Network


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

Hotspot


Dive into the research topics where Weiguo Chen is active.

Publication


Featured researches published by Weiguo Chen.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2008

Endothelial cell barrier protection by simvastatin: GTPase regulation and NADPH oxidase inhibition

Weiguo Chen; Srikanth Pendyala; Viswanathan Natarajan; Joe G. N. Garcia; Jeffrey R. Jacobson

The statins, hydroxy-3-methylglutaryl-CoA reductase inhibitors that lower serum cholesterol, exhibit myriad clinical benefits, including enhanced vascular integrity. One potential mechanism underlying increased endothelial cell (EC) barrier function is inhibition of geranylgeranylation, a covalent modification enabling translocation of the small GTPases Rho and Rac to the cell membrane. While RhoA inhibition attenuates actin stress fiber formation and promotes EC barrier function, Rac1 inhibition at the cell membrane potentially prevents activation of NADPH oxidase and subsequent generation of superoxides known to induce barrier disruption. We examined the relative regulatory effects of simvastatin on RhoA, Rac1, and NADPH oxidase activities in the context of human pulmonary artery EC barrier protection. Confluent EC treated with simvastatin demonstrated significantly decreased thrombin-induced FITC-dextran permeability, a reflection of vascular integrity, which was linked temporally to simvastatin-mediated actin cytoskeletal rearrangement. Compared with Rho inhibition alone (Y-27632), simvastatin afforded additional protection against thrombin-mediated barrier dysfunction and attenuated LPS-induced EC permeability and superoxide generation. Statin-mediated inhibition of both Rac translocation to the cell membrane and superoxide production were attenuated by geranylgeranyl pyrophosphate (GGPP), indicating that these effects are due to geranylgeranylation inhibition. Finally, thrombin-induced EC permeability was modestly attenuated by reduced Rac1 expression (small interfering RNA), whereas these effects were made more pronounced by simvastatin pretreatment. Together, these data suggest EC barrier protection by simvastatin is due to dual inhibitory effects on RhoA and Rac1 as well as the attenuation of superoxide generation by EC NADPH oxidase and contribute to the molecular mechanistic understanding of the modulation of EC barrier properties by simvastatin.


Journal of Biological Chemistry | 2013

Critical Role of S1PR1 and Integrin β4 in HGF/c-Met-Mediated Increases in Vascular Integrity.

Yulia Ephstein; Patrick A. Singleton; Weiguo Chen; Lichun Wang; Ravi Salgia; Prasad Kanteti; Steven M. Dudek; Joe G. N. Garcia; Jeffrey R. Jacobson

Background: Hepatocyte growth factor (HGF) is a potent mediator of endothelial barrier enhancement, however, the mechanisms are poorly understood. Results: HGF-induced endothelial barrier enhancement is mediated via sphingosine 1-phosphate receptor 1 (S1PR1) and integrin β4 (ITGB4) transactivation. Conclusion: ITGB4 and S1PR1 are essential for HGF-induced endothelial barrier augmentation. Significance: Our findings identify novel mechanisms of endothelial barrier enhancement by HGF. Vascular endothelial cell (EC) barrier integrity is critical to vessel homeostasis whereas barrier dysfunction is a key feature of inflammatory disorders and tumor angiogenesis. We previously reported that hepatocyte growth factor (HGF)-mediated increases in EC barrier integrity are signaled through a dynamic complex present in lipid rafts involving its receptor, c-Met (1). We extended these observations to confirm that S1PR1 (sphingosine 1-phosphate receptor 1) and integrin β4 (ITGB4) are essential participants in HGF-induced EC barrier enhancement. Immunoprecipitation experiments demonstrated HGF-mediated recruitment of c-Met, ITGB4 and S1PR1 to caveolin-enriched lipid rafts in human lung EC with direct interactions of c-Met with both S1PR1 and ITGB4 accompanied by c-Met-dependent S1PR1 and ITGB4 transactivation. Reduced S1PR1 expression (siRNA) attenuated both ITGB4 and Rac1 activation as well as c-Met/ITGB4 interaction and resulted in decreased transendothelial electrical resistance. Furthermore, reduced ITGB4 expression attenuated HGF-induced c-Met activation, c-Met/S1PR1 interaction, and effected decreases in S1P- and HGF-induced EC barrier enhancement. Finally, the c-Met inhibitor, XL880, suppressed HGF-induced c-Met activation as well as S1PR1 and ITGB4 transactivation. These results support a critical role for S1PR1 and ITGB4 transactivation as rate-limiting events in the transduction of HGF signals via a dynamic c-Met complex resulting in enhanced EC barrier integrity.


American Journal of Respiratory Cell and Molecular Biology | 2013

Role of Claudin-5 in the Attenuation of Murine Acute Lung Injury by Simvastatin

Weiguo Chen; Rajesh Sharma; Alicia N. Rizzo; Jessica Siegler; Joe G. N. Garcia; Jeffrey R. Jacobson

The statins are now recognized to have pleiotropic properties, including augmentation of endothelial barrier function. To explore the mechanisms involved, we investigated the effect of simvastatin on endothelial cell (EC) tight junctions. Western blotting of human pulmonary artery ECs treated with simvastatin (5 μM) confirmed a significant time-dependent increase (16-48 h) in claudin-5 protein expression compared with controls, without detectable alterations in zonula occludens-1 or occludin. These effects were associated with membrane translocation of VE-cadherin, whereas translocation of vascular endothelial cadherin (VE-cadherin; silencing RNA) inhibited simvastatin-induced claudin-5 up-regulation. Moreover, simvastatin treatment of ECs induced increased phosphorylation of both FoxO1 and β-catenin, transcriptional regulators of claudin-5 expression mediated by VE-cadherin. Subsequently, we found no effect of claudin-5 silencing on EC barrier protection by simvastatin in response to thrombin stimulation, as measured by either transendothelial electrical resistance or by EC monolayer flux of FITC-dextran (2,000 kD). However, silencing of claudin-5 did significantly attenuate simvastatin-mediated EC barrier protection in response to thrombin, as measured by monolayer flux of sodium fluorescein (376 Da). Finally, employing a murine model of LPS-induced acute lung injury, there was no effect of claudin-5 silencing in vivo (intratracheal injection) on bronchoalveolar lavage fluid protein or cell counts, but LPS-induced lung tissue extravasation of the small molecular weight markers, sodium fluorescein and Hochst stain (562 Da), were significantly increased in claudin-5-silenced animals compared with simvastatin-treated control animals. These findings implicate a distinct mechanism underlying size-selective endothelial barrier-protective properties of statins, and may ultimately lead to new novel therapeutic targets for patients with acute lung injury.


PLOS ONE | 2014

Endothelial Nitric Oxide Synthase Dimerization Is Regulated by Heat Shock Protein 90 Rather than by Phosphorylation

Weiguo Chen; Hongbing Xiao; Alicia N. Rizzo; Wei Zhang; Yifeng Mai; Meng Ye

Endothelial nitric oxide synthase (eNOS) is a multifunctional enzyme with roles in diverse cellular processes including angiogenesis, tissue remodeling, and the maintenance of vascular tone. Monomeric and dimeric forms of eNOS exist in various tissues. The dimeric form of eNOS is considered the active form and the monomeric form is considered inactive. The activity of eNOS is also regulated by many other mechanisms, including amino acid phosphorylation and interactions with other proteins. However, the precise mechanisms regulating eNOS dimerization, phosphorylation, and activity remain incompletely characterized. We utilized purified eNOS and bovine aorta endothelial cells (BAECs) to investigate the mechanisms regulating eNOS degradation. Both eNOS monomer and dimer existed in purified bovine eNOS. Incubation of purified bovine eNOS with protein phosphatase 2A (PP2A) resulted in dephosphorylation at Serine 1179 (Ser1179) in both dimer and monomer and decrease in eNOS activity. However, the eNOS dimer∶monomer ratio was unchanged. Similarly, protein phosphatase 1 (PP1) induced dephosphorylation of eNOS at Threonine 497 (Thr497), without altering the eNOS dimer∶monomer ratio. Different from purified eNOS, in cultured BAECs eNOS existed predominantly as dimers. However, eNOS monomers accumulated following treatment with the proteasome inhibitor lactacystin. Additionally, treatment of BAECs with vascular endothelial growth factor (VEGF) resulted in phosphorylation of Ser1179 in eNOS dimers without altering the phosphorylation status of Thr497 in either form. Inhibition of heat shock protein 90 (Hsp90) or Hsp90 silencing destabilized eNOS dimers and was accompanied by dephosphorylation both of Ser1179 and Thr497. In conclusion, our study demonstrates that eNOS monomers, but not eNOS dimers, are degraded by ubiquitination. Additionally, the dimeric eNOS structure is the predominant condition for eNOS amino acid modification and activity regulation. Finally, destabilization of eNOS dimers not only results in eNOS degradation, but also causes changes in eNOS amino acid modifications that further affect eNOS activity.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2012

Critical role for integrin-β4 in the attenuation of murine acute lung injury by simvastatin

Weiguo Chen; Saad Sammani; Sumegha Mitra; Shwu Fan Ma; Joe G. N. Garcia; Jeffrey R. Jacobson

The statins are a class of 3-hydroxy-3-methylglutaryl-coenzyme A-reductase inhibitors that are recognized to have pleiotropic properties. We previously reported the attenuation of LPS-induced murine acute lung injury (ALI) by simvastatin in vivo and identified relevant effects of simvastatin on endothelial cell (EC) signaling, activation, and barrier function in vitro. In particular, simvastatin induces the upregulation of integrin-β4, which in turn inhibits EC inflammatory responses via attenuation of MAPK signaling. The role of integrin-β4 in murine ALI protection by simvastatin, however, is unknown. We initially confirmed a time- and dose-dependent effect of simvastatin on increased integrin-β4 mRNA expression in human lung EC with peak protein expression evident at 16 h. Subsequently, reciprocal immunoprecipitation demonstrated an attenuation of LPS-induced integrin-β4 tyrosine phosphorylation by simvastatin (5 μM, 16 h). Increased expression of EC inflammatory cytokines [IL-6, IL-8, monocyte chemoattractant protein (MCP)-1, regulated on activation normal T cell expressed and secreted (RANTES)] by LPS (500 ng/ml, 4 h) was also significantly attenuated by simvastatin pretreatment (5 μM, 16 h), but this effect was reversed by cotreatment with an integrin-β4-blocking antibody. Finally, although simvastatin (20 mg/kg) conferred significant protection in murine ALI as evidenced by decreased bronchoalveolar lavage fluid cell counts, protein, inflammatory cytokines (IL-6, IL-1β, MCP-1, RANTES), decreased Evans blue dye albumin extravasation in lung tissue, and changes on lung histology, these effects were reversed by the integrin-β4-blocking antibody (IV, 1 mg/kg, 2 h before LPS). These findings support integrin-β4 as an important mediator of ALI protection by simvastatin and implicate signaling by integrin-β4 as a novel therapeutic target in patients with ALI.


Journal of Cellular Biochemistry | 2010

Integrin β4 attenuates SHP-2 and MAPK signaling and reduces human lung endothelial inflammatory responses

Weiguo Chen; Joe G. N. Garcia; Jeffrey R. Jacobson

We previously identified the marked upregulation of integrin β4 in human lung endothelial cells (EC) treated with simvastatin, an HMG coA‐reductase inhibitor with vascular‐protective and anti‐inflammatory properties in murine models of acute lung injury (ALI). We now investigate the role of integrin β4 as a novel mediator of vascular inflammatory responses with a focus on mitogen‐activated protein kinases (MAPK) signaling and the downstream expression of the inflammatory cytokines (IL‐6 and IL‐8) essential for the full elaboration of inflammatory lung injury. Silencing of integrin β4 (siITGB4) in human lung EC resulted in significant increases in both basal and LPS‐induced phosphorylation of ERK 1/2, JNK, and p38 MAPK, consistent with robust integrin β4 regulation of MAPK activation. In addition, siITB4 increased both basal and LPS‐induced expression of IL‐6 and IL‐8 mRNA and protein secretion into the media. We next observed that integrin β4 silencing increased basal and LPS‐induced phosphorylation of SHP‐2, a protein tyrosine phosphatase known to modulate MAPK signaling. In contrast, inhibition of SHP‐2 enzymatic activity (sodium stibogluconate) abrogated the increased ERK phosphorylation associated with integrin β4 silencing in LPS‐treated EC and attenuated the increases in levels of IL‐6 and IL‐8 in integrin‐β4‐silenced EC. These findings highlight a novel negative regulatory role for integrin β4 in EC inflammatory responses involving SHP‐2‐mediated MAPK signaling. Upregulation of integrin β4 may represent an important element of the anti‐inflammatory and vascular‐protective properties of statins and provides a novel strategy to limit inflammatory vascular syndromes. J. Cell. Biochem. 110: 718–724, 2010.


Mediators of Inflammation | 2016

MicroRNA Regulation of Endothelial Junction Proteins and Clinical Consequence.

Yugang Zhuang; Hu Peng; Victoria Mastej; Weiguo Chen

Cellular junctions play a critical role in structural connection and signal communication between cells in various tissues. Although there are structural and functional varieties, cellular junctions include tight junctions, adherens junctions, focal adhesion junctions, and tissue specific junctions such as PECAM-1 junctions in endothelial cells (EC), desmosomes in epithelial cells, and hemidesmosomes in EC. Cellular junction dysfunction and deterioration are indicative of clinical diseases. MicroRNAs (miRNA) are ~20 nucleotide, noncoding RNAs that play an important role in posttranscriptional regulation for almost all genes. Unsurprisingly, miRNAs regulate junction protein gene expression and control junction structure integrity. In contrast, abnormal miRNA regulation of junction protein gene expression results in abnormal junction structure, causing related diseases. The major components of tight junctions include zonula occluden-1 (ZO-1), claudin-1, claudin-5, and occludin. The miRNA regulation of ZO-1 has been intensively investigated. ZO-1 and other tight junction proteins such as claudin-5 and occludin were positively regulated by miR-126, miR-107, and miR21 in different models. In contrast, ZO-1, claudin-5, and occludin were negatively regulated by miR-181a, miR-98, and miR150. Abnormal tight junction miRNA regulation accompanies cerebral middle artery ischemia, brain trauma, glioma metastasis, and so forth. The major components of adherens junctions include VE-cadherin, β-catenin, plakoglobin, P120, and vinculin. VE-cadherin and β-catenin were regulated by miR-9, miR-99b, miR-181a, and so forth. These regulations directly affect VE-cadherin-β-catenin complex stability and further affect embryo and tumor angiogenesis, vascular development, and so forth. miR-155 and miR-126 have been shown to regulate PECAM-1 and affect neutrophil rolling and EC junction integrity. In focal adhesion junctions, the major components are integrin β4, paxillin, and focal adhesion kinase (FAK). Integrin β4 has been regulated by miR-184, miR-205, and miR-9. Paxillin has been regulated by miR-137, miR-145, and miR-218 in different models. FAK has been regulated by miR-7, miR-138, and miR-135. Deregulation of miRNAs is caused by viral infections, tumorigenesis, and so forth. By regulation of posttranscription, miRNAs manipulate junction protein expression in all cellular processes and further determine cellular fate and development. Elucidation of these regulatory mechanisms will become a new alternative therapy for many diseases, such as cancers and inflammatory diseases.


Translational Research | 2013

MicroRNA regulation of integrins.

Weiguo Chen; Mark Harbeck; Wei Zhang; Jeffrey R. Jacobson

MicroRNAs (miRNAs) are a family of small RNAs that are ∼20 nucleotides in length and are nontranslated. To date, more than 700 miRNAs have been identified, and their involvement in many essential cellular processes is now apparent. By binding with target messenger RNAs (mRNA), miRNAs are able to regulate both mRNA stability and mRNA translational efficiency. Integrins are a family of transmembrane proteins that both regulate cell matrix interactions and serve as receptors that mediate intracellular signaling and a variety of cellular processes, including inflammatory responses, immunoresponses, and tumorigenesis. Integrin expression may also be regulated by miRNAs, which can also modulate integrin signaling and function. Integrins are heterodimer adhesion proteins comprised of an α and a β subunit. Cumulatively, there are 18 α subunits and 8 β subunits that can combine to form 24 distinct αβ receptor complexes. In addition, each integrin can be classified into 1 of 4 groups based on its extracellular binding ligand: collagen, laminin, RGD (Arg-Gly-Asp) or leukocyte-specific receptors. Collagen ligand integrins include integrins α1 and α2 subunits, known to be regulated by specific miRNAs. Among the laminin ligand integrins, there are no integrin α subunits known to be regulated by miRNA. As for the RGD ligand integrins, integrin α5 is the only α subunit found to be regulated by miRNAs (miR-31, miR-17-92 cluster, and miR-148 b). Finally, among the α subunits that comprise the leukocyte-specific receptor ligand integrins, integrins αD, αL, αM, and αX have shown regulation by different miRNAs. As for the integrin β subunits, regulation by miRNAs has been reported for all but β5 and β6 to date. However, computational predictions suggest that numerous miRNAs potentially regulate a variety of target integrins. These predictions will undoubtedly guide future investigations of mechanisms underlying integrin expression mechanism and may ultimately yield new therapeutic tools.


Scientific Reports | 2015

Role of Integrin β4 in Lung Endothelial Cell Inflammatory Responses to Mechanical Stress

Weiguo Chen; Yulia Epshtein; Xiuquin Ni; Randal O. Dull; Anne E. Cress; Joe G. N. Garcia; Jeffrey R. Jacobson

Simvastatin, an HMG-CoA reductase inhibitor, has lung vascular-protective effects that are associated with decreased agonist-induced integrin β4 (ITGB4) tyrosine phosphorylation. Accordingly, we hypothesized that endothelial cell (EC) protection by simvastatin is dependent on these effects and sought to further characterize the functional role of ITGB4 as a mediator of EC protection in the setting of excessive mechanical stretch at levels relevant to ventilator-induced lung injury (VILI). Initially, early ITGB4 tyrosine phosphorylation was confirmed in human pulmonary artery EC subjected to excessive cyclic stretch (18% CS). EC overexpression of mutant ITGB4 with specific tyrosines mutated to phenylalanine (Y1440, Y1526 Y1640, or Y1422) resulted in significantly attenuated CS-induced cytokine expression (IL6, IL-8, MCP-1, and RANTES). In addition, EC overexpression of ITGB4 constructs with specific structural deletions also resulted in significantly attenuated CS-induced inflammatory cytokine expression compared to overexpression of wildtype ITGB4. Finally, mice expressing a mutant ITGB4 lacking a cytoplasmic signaling domain were found to have attenuated lung injury after VILI-challenge (VT = 40 ml/kg, 4 h). Our results provide mechanistic insights into the anti-inflammatory properties of statins and may ultimately lead to novel strategies targeted at ITGB4 signaling to treat VILI.


Journal of Allergy and Therapy | 2013

The Importance of Bronchial Epithelial Junction Integrity in Asthma

Hongbing Xiao; Alicia N. Rizzo; Jessica Siegler; Weiguo Chen

Bronchial epithelial junctions provide not only a physical barrier, but also an immune barrier against the allergens of asthma. Epithelial junction integrity is closely related to the severity and progression of asthma. The bronchial epithelial barrier consists of tight junctions, adherens junctions, desmosomes, hemidesmosomes and gap junctions, all of which are potentially implicated in asthma pathophysiology. In tight junctions, claudins, occludens, ZO-1 and β-catenin expression have been shown to be decreased by asthma allergens, resulting tight junction disruption. Similarly, E-cadherin and α-catenin levels have also been reported to be dysregulated in response to asthma allergens, resulting in alterations in adherens junction structure. Asthma allergens also alter desmosome and hemidesmosome structure; however, no reports have shown that desmosome or hemidesmosome junction protein expression is altered in response to asthma allergens. Finally, in gap junctions, connexin 37 mRNA and protein were found to be decreased in the ovalbumin (OVA) induced allergic model. In summary, the regulation of bronchial junction protein expression and structure is an important and presently understudied component of asthma pathophysiology. We believe that further investigation in this area has the potential to aid in the development of novel asthma treatments.

Collaboration


Dive into the Weiguo Chen's collaboration.

Top Co-Authors

Avatar

Jeffrey R. Jacobson

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Saad Sammani

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Alicia N. Rizzo

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Biji Mathew

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jessica Siegler

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Lishi Xie

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge