Network


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

Hotspot


Dive into the research topics where Ming-Fang Wu is active.

Publication


Featured researches published by Ming-Fang Wu.


Autophagy | 2011

GMI, an immunomodulatory protein from Ganoderma microsporum, induces autophagy in non-small cell lung cancer cells

I-Lun Hsin; Chu-Chyn Ou; Tzu-Chin Wu; Ming-Shiou Jan; Ming-Fang Wu; Ling-Yen Chiu; Ko-Huang Lue; Jiunn-Liang Ko

Autophagy is a self-digestive process that degrades the cytoplasmic constituents. Immunomodulatory protein, one major bioactive component of Ganoderma, has antitumor activity. In this study, recombinant fungal immunomodulatory protein, GMI, was cloned from Ganoderma microsporum and purified. We demonstrated that GMI induces lung cancer cell death by activating autophagy, but does not induce apoptotic cell death. On western blot, GMI increased LC3 conversion and decreased p53 expression in a time- and concentration-dependent manner. Cytoplasmic calcium chelator BAPTA-AM was used to prove that GMI promotes autophagy via a calcium-mediated signaling pathway. 3-methyladenine (3-MA), an autophagy inhibitor, enhanced the cytotoxicity of GMI on cell viability assay. Using VZV-G pseudotyped lentivirus-shRNA system for autophagy-related genes silencing, the capabilities of GMI to reduce cell viability and colony formation were abolished in autophagy-defective cells. Furthermore, GMI did not stimulate apoptosis after blocking of autophagy by 3-MA or shRNA knockdown system. In xenograft studies, oral administration of GMI inhibited the tumor growth and induced autophagy significantly in nude mice that had received a subcutaneous injection of A549 cells. This is the first study to reveal the novel function of GMI in activating autophagy. GMI may be a potential chemopreventive agent against non-small cell lung cancer.


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.


Cancer | 2011

Epigenetic mechanisms for silencing glutathione S‐transferase m2 expression by hypermethylated specificity protein 1 binding in lung cancer

Sheau-Chung Tang; Ming-Fang Wu; Ruey-Hong Wong; Yu‐Fan Liu; Lee‐Chun Tang; Chien‐Hung Lai; Chung-Ping Hsu; Jiunn-Liang Ko

Glutathione S‐transferases M2 (GST‐M2) is a detoxifying enzyme. Low expression levels of GST‐M2 have been detected in lung cancer cells. However, little is known about the regulation of GST‐M2 in lung cancer cells. In this study, the authors investigated the epigenetic regulatory mechanisms of GST‐M2 in lung cancer cells.


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 Neurosurgery | 2010

Gamma Knife surgery for recurrent or residual trigeminal neuralgia after a failed initial procedure

Chuan-Fu Huang; Shyh-Ying Chiou; Ming-Fang Wu; Hsien-Tang Tu; Wen-Shan Liu

OBJECT The purpose of this study was to assess outcomes of Gamma Knife surgery (GKS) as a second treatment for recurrent or residual trigeminal neuralgia (TN) after failure of 3 initial procedures: microvascular decompression (MVD), GKS, and percutaneous radiofrequency rhizotomy (PRR). METHODS Between 1999 and 2008, 65 patients (31 men [48%] and 34 women [52%]) with recurrent TN were treated with GKS. All 65 patients had undergone previous medical procedures that failed to achieve sufficient pain relief: 27 patients (42%) had undergone MVD, 8 (12%) had undergone PRR, and 30 (46%) had undergone GKS as the initial treatment. The entry zone of the trigeminal nerve was targeted using a 4-mm collimator and treated with 35-90 Gy. The isocenter was positioned so that the brainstem surface was usually irradiated at an isodose no greater than 20% (59 patients) to 30% (6 patients). The median duration of TN symptoms in these patients was 39 months (range 1-192 months). RESULTS At the clinical evaluation, 42 patients (65%) with idiopathic TN reported successful pain control at a median follow-up point of 64 months (range 18-132 months). Of these patients, 33 (51%) were no longer using medication. At the 1-, 2-, and 3-year follow-up examinations, 74%, 71%, and 66% of patients experienced successful pain control, respectively. There was no significant difference in pain relief in the initial MVD group compared with the initial GKS and initial PRR groups (74% vs 59% and 50%, respectively; p = 0.342). Recurrence of pain was noted in 23 patients. Twelve of these 23 patients underwent another GKS, resulting in pain control in 8 patients (67%); 8 other patients underwent MVD, resulting in pain relief in 7 patients (87.5%). The median time from GKS to pain recurrence was 7 months (range 3-48 months). There was no significant difference in new facial numbness among the 3 groups (p = 0.24); however, in the initial GKS group, facial numbness was significantly associated with freedom from pain (p = 0.0012). There was a significant correlation between the total radiation dose and facial numbness. The cutoff value for facial numbness ranged from 115 to 120 Gy (p = 0.037). CONCLUSIONS Gamma Knife surgery as a second treatment achieved acceptable levels of pain control in 65% of patients with residual or recurrent TN after long-term follow-up. Initial treatment was not a factor that affected pain control, but salvage surgery may be considered separately for each group.


Toxicology and Applied Pharmacology | 2012

Lipocalin 2, a new GADD153 target gene, as an apoptosis inducer of endoplasmic reticulum stress in lung cancer cells.

I-Lun Hsin; Yueh-Chieh Hsiao; Ming-Fang Wu; Ming-Shiou Jan; Sheau-Chung Tang; Yu-Wen Lin; Chung-Ping Hsu; Jiunn-Liang Ko

Endoplasmic reticulum (ER) stress is activated under severe cellular conditions. GADD153, a member of the C/EBP family, is an unfolded protein response (UPR) responsive transcription factor. Increased levels of lipocalin 2, an acute phase protein, have been found in several epithelial cancers. The aim of this study is to investigate the function of lipocalin 2 in lung cancer cells under ER stress. Treatment with thapsigargin, an ER stress activator, led to increases in cytotoxicity, ER stress, apoptosis, and lipocalin 2 expression in A549 cells. GADD153 silencing decreased lipocalin 2 expression in A549 cells. On chromatin immunoprecipitation assay, ER stress increased GADD153 DNA binding to lipocalin 2 promoter. Furthermore, silencing of lipocalin 2 mitigated ER stress-mediated apoptosis in A549 cells. Our findings demonstrated that lipocalin 2 is a new GADD153 target gene that mediates ER stress-induced apoptosis.


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.


Cancer Chemotherapy and Pharmacology | 2012

Regulation of chemosensitivity and migration by clusterin in non-small cell lung cancer cells

Ching-Yuan Cheng; Shur-Hueih Cherng; Wen-Jun Wu; Tsung-Ying Yang; Xin-Yi Huang; Fu-Tien Liao; Ming-Fang Wu; Gwo-Tarng Sheu

PurposeIn terms of drug resistance, cancer cells usually benefit from high clusterin (CLU) expression on chemotherapy. In contrast, CLU expression has been found to be a favorable prognostic factor in lung cancer patients. The aims of this study are to determine the association between CLU expression and chemotherapeutic sensitivity and the potential role of CLU in migration in human non-small-cell lung cancer (NSCLC) cell lines.MethodsThe levels of clusterin in NSCLC cell lines were altered by short hairpin RNA interference (shRNAi) and overexpression on chemosensitivity assay. Migratory ability of these cell lines was also investigated.ResultsH1355 cells with the highest level of CLU demonstrated the lowest sensitivities to Adriamycin (ADR), docetaxel (DOC), and gemcitabine (GEM) treatment. Inhibition of CLU expression in H1355 cells resulted in higher chemosensitivities. When CLU was stably overexpressed in A549 and H1299 cells, only the chemosensitivity to ADR was reduced. The migratory ability of CLU-overexpressing cells significantly decreased. Moreover, MMP2 transcription was inhibited in CLU-overexpressing H1299 cells. These results indicated lower metastatic potential for cancer cells with high CLU level.ConclusionLung cancer cells with high level of CLU have reduced chemosensitivity. High level of CLU may result in migratory inhibition and thus favorable prognosis in lung cancer.


Toxicology in Vitro | 2012

Docetaxel and 5-fluorouracil induce human p53 tumor suppressor gene transcription via a short sequence at core promoter element.

Jui-Chuan Chuang; Gwo-Tarng Sheu; Pi-Chieh Wang; Fu-Tien Liao; Wen-Shan Liu; Chuan-Fu Huang; Mei-Hui Tseng; Ming-Fang Wu

The p53 tumor suppressor protein is involved in cellular defense against agents that can cause genetic damage. Induction of p53 gene expression at transcriptional and post-transcriptional levels by such agents results in p53-regulated gene activation or suppression. Docetaxel (DOC), a member of the taxanes family that is widely used in cancer chemotherapy, activates p53 at the transcriptional level. We demonstrated that p53 is induced by low dose DOC treatment, resulting in MDR-1 gene suppression in human lung cancer cells. To identify the cis-element of p53 promoter that responds to DOC, p53 promoter region was cloned and promoter activity was analyzed on luciferase gene reporter assay. Promoter region (-78 to +129) contained the highest basal p53 promoter activity and deletion of +86 to +129 severely reduced basal promoter activity. Basal promoter region included the 21-bp element (PE21) that determines UV-inducible expression of p53 and mediates DOC-inducible p53 expression. On site-specific mutagenesis of PE21 (-78 to -58), with mutation of ATTG (-62 to -59) to CGGT, completely diminished the response to DOC. The same mutations also inhibited 5-fluorouracil (5-FU)-inducible p53 expression. Our data revealed that a sequence located at PE21 of p53 core promoter regulates p53 induction by chemotherapeutic agents.

Collaboration


Dive into the Ming-Fang Wu's collaboration.

Top Co-Authors

Avatar

Jiunn-Liang Ko

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Gwo-Tarng Sheu

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Yi-Min Hsiao

Central Taiwan University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Chu-Chyn Ou

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Chuan-Fu Huang

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Chung-Ping Hsu

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Fu-Tien Liao

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

I-Lun Hsin

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Ling-Yen Chiu

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Ming-Shiou Jan

Chung Shan Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge