36 results on '"Zhihu Ding"'
Search Results
2. Supplementary Table S1 from Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Ronald A. DePinho, Y. Alan Wang, Lynda Chin, Mark J. McArthur, Christopher J. Logothetis, Patricia Troncoso, Qing Chang, Liren Li, Yanxia Shi, Zhihu Ding, Xiaolu Pan, Wantong Yao, Eun-Jung Jin, Baoli Hu, Pingping Hou, Sunada Khadka, Xiaoying Shang, Di Zhao, Tim Heffernan, Trang N. Tieu, Vandhana Ramamoorthy, Zhenglin Guo, Neelay Bhaskar Patel, Chang-Jiun Wu, Avnish Kapoor, Elsa M. Li-Ning-Tapia, Jianhua Zhang, Sujun Hua, Ramakrishna Konaparthi, Kun Zhao, Zhuangna Fang, Shan Jiang, Chia Chin Wu, Pingna Deng, Prasenjit Dey, Xin Lu, and Guocan Wang
- Abstract
Markers used in CyTOF analysis.
- Published
- 2023
3. Supplementary Table S3 from Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Ronald A. DePinho, Y. Alan Wang, Lynda Chin, Mark J. McArthur, Christopher J. Logothetis, Patricia Troncoso, Qing Chang, Liren Li, Yanxia Shi, Zhihu Ding, Xiaolu Pan, Wantong Yao, Eun-Jung Jin, Baoli Hu, Pingping Hou, Sunada Khadka, Xiaoying Shang, Di Zhao, Tim Heffernan, Trang N. Tieu, Vandhana Ramamoorthy, Zhenglin Guo, Neelay Bhaskar Patel, Chang-Jiun Wu, Avnish Kapoor, Elsa M. Li-Ning-Tapia, Jianhua Zhang, Sujun Hua, Ramakrishna Konaparthi, Kun Zhao, Zhuangna Fang, Shan Jiang, Chia Chin Wu, Pingna Deng, Prasenjit Dey, Xin Lu, and Guocan Wang
- Abstract
Genes upregulated in Ptenpc-/-Smad4pc-/- tumors as compared to Ptenpc-/- tumors ({greater than or equal to}2 fold).
- Published
- 2023
4. Supplementary Table S2 from Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Ronald A. DePinho, Y. Alan Wang, Lynda Chin, Mark J. McArthur, Christopher J. Logothetis, Patricia Troncoso, Qing Chang, Liren Li, Yanxia Shi, Zhihu Ding, Xiaolu Pan, Wantong Yao, Eun-Jung Jin, Baoli Hu, Pingping Hou, Sunada Khadka, Xiaoying Shang, Di Zhao, Tim Heffernan, Trang N. Tieu, Vandhana Ramamoorthy, Zhenglin Guo, Neelay Bhaskar Patel, Chang-Jiun Wu, Avnish Kapoor, Elsa M. Li-Ning-Tapia, Jianhua Zhang, Sujun Hua, Ramakrishna Konaparthi, Kun Zhao, Zhuangna Fang, Shan Jiang, Chia Chin Wu, Pingna Deng, Prasenjit Dey, Xin Lu, and Guocan Wang
- Abstract
Detailed pathology description of the Gr1 treated mice and Cxcr2 inhibitor treated mice.
- Published
- 2023
5. Supplementary Table S4 from Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Ronald A. DePinho, Y. Alan Wang, Lynda Chin, Mark J. McArthur, Christopher J. Logothetis, Patricia Troncoso, Qing Chang, Liren Li, Yanxia Shi, Zhihu Ding, Xiaolu Pan, Wantong Yao, Eun-Jung Jin, Baoli Hu, Pingping Hou, Sunada Khadka, Xiaoying Shang, Di Zhao, Tim Heffernan, Trang N. Tieu, Vandhana Ramamoorthy, Zhenglin Guo, Neelay Bhaskar Patel, Chang-Jiun Wu, Avnish Kapoor, Elsa M. Li-Ning-Tapia, Jianhua Zhang, Sujun Hua, Ramakrishna Konaparthi, Kun Zhao, Zhuangna Fang, Shan Jiang, Chia Chin Wu, Pingna Deng, Prasenjit Dey, Xin Lu, and Guocan Wang
- Abstract
Table S4. Genes upregulated in GFP+ tumors cells from Ptenpc-/-Smad4pc-/- mice as compared to Tomato+ cells ({greater than or equal to}4 fold).
- Published
- 2023
6. Supplementary Methods, Figure Legends, Figures S1 - S7 from Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Ronald A. DePinho, Y. Alan Wang, Lynda Chin, Mark J. McArthur, Christopher J. Logothetis, Patricia Troncoso, Qing Chang, Liren Li, Yanxia Shi, Zhihu Ding, Xiaolu Pan, Wantong Yao, Eun-Jung Jin, Baoli Hu, Pingping Hou, Sunada Khadka, Xiaoying Shang, Di Zhao, Tim Heffernan, Trang N. Tieu, Vandhana Ramamoorthy, Zhenglin Guo, Neelay Bhaskar Patel, Chang-Jiun Wu, Avnish Kapoor, Elsa M. Li-Ning-Tapia, Jianhua Zhang, Sujun Hua, Ramakrishna Konaparthi, Kun Zhao, Zhuangna Fang, Shan Jiang, Chia Chin Wu, Pingna Deng, Prasenjit Dey, Xin Lu, and Guocan Wang
- Abstract
Supplementary Figure S1. CyTOF analysis of biological samples from Ptenpc-/-Smad4pc-/- mice (Related to Figure 2). Supplementary Figure S2. Strategy used for MDSCs Isolation (Related to Figure 3). Supplementary Figure S3. Treatment scheme for Gr-1 antibody, peptibody, and Cxcr2 inhibitor SB225002. Supplementary Figure S4. IHC staining of Ki67, CD45, Vimentin, Smooth muscle actin (SMA) and Trichrome staining of mouse prostate tissues treated with IgG control or Gr1 antibody. Supplementary Figure S5. The top 10 differentially expressed genes in Ptenpc-/-Smad4pc-/- tumors as compared to Ptenpc-/- tumors, identified by microarray analysis (n=5). Figure S6. Top 10 activated oncogenic signatures identified by GSEA analysis in Ptenpc-/- Smad4pc-/- tumors as compared to Ptenpc-/- tumors (n=5). Figure S7. Clustering of primary prostate tumors from Wallace et al into MDSC-high and MDSC-low subtypes.
- Published
- 2023
7. Supplementary Table S6 from Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Ronald A. DePinho, Y. Alan Wang, Lynda Chin, Mark J. McArthur, Christopher J. Logothetis, Patricia Troncoso, Qing Chang, Liren Li, Yanxia Shi, Zhihu Ding, Xiaolu Pan, Wantong Yao, Eun-Jung Jin, Baoli Hu, Pingping Hou, Sunada Khadka, Xiaoying Shang, Di Zhao, Tim Heffernan, Trang N. Tieu, Vandhana Ramamoorthy, Zhenglin Guo, Neelay Bhaskar Patel, Chang-Jiun Wu, Avnish Kapoor, Elsa M. Li-Ning-Tapia, Jianhua Zhang, Sujun Hua, Ramakrishna Konaparthi, Kun Zhao, Zhuangna Fang, Shan Jiang, Chia Chin Wu, Pingna Deng, Prasenjit Dey, Xin Lu, and Guocan Wang
- Abstract
Detailed information for the YAP1 IHC staining in human prostate cancers.
- Published
- 2023
8. Application of VR technology in simulated attack training of armed helicopters
- Author
-
Xiaoqin Liu, Xun Yan, Zhihu Ding, and Jun Zhang
- Published
- 2022
9. Oncogenic dependency on β-catenin in liver cancer cell lines correlates with pathway activation
- Author
-
Chaomei Shi, Mikhail Levit, Tai-Guang Jin, Dinesh S. Bangari, Zhihu Ding, Tatiana Tolstykh, Karl Mamaat, Joern Hopke, Susan Ryan, Lan Jiang, Hui Cao, Qunyan Yu, Dietmar Hoffmann, Hui Qu, Liang Schweizer, Sabine Scheidler, Fangxian Sun, Douglas D. Fang, May Cindhuchao, Christopher Winter, Dmitri Wiederschain, Jack Pollard, Mike W. Helms, and Kerstin Jahn-Hofmann
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Cell growth ,Cancer ,hepatocellular carcinoma ,β-catenin ,Biology ,medicine.disease ,phenotypic rescue ,Small hairpin RNA ,03 medical and health sciences ,cell proliferation ,030104 developmental biology ,Downregulation and upregulation ,siRNA ,Hepatocellular carcinoma ,Catenin ,Internal medicine ,AXIN2 ,medicine ,Liver cancer ,Research Paper - Abstract
Hepatocellular carcinoma (HCC) represents a serious public health challenge with few therapeutic options available to cancer patients.Wnt/β-catenin pathway is thought to play a significant role in HCC pathogenesis. In this study, we confirmed high frequency of CTNNB1 (β-catenin) mutations in two independent cohorts of HCC patients and demonstrated significant upregulation of β-catenin protein in the overwhelming majority of HCC patient samples, patient-derived xenografts (PDX) and established cell lines. Using genetic tools validated for target specificity through phenotypic rescue experiments, we went on to investigate oncogenic dependency on β-catenin in an extensive collection of human HCC cells lines. Our results demonstrate that dependency on β-catenin generally tracks with its activation status. HCC cell lines that harbored activating mutations in CTNNB1 or displayed elevated levels of non-phosphorylated (active) β-catenin were significantly more sensitive to β-catenin siRNA treatment than cell lines with wild-type CTNNB1 and lower active β-catenin. Finally, significant therapeutic benefit of β-catenin knock-down was demonstrated in established HCC tumor xenografts using doxycycline-inducible shRNA system. β-catenin downregulation and tumor growth inhibition was associated with reduction in AXIN2, direct transcriptional target of β-catenin, and decreased cancer cell proliferation as measured by Ki67 staining. Taken together, our data highlight fundamental importance of aberrant β-catenin signaling in the maintenance of oncogenic phenotype in HCC.
- Published
- 2017
10. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression
- Author
-
Lynda Chin, Sujun Hua, Y. Alan Wang, Jianhua Zhang, Kun Zhao, Timothy P. Heffernan, Elsa M. Li-Ning-Tapia, Xiaolu Pan, Trang N. Tieu, Eun Jung Jin, Zhihu Ding, Vandhana Ramamoorthy, Prasenjit Dey, Ronald A. DePinho, Wantong Yao, Patricia Troncoso, Sunada Khadka, Pingping Hou, Shan Jiang, Zhuangna Fang, Avnish Kapoor, Ramakrishna Konaparthi, Qing Chang, Yanxia Shi, Christopher J. Logothetis, Xiaoying Shang, Chia Chin Wu, Neelay Bhaskar Patel, Guocan Wang, Xin Lu, Pingna Deng, Mark J. McArthur, Chang-Jiun Wu, Baoli Hu, Zhenglin Guo, Di Zhao, and Liren Li
- Subjects
Male ,0301 basic medicine ,Chemokine CXCL5 ,Stromal cell ,Protein Serine-Threonine Kinases ,Article ,Receptors, Interleukin-8B ,Mice ,03 medical and health sciences ,Prostate cancer ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,PTEN ,Hippo Signaling Pathway ,Myeloid Cells ,CXC chemokine receptors ,Adaptor Proteins, Signal Transducing ,Smad4 Protein ,YAP1 ,biology ,business.industry ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,YAP-Signaling Proteins ,Phosphoproteins ,medicine.disease ,030104 developmental biology ,Oncology ,Tumor progression ,CXCL5 ,Immunology ,Cancer cell ,Disease Progression ,Cancer research ,biology.protein ,business ,Signal Transduction ,Transcription Factors - Abstract
The signaling mechanisms between prostate cancer cells and infiltrating immune cells may illuminate novel therapeutic approaches. Here, utilizing a prostate adenocarcinoma model driven by loss of Pten and Smad4, we identify polymorphonuclear myeloid-derived suppressor cells (MDSC) as the major infiltrating immune cell type, and depletion of MDSCs blocks progression. Employing a novel dual reporter prostate cancer model, epithelial and stromal transcriptomic profiling identified CXCL5 as a cancer-secreted chemokine to attract CXCR2-expressing MDSCs, and, correspondingly, pharmacologic inhibition of CXCR2 impeded tumor progression. Integrated analyses identified hyperactivated Hippo–YAP signaling in driving CXCL5 upregulation in cancer cells through the YAP–TEAD complex and promoting MDSC recruitment. Clinicopathologic studies reveal upregulation and activation of YAP1 in a subset of human prostate tumors, and the YAP1 signature is enriched in primary prostate tumor samples with stronger expression of MDSC-relevant genes. Together, YAP-driven MDSC recruitment via heterotypic CXCL5–CXCR2 signaling reveals an effective therapeutic strategy for advanced prostate cancer. Significance: We demonstrate a critical role of MDSCs in prostate tumor progression and discover a cancer cell nonautonomous function of the Hippo–YAP pathway in regulation of CXCL5, a ligand for CXCR2-expressing MDSCs. Pharmacologic elimination of MDSCs or blocking the heterotypic CXCL5–CXCR2 signaling circuit elicits robust antitumor responses and prolongs survival. Cancer Discov; 6(1); 80–95. ©2015 AACR. This article is highlighted in the In This Issue feature, p. 1
- Published
- 2016
11. Abstract 1377: Identification and validation of WRN as a novel synthetic lethality target in context of microsatellite instability
- Author
-
Bailin Zhang, Jing Zhang, Shujia Dai, Zhuyan Guo, Zhihu Ding, Jennifer Buell, Emma Wang, Steve Rowley, Jack Pollard, Dietmar Hoffmann, May Cindhuchao, Fangxian Sun, Jane Cheng, Alexei Protopopov, Stephane Poirier, Karen Wong, Amanda Lennon, Chandra Sekhar Pedamallu, Bridget Zhou, Joern Hopke, Laurent Debussche, Monsif Bouaboula, and Malvika Koundinya
- Subjects
Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,DNA repair ,nutritional and metabolic diseases ,Microsatellite instability ,Context (language use) ,Synthetic lethality ,Biology ,medicine.disease ,Telomere ,Oncology ,RNA interference ,medicine ,Gene silencing ,Werner syndrome - Abstract
Despite the significant clinical success of the Immuno-Oncology treatment of microsatellite instability (MSI) patients, there remains huge unmet medical need because of mechanisms of resistance. Through an effort to analyze publicly available large-scale shRNA screening and CRISPR screening, we surprisingly found that WRN is a potent synthetic lethality target in the context of MSI. WRN is an enzyme known as the “Werner syndrome ATP-dependent helicase”. WRN is involved in multiple cellular functions, including DNA repair and telomere maintenance. Silencing of WRN by RNAi and CRISPRi in a panel of MSI-H cells lead to tumor cell growth inhibition in vitro and in vivo, thus demonstrated WRN is a novel synthetic lethality target in context of MSI. Our discovery has recently been cross-validated by multiple independent studies1-4. Furthermore, we also report here the first time that silencing of WRN by RNAi and CRISPRi in DLD1, a MSI-H cell, does not lead to tumor cell inhibition. Taken together, our data using independent genetic approach CRISPRi in vitro and in vivo, further highlight fundamental importance of WRN as a synthetic lethality target in some, but not all, MSI context. References: 1 Chan E.M. et al. (2019). Nature. 568(7753):551-556. 2 Behan F.M. et al (2019). Nature. 2019 568(7753):511-516. 3 Kategaya L. et al. (2019). iScience. 13:488-497. 4 Lieb S. et al. (2019). Elife. 8. pii: e43333. Citation Format: Zhihu (Jeff) Ding, Jing Zhang, Chandra Sekhar Pedamallu, Steve Rowley, Jane Cheng, Shujia Dai, Bridget Zhou, Malvika Koundinya, Zhuyan Guo, Stephane Poirier, Joern Hopke, Amanda Lennon, Jennifer Buell, May Cindhuchao, Karen Wong, Emma Wang, Alexei Protopopov, Bailin Zhang, Dietmar Hoffmann, Fangxian Sun, Jack Pollard, Laurent Debussche, Monsif Bouaboula. Identification and validation of WRN as a novel synthetic lethality target in context of microsatellite instability [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 1377.
- Published
- 2020
12. Evaluating a 4-marker signature of aggressive prostate cancer using time-dependent AUC
- Author
-
Meir J. Stampfer, Elizabeth Nuttall, Rosina T. Lis, P.W. Kantoff, Edward C. Stack, Massimo Loda, Zhihu Ding, Travis Gerke, Edward Giovannucci, Lorelei A. Mucci, Neil E. Martin, and Giovanni Parmigiani
- Subjects
Oncology ,medicine.medical_specialty ,education.field_of_study ,biology ,business.industry ,Urology ,Population ,Cancer ,Disease ,medicine.disease ,Bioinformatics ,Prostate cancer ,medicine.anatomical_structure ,Prostate ,Internal medicine ,Cohort ,biology.protein ,Medicine ,PTEN ,business ,education ,Survival analysis - Abstract
BACKGROUND We previously identified a protein tumor signature of PTEN, SMAD4, SPP1, and CCND1 that, together with clinical features, was associated with lethal outcomes among prostate cancer patients. In the current study, we sought to validate the molecular model using time-dependent measures of AUC and predictive values for discriminating lethal from non-lethal prostate cancer. METHODS Using data from the initial study, we fit survival models for men with prostate cancer who were participants in the Physicians' Health Study (PHS; n = 276). Based on these models, we generated prognostic risk scores in an independent population, the Health Professionals Follow-up Study (HPFS; n = 347) to evaluate external validity. In each cohort, men were followed prospectively from cancer diagnosis through 2011 for development of distant metastasis or cancer mortality. We measured protein tumor expression of PTEN, SMAD4, SPP1, and CCND1 on tissue microarrays. RESULTS During a median of 11.9 and 14.3 years follow-up in the PHS and HPFS cohorts, 24 and 32 men (9%) developed lethal disease. When used as a prognostic factor in a new population, addition of the four markers to clinical variables did not improve discriminatory accuracy through 15 years of follow-up. CONCLUSIONS Although the four markers have been identified as key biological mediators in metastatic progression, they do not provide independent, long-term prognostic information beyond clinical factors when measured at diagnosis. This finding may underscore the broad heterogeneity in aggressive prostate tumors and highlight the challenges that may result from overfitting in discovery-based research. Prostate 75:1926–1933, 2015. © 2015 Wiley Periodicals, Inc.
- Published
- 2015
13. Antitelomerase Therapy Provokes ALT and Mitochondrial Adaptive Mechanisms in Cancer
- Author
-
Hailei Zhang, Ronald A. DePinho, Yaoqi Alan Wang, Maria Kost-Alimova, Marc Liesa, Soyoon Sarah Hwang, Lynda Chin, Haoqiang Ying, Mariela Jaskelioff, Adam T. Boutin, Zhihu Ding, Shawn F. Johnson, Orian S. Shirihai, Elena Ivanova, Ergun Sahin, Boyi Gan, Alexei Protopopov, and Jian Hu
- Subjects
0303 health sciences ,Telomerase ,Biochemistry, Genetics and Molecular Biology(all) ,SOD2 ,Cancer ,Mitochondrion ,Biology ,medicine.disease ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,3. Good health ,Telomere ,03 medical and health sciences ,0302 clinical medicine ,Telomere Homeostasis ,Mitochondrial biogenesis ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Telomerase reverse transcriptase ,030304 developmental biology - Abstract
SummaryTo assess telomerase as a cancer therapeutic target and determine adaptive mechanisms to telomerase inhibition, we modeled telomerase reactivation and subsequent extinction in T cell lymphomas arising in Atm−/− mice engineered with an inducible telomerase reverse transcriptase allele. Telomerase reactivation in the setting of telomere dysfunction enabled full malignant progression with alleviation of telomere dysfunction-induced checkpoints. These cancers possessed copy number alterations targeting key loci in human T cell lymphomagenesis. Upon telomerase extinction, tumor growth eventually slowed with reinstatement of telomere dysfunction-induced checkpoints, yet growth subsequently resumed as tumors acquired alternative lengthening of telomeres (ALT) and aberrant transcriptional networks centering on mitochondrial biology and oxidative defense. ALT+ tumors acquired amplification/overexpression of PGC-1β, a master regulator of mitochondrial biogenesis and function, and they showed marked sensitivity to PGC-1β or SOD2 knockdown. Genetic modeling of telomerase extinction reveals vulnerabilities that motivate coincidental inhibition of mitochondrial maintenance and oxidative defense mechanisms to enhance antitelomerase cancer therapy.
- Published
- 2012
- Full Text
- View/download PDF
14. SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression
- Author
-
Sabina Signoretti, Yujin Hoshida, Hongwu Zheng, Samuel R. Perry, Gerald C. Chu, Emma S. Labrot, Todd R. Golub, Dennis Ho, Kenneth L. Scott, Massimo Loda, David Hiller, Ronald A. DePinho, Y. Alan Wang, Wing Hung Wong, Alexander H. Stegh, Chang-Jiun Wu, Xiaoqiu Wu, Shan Jiang, Lorelei A. Mucci, Lynda Chin, J. Zhang, Nabeel Bardeesy, Baoli Hu, Rosina T. Lis, Zhihu Ding, Yonghong Xiao, David E. Hill, and Meir J. Stampfer
- Subjects
Male ,Biochemical recurrence ,Lung Neoplasms ,Mice, Transgenic ,Penetrance ,Models, Biological ,Metastasis ,Mice ,Prostate cancer ,Transforming Growth Factor beta ,Prostate ,medicine ,Animals ,Humans ,PTEN ,Cyclin D1 ,Genes, Tumor Suppressor ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Cell Proliferation ,Smad4 Protein ,Multidisciplinary ,biology ,Gene Expression Profiling ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,Cancer ,Prostate-Specific Antigen ,Prognosis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Prostate-specific antigen ,medicine.anatomical_structure ,Tumor progression ,Lymphatic Metastasis ,Bone Morphogenetic Proteins ,Immunology ,Disease Progression ,biology.protein ,Cancer research ,Osteopontin - Abstract
Effective clinical management of prostate cancer (PCA) has been challenged by significant intratumoural heterogeneity on the genomic and pathological levels and limited understanding of the genetic elements governing disease progression. Here, we exploited the experimental merits of the mouse to test the hypothesis that pathways constraining progression might be activated in indolent Pten-null mouse prostate tumours and that inactivation of such progression barriers in mice would engender a metastasis-prone condition. Comparative transcriptomic and canonical pathway analyses, followed by biochemical confirmation, of normal prostate epithelium versus poorly progressive Pten-null prostate cancers revealed robust activation of the TGFβ/BMP-SMAD4 signalling axis. The functional relevance of SMAD4 was further supported by emergence of invasive, metastatic and lethal prostate cancers with 100% penetrance upon genetic deletion of Smad4 in the Pten-null mouse prostate. Pathological and molecular analysis as well as transcriptomic knowledge-based pathway profiling of emerging tumours identified cell proliferation and invasion as two cardinal tumour biological features in the metastatic Smad4/Pten-null PCA model. Follow-on pathological and functional assessment confirmed cyclin D1 and SPP1 as key mediators of these biological processes, which together with PTEN and SMAD4, form a four-gene signature that is prognostic of prostate-specific antigen (PSA) biochemical recurrence and lethal metastasis in human PCA. This model-informed progression analysis, together with genetic, functional and translational studies, establishes SMAD4 as a key regulator of PCA progression in mice and humans.
- Published
- 2011
15. Abstract 2489: Sensitivity of liver cancer cell lines to B-catenin knock-down correlates with pathway activation
- Author
-
Dinesh S. Bangari, Zhihu Ding, Joern Hopke, Tatiana Tolstykh, Dmitri Wiederschain, May Cindhuchao, Hui Qu, Chaomei Shi, Mike W Helms, Christopher Winter, Kerstin Jahn-Hofmann, Karl Mamaat, Susan Ryan, Liang Schweizer, Lan Jiang, Fangxian Sun, Qunyan Yu, Sabine Scheidler, Mikhail Levit, Jack Pollard, Dietmar Hoffmann, Hui Cao, Tai-Guang Jin, and Douglas D. Fang
- Subjects
Sorafenib ,Cancer Research ,Cell growth ,business.industry ,Wnt signaling pathway ,medicine.disease ,chemistry.chemical_compound ,Oncology ,chemistry ,Catenin ,Hepatocellular carcinoma ,Regorafenib ,AXIN2 ,Cancer research ,Medicine ,business ,Liver cancer ,medicine.drug - Abstract
Introduction: Hepatocellular carcinoma (HCC) is the 3rd leading cause of cancer-related mortality. Currently there are few treatment options available for advanced HCC patients. Multi-kinase inhibitors sorafenib and regorafenib are the only approved systemic HCC therapies and are only marginally effective in extending survival. HCC is largely driven by difficult-to-drug oncogenes, including β-catenin, and none of the current HCC therapeutics on the market or in development addresses these major genetic drivers of disease. β-catenin is an integral part of the WNT signaling pathway and plays a major role in regulation of cell survival, apoptosis and developmental processes. The goal of this study was to systematically interrogate contribution of β-catenin to HCC pathogenesis. Methods: The extent of genomic alterations in β-catenin itself or components of its regulatory network was interrogated in two large HCC data sets, The Cancer Genome Atlas and HCC cohort from Asan Medical Center. β-catenin in vitro credentialing (target knock-down, PD modulation and cell growth inhibition) was carried out in cultured HCC cell lines using multiple siRNAs against β-catenin. Stably expressed doxycycline-inducible shRNAs targeting β-catenin were used to establish in vivo dependency in HCC tumor xenografts. Results: Building on previous observations, we detected high frequency of somatic mutations in CTNNB1/β-catenin itself and in constituent members of its regulatory network in two independent cohorts of HCC patients. Furthermore, significant upregulation in β-catenin protein levels was detected in the overwhelming majority of HCC patient samples, patient-derived xenografts and established cell lines. Using genetic tools validated for target specificity through phenotypic rescue experiments, we discovered that dependency on β-catenin in human HCC cell lines generally tracks with its activation status. HCC cell lines that harbored activating mutations in CTNNB1/β-catenin or displayed elevated levels of non-phosphorylated (active) β-catenin were significantly more sensitive to β-catenin siRNA treatment than cell lines with wild-type CTNNB1/β-catenin and lower active β-catenin. Finally, significant therapeutic benefit of β-catenin knock-down was demonstrated in established HCC tumor xenografts using doxycycline-inducible shRNA system. β-catenin downregulation and tumor growth inhibition was associated with reduction in PD marker AXIN2, and decreased cancer cell proliferation. Conclusions: β-catenin is an important therapeutic target in HCC that could potentially be targeted using therapeutic siRNA suitably formulated for delivery to human liver tumors. Genomic alterations in CTNNB1/β-catenin, as well as increased abundance of non-phosphorylated (active) β-catenin, may serve as predictive biomarkers for patient selection. Citation Format: Zhihu (Jeff) Ding, Chaomei Shi, Lan Jiang, Tatiana Tolstykh, Hui Cao, Dinesh Bangari, Susan Ryan, Taiguang Jin, Mikhail Levit, Karl Mamaat, Qunyan Yu, Hui Qu, Joern Hopke, May Cindhuchao, Dietmar Hoffmann, Fangxian Sun, Mike Helms, Kerstin Jahn-Hofmann, Sabine Scheidler, Douglas Fang, Liang Schweizer, Jack Pollard, Christopher Winter, Dmitri Wiederschain. Sensitivity of liver cancer cell lines to B-catenin knock-down correlates with pathway activation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2489.
- Published
- 2018
16. FoxOs Cooperatively Regulate Diverse Pathways Governing Neural Stem Cell Homeostasis
- Author
-
Wing Hung Wong, Walid S. Kamoun, Lynda Chin, Hongwu Zheng, Jed Mahoney, Jihye Paik, Shan Jiang, Shakti Ramkissoon, Alexei Protopopov, Florian L. Muller, Rujuta Narurkar, Zhihu Ding, David Hiller, Haoqiang Ying, Sung Suk Chae, Ronald A. DePinho, and Keith L. Ligon
- Subjects
Regulation of gene expression ,Cellular differentiation ,Neurogenesis ,FOXO Family ,Cell Biology ,Biology ,STEMCELL ,Article ,Neural stem cell ,Cell biology ,Genetics ,Molecular Medicine ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Progenitor cell ,Protein kinase B - Abstract
SummaryThe PI3K-AKT-FoxO pathway is integral to lifespan regulation in lower organisms and essential for the stability of long-lived cells in mammals. Here, we report the impact of combined FoxO1, 3, and 4 deficiencies on mammalian brain physiology with a particular emphasis on the study of the neural stem/progenitor cell (NSC) pool. We show that the FoxO family plays a prominent role in NSC proliferation and renewal. FoxO-deficient mice show initial increased brain size and proliferation of neural progenitor cells during early postnatal life, followed by precocious significant decline in the NSC pool and accompanying neurogenesis in adult brains. Mechanistically, integrated transcriptomic, promoter, and functional analyses of FoxO-deficient NSC cultures identified direct gene targets with known links to the regulation of human brain size and the control of cellular proliferation, differentiation, and oxidative defense. Thus, the FoxO family coordinately regulates diverse genes and pathways to govern key aspects of NSC homeostasis in the mammalian brain.
- Published
- 2009
17. Pten and p53 Converge on c-Myc to Control Differentiation, Self-renewal, and Transformation of Normal and Neoplastic Stem Cells in Glioblastoma
- Author
-
Wing Hung Wong, Mingjian James You, Yaoqi Alan Wang, Giovanni Tonon, Cameron Brennan, Gerald C. Chu, Ruprecht Wiedemeyer, Haoqiang Ying, An Jou Chen, Samuel R. Perry, Haiyan Yan, Lynda Chin, Jayne M. Stommel, Hongwu Zheng, Ronald A. DePinho, David Hiller, Keith L. Ligon, Zhihu Ding, Alec C. Kimmelman, and Katherine Dunn
- Subjects
Angiogenesis ,Models, Neurological ,Genes, myc ,Mice, Transgenic ,Biochemistry ,Transcriptome ,Mice ,Species Specificity ,Glioma ,Genetics ,medicine ,Animals ,Humans ,PTEN ,Progenitor cell ,Molecular Biology ,Cell Proliferation ,biology ,Brain Neoplasms ,PTEN Phosphohydrolase ,Cell Differentiation ,Genes, p53 ,medicine.disease ,Phenotype ,Mice, Mutant Strains ,Neural stem cell ,nervous system diseases ,Disease Models, Animal ,Cell Transformation, Neoplastic ,Mutation ,Immunology ,Neoplastic Stem Cells ,Cancer research ,biology.protein ,Stem cell ,Glioblastoma - Abstract
Glioblastoma (GBM) is a highly lethal primary brain cancer with hallmark features of diffuse invasion, intense apoptosis resistance and florid necrosis, robust angiogenesis, and an immature profile with developmental plasticity. In the course of assessing the developmental consequences of central nervous system (CNS)-specific deletion of p53 and Pten, we observed a penetrant acute-onset malignant glioma phenotype with striking clinical, pathological, and molecular resemblance to primary GBM in humans. This primary, as opposed to secondary, GBM presentation in the mouse prompted genetic analysis of human primary GBM samples that revealed combined p53 and Pten mutations as the most common tumor suppressor defects in primary GBM. On the mechanistic level, the "multiforme" histopathological presentation and immature differentiation marker profile of the murine tumors motivated transcriptomic promoter-binding element and functional studies of neural stem cells (NSCs), which revealed that dual, but not singular, inactivation of p53 and Pten promotes cellular c-Myc activation. This increased c-Myc activity is associated not only with impaired differentiation, enhanced self-renewal capacity of NSCs, and tumor-initiating cells (TICs), but also with maintenance of TIC tumorigenic potential. Together, these murine studies have provided a highly faithful model of primary GBM, revealed a common tumor suppressor mutational pattern in human disease, and established c-Myc as a key component of p53 and Pten cooperative actions in the regulation of normal and malignant stem/progenitor cell differentiation, self-renewal, and tumorigenic potential.
- Published
- 2008
18. FoxOs Are Lineage-Restricted Redundant Tumor Suppressors and Regulate Endothelial Cell Homeostasis
- Author
-
Gerald C. Chu, Daniel R. Carrasco, Diego H. Castrillon, Zuzana Tothova, Ronald A. DePinho, James W. Horner, Yonghong Xiao, Zhihu Ding, Hongkai Ji, Wing Hung Wong, Ramya Kollipara, D. Gary Gilliland, Lili Miao, Lynda Chin, Shan Jiang, and Jihye Paik
- Subjects
Cell type ,endocrine system ,Lymphoma ,Somatic cell ,Cellular differentiation ,Endothelial cell morphogenesis ,Neovascularization, Physiologic ,Cell Cycle Proteins ,Nerve Tissue Proteins ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Phosphatidylinositol 3-Kinases ,Animals ,Drosophila Proteins ,Homeostasis ,Cell Lineage ,Transcription factor ,PI3K/AKT/mTOR pathway ,Homeodomain Proteins ,Mice, Knockout ,Forkhead Box Protein O1 ,Biochemistry, Genetics and Molecular Biology(all) ,Tumor Suppressor Proteins ,fungi ,Forkhead Box Protein O3 ,Pre-B-Cell Leukemia Transcription Factor 1 ,FOXO Family ,Endothelial Cells ,Cell Differentiation ,Forkhead Transcription Factors ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,FOXO4 ,embryonic structures ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Hemangioma ,Proto-Oncogene Proteins c-akt ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction ,Transcription Factors - Abstract
SummaryActivated phosphoinositide 3-kinase (PI3K)-AKT signaling appears to be an obligate event in the development of cancer. The highly related members of the mammalian FoxO transcription factor family, FoxO1, FoxO3, and FoxO4, represent one of several effector arms of PI3K-AKT signaling, prompting genetic analysis of the role of FoxOs in the neoplastic phenotypes linked to PI3K-AKT activation. While germline or somatic deletion of up to five FoxO alleles produced remarkably modest neoplastic phenotypes, broad somatic deletion of all FoxOs engendered a progressive cancer-prone condition characterized by thymic lymphomas and hemangiomas, demonstrating that the mammalian FoxOs are indeed bona fide tumor suppressors. Transcriptome and promoter analyses of differentially affected endothelium identified direct FoxO targets and revealed that FoxO regulation of these targets in vivo is highly context-specific, even in the same cell type. Functional studies validated Sprouty2 and PBX1, among others, as FoxO-regulated mediators of endothelial cell morphogenesis and vascular homeostasis.
- Published
- 2007
- Full Text
- View/download PDF
19. Evaluating a 4-marker signature of aggressive prostate cancer using time-dependent AUC
- Author
-
Travis A, Gerke, Neil E, Martin, Zhihu, Ding, Elizabeth J, Nuttall, Edward C, Stack, Edward, Giovannucci, Rosina T, Lis, Meir J, Stampfer, Phillip W, Kantoff, Giovanni, Parmigiani, Massimo, Loda, and Lorelei A, Mucci
- Subjects
Aged, 80 and over ,Male ,Gene Expression Profiling ,PTEN Phosphohydrolase ,Prostate ,Prostatic Neoplasms ,Middle Aged ,Prognosis ,Article ,Area Under Curve ,Biomarkers, Tumor ,Disease Progression ,Humans ,Cyclin D1 ,Neoplasm Invasiveness ,Osteopontin ,Aged ,Follow-Up Studies ,Smad4 Protein - Abstract
We previously identified a protein tumor signature of PTEN, SMAD4, SPP1, and CCND1 that, together with clinical features, was associated with lethal outcomes among prostate cancer patients. In the current study, we sought to validate the molecular model using time-dependent measures of AUC and predictive values for discriminating lethal from non-lethal prostate cancer.Using data from the initial study, we fit survival models for men with prostate cancer who were participants in the Physicians' Health Study (PHS; n = 276). Based on these models, we generated prognostic risk scores in an independent population, the Health Professionals Follow-up Study (HPFS; n = 347) to evaluate external validity. In each cohort, men were followed prospectively from cancer diagnosis through 2011 for development of distant metastasis or cancer mortality. We measured protein tumor expression of PTEN, SMAD4, SPP1, and CCND1 on tissue microarrays.During a median of 11.9 and 14.3 years follow-up in the PHS and HPFS cohorts, 24 and 32 men (9%) developed lethal disease. When used as a prognostic factor in a new population, addition of the four markers to clinical variables did not improve discriminatory accuracy through 15 years of follow-up.Although the four markers have been identified as key biological mediators in metastatic progression, they do not provide independent, long-term prognostic information beyond clinical factors when measured at diagnosis. This finding may underscore the broad heterogeneity in aggressive prostate tumors and highlight the challenges that may result from overfitting in discovery-based research.
- Published
- 2015
20. Utilizing Murine Inducible Telomerase Alleles in the Studies of Tissue Degeneration/Regeneration and Cancer
- Author
-
Alexei Protopopov, Mariela Jaskelioff, Zhihu Ding, Liang Yuan, Jian Hu, Takashi Shingu, and Maria Kost-Alimova
- Subjects
Genome instability ,Male ,Telomerase ,General Chemical Engineering ,mTERT-ER ,In situ hybridization ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Neoplasms ,medicine ,Animals ,Regeneration ,Telomerase reverse transcriptase ,Gene Knock-In Techniques ,Alleles ,Cells, Cultured ,In Situ Hybridization, Fluorescence ,030304 developmental biology ,Issue 98 ,LSL-mTERT ,Cancer ,0303 health sciences ,General Immunology and Microbiology ,medicine.diagnostic_test ,General Neuroscience ,Telomere ,Molecular biology ,Neural stem cell ,3. Good health ,Mice, Inbred C57BL ,Ageing ,Cancer research ,Medicine ,Female ,Carcinogenesis ,030217 neurology & neurosurgery ,Fluorescence in situ hybridization - Abstract
Telomere dysfunction-induced loss of genome integrity and its associated DNA damage signaling and checkpoint responses are well-established drivers that cause tissue degeneration during ageing. Cancer, with incidence rates greatly increasing with age, is characterized by short telomere lengths and high telomerase activity. To study the roles of telomere dysfunction and telomerase reactivation in ageing and cancer, the protocol shows how to generate two murine inducible telomerase knock-in alleles 4-Hydroxytamoxifen (4-OHT)-inducible TERT-Estrogen Receptor (mTERT-ER) and Lox-Stopper-LoxTERT (LSL-mTERT). The protocol describes the procedures to induce telomere dysfunction and reactivate telomerase activity in mTERT-ER and LSL-mTERT mice in vivo. The representative data show that reactivation of telomerase activity can ameliorate the tissue degenerative phenotypes induced by telomere dysfunction. In order to determine the impact of telomerase reactivation on tumorigenesis, we generated prostate tumor model G4 PB-Cre4 Pten(L/L) p53(L/L) LSL-mTERT(L/L) and thymic T-cell lymphoma model G4 Atm(-/-) mTERT(ER/ER). The representative data show that telomerase reactivation in the backdrop of genomic instability induced by telomere dysfunction can greatly enhance tumorigenesis. The protocol also describes the procedures used to isolate neural stem cells (NSCs) from mTERT-ER and LSL-mTERT mice and reactivate telomerase activity in NSCs in vitro. The representative data show that reactivation of telomerase can enhance the self-renewal capability and neurogenesis in vitro. Finally, the protocol describes the procedures for performing telomere FISH (Fluorescence In Situ Hybridization) on both mouse FFPE (Formalin Fixed and Paraffin Embedded) brain tissues and metaphase chromosomes of cultured cells.
- Published
- 2015
21. Haploinsufficiency of the mSds3 chromatin regulator promotes chromosomal instability and cancer only upon complete neutralization of p53
- Author
-
Lili Miao, Patricia M. Finnerty, G. M. Turner, Ronald A. DePinho, Zhihu Ding, Ruben D. Carrasco, Natalie E. Simpson, Jan-Hermen Dannenberg, and Gregory David
- Subjects
Mice, Knockout ,Cancer Research ,Tumor suppressor gene ,Aneuploidy ,Biology ,medicine.disease ,medicine.disease_cause ,Molecular biology ,Cell Line ,Chromatin ,Cell biology ,Repressor Proteins ,Mice ,Haplotypes ,Chromosomal Instability ,Chromosome instability ,Genetics ,medicine ,Animals ,Histone deacetylase activity ,Tumor Suppressor Protein p53 ,Haploinsufficiency ,Carcinogenesis ,Molecular Biology ,Pericentric heterochromatin - Abstract
The mSin3 corepressor complex has been linked to diverse cancer signaling pathways through its capacity to regulate target gene expression via chromatin modification. mSds3, a cell essential gene, is a key component of the mSin3 complex serving to maintain its inherent histone deacetylase activity. mSds3 also serves an essential role in the establishment of pericentric heterochromatin, and genetic ablation of mSds3 results in chromosome missegregation. In contrast, mSin3A nullizygous cells show normal chromosome dynamics and cytogenetic profiles. The integral role of mSds3 in controlling chromosome segregation and mSin3-regulated transcriptional networks prompted efforts to determine the neoplastic impact of loss of one copy of mSds3 or mSin3A. In particular, we assessed whether loss of one copy of mSds3, alone or in combination with p53 mutation, results in aneuploidy and promotes a cancer-prone condition in the mouse. We observe that, in a p53 null background, loss of one mSds3 allele results in accelerated tumor onset and increased tumor burden. Notably, these mSds3(+/-) p53(-/-) tumors exhibit a more complex cytogenetic profile characterized by marked aneuploidy and centromeric associations. The presence of even one copy of p53 is sufficient to suppress the accelerated tumorigenesis in mSds3(+/-) mice, consistent with a key role for p53 in monitoring mitotic fidelity. These observations with Sds3 mutant mice contrast with mSin3A(+/-) p53(-/-) mice, which do not show an accelerated or increased tumor incidence relative to mSin3A(+/+)p53(-/-) controls, correlating with the absence of aneuploidy detected upon mSin3A genetic inactivation. This genetic study establishes that the capacity of mSds3 to cooperate with p53 deficiency in cancer predisposition relates to its specific role in chromosome segregation, rather than its central role in maintaining a functional mSin3A complex.
- Published
- 2006
22. The alkaloid sanguinarine is effective against multidrug resistance in human cervical cells via bimodal cell death
- Author
-
Priya Weerasinghe, Xiaolong Yang, Zhihu Ding, Alan Pater, Shou Ching Tang, and Andrejs Liepins
- Subjects
Programmed cell death ,Cell Survival ,Poly ADP ribose polymerase ,Cell ,Apoptosis ,Cervix Uteri ,Biochemistry ,chemistry.chemical_compound ,Alkaloids ,medicine ,Humans ,Sanguinarine ,Cells, Cultured ,Caspase ,Benzophenanthridines ,Pharmacology ,Dose-Response Relationship, Drug ,biology ,Caspase 3 ,Isoquinolines ,Drug Resistance, Multiple ,Intercalating Agents ,Enzyme Activation ,Multiple drug resistance ,medicine.anatomical_structure ,chemistry ,Caspases ,Cancer research ,biology.protein ,Female ,Trypan blue - Abstract
Sanguinarine, a benzophenanthrine alkaloid, is potentially antineoplastic through induction of cell death pathways. The development of multidrug resistance (MDR) is a major obstacle to the success of chemotherapeutic agents. The aim of this study was to investigate whether sanguinarine is effective against uterine cervical MDR and, if so, by which mechanism. The effects of treatment with sanguinarine on human papillomavirus (HPV) type 16-immortalized endocervical cells and their MDR counterpart cells were compared. Trypan blue exclusion assays and clonogenic survival assays demonstrated that MDR human cervical cells are as sensitive as their drug-sensitive parental cells to death induced by sanguinarine. Upon treatment of both types of cells with sanguinarine, two distinct concentration-dependent modes of cell death were observed. Treatment with 2.12 or 4.24 microM sanguinarine induced death in most cells that was characterized as apoptosis using the criteria of cell surface blebbing, as determined by light and scanning electron microscopy, and proteolytic activation of caspase-3 and cleavage of the caspase-3 substrate poly(ADP-ribose) polymerase (PARP), as detected by Western blot analysis. However, 8.48 and 16.96 microM sanguinarine caused a second mode of cell death, oncosis, distinguished by cell surface blistering, and neither caspase-3 activation nor PARP cleavage. This study provides the first evidence that sanguinarine is effective against MDR in cervical cells via bimodal cell death, which displays alternative mechanisms involving different morphologies and caspase-3 activation status.
- Published
- 2002
23. Retinoic Acid Inhibits Telomerase Activity and Downregulates Expression but Does Not Affect Splicing of hTERT: Correlation with Cell Growth Rate Inhibition in an in Vitro Cervical Carcinogenesis/Multidrug-Resistance Model
- Author
-
Xiaolong Yang, Garry Chernenko, Zhihu Ding, Adam G Green, Alan Pater, and Shou Ching Tang
- Subjects
Telomerase ,Retinoic acid ,Down-Regulation ,Uterine Cervical Neoplasms ,Antineoplastic Agents ,Tretinoin ,Cervix Uteri ,Biology ,medicine.disease_cause ,Models, Biological ,chemistry.chemical_compound ,medicine ,Humans ,Telomerase reverse transcriptase ,neoplasms ,Cells, Cultured ,Cell Line, Transformed ,Gene Expression Profiling ,RNA-Binding Proteins ,Cell Biology ,Molecular biology ,Drug Resistance, Multiple ,Reverse transcriptase ,In vitro ,DNA-Binding Proteins ,Enzyme Activation ,Multiple drug resistance ,Alternative Splicing ,Cell Transformation, Neoplastic ,chemistry ,Drug Resistance, Neoplasm ,Cell culture ,Female ,Carrier Proteins ,Carcinogenesis ,Cell Division ,HeLa Cells - Abstract
Telomerase, a ribonucleoprotein complex of hTERT, hTR, and TP1, has been reported to be associated with carcinogenesis and multidrug resistance (MDR). This study used our in vitro human cervical multistep carcinogenesis/MDR model system in which normal human ectocervical and endocervical (HEN) cells were immortalized by HPV18 or 16, respectively, and subsequently transformed. The first evidence was found that immortalization and telomerase activation were correlated with increased expression specifically of two of the hTERT alternatively spliced mRNAs, one encoding wild-type protein containing the full-length functional reverse transcriptase (RT) region and one encoding a defective RT protein. Expression of neither hTERT mRNA containing full-length functional or defective RT motif was affected by transformation/MDR. All-trans-retinoic acid (ATRA) treatment of HPV-immortalized HEN-16-2 cells and transformed/MDR HEN-16-2/CDDP cells inhibited telomerase activity and downregulated expression of hTERT mRNAs containing full-length functional and a defective RT motif, but there were no changes in hTR and TP1 expression. Moreover, ATRA inhibited cell growth rate of HEN-16-2 and HEN-16-2/CDDP cells equally. These results provided the first evidence that ATRA equally in both immortalized and transformed/MDR cell lines inhibits telomerase activity and downregulates expression, but not splicing, of hTERT, and this is correlated with cell growth rate inhibition; the potential is implicated for applying ATRA to hTERT-targeted treatment of cervical cell carcinogenesis/MDR.
- Published
- 2002
24. BAG-1 Promotes Apoptosis Induced by N-(4-hydroxyphenyl)retinamide in Human Cervical Carcinoma Cells
- Author
-
Alan Pater, Xiaolong Yang, Yawei Hao, and Zhihu Ding
- Subjects
Fenretinide ,medicine.drug_class ,Retinoic acid ,Antineoplastic Agents ,Apoptosis ,macromolecular substances ,Biology ,Transfection ,chemistry.chemical_compound ,Ubiquitin ,Tumor Cells, Cultured ,medicine ,Humans ,Retinoid ,Caspase 3 ,Cell Biology ,Staurosporine ,Molecular biology ,In vitro ,Protein Structure, Tertiary ,Hsp70 ,DNA-Binding Proteins ,carbohydrates (lipids) ,chemistry ,Cell culture ,Caspases ,Cancer cell ,Cancer research ,biology.protein ,bacteria ,Carrier Proteins ,Transcription Factors - Abstract
N-(4-hydroxyphenyl)retinamide (4-HPR) is a synthetic apoptosis-inducing retinoid with cancer chemopreventive properties and lower toxicity than all-trans retinoic acid. BAG-1 is an antiapoptotic gene that is overexpressed in cervical and other cancers. In this study, we examined whether BAG-1 can inhibit 4-HPR-induced apoptosis in the C33A cervical carcinoma cell line. Surprisingly, although it inhibited apoptosis induced by five different apoptotic stimuli, overexpression of BAG-1 enhanced apoptosis induced by 4-HPR, producing a 2.5-fold lower IC(50) of 4-HPR. The effects of BAG-1 on 4-HPR-induced apoptosis were mediated by enhancing the caspase-3 activation pathway. Deletion mutation experiments showed that the central ubiquitin homology domain of BAG-1 protein was necessary for its promotion of 4-HPR-induced apoptosis, whereas its C-terminal Hsp70/Hsc70-interacting domain was required for its inhibition of staurosporine-induced apoptosis. These in vitro results suggest that the effectiveness of 4-HPR against the development of malignancy may be due to the overexpression of BAG-1 in cancer cells.
- Published
- 2000
25. Human papillomavirus type 16-immortalized endocervical cells selected for resistance to cisplatin are malignantly transformed and have a multidrug resistance phenotype
- Author
-
Zhihu Ding, Garry Chernenko, Shou Ching Tang, Xiaolong Yang, and Alan Pater
- Subjects
Cisplatin ,Cancer Research ,Drug resistance ,Biology ,Virology ,In vitro ,Malignant transformation ,Multiple drug resistance ,chemistry.chemical_compound ,Oncology ,Paclitaxel ,chemistry ,medicine ,Cancer research ,Doxorubicin ,Etoposide ,medicine.drug - Abstract
Cis-diamminedichloroplatinum (II) (cisplatin, CDDP) is a highly effective chemotherapeutic agent against cervical cancer, but drug resistance is a major obstacle in its clinical application. The mechanism of drug resistance in human cervical cancer is not well understood. Here, we established an in vitro endocervical, cisplatin-resistant cell system that mimics the development of cisplatin resistance in the human cervix. Human papillomavirus (HPV) type 16-immortalized human endocervical cells (HEN-16-2) were treated with cisplatin, and the cisplatin-selected cells (HEN-16-2/CDDP) were resistant to cisplatin, paclitaxel, actinomycin D, doxorubicin, etoposide, and 5-fluorouracil, thus demonstrating a multidrug resistance (MDR) phenotype. Furthermore, compared with a similar passage of drug-sensitive HEN-16-2 cells, HEN-16-2/CDDP cells exhibited the general growth characteristics of cancer cell lines: faster growth in medium containing serum and high calcium levels, higher saturation density, anchorage-independent growth, and formation of tumors in nude mice. These results provided the first in vitro evidence that cisplatin selection can transform HPV-immortalized endocervical cells and cause a phenotype of MDR.
- Published
- 2000
26. New and newly assigned species of the genusDentatisyllis(Polychaeta, Syllidae, Syllinae), with comments on the reproduction, together with a key and a synoptic table of all species of the genus
- Author
-
Wilfried Westheide, Frank Licher, and Zhihu Ding
- Subjects
Dorsum ,Ecology ,media_common.quotation_subject ,Zoology ,Aquatic Science ,Biology ,Chaeta ,Secondary tooth ,Typosyllis ,Genus ,Table (landform) ,Key (lock) ,Reproduction ,media_common - Abstract
Three new Dentatisyllis species, D. hongkongensis sp. nov. and D. mortoni sp. nov., each from sandy tidal beaches of Hong Kong, and D. uebelackerae sp. nov. from Florida are described. D. hongkongensis sp. nov. is distinguished from other species of the genus in having antennae, peristomial and dorsal cirri composed of numerous articles, falcigerous blades of compound chaetae with enlarged secondary tooth, and lacking spines subdistally. D. mortoni sp. nov. has the unique combination of compound dorsalmost pseudospinigers, simple bifid chaetae, and antennae, peristomial and dorsal cirri possessing few articles; details of the viviparous reproduction of this species are given. D. uebelackerae sp. nov. is the Dentatisyllis sp. A of UEBELACKER (1984); it differs from all other species of the genus in the high number of proventricular muscle cell rings. Syllis inflata MARENZELLER, 1879 and Typosyllis (Langerhansia) kiaorensis HARTMANN-SCHRoDER, 1992 are assigned to the genus Dentatisyllis. A key to a...
- Published
- 1998
27. FoxOs enforce a progression checkpoint to constrain mTORC1-activated renal tumorigenesis
- Author
-
Boyi Gan, Eliot Fletcher-Sananikone, Ronald A. DePinho, Hongwu Zheng, Gerald C. Chu, Li Zhuang, Sabina Signoretti, Michelle Chang, William G. Kaelin, Shan Jiang, Sujun Hua, Zhihu Ding, David J. Kwiatkowski, Carol Lim, Michael Collins, Y. Alan Wang, and Jian Hu
- Subjects
Transcriptional Activation ,Cancer Research ,Apoptosis ,mTORC1 ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,medicine.disease_cause ,urologic and male genital diseases ,Article ,Tuberous Sclerosis Complex 1 Protein ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Renal cell carcinoma ,microRNA ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Tumor Cells, Cultured ,Animals ,Humans ,Carcinoma, Renal Cell ,030304 developmental biology ,0303 health sciences ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,fungi ,Cancer ,Proteins ,Forkhead Transcription Factors ,Cell Biology ,medicine.disease ,Kidney Neoplasms ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Multiprotein Complexes ,embryonic structures ,Cancer research ,TSC1 ,biological phenomena, cell phenomena, and immunity ,Carcinogenesis ,Clear cell ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
SummarymTORC1 is a validated therapeutic target for renal cell carcinoma (RCC). Here, analysis of Tsc1-deficient (mTORC1 hyperactivation) mice uncovered a FoxO-dependent negative feedback circuit constraining mTORC1-mediated renal tumorigenesis. We document robust FoxO activation in Tsc1-deficient benign polycystic kidneys and FoxO extinction on progression to murine renal tumors; murine renal tumor progression on genetic deletion of both Tsc1 and FoxOs; and downregulated FoxO expression in most human renal clear cell and papillary carcinomas, yet continued expression in less aggressive RCCs and benign renal tumor subtypes. Mechanistically, integrated analyses revealed that FoxO-mediated block operates via suppression of Myc through upregulation of the Myc antagonists, Mxi1-SRα and mir-145, establishing a FoxO-Mxi1-SRα/mir-145 axis as a major progression block in renal tumor development.
- Published
- 2010
28. BRAF activation initiates but does not maintain invasive prostate adenocarcinoma
- Author
-
Cory Abate-Shen, Zhenxiong Wang, Gyeong Hoon Kang, Massimo Loda, Isil Guney, Xuesong Ouyang, William C. Hahn, Eyoung Shin, Alexander S. Guimaraes, Joseph H. Jeong, Ralph Weissleder, Zhihu Ding, Lynda Chin, Jose L. Figueiredo, and Shan Jiang
- Subjects
Male ,Pathology ,lcsh:Medicine ,medicine.disease_cause ,Prostate cancer ,Mice ,0302 clinical medicine ,Prostate ,Transgenes ,lcsh:Science ,Oncology/Prostate Cancer ,0303 health sciences ,Multidisciplinary ,Melanoma ,Urology/Prostate Cancer ,Chromoplexy ,3. Good health ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Androgens ,Adenocarcinoma ,Research Article ,PCA3 ,Proto-Oncogene Proteins B-raf ,medicine.medical_specialty ,Mice, Transgenic ,Biology ,03 medical and health sciences ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Cell Lineage ,Neoplasm Invasiveness ,Castration ,Protein kinase B ,Molecular Biology ,030304 developmental biology ,Cell Proliferation ,Hyperplasia ,lcsh:R ,Prostatic Neoplasms ,Epithelial Cells ,medicine.disease ,Phosphoproteins ,Rats ,Cancer research ,Trans-Activators ,lcsh:Q ,Urothelium ,Carcinogenesis ,Proto-Oncogene Proteins c-akt - Abstract
Prostate cancer is the second leading cause of cancer-related deaths in men. Activation of MAP kinase signaling pathway has been implicated in advanced and androgen-independent prostate cancers, although formal genetic proof has been lacking. In the course of modeling malignant melanoma in a tyrosinase promoter transgenic system, we developed a genetically-engineered mouse (GEM) model of invasive prostate cancers, whereby an activating mutation of BRAF(V600E)--a mutation found in approximately 10% of human prostate tumors--was targeted to the epithelial compartment of the prostate gland on the background of Ink4a/Arf deficiency. These GEM mice developed prostate gland hyperplasia with progression to rapidly growing invasive adenocarcinoma without evidence of AKT activation, providing genetic proof that activation of MAP kinase signaling is sufficient to drive prostate tumorigenesis. Importantly, genetic extinction of BRAF(V600E) in established prostate tumors did not lead to tumor regression, indicating that while sufficient to initiate development of invasive prostate adenocarcinoma, BRAF(V600E) is not required for its maintenance.
- Published
- 2008
29. p53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation
- Author
-
Giovanni Tonon, Gerald C. Chu, Samuel R. Perry, David Hiller, Hongwu Zheng, Ronald A. DePinho, An Jou Chen, Y. Alan Wang, Wing Hung Wong, Lynda Chin, Jayne M. Stommel, Keith L. Ligon, Cameron Brennan, Ruprecht Wiedemeyer, Katherine Dunn, Zhihu Ding, Mingjian James You, Haoqiang Ying, Alec C. Kimmelman, Haiyan Yan, Zheng, H, Ying, H, Yan, H, Kimmelman, Ac, Hiller, Dj, Chen, A, Perry, Sr, Tonon, G, Chu, Gc, Ding, Z, Stommel, Jm, Dunn, Kl, Wiedemeyer, R, You, Mj, Brennan, C, Wang, Ya, Ligon, Kl, Wong, Wh, Chin, L, and Depinho, Ra.
- Subjects
Tumor suppressor gene ,Cellular differentiation ,Biology ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neurosphere ,medicine ,PTEN ,Animals ,Humans ,Progenitor cell ,030304 developmental biology ,Cell Proliferation ,Neurons ,0303 health sciences ,Multidisciplinary ,Brain Neoplasms ,PTEN Phosphohydrolase ,Cell Differentiation ,Glioma ,Immunohistochemistry ,Neural stem cell ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Neoplastic Stem Cells ,Stem cell ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Glioblastoma - Abstract
Glioblastoma (GBM) is a highly lethal brain tumour presenting as one of two subtypes with distinct clinical histories and molecular profiles. The primary GBM subtype presents acutely as a high-grade disease that typically harbours mutations in EGFR, PTEN and INK4A/ARF (also known as CDKN2A), and the secondary GBM subtype evolves from the slow progression of a low-grade disease that classically possesses PDGF and TP53 events. Here we show that concomitant central nervous system (CNS)-specific deletion of p53 and Pten in the mouse CNS generates a penetrant acute-onset high-grade malignant glioma phenotype with notable clinical, pathological and molecular resemblance to primary GBM in humans. This genetic observation prompted TP53 and PTEN mutational analysis in human primary GBM, demonstrating unexpectedly frequent inactivating mutations of TP53 as well as the expected PTEN mutations. Integrated transcriptomic profiling, in silico promoter analysis and functional studies of murine neural stem cells (NSCs) established that dual, but not singular, inactivation of p53 and Pten promotes an undifferentiated state with high renewal potential and drives increased Myc protein levels and its associated signature. Functional studies validated increased Myc activity as a potent contributor to the impaired differentiation and enhanced renewal of NSCs doubly null for p53 and Pten (p53(-/-) Pten(-/-)) as well as tumour neurospheres (TNSs) derived from this model. Myc also serves to maintain robust tumorigenic potential of p53(-/-) Pten(-/-) TNSs. These murine modelling studies, together with confirmatory transcriptomic/promoter studies in human primary GBM, validate a pathogenetic role of a common tumour suppressor mutation profile in human primary GBM and establish Myc as an important target for cooperative actions of p53 and Pten in the regulation of normal and malignant stem/progenitor cell differentiation, self-renewal and tumorigenic potential.
- Published
- 2008
30. The SANT domain of human MI-ER1 interacts with Sp1 to interfere with GC box recognition and repress transcription from its own promoter
- Author
-
Gary D. Paterno, F. Corinne Mercer, Zhihu Ding, and Laura L. Gillespie
- Subjects
Transcription, Genetic ,Sp1 Transcription Factor ,Recombinant Fusion Proteins ,Response element ,Molecular Sequence Data ,CAAT box ,Down-Regulation ,Biology ,Transfection ,Biochemistry ,Histone Deacetylases ,Cell Line ,Immediate-Early Proteins ,Epigenetics of physical exercise ,Transcription (biology) ,Genes, Reporter ,Humans ,RNA, Messenger ,Cloning, Molecular ,Promoter Regions, Genetic ,Molecular Biology ,Glutathione Transferase ,Binding Sites ,Base Sequence ,Dose-Response Relationship, Drug ,Reverse Transcriptase Polymerase Chain Reaction ,Downstream promoter element ,Nuclear Proteins ,Promoter ,Cell Biology ,DNA ,Sequence Analysis, DNA ,Molecular biology ,Precipitin Tests ,Chromatin ,Protein Structure, Tertiary ,DNA-Binding Proteins ,CpG Islands ,Chromatin immunoprecipitation ,SANT domain ,HeLa Cells ,Plasmids ,Protein Binding ,Transcription Factors - Abstract
ld;&.2qTo gain insight into the regulation of hmi-er1 expression, we cloned a human genomic DNA fragment containing one of the two hmi-er1 promoters and consisting of 1460 bp upstream of the translation initiation codon of hMI-ER1. Computer-assisted sequence analysis revealed that the hmi-er1 promoter region contains a CpG island but lacks an identifiable TATA element, initiator sequence and downstream promoter element. This genomic DNA was able to direct transcription of a luciferase reporter gene in a variety of human cell lines, and the minimal promoter was shown to be located within–68/+144 bp. Several putative Sp1 binding sites were identified, and we show that Sp1 can bind to the hmi-er1 minimal promoter and increase transcription, suggesting that the level of hmi-er1 expression may depend on the availability of Sp1 protein. Functional analysis revealed that hMI-ER1 represses Sp1-activated transcription from the minimal promoter by a histone deacetylase-independent mechanism. Chromatin immunoprecipitation analysis demonstrated that both Sp1 and hMI-ER1 are associated with the chromatin of the hmi-er1 promoter and that overexpression of hMI-ER1 in cell lines that allow Tet-On-inducible expression resulted in loss of detectable Sp1 from the endogenous hmi-er1 promoter. The mechanism by which this occurs does not involve binding of hMI-ER1 to cis-acting elements. Instead, we show that hMI-ER1 physically associates with Sp1 and that endogenous complexes containing the two proteins could be detected in vivo. Furthermore, hMI-ER1 specifically interferes with binding of Sp1 to the hmi-er1 minimal promoter as well as to an Sp1 consensus oligonucleotide. Deletion analysis revealed that this interaction occurs through a region containing the SANT domain of hMI-ER1. Together, these data reveal a functional role for the SANT domain in the action of co-repressor regulatory factors and suggest that the association of hMI-ER1 with Sp1 represents a novel mechanism for the negative regulation of Sp1 target promoters.
- Published
- 2004
31. Human MI-ER1 alpha and beta function as transcriptional repressors by recruitment of histone deacetylase 1 to their conserved ELM2 domain
- Author
-
Laura L. Gillespie, Gary D. Paterno, and Zhihu Ding
- Subjects
Gene isoform ,Transcription, Genetic ,Molecular Sequence Data ,Repressor ,Uterine Cervical Neoplasms ,Histone Deacetylase 1 ,Biology ,Hydroxamic Acids ,DNA-binding protein ,Histone Deacetylases ,Mice ,Sp3 transcription factor ,Transcription (biology) ,Tumor Cells, Cultured ,Animals ,Humans ,Protein Isoforms ,Amino Acid Sequence ,Enzyme Inhibitors ,Caenorhabditis elegans Proteins ,Molecular Biology ,Transcription factor ,Conserved Sequence ,Transcriptional Regulation ,Base Sequence ,Carcinoma ,Cell Biology ,DNA-binding domain ,Helminth Proteins ,Molecular biology ,HDAC4 ,Recombinant Proteins ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Histone Deacetylase Inhibitors ,Repressor Proteins ,Alternative Splicing ,Female ,Transcription Factors - Abstract
mi-er1 (previously called er1) was first isolated from Xenopus laevis embryonic cells as a novel fibroblast growth factor-regulated immediate-early gene. Xmi-er1 was shown to encode a nuclear protein with an N-terminal acidic transcription activation domain. The human orthologue of mi-er1 (hmi-er1) displays 91% similarity to the Xenopus sequence at the amino acid level and was shown to be upregulated in breast carcinoma cell lines and tumors. Alternative splicing at the 3' end of hmi-er1 produces two major isoforms, hMI-ER1alpha and hMI-ER1beta, which contain distinct C-terminal domains. In this study, we investigated the role of hMI-ER1alpha and hMI-ER1beta in the regulation of transcription. Using fusion proteins of hMI-ER1alpha or hMI-ER1beta tethered to the GAL4 DNA binding domain, we show that both isoforms, when recruited to the G5tkCAT minimal promoter, function to repress transcription. We demonstrate that this repressor activity is due to interaction and recruitment of a trichostatin A-sensitive histone deacetylase 1 (HDAC1). Furthermore, deletion analysis revealed that recruitment of HDAC1 to hMI-ER1alpha and hMI-ER1beta occurs through their common ELM2 domain. The ELM2 domain was first described in the Caenorhabditis elegans Egl-27 protein and is present in a number of SANT domain-containing transcription factors. This is the first report of a function for the ELM2 domain, highlighting its role in the regulation of transcription.
- Published
- 2002
32. Genomic organization of the human mi-er1 gene and characterization of alternatively spliced isoforms: regulated use of a facultative intron determines subcellular localization
- Author
-
Gary D. Paterno, Laura L. Gillespie, Yuan-Y. Lew, Zhihu Ding, F. Corinne Mercer, and Gord W. Nash
- Subjects
Gene isoform ,Male ,Cytoplasm ,Polyadenylation ,Transcription, Genetic ,Molecular Sequence Data ,Gene Expression ,Biology ,Immediate-Early Proteins ,Exon ,Mice ,Genetics ,Animals ,Humans ,Protein Isoforms ,Amino Acid Sequence ,Nuclear protein ,Gene ,Genomic organization ,Cell Nucleus ,Base Sequence ,Sequence Homology, Amino Acid ,Intron ,Nuclear Proteins ,General Medicine ,3T3 Cells ,Molecular biology ,Introns ,DNA-Binding Proteins ,Alternative Splicing ,Genes ,Female ,Nuclear localization sequence ,Transcription Factors - Abstract
mi-er1 (previously called er1) is a fibroblast growth factor-inducible early response gene activated during mesoderm induction in Xenopus embryos and encoding a nuclear protein that functions as a transcriptional activator. The human orthologue of mi-er1 was shown to be upregulated in breast carcinoma cell lines and breast tumours when compared to normal breast cells. In this report, we investigate the structure of the human mi-er1 (hmi-er1) gene and characterize the alternatively spliced transcripts and protein isoforms. hmi-er1 is a single copy gene located at 1p31.2 and spanning 63 kb. It contains 17 exons and includes one skipped exon, a facultative intron and three polyadenylation signals to produce 12 transcripts encoding six distinct proteins. hmi-er1 transcripts were expressed at very low levels in most human adult tissues and the mRNA isoform pattern varied with the tissue. The 12 transcripts encode proteins containing a common internal sequence with variable N- and C-termini. Three distinct N- and two distinct C-termini were identified, giving rise to six protein isoforms. The two C-termini differ significantly in size and sequence and arise from alternate use of a facultative intron to produce hMI-ER1alpha and hMI-ER1beta. In all tissues except testis, transcripts encoding the beta isoform were predominant. hMI-ER1alpha lacks the predicted nuclear localization signal and transfection assays revealed that, unlike hMI-ER1beta, it is not a nuclear protein, but remains in the cytoplasm. Our results demonstrate that alternate use of a facultative intron regulates the subcellular localization of hMI-ER1 proteins and this may have important implications for hMI-ER1 function.
- Published
- 2002
33. Abstract 5448: PTK7 as a potential therapeutic target in ovarian cancer
- Author
-
Stuart Licht, Zhihu Ding, Hui Su, William Brondyk, Serena J. Silver, Venkat Reddy, Joshua Murtie, Kimberly Bishop, Keli Perron, William Weber, Raffaele Baffa, Christopher Winter, Julie-Ann Gavigan, Pinckney Jason Robert, Vicky Drewett, David P. Harper, Amanda Lennon, Richard Gregory, Meredith Wolfram, Dietmar Hoffmann, David Reczek, Maureen Magnay, Marc Trombe, Victoria M. Richon, Helene Simonds-Mannes, and Dinesh S. Bangari
- Subjects
Cancer Research ,Cell growth ,Angiogenesis ,Cell ,Wnt signaling pathway ,Cancer ,Biology ,medicine.disease ,Ovarian tumor ,medicine.anatomical_structure ,Oncology ,Immunology ,medicine ,Cancer research ,PTK7 ,Ovarian cancer - Abstract
The identification of proteins that selectively discriminate between tumor cells and normal adult cells allows for the specific targeting of diseased cells with antibody therapeutics. One such recently identified protein, PTK7, is an onco-fetal membrane protein which exhibits limited expression and function in adults. PTK7 was identified as a member of the RTK super family but lacks a functional kinase domain. Normally, PTK7 is expressed early in development and its loss is associated with severe defects in neural tube closure and sensory hair cell bundle formation. Functionally, little is known about the signaling involving PTK7, but it has been linked to both the canonical and noncanonical WNT pathways. Recently, PTK7 expression has been shown to be upregulated in a number of cancers including: ovarian, melanoma, leukemia, lung, pancreatic, colon, renal and breast. In vitro and in vivo studies support a role in regulating angiogenesis, invasion & survival. To further validate PTK7 as a potential cancer target that may be required for tumor maintenance and progression, we analyzed the expression of PTK7 in normal and tumor samples, and validated an in vitro and in vivo role of PTK7 on cell growth in ovarian cancer cell lines using both genetic tools and polyclonal antibodies. Silencing PTK7 with stably expressed inducible shRNAs is shown to inhibit the growth of ovarian cancer cell lines in vitro and to lead to delayed tumor growth upon PTK7 knockdown in murine tumor xenograft models. Further supporting the role of PTK7 as a potential antibody target, polyclonal antibodies to PTK7 are shown to inhibit the growth of SKOV3 and OVCAR8 cells in vitro. Although 4 human antibodies derived from phage display failed to inhibit in vitro cell growth, these results suggest that functionally blocking PTK7 may lead to the inhibition of ovarian tumor growth and is a potential target for antibody therapies. Citation Format: Zhihu Ding, Amanda Lennon, Keli Perron, David Harper, Hui Su, Meredith Wolfram, Joshua Murtie, Stuart Licht, Jason Pinckney, Helene Simonds-Mannes, Kimberly Bishop, Julie-Ann Gavigan, Dinesh Bangari, Maureen Magnay, William Weber, David Reczek, William Brondyk, Vicky Drewett, Marc Trombe, Dietmar Hoffmann, Raffaele Baffa, Serena Silver, Victoria Richon, Christopher Winter, Venkat Reddy, Richard C. Gregory. PTK7 as a potential therapeutic target in ovarian cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5448. doi:10.1158/1538-7445.AM2014-5448
- Published
- 2014
34. Resistance to apoptosis is correlated with the reduced caspase-3 activation and enhanced expression of antiapoptotic proteins in human cervical multidrug-resistant cells
- Author
-
Alan Pater, Xiaolong Yang, Zhihu Ding, and Shou Ching Tang
- Subjects
P50 ,Poly ADP ribose polymerase ,Biophysics ,bcl-X Protein ,Caspase 3 ,Apoptosis ,Cervix Uteri ,Biology ,Biochemistry ,Western blot ,Acetyltransferases ,medicine ,Cytotoxic T cell ,Humans ,Molecular Biology ,Cell Line, Transformed ,Cisplatin ,medicine.diagnostic_test ,Proteins ,Cell Biology ,Molecular biology ,Drug Resistance, Multiple ,Multiple drug resistance ,DNA-Binding Proteins ,Enzyme Activation ,Proto-Oncogene Proteins c-bcl-2 ,Caspases ,Female ,Carrier Proteins ,medicine.drug ,Transcription Factors - Abstract
Recent studies have indicated that induction of apoptosis is the primary cytotoxic mechanism of most cancer chemotherapeutic agents, and abnormalities in the control of apoptosis can affect the sensitivity of malignant cells to multiple drugs. Here, we treated cells with cisplatin and other apoptotic stimuli and found that multidrug-resistant (MDR) endocervical HEN-16-2/CDDP cells, compared with drug-sensitive parental cells, were significantly more resistant to apoptosis and exhibited decreased proteolytic activation of caspase-3. The latter was further demonstrated by decreased cleavage of its substrate poly(ADP-ribose) polymerase (PARP). Further, Western blot analysis showed that MDR HEN-16-2/CDDP cells had significantly higher levels of the apoptosis-inhibiting proteins BAG-1 p50 and p33 isoforms and Bcl-XL. This study provided the first evidence that overexpression of antiapoptotic BAG-1 p50 and p33 and Bcl-XL may cause resistance to apoptosis through reduction of caspase-3 activity in human cervical cells having an MDR phenotype.
- Published
- 2001
35. Abstract 3135: In vitro and in vivo synthetic lethal screens to identify novel targets in the context of PTEN deficiency
- Author
-
Joachim Theihaber, Zhihu Ding, Yu-an Zhang, Serena J. Silver, James Watters, Oleg Iartchouk, Joshua Murtie, Zhifang Li, Hongyun Wang, Gary Shapiro, Hui Cao, Stephan Reiling, Jack Pollard, Saurin Jani, Christoph Lengauer, Dietmar Hoffman, Angela Virone-Oddos, Venkat Reddy, Bin Wu, Jingxin Zhang, Richard Newcombe, Madelyn Light, May Cindhuchao, and Eva Bric-Furlong
- Subjects
Genetics ,Cancer Research ,Gene knockdown ,Mutation ,biology ,Drug discovery ,Context (language use) ,medicine.disease_cause ,Small hairpin RNA ,Oncology ,Essential gene ,In vivo ,biology.protein ,medicine ,PTEN - Abstract
Functional genomic screens have been employed by many groups to identify novel targets for cancer therapeutics. However, translation of these data sets into new drug discovery programs has proved challenging, in part due to difficulties in obtaining strong on-target knockdown, coupled with misleading off-target effects. Most commercially available pooled shRNA screening libraries provide 5-6 shRNAs per gene, with little knowledge of knock-down efficiency. In an effort to overcome these issues, we designed a custom shRNA library targeting 580 human genes, including key nodes in signal transduction pathways and genes of interest for oncology drug development. This pathway-centric design allows us to determine critical pathways for cancer cell growth in addition to individual gene phenotypes. For each gene, we have total of 11 shRNAs, a subset of which have been previously shown high efficiency knock-down. In particular, we emphasized genes where tool or lead compounds were available for rapid follow-up. Both in size and scope, this library was designed to quickly identify important signaling pathways that are essential on their own or when perturbed in the presence of small molecule drugs in our pipeline. With this focused library, we performed functional genomic shRNA screens in PTEN mutant or null lines across cancer types in vitro, including four cell lines that are PTEN deficient (UACC62, MDA-MB-468, U87-MG and PC3), and one cell line, (HCT-116) that is PTEN wild type. Meanwhile, we performed in vivo screens with PC3 cells. To identify hits from our screens, we used an internally developed algorithm, c-FOLD, to calculate p-value and fold changes for each shRNA. As confirmation that our library and methods can identify essential genes in a context specific manner, we observe BRAF as the top essential gene hit in the cell line which contains an activated BRAF allele, UACC62, but not in other lines tested. We then examined the concordance of hits in our in vitro and in vivo experiments with PC3 cells. In general, individual shRNAs were highly correlated between in vitro and in vivo assays, but a subset is specific to one setting, and may present particularly interesting targets. To discover synthetic lethal partners to PTEN deficiency, we required that a gene is essential in all four PTEN deficient lines in vitro, as well as in PC3 in vivo, but not essential in HCT-116. Among the top hits are multiple shRNAs against ENTPD5, an ER enzyme hydrolyzing UDP to UMP. In addition, using our pathway approach, we identified components in JNK pathways as synthetic lethal partners to PTEN deficiency. The results obtained from these studies established a strong platform for in vivo and in vitro identification of novel targets and novel combination partners. These approaches can also be integrated with other Omics data, such as mutation and expression, to ultimately find promising targets to develop novel cancer therapeutics. Citation Format: Jing-xin Zhang, Joshua Murtie, Oleg Iartchouk, Hui Cao, Gary Shapiro, Zhifang Li, Hongyun Wang, Zhihu Ding, Yu-an Zhang, Madelyn Light, Dietmar Hoffman, May Cindhuchao, Saurin Jani, Richard Newcombe, Eva Bric-Furlong, Bin Wu, Angela Virone-Oddos, Stephan Reiling, Joachim Theihaber, Christoph Lengauer, Jack Pollard, James Watters, Serena Silver, Venkat Reddy. In vitro and in vivo synthetic lethal screens to identify novel targets in the context of PTEN deficiency. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3135. doi:10.1158/1538-7445.AM2013-3135
- Published
- 2013
36. Telomerase Reactivation following Telomere Dysfunction Yields Murine Prostate Tumors with Bone Metastases
- Author
-
Wei Wang, Elena Ivanova, Alexei Protopopov, Y. Alan Wang, Chang-Jiun Wu, Gerald C. Chu, Shan Jiang, Liren Li, Jianhua Zhang, Hailei Zhang, Lynda Chin, Guocan Wang, Boyi Gan, Samuel R. Perry, Emma S. Labrot, Maria Kost-Alimova, J. Zhang, James W. Horner, Yonghong Xiao, Zhihu Ding, Ronald A. DePinho, Mariela Jaskelioff, Jian Hu, and Xin Lu
- Subjects
Genome instability ,0303 health sciences ,Telomerase ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Cancer ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,3. Good health ,Telomere ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,medicine.anatomical_structure ,Prostate ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,biology.protein ,PTEN ,Telomerase reverse transcriptase ,030304 developmental biology - Abstract
SummaryTo determine the role of telomere dysfunction and telomerase reactivation in generating pro-oncogenic genomic events and in carcinoma progression, an inducible telomerase reverse transcriptase (mTert) allele was crossed onto a prostate cancer-prone mouse model null for Pten and p53 tumor suppressors. Constitutive telomerase deficiency and associated telomere dysfunction constrained cancer progression. In contrast, telomerase reactivation in the setting of telomere dysfunction alleviated intratumoral DNA-damage signaling and generated aggressive cancers with rearranged genomes and new tumor biological properties (bone metastases). Comparative oncogenomic analysis revealed numerous recurrent amplifications and deletions of relevance to human prostate cancer. Murine tumors show enrichment of the TGF-β/SMAD4 network, and genetic validation studies confirmed the cooperative roles of Pten, p53, and Smad4 deficiencies in prostate cancer progression, including skeletal metastases. Thus, telomerase reactivation in tumor cells experiencing telomere dysfunction enables full malignant progression and provides a mechanism for acquisition of cancer-relevant genomic events endowing new tumor biological capabilities.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.