Network


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

Hotspot


Dive into the research topics where Yi-Min Hsiao is active.

Publication


Featured researches published by Yi-Min Hsiao.


Molecular Carcinogenesis | 2006

Transcriptionally Mediated Inhibition of Telomerase of Fungal Immunomodulatory Protein From Ganoderma tsugae in A549 Human Lung Adenocarcinoma Cell Line

Chien-Huang Liao; Yi-Min Hsiao; Chung-Ping Hsu; Meei-Yn Lin; James Chun-Huan Wang; Yu-Lu Huang; Jiunn-Liang Ko

Telomerase expression is the hallmark of tumor cells, and activation of this ribonucleoprotein complex may be a rate‐limiting or critical step in cellular immortalization and oncogenesis. Fungal immunomodulatory protein, FIP‐gts, has been isolated from Ganoderma tsugae. In the present study, we expressed and purified the recombinant fungal immunomodulatory protein reFIP‐gts in E. coli. We found that reFIP‐gts significantly and selectively inhibits the growth of A549 cancer cells while not affecting the growth of normal MRC‐5 fibroblasts. The reFIP‐gts suppression of telomerase activity is concentration‐dependent, due to the downregulation of the telomerase catalytic subunit (hTERT). It also happens at the mRNA level. These results were confirmed by transient transfections of A549 cells with pGL3‐Basic plasmid constructs containing the functional hTERT promoter and its E‐box‐deleted sequences cloned upstream of a luciferase reporter gene. With electrophoretic mobility shift assays and Western blotting, we demonstrated that in response to reFIP‐gts, binding of c‐myc transcriptional factor to the E‐box sequence on the hTERT promoter is inhibited. These results show that reFIP‐gts suppresses telomerase activity and inhibits transcriptional regulation of hTERT via a c‐myc‐responsive element‐dependent mechanism. Our findings provide new insight into both the anticancer function of reFIP‐gts and the regulation of hTERT/telomerase expression, which may be valuable in the development of a promising chemopreventive agent.


Food and Chemical Toxicology | 2008

Induction of premature senescence in human lung cancer by fungal immunomodulatory protein from Ganoderma tsugae.

Chien-Huang Liao; Yi-Min Hsiao; Ching-Hsiung Lin; Chin-Shui Yeh; James Chun-Huan Wang; Chia-Hung Ni; Chung-Ping Hsu; Jiunn-Liang Ko

Purified recombinant fungal immunomodulatory protein from Ganoderma tsugae (reFIP-gts) has anti-telomerase effects in human lung adenocarcinoma A549 cells. However, how reFIP-gts affects cancer cell fates remains unclear. Here, we demonstrated that reFIP-gts-treated lung cancer cells are arrested at G1 phase by flow cytometry and possess morphological phenotype consistent with cellular senescence. The senescent nature of these cells was supported by positive staining for senescence-associated beta-galactosidase activity and increased lysosomal content in A549 and CaLu-1 lung cancer cells. Arrest of cells at G1 appears to be the key means through which reFIP-gts induces premature cellular senescence in A549 cells. Finally, reFIP-gts- treated A549 cells grew more slowly and formed significantly fewer cell colonies in soft agar than untreated A549 cells. In an in vivo mouse model, A549 cells treated with reFIP-gts grew significantly slower than cells treated with PBS alone, confirming that lung tumor can be inhibited by reFIP-gts. The use of reFIP-gts may be a powerful new strategy for chemoprevention and antineoplastic therapy.


Journal of Agricultural and Food Chemistry | 2010

GMI, a Ganoderma Immunomodulatory Protein, Down-regulates Tumor Necrosis Factor α-Induced Expression of Matrix Metalloproteinase 9 via NF-κB Pathway in Human Alveolar Epithelial A549 Cells

Ching-Hsiung Lin; Yi-Min Hsiao; Chu-Chyn Ou; Yu-Wen Lin; Yi-Lin Chiu; Ko-Huang Lue; Jan-Gowth Chang; Jiunn-Liang Ko

Matrix metalloproteinase 9 (MMP-9) has been implicated in airway injury in chronic obstructive pulmonary disease (COPD), lung inflammation, and lung cancer and plays a major role in tumor necrosis factor-α (TNF-α)-stimulated tumor invasion and lung inflammation. MMP-9 activity is promoted by the pro-inflammatory cytokine TNF-α. GMI, cloned from Ganoderma microsporum and purified, is one of the recombinant fungal immunomodulatory proteins. To understand the molecular mechanisms involved in the suppression of TNF-α-mediated tumor invasion and inflammation, GMI modulation of this pathway was investigated in human alveolar epithelial A549 cells in this study. GMI exhibited an inhibitory effect on TNF-α-induced invasion, with GMI treatment and TNF-α exposure presenting the most anti-invasive properties on Boyden chamber assay. GMI reduced TNF-α-induced MMP-9 activities on gelatin zymography assay through inhibition of MMP-9 transcriptional activity. RT-PCR and MMP-9 promoter luciferase analysis revealed that GMI inhibits the transcription of MMP-9 mRNA. Moreover, in vitro and in vivo binding experiments, an electrophoretic mobility shift assay (EMSA), and chromatin immunoprecipitation assay (ChIP) demonstrated that GMI suppresses DNA binding of nuclear factor (NF)-κB transcription factors to MMP-9 promoter. Western blot analysis indicated that GMI blocks the phosphorylation and degradation of IκBα, which in turn leads to suppression of the phosphorylation and nuclear translocation of p65. Thus, overall, our results indicated that GMI mediates antitumor invasion and anti-inflammatory effects through modulation of NF-κB/MMP-9 pathways.


Journal of Thoracic Oncology | 2010

Genetic Determinants of Pemetrexed Responsiveness and Nonresponsiveness in Non-small Cell Lung Cancer Cells

Ming-Fang Wu; Yi-Min Hsiao; Chuan-Fu Huang; Yu-Hsin Huang; Wan-Jung Yang; Hsiu-Wen Chan; Jinghua Tsai Chang; Jiunn-Liang Ko

Background: Pemetrexed disodium (Alimta®), LY231514, is an antifolate that is able to simultaneously inhibit the synthesis of purines and pyrimidines. Pemetrexed has been approved for first- and second-line treatment in patients with non-small cell lung cancer (NSCLC). However, there is still a lack of clinical biomarkers for predicting the therapeutic response to pemetrexed. The aim of this study is to establish new biomarkers for pemetrexed treatment in NSCLC. Methods: Human NSCLC cell lines were exposed to pemetrexed. The antitumor effect was measured by growth inhibition with MTT assay and expression of cell cycle mediators with immunoblots. Using the Superarray cancer pathway gene array, 482 genes were screened for differential expression in A549 cells that were untreated or treated with pemetrexed. Results: A549 cells exhibited sensitivity but H1355 cells showed resistance to pemetrexed. To investigate the mechanisms of responsiveness and nonresponsiveness to pemetrexed in these cell lines, we measured the expression levels of thymidylate synthase (TS), dihydrofolate reductase (DHFR), reduced folate carrier, and folylpoly-gamma-glutamate synthetase genes. TS, DHFR, and reduced folate carrier gene expressions were significantly reduced in A549 and H1355 cells. Pemetrexed caused cell cycle arrest in the G1 phase and S phase in H1355 and A549 cells, respectively. Significantly higher expressions of many genes, especially lipocalin-2 (Lcn-2) and nm23-H1 proteins, were noted in A549 cells treated with pemetrexed in comparison with untreated cells. Furthermore, reverse transcriptase polymerase chain reaction and Western blot showed that Lcn-2 and nm23-H1 expressions increase in response to pemetrexed treatment in a dose-responsive manner in pemetrexed-sensitive A549 cells but not in resistant H1355 cells. Conclusions: Our results indicated that downregulation of TS and DHFR genes and upregulation of p21, p27, Lcn-2, and nm23-H1 genes may serve as new biomarkers for predicting responsiveness to pemetrexed.


Molecular Pharmaceutics | 2015

GMI, an Immunomodulatory Protein from Ganoderma microsporum, Potentiates Cisplatin-Induced Apoptosis via Autophagy in Lung Cancer Cells.

I-Lun Hsin; Chu-Chyn Ou; Ming-Fang Wu; Ming-Shiou Jan; Yi-Min Hsiao; Ching-Hsiung Lin; Jiunn-Liang Ko

Cisplatin-based therapy is common in the treatment of several types of cancers, including lung cancers. In our previous study, GMI, an immunomodulatory protein cloned from Ganoderma microsporum, induced a cytotoxic effect in lung cancer cells via autophagy. The aim of this study is to examine the role of GMI in enhancing cisplatin-mediated cell death. On the basis of MTT assay and Combination Index, GMI and cisplatin cotreatment induced a synergistic cytotoxic effect. GMI and cisplatin-induced apoptosis was determined by sub-G1, nuclear condensation, and annexin-V/propidium iodide analyses. On Western blot, expressions of γH2AX and cleaved forms of PARP, caspase-3, and caspase-7 were induced by combined treatment. Akt/mTOR pathway activity, LC3-II expression, and acidic vesicular organelle development demonstrated that cisplatin does not abolish GMI-mediated autophagy. Cyto-ID Green/hoechst 33342 double staining and time-dependent experiment indicated that GMI and cisplatin-treated A549 cells simultaneously express autophagosomes and apoptotic nuclei. To elucidate the role of autophagy in inducing apoptosis by GMI and cisplatin, chemical inhibitors and LC3 shRNA were used to inhibit autophagy. The results showed that 3-methyladenine decreases, while chloroquine increases GMI and cisplatin cotreatment-induced cleavage of caspase-7 and PARP. LC3 silencing abolished activation of apoptosis in A549 cells. Caspase inhibitors and caspase-7 silencing mitigated GMI and cisplatin-elicited cell viability inhibition and apoptosis. This is the first study to reveal the novel function of GMI in potentiating cisplatin-mediated apoptosis. GMI and cisplatin induce apoptosis via autophagy/caspase-7-dependent and survivin- and ERCC1-independent pathway. GMI may be a potential cisplatin adjuvant against lung cancer.


Journal of Agricultural and Food Chemistry | 2013

Interruption of Lung Cancer Cell Migration and Proliferation by Fungal Immunomodulatory Protein FIP-fve from Flammulina velutipes

Yu-Chi Chang; Yi-Min Hsiao; Ming-Fang Wu; Chu-Chyn Ou; Yu-Wen Lin; Ko-Huang Lue; Jiunn-Liang Ko

FIP-fve is an immunomodulatory protein isolated from Flammulina velutipes that possesses anti-inflammatory and immunomodulatory activities. However, little is known about its anticancer effects. It is suppressed cell proliferation of A549 lung cancer cells on MTT assay following 48 h treatment of FIP-fve. FIP-fve treatment also resulted in cell cycle arrest but not apoptosis on flow cytometry. This immunomodulatory protein was observed to increase p53 expression, as well as the expression of its downstream gene p21, on Western blot. FIP-fve inhibited migration of A549 cells on wound healing assay and decreased filopodia fiber formation on labeling with Texas Red-X phalloidin. To confirm the effect of FIP-fve on the role of Rac1 in filopodia formation, we investigated the activity of Rac1 in A549 cells following FIP-fve treatment. FIP-fve inhibited EGF-induced activation of Rac1. We demonstrated that FIP-fve decreases RACGAP1 mRNA and protein levels on RT-PCR and Western blot. In addition, the reporter activity of RACGAP1 was reduced by FIP-fve on RacGAP1 promoter assay. Silencing of RacGAP1 decreased cell migration, and overexpression of RacGAP1 increased cell migration in A549 cells. In conclusion, FIP-fve inhibits lung cancer cell migration via RacGAP1 and suppresses the proliferation of A549 via p53 activation pathway.


Journal of Agricultural and Food Chemistry | 2009

FIP-fve stimulates interferon-gamma production via modulation of calcium release and PKC-α activation.

Chu-Chyn Ou; Yi-Min Hsiao; Wen-Jun Wu; Gregory J. Tasy; Jiunn-Liang Ko; Meei-Yn Lin

Fungal immunomodulatory protein, FIP-fve, has been isolated from Flammulina velutipes, and its immunomodulatory effects are believed to be associated with the enhanced activation of IFN-gamma-releasing Th1 cells. However, the mechanisms of FIP-fve-mediated signal transduction in the regulation of interferon-gamma (IFN-gamma) gene expression in human peripheral blood mononuclear cells (PBMCs) are still poorly understood. Using fluo-3 AM, we found that FIP-fve induces a rapid elevation in calcium concentration. ELISA, RT-PCR and Western blot assays demonstrated significant increases in the production and mRNA expression of IFN-gamma and protein kinase C-alpha (PKC-alpha) activation in activated PBMCs, which were abolished by EGTA, nifedipine and GO6976. In conclusion, Ca2+ release and PKC-alpha activation are required for IFN-gamma production induced by FIP-fve in PBMCs.


Mutation Research | 2015

Benzo[a]pyrene-induced cell cycle progression occurs via ERK-induced Chk1 pathway activation in human lung cancer cells

Bing-Yen Wang; Sung-Yu Wu; Sheau-Chung Tang; Chien-Hung Lai; Chu-Chyn Ou; Ming-Fang Wu; Yi-Min Hsiao; Jiunn-Liang Ko

Benzo[a]pyrene (B[a]P) is a potent lung carcinogen derived from tobacco smoking and environmental contamination. This study aimed to investigate the signal transduction pathway responsible for B[a]P-induced non-small cell lung cancer (NSCLC) development. We exposed the human NSCLC cell lines Calu-1, CL3, H1299, CH27, H23, and H1355 to B[a]P and assessed cell cycle progression using flow cytometry. Expression of cell cycle mediators was measured using Western blot analyses and electrophoretic mobility shift assays (EMSAs). B[a]P exposure dramatically induced S-phase accumulation in H1355 cells. Phospho-p53 (Ser15 and Ser20), phospho-ERK, phospho-p38, and Bax were significantly increased in H1355 cells whereas phospho-Rb was decreased in these cells. In addition, B[a]P induced phosphorylation of checkpoint kinase-1 (Chk1) but not Chk2. EMSA experiments revealed a slower migrating band after c-Myc bound the E-box in response to B[a]P treatment, which was abolished upon the addition of the ERK inhibitor PD98059 in H1355 cells. Phospho-ERK inhibition and dominant negative mutant Chk1 expression reversed B[a]P-induced S phase accumulation and downregulated phospho-Chk1 and phospho-ERK expression. Taken together, these results suggest that activation of ERK and its downstream mediator Chk1 may contribute to B[a]P-induced S phase accumulation in H1355 cells. The results could help in the development of lung cancer treatments that target the Chk1 pathway through ERK.


Food and Agricultural Immunology | 2009

Stability of fungal immunomodulatory protein, FIP-gts and FIP-fve, in IFN-γ production.

Chu-Chyn Ou; Yi-Min Hsiao; Wen-Hsien Wang; Jiunn-Liang Ko; Meei-Yn Lin

Abstract Fungal immunomodulatory proteins (FIPs) are a group of novel proteins, purified from medicinal fungi or edible mushrooms that possess immunomodulatory properties. FIP-gts and FIP-fve have been isolated and purified from Ganoderma tsugae and Flammunlina velutipes, respectively. The evaluation of FIP immunomodulatory activity was based on their ability to stimulate human peripheral blood lymphocytes to release interferon-gamma (IFN-γ). We found that FIP-gts exhibited better immunomodulatory activity than FIP-fve. Activities were both greatly reduced with duration of heating. For digestibility, FIP-fve was more resistant than FIP-gts to digestive enzymes in simulated gastric fluid and simulated intestinal fluid. IFN-γ production is only detectable in dimers of FIP-gts as opposed to polymer of FIP-fve. These results suggest that FIP-gts and FIP-fve have activities that are stable and have a strong potential of being applied to food or pharmaceutical products for commercial development.


Bioscience, Biotechnology, and Biochemistry | 2015

Alleviation of Dermatophagoides microceras-induced allergy by an immunomodulatory protein, FIP-fve, from Flammulina velutipes in mice

Yu-Chi Chang; Yi-Min Hsiao; Shao-Chi Hung; Ya-Wen Chen; Chu-Chyn Ou; Wei-Ting Chang; Ko-Huang Lue; Jiunn-Liang Ko

Asthma is a major public health concern. Its greatest risk factor is house dust mite (HDM). Dermatophagoides microceras (Der m) is a type of HDM, and in central Taiwan, there is approximately 80% prevalence of sensitization to Der m. FIP-fve is a fungal immunomodulatory protein (FIP) isolated from the fungus Flammulina velutipes, and exhibits anti-inflammatory properties. To investigate whether FIP-fve affects Der m-induced asthma and inflammation, we evaluated hyper-responsiveness (AHR), pathological changes, and cytokines in mice. We demonstrated that oral FIP-fve decreased Der m-induced airway AHR, airway inflammation, cell infiltration, and expression of cytokines in the bronchoalveolar lavage fluid of Balb/c mice. The results of this study suggest that FIP-fve suppresses asthma, inflammation, and respiratory pathogenesis stimulated by Der m. FIP-fve is able to maintain immunomodulatory activity even in simulated gastric fluid and intestinal fluid. FIP-fve could be a safe and stable agent for suppression of allergic asthma. Graphic Abstract Dermatophagoides Microceras (Der m) induced IL-6, TNF-α and MIP-1α secretion in bronchoalveolar lavage fluid. Der m-induced neutrophils and lung inflammation resulted in asthma. FIP-fve suppressed Der m-induced asthma and inflammation-related cytokines.

Collaboration


Dive into the Yi-Min Hsiao's collaboration.

Top Co-Authors

Avatar

Jiunn-Liang Ko

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Chu-Chyn Ou

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Ming-Fang Wu

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Chien-Huang Liao

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Chung-Ping Hsu

Taipei Veterans General Hospital

View shared research outputs
Top Co-Authors

Avatar

Ko-Huang Lue

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Meei-Yn Lin

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar

Gow-Jen Shieh

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

James Chun-Huan Wang

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Jinghua Tsai Chang

Chung Shan Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge