Network


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

Hotspot


Dive into the research topics where Kuo-Hui Su is active.

Publication


Featured researches published by Kuo-Hui Su.


Journal of Cellular Physiology | 2011

β Common receptor integrates the erythropoietin signaling in activation of endothelial nitric oxide synthase.

Kuo-Hui Su; Song-Kun Shyue; Yu Ru Kou; Li-Chieh Ching; An-Na Chiang; Yuan-Bin Yu; Chien-Yu Chen; Ching-Chian Pan; Tzong-Shyuan Lee

Erythropoietin (EPO), the key hormone for erythropoiesis, also increases nitric oxide (NO) bioavailability in endothelial cells (ECs), yet the definitive mechanisms are not fully understood. Increasing evidence has demonstrated that β common receptor (βCR) plays a crucial role in EPO‐mediated non‐hematopoietic effects. We investigated the role of βCR in EPO‐induced endothelial NO synthase (eNOS) activation in bovine aortic ECs (BAECs) and the molecular mechanisms involved. Results of confocal microscopy and immunoprecipitation analyses revealed that βCR was colocalized and interacted with EPO receptor (EPOR) in ECs. Inhibition of βCR or EPOR by neutralizing antibodies or small interfering RNA abolished the EPO‐induced NO production. Additionally, blockage of βCR abrogated the EPO‐induced increase in the phosphorylation of eNOS, Akt, Src, or Janus kinase 2 (JAK2). Immunoprecipitation analysis revealed that treatment with EPO increased the interaction between βCR and eNOS, which was suppressed by inhibition of Src, JAK2, or Akt signaling with specific pharmacological inhibitors. Furthermore, EPO‐induced EC proliferation, migration, and tube formation were blocked by pretreatment with βCR antibody and Src, JAK2, or PI3K/Akt inhibitors. Moreover, in vivo experiments showed that EPO increased the level of phosphorylated eNOS, Src, JAK2, and Akt, as well as βCR–eNOS association in aortas and promoted the angiogenesis in Matrigel plug, which was diminished by βCR or EPOR neutralizing antibodies. Our findings suggest that βCR may play an integrative role in the EPO signaling‐mediated activation of eNOS in ECs. J. Cell. Physiol. 226: 3330–3339, 2011.


Cardiovascular Research | 2011

Molecular mechanisms of activation of endothelial nitric oxide synthase mediated by transient receptor potential vanilloid type 1

Li-Chieh Ching; Yu Ru Kou; Song-Kun Shyue; Kuo-Hui Su; Jeng Wei; Li-Ching Cheng; Yuan-Bin Yu; Ching-Chian Pan; Tzong-Shyuan Lee

AIMS We investigated the molecular mechanism underlying the role of transient receptor potential vanilloid type 1 (TRPV1), a Ca(2+)-permeable non-selective cation channel, in the activation of endothelial nitric oxide (NO) synthase (eNOS) in endothelial cells (ECs) and mice. METHODS AND RESULTS In ECs, TRPV1 ligands (evodiamine or capsaicin) promoted NO production, eNOS phosphorylation, and the formation of a TRPV1-eNOS complex, which were all abrogated by the TRPV1 antagonist capsazepine. TRPV1 ligands promoted the phosphorylation of Akt, calmodulin-dependent protein kinase II (CaMKII) and TRPV1, and increased the formation of a TRPV1-Akt-CaMKII complex. Removal of extracellular Ca(2+) abolished the ligand-induced increase in the phosphorylation of Akt and CaMKII, formation of a TRPV1-eNOS complex, and eNOS activation. Inhibition of PI3K and CaMKII suppressed the ligand-induced increase in TRPV1 phosphorylation, formation of a TRPV1-eNOS complex, and eNOS activation. TRPV1 activation increased the phosphorylation of Akt, CaMKII, and eNOS in the aortas of wild-type mice but failed to activate eNOS in TRPV1-deficient aortas. Additionally, TRPV1 ligand-induced angiogenesis was diminished in eNOS- or TRPV1-deficient mice. When compared with apolipoprotein E (ApoE)-deficient mice, ApoE/TRPV1-double-knockout mice displayed reduced phosphorylation of eNOS, Akt, and CaMKII in aortas but worsened atherosclerotic lesions. CONCLUSION TRPV1 activation in ECs may trigger Ca(2+)-dependent PI3K/Akt/CaMKII signalling, which leads to enhanced phosphorylation of TRPV1, increased TRPV1-eNOS complex formation, eNOS activation and, ultimately, NO production.


Life Sciences | 2009

Resistin increases lipid accumulation by affecting class A scavenger receptor, CD36 and ATP-binding cassette transporter-A1 in macrophages

Tzong-Shyuan Lee; Chun-Yueh Lin; Jin-Yi Tsai; Yuh-Lin Wu; Kuo-Hui Su; Kuo-Yun Lu; Sheng-Huang Hsiao; Ching-Chian Pan; Yu Ru Kou; Yung-Pei Hsu; Low-Tone Ho

AIMS Resistin promotes macrophage-foam cell formation, but the mechanisms are unclear. In macrophages, lipid uptake is regulated by scavenger receptors (SR-A and CD36), while the cholesterol efflux is regulated by SR-BI, ATP-binding cassette transporter-A1 (ABCA1) and ABCG1. We investigated the mechanisms underlying the dysregulation by resistin of these regulators leading to promotion of lipid accumulation in bone marrow-derived macrophages. MAIN METHODS Western blotting, real-time PCR and oil red O staining were performed. KEY FINDINGS Resistin exacerbated lipid accumulation in oxLDL-treated macrophages. Resistin treatment of oxLDL-untreated macrophages showed increased SR-A and CD36 mRNA and protein levels, and decreased ABCA1 protein level, while having no effect on SR-BI or ABCG1 expression. Up-regulation of SR-A and CD36 by resistin resulted from activation of AP-1 and PPARgamma, respectively, and this was confirmed by the lack of activation of either after AP-1 inhibition using curcumin or SP600125, or PPARgamma inhibition using GW9662, respectively. The down-regulation of ABCA1 by resistin was not accompanied by a reduced mRNA level or an activation of LXRalpha/RXR, but resulted from enhanced protein degradation as revealed by the abolition of the down-regulation after inhibition of the proteasome pathway using ALLN or MG-132. A combined inhibition by SP600125, GW9662 and ALLN prevented resistin-induced exacerbation of lipid accumulation in oxLDL-treated macrophages. SIGNIFICANCE Resistin promotes foam cell formation via dysregulation of SR-A, CD36 and ABCA1. SR-A and CD36 are transcriptionally up-regulated by resistin through AP-1 and PPARgamma, respectively, whereas ABCA1 is down-regulated by resistin through proteasome-mediated enhancement of protein degradation.


Circulation | 2010

Erythropoietin Suppresses the Formation of Macrophage Foam Cells: Role of Liver X Receptor α

Kuo-Yun Lu; Li-Chieh Ching; Kuo-Hui Su; Yuan-Bin Yu; Yu Ru Kou; Sheng-Huang Hsiao; Yu-Chu Huang; Chien-Yu Chen; Li-Ching Cheng; Ching-Chian Pan; Tzong-Shyuan Lee

