Network


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

Hotspot


Dive into the research topics where Ming-Teh Chen is active.

Publication


Featured researches published by Ming-Teh Chen.


Cytotherapy | 2008

Isolation and characterization of mesenchymal stromal cells from human anterior cruciate ligament

Tung-Fu Huang; Chen Yt; Yang Th; Ling-Lan Chen; Shih-Hwa Chiou; Tung-Hu Tsai; Chih-Chien Tsai; Ming-Teh Chen; Hsiao-Li Ma; Shih-Chieh Hung

BACKGROUND The anterior cruciate ligament (ACL) is one of the most commonly injured ligaments of the knee. Because the torn ACL is always discarded during ACL reconstruction, it may be a potential source for isolating mesenchymal stromal cells (MSC). METHODS To characterize MSC from human ACL, cells were enzymatically released from the ACL of adult human donors and seeded in plastic dishes with serial passages at confluence. At different passages, ACL-derived cells were subjected to in vitro assays to investigate their multilineage potential. Upon treatment, the phenotypes of the cell cultures were analyzed by histo- and immunohistochemistry and semi-quantitative reverse transcription-polymerase chain reaction for the expression of lineage-specific genes. RESULTS Six independent cell lines from individual donors showed diversity in multilineage potential. Interestingly, five of the six lines displayed adipogenic potential, four had osteogenic and adipogenic potential, and only one cell line was tripotent. Both bone marrow (BM)- and ACL-derived MSC expressed marker genes for ligament fibroblasts, whereas the mRNA levels of collagen I and III were more abundant in ACL-derived MSC. DISCUSSION Our study demonstrates that human MSC can be isolated from ACL with diversity in the potential to form bone, fat and cartilage and an increase as compared to BM MSC, in the potential to form ligament fibroblasts.


Stereotactic and Functional Neurosurgery | 2011

Gamma Knife Radiosurgery for Central Neurocytoma: Retrospective Analysis of Fourteen Cases with a Median Follow-Up Period of Sixty-Five Months

Meng-Chao Chen; David Hung-Chi Pan; Wen-Yuh Chung; Kang-Du Liu; Yu-Shu Yen; Ming-Teh Chen; Tai-Tong Wong; Yang-Hsin Shih; Hsiu-Mei Wu; Wan-Yuo Guo; Cheng-Ying Shiau; Ling-Wei Wang; Chii-Wann Lin

Object: Central neurocytoma (CN) is considered to be a benign neuronal tumor with possible atypical behavior. Microsurgery, radiation therapy (RT) and radiosurgery all have been used in treating this rare disease during the past decade. In this study, the authors present the experience with gamma knife radiosurgery (GKRS) on 14 patients with CN during a median follow-up period of 65 months and document the safety and efficacy of GKRS in the treatment of CN. Methods: Between November 1997 and December 2009, 14 patients pathologically diagnosed with CN were treated with GKRS. Follow-up magnetic resonance imaging (MRI) was performed at 6-month intervals. Tumor volume and adverse radiation effects (ARE) were documented to evaluate tumor response to GKRS. The Karnofsky Performance Scale (KPS) and neurological status were used to assess clinical outcome. The mean radiation dose prescribed to the tumor margin was 12.1 Gy (ranging from 11 to 13 Gy). The mean tumor volume was 19.6 ml (ranging from 3.5 to 48.9 ml). The mean follow-up period was 70 months (ranging from 30 to 140 months), and the median follow-up period was 65 months. Results: Tumor shrinkage was found in all patients at the final MRI follow-up. The mean volume reduction was 69% (ranging from 47 to 87%). No tumor progression, ARE or radiation-related toxicity developed in any of the cases. The KPS scores of all patients were the same or had increased, and the neurological functions were all stable without deterioration at the final follow-up. Conclusion: In our observations, GKRS was found to be an effective and safe alternative as adjuvant therapy for pathology-confirmed CN. The tumor volume and functional outcome can be controlled with a favorable result in long-term observation. Compared with RT and microsurgery, GKRS plays an important role in the treatment of CN as a minimally invasive technique with low morbidity. Regular long-term MRI follow-up should be mandatory to document the tumor response and possible recurrence. Multicenter consortia should be considered for further investigation and evaluation of GKRS for such a rare tumor.


Radiation Oncology | 2011

Enhancement of radiosensitivity in human glioblastoma cells by the DNA N-mustard alkylating agent BO-1051 through augmented and sustained DNA damage response

Pei Ming Chu; Shih-Hwa Chiou; Tsann-Long Su; Yi-Jang Lee; Li-Hsin Chen; Yi-Wei Chen; Sang-Hue Yen; Ming-Teh Chen; Ming-Hsiung Chen; Yang-Hsin Shih; Pang-Hsien Tu; Hsin-I Ma

Background1-{4-[Bis(2-chloroethyl)amino]phenyl}-3-[2-methyl-5-(4-methylacridin-9-ylamino)phenyl]urea (BO-1051) is an N-mustard DNA alkylating agent reported to exhibit antitumor activity. Here we further investigate the effects of this compound on radiation responses of human gliomas, which are notorious for the high resistance to radiotherapy.MethodsThe clonogenic assay was used to determine the IC50 and radiosensitivity of human glioma cell lines (U87MG, U251MG and GBM-3) following BO-1051. DNA histogram and propidium iodide-Annexin V staining were used to determine the cell cycle distribution and the apoptosis, respectively. DNA damage and repair state were determined by γ-H2AX foci, and mitotic catastrophe was measure using nuclear fragmentation. Xenograft tumors were measured with a caliper, and the survival rate was determined using Kaplan-Meier method.ResultsBO-1051 inhibited growth of human gliomas in a dose- and time-dependent manner. Using the dosage at IC50, BO-1051 significantly enhanced radiosensitivity to different extents [The sensitizer enhancement ratio was between 1.24 and 1.50 at 10% of survival fraction]. The radiosensitive G2/M population was raised by BO-1051, whereas apoptosis and mitotic catastrophe were not affected. γ-H2AX foci was greatly increased and sustained by combined BO-1051 and γ-rays, suggested that DNA damage or repair capacity was impaired during treatment. In vivo studies further demonstrated that BO-1051 enhanced the radiotherapeutic effects on GBM-3-beared xenograft tumors, by which the sensitizer enhancement ratio was 1.97. The survival rate of treated mice was also increased accordingly.ConclusionsThese results indicate that BO-1051 can effectively enhance glioma cell radiosensitivity in vitro and in vivo. It suggests that BO-1051 is a potent radiosensitizer for treating human glioma cells.


Oncotarget | 2016

Musashi-1 regulates AKT-derived IL-6 autocrinal/paracrinal malignancy and chemoresistance in glioblastoma

Hsiao-Yun Chen; Liang-Ting Lin; Mong-Lien Wang; Shu-Hsien Lee; Ming-Long Tsai; Chi-Chang Tsai; Wei-Hsiu Liu; Tzu-Chien Chen; Yi-Ping Yang; Yi-Yen Lee; Yuh-Lih Chang; Pin-I Huang; Yi-Wei Chen; Wen-Liang Lo; Shih-Hwa Chiou; Ming-Teh Chen

Glioblastoma multiform (GBM) is one of the most lethal human malignant brain tumors with high risks of recurrence and poor treatment outcomes. The RNA-binding protein Musashi-1 (MSI1) is a marker of neural stem/progenitor cells. Recent study showed that high expression level of MSI1 positively correlates with advanced grade of GBM, where MSI1 increases the growth of GBM. Herein, we explore the roles of MSI1 as well as the underlying mechanisms in the regulation of drug resistance and tumorigenesis of GBM cells. Our results demonstrated that overexpression of MSI1 effectively protected GBM cells from drug-induced apoptosis through down-regulating pro-apoptotic genes; whereas inhibition of AKT withdrew the MSI1-induced anti-apoptosis and cell survival. We further showed that MSI1 robustly promoted the secretion of the pro-inflammatory cytokine IL-6, which was governed by AKT activity. Autonomously, the secreted IL-6 enhanced AKT activity in an autocrine/paracrine manner, forming a positive feedback regulatory loop with the MSI1-AKT pathway. Our results conclusively demonstrated a novel drug resistance mechanism in GBM cells that MSI1 inhibits drug-induced apoptosis through AKT/IL6 regulatory circuit. MSI1 regulates both cellular signaling and tumor-microenvironmental cytokine secretion to create an intra- and intercellular niche for GBM to survive from chemo-drug attack.


Journal of The Chinese Medical Association | 2016

Sox2, a stemness gene, regulates tumor-initiating and drug-resistant properties in CD133-positive glioblastoma stem cells

Wen-Shin Song; Yi-Ping Yang; Chi-Shuan Huang; Kai-Hsi Lu; Wei-Hsiu Liu; Wai-Wah Wu; Yi-Yen Lee; Wen-Liang Lo; Shou-Dong Lee; Yi-Wei Chen; Pin-I Huang; Ming-Teh Chen

Background Glioblastoma multiforme (GBM) is the most lethal type of adult brain cancer and performs outrageous growth and resistance regardless of adjuvant chemotherapies, eventually contributing to tumor recurrence and poor outcomes. Considering the common heterogeneity of cancer cells, the imbalanced regulatory mechanism could be switched on/off and contribute to drug resistance. Moreover, the subpopulation of GBM cells was recently discovered to share similar phenotypes with neural stem cells. These cancer stem cells (CSCs) promote the potency of tumor initiation. As a result, targeting of glioma stem cells has become the dominant way of improving the therapeutic outcome against GBM and extending the life span of patients. Among the biomarkers of CSCs, CD‐133 (prominin‐1) has been known to effectively isolate CSCs from cancer population, including GBM; however, the underlying mechanism of how stemness genes manipulate CSC‐associated phenotypes, such as tumor initiation and relapse, is still unclear. Methods Tumorigenicity, drug resistance and embryonic stem cell markers were examined in primary CD133‐positive (CD133+) GBM cells and CD133+ subpopulation. Stemness signature of CD133+ GBM cells was identified using microarray analysis. Stem cell potency, tumorigenicity and drug resistance were also tested in differential expression of SOX2 in GBM cells. Results In this study, high tumorigenic and drug resistance was noticed in primary CD‐133+ GBM cells; meanwhile, plenty of embryonic stem cell markers were also elevated in the CD‐133+ subpopulation. Using microarray analysis, we identified SOX2 as the most enriched gene among the stemness signature in CD133+ GBM cells. Overexpression of SOX2 consistently enhanced the stem cell potency in the GBM cell lines, whereas knockdown of SOX2 dramatically withdrew CD133 expression in CD133+ GBM cells. Additionally, we silenced SOX2 expression using RNAi system, which abrogated the ability of tumor initiation as well as drug resistance of CD133+ GBM cells, suggesting that SOX2 plays a crucial role in regulating tumorigenicity in CD133+ GBM cells. Conclusion SOX2 plays a crucial role in regulating tumorigenicity in CD133+ GBM cells. Our results not only revealed the genetic plasticity contributing to drug resistance and stemness but also demonstrated the dominant role of SOX2 in maintenance of GBM CSCs, which may provide a novel therapeutic target to overcome the conundrum of poor survival of brain cancers.


Cancer Chemotherapy and Pharmacology | 2012

Targeting autophagy enhances BO-1051-induced apoptosis in human malignant glioma cells

Pei Ming Chu; Li-Hsin Chen; Ming-Teh Chen; Hsin-I Ma; Tsann-Long Su; Pei-Chen Hsieh; Chian-Shiu Chien; Bo-Hua Jiang; Yu-Chih Chen; Yi-Hui Lin; Yang-Hsin Shih; Pang-Hsien Tu; Shih-Hwa Chiou

PurposeBO-1051 is an N-mustard derivative that is conjugated with DNA-affinic 9-anilinoacridine. Since BO-1051 was reported to have strong anticancer activity, we investigated the effect and underlying mechanism of BO-1051 in human glioma cell lines.MethodsHuman glioma cell lines U251MG and U87MG were studied with BO-1051 or the combination of BO-1051 and autophagic inhibitors. Growth inhibition was assessed by MTT assay. Apoptosis was measured by annexin V staining followed by flow cytometry and immunoblotting for apoptosis-related molecules. Induction of autophagy was detected by acridine orange labeling, electron microscopy, LC3 localization and its conversion. Transfection of shRNA was used to determine the involvement of Beclin1 in apoptotic cell death.ResultsMTT assay showed that BO-1051 suppressed the viability of four glioma cell lines (U251MG, U87MG, GBM-3 and DBTRG-05MG) in a dose-dependent manner. The IC50 values of BO-1051 for the glioma cells were significantly lower than the values for primary neurons cultures and normal fibroblast cells. Moreover, BO-1051 not only induced apoptotic cell death, but also enhanced autophagic flux via inhibition of Akt/mTOR and activation of Erk1/2. Importantly, suppression of autophagy by 3-methyladenine or bafilomycin A1 significantly increased BO-1051-induced apoptotic cell death in U251MG and U87MG cells. In addition, the proportion of apoptotic cells after BO-1051 treatment was enhanced by co-treatment with shRNA against Beclin1.ConclusionsBO-1051 induced both apoptosis and autophagy, and inhibition of autophagy significantly augmented the cytotoxic effect of BO-1051. Thus, a combination of BO-1051 and autophagic inhibitors offers a potentially new therapeutic modality for the treatment of malignant glioma.


Cell Transplantation | 2014

MicroRNA142-3p Promotes Tumor-Initiating and Radioresistant Properties in Malignant Pediatric Brain Tumors

Yi-Yen Lee; Yi-Ping Yang; Ming-Chao Huang; Mong-Lien Wang; Sang-Hue Yen; Pin-I Huang; Yi-Wei Chen; Shih-Hwa Chiou; Yuan-Tzu Lan; Hsin-I Ma; Yang-Hsin Shih; Ming-Teh Chen

Primary central nervous system (CNS) atypical teratoid/rhabdoid tumor (ATRT) is an extremely malignant pediatric brain tumor observed in infancy and childhood. It has been reported that a subpopulation of CD133+ cells isolated from ATRT tumors present with cancer stem-like and radioresistant properties. However, the exact biomolecular mechanisms of ATRT or CD133-positive ATRT (ATRT-CD133+) cells are still unclear. We have previously shown that ATRT-CD133+ cells have pluripotent differentiation ability and the capability of malignant cells to be highly resistant to ionizing radiation (IR). By using microRNA array and quantitative RT-PCR in this study, we showed that expression of miR142-3p was lower in ATRT-CD133+ cells than in ATRT-CD133- cells. miR142-3p overexpression significantly inhibited the self-renewal and tumorigenicity of ATRT-CD133+ cells. On the contrary, silencing of endogenous miR142-3p dramatically increased the tumor-initiating and stem-like cell capacities in ATRT cells or ATRT-CD133- cells and further promoted the mesenchymal transitional and radioresistant properties of ATRT cells. Most importantly, therapeutic delivery of miR142-3p in ATRT cells effectively reduced its lethality by blocking tumor growth, repressing invasiveness, increasing radiosensitivity, and prolonging survival time in orthotropic-transplanted immunocompromised mice. These results demonstrate the prospect of developing novel miRNA-based strategies to block the stem-like and radioresistant properties of malignant pediatric brain cancer stem cells.


Brain Research Bulletin | 2015

Exercise suppresses COX-2 pro-inflammatory pathway in vestibular migraine

Yi-Yen Lee; Yi-Ping Yang; Pin-I Huang; Wen-Cheng Li; Ming-Chao Huang; Chung-Lan Kao; Yann-Jang Chen; Ming-Teh Chen

Migraine and dizziness are relatively common disorders. Patients with dizziness have a higher incidence of migraines than the general population. The discomfort experienced by these patients is often poorly controlled by medication. However, the pathophysiology of vestibular migraine (VM) remains unclear. We hypothesized that patients with VM would experience remission from symptoms after exercise training and that this effect may be mediated through the suppression of cyclooxygenase-2 (COX-2)-mediated inflammation. Thus, the aim of the present study was to investigate the efficacy and possible anti-inflammatory benefits of exercise in patients with VM. We assessed the level of soluble inflammatory mediators in plasma from VM patients and control subjects. Our analysis of cytokine expression in the patients with VM undergoing exercise treatment revealed a significant reduction in pro-inflammatory cytokines and/or cytotoxic factors, such as tumor necrosis factor-α, interleukins, nitric oxide (NO), inducible NO synthase, and reactive oxygen species. In contrast, we found an increase in the level of anti-inflammatory cytokines after exercise. Moreover, the group undergoing exercise training showed significant symptomatic improvement and demonstrated suppressed antioxidant enzyme activity. To summarize, our data suggest that exercise significantly inhibits COX-2 activity, leading to the suppression of pro-inflammatory cytokines and changes in redox status. These results suggest that there is a molecular link between the central nervous system and the immune system. Furthermore, elucidation of the neurobiological mechanisms underlying VM could potentially lead to the development of novel therapeutic interventions for these patients.


Cell Transplantation | 2015

Nuclear Localization Signal-Enhanced Polyurethane-Short Branch Polyethylenimine-Mediated Delivery of Let-7a Inhibited Cancer Stem-Like Properties by Targeting the 3′-UTR of HMGA2 in Anaplastic Astrocytoma:

Meng-Yin Yang; Ming-Teh Chen; Pin-I Huang; Chien-Ying Wang; Yun-Chin Chang; Yi-Ping Yang; Wen-Liang Lo; Wen-Hsing Sung; Yi-Wen Liao; Yi-Yen Lee; Yuh-Lih Chang; Ling-Ming Tseng; Yi-Wei Chen; Hsin-I Ma

Anaplastic astrocytoma (AA) is a grade III glioma that often occurs in middle-aged patients and presents a uniformly poor prognosis. A small subpopulation of cancer stem cells (CSCs) possessing a self-renewing capacity is reported to be responsible for tumor recurrence and therapeutic resistance. An accumulating amount of microRNAs (miRNA) were found aberrantly expressed in human cancers and regulate CSCs. Efforts have been made to couple miRNAs with nonviral gene delivery approaches to target specific genes in cancer cells. However, the efficiency of delivery of miRNAs to AA-derived CSCs is still an applicability hurdle. The present study aimed to investigate the effectiveness and applicability of nonviral vector-mediated delivery of Let-7a with regard to eradication of AA and AA-derived CSC cells. Herein, our miRNA/mRNA microarray and RT-PCR analysis showed that the expression of Let-7a, a tumor-suppressive miRNA, is inversely correlated with the levels of HMGA2 and Sox2 in the AA side population (SP+) cells. Luciferase reporter assay showed that Let-7a directly targets the 3′-UTRs of HMGA2 in AA-SP+ cells. Knockdown of HMGA2 significantly suppressed the protein expression of Sox2 in AA-SP+ cells, whereas overexpression of HMGA2 upregulated Sox2 expression in AA-SP-. Nuclear localization signal (NLS) peptides can facilitate nuclear targeting of DNA and are used to improve gene delivery. Using polyurethane-short branch polyethylenimine (PU-PEI) as a therapeutic delivery vehicle, we conjugated NLS with Let-7 and successfully delivered it to AA-SP+ cells, resulting in significantly suppressed expression of HMGA2 and Sox2, tumorigenicity, and CSC-like abilities. This treatment facilitated the differentiation of AA-SP+ cells into non-SP CSCs. Furthermore, PU-PEI-mediated delivery of NLS-conjugated Let-7a in AA-SP+ cells suppressed the expression of drug-resistant and antiapoptotic genes, and increased cell sensitivity to radiation. Finally, the in vivo delivery of PU-PEI-NLS-Let-7a significantly suppressed the tumorigenesis of AA-SP+ cells and synergistically improved the survival rate of orthotopically AA-SP+-transplanted immunocompromised mice when combined with radiotherapy. Therefore, PU-PEI-NLS-Let-7a is a potential novel therapeutic approach for AA.


Oncotarget | 2017

TDP-43/HDAC6 axis promoted tumor progression and regulated nutrient deprivation-induced autophagy in glioblastoma

Tzu-Wei Lin; Ming-Teh Chen; Liang-Tin Lin; Pin-I Huang; Wen-Liang Lo; Yi-Ping Yang; Kai-Hsi Lu; Yi-Wei Chen; Shih-Hwa Chiou; Cheng-Wen Wu

Glioblastoma Multiforme (GBM) is a lethal primary brain tumor with poor survival lifespan and dismal outcome. Surgical resection of GBM is greatly limited due to the biological significance of brain, giving rise to tumor relapse in GBM patients. Transactive response DNA binding protein-43 (TDP-43) is a DNA/RNA-binding protein known for causing neurodegenerative diseases through post-translational modification; but little is known about its involvement in cancer development. In this study, we found that nutrient deprivation in GBM cell lines elevated TDP-43 expression by a mechanism of evasion from ubiquitin-dependent proteolytic pathway, and subsequently activated the autophagy process. Exogenous overexpression of TDP-43 consistently activated autophagy and suppressed stress-induced apoptosis. The inhibition of autophagy in TDP-43-overexpressing cells effectively increased the apoptotic population under nutrition shortage. Furthermore, we demonstrated that HDAC6 was involved in the activation of autophagy in TDP-43-overexpressing GBM cell lines. The treatment with SAHA, a universal HDAC inhibitor, significantly reduced TDP-43-mediated anti-apoptotic effect. Additionally, the results of immunohistochemistry showed that TDP-43 and HDAC6 collaborated in GBM-tumor lesions and negatively correlated with the relapse-free survival of GBM patients. Taken together, our results suggest that the TDP-43-HDAC6 signaling axis functions as a stress responsive pathway in GBM tumorigenesis and combats nutrient deprivation stress via activating autophagy, while inhibition of HDAC6 overpowers the pathway and provides a novel therapeutic strategy against GBM.Glioblastoma Multiforme (GBM) is a lethal primary brain tumor with poor survival lifespan and dismal outcome. Surgical resection of GBM is greatly limited due to the biological significance of brain, giving rise to tumor relapse in GBM patients. Transactive response DNA binding protein-43 (TDP-43) is a DNA/RNA-binding protein known for causing neurodegenerative diseases through post-translational modification; but little is known about its involvement in cancer development. In this study, we found that nutrient deprivation in GBM cell lines elevated TDP-43 expression by a mechanism of evasion from ubiquitin-dependent proteolytic pathway, and subsequently activated the autophagy process. Exogenous overexpression of TDP-43 consistently activated autophagy and suppressed stress-induced apoptosis. The inhibition of autophagy in TDP-43-overexpressing cells effectively increased the apoptotic population under nutrition shortage. Furthermore, we demonstrated that HDAC6 was involved in the activation of autophagy in TDP-43-overexpressing GBM cell lines. The treatment with SAHA, a universal HDAC inhibitor, significantly reduced TDP-43-mediated anti-apoptotic effect. Additionally, the results of immunohistochemistry showed that TDP-43 and HDAC6 collaborated in GBM-tumor lesions and negatively correlated with the relapse-free survival of GBM patients. Taken together, our results suggest that the TDP-43-HDAC6 signaling axis functions as a stress responsive pathway in GBM tumorigenesis and combats nutrient deprivation stress via activating autophagy, while inhibition of HDAC6 overpowers the pathway and provides a novel therapeutic strategy against GBM.

Collaboration


Dive into the Ming-Teh Chen's collaboration.

Top Co-Authors

Avatar

Yi-Wei Chen

Taipei Veterans General Hospital

View shared research outputs
Top Co-Authors

Avatar

Pin-I Huang

Taipei Veterans General Hospital

View shared research outputs
Top Co-Authors

Avatar

Shih-Hwa Chiou

Taipei Veterans General Hospital

View shared research outputs
Top Co-Authors

Avatar

Yi-Yen Lee

Taipei Veterans General Hospital

View shared research outputs
Top Co-Authors

Avatar

Yi-Ping Yang

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Hsin-I Ma

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yang-Hsin Shih

Taipei Veterans General Hospital

View shared research outputs
Top Co-Authors

Avatar

Mong-Lien Wang

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Wen-Liang Lo

Taipei Veterans General Hospital

View shared research outputs
Top Co-Authors

Avatar

Ming-Chao Huang

Taipei Veterans General Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge