Wen-Yi Tseng
Memorial Hospital of South Bend
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Wen-Yi Tseng.
Joint Bone Spine | 2011
Chang-Fu Kuo; Kuang-Hui Yu; Lai-Chu See; I-Jun Chou; Wen-Yi Tseng; Hsiao-Chun Chang; Yu-Ming Shen; Shue-Fen Luo
OBJECTIVES Accumulating evidence suggests that gout is associated with increased overall and cardiovascular mortality. Using data from a large consecutive cohort of gout sufferers who were followed for 8 years, we investigated mortality among gout sufferers and compared the results with data from the general population of Taiwan. METHODS The study comprised 6631 people who had gout diagnosis at outpatient department of the Chang Gung Memorial Hospital in 2000. Survival status and cause of death were ascertained by examining the National Death Registry of Taiwan through 2008. RESULTS During 53,048 person-years of follow-up in 6631 gout patients, 1273 (male, 941; female, 332) deaths were identified, corresponding to the crude mortality rate was 21.3 (male, 20.0; female, 26.1) per 1000 patient-years, which was significantly higher than that of the national population. As compared with the national population of Taiwan in 2000, the all-cause standardized mortality ratio (95% confidence interval) was 1.29 (1.21-1.37) for men and 1.70 (1.53-1.89) for women. Both men and women in this cohort had higher standardized mortality ratios for death due to kidney diseases (men, 3.10; women, 3.54), endocrine and metabolic diseases (men, 2.24; women, 2.71), and cardiovascular diseases (men, 1.58; women, 1.81). Multivariate Cox regression showed no difference in mortality between genders. DISCUSSION/CONCLUSIONS Individuals with gout have increased mortality risk in both sexes; however, the extent of risk increment is higher in women. Kidney diseases, endocrine and metabolic diseases, and cardiovascular diseases are primary causes of death.
PLOS ONE | 2013
Wen-Yi Tseng; Yi-Shu Huang; Nien-Yi Chiang; Yeh-Pin Chou; Yeong-Jian Jan Wu; Shue-Fen Luo; Chang-Fu Kuo; Ko-Ming Lin; Hsi-Hsien Lin
Higher soluble CD4 (sCD4) levels in serum have been detected in patients of infectious and chronic inflammatory diseases. However, how and why sCD4 is produced remains poorly understood. We establish sensitive ELISA and WB assays for sCD4 detection in conditioned medium of in vitro cell culture system and serum of chronic inflammatory patients. Serum samples from patients with systemic lupus erythematosus (SLE) (n = 79), rheumatoid arthritis (RA) (n = 59), ankylosing spondylitis (AS) (n = 25), gout (n = 31), and normal controls (n = 99) were analyzed using ELISA for sCD4 detection. Results from each assay were analyzed by the Kruskal-Wallis test. Dunn’s multiple comparison post-test was then applied between groups. We confirm that cells expressing exogenous CD4 produce sCD4 in a constitutive and PMA-induced manner. Importantly, sCD4 production in a heterologous expression system is inhibited by GM6001 and TAPI-0, suggesting receptor shedding by matrix metalloproteinase (MMP)-like proteinases. Moreover, similar findings are recapitulated in human primary CD4+ T cells. Finally, we show that serum sCD4 levels are increased in patients of chronic inflammatory diseases including RA and SLE, but not in those with gout. Intriguingly, sCD4 levels in RA patients are correlated positively with the disease activities and higher sCD4 levels seem to associate with poor prognosis. Taken together, we conclude that CD4 is shed from cell surface by a MMP-like sheddase and sCD4 level is closely related with the inflammatory condition in certain chronic diseases. Hence, sCD4 might be considered an important parameter for RA disease progression with potential diagnostic importance.
Schizophrenia Bulletin | 2017
I-Jun Chou; Chang-Fu Kuo; Yu-Shu Huang; Matthew J. Grainge; Ana M. Valdes; Lai-Chu See; Kuang-Hui Yu; Shue-Fen Luo; Lu-Shuang Huang; Wen-Yi Tseng; Weiya Zhang; Michael Doherty
Abstract Strong familial aggregation of schizophrenia has been reported but there is uncertainty concerning the degree of genetic contribution to the phenotypic variance of the disease. This study aimed to examine the familial aggregation and heritability of schizophrenia, and the relative risks (RRs) of other psychiatric diseases, in relatives of people with schizophrenia using the Taiwan National Health Insurance Database. The study population included individuals with affected first-degree or second-degree relatives identified from all beneficiaries (n = 23 422 955) registered in 2013. Diagnoses of schizophrenia made by psychiatrists were ascertained between January 1, 1996 and December 31, 2013. Having an affected co-twin, first-degree relative, second-degree relative, or spouse was associated with an adjusted RR (95% CI) of 37.86 (30.55–46.92), 6.30 (6.09–6.53), 2.44 (1.91–3.12), and 1.88 (1.64–2.15), respectively. Compared with the general population, individuals with one affected first-degree relative had a RR (95% CI) of 6.00 (5.79–6.22) and those with 2 or more had a RR (95% CI) of 14.66 (13.00–16.53) for schizophrenia. The accountability for the phenotypic variance of schizophrenia was 47.3% for genetic factors, 15.5% for shared environmental factors, and 37.2% for non-shared environmental factors. The RR (95% CI) in individuals with a first-degree relative with schizophrenia was 3.49 (3.34–3.64) for mood disorders and 3.91 (3.35–4.57) for delusional disorders. A family history of schizophrenia is therefore associated with a higher risk of developing schizophrenia, mood disorders, and delusional disorders. Heritability and environmental factors each account for half of the phenotypic variance of schizophrenia.
Protein Expression and Purification | 2015
Tai-Yun Yang; Nien-Yi Chiang; Wen-Yi Tseng; Hsiao-Lin Pan; Yen-Ming Peng; Jiann-Jong Shen; Kuo-An Wu; Ming-Ling Kuo; Gin-Wen Chang; Hsi-Hsien Lin
GPR56 is a multi-functional adhesion-class G protein-coupled receptor involved in biological systems as diverse as brain development, male gonad development, myoblast fusion, hematopoietic stem cell maintenance, tumor growth and metastasis, and immune-regulation. Ectodomain shedding of human GPR56 receptor has been demonstrated previously, however the quantitative detection of GPR56 receptor shedding has not been investigated fully due to the lack of appropriate assays. Herein, an efficient system of expression and immune-affinity purification of the recombinant soluble extracellular domain of human GPR56 (sGPR56) protein from a stably transduced human melanoma cell line was established. The identity and functionality of the recombinant human sGPR56 protein were verified by Western blotting and mass spectrometry, and ligand-binding assays, respectively. Combined with the use of two recently generated anti-GPR56 monoclonal antibodies, a sensitive sandwich ELISA assay was successfully developed for the quantitative detection of human sGPR56 molecule. We found that GPR56 receptor shedding occurred constitutively and was further increased in activated human melanoma cells expressing endogenous GPR56. In conclusion, we report herein an efficient system for the production and purification of human sGPR56 protein for the establishment of a quantitative ELISA analysis of GPR56 receptor shedding.
Cytokine | 2018
Wen-Yi Tseng; Yi-Shu Huang; Hsi-Hsien Lin; Shue-Fen Luo; Fiona E. McCann; Kay McNamee; Felix I.L. Clanchy; Richard O. Williams
HighlightsTNFR1 is ubiquitously expressed, whereas TNFR2 is expressed on a restricted range of cell types.TNFR1 and TNFR2 signalling mediate different pathophysiologic effects.TNFR2 signalling may be beneficial for autoimmune disease. ABSTRACT Tumour necrosis factor‐&agr; (TNF‐&agr;) is a highly pleiotropic cytokine with effects on multiple pathological and physiological functions via two distinct receptors, TNFR1 and TNFR2. Much of the pro‐ inflammatory action of TNF‐&agr; is mediated by TNFR1 whereas TNFR2 is thought to play an immunoregulatory and tissue protective role. Anti‐TNF‐ &agr; biologics have been extremely successful in treating a number of immune mediated pathologies, including rheumatoid arthritis, ankylosing spondylitis, psoriasis, psoriatic arthritis and inflammatory bowel disease. However, anti‐TNF therapy has been shown to induce systemic lupus erythematosus and psoriasis in some patients, and to be deleterious in multiple sclerosis. It is hypothesized that these paradoxical effects of anti‐TNF‐&agr; are due to inhibition of TNFR2 signalling. In this review, we will focus on the biology and pathophysiologic role of TNF‐&agr; and on the therapeutic implications of targeting TNF‐&agr; receptor signalling.
Frontiers in Immunology | 2017
Kuan-Yu I; Yi-Shu Huang; Ching-Hsun Hu; Wen-Yi Tseng; Chia-Hsin Cheng; Martin Stacey; Siamon Gordon; Gin-Wen Chang; Hsi-Hsien Lin
EMR2/ADGRE2 is a human myeloid-restricted adhesion G protein-coupled receptor critically implicated in vibratory urticaria, a rare type of allergy caused by vibration-induced mast cell activation. In addition, EMR2 is also highly expressed by monocyte/macrophages and has been linked to neutrophil migration and activation. Despite these findings, little is known of EMR2-mediated signaling and its role in myeloid biology. In this report, we show that activation of EMR2 via a receptor-specific monoclonal antibody promotes the differentiation of human THP-1 monocytic cell line and induces the expression of pro-inflammatory mediators, including IL-8, TNF-α, and MMP-9. Using specific signaling inhibitors and siRNA knockdowns, biochemical and functional analyses reveal that the EMR2-mediated signaling is initiated by Gα16, followed by the subsequent activation of Akt, extracellular signal-regulated kinase, c-Jun N-terminal kinase, and nuclear factor kappa-light-chain-enhancer of activated B cells. Our results demonstrate a functional role for EMR2 in the differentiation and inflammatory activation of human monocytic cells and provide potential targets for myeloid cell-mediated inflammatory disorders.
Joint Bone Spine | 2017
Ping-Han Tsai; Kuang-Hui Yu; I-Jun Chou; Shue-Fen Luo; Wen-Yi Tseng; Lu-Hsiang Huang; Chang-Fu Kuo
OBJECTIVES To determine whether offspring of Taiwanese mothers with systemic lupus erythematosus or rheumatoid arthritis have a higher risk of autism spectrum disorder. METHODS Using the National Health Insurance database and National Birth Registry, we identified a cohort of all live births in Taiwan between 2001 and 2012. Children born to mothers with systemic lupus erythematosus or rheumatoid arthritis were identified and matched with up to 8 controls by maternal age, 1-minute Apgar score, 5-minute Apgar score, mode of delivery, sex of the child, gestational age, birth weight and place of residence. Marginal Cox proportional hazard models were used to estimate relative risk (RR) with 95% confidence intervals (CI) for ASD in offspring. RESULTS Of 1,893,244 newborns, 0.08% (n=1594) were born to systemic lupus erythematosus mothers, and 0.04% (n=673) were born to rheumatoid arthritis mothers. Overall, 5 of 673 (0.74%) offspring of rheumatoid arthritis mothers, 7 of 1594 (0.44%) offspring of systemic lupus erythematosus mothers and 10,631 of 1,893,244 (0.56%) offspring of all mothers developed autism spectrum disorder. Autism spectrum disorder incidence (per 100,000 person-years) was 140.39 (95% CI, 45.58-327.62) for the rheumatoid arthritis group and 76.19 (95% CI, 30.63-156.97) for the systemic lupus erythematosus group. Autism spectrum disorder risk was not significantly higher for children born to mothers with rheumatoid arthritis (HR, 1.42; 95% CI, 0.60-3.40) or systemic lupus erythematosus (HR, 0.76; 95% CI, 0.36-1.59). CONCLUSIONS Children born to women with systemic lupus erythematosus or rheumatoid arthritis do not have a higher risk of autism spectrum disorder.
Annals of the Rheumatic Diseases | 2017
Wen-Yi Tseng; Huang I-S.; Felix I.L. Clanchy; Kay McNamee; D Perocheau; P Ericsson; Sjogren H-O.; Z Xue; L Salford; A Sundstedt; Richard O. Williams
Background Dysfunction of Tregs results in a breakdown of immunological tolerance and has been implicated in the pathogenesis of autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis (RA) and type 1 diabetes. Treg function is regulated by epigenetic factors and we have previously reported the presence of Tregs expressing an aberrant DNA methylation profile in RA. Objectives The aim of this study was to assess the potential utility of DNA methylation inhibitors for the treatment of RA, using collagen-induced arthritis (CIA) as an animal model. Methods DBA/1 mice were immunised with bovine type II collagen emulsified in complete Freunds adjuvant. The mice were treated with zebularine (400 mg/kg), decitabine (1 mg/kg) or psammaplin A (10 mg/kg) for 4 days, starting on the day of arthritis onset. Treatment was then stopped and the disease was monitored up to day 10 of arthritis. The expression of Treg genes was measured in lymph nodes on day 10 by qPCR. To assess the effect of DNA methylation inhibitors on generation of Tregs, naïve CD4+CD25- T cells were cultured with mitomycin C treated APCs plus IL-2, TGFβ and anti-CD3 in the presence or absence of DNA demethylating agents and numbers of CD4+FoxP3+ Tregs were determined by FACS after 72h. Results Treatment with zebularine resulted in a sustained reduction of arthritis severity, accompanied by an increase in the expression of Treg associated genes, Foxp3, Ctla4 and Tgfb1, in draining lymph nodes. Treatment with decitabine produced a more profound reduction in disease severity whereas the therapeutic effect of psammaplin A was more transient. All three DNA methylation inhibitors could convert CD4+CD25- T cells into CD4+FoxP3+ Tregs in a dose-dependent manner in vitro. Conclusions This study has shown that pulse treatment with DNA demethylating drugs produces a sustained reduction in the severity of arthritis and promotes the generation of Tregs. The findings raise the possibility that epigenetic drugs can be used on a short-term basis for re-setting tolerance and boosting Treg responses in human autoimmune disease. Acknowledgements This work was supported by grants from Chang Gung Memorial Hospital (Grant Code: CMRPG2D0251) and Idogen. Disclosure of Interest W. Tseng: None declared, I.-S. Huang: None declared, F. Clanchy: None declared, K. McNamee: None declared, D. Perocheau: None declared, P. Ericsson: None declared, H.-O. Sjogren Shareholder of: Hans Olov Sjogren is a co-founder of Idogen and a co-inventor of a patent on the use of zeburaline for the treatment of autoimmune diseases., Z. Xue: None declared, L. Salford Shareholder of: Leif Salford is a co-founder of Idogen and member of the board of Idogen and a co-inventor of a patent on the use of zeburaline for the treatment of autoimmune diseases, A. Sundstedt Employee of: Anette Sundstedt is an employee of Idogen, R. Williams Consultant for: Dr. Richard Williams is on scientific board of Idogen
Journal of Microbiology Immunology and Infection | 2017
Wen-Yi Tseng; Yeong-Jian Jan Wu; Tai-Yun Yang; Nien-Yi Chiang; Wen-Pin Tsai; Siamon Gordon; Gin-Wen Chang; Chang-Fu Kuo; Shue-Fen Luo; Hsi-Hsien Lin
Journal of Microbiology Immunology and Infection | 2018
Shue-Fen Luo; Chia-Yin Chin; Ling-Jun Ho; Wen-Yi Tseng; Chang-Fu Kuo; Jenn-Haung Lai