis in neonate. The molecular mechanism involved in the pathogenesis of C. sakazakii meningitis remains unclear. In this study, we found that C. sakazakii invasion was significantly decreased in human brain microvascular endothelial cells (HBMEC) treated with cytosolic phospholipases A(2)α (cPLA(2)α) inhibitor. Increased phosphorylation of cPLA(2)α was observed in HBMEC infected with C. sakazakii, which was prevented by treatment with cPLA(2)α inhibitor. cPLA(2)α knockdown in HBMEC significantly attenuated C. sakazakii invasion into HBMEC. Immunofluorescence demonstrated that the rearrangements of actin filaments in HBMEC induced by C. sakazakii were effectively blocked by either treatment with cPLA(2)α inhibitor or transfection with cPLA(2)α siRNA. Interestingly, we found that C. sakazakii infection promoted the aggregation of phosphorylated cPLA(2)α, which was associated with depolymerized actin filaments in HBMEC. Furthermore, our data revealed that cPLA(2)α acts downstream of Akt signaling pathway in HBMEC stimulated with C. sakazakii. Taken together, our results illustrated that cPLA(2)α-mediated actin filament rearrangements downstream of Akt activation is required for C. sakazakii invasion into brain endothelial cells.


PLOS ONE | 2016

Cystatin C Shifts APP Processing from Amyloid-β Production towards Non-Amyloidgenic Pathway in Brain Endothelial Cells

Xia-Fei Wang; Dong-Xin Liu; Yue Liang; Li-Li Xing; Wenhui Zhao; Xiao-Xue Qin; De-Shu Shang; Bo Li; Wen-Gang Fang; Liu Cao; Wei-Dong Zhao; Yu-Hua Chen

Amyloid-β (Aβ), the major component of neuritic plaques in Alzheimer’s disease (AD), is derived from sequential proteolytic cleavage of amyloid protein precursor (APP) by secretases. In this study, we found that cystatin C (CysC), a natural cysteine protease inhibitor, is able to reduce Aβ40 secretion in human brain microvascular endothelial cells (HBMEC). The CysC-induced Aβ40 reduction was caused by degradation of β-secretase BACE1 through the ubiquitin/proteasome pathway. In contrast, we found that CysC promoted secretion of soluble APPα indicating the activated non-amyloidogenic processing of APP in HBMEC. Further results revealed that α-secretase ADAM10, which was transcriptionally upregulated in response to CysC, was required for the CysC-induced sAPPα secretion. Knockdown of SIRT1 abolished CysC-triggered ADAM10 upregulation and sAPPα production. Taken together, our results demonstrated that exogenously applied CysC can direct amyloidogenic APP processing to non-amyloidgenic pathway in brain endothelial cells, mediated by proteasomal degradation of BACE1 and SIRT1-mediated ADAM10 upregulation. Our study unveils previously unrecognized protective role of CysC in APP processing.


Alzheimer Disease & Associated Disorders | 2015

Alterations in cholesterol and ganglioside GM1 content of lipid rafts in platelets from patients with Alzheimer disease.

Li Liu; Ke Zhang; Liang Tan; Yu-Hua Chen; Yun-Peng Cao

The aim of this study was to investigate the changes in the protein, cholesterol, and ganglioside GM1 content of lipid rafts in platelets from patients with Alzheimer disease (AD), and identify potential blood biomarkers of the disease. A total of 31 Chinese patients with AD and 31 aged-matched control subjects were selected. Lipid rafts were isolated from platelets using Optiprep gradient centrifugation. The protein content of lipid rafts was evaluated using Micro BCA assay, the cholesterol content using molecular probes, ganglioside GM1 content using colorimetry and dot-blotting analysis. The results showed that the cholesterol and ganglioside GM1 content of lipid rafts from platelets was significantly higher in patients with AD than aged-matched control subjects, whereas the protein content of lipid rafts did not show any differences between the 2 groups. These results indicate that the increases in the cholesterol and ganglioside GM1 content of lipid rafts from the platelets of patients with AD might serve as a biochemical adjunct to the clinical diagnosis of AD.

Collaboration


Dive into the Yu-Hua Chen's collaboration.

Top Co-Authors

Avatar

Wen-Gang Fang

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Wei-Dong Zhao

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Bo Li

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Dong-Xin Liu

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

De-Shu Shang

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Ke Zhang

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Jia-Yi Wei

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Liu Cao

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Zi-Wei Miao

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Xiao-Xue Qin

Chinese Ministry of Education

View shared research outputs
Researchain Logo
Decentralizing Knowledge