Network


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

Hotspot


Dive into the research topics where Jih-Hwa Guh is active.

Publication


Featured researches published by Jih-Hwa Guh.


Oncogene | 2007

YC-1 inhibits HIF-1 expression in prostate cancer cells: Contribution of Akt/NF-κB signaling to HIF-1α accumulation during hypoxia

Hui Lung Sun; Yi Nan Liu; Yun-Ju Huang; Shiow-Lin Pan; Dong-Ming Huang; Jih-Hwa Guh; Fang-Yu Lee; S C Kuo; Che-Ming Teng

Hypoxia-inducible factor 1 (HIF-1), a transcription factor that is critical for tumor adaptation to microenvironmental stimuli, represents an attractive chemotherapeutic target. YC-1 is a novel antitumor agent that inhibits HIF-1 through previously unexplained mechanisms. In the present study, YC-1 was found to prevent HIF-1α and HIF-1β accumulation in response to hypoxia or mitogen treatment in PC-3 prostate cancer cells. Neither HIF-1α protein half-life nor mRNA level was affected by YC-1. However, YC-1 was found to suppress the PI3K/Akt/mTOR/4E-BP pathway, which serves to regulate HIF-1α expression at the translational step. We demonstrated that YC-1 also inhibited hypoxia-induced activation of nuclear factor (NF)-κB, a downstream target of Akt. Two modulators of the Akt/NF-κB pathway, caffeic acid phenethyl ester and evodiamine, were observed to decrease HIF-1α expression. Additionally, overexpression of NF-κB partly reversed the ability of wortmannin to inhibit HIF-1α-dependent transcriptional activity, suggesting that NF-κB contributes to Akt-mediated HIF-1α accumulation during hypoxia. Overall, we identify a potential molecular mechanism whereby YC-1 serves to reduce HIF-1 expression.


Biochemical Pharmacology | 2003

Investigation of anticancer mechanism of thiadiazole-based compound in human non-small cell lung cancer A549 cells.

Jui Yi Chou; Shin Yu Lai; Shiow Lin Pan; Guey Mei Jow; Ji Wang Chern; Jih-Hwa Guh

In this study, we have synthesized several compounds and examined their cytotoxic effects on human non-small cell lung cancer A549 cells. We found that GO-13 ((E,E)-2,5-bis[4-(3-dimethyl-aminopropoxy)styryl]-1,3,4-thiadiazole) is the most effective one by the MTT assay. Furthermore, the GO-13-induced apoptotic reaction was identified based on several criteria, such as negative release reaction of lactate dehydrogenase and positive labeling of annexin V and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) techniques. GO-13 induced the apoptosis in A549 cells in a concentration- and time-dependent manner. The data demonstrate that the regulations of p38 mitogen-activated protein kinase and protein kinase C was not involved in the GO-13-mediated mechanism. However, GO-13 significantly induced a down-regulation of Bcl-X(L) expression in a short-term treatment (less than 3hr), whereas stimulated up-regulation of Bax expression in a long-term treatment (24hr) indicating their involvement in GO-13 action. GO-13-mediated apoptosis is also positively correlated with the increase in caspase-3 activity. Worth noting is the fact that GO-13 did not modify the phosphorylation level of Akt/protein kinase B (PKB) until a 24-hr exposure was carried out indicating that the inhibition of Akt/PKB activation was involved in the late-phase apoptosis. Besides the anticancer activity, GO-13 also showed equivalent anti-angiogenic activity in the nude mice angiogenesis model. In summary, we conclude that GO-13 is the most effective anticancer compound in our screening tests. It induced the early-phase apoptosis in A549 cells via the Bcl-X(L) down-regulation, and that of the late-phase through up-regulation of Bax expression as well as inhibition of Akt/PKB activation.


Journal of Pharmacy and Pharmacology | 1995

Antiplatelet effect of gingerol isolated from Zingiber officinale

Jih-Hwa Guh; Feng-Nien Ko; Ting-Ting Jong; Che-Ming Teng

The purpose of this investigation was to determine the antiplatelet mechanism of gingerol.


Circulation | 1996

Mechanism of Catecholamine-Induced Proliferation of Vascular Smooth Muscle Cells

Sheu-Meei Yu; Sheng-Yi Tsai; Jih-Hwa Guh; Feng-Nien Ko; Che-Ming Teng; Jonathan T. Ou

BACKGROUND Catecholamines have been shown to aggravate atherosclerosis in animals and humans, and abnormal proliferation of vascular smooth muscle cells (VSMC) is a key event in the early stage of atherosclerosis. Catecholamines may be involved in such cell growth. Therefore, a series of experiments using cultured VSMC was performed to elucidate their possible mitogenic effect. METHODS AND RESULTS We examined the mitogenic effect of catecholamines using rat aortic smooth muscle cells (VSMC) by measuring [3H]thymidine incorporation, checking with flow cytometry, and counting the cell number directly. Furthermore, the catecholamine-activated signal transduction pathway was assessed by measurement of the formation of inositol 1, 4, 5-triphosphate, intracellular Ca2+ concentration, mitogen-activated protein kinase (MAPK) activity, and mitogenic gene expression. Norepinephrine (NE) and phenylephrine stimulated [3H]thymidine incorporation and cell growth. Clonidine and isoproterenol showed little of such effects. Prazosin was more effective than either yohimbine or propranolol in suppressing the mitogenic effect of NE, indicating that catecholamine-induced VSMC proliferation is mediated by alpha 1-adrenoceptors. The alpha 1-adrenoceptor activation was coupled to pertussis toxin-insensitive Gq-protein and triggered phosphoinositide hydrolysis with subsequent activation of protein kinase C and MAPK in VSMC. In response to NE, both 42- and 44-kD MAPK were activated and tyrosine was phosphorylated. alpha 1-Adrenoceptor stimulation with NE also caused accumulation of c-fos, c-jun, and c-myc mRNA. Chloroethylclonidine completely blocked the alpha 1-adrenoceptor-mediated mitogenesis. CONCLUSIONS The effect of catecholamines appears to be mediated via the activation of the chloroethylclonidine-sensitive alpha 1-adrenoceptors that triggers the phosphoinositide hydrolysis and activates the MAPK pathway, leading to DNA synthesis and cell proliferation.


The Journal of Urology | 2001

DUAL EFFECTS OF OUABAIN ON THE REGULATION OF PROLIFERATION AND APOPTOSIS IN HUMAN PROSTATIC SMOOTH MUSCLE CELLS

Shih-Chieh Chueh; Jih-Hwa Guh; Jun Chen; Ming-Kuen Lai; Che-Ming Teng

PURPOSE To elucidate the role of ouabain in the pathophysiology of benign prostatic hyperplasia we examined the effects of ouabain on the proliferation and apoptosis of human prostatic smooth muscle cells. MATERIALS AND METHODS Primary cultures of human prostatic smooth muscle cells were obtained from 7 patients with bladder outlet obstruction caused by benign prostatic enlargement. A cell proliferation study was performed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay method to examine the effects of different concentrations of ouabain and various inhibitors. Western blot analysis was done to determine mitogen activated protein kinase (MAPK) activation. The terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling reaction method and caspase-3 activity assay were also performed to examine the apoptotic mechanism. RESULTS Ouabain exhibited a modest but significant proliferative effect in nanomolar concentrations; whereas it induced cell apoptosis at higher concentrations. Ouabain caused rapid activation of p42/44 MAPKs. The proliferative effect of ouabain was completely flattened by W-7 and MAPK kinase (MEK) inhibitor, suggesting the requirement of Ca(2+) mobilization and the involvement of the MEK-p42/44 MAPK cascade. The cytotoxic effect by ouabain was defined as apoptosis and necrosis using the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling reaction technique and lactate dehydrogenase release assay, respectively. In addition, ouabain induced profound caspase-3 activity in the cytotoxic concentrations and DEVD-CHO reversed the cytotoxic action to ouabain, demonstrating the involvement of caspase-3 activation in the cytotoxic action. CONCLUSIONS Ouabain at different concentrations caused dual effects on proliferation and apoptosis in human prostatic smooth muscle cells. At low concentrations ouabain promoted cell proliferation via a Ca(2+) dependent mechanism and activation of the MEK-p42/44 MAPK pathway; whereas it induced cell apoptosis via the activation of caspase-3 activity at higher concentrations.


Journal of Biological Chemistry | 2011

Targeting Energy Metabolic and Oncogenic Signaling Pathways in Triple-negative Breast Cancer by a Novel Adenosine Monophosphate-activated Protein Kinase (AMPK) Activator

Kuen Haur Lee; En-Chi Hsu; Jih-Hwa Guh; Hsiao Ching Yang; Dasheng Wang; Samuel K. Kulp; Charles L. Shapiro; Ching-Shih Chen

Background: Adenosine monophosphate-activated protein kinase (AMPK) modulates cancer cell metabolism and survival. Results: The novel compound OSU-53 directly activates AMPK, inhibits multiple metabolic and oncogenic pathways, and induces apoptosis in triple-negative breast cancer cells. Conclusion: OSU-53 acts through a broad spectrum of antitumor activities downstream of AMPK activation. Significance: OSU-53 is a potent small molecule AMPK activator with translational potential for breast cancer therapy. The antitumor activities of the novel adenosine monophosphate-activated protein kinase (AMPK) activator, OSU-53, were assessed in in vitro and in vivo models of triple-negative breast cancer. OSU-53 directly stimulated recombinant AMPK kinase activity (EC50, 0.3 μm) and inhibited the viability and clonogenic growth of MDA-MB-231 and MDA-MB-468 cells with equal potency (IC50, 5 and 2 μm, respectively) despite lack of LKB1 expression in MDA-MB-231 cells. Nonmalignant MCF-10A cells, however, were unaffected. Beyond AMPK-mediated effects on mammalian target of rapamycin signaling and lipogenesis, OSU-53 also targeted multiple AMPK downstream pathways. Among these, the protein phosphatase 2A-dependent dephosphorylation of Akt is noteworthy because it circumvents the feedback activation of Akt that results from mammalian target of rapamycin inhibition. OSU-53 also modulated energy homeostasis by suppressing fatty acid biosynthesis and shifting the metabolism to oxidation by up-regulating the expression of key regulators of mitochondrial biogenesis, such as a peroxisome proliferator-activated receptor γ coactivator 1α and the transcription factor nuclear respiratory factor 1. Moreover, OSU-53 suppressed LPS-induced IL-6 production, thereby blocking subsequent Stat3 activation, and inhibited hypoxia-induced epithelial-mesenchymal transition in association with the silencing of hypoxia-inducible factor 1a and the E-cadherin repressor Snail. In MDA-MB-231 tumor-bearing mice, daily oral administration of OSU-53 (50 and 100 mg/kg) suppressed tumor growth by 47–49% and modulated relevant intratumoral biomarkers of drug activity. However, OSU-53 also induced protective autophagy that attenuated its antiproliferative potency. Accordingly, cotreatment with the autophagy inhibitor chloroquine increased the in vivo tumor-suppressive activity of OSU-53. OSU-53 is a potent, orally bioavailable AMPK activator that acts through a broad spectrum of antitumor activities.


British Journal of Pharmacology | 1994

(−)‐Discretamine, a selective α1D‐adrenoceptor antagonist, isolated from Fissistigma glaucescens

Feng‐Nien ‐N Ko; Jih-Hwa Guh; Sheu‐Meei ‐M Yu; Yu‐Sheng ‐S Hou; Yang‐Chang ‐C Wu; Che-Ming Teng

1 The selectivity of (−)‐discretamine for α1‐adrenoceptor subtypes was investigated by use of functional and binding studies in rat vas deferens, spleen and aorta, and in cultured DDT1MF‐2 and A10 cells. 2 In prostatic portions of rat vas deferens, the competitive antagonists (−)‐discretamine, 5‐methyl‐urapidil (5‐MU) and prazosin inhibited contractions to noradrenaline (NA) with pA2 values of 6.21, 8.71 and 9.27, respectively. The irreversible antagonist, chloroethylclonidine (CEC, 100 μm) failed to affect contractions to NA while nifedipine (1 μm) blocked them almost completely. 3 In rat spleen, the competitive antagonists (−)‐discretamine, 5‐MU and prazosin inhibited contractions to phenylephrine with pA2 values of 6.44, 7.19 and 9.45, respectively. CEC (100 μm) significantly reduced the maximum contraction to phenylephrine while nifedipine (1 μm) did not affect it. 4 In rat aorta, the competitive antagonists (−)‐discretamine, 5‐MU and prazosin inhibited contractions to NA with pA2 values of 7.60, 8.00 and 9.40, respectively. CEC also antagonized the contractions to NA in a competitive manner with a pA2 value of 6.10. 5 The specific binding of [3H]‐prazosin to DDT1MF‐2 and A10 cells was concentration‐dependent and saturated at 3–5 nM with KD values of 0.24 ± 0.02 and 0.20 ± 0.02 nM, respectively. (−)‐Discretamine, 5‐MU, CEC and prazosin inhibited specific [3H]‐prazosin binding to DDT1MF‐2 and A10 cells in a concentration‐dependent manner with IC50 values of 390.8 ± 20.6, 43.6 ± 3.9, 200.0 ± 30.0 and 0.8 ± 0.1 nM, respectively in DDT1MF‐2 cells, and 25.0 ± 3.2, 8.6 ± 1.4, 1000.0 ± 30.8 and 0.52 ± 0.03 nM, respectively in A10 cells. 6 Pretreatment of A10 cells with CEC (10 μm) for 30 min and then washed out thoroughly, reduced specific [3H]‐prazosin binding by 30%. The CEC‐insensitive [3H]‐prazosin binding was inhibited by (−)‐discretamine with an IC50 value of 7.0 ± 0.3 nM. 7 5‐MU (100 nM), CEC (1 μm) and prazosin (10 nM) markedly inhibited NA (3 μm)‐induced [3H]‐inositol monophosphate formation in DDT1MF‐2 and A10 cells, while (−)‐discretamine (100 nM) inhibited NA‐induced [3H]‐inositol monophosphate formation only in A10 cells. 8 In conclusion, (−)‐discretamine is a selective α1D‐adrenoceptor antagonist in vascular smooth muscle. Its selectivity among various α1‐adrenoceptor subtypes is α1A:α1B:α1D = 0.04:0.07:1.0.


Biochemical Pharmacology | 2003

Esculetin inhibits Ras-mediated cell proliferation and attenuates vascular restenosis following angioplasty in rats

Shiow Lin Pan; Ying Wen Huang; Jih-Hwa Guh; Ya Ling Chang; Chieh Yu Peng; Che-Ming Teng

The proliferation of vascular smooth muscle cells (VSMCs) induced by injury to the intima of arteries is an important etiologic factor in vascular proliferative disorders such as atherosclerosis and restenosis. Esculetin, derived from the Chinese herb Artemisia scoparia, is well known as a lipoxygenase inhibitor. We have investigated the inhibitory effects of esculetin on VSMC proliferation and intimal hyperplasia by balloon angioplasty in the rat. We determined, using [3H]thymidine incorporation and the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, that esculetin inhibited the proliferation of VSMCs via a lipoxygenase-independent pathway. Three predominant signaling pathways were identified to be inhibited by esculetin: (a) the activation of p42/44 mitogen-activated protein kinase (MAPK) and the downstream effectors of c-fos and c-jun immediate early genes by means of western and reverse transcription-polymerase chain reaction (RT-PCR) analyses; (b) the activation of nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1), using the electrophoretic mobility shift assay; and (c) the activation of phosphoinositide 3-kinase (PI 3-kinase) and cell cycle progression, by western blot analysis and flow cytometric detection. Furthermore, esculetin also profoundly inhibited Ras activation, a shared upstream event of the above signaling cascades. In vascular injury studies, intraperitoneal administration of esculetin significantly suppressed intimal hyperplasia induced by balloon angioplasty. We conclude that esculetin blocks cell proliferation via the inhibition of an upstream effector of Ras and downstream events including p42/44 MAPK activation, PI 3-kinase activation, immediate early gene expression, as well as NF-kappaB and AP-1 activation. It also inhibits intimal hyperplasia after balloon vascular injury in the rat, indicating the therapeutic potential for treating restenosis after arterial injury.


European Journal of Pharmaceutical Sciences | 2003

Pharmacological evaluation of several major ingredients of Chinese herbal medicines in human hepatoma Hep3B cells

C.C. Chou; Shiow-Lin Pan; Che-Ming Teng; Jih-Hwa Guh

Long-dan-tan (Chinese name) is one of the most common herbal medicines used by Chinese people with chronic liver disease. Accumulated anecdotal evidence suggests that Long-dan-tan may show a beneficial effect in patients with hepatocellular carcinoma. Long-dan-tan is made from five plants: Gentiana root, Scutellaria root, Gardenia fruit, Alisma rhizome, and Bupleurum root. In this study, we have examined the cytotoxic effects of the five major ingredients isolated from the above plants, i.e. gentiopicroside, baicalein, geniposide, alisol B acetate and saikosaponin-d, respectively, on human hepatoma Hep3B cells. Annexin V immunofluorescence detection, DNA fragmentation assays and FACScan analysis of propidium iodide-staining cells showed that gentiopicroside, baicalein, and geniposide had little effect, whereas alisol B acetate and saikosaponin-d profoundly induced apoptosis in Hep3B cells. Alisol B acetate, but not saikosaponin-d, induced G2/M arrest of the cell cycle as well as a significant increase in caspase-3 activity. Interestingly, baicalein by itself induced an increase in H(2)O(2) generation and the subsequent NF-kappaB activation; furthermore, it effectively inhibited the transforming growth factor-beta(1) (TGF-beta(1))-induced caspase-3 activation and cell apoptosis. We suggest that alisol B acetate and saikosaponin-d induced cell apoptosis through the caspase-3-dependent and -independent pathways, respectively. Instead of inducing apoptosis, baicalein inhibits TGF-beta(1)-induced apoptosis via increase in cellular H(2)O(2) formation and NF-kappaB activation in human hepatoma Hep3B cells.


Molecular Cancer Therapeutics | 2005

YC-1 suppresses constitutive nuclear factor-κB activation and induces apoptosis in human prostate cancer cells

Yao Ting Huang; Shiow Lin Pan; Jih-Hwa Guh; Ya Ling Chang; Fang Yu Lee; Sheng Chu Kuo; Cheng Ming Teng

Although the indazole compound, YC-1, is reported to exert anticancer activities in several cancer cell types, its target and mechanism of action have not been well explored. The objectives of this study were to ascertain whether YC-1 directly induces apoptosis in prostate cancer cells and to explore the mechanism(s) whereby YC-1 causes cell death. Hormone-refractory metastatic human prostate cancer PC-3 cells were selected for this study. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay indicated that YC-1 suppresses growth of PC-3 cells in a concentration-dependent and time-dependent manner. Apoptosis was determined using 4′,6-diamidino-2-phenylindole staining, and cell cycle progression was examined by FACScan flow cytometry. YC-1 treatment showed chromatin condensation and increased the percentage of PC-3 cells in the hypodiploid sub-G0-G1 phase, indicative of apoptosis. Additionally, exposure to YC-1 was found to induce activation of caspase-3 and cleavage of poly(ADP-ribose) polymerase. Translocation and activation of nuclear factor-κB (NF-κB) were determined by immunofluorescent staining and ELISA, respectively. The results showed that YC-1 abolished constitutive nuclear translocation and activation of NF-κB/p65. Furthermore, inhibition of inhibitor of κBα (IκBα) phosphorylation and accumulation of IκBα were observed. The antitumor effects of YC-1 were evaluated by measuring the growth of tumor xenografts in YC-1-treated severe combined immunodeficient mice. The volumes of PC-3 tumors produced in severe combined immunodeficient mice were observed to decline significantly after treatment with YC-1 compared with vehicle controls. We concluded that the antitumor effects of YC-1 in PC-3 cells include the induction of apoptosis and the suppression of NF-κB activation. Given these unique actions, further investigations of the effects of YC-1 against hormone-refractory prostate cancer are warranted.

Collaboration


Dive into the Jih-Hwa Guh's collaboration.

Top Co-Authors

Avatar

Che-Ming Teng

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Shiow Lin Pan

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

Jui-Ling Hsu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Shiow-Lin Pan

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Lih-Ching Hsu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Ya-Ching Shen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Shih-Chieh Chueh

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chia-Chun Yu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Ya Ling Chang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Wohn-Jenn Leu

National Taiwan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge