Network


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

Hotspot


Dive into the research topics where Cheng Hsiang Kuo is active.

Publication


Featured researches published by Cheng Hsiang Kuo.


Blood | 2008

Lectin-like domain of thrombomodulin binds to its specific ligand Lewis Y antigen and neutralizes lipopolysaccharide-induced inflammatory response

Chung-Sheng Shi; Guey-Yueh Shi; Shi-Ming Hsiao; Yuan-Chung Kao(高遠忠); Kuan-Lin Kuo; Chih-Yuan Ma; Cheng Hsiang Kuo; Bi-Ing Chang; Chuan Fa Chang; Chun-Hung Lin; Chi-Huey Wong; Hua-Lin Wu

Thrombomodulin (TM), a widely expressing glycoprotein originally identified in vascular endothelium, is an important cofactor in the protein C anticoagulant system. TM appears to exhibit anti-inflammatory ability through both protein C-dependent and -independent pathways. We presently have demonstrated that recombinant N-terminal lectinlike domain of TM (rTMD1) functions as a protective agent against sepsis caused by Gram-negative bacterial infections. rTMD1 caused agglutination of Escherichia coli and Klebsiella pneumoniae and enhanced the macrophage phagocytosis of these Gram-negative bacteria. Moreover, rTMD1 bound to the Klebsiella pneumoniae and lipopolysaccharide (LPS) by specifically interacting with Lewis Y antigen. rTMD1 inhibited LPS-induced inflammatory mediator production via interference with CD14 and LPS binding. Furthermore, rTMD1 modulated LPS-induced mitogen-activated protein kinase and nuclear factor-kappaB signaling pathway activations and inducible nitric oxide synthase expression in macrophages. Administration of rTMD1 protected the host by suppressing inflammatory responses induced by LPS and Gram-negative bacteria, and enhanced LPS and bacterial clearance in sepsis. Thus, rTMD1 can be used to defend against bacterial infection and inhibit LPS-induced inflammatory responses, suggesting that rTMD1 may be valuable in the treatment of severe inflammation in sepsis, especially in Gram-negative bacterial infections.


Circulation | 2005

Evidence of human thrombomodulin domain as a novel angiogenic factor.

Chung Sheng Shi; Guey-Yueh Shi; Yi Sheng Chang; Huai Song Han; Cheng Hsiang Kuo; Che Liu; Huey-Chun Huang; Yu Jia Chang; Pin Shern Chen; Hua-Lin Wu

Background—Thrombomodulin is an anticoagulant, endothelial-cell-membrane glycoprotein. A recombinant thrombomodulin domain containing 6 epidermal growth factor–like structures exhibits mitogenic activity. This study explored the novel angiogenic effects of the recombinant domain using in vitro and in vivo models. Methods and Results—Human recombinant thrombomodulin containing 6 epidermal growth factor–like structures (TMD2) and TMD2 plus a serine and threonine-rich domain (TMD23) were prepared using the Pichia pastoris expression system. Combined with purified TMD2 or TMD23, thrombin effectively activated protein C. TMD23 had higher activity than TMD2 in stimulating DNA synthesis in cultured human umbilical vein endothelial cells. Additionally, TMD23 stimulated chemotactic motility and capillarylike tube formation in human umbilical vein endothelial cells, an effect mediated through phosphorylation of extracellular signal–regulated kinase 1/2 and p38 mitogen-activated protein kinase and the phosphatidylinositol-3 kinase/Akt/endothelial nitric oxide synthase pathway. TMD23 also stimulated endothelial cell expression of matrix metalloproteinases and plasminogen activators, which mediated extracellular proteolysis, leading to endothelial cell invasion and migration during angiogenesis. Furthermore, TMD23-containing implants in rat cornea induced ingrowth of new blood vessels from the limbus. With the murine angiogenesis assay, TMD23 not only induced neovascularization coinjected with Matrigel and heparin but also enhanced angiogenesis in Matrigel containing melanoma A2058 cells in nude mice. Conclusions—The recombinant thrombomodulin domain TMD23 enhanced the angiogenic response in vitro and in vivo, suggesting that thrombomodulin fragments may play a role in the formation of new vessels. These findings may provide a new therapeutic option for treating ischemic diseases.