Background— In addition to the hematopoietic effect of erythropoietin, increasing evidence suggests that erythropoietin also exerts protective effects for cardiovascular diseases. However, the role of erythropoietin and its underlying mechanism in macrophage foam cell formation are poorly understood. Methods and Results— Compared with wild-type specimens, erythropoietin was increased in atherosclerotic aortas of apolipoprotein E–deficient (apoE−/−) mice, mainly in the macrophage foam cells of the lesions. Erythropoietin levels in culture medium and macrophages were significantly elevated in response to oxidized low-density lipoprotein in a dose-dependent manner. Furthermore, erythropoietin markedly attenuated lipid accumulation in oxidized low-density lipoprotein–treated macrophages, a result that was due to an increase in cholesterol efflux. Erythropoietin treatment significantly increased ATP-binding cassette transporters (ABC) A1 and ABCG1 mRNA and protein levels without affecting protein expression of scavenger receptors, including scavenger receptor-A, CD36, and scavenger receptor-BI. The upregulation of ABCA1 and ABCG1 by erythropoietin resulted from liver X receptor &agr; activation, which was confirmed by its prevention on expression of ABCA1 and ABCG1 after pharmacological or small interfering RNA inhibition of liver X receptor &agr;. Moreover, the erythropoietin-mediated attenuation on lipid accumulation was abolished by such inhibition. Finally, reduced lipid accumulation and marked increase in ABCA1 and ABCG1 were demonstrated in erythropoietin-overexpressed macrophages. Conclusion— Our data suggest that erythropoietin suppresses foam cell formation via the liver X receptor &agr;–dependent upregulation of ABCA1 and ABCG1.


Cardiovascular Research | 2010

EGb761 ameliorates the formation of foam cells by regulating the expression of SR-A and ABCA1: role of haem oxygenase-1

Jin-Yi Tsai; Kuo-Hui Su; Song-Kun Shyue; Yu Ru Kou; Yuan-Bin Yu; Sheng-Huang Hsiao; An-Na Chiang; Yuh-Lin Wu; Li-Chieh Ching; Tzong-Shyuan Lee

AIMS Accumulation of foam cells in the intima is a hallmark of early-stage atherosclerotic lesions. Ginkgo biloba extract (EGb761) has been reported to exert anti-oxidative and anti-inflammatory properties in atherosclerosis, yet the significance and the molecular mechanisms of action of EGb761 in the formation of macrophage foam cells are not fully understood. METHODS AND RESULTS Treatment with EGb761 resulted in a dose-dependent decrease in oxidized low-density lipoprotein (oxLDL)-mediated cholesterol accumulation in macrophages, a consequence that was due to a decrease in cholesterol uptake and an increase in cholesterol efflux. Additionally, EGb761 significantly down-regulated the mRNA and protein expression of class A scavenger receptor (SR-A) by decreasing expression of activator protein 1 (AP-1); however, EGb761 increased the protein stability of ATP-binding cassette transporter A1 (ABCA1) by reducing calpain activity without affecting ABCA1 mRNA expression. Small interfering RNA (siRNA) targeting haem oxygenase-1 (HO-1) abolished the EGb761-induced protective effects on the expression of AP-1, SR-A, ABCA1, and calpain activity. Accordingly, EGb761-mediated suppression of lipid accumulation in foam cells was also abrogated by HO-1 siRNA. Moreover, the lesion size of atherosclerosis was smaller in EGb761-treated, apolipoprotein E-deficient mice compared with the vehicle-treated mice, and the expression of HO-1, SR-A, and ABCA1 in aortas was modulated similar to that observed in macrophages. CONCLUSION These findings suggest that EGb761 confers a protection from the formation of foam cells by a novel HO-1-dependent regulation of cholesterol homeostasis in macrophages.


Cardiovascular Research | 2009

Valsartan regulates the interaction of angiotensin II type 1 receptor and endothelial nitric oxide synthase via Src/PI3K/Akt signalling

Kuo-Hui Su; Jin-Yi Tsai; Yu Ru Kou; An-Na Chiang; Sheng-Huang Hsiao; Yuh-Lin Wu; Hsin-Han Hou; Ching-Chian Pan; Song-Kun Shyue; Tzong-Shyuan Lee

AIMS Valsartan, a selective angiotensin II type 1 receptor (AT1R) blocker, has beneficial effects in the cardiovascular system in part by its increase of nitric oxide (NO) bioavailability, yet the mechanisms are unclear. We investigated the molecular mechanisms underlying this effect in endothelial cells (ECs). METHODS AND RESULTS NO production was examined by Griess reagent assay, DAF-2 DA fluorescence staining and cGMP ELISA kits. Protein interaction was determined by western blotting and immunoprecipitation. Treating bovine or human aortic ECs with valsartan increased NO production, as evidenced by elevated level of stable NO metabolites and intracellular cGMP. Valsartan increased the phosphorylation but not the protein level of endothelial NO synthase (eNOS). Inhibition of phosphoinositide-3 kinase (PI3K)/Akt and Src pathways by specific inhibitors suppressed valsartan-induced NO release. In addition, valsartan increased the tyrosine residue phosphorylation of AT1R, which was attenuated by inhibition of Src but not PI3K activities. Valsartan also suppressed the interaction of eNOS and AT1R, which was blocked by Src or PI3K inhibition. CONCLUSION Valsartan-induced NO production in ECs is mediated through Src/PI3K/Akt-dependent phosphorylation of eNOS. Valsartan-induced AT1R phosphorylation depends on Src but not PI3K, whereas valsartan-induced suppression of AT1R-eNOS interaction depends on Src/PI3K/Akt signalling. These results indicate a novel vasoprotective mechanism of valsartan in upregulating NO production in ECs.


Molecular Medicine | 2012

Implication of AMP-Activated Protein Kinase in Transient Receptor Potential Vanilloid Type 1-Mediated Activation of Endothelial Nitric Oxide Synthase

Li-Chieh Ching; Chien-Yu Chen; Kuo-Hui Su; Hsin-Han Hou; Song-Kun Shyue; Yu Ru Kou; Tzong-Shyuan Lee

We investigated whether AMP-activated protein kinase (AMPK), a multifunctional regulator of energy homeostasis, is involved in transient receptor potential vanilloid type 1 (TRPV1)-mediated activation of endothelial nitric oxide synthase (eNOS) in endothelial cells (ECs) and mice. In ECs, treatment with evodiamine, the activator of TRPV1, increased the phosphorylation of AMPK, acetyl-CoA carboxylase (ACC) and eNOS, as revealed by Western blot analysis. Inhibition of AMPK activation by compound C or dominant-negative AMPK mutant abrogated the evodiamine-induced increase in phosphorylation of AMPK and eNOS and NO bioavailability, as well as tube formation in ECs. Immunoprecipitation and two-hybrid analysis demonstrated that AMPK mediated the evodiamine-induced increase in the formation of a TRPVl-eNOS complex. Additionally, TRPV1 activation by evodiamine increased the phosphorylation of AMPK and eNOS in aortas of wild-type mice but did not activate eNOS in aortas of TRPV1-deficient mice. In mice, inhibition of AMPK activation by compound C markedly decreased evodiamine-evoked angiogenesis in matrigel plugs and in a hind-limb ischemia model. Moreover, evodiamine-induced phosphorylation of AMPK and eNOS in aortas of apolipoprotein E-deficient (ApoE−/−) mice was abrogated in TRPVl-deficient ApoE−/− mice. In conclusion, TRPV1 activation may trigger AMPK-dependent signaling, which leads to enhanced activation of AMPK and eNOS and retarded development of atherosclerosis.


Journal of Cellular Physiology | 2012

AMP-activated protein kinase mediates erythropoietin-induced activation of endothelial nitric oxide synthase†

Kuo-Hui Su; Yuan-Bin Yu; Hsin-Han Hou; Jin-Feng Zhao; Yu Ru Kou; Li-Ching Cheng; Song-Kun Shyue; Tzong-Shyuan Lee

We investigated whether AMP‐activated protein kinase (AMPK), a multi‐functional regulator of energy homeostasis, participates in the regulation of erythropoietin (EPO)‐mediated activation of endothelial nitric oxide synthase (eNOS) in endothelial cells (ECs) and mice. In ECs, treatment with EPO increased the phosphorylation of AMPK, acetyl‐CoA carboxylase (ACC), and eNOS, as revealed by Western blot analysis. Inhibition of AMPK activation by compound C or dominant‐negative AMPK mutant abrogated the EPO‐induced increase in the phosphorylation of AMPK, ACC, and eNOS, as well as nitric oxide (NO) production. Additionally, suppression of AMPK activation abolished EPO‐induced EC proliferation, migration and tube formation. Immunoprecipitation analysis demonstrated that AMPK mediated the EPO‐induced increase in the phosphorylation of β common receptor (βCR) and the formation of a βCR–AMPK–eNOS complex. In mice, inhibition of AMPK activation by compound C markedly decreased EPO‐elicited angiogenesis in Matrigel plugs. Furthermore, the phosphorylation of AMPK and eNOS was significantly higher in aortas from EPO transgenic mice than wild‐type mice. Moreover, treatment with EPO neutralizing antibody greatly reduced the exercise training‐induced increase in phosphorylation of AMPK and eNOS in aortas of wild‐type mice. Taken together, EPO may trigger AMPK‐dependent signaling, which leads to enhanced phosphorylation of βCR and eNOS, increased βCR–AMPK–eNOS complex formation, NO production, and, ultimately, angiogenesis. J. Cell. Physiol. 227: 3053–3062, 2012.


Acta Physiologica | 2014

The essential role of transient receptor potential vanilloid 1 in simvastatin-induced activation of endothelial nitric oxide synthase and angiogenesis

Kuo-Hui Su; Shing-Jong Lin; Jeng Wei; Kuan-I Lee; Jin-Feng Zhao; Song-Kun Shyue; Tzong-Shyuan Lee

We investigated the role of transient receptor potential vanilloid receptor type 1 (TRPV1) in simvastatin‐mediated activation of endothelial nitric oxide synthase (eNOS) and angiogenesis.


Free Radical Biology and Medicine | 2011

α-Lipoic acid ameliorates foam cell formation via liver X receptor α-dependent upregulation of ATP-binding cassette transporters A1 and G1

Li-Ching Cheng; Kuo-Hui Su; Yu Ru Kou; Song-Kun Shyue; Li-Chieh Ching; Yuan-Bin Yu; Yuh-Lin Wu; Ching-Chian Pan; Tzong-Shyuan Lee

α-Lipoic acid (α-LA), a key cofactor in cellular energy metabolism, has protective activities in atherosclerosis, yet the detailed mechanisms are not fully understood. In this study, we examined whether α-LA affects foam cell formation and its underlying molecular mechanisms in murine macrophages. Treatment with α-LA markedly attenuated oxidized low-density lipoprotein (oxLDL)-mediated cholesterol accumulation in macrophages, which was due to increased cholesterol efflux. Additionally, α-LA treatment dose-dependently increased protein levels of ATP-binding cassette transporter A1 (ABCA1) and ABCG1 but had no effect on the protein expression of SR-A, CD36, or SR-BI involved in cholesterol homeostasis. Furthermore, α-LA increased the mRNA expression of ABCA1 and ABCG1. The upregulation of ABCA1 and ABCG1 by α-LA depended on liver X receptor α (LXRα), as evidenced by an increase in the nuclear levels of LXRα and LXRE-mediated luciferase activity and its prevention of the expression of ABCA1 and ABCG1 after inhibition of LXRα activity by the pharmacological inhibitor geranylgeranyl pyrophosphate (GGPP) or knockdown of LXRα expression with small interfering RNA (siRNA). Consistently, α-LA-mediated suppression of oxLDL-induced lipid accumulation was abolished by GGPP or LXRα siRNA treatment. In conclusion, LXRα-dependent upregulation of ABCA1 and ABCG1 may mediate the beneficial effect of α-LA on foam cell formation.

Collaboration


Dive into the Kuo-Hui Su's collaboration.

Top Co-Authors

Avatar

Tzong-Shyuan Lee

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yu Ru Kou

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Li-Chieh Ching

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Yuan-Bin Yu

Taipei Veterans General Hospital

View shared research outputs
Top Co-Authors

Avatar

An-Na Chiang

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Ching-Chian Pan

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Jin-Feng Zhao

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Jin-Yi Tsai

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Sheng-Huang Hsiao

National Yang-Ming University

View shared research outputs
Researchain Logo
Decentralizing Knowledge