Network


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

Hotspot


Dive into the research topics where Jaw Ji Yang is active.

Publication


Featured researches published by Jaw Ji Yang.


Experimental Physiology | 2007

Eccentric cardiac hypertrophy was induced by long‐term intermittent hypoxia in rats

Li Mien Chen; Wei Wen Kuo; Jaw Ji Yang; Shyi Gang P Wang; Yu Lan Yeh; Fuu Jen Tsai; Ying Jui Ho; Mu Hsin Chang; Chih Yang Huang; Shin Da Lee

It is unclear whether cardiac hypertrophy and hypertrophy‐related pathways will be induced by long‐term intermittent hypoxia. Thirty‐six Sprague–Dawley rats were randomly assigned into three groups: normoxia, and long‐term intermittent hypoxia (12% O2, 8 h per day) for 4 weeks (4WLTIH) or for 8 weeks (8WLTIH). Myocardial morphology, trophic factors and signalling pathways in the three groups were determined by heart weight index, histological analysis, Western blotting and reverse transcriptase‐polymerase chain reaction from the excised left ventricle. The ratio of whole heart weight to body weight, the ratio of left ventricular weight to body weight, the gross vertical cross‐section of the heart and myocardial morphological changes were increased in the 4WLTIH group and were further augmented in the 8WLTIH group. In the 4WLTIH group, tumour necrosis factor‐α(TNFα), insulin‐like growth factor (IGF)‐II, phosphorylated p38 mitogen‐activated protein kinase (P38), signal transducers and activators of transcription (STAT)‐1 and STAT‐3 were significantly increased in the cardiac tissues. However, in the 8WLTIH group, in addition to the above factors, interleukin‐6, mitogen‐activated protein kinase (MEK)5 and extracellular signal‐regulated kinase (ERK)5 were significantly increased compared with the normoxia group. We conclude that cardiac hypertrophy associated with TNFα and IGF‐II was induced by intermittent hypoxia. The longer duration of intermittent hypoxia further activated the eccentric hypertrophy‐related pathway, as well as the interleukin 6‐related MEK5–ERK5 and STAT‐3 pathways, which could result in the development of cardiac dilatation and pathology.


Molecular and Cellular Biochemistry | 2009

ZAK induces MMP-2 activity via JNK/p38 signals and reduces MMP-9 activity by increasing TIMP-1/2 expression in H9c2 cardiomyoblast cells

Yi Chang Cheng; Wei Wen Kuo; Hsi Chin Wu; Tung Yuan Lai; Chun Hsien Wu; Jin Ming Hwang; Wang W; Fuu Jen Tsai; Jaw Ji Yang; Chih Yang Huang; Chun Hsien Chu

Leucine-zipper and sterile-alpha motif kinase (ZAK) is the key intra-cellular mediator protein in cardiomyocyte hypertrophy induction by transforming growth factor beta 1 (TGF-β1) which has also been identified as a profibrotic cytokine involved in cardiac fibrosis progression. We hypothesized whether ZAK over-expression causes cardiac scar formation due to the extra-cellular matrix (ECM) degraded enzyme regulation in this paper. Using immuno-histochemical analysis of the human cardiovascular tissue array, we found a positively significant association between ZAK over-expression and myocardial scars. ZAK over-expression in H9c2 cardiomyoblast cells increases the metalloproteinase tissue inhibitor 1/2 (TIMP-1/2) protein level, which reduces matria metalloproteinase-9 (MMP-9) activity and also activates c-JNK N-terminal kinase 1/2 (JNK1/2) and p38 signaling, which induces MMP-2, possibly resulting in cardiac fibrosis. Taken together, ZAK activity inhibition may be a good strategy to prevent the cardiac fibrosis progression.


Journal of Biomedical Science | 2014

A putative novel protein, DEPDC1B, is overexpressed in oral cancer patients, and enhanced anchorage-independent growth in oral cancer cells that is mediated by Rac1 and ERK

Ying Fang Su; Chi Yen Liang; Chih Yang Huang; Chih Yu Peng; Claire Chen; Ming Cheng Lin; Rong Kai Lin; Wei Wen Lin; Ming Yung Chou; Pao Hsin Liao; Jaw Ji Yang

BackgroundThe DEP domain is a globular domain containing approximately 90 amino acids, which was first discovered in 3 proteins: Drosophila disheveled, Caenorhabditis elegans EGL-10, and mammalian Pleckstrin; hence the term, DEP. DEPDC1B is categorized as a potential Rho GTPase-activating protein. The function of the DEP domain in signal transduction pathways is not fully understood. The DEPDC1B protein exhibits the characteristic features of a signaling protein, and contains 2 conserved domains (DEP and RhoGAP) that are involved in Rho GTPase signaling. Small GTPases, such as Rac, CDC42, and Rho, regulate a multitude of cell events, including cell motility, growth, differentiation, cytoskeletal reorganization and cell cycle progression.ResultsIn this study, we found that it was a guanine nucleotide exchange factor and induced both cell migration in a cultured embryonic fibroblast cell line and cell invasion in cancer cell lines; moreover, it was observed to promote anchorage-independent growth in oral cancer cells. We also demonstrated that DEPDC1B plays a role in regulating Rac1 translocated onto cell membranes, suggesting that DEPDC1B exerts a biological function by regulating Rac1. We examined oral cancer tissue; 6 out of 7 oral cancer tissue test samples overexpressed DEPDC1B proteins, compared with normal adjacent tissue.ConclusionsDEPDC1B was a guanine nucleotide exchange factor and induced both cell migration in a cultured embryonic fibroblast cell line and cell invasion in cancer cell lines; moreover, it was observed to promote anchorage-independent growth in oral cancer cells. We also demonstrated that DEPDC1B exerts a biological function by regulating Rac1. We found that oral cancer samples overexpressed DEPDC1B proteins, compared with normal adjacent tissue. Suggest that DEPDC1B plays a role in the development of oral cancer. We revealed that proliferation was linked to a novel DEPDC1B-Rac1-ERK1/2 signaling axis in oral cancer cell lines.


Journal of Biomedical Science | 2009

RhoGDIβ-induced hypertrophic growth in H9c2 cells is negatively regulated by ZAK

Chih Yang Huang; Li Chiu Yang; Kuan Yu Liu; Pao Hsin Liao; Janet Ing-Yuh Chou; Ming Yung Chou; Wei Wen Lin; Jaw Ji Yang

We found that overexpression of RhoGDIβ, a Rho GDP dissociation inhibitor, induced hypertrophic growth and suppressed cell cycle progression in a cultured cardiomyoblast cell line. Knockdown of RhoGDIβ expression by RNA interference blocked hypertrophic growth. We further demonstrated that RhoGDIβ physically interacts with ZAK and is phosphorylated by ZAK in vitro, and this phosphorylation negatively regulates RhoGDIβ functions. Moreover, the ZAK-RhoGDIβ interaction may maintain ZAK in an inactive hypophosphorylated form. These two proteins could negatively regulate one another such that ZAK suppresses RhoGDIβ functions through phosphorylation and RhoGDIβ counteracts the effects of ZAK by physical interaction. Knockdown of ZAK expression in ZAK- and RhoGDIβ-expressing cells by ZAK-specific RNA interference restored the full functions of RhoGDIβ.


International Journal of Medical Sciences | 2017

Tanshinone IIA inhibits β-catenin nuclear translocation and IGF-2R activation via estrogen receptors to suppress angiotensin II-induced H9c2 cardiomyoblast cell apoptosis

Ya Fang Chen; Cecilia Hsuan Day; Nien Hung Lee; Yu Feng Chen; Jaw Ji Yang; Chih Hsueh Lin; Ray Jade Chen; Peramaiyan Rajendran; Vijaya Padma Viswanadha; Chih Yang Huang

Cardiomyopathy involves changes in the myocardial ultra-structure, hypertrophy, apoptosis, fibrosis and inflammation. Angiotensin II (AngII) stimulates the expression of insulin like-growth factors (IGF-2) and IGF-2 receptor (IGF-2R) in H9c2 cardiomyoblasts and subsequently leads to apoptosis. Estrogen receptors protect cardiomyocytes from apoptosis and fibrosis. Tanshinone IIA (TSN), a main active ingredient from Danshen, has been shown to protect cardiomyocytes from death caused by different stress signals. Estrogen receptor α (ER) is required for the rapid activation of the IGF-1R signaling cascade. This study aimed to investigate whether TSN protected H9c2 cardiomyocytes from AngII-induced activation of IGF-2R pathway and hypertrophy via ERs. We found that AngII caused the reduction in IGF-1R phosphorylation and the elevation of β-catenin and IGF-2R levels. This was reversed by increasing doses of TSN and of caspase-3 and ERK1/2 phosphorylation mediated by ERs. The phytoestrogen significantly attenuated AngII-induced apoptosis and suppressed the subsequent cardiac remodeling effect. Therefore, TSN reduced the AngII-induced activation of β-catenin and IGF-2R pathways, apoptosis and cardiac remodeling via ERs in H9c2 cardiomyoblasts.


Cell Biochemistry and Function | 2016

ZAKβ antagonizes and ameliorates the cardiac hypertrophic and apoptotic effects induced by ZAKα

Chien Yao Fu; Wei Wen Kuo; Tsung Jung Ho; Su Ying Wen; Ling Chun Lin; Yan Shen Tseng; Hui Chuan Hung; Vijaya Padma Viswanadha; Jaw Ji Yang; Chih Yang Huang

ZAK (sterile alpha motif and leucine zipper containing kinase AZK), a serine/threonine kinase with multiple biochemical functions, has been associated with various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy. In our previous reports, we found that the activation of ZAKα signaling was critical for cardiac hypertrophy. In this study, we show that the expression of ZAKα activated apoptosis through both a FAS‐dependent pathway and a mitochondria‐dependent pathway by subsequently inducing caspase‐3. ZAKβ, an isoform of ZAKα, is dramatically expressed during cardiac hypertrophy and apoptosis. The interaction between ZAKα and ZAKβ was demonstrated here using immunoprecipitation. The results show that ZAKβ has the ability to diminish the expression level of ZAKα. These findings reveal an inherent regulatory role of ZAKβ to antagonize ZAKα and to subsequently downregulate the cardiac hypertrophy and apoptosis induced by ZAKα.


Environmental Toxicology | 2018

Doxorubicin induces ZAKα overexpression with a subsequent enhancement of apoptosis and attenuation of survivability in human osteosarcoma cells

Chien Yao Fu; Yan Shen Tseng; Ming Cheng Chen; Hsi Hsien Hsu; Jaw Ji Yang; Chuan Chou Tu; Yueh Min Lin; Vijaya Padma Viswanadha; Wei Wen Kuo; Chih Yang Huang

Human osteosarcoma (OS) is a malignant cancer of the bone. It exhibits a characteristic malignant osteoblastic transformation and produces a diseased osteoid. A previous study demonstrated that doxorubicin (DOX) chemotherapy decreases human OS cell proliferation and might enhance the relative RNA expression of ZAK. However, the impact of ZAKα overexpression on the OS cell proliferation that is inhibited by DOX and the molecular mechanism underlying this effect are not yet known. ZAK is a protein kinase of the MAPKKK family and functions to promote apoptosis. In our study, we found that ZAKα overexpression induced an apoptotic effect in human OS cells. Treatment of human OS cells with DOX enhanced ZAKα expression and decreased cancer cell viability while increasing apoptosis of human OS cells. In the meantime, suppression of ZAKα expression using shRNA and inhibitor D1771 both suppressed the DOX therapeutic effect. These findings reveal a novel molecular mechanism underlying the DOX effect on human OS cells. Taken together, our findings demonstrate that ZAKα enhances the apoptotic effect and decreases cell viability in DOX‐treated human OS cells.


Cell Biochemistry and Function | 2018

Overexpression of ZAKβ in human osteosarcoma cells enhances ZAKα expression, resulting in a synergistic apoptotic effect: ZAKβ enhances ZAKα expression, resulting in a synergistic apoptotic effect.

Chien Yao Fu; Yan Shen Tseng; Ming Cheng Chen; Hsi Hsien Hsu; Jaw Ji Yang; Chuan Chou Tu; Yueh Min Lin; Vijaya Padma Viswanadha; Ke Ding; Wei Wen Kuo; Chih Yang Huang

ZAK is a novel mixed lineage kinase‐like protein that contains a leucine‐zipper and a sterile‐alpha motif as a protein‐protein interaction domain, and it is located in the cytoplasm. There are 2 alternatively spliced forms of ZAK: ZAKα and ZAKβ. Previous studies showed that ZAKα is involved in various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy, but the molecular mechanism of ZAKβ is not yet known. In a recent study in our laboratory, we found that ZAKβ can ameliorate the apoptotic effect induced by ZAKα in H9c2 cells. We further hypothesized that ZAKβ could also improve the apoptotic effect induced by ZAKα in human osteosarcoma cells. The results of this study show that ZAKβ can induce apoptosis and decrease cell viability similar to the effects of ZAKα. Interestingly, our ZAKα‐specific inhibitor assay shows that the expression of ZAKβ is highly dependent on ZAKα expression. However, ZAKβ expression effectively induces ZAKα expression and results in synergistic enhancement of apoptosis in human osteosarcoma cells. Furthermore, co‐immunoprecipitation results revealed that ZAKα can directly interact with ZAKβ, and this interaction may contribute to the enhanced apoptotic effects.


Biochemical and Biophysical Research Communications | 2004

Transforming growth factor-β induces the expression of ANF and hypertrophic growth in cultured cardiomyoblast cells through ZAK

Chih Yang Huang; Wei Wen Kuo; Pin Ju Chueh; Chien Tang Tseng; Ming Yung Chou; Jaw Ji Yang


Biochemical and Biophysical Research Communications | 2004

ZAK re-programs atrial natriuretic factor expression and induces hypertrophic growth in H9c2 cardiomyoblast cells.

Chih Yang Huang; Pin Ju Chueh; Chien Tang Tseng; Kuan Yu Liu; Hsiao Ya Tsai; Wei Wen Kuo; Ming Yung Chou; Jaw Ji Yang

Collaboration


Dive into the Jaw Ji Yang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ming Yung Chou

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chien Tang Tseng

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Hsi Hsien Hsu

Mackay Memorial Hospital

View shared research outputs
Top Co-Authors

Avatar

Kuan Yu Liu

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Pao Hsin Liao

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Pin Ju Chueh

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Su Ying Wen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Wei Wen Kuo

China Medical University (PRC)

View shared research outputs
Researchain Logo
Decentralizing Knowledge