Journal of Biomedical Science | 2012

The role of thrombomodulin lectin-like domain in inflammation

Yi-Heng Li; Cheng Hsiang Kuo; Guey-Yueh Shi; Hua-Lin Wu

Thrombomodulin (TM) is a cell surface glycoprotein which is widely expressed in a variety of cell types. It is a cofactor for thrombin binding that mediates protein C activation and inhibits thrombin activity. In addition to its anticoagulant activity, recent evidence has revealed that TM, especially its lectin-like domain, has potent anti-inflammatory function through a variety of molecular mechanisms. The lectin-like domain of TM plays an important role in suppressing inflammation independent of the TM anticoagulant activity. This article makes an extensive review of the role of TM in inflammation. The molecular targets of TM lectin-like domain have also been elucidated. Recombinant TM protein, especially the TM lectin-like domain may play a promising role in the management of sepsis, glomerulonephritis and arthritis. These data demonstrated the potential therapeutic role of TM in the treatment of inflammatory diseases.


Diabetologia | 2014

Thrombomodulin domain 1 ameliorates diabetic nephropathy in mice via anti-NF-κB/NLRP3 inflammasome-mediated inflammation, enhancement of NRF2 antioxidant activity and inhibition of apoptosis

Shun Min Yang; Shuk Man Ka; Hua-Lin Wu; Yu Chuan Yeh; Cheng Hsiang Kuo; Kuo Feng Hua; Guey-Yueh Shi; Yi Jen Hung; Fone Ching Hsiao; Sung Sen Yang; Yi Shing Shieh; Lin Sh; Chyou Wei Wei; Jeng Shin Lee; Chu Yi Yang; Ann Chen

Aims/hypothesisChronic inflammatory processes have been increasingly shown to be involved in the pathogenesis of diabetes and diabetic nephropathy. Recently, we demonstrated that a lectin-like domain of thrombomodulin (THBD), which is known as THBD domain 1 (THBDD1) and which acts independently of protein C activation, neutralised an inflammatory response in a mouse model of sepsis. Here, therapeutic effects of gene therapy with adeno-associated virus (AAV)-carried THBDD1 (AAV-THBDD1) were tested in a mouse model of type 2 diabetic nephropathy.MethodsTo assess the therapeutic potential of THBDD1 and the mechanisms involved, we delivered AAV-THBDD1 (1011 genome copies) into db/db mice and tested the effects of recombinant THBDD1 on conditionally immortalised podocytes.ResultsA single dose of AAV-THBDD1 improved albuminuria, renal interstitial inflammation and glomerular sclerosis, as well as renal function in db/db mice. These effects were closely associated with: (1) inhibited activation of the nuclear factor κB (NF-κB) pathway and the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome; (2) promotion of nuclear factor (erythroid-derived 2)-like 2 (NRF2) nuclear translocation; and (3) suppression of mitochondria-derived apoptosis in the kidney of treated mice.Conclusions/interpretationAAV-THBDD1 gene therapy resulted in improvements in a model of diabetic nephropathy by suppressing the NF-κB–NLRP3 inflammasome-mediated inflammatory process, enhancing the NRF2 antioxidant pathway and inhibiting apoptosis in the kidney.


Journal of Materials Chemistry B | 2014

Inhibition of atherosclerosis-promoting microRNAs via targeted polyelectrolyte complex micelles.

Cheng Hsiang Kuo; Lorraine Leon; Eun Ji Chung; Ru-Ting Huang; Timothy J. Sontag; Catherine A. Reardon; Godfrey S. Getz; Matthew Tirrell; Yun Fang

Polyelectrolyte complex micelles have great potential as gene delivery vehicles because of their ability to encapsulate charged nucleic acids forming a core by neutralizing their charge, while simultaneously protecting the nucleic acids from non-specific interactions and enzymatic degradation. Furthermore, to enhance specificity and transfection efficiency, polyelectrolyte complex micelles can be modified to include targeting capabilities. Here, we describe the design of targeted polyelectrolyte complex micelles containing inhibitors against dys-regulated microRNAs (miRNAs) that promote atherosclerosis, a leading cause of human mortality and morbidity. Inhibition of dys-regulated miRNAs in diseased cells associated with atherosclerosis has resulted in therapeutic efficacy in animal models and has been proposed to treat human diseases. However, the non-specific targeting of microRNA inhibitors via systemic delivery has remained an issue that may cause unwanted side effects. For this reason, we incorporated two different peptide sequences to our miRNA inhibitor containing polyelectrolyte complex micelles. One of the peptides (Arginine-Glutamic Acid-Lysine-Alanine or REKA) was used in another micellar system that demonstrated lesion-specific targeting in a mouse model of atherosclerosis. The other peptide (Valine-Histidine-Proline-Lysine-Glutamine-Histidine-Arginine or VHPKQHR) was identified via phage display and targets vascular endothelial cells through the vascular cell adhesion molecule-1 (VCAM-1). In this study we have tested the in vitro efficacy and efficiency of lesion- and cell-specific delivery of microRNA inhibitors to the cells associated with atherosclerotic lesions via peptide-targeted polyelectrolyte complex micelles. Our results show that REKA-containing micelles (fibrin-targeting) and VHPKQHR-containing micelles (VCAM-1 targeting) can be used to carry and deliver microRNA inhibitors into macrophages and human endothelial cells, respectively. Additionally, the functionality of miRNA inhibitors in cells was demonstrated by analyzing miRNA expression as well as the expression or the biological function of its downstream target protein. Our study provides the first demonstration of targeting dys-regulated miRNAs in atherosclerosis using targeted polyelectrolyte complex micelles and holds promising potential for translational applications.


Cardiovascular Research | 2011

Thrombomodulin domains attenuate atherosclerosis by inhibiting thrombin-induced endothelial cell activation

Hsi Ju Wei; Yi-Heng Li; Guey-Yueh Shi; Shu Lin Liu; Po Chiao Chang; Cheng Hsiang Kuo; Hua-Lin Wu

AIMSnThrombin modulates the formation of atherosclerotic lesions by stimulating a variety of cellular effects through protease-activated receptor-1 (PAR-1) activation. Thrombomodulin (TM) inhibits thrombin effects by binding thrombin through its domains 2 and 3 (TMD23). We investigated whether recombinant TMD23 (rTMD23) could inhibit atherosclerosis via its thrombin-binding ability.nnnMETHODS AND RESULTSnWild-type mouse rTMD23 and three mutants with altered thrombin-binding sites, rTMD23 (I425A), rTMD23 (D424A/D426A), and rTMD23 (D424A/I425A/D426A), were expressed and purified in the Pichia pastoris expression system. Wild-type rTMD23 and rTMD23 (D424A/D426A) could effectively bind thrombin, activate protein C, and prolong thrombin clotting time, whereas rTMD23 (I425A) and rTMD23 (D424A/I425A/D426A) lost these functions. Wild-type rTMD23, but not rTMD23 (I425A), decreased both the thrombin-induced surface PAR-1 internalization and the increase in cytoplasmic Ca(2+) concentrations in endothelial cells (ECs). Wild-type rTMD23 and rTMD23 (D424A/D426A) also inhibited thrombin-induced adhesion molecules and monocyte chemoattractant protein-1 expression and increased permeability in ECs, whereas rTMD23 (I425A) and rTMD23 (D424A/I425A/D426A) had no such effects. Furthermore, wild-type rTMD23 and rTMD23 (D424A/D426A) were effective in reducing carotid ligation-induced neointima formation in C57BL/6 mice and atherosclerotic lesion formation in apolipoprotein E-deficient (ApoE-/-) mice, whereas rTMD23 with the I425A mutation showed impairment of this function. Wild-type rTMD23, but not rTMD23 (I425A), also markedly suppressed the PAR-1, the adhesion molecules expression, and the macrophage content in the carotid ligation model and ApoE-/- mice.nnnCONCLUSIONnrTMD23 protein significantly reduces atherosclerosis and neointima formation through its thrombin-binding ability.


Circulation Research | 2015

Mechano-Sensitive PPAP2B Regulates Endothelial Responses to Athero-Relevant Hemodynamic Forces

Congqing Wu; Ru-Ting Huang; Cheng Hsiang Kuo; Sandeep Kumar; Chan Woo Kim; Yen-Chen Lin; Yen-Ju Chen; Anna A. Birukova; Konstantin G. Birukov; Nickolai O. Dulin; Mete Civelek; Aldons J. Lusis; Xavier Loyer; Alain Tedgui; Guohao Dai; Hanjoong Jo; Yun Fang

RATIONALEnPhosPhatidic Acid Phosphatase type 2B (PPAP2B), an integral membrane protein known as lipid phosphate phosphatase (LPP3) that inactivates lysophosphatidic acid, was implicated in coronary artery disease (CAD) by genome-wide association studies. However, it is unclear whether genome-wide association studies-identified coronary artery disease genes, including PPAP2B, participate in mechanotransduction mechanisms by which vascular endothelia respond to local atherorelevant hemodynamics that contribute to the regional nature of atherosclerosis.nnnOBJECTIVEnTo establish the critical role of PPAP2B in endothelial responses to hemodynamics.nnnMETHODS AND RESULTSnReduced PPAP2B was detected in vivo in mouse and swine aortic arch (AA) endothelia exposed to chronic disturbed flow, and in mouse carotid artery endothelia subjected to surgically induced acute disturbed flow. In humans, PPAP2B was reduced in the downstream part of carotid plaques where low shear stress prevails. In culture, reduced PPAP2B was measured in human aortic endothelial cells under atherosusceptible waveform mimicking flow in human carotid sinus. Flow-sensitive microRNA-92a and transcription factor KLF2 were identified as upstream inhibitor and activator of endothelial PPAP2B, respectively. PPAP2B suppression abrogated atheroprotection of unidirectional flow; inhibition of lysophosphatidic acid receptor 1 restored the flow-dependent, anti-inflammatory phenotype in PPAP2B-deficient cells. PPAP2B inhibition resulted in myosin light-chain phosphorylation and intercellular gaps, which were abolished by lysophosphatidic acid receptor 1/2 inhibition. Expression quantitative trait locus mapping demonstrated PPAP2B coronary artery disease risk allele is not linked to PPAP2B expression in various human tissues but significantly associated with reduced PPAP2B in human aortic endothelial cells.nnnCONCLUSIONSnAtherorelevant flows dynamically modulate endothelial PPAP2B expression through miR-92a and KLF2. Mechanosensitive PPAP2B plays a critical role in promoting anti-inflammatory phenotype and maintaining vascular integrity of endothelial monolayer under atheroprotective flow.


Biochemical and Biophysical Research Communications | 2008

Lysophosphatidic acid stimulates thrombomodulin lectin-like domain shedding in human endothelial cells.

Hua-Lin Wu; Chi-Iou Lin; Yuan-Li Huang; Pin-Shern Chen(陳品晟); Cheng Hsiang Kuo; Mei-Shing Chen; Georgiana Cho-Chen Wu; Guey-Yueh Shi; Hsi-Yuan Yang; Hsinyu Lee

Thrombomodulin (TM) is an anticoagulant glycoprotein highly expressed on endothelial cell surfaces. Increased levels of soluble TM in circulation have been widely accepted as an indicator of endothelial damage or dysfunction. Previous studies indicated that various proinflammatory factors stimulate TM shedding in various cell types such as smooth muscle cells and epithelial cells. Lysophosphatidic acid (LPA) is a bioactive lipid mediator present in biological fluids during endothelial damage or injury. In the present study, we first observed that LPA triggered TM shedding in human umbilical vein endothelial cells (HUVECs). By Cyflow analysis, we showed that the LPA-induced accessibility of antibodies to the endothelial growth factor (EGF)-like domain of TM is independent of matrix metalloproteinases (MMPs), while LPA-induced TM lectin-like domain shedding is MMP-dependent. Furthermore, a stable cell line expressing TM without its lectin-like domain exhibited a higher cell proliferation rate than a stable cell line expressing full-length TM. These results imply that LPA induces TM lectin-like domain shedding, which might contribute to the exposure of its EGF-like domain for EGF receptor (EGFR) binding, thereby stimulating subsequent cell proliferation. Based on our findings, we propose a novel mechanism for the exposure of TM EGF-like domain, which possibly mediates LPA-induced EGFR transactivation.


Blood | 2012

The recombinant lectin-like domain of thrombomodulin inhibits angiogenesis through interaction with Lewis Y antigen

Cheng Hsiang Kuo; Po Ku Chen; Bi Ing Chang; Meng Chen Sung; Chung Sheng Shi; Jeng Shin Lee; Chuan Fa Chang; Guey-Yueh Shi; Hua-Lin Wu

Lewis Y Ag (LeY) is a cell-surface tetrasaccharide that participates in angiogenesis. Recently, we demonstrated that LeY is a specific ligand of the recombinant lectin-like domain of thrombomodulin (TM). However, the biologic function of interaction between LeY and TM in endothelial cells has never been investigated. Therefore, the role of LeY in tube formation and the role of the recombinant lectin-like domain of TM-TM domain 1 (rTMD1)-in antiangiogenesis were investigated. The recombinant TM ectodomain exhibited lower angiogenic activity than did the recombinant TM domains 2 and 3. rTMD1 interacted with soluble LeY and membrane-bound LeY and inhibited soluble LeY-mediated chemotaxis of endothelial cells. LeY was highly expressed on membrane ruffles and protrusions during tube formation on Matrigel. Blockade of LeY with rTMD1 or Ab against LeY inhibited endothelial tube formation in vitro. Epidermal growth factor (EGF) receptor in HUVECs was LeY modified. rTMD1 inhibited EGF receptor signaling, chemotaxis, and tube formation in vitro, and EGF-mediated angiogenesis and tumor angiogenesis in vivo. We concluded that LeY is involved in vascular endothelial tube formation and rTMD1 inhibits angiogenesis via interaction with LeY. Administration of rTMD1 or recombinant adeno-associated virus vector carrying TMD1 could be a promising antiangiogenesis strategy.


The FASEB Journal | 2012

Thrombomodulin is an ezrin-interacting protein that controls epithelial morphology and promotes collective cell migration

Yun Yan Hsu; Guey-Yueh Shi; Cheng Hsiang Kuo; Shu Lin Liu; Ching Ming Wu; Chih Yuan Ma; Feng Yi Lin; Hsi-Yuan Yang; Hua-Lin Wu

Adhesive interactions between cells are needed to maintain tissue architecture during development, tissue renewal and wound healing. Thrombo‐modulin (TM) is an integral membrane protein that participates in cell–cell adhesion through its extracellular lectin‐like domain. However, the molecular basis of TM‐mediated cell–cell adhesion is poorly understood. Here, we demonstrate that TM is linked to the actin cytoskeleton via ezrin. In vitro binding assays showed that the TM cytoplasmic domain bound directly to the N‐terminal domain of ezrin. Mutational analysis of the TM cytoplasmic domain identified 522RKK524 as important ezrin‐binding residues. In epidermal epithelial A431 cells, TM colocalized with ezrin and actin filaments at cell–cell contacts. Knockdown of endogenous TM expression by RNA interference induced morphological changes and accelerated cell migration in A431 cells. Moreover, epidermal growth factor, upstream of ezrin activation, stimulated the interaction between ezrin and TM. In skin wound healing of mice, TM and ezrin were highly expressed in neoepidermis, implying that both proteins are key molecules in reepithelialization that requires collective cell migration of epithelial cells. Finally, exogenous expression of TM in TM‐deficient melanoma A2058 cells promoted collective cell migration. In summary, TM, which associates with ezrin and actin filaments, maintains epithelial morphology and promotes collective cell migration.—Hsu, Y.‐Y., Shi, G.‐Y., Kuo, C.‐H., Liu, S.‐L., Wu, C.‐M., Ma, C.‐Y., Lin, F.‐Y., Yang, H.‐Y., Wu, H.‐L. Thrombomodulin is an ezrin‐interacting protein that controls epithelial morphology and promotes collective cell migration. FASEB J. 26, 3440–3452 (2012). www.fasebj.org

Collaboration


Dive into the Cheng Hsiang Kuo's collaboration.

Top Co-Authors

Avatar

Guey-Yueh Shi

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Hua-Lin Wu

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Bi Ing Chang

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Fang Tzu Lee

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Po Ku Chen

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Yun Fang

University of Chicago

View shared research outputs
Top Co-Authors

Avatar

Chao Han Lai

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Kuan Chieh Wang

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Yi-Heng Li

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge