Network


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

Hotspot


Dive into the research topics where Ya Wen Chen is active.

Publication


Featured researches published by Ya Wen Chen.


Islets | 2009

Heavy metals, islet function and diabetes development

Ya Wen Chen; Ching-Yao Yang; Chun Fa Huang; Dong-Zong Hung; Yuk Man Leung; Shing-Hwa Liu

Heavy metals have been known to possess many adverse health effects for a long time. Uncontrolled industrialization breaks out heavy metal pollution in the world. Heavy metal pollutants damage organ functions and disrupt physiological homeostasis. Diabetes mellitus is growing in prevalence worldwide. Several studies have indicated that the deficiency and efficiency of some essential trace metals may play a role in the islet function and development of diabetes mellitus. Some toxic metals have also been shown to be elevated in biological samples of diabetes mellitus patients. In the present work, we review the important roles of heavy metals in islet function and diabetes development in which the in vitro, in vivo, or human evidences are associated with exposure to zinc, arsenic, cadmium, mercury, and nickel. Through this work, we summarize the evidence which suggests that some heavy metals may play an important role in diabetes mellitus as environmental risk factors.


Diabetes | 2006

The Role of Phosphoinositide 3-Kinase/Akt Signaling in Low-Dose Mercury–Induced Mouse Pancreatic β-Cell Dysfunction In Vitro and In Vivo

Ya Wen Chen; Chun Fa Huang; Keh-Sung Tsai; Rong-Sen Yang; Cheng Chieh Yen; Ching-Yao Yang; Shoei Yn Lin-Shiau; Shing-Hwa Liu

The relationship between oxidation stress and phosphoinositide 3-kinase (PI3K) signaling in pancreatic β-cell dysfunction remains unclear. Mercury is a well-known toxic metal that induces oxidative stress. Submicromolar-concentration HgCl2 or methylmercury triggered reactive oxygen species (ROS) production and decreased insulin secretion in β-cell–derived HIT-T15 cells and isolated mouse islets. Mercury increased PI3K activity and its downstream effector Akt phosphorylation. Antioxidant N-acetyl-l-cysteine (NAC) prevented mercury-induced insulin secretion inhibition and Akt phosphorylation but not increased PI3K activity. Inhibition of PI3K/Akt activity with PI3K inhibitor or by expressing the dominant-negative p85 or Akt prevented mercury-induced insulin secretion inhibition but not ROS production. These results indicate that both PI3K and ROS independently regulated Akt signaling–related, mercury-induced insulin secretion inhibition. We next observed that 2- or 4-week oral exposure to low-dose mercury to mice significantly caused the decrease in plasma insulin and displayed the elevation of blood glucose and plasma lipid peroxidation and glucose intolerance. Akt phosphorylation was shown in islets isolated from mercury-exposed mice. NAC effectively antagonized mercury-induced responses. Mercury-induced in vivo effects and increased blood mercury were reversed after mercury exposure was terminated. These results demonstrate that low-dose mercury–induced oxidative stress and PI3K activation cause Akt signaling–related pancreatic β-cell dysfunction.


Toxicology Letters | 2011

Involvement of oxidative stress-mediated ERK1/2 and p38 activation regulated mitochondria-dependent apoptotic signals in methylmercury-induced neuronal cell injury

Tien Hui Lu; Shan Yu Hsieh; Cheng Chien Yen; Hsi Chin Wu; Kuo Liang Chen; Dong-Zong Hung; Chun-Hung Chen; Chin Ching Wu; Yi-Chang Su; Ya Wen Chen; Shing-Hwa Liu; Chun Fa Huang

Methylmercury (MeHg) is well-known for causing irreversible damage in the central nervous system as well as a risk factor for inducing neuronal degeneration. However, the molecular mechanisms of MeHg-induced neurotoxicity remain unclear. Here, we investigated the effects and possible mechanisms of MeHg in the mouse cerebrum (in vivo) and in cultured Neuro-2a cells (in vitro). In vivo study showed that the levels of LPO in the plasma and cerebral cortex significantly increased after administration of MeHg (50μg/kg/day) for 7 consecutive weeks. MeHg could also decrease glutathione level and increase the expressions of caspase-3, -7, and -9, accompanied by Bcl-2 down-regulation and up-regulation of Bax, Bak, and p53. Moreover, treatment of Neuro-2a cells with MeHg significantly reduced cell viability, increased oxidative stress damage, and induced several features of mitochondria-dependent apoptotic signals, including increased sub-G1 hypodiploids, mitochondrial dysfunctions, and the activation of PARP, and caspase cascades. These MeHg-induced apoptotic-related signals could be remarkably reversed by antioxidant NAC. MeHg also increased the phosphorylation of ERK1/2 and p38, but not JNK. Pharmacological inhibitors NAC, PD98059, and SB203580 attenuated MeHg-induced cytotoxicity, ERK1/2 and p38 activation, MMP loss, and caspase-3 activation in Neuro-2a cells. Taken together, these results suggest that the signals of ROS-mediated ERK1/2 and p38 activation regulated mitochondria-dependent apoptotic pathways that are involved in MeHg-induced neurotoxicity.


Archives of Toxicology | 2011

Inorganic arsenic causes cell apoptosis in mouse cerebrum through an oxidative stress-regulated signaling pathway.

Cheng Chien Yen; Tsung Jung Ho; Chin Ching Wu; Chun Fang Chang; Chin Chuan Su; Ya Wen Chen; Tzyy Rong Jinn; Tien Hui Lu; Po-Wen Cheng; Yi-Chang Su; Shing-Hwa Liu; Chun Fa Huang

Arsenic pollution is a major public health problem worldwide. Inorganic arsenic (iAs) is usually more harmful than organic ones. iAs pollution increases the risk of human diseases such as peripheral vascular disease and cancer. However, the toxicological effects of iAs in the brain are mostly unclear. Here, we investigated the toxic effects and possible mechanisms of iAs in the cerebrum of mice after exposure to iAs (0.5 and 5xa0ppm (mg/l) As2O3, via the drinking water), which was the possible human exposed dose via the ingestion in iAs-contaminated areas, for 6xa0consecutive weeks. iAs dose-dependently caused an increase of LPO production in the plasma and cerebral cortex. iAs also decreased the reduced glutathione levels and the expressions of NQO1 and GPx mRNA in the cerebral cortex. These impairments in the cerebral cortex caused by iAs exposure were significantly correlated with the accumulation of As. Moreover, iAs induced the production of apoptotic cells and activation of caspase-3, up-regulation of Bax and Bak, and down-regulation of Mcl-1 in the cerebral cortex. Exposure to iAs also triggered the expression of ER stress-related genes, including GRP78, GRP94, and CHOP. Meanwhile, an increase of p38 activation and dephosphorylation of ERK1/2 were shown in the cerebral cortex as a result of iAs-exposed mice. These iAs-induced damages and apoptosis-related signals could be significantly reversed by NAC. Taken together, these results suggest that iAs-induced oxidative stress causes cellular apoptosis in the cerebrum, signaling of p38 and ERK1/2, and ER stress may be involved in iAs-induced cerebral toxicity.


Toxicology and Applied Pharmacology | 2010

Inorganic mercury causes pancreatic β-cell death via the oxidative stress-induced apoptotic and necrotic pathways

Ya Wen Chen; Chun Fa Huang; Ching-Yao Yang; Cheng Chieh Yen; Keh-Sung Tsai; Shing-Hwa Liu

Mercury is a well-known highly toxic metal. In this study, we characterize and investigate the cytotoxicity and its possible mechanisms of inorganic mercury in pancreatic beta-cells. Mercury chloride (HgCl2) dose-dependently decreased the function of insulin secretion and cell viability in pancreatic beta-cell-derived HIT-T15 cells and isolated mouse pancreatic islets. HgCl2 significantly increased ROS formation in HIT-T15 cells. Antioxidant N-acetylcysteine effectively reversed HgCl2-induced insulin secretion dysfunction in HIT-T15 cells and isolated mouse pancreatic islets. Moreover, HgCl2 increased sub-G1 hypodiploids and annexin-V binding in HIT-T15 cells, indicating that HgCl2 possessed ability in apoptosis induction. HgCl2 also displayed several features of mitochondria-dependent apoptotic signals including disruption of the mitochondrial membrane potential, increase of mitochondrial cytochrome c release and activations of poly (ADP-ribose) polymerase (PARP) and caspase 3. Exposure of HIT-T15 cells to HgCl2 could significantly increase both apoptotic and necrotic cell populations by acridine orange/ethidium bromide dual staining. Meanwhile, HgCl2 could also trigger the depletion of intracellular ATP levels and increase the LDH release from HIT-T15 cells. These HgCl2-induced cell death-related signals could be significantly reversed by N-acetylcysteine. The intracellular mercury levels were markedly elevated in HgCl2-treated HIT-T15 cells. Taken together, these results suggest that HgCl2-induced oxidative stress causes pancreatic beta-cell dysfunction and cytotoxicity involved the co-existence of apoptotic and necrotic cell death.


Journal of Agricultural and Food Chemistry | 2011

Extract of lotus leaf ( Nelumbo nucifera ) and its active constituent catechin with insulin secretagogue activity.

Chun Fa Huang; Ya Wen Chen; Ching-Yao Yang; Hui-Yi Lin; Tzong Der Way; Wenchang Chiang; Shing-Hwa Liu

The effect of lotus leaf ( Nelumbo nucifera Gaertn.) on diabetes is unclear. We hypothesized that lotus leaf can regulate insulin secretion and blood glucose levels. The in vitro and in vivo effects of lotus leaf methanolic extract (NNE) on insulin secretion and hyperglycemia were investigated. NNE increased insulin secretion from β cells (HIT-T15) and human islets. NNE enhanced the intracellular calcium levels in β cells. NNE could also enhance phosphorylation of extracellular signal-regulated protein kinases (ERK)1/2 and protein kinase C (PKC), which could be reversed by a PKC inhibitor. The in vivo studies showed that NNE possesses the ability to regulate blood glucose levels in fasted normal mice and high-fat-diet-induced diabetic mice. Furthermore, the in vitro and in vivo effects of the active constituents of NNE, quercetin, and catechin, on glucose-induced insulin secretion and blood glucose regulation were evaluated. Quercetin did not affect insulin secretion, but catechin significantly and dose-dependently enhanced insulin secretion. Orally administered catechin significantly reversed the glucose intolerance in high-fat-diet-induced diabetic mice. These findings suggest that NNE and its active constituent catechin are useful in the control of hyperglycemia in non-insulin-dependent diabetes mellitus through their action as insulin secretagogues.


Kaohsiung Journal of Medical Sciences | 2011

Arsenic and diabetes: Current perspectives

Chun Fa Huang; Ya Wen Chen; Ching-Yao Yang; Keh-Sung Tsai; Rong-Sen Yang; Shing-Hwa Liu

Arsenic is a naturally occurring toxic metalloid of global concern. Many studies have indicated a dose–response relationship between accumulative arsenic exposure and the prevalence of diabetes mellitus (DM) in arseniasis‐endemic areas in Taiwan and Bangladesh, where arsenic exposure occurs through drinking water. Epidemiological researches have suggested that the characteristics of arsenic‐induced DM observed in arseniasis‐endemic areas in Taiwan and Mexico are similar to those of non‐insulin‐dependent DM (Type 2 DM). These studies analyzed the association between high and chronic exposure to inorganic arsenic in drinking water and the development of DM, but the effect of exposure to low to moderate levels of inorganic arsenic on the risk of DM is unclear. Navas‐Acien etu2009al. recently proposed that a positive association existed between total urine arsenic and the prevalence of Type 2 DM in people exposed to low to moderate levels of arsenic. However, the diabetogenic role played by arsenic is still debated upon. An increase in the prevalence of DM has been observed among residents of highly arsenic‐contaminated areas, whereas the findings from community‐based and occupational studies in low‐arsenic‐exposure areas have been inconsistent. Recently, a population‐based cross‐sectional study showed that the current findings did not support an association between arsenic exposure from drinking water at levels less than 300 μg/L and a significantly increased risk of DM. Moreover, although the precise mechanisms for the arsenic‐induced diabetogenic effect are still largely undefined, recent inu2009vitro experimental studies indicated that inorganic arsenic or its metabolites impair insulin‐dependent glucose uptake or glucose‐stimulated insulin secretion. Nevertheless, the dose, the form of arsenic used, and the experimental duration in the inu2009vivo studies varied greatly, leading to conflicting results and ambiguous interpretation of these data with respect to human exposure to arsenic in the environment. Moreover, the experimental studies were limited to the use of arsenic concentrations much higher than those relevant to human exposure. Further prospective epidemiological studies might help to clarify this controversy. The issues about environmental exposure assessment and appropriate biomarkers should also be considered. Here, we focus on the review of mechanism studies and discuss the currently available evidence and conditions for the association between environmental arsenic exposure and the development of DM.


Toxicology Letters | 2010

Methylmercury chloride induces alveolar type II epithelial cell damage through an oxidative stress-related mitochondrial cell death pathway

Tien Hui Lu; Chun-Hung Chen; Ming Jye Lee; Tsung Jung Ho; Yuk Man Leung; Dong-Zong Hung; Cheng Chien Yen; Tsung Ying He; Ya Wen Chen

Mercury, one of the widespread pollutants in the world, induces oxidative stress and dysfunction in many cell types. Alveolar type II epithelial cells are known to be vulnerable to oxidative stress. Alveolar type II epithelial cells produce and secrete surfactants to maintain morphological organization, biophysical functions, biochemical composition, and immunity in lung tissues. However, the precise action and mechanism of mercury on alveolar type II epithelial cell damage remains unclear. In this study, we investigate the effect and possible mechanism of methylmercury chloride (MeHgCl) on the human lung invasive carcinoma cell line (Cl1-0) and mouse lung tissue. Cl1-0 cells were exposed to MeHgCl (2.5-10 microM) for 24-72 h. The results showed a decrease in cell viability and an increase in malondialdehyde (MDA) level and ROS production at 72 h after MeHgCl exposure in a dose-dependent manner. Caspase-3 activity, sub-G1 contents and annexin-V binding were dramatically enhanced in Cl1-0 cells treated with MeHgCl. MeHgCl could also activate Bax, release cytochrome c, and cleave poly(ADP-Ribose) polymerase (PARP), and decrease surfactant proteins mRNA levels. Moreover, in vivo study showed that mercury contents of blood and lung tissues were significantly increased after MeHgCl treatment in mice. The MDA levels in plasma and lung tissues were also dramatically raised after MeHgCl treatment. Lung tissue sections of MeHgCl-treated mice showed pathological fibrosis as compared with vehicle control. The mRNA levels of proteins in apoptotic signaling, including p53, mdm-2, Bax, Bad, and caspase-3 were increased in mice after exposure to MeHgCl. In addition, the mRNA levels of surfactant proteins (SPs), namely, SP-A, SP-B, SP-C, and SP-D (alveolar epithelial cell functional markers) were significantly decreased. These results suggest that MeHgCl activates an oxidative stress-induced mitochondrial cell death in alveolar epithelial cells.


Toxicology Letters | 2010

Pyrrolidine dithiocarbamate (PDTC)/Cu complex induces lung epithelial cell apoptosis through mitochondria and ER-stress pathways

Ya Wen Chen; Kuo Liang Chen; Chun-Hung Chen; Hsi Chin Wu; Chin Chuan Su; Chin Ching Wu; Tzong Der Way; Dong-Zong Hung; Cheng Chien Yen; Yuan Ting Yang; Tien Hui Lu

Pyrrolidine dithiocarbamate (PDTC) is widely used in pesticides, fungicides, insecticides, and herbicides. Copper (Cu) is a toxic heavy metal in the environment, and an essential trace metal element in the body, which is involved in many biological processes as a catalytic cofactor. The present study is designed to investigate the cellular toxicity of PDTC, CuCl(2), and PDTC/Cu complex exposure in lung alveolar epithelial cells that serve primary structural and functional roles in the lungs. The results showed that PDTC or CuCl(2) alone did not affect cell viability, but PDTC/Cu complex significantly decreased lung alveolar epithelial cell viability. PDTC/Cu complex also significantly increased intracellular copper concentration, but PDTC or CuCl(2) alone had low levels of copper. PDTC/Cu complex dramatically enhanced the JNK protein phosphorylation and ERK protein phosphorylation proteins. PDTC/Cu complex did not affect the p38 protein phosphorylation. PDTC/Cu complex was capable of activating the apoptosis-related caspases including caspase-9, caspase-7, and caspase-3, which could be reversed by the addition of JNK inhibitor SP600125 or transfection of MAPK8 short hairpin RNA. PDTC/Cu complex also increased cytosolic cytochrome c and decreased mitochondrial transmembrane potential. The Bcl-2 mRNA and protein expressions were decreased in lung epithelial cells treated with PDTC/Cu complex, which could be reversed by SP600125. Furthermore, PDTC/Cu complex could trigger the expressions of ER stress-associated signaling molecules including Grp78, Grp94, caspase-12, ATF4, and CHOP, which could be reversed by SP600125. Taken together, these results indicate that exposure to PDTC/Cu complex induces cytotoxicity and apoptosis in alveolar epithelial cells via the mitochondria- and ER-stress-related signaling pathways.


Toxicology | 2011

Nickel(II) induced JNK activation-regulated mitochondria-dependent apoptotic pathway leading to cultured rat pancreatic β-cell death.

Hsi Chin Wu; Ching-Yao Yang; Dong-Zong Hung; Chin Chuan Su; Kuo Liang Chen; Cheng Chieh Yen; Tsung Jung Ho; Yi-Chang Su; Chun Fa Huang; Chun-Hung Chen; Ling Mei Tsai; Ya Wen Chen

Nickel (Ni), a well-known toxic metal, is widely used in electroplating and alloy production. It is also significantly implicated in industrial and environmental pollution caused by uncontrolled industrial and municipal discharges. In this study, we characterized and investigated the cytotoxic effects of Ni exposure and their probable toxicological mechanisms in the pancreatic β-cells. The results showed that it was significantly decreased cell viability after exposing pancreatic β-cell-derived RIN-m5F cells to NiCl(2) for 24h in a dose-dependent manner. NiCl(2) also increased sub-G1 hypodiploid cells and Annexin V-Cy3 binding population in RIN-m5F cells, indicating that it has apoptosis-inducing ability. Moreover, the exposure of RIN-m5F cells to NiCl(2) induced distinct signals of mitochondria-dependent apoptosis, including mitochondrial dysfunction (the disruption of mitochondrial membrane potential (MMP) and increase in mitochondrial cytochrome c release into the cytosol), Bak and Bid mRNA up-regulation, and activation of caspase-3, caspase-7, and caspase-9, and poly(ADP-ribose) polymerase (PARP) degradation. In addition, NiCl(2) also markedly induced the activation of c-Jun N-terminal kinases (JNK), but not of extracellular signal-regulated kinase (ERK)1/2 and p38. These NiCl(2)-induced apoptosis-related signaling responses could be effectively reversed by specific JNK inhibitor SP600125. To the best of our knowledge, this study is the first to show that Ni causes pancreatic β-cell death through a JNK activation-regulated mitochondria-dependent apoptosis-signaling pathway.

Collaboration


Dive into the Ya Wen Chen's collaboration.

Top Co-Authors

Avatar

Chun Fa Huang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Shing-Hwa Liu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Ching-Yao Yang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Keh-Sung Tsai

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Rong-Sen Yang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Cheng Chieh Yen

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Cheng Chien Yen

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo Lin Chen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chia Chi Chuang

National Taiwan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge