hosted by
publicationslist.org
    

Ming Tan

Center for Basic & Translational Research
USA Mitchell Cancer Institute
1660 Springhill Avenue, Mobile, AL 36604
Telephone: (251) 460-6993
Fax: (251) 460-6994
mtan@usouthal.edu
Ming Tan, MD, PhD
Associate Professor of Oncologic Sciences
Associate Professor of Cell Biology
Vincent F. Kilborn, Jr. Cancer Research Scholar
University of South Alabama


Education:

Postdoctoral Fellow, University of Texas M.D. Anderson Cancer Center , Houston, Texas
Ph.D., University of Texas M.D. Anderson Cancer Center and Health Science Center at Houston, Texas
M.D., West China University of Medical Sciences, Chengdu, China

Scientific Focus:

Dr. Tan's laboratory focuses on the molecular mechanisms of tumor progression, metastasis, and therapeutic resistance of human breast cancer, melanoma, and other cancer types. Currently Dr. Tan’s lab is studying growth factor receptors, such as HER-2/ErbB2, and transcription factors, such as HSF1, in mediating cancer metabolism, signal transduction, apoptosis, metastasis, and resistance to therapeutic agents. Another emphasis of his study is on the deregulation of microRNA in cancer progression and therapeutic resistance. The ultimate goal of Dr. Tan’s research is to translate the knowledge acquired in the laboratory into future novel therapeutics.


Honors and Academic Achievements:

Member of the peer review panel, U.S. Department of Defese Cancer Research Programs
Vincent F. Kilborn, Jr. Cancer Research Scholar, USA Mitchell Cancer Institute
Second Prize, Alexander Y-M Wang Memorial Awards, SCBA Annual Scientific Symposium Oral Competition
Sowell-Huggins Scholarship for Excellence in Cancer Research, University of Texas M.D. Anderson Cancer Center and Health Sciences Center at Houston
U.S. Department of Defense Breast Cancer Research Fellowship
Brenda Graham Travel Awards, University of Texas Health Sciences Center at Houston
Research Fellowship, Graduate School, University of Texas Health Science Center at Houston
Finalist, Huo Ying-Dong Young Teacher Award for Teaching Excellence, West China University of Medical Sciences, China
Excellent Research Award for Young Scientist, Science and Technology Association of Sichuan Province, China
Excellent Medical Student Awards, School of Medicine, West China University of Medical Sciences, China

Journal articles

2012
J Huang, D Dou, J Dang, J H Pardue, X Qin, J Huan, W T Gerthoffer, M Tan (2012)  Knowledge acquisition, semantic text mining, and security risks in health and biomedical informatics.   World J Biol Chem. 3: 2. 27-33  
Abstract: Abstract Computational techniques have been adopted in medical and biological systems for a long time. There is no doubt that the development and application of computational methods will render great help in better understanding biomedical and biological functions. Large amounts of datasets have been produced by biomedical and biological experiments and simulations. In order for researchers to gain knowledge from original data, nontrivial transformation is necessary, which is regarded as a critical link in the chain of knowledge acquisition, sharing, and reuse. Challenges that have been encountered include: how to efficiently and effectively represent human knowledge in formal computing models, how to take advantage of semantic text mining techniques rather than traditional syntactic text mining, and how to handle security issues during the knowledge sharing and reuse. This paper summarizes the state-of-the-art in these research directions. We aim to provide readers with an introduction of major computing themes to be applied to the medical and biological research.
Notes:
2011
Yuhua Zhao, Hao Liu, Zixing Liu, Yan Ding, Susan P LeDoux, Glenn L Wilson, Richard Voellmy, Yifeng Lin, Wensheng Lin, Rita Nahta, Bolin Liu, Øystein Fodstad, Jieqing Chen, Yun Wu, Janet E Price, Ming Tan (2011)  Overcoming Trastuzumab Resistance in Breast Cancer by Targeting Dysregulated Glucose Metabolism.   Cancer Research (Published OnlineFirst April 15, 2011)  
Abstract: Trastuzumab shows remarkable efficacy in treatment of ErbB2-positive breast cancers when used alone or in combination with other chemotherapeutics. However, acquired resistance develops in most treated patients, necessitating alternate treatment strategies. Increased aerobic glycolysis is a hallmark of cancer and inhibition of glycolysis may offer a promising strategy to preferentially kill cancer cells. In this study, we investigated the antitumor effects of trastuzumab in combination with glycolysis inhibitors in ErbB2-positive breast cancer. We found that trastuzumab inhibits glycolysis via downregulation of heat shock factor 1 (HSF1) and lactate dehydrogenase A (LDH-A) in ErbB2-positive cancer cells, resulting in tumor growth inhibition. Moreover, increased glycolysis via HSF1 and LDH-A contributes to trastuzumab resistance. Importantly, we found that combining trastuzumab with glycolysis inhibition synergistically inhibited trastuzumab-sensitive and -resistant breast cancers in vitro and in vivo, due to more efficient inhibition of glycolysis. Taken together, our findings show how glycolysis inhibition can dramatically enhance the therapeutic efficacy of trastuzumab in ErbB2-positive breast cancers, potentially useful as a strategy to overcome trastuzumab resistance.
Notes:
2010
2009
M Tan*, J Lu*, W C Huang, P Li, H Guo, L M Tseng, X H Su, W T Yang, W Treekitkarnmongkol, M Andreeff, F Symmans, D Yu (2009)  Mitotic deregulation by survivin in ErbB2-overexpressing breast cancer cells contributes to Taxol resistance   Clin Cancer Res 15: 4. 1326-34 (*Co-First Authors)  
Abstract: PURPOSE: Taxol resistance remains a major obstacle to improve the benefit of breast cancer patients. Here, we studied whether overexpression of ErbB2 may lead to mitotic deregulation in breast cancer cells via up-regulation of survivin that confers Taxol resistance. EXPERIMENTAL DESIGN: ErbB2-overexpressing and ErbB2-low-expressing breast cancer cell lines were used to compare their mitotic exit rate, survivin expression level, and apoptosis level in response to Taxol. Survivin was then down-regulated by antisense oligonucleotides to evaluate its contribution to mitotic exit and Taxol resistance in ErbB2-overexpressing breast cancer cells. At last, specific PI3K/Akt and Src inhibitors were used to investigate the involvement of these two pathways in ErbB2-mediated survivin up-regulation and Taxol resistance. RESULTS: We found that ErbB2-overexpressing cells expressed higher levels of survivin in multiple breast cancer cell lines and patient samples. ErbB2-overexpressing cells exited M phase faster than ErbB2-low-expressing cells, which correlated with the increased resistance to Taxol-induced apoptosis. Down-regulation of survivin by antisense oligonucleotide delayed mitotic exit of ErbB2-overexpressing cells and also sensitized ErbB2-overexpressing cells to Taxol-induced apoptosis. Moreover, ErbB2 up-regulated survivin at translational level and PI3K/Akt and Src activation are involved. In addition, combination treatment of Taxol with PI3K/Akt and Src inhibitor led to increased apoptosis in ErbB2-overexpressing breast cancer cells than single treatment. CONCLUSIONS: Survivin up-regulation by ErbB2 is a critical event in ErbB2-mediated faster mitotic exit and contributes to Taxol resistance.
Notes: P01-CA099031/CA/NCI NIH HHS/United States xD;P30-CA16672/CA/NCI NIH HHS/United States xD;P50-CA116199/CA/NCI NIH HHS/United States xD;R01-CA109570/CA/NCI NIH HHS/United States xD;R01-CA112567/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, N.I.H., Extramural xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, Non-P.H.S. xD;United States xD;an official journal of the American Association for Cancer Research
Y H Zhao, M Zhou, H Liu, Y Ding, H T Khong, D Yu, O Fodstad, M Tan (2009)  Upregulation of lactate dehydrogenase A by ErbB2 through heat shock factor 1 promotes breast cancer cell glycolysis and growth   Oncogene 28: 42. 3689–3701  
Abstract: ErbB2 has been shown to activate signaling molecules that may regulate glucose metabolism. However, there is no evidence reported to directly link ErbB2 to glycolysis, and the mechanism underlying ErbB2-enhanced glycolysis is poorly understood. In this study, we investigated the role and mechanism of ErbB2 in regulating glycolysis. We found that ErbB2-overexpressing cells possessed a significantly higher level of glycolysis when compared to the ErbB2-low-expressing cells, and the downregulation of ErbB2 markedly decreased glycolysis. Overexpression of ErbB2 increased the expression of glycolysis-regulating molecules lactate dehydrogenase A (LDH-A) and heat shock factor 1 (HSF1). ErbB2 activated HSF1, indicated by the increased HSF1 trimer formation, and promoted HSF1 protein synthesis. HSF1 bound to LDH-A promoter and the downregulation of HSF1 reduced the expression of LDH-A and subsequently decreased cancer cell glycolysis and growth. Moreover, the glycolysis inhibitors, 2-deoxyglucose and oxamate, selectively inhibited the growth of ErbB2-overexpressing cells. Taken together, this study shows that in human breast cancer cells, ErbB2 promotes glycolysis at least partially through the HSF1-mediated upregulation of LDH-A. This pathway may have a major role in regulating glucose metabolism in breast cancer cells. These novel findings have important implications for the design of new approaches to target ErbB2-overexpressing breast cancers.
Notes: News on this study: http://www.baldwincountynow.com/articles/2009/08/28/local_news/doc4a8dbf00d813e083396309.txt
2007
M Tan, D Yu (2007)  Molecular mechanisms of erbB2-mediated breast cancer chemoresistance   Adv Exp Med Biol 608: 119-29  
Abstract:
Notes: 1P01-CA099031-1/CA/NCI NIH HHS/United States xD;R01-CA109570/CA/NCI NIH HHS/United States xD;R01-CA60488/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, N.I.H., Extramural xD;Research Support, U.S. Gov't, Non-P.H.S. xD;Review xD;United States
2006
M Tan*, K H Lan*, J Yao, C H Lu, M Sun, C L Neal, J Lu, D Yu (2006)  Selective inhibition of ErbB2-overexpressing breast cancer in vivo by a novel TAT-based ErbB2-targeting signal transducers and activators of transcription 3-blocking peptide   Cancer Res 66: 7. 3764-72 (*Co-First Authors)  
Abstract: ErbB2 is an excellent target for cancer therapies. Unfortunately, the outcome of current therapies for ErbB2-positive breast cancers remains unsatisfying due to resistance and side effects. New therapies for ErbB2-overexpressing breast cancers continue to be in great need. Peptide therapy using cell-penetrating peptides (CPP) as peptide carriers is promising because the internalization is highly efficient, and the cargoes delivered can be bioactive. However, the major obstacle in using these powerful CPPs for therapy is their lack of specificity. Here, we sought to develop a peptide carrier that could introduce therapeutics specifically to ErbB2-overexpressing breast cancer cells. By modifying the HIV TAT-derived CPP and conjugating anti-HER-2/neu peptide mimetic (AHNP), we developed the peptide carrier (P3-AHNP) that specifically targeted ErbB2-overexpressing breast cancer cells in vitro and in vivo. A signal transducers and activators of transcription 3 (STAT3)-inhibiting peptide conjugated to this peptide carrier (P3-AHNP-STAT3BP) was delivered more efficiently into ErbB2-overexpressing than ErbB2 low-expressing cancer cells in vitro and successfully decreased STAT3 binding to STAT3-interacting DNA sequence. P3-AHNP-STAT3BP inhibited cell growth in vitro, with ErbB2-overexpressing 435.eB breast cancer cells being more sensitive to the treatment than the ErbB2 low-expressing MDA-MB-435 cells. Compared with ErbB2 low-expressing MDA-MB-435 xenografts, i.p. injected P3-AHNP-STAT3BP preferentially accumulated in 435.eB xenografts, which led to more reduction of proliferation and increased apoptosis and targeted inhibition of tumor growth. This novel peptide delivery system provided a sound basis for the future development of safe and effective new-generation therapeutics to cancer-specific molecular targets.
Notes: 1R01 CA 109570/CA/NCI NIH HHS/United States xD;1R01 CA 119127-01/CA/NCI NIH HHS/United States xD;P01 CA 099031/CA/NCI NIH HHS/United States xD;P30 CA 16672/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, N.I.H., Extramural xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, Non-P.H.S. xD;United States
K S Klos, S L Wyszomierski, M Tan, X Zhou, P Li, W Yang, G Yin, W N Hittelman, D Yu (2006)  ErbB2 increases VEGF protein synthesis via activation of mammalian target of rapamycin/p70S6K leading to increased angiogenesis and spontaneous metastasis of human breast cancer cells   Cancer Res 66: 4. 2028-37  
Abstract: ErbB2 overexpression in breast tumors results in increased metastasis and angiogenesis and reduced survival. To study ErbB2 signaling mechanisms in metastasis and angiogenesis, we did a spontaneous metastasis assay using MDA-MB-435 human breast cancer cells stably transfected with constitutively active ErbB2 kinase (V659E), a kinase-dead mutant of ErbB2 (K753M), or vector control (neo). Mice injected with V659E had increased metastasis incidence and tumor microvessel density than mice injected with K753M or control. Increased angiogenesis in vivo from the V659E transfectants paralleled increased angiogenic potential in vitro. V659E produced increased vascular endothelial growth factor (VEGF) through increased VEGF protein synthesis. This was mediated through signaling events involving extracellular signal-regulated kinase, phosphatidylinositol 3-kinase/Akt, mammalian target of rapamycin (mTOR), and p70S6K. The V659E xenografts also had significantly increased phosphorylated Akt, phosphorylated p70S6K, and VEGF compared with controls. To validate the clinical relevance of these findings, we examined 155 human breast tumor samples. Human tumors that overexpressed ErbB2, which have been previously shown to have higher VEGF expression, showed significantly higher p70S6K phosphorylation as well. Increased VEGF expression also significantly correlated with higher levels of Akt and mTOR phosphorylation. Additionally, patients with tumors having increased p70S6K phosphorylation showed a trend for worse disease-free survival and increased metastasis. Our findings show that ErbB2 increases VEGF protein production by activating p70S6K in cell lines, xenografts, and in human cancers and suggest that these signaling molecules may serve as targets for antiangiogenic and antimetastatic therapies.
Notes: 1R01-CA109570/CA/NCI NIH HHS/United States xD;2R01-CA60448/CA/NCI NIH HHS/United States xD;P01-CA099031/CA/NCI NIH HHS/United States xD;P30-CA 16672/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, N.I.H., Extramural xD;Research Support, U.S. Gov't, Non-P.H.S. xD;United States
M Tan, P Li, M Sun, G Yin, D Yu (2006)  Upregulation and activation of PKC alpha by ErbB2 through Src promotes breast cancer cell invasion that can be blocked by combined treatment with PKC alpha and Src inhibitors   Oncogene 25: 23. 3286-95  
Abstract: Although ErbB2 is known to enhance breast cancer metastasis, the signaling events responsible for this remain elusive. Alpha-isozyme of protein kinase C (PKCalpha), which is involved in cancer development and progression, has been suggested to be activated by ErbB2 without direct evidence. In addition, the roles of PKCalpha in ErbB2-mediated cancer cell malignancy have not been clearly identified. In this study, we investigated whether ErbB2 can activate PKCalpha and determined what role PKCalpha plays in ErbB2-mediated breast cancer cell invasion. We expressed wild-type and mutant ErbB2 with altered signaling capacities in MDA-MB-435 breast cancer cells and revealed that overexpression or activation of ErbB2 in MDA-MB-435 cells upregulated and activated PKCalpha and that downregulation of ErbB2 by small-interfering RNA decreased the expression and activity of PKCalpha in BT474 breast cancer cells. These in vitro results were supported by data from breast cancer patient samples. In 150 breast cancer tumor samples, ErbB2-overexpressing tumors showed significantly higher positive rates of PKCalpha membrane immunohistochemistry staining than that of ErbB2-low-expressing tumors. Mechanistically, we found that PKCalpha is co-immunoprecipitated with Src and PKCalpha expression and activity can be decreased by Src inhibitor PP2 and by the expression of a dominant-negative mutant of Src. Moreover, ErbB2-mediated upregulation of urokinase-type plasminogen activator receptor (uPAR) is reduced by either the PKCalpha inhibitor Go6976 or the Src inhibitor PP2, and the combination of Go6976 with PP2 is superior to either agent alone in suppressing uPAR expression and cell invasion. These results demonstrate that PKCalpha is critical for ErbB2-mediated cancer cell invasion and provide valuable insights for current and future PKCalpha and Src inhibitor clinical trials.
Notes: 1R01-CA109570/CA/NCI NIH HHS/United States xD;2R01-CA60488/CA/NCI NIH HHS/United States xD;P01-CA099031/CA/NCI NIH HHS/United States xD;P30-CA16672/CA/NCI NIH HHS/United States xD;R01-CA112567/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, N.I.H., Extramural xD;England
2005
M Tan, P Li, K S Klos, J Lu, K H Lan, Y Nagata, D Fang, T Jing, D Yu (2005)  ErbB2 promotes Src synthesis and stability : novel mechanisms of Src activation that confer breast cancer metastasis   Cancer Res 65: 5. 1858-67  
Abstract: Activation of Src kinase plays important roles in the development of many neoplasias. Most of the previous Src studies focused on the deregulation of Src kinase activity. The deregulated Src protein synthesis and stability in mediating malignant phenotypes of cancer cells, however, have been neglected. While investigating the signal transduction pathways contributing to ErbB2-mediated metastasis, we found that ErbB2-activated breast cancer cells that had higher metastatic potentials also had increased Src activity compared with ErbB2 low-expressing cells. The increased Src activity in ErbB2-activated cells paralleled higher Src protein levels, whereas Src RNA levels were not significantly altered. Our studies revealed two novel mechanisms that are involved in Src protein up-regulation and activation by ErbB2: (a) ErbB2 increased Src translation through activation of the Akt/mammalian target of rapamycin/4E-BP1 pathway and (b) ErbB2 increased Src stability most likely through the inhibition of the calpain protease. Furthermore, inhibition of Src activity by a Src-specific inhibitor, PP2, or a Src dominant-negative mutant dramatically reduced ErbB2-mediated cancer cell invasion in vitro and metastasis in an experimental metastasis animal model. Together, activation of ErbB2 and downstream signaling pathways can lead to increased Src protein synthesis and decreased Src protein degradation resulting in Src up-regulation and activation, which play critical roles in ErbB2-mediated breast cancer invasion and metastasis.
Notes: 1P01-CA099031-1/CA/NCI NIH HHS/United States xD;2R01-CA60488/CA/NCI NIH HHS/United States xD;P30-CA16672/CA/NCI NIH HHS/United States xD;R01-CA109570/CA/NCI NIH HHS/United States xD;Comparative Study xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, P.H.S. xD;United States
2004
Y Nagata, K H Lan, X Zhou, M Tan, F J Esteva, A A Sahin, K S Klos, P Li, B P Monia, N T Nguyen, G N Hortobagyi, M C Hung, D Yu (2004)  PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients   Cancer Cell 6: 2. 117-27  
Abstract: The ErbB2-targeting antibody, trastuzumab (Herceptin), has remarkable therapeutic efficacy in certain patients with ErbB2-overexpressing tumors. The overall trastuzumab response rate, however, is limited and what determines trastuzumab response is poorly understood. Here we report that PTEN activation contributes to trastuzumab's antitumor activity. Trastuzumab treatment quickly increased PTEN membrane localization and phosphatase activity by reducing PTEN tyrosine phosphorylation via Src inhibition. Reducing PTEN in breast cancer cells by antisense oligonucleotides conferred trastuzumab resistance in vitro and in vivo. Patients with PTEN-deficient breast cancers had significantly poorer responses to trastuzumab-based therapy than those with normal PTEN. Thus, PTEN deficiency is a powerful predictor for trastuzumab resistance. Additionally, PI3K inhibitors rescued PTEN loss-induced trastuzumab resistance, suggesting that PI3K-targeting therapies could overcome this resistance.
Notes: 2R01-CA60488/CA/NCI NIH HHS/United States xD;P01-CA099031/CA/NCI NIH HHS/United States xD;P30-CA16672/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, Non-P.H.S. xD;Research Support, U.S. Gov't, P.H.S. xD;United States
Y M Li, Y Pan, Y Wei, X Cheng, B P Zhou, M Tan, X Zhou, W Xia, G N Hortobagyi, D Yu, M C Hung (2004)  Upregulation of CXCR4 is essential for HER2-mediated tumor metastasis   Cancer Cell 6: 5. 459-69  
Abstract: The receptor tyrosine kinase HER2 enhances tumor metastasis; however, its role in homing to metastatic organs is poorly understood. The chemokine receptor CXCR4 has recently been shown to mediate the movement of malignant cancer cells to specific organs. Here, we show that HER2 enhances the expression of CXCR4, which is required for HER2-mediated invasion in vitro and lung metastasis in vivo. HER2 also inhibits ligand-induced CXCR4 degradation. Finally, a significant correlation between HER2 and CXCR4 expression was observed in human breast tumor tissues, and CXCR4 expression correlated with a poor overall survival rate in patients with breast cancer. These results provide a plausible mechanism for HER2-mediated breast tumor metastasis and establish a functional link between HER2 and CXCR4 signaling pathways.
Notes: CA16672/CA/NCI NIH HHS/United States xD;P01 CA099031/CA/NCI NIH HHS/United States xD;R01 CA058880/CA/NCI NIH HHS/United States xD;R01 CA109311/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, Non-P.H.S. xD;Research Support, U.S. Gov't, P.H.S. xD;United States
X Zhou, M Tan, V Stone Hawthorne, K S Klos, K H Lan, Y Yang, W Yang, T L Smith, D Shi, D Yu (2004)  Activation of the Akt/mammalian target of rapamycin/4E-BP1 pathway by ErbB2 overexpression predicts tumor progression in breast cancers   Clin Cancer Res 10: 20. 6779-88  
Abstract: The Akt/mammalian target of rapamycin (mTOR)/4E-BP1 pathway is considered to be a central regulator of protein synthesis, involving the regulation of cell proliferation, differentiation, and survival. The inhibitors of mTOR as anticancer reagents are undergoing active evaluation in various malignancies including breast cancer. However, the activation status of the Akt/mTOR/4E-BP1 pathway and its potential roles in breast cancers remain unknown. Thus, we examined 165 invasive breast cancers with specific antibodies for the phosphorylation of Akt, mTOR, and 4E-BP1 by immunohistochemistry and compared them with normal breast epithelium, fibroadenoma, intraductal hyperplasia, and ductal carcinoma in situ. We discovered that the phosphorylation of Akt, mTOR, and 4E-BP1 increased progressively from normal breast epithelium to hyperplasia and abnormal hyperplasia to tumor invasion. Phosphorylated Akt, mTOR, and 4E-BP1 were positively associated with ErbB2 overexpression. Survival analysis showed that phosphorylation of each of these three markers was associated with poor disease-free survival independently. In vitro, we further confirmed the causal relationship between ErbB2 overexpression and mTOR activation, which was associated with enhanced invasive ability and sensitivity to a mTOR inhibitor, rapamycin. Our results, for the first time, demonstrate the following: (a) high levels of phosphorylation of Akt, mTOR, and 4E-BP1 in breast cancers, indicating activation of the Akt/mTOR/4E-BP1 pathway in breast cancer development and progression; (b) a link between ErbB2 and the Akt/mTOR/4E-BP1 pathway in breast cancers in vitro and in vivo, indicating the possible role of Akt/mTOR activation in ErbB2-mediated breast cancer progression; and (c) a potential role for this pathway in predicting the prognosis of patients with breast cancer, especially those treated with mTOR inhibitors.
Notes: 2R01-CA60488/CA/NCI NIH HHS/United States xD;5PO1-CA099031/CA/NCI NIH HHS/United States xD;P30-CA16672/CA/NCI NIH HHS/United States xD;Comparative Study xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, Non-P.H.S. xD;Research Support, U.S. Gov't, P.H.S. xD;United States xD;an official journal of the American Association for Cancer Research
2002
M Tan, T Jing, K H Lan, C L Neal, P Li, S Lee, D Fang, Y Nagata, J Liu, R Arlinghaus, M C Hung, D Yu (2002)  Phosphorylation on tyrosine-15 of p34(Cdc2) by ErbB2 inhibits p34(Cdc2) activation and is involved in resistance to taxol-induced apoptosis   Molecular Cell 9: 5. 993-1004  
Abstract: ErbB2 overexpression confers resistance to taxol-induced apoptosis by inhibiting p34(Cdc2) activation. One mechanism is via ErbB2-mediated upregulation of p21(Cip1), which inhibits Cdc2. Here, we report that the inhibitory phosphorylation on Cdc2 tyrosine (Y)15 (Cdc2-Y15-p) is elevated in ErbB2-overexpressing breast cancer cells and primary tumors. ErbB2 binds to and colocalizes with cyclin B-Cdc2 complexes and phosphorylates Cdc2-Y15. The ErbB2 kinase domain is sufficient to directly phosphorylate Cdc2-Y15. Increased Cdc2-Y15-p in ErbB2-overexpressing cells corresponds with delayed M phase entry. Expressing a nonphosphorylatable mutant of Cdc2 renders cells more sensitive to taxol-induced apoptosis. Thus, ErbB2 membrane RTK can confer resistance to taxol-induced apoptosis by directly phosphorylating Cdc2.
Notes: 2R01-CA60488/CA/NCI NIH HHS/United States xD;P30-CA16672/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, Non-P.H.S. xD;Research Support, U.S. Gov't, P.H.S. xD;United States
1999
M Tan, R Grijalva, D Yu (1999)  Heregulin beta1-activated phosphatidylinositol 3-kinase enhances aggregation of MCF-7 breast cancer cells independent of extracellular signal-regulated kinase   Cancer Res 59: 7. 1620-5  
Abstract: Heregulin (HRG) is a family of polypeptide growth factors derived from alternatively spliced genes. HRG can bind to receptor tyrosine kinases erbB3 and erbB4, thereby inducing erbB3 and erbB4 heterodimerization with erbB2, leading to receptor tyrosine phosphorylation and activating downstream signal transduction. Cell-cell homophilic adhesion (cell aggregation) is important in determining the structural organization and behavior of cells in tissues. In addition, tumor cell homophilic adhesion may affect invasive and metastatic potentials of cells. We report that HRG-beta1 can enhance aggregation of MCF-7 and SKBR3 human breast cancer cells. While investigating the downstream signals involved in HRG-beta1-enhanced cell aggregation, we observed that HRG-beta1 induced tyrosine phosphorylation of erbB2 and crbB3 receptor heterodimers and increased the association of the dimerized receptors with the 85-kDa subunit of phosphatidylinositol 3-kinase (PI3K). HRG-beta also increased the kinase activities of extracellular signal-regulated protein kinase (ERK) and PI3K in these cells. By using the mitogen-activated protein kinase/ERK 1 (MEK1) inhibitor PD98059 and PI3K inhibitors wortmannin and LY294002, we found that blocking the MEK1-ERK pathway had no effect on HRG-pbeta1-enhanced cell aggregation; however, blocking the PI3K pathway greatly inhibited HRG-beta1-mediated cell aggregation. Our study indicated that the HRG-beta1-activated MEK1-ERK pathway has no demonstrable role in the induction of cell aggregation, whereas HRG-beta1-activated PI3K is required for enhancing breast cancer cell aggregation. Because aggregation can contribute to invasion/metastasis phenotype of cancer cells, our results have provided one mechanism by which HRG-beta1-activated signaling of erbB receptors may affect invasive/metastatic properties of MCF-7 and SKBR3 breast cancer cells.
Notes: CA60488/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, Non-P.H.S. xD;Research Support, U.S. Gov't, P.H.S. xD;United states
1998
J Yao, R E Pollock, A Lang, M Tan, P W Pisters, D Goodrich, A El-Naggar, D Yu (1998)  Infrequent mutation of the p16/MTS1 gene and overexpression of cyclin-dependent kinase 4 in human primary soft-tissue sarcoma   Clin Cancer Res 4: 4. 1065-70  
Abstract: The pl6INK4a/MTS1 (p16) gene encodes a specific inhibitor of cyclin-dependent kinase (CDK)4 and CDK6. The p16 gene is frequently mutated or deleted in many types of cancer cell lines as well as in certain types of primary tumors. p16 knockout mice are viable but predisposed to sarcoma and B-cell lymphoma. To investigate the role of p16 in human soft-tissue sarcoma tumor progression, we examined the p16 gene by Southern blot analysis and PCR sequencing in 30 pairs of primary soft-tissue sarcomas and autologous normal tissue. Only one tumor sample showed possible rearrangement of the p16 gene. In contrast, Western blot analysis of the p16 protein in 20 pairs of samples showed decreased p16 expression in only 20% of the tumors but elevated p16 expression in 40% of the tumors when compared with the autologous normal controls. Overexpression of p16 was not concomitant with loss of the RB protein as is found in several other types of cancers, because more than one-half of the tumors with increased p16 expression also had high levels of RB protein. On the other hand, the p16 target protein CDK4 was overexpressed in at least 60% of the tumors. In the majority of cases, CDK4 overexpression accompanied elevated p16 and/or RB levels. Our results suggest that: (a) alteration of the p16 gene is infrequent in primary soft-tissue sarcoma; (b) Cdk4 may act as an oncogene in soft-tissue sarcoma; and (c) elevated p16 and RB levels might be the result of compensatory up-regulation of these proteins to counteract CDK4 overexpression in these tumors. Our results also suggest that it is more informative to examine aberrations in the "p16-CDK4/cyclin D-RB" pathway than to selectively examine individual components in this pathway when investigating genetic changes involved in human malignancy.
Notes: CA60488/CA/NCI NIH HHS/United States xD;CA67802/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, P.H.S. xD;United states xD;an official journal of the American Association for Cancer Research
R Pollock, A Lang, T Ge, D Sun, M Tan, D Yu (1998)  Wild-type p53 and a p53 temperature-sensitive mutant suppress human soft tissue sarcoma by enhancing cell cycle control   Clin Cancer Res 4: 8. 1985-94  
Abstract: Soft-tissue sarcomas are a heterogeneous group of tumors that are putatively mesenchymal in origin. Therapeutic advances in this disease have been limited over the past several decades. Approximately one-half of all patients will ultimately succumb, usually to uncontrollable pulmonary metastases. Although little is known about the underlying molecular determinants driving soft-tissue sarcoma inception, proliferation, and metastasis, mutation of the p53 gene is the most frequently detected molecular alteration in this disease. Accordingly, we were interested in determining whether transduction of wild-type (wt) p53 into soft-tissue sarcomas bearing mutated p53 genes might alter the malignant phenotype. SKLMS-1 is a human-derived leiomyosarcoma cell line with a codon 245 p53 point mutation. Cationic liposome was used to transfect wt p53 or 143Ala temperature-sensitive mutant p53 into this cell line. SKLMS-1 stable transfectants expressing wt p53 had decreased cell proliferation in vitro, decreased in vitro colony formation in soft agar, and decreased tumorigenicity in severe combined immunodeficient mice in vivo. Flow cytometric analysis of cell cycle components demonstrated markedly increased G1 cell cycle arrest and decreased entry into S phase, which corresponded to the induction of p21cip1 protein in the transfectants. Using SKLMS-1 stable transfectants expressing the 143Ala p53 temperature-sensitive mutant, we demonstrated the kinetics of and the causal relationship between wt p53 expression, the wt p53-dependent induction of cell cycle inhibitor p21cip1, and inhibition of cell cycle progression in p53-transfected SKLMS-1 cells. The ability to restore wt p53 growth-regulatory functions in soft-tissue sarcoma may ultimately be useful as a future therapy in patients with soft-tissue sarcomas.
Notes: CA60488/CA/NCI NIH HHS/United States xD;CA67802/CA/NCI NIH HHS/United States xD;P30-CA16672/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, U.S. Gov't, P.H.S. xD;United states xD;an official journal of the American Association for Cancer Research
D Yu, T Jing, B Liu, J Yao, M Tan, T J McDonnell, M C Hung (1998)  Overexpression of ErbB2 blocks Taxol-induced apoptosis by upregulation of p21Cip1, which inhibits p34Cdc2 kinase   Molecular Cell 2: 5. 581-91  
Abstract: Overexpression of the receptor tyrosine kinase p185ErbB2 confers Taxol resistance in breast cancers. Here, we investigated the underlying mechanisms and found that overexpression of p185ErbB2 inhibits Taxol-induced apoptosis. Taxol activates p34Cdc2 kinase in MDA-MB-435 breast cancer cells, leading to cell cycle arrest at the G2/M phase and, subsequently, apoptosis. A chemical inhibitor of p34Cdc2 and a dominant-negative mutant of p34Cdc2 blocked Taxol-induced apoptosis in these cells. Overexpression of p185ErbB2 in MDA-MB-435 cells by transfection transcriptionally upregulates p21Cip1, which associates with p34Cdc2, inhibits Taxol-mediated p34Cdc2 activation, delays cell entrance to G2/M phase, and thereby inhibits Taxol-induced apoptosis. In p21Cip1 antisense-transfected MDA-MB-435 cells or in p21-/- MEF cells, p185ErbB2 was unable to inhibit Taxol-induced apoptosis. Therefore, p21Cip1 participates in the regulation of a G2/M checkpoint that contributes to resistance to Taxol-induced apoptosis in p185ErbB2-overexpressing breast cancer cells.
Notes: CA60488/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, Non-P.H.S. xD;Research Support, U.S. Gov't, P.H.S. xD;United states
1997
M Tan, J Yao, D Yu (1997)  Overexpression of the c-erbB-2 gene enhanced intrinsic metastasis potential in human breast cancer cells without increasing their transformation abilities   Cancer Res 57: 6. 1199-205  
Abstract: Overexpression of the c-erbB-2 gene-encoded p185 has been reported in approximately 30% of human breast cancers and has been correlated with lymph node metastasis and poor prognosis in breast cancer patients. To investigate whether overexpression of p185 can enhance the metastatic potential of human breast cancer cells, we have introduced the human c-erbB-2 gene into the very low p185-expressing MDA-MB-435 human breast cancer cells and established 435.eB transfectants that express higher levels of p185. In this study, we compared the metastatic phenotypes of the parental MDA-MB-435 cells and the 435.eB transfectants. In vivo experimental metastasis assays in which we injected MDA-MB-435 parental cells or 435.eB transfectants into the tail veins of ICR-SCID mice demonstrated that mice injected with p185-overexpressing 435.eB transfectants formed significantly more metastatic tumors than the mice injected with parental and control cells. The changes in experimental metastatic potential in vivo were accompanied by increased invasiveness in vitro. In addition, the secretion of basement membrane-degradative enzymes, which is an important step in the invasion and metastasis process, was also increased in the p185-overexpressing 435.eB transfectants. These results indicated that p185 overexpression can enhance the metastatic potential of MDA-MB-435 human breast cancer cells. To investigate whether enhanced metastatic potential in the p185-overexpressing 435.eB transfectants was the result of increased cancer cell growth and transformation potential, we compared the growth rate, anchorage-independent growth ability, and tumorigenicity of the 435.eB transfectants with that of the parental cells. The transfectants and the parental cells all had similar growth rates and anchorage-independent growth abilities and demonstrated similar tumorigenic potential. These findings suggest that c-erbB-2 is a metastasis-promoting gene for breast cancers that is distinct from other tumor-promoting genes in that the c-erbB-2 gene can enhance the intrinsic metastatic potentials of MDA-MB-435 cells without increasing their transformation abilities.
Notes: CA60488/CA/NCI NIH HHS/United States xD;P30-CA16672/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, Non-U.S. Gov't xD;Research Support, U.S. Gov't, P.H.S. xD;United states
1996
D Yu, B Liu, M Tan, J Li, S S Wang, M C Hung (1996)  Overexpression of c-erbB-2/neu in breast cancer cells confers increased resistance to Taxol via mdr-1-independent mechanisms   Oncogene 13: 6. 1359-65  
Abstract: It has been reported that breast tumors that overexpress c-erbB-2/neu are less responsive to certain adjuvant chemotherapy regimens than those that express a normal amount of the gene product. To investigate whether overexpression of the c-erbB-2/neu-encoded p185 can indeed lead to increased chemoresistance in breast cancers, we introduced the human c-erbB-2/neu gene into the very low p185-expressing MDA-MB435 human breast cancer cells and examined Taxol sensitivities among the parental MDA-MB-435 cells and stable transfectants which express increased levels of p185. The p185-overexpressing MDA-MB-435 transfectants were more resistant to Taxol than the parental cells. The increased Taxol resistance was not accompanied by changes in doubling time and S-phase fraction. The increased Taxol resistance was independent from oncogenic transformation since it was observed only in c-erbB-2/neu-transformed cells and not ras-transformed cells when oncogene-transformed NIH3T3 cells were examined. To study whether p185 induced Taxol resistance through the mdr-1 pathway, we examined the mdr-l-encoded p170 levels in these transfectants. The MDA-MB-435 cells expressed very low levels of p170 and there was no increase of p170 expression in the p185-overexpressing MDA-MB-435 transfectants. Furthermore, these transfectants were not sensitized to Taxol treatment by mdr-1 blocker thioradazine. These data demonstrated that overexpression of c-erbB-2/neu can lead to intrinsic Taxol resistance independent from mdr-1 mechanisms.
Notes: CA58880/CA/NCI NIH HHS/United States xD;CA60488/CA/NCI NIH HHS/United States xD;CA60856/CA/NCI NIH HHS/United States xD;Journal Article xD;Research Support, U.S. Gov't, P.H.S. xD;England

Book chapters

2010
2007

Conference papers

2009
Yuhua Zhao, Ming Zhou, Hao Liu, Hung T Khong, Dihua Yu, Oystein Fodstad, Ming Tan (2009)  Upregulation of lactate dehydrogenase A by ErbB2 through heat shock factor 1 promotes breast cancer cell glycolysis and growth   In: Proceedings for the American Association for Cancer Research 100th Annual Meeting 2009 Edited by:AACR. Abstract Number:4423  
Abstract: Overexpression of ErbB2 has been shown to activate signaling molecules that are known to regulate glucose metabolism. However, there is no evidence that directly links ErbB2 to increased glycolysis in human breast cancer, and the impact on cancer progression of ErbB2-mediated changes in energy dependency is poorly understood. The role of ErbB2 in regulating glycolysis was studied in isogenic ErbB2-low and -overexpressing breast cancer cells. Compared to ErbB2-low cells, ErbB2-overexpressing breast cancer cells possessed significantly higher glucose uptake, lactate production, and lower oxygen consumption; the downregulation of ErbB2 in these cells decreased glycolysis, indicating that ErbB2 promotes glycolysis. To identify molecules that may contribute to this, the glycolytic regulating molecules lactate dehydrogenase A (LDH-A) and heat shock factor 1 (HSF1) were studied. Overexpression of ErbB2 increased the expression of both molecules, whereas downregulation of ErbB2 inhibited LDH-A and HSF1 and subsequently decreased glycolysis. Furthermore, the downregulation of HSF1 inhibited LDH-A expression, decreased cell glycolysis and cell growth, indicating that HSF1 is critical to ErbB2-mediated upregulation of LDH-A and glycolysis. In addition, the glycolysis inhibitors 2-deoxyglucose (2-DG) and oxamate selectively inhibited the growth of ErbB2-overexpressing cells and downregulation of HSF1 resulted in the cells to be less sensitive to glycolysis inhibitors. This study provides direct evidence to support a causal relationship between ErbB2 and glycolysis in human breast cancer cells. To our knowledge, this is the first report demonstrating that ErbB2 upregulates HSF1 in human breast cancer cells, and the glycolysis inhibitors 2-DG and oxamate can selectively inhibit ErbB2-overexpressing human breast cancer cells. In summary, this study demonstrates that in human breast cancer cells, overexpression of ErbB2 promotes glycolysis at least partially through HSF1-mediated upregulation of LDH-A, and glycolysis inhibitors can selectively inhibit the growth of ErbB2-overexpressing cells. These novel findings have important implications for the design of new approaches to target ErbB2-overexpressing breast cancers.
Notes:
Ming Zhou, Yuhua Zhao, Hao Liu, Oystein Fodstad, Ming Tan (2009)  Elevated expression of LDHA in Taxol-resistant breast cancer cells contributes to the increased sensitivity to Oxamate   In: Proceedings for the American Association for Cancer Research 100th Annual Meeting 2009 Edited by:AACR. Abstract Number:2943  
Abstract: Taxol is one of the major chemotherapeutic agents for the treatment of breast cancer. Despite a remarkable response of tumors to initial treatment, the majority of patients eventually develop resistance. The resistance has been proved to be a major obstacle to improving the response and survival of Taxol-treated breast cancer patients. Lactate dehydrogenase A (LDH-A) is a major isoform of LDH that is expressed in breast cancer and it controls the conversion of pyruvate to lactate. This study focused on the role of LDH-A in mediating Taxol resistance in breast cancer cells. MDA-MB-435 human breast cancer cells were treated with gradually increasing concentrations of Taxol. A series of subclones, 435TR1, 435TR2, 435TR3, 435TR4 and 435TRP (pool clones), developed Taxol resistance. Compared to the parental cells, the IC50 of the Taxol-resistant subclones increased more than 30 folds. The growth properties of the Taxol-resistant 435TR1 and 435TRP cells were examined by MTS cell proliferation assay and flow cytometry analysis. Compared to the parental cells (37.08 % in G1 phase / 36.21% in S phase), we found that Taxol-resistant cells have decreased cell growth rate and more cells are arrested at the transition of G1 to S phase (57.66% in G1 / 16.85% in S for 435TR1, and 55.67% in G1 / 16.09% in S for 435TRP). Interestingly, Taxol-resistant cells have increased expression and activity of LDH-A when compared with their parental cells. In addition, higher sensitivity to specific LDH inhibitor oxamate were found in Taxol-resistant 435TR1 and 435TRP cells and the downregulation of LDH-A by siRNA decreased the sensitivity of these cells to Taxol. Furthermore, when compared to the treatment using single agent, treating cells with the combination of Taxol and oxamate shows stronger inhibitory effect on both Taxol-resistant and sensitive cancer cells by synergistically promoting apoptosis in vitro. Taken together, LDH-A plays an important role in Taxol-resistance and in cancer treatment that is consistent with the role of LDH-A as an essential tumor maintenance gene. This study provides valuable information for the development of therapeutics for Taxol-resistant breast cancer.
Notes:

Other

2005
1998
1997

Patent

Powered by PublicationsList.org.