81 results on '"Kurtis E. Bachman"'
Search Results
2. Data from DFMO and 5-Azacytidine Increase M1 Macrophages in the Tumor Microenvironment of Murine Ovarian Cancer
- Author
-
Cynthia A. Zahnow, Robert A. Casero, Stephen B. Baylin, Meredith L. Stone, Jackson R. Foley, Kurtis E. Bachman, Karla R. Wiehagen, Cassandra E. Holbert, Matthew Dunworth, Stephen M. Brown, and Meghan Travers
- Abstract
Although ovarian cancer has a low incidence rate, it remains the most deadly gynecologic malignancy. Previous work has demonstrated that the DNMTi 5-Azacytidine (5AZA-C) activates type I interferon signaling to increase IFNγ+ T cells and natural killer (NK) cells and reduce the percentage of macrophages in the tumor microenvironment. To improve the efficacy of epigenetic therapy, we hypothesized that the addition of α-difluoromethylornithine (DFMO), an ornithine decarboxylase inhibitor, may further decrease immunosuppressive cell populations improving outcome. We tested this hypothesis in an immunocompetent mouse model for ovarian cancer and found that in vivo, 5AZA-C and DFMO, either alone or in combination, significantly increased survival, decreased tumor burden, and caused recruitment of activated (IFNγ+) CD4+ T cells, CD8+ T cells, and NK cells. The combination therapy had a striking increase in survival when compared with single-agent treatment, despite a smaller difference in recruited lymphocytes. Instead, combination therapy led to a significant decrease in immunosuppressive cells such as M2 polarized macrophages and an increase in tumor-killing M1 macrophages. In this model, depletion of macrophages with a CSF1R-blocking antibody reduced the efficacy of 5AZA-C + DFMO treatment and resulted in fewer M1 macrophages in the tumor microenvironment. These observations suggest our novel combination therapy modifies macrophage polarization in the tumor microenvironment, recruiting M1 macrophages and prolonging survival.Significance:Combined epigenetic and polyamine-reducing therapy stimulates M1 macrophage polarization in the tumor microenvironment of an ovarian cancer mouse model, resulting in decreased tumor burden and prolonged survival.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Data from DFMO and 5-Azacytidine Increase M1 Macrophages in the Tumor Microenvironment of Murine Ovarian Cancer
- Author
-
Cynthia A. Zahnow, Robert A. Casero, Stephen B. Baylin, Meredith L. Stone, Jackson R. Foley, Kurtis E. Bachman, Karla R. Wiehagen, Cassandra E. Holbert, Matthew Dunworth, Stephen M. Brown, and Meghan Travers
- Abstract
Supplementary data including polyamine levels, addition of anti-PD-1 to therapy, and flow cytometry plots.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure Legends 1-4 from Knock-in of Mutant K-ras in Nontumorigenic Human Epithelial Cells as a New Model for Studying K-ras–Mediated Transformation
- Author
-
Ben Ho Park, Kurtis E. Bachman, Eike Gallmeier, Yuko Konishi, Joseph P. Garay, John P. Gustin, Josh Lauring, Abde M. Abukhdeir, Bedri Karakas, and Hiroyuki Konishi
- Abstract
Supplementary Figure Legends 1-4 from Knock-in of Mutant K-ras in Nontumorigenic Human Epithelial Cells as a New Model for Studying K-ras–Mediated Transformation
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 1 from Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells
- Author
-
Kurtis E. Bachman, David J. Weber, Stuart S. Martin, Ben Ho Park, Todd Waldman, Khola Tahir, Marta Szmacinski, Michele B. Weiss, and Michele I. Vitolo
- Abstract
Supplementary Figure 1 from Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure Legends 1-2 from Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells
- Author
-
Kurtis E. Bachman, David J. Weber, Stuart S. Martin, Ben Ho Park, Todd Waldman, Khola Tahir, Marta Szmacinski, Michele B. Weiss, and Michele I. Vitolo
- Abstract
Supplementary Figure Legends 1-2 from Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 2 from Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells
- Author
-
Kurtis E. Bachman, David J. Weber, Stuart S. Martin, Ben Ho Park, Todd Waldman, Khola Tahir, Marta Szmacinski, Michele B. Weiss, and Michele I. Vitolo
- Abstract
Supplementary Figure 2 from Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 1 from Molecular Target Class Is Predictive of In vitro Response Profile
- Author
-
Richard Wooster, Mary Ann Hardwicke, Kurt Auger, Ronald Wegrzyn, Christopher Moy, Elizabeth McNeil, Stephen Eastman, Yuan H. Wen, Junping Jing, Yan Y. Degenhardt, Kurtis E. Bachman, and Joel Greshock
- Abstract
Supplementary Figure 1 from Molecular Target Class Is Predictive of In vitro Response Profile
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table 1, Figures 1-4 from Knock-in of Mutant K-ras in Nontumorigenic Human Epithelial Cells as a New Model for Studying K-ras–Mediated Transformation
- Author
-
Ben Ho Park, Kurtis E. Bachman, Eike Gallmeier, Yuko Konishi, Joseph P. Garay, John P. Gustin, Josh Lauring, Abde M. Abukhdeir, Bedri Karakas, and Hiroyuki Konishi
- Abstract
Supplementary Table 1, Figures 1-4 from Knock-in of Mutant K-ras in Nontumorigenic Human Epithelial Cells as a New Model for Studying K-ras–Mediated Transformation
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Knock-in of Mutant K-ras in Nontumorigenic Human Epithelial Cells as a New Model for Studying K-ras–Mediated Transformation
- Author
-
Ben Ho Park, Kurtis E. Bachman, Eike Gallmeier, Yuko Konishi, Joseph P. Garay, John P. Gustin, Josh Lauring, Abde M. Abukhdeir, Bedri Karakas, and Hiroyuki Konishi
- Abstract
The oncogenic function of mutant ras in mammalian cells has been extensively investigated using multiple human and animal models. These systems include overexpression of exogenous mutant ras transgenes, conditionally expressed knock-in mouse models, and somatic cell knockout of mutant and wild-type ras genes in human cancer cell lines. However, phenotypic discrepancies between knock-in mice and transgenic mutant ras overexpression prompted us to evaluate the consequences of targeted knock-in of an oncogenic K-ras mutation in the nontumorigenic human breast epithelial cell line MCF-10A and hTERT-immortalized human mammary epithelial cells. Our results show several significant differences between mutant K-ras knock-in cells versus their transgene counterparts, including limited phosphorylation of the downstream molecules extracellular signal-regulated kinase and AKT, minor proliferative capacity in the absence of an exogenous growth factor, and the inability to form colonies in semisolid medium. Analysis of 16 cancer cell lines carrying mutant K-ras genes indicated that 50% of cancer cells harbor nonoverexpressed heterozygous K-ras mutations similar to the expression seen in our knock-in cell lines. Thus, this system serves as a new model for elucidating the oncogenic contribution of mutant K-ras as expressed in a large fraction of human cancer cells. [Cancer Res 2007;67(18):8460–7]
- Published
- 2023
- Full Text
- View/download PDF
11. Data from MCT-1 Protein Interacts with the Cap Complex and Modulates Messenger RNA Translational Profiles
- Author
-
Ronald B. Gartenhaus, Huili He, Kurtis E. Bachman, Michele Vitolo, Krystyna Mazan-Mamczarz, Suvobroto Nandi, Bo Shi, and Line S. Reinert
- Abstract
MCT-1 is an oncogene that was initially identified in a human T cell lymphoma and has been shown to induce cell proliferation as well as activate survival-related pathways. MCT-1 contains the PUA domain, a recently described RNA-binding domain that is found in several tRNA and rRNA modification enzymes. Here, we established that MCT-1 protein interacts with the cap complex through its PUA domain and recruits the density-regulated protein (DENR/DRP), containing the SUI1 translation initiation domain. Through the use of microarray analysis on polysome-associated mRNAs, we showed that up-regulation of MCT-1 was able to modulate the translation profiles of BCL2L2, TFDP1, MRE11A, cyclin D1, and E2F1 mRNAs, despite equivalent levels of mRNAs in the cytoplasm. Our data establish a role for MCT-1 in translational regulation, and support a linkage between translational control and oncogenesis. (Cancer Res 2006; 66(18): 8994-9001)
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Methods, Figure Legend from Molecular Target Class Is Predictive of In vitro Response Profile
- Author
-
Richard Wooster, Mary Ann Hardwicke, Kurt Auger, Ronald Wegrzyn, Christopher Moy, Elizabeth McNeil, Stephen Eastman, Yuan H. Wen, Junping Jing, Yan Y. Degenhardt, Kurtis E. Bachman, and Joel Greshock
- Abstract
Supplementary Methods, Figure Legend from Molecular Target Class Is Predictive of In vitro Response Profile
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure Legend and Table 1 from MCT-1 Protein Interacts with the Cap Complex and Modulates Messenger RNA Translational Profiles
- Author
-
Ronald B. Gartenhaus, Huili He, Kurtis E. Bachman, Michele Vitolo, Krystyna Mazan-Mamczarz, Suvobroto Nandi, Bo Shi, and Line S. Reinert
- Abstract
Supplementary Figure Legend and Table 1 from MCT-1 Protein Interacts with the Cap Complex and Modulates Messenger RNA Translational Profiles
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table 1 from Molecular Target Class Is Predictive of In vitro Response Profile
- Author
-
Richard Wooster, Mary Ann Hardwicke, Kurt Auger, Ronald Wegrzyn, Christopher Moy, Elizabeth McNeil, Stephen Eastman, Yuan H. Wen, Junping Jing, Yan Y. Degenhardt, Kurtis E. Bachman, and Joel Greshock
- Abstract
Supplementary Table 1 from Molecular Target Class Is Predictive of In vitro Response Profile
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Table 3 from Molecular Target Class Is Predictive of In vitro Response Profile
- Author
-
Richard Wooster, Mary Ann Hardwicke, Kurt Auger, Ronald Wegrzyn, Christopher Moy, Elizabeth McNeil, Stephen Eastman, Yuan H. Wen, Junping Jing, Yan Y. Degenhardt, Kurtis E. Bachman, and Joel Greshock
- Abstract
Supplementary Table 3 from Molecular Target Class Is Predictive of In vitro Response Profile
- Published
- 2023
- Full Text
- View/download PDF
16. Data from Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells
- Author
-
Kurtis E. Bachman, David J. Weber, Stuart S. Martin, Ben Ho Park, Todd Waldman, Khola Tahir, Marta Szmacinski, Michele B. Weiss, and Michele I. Vitolo
- Abstract
Many cancers, including breast cancer, harbor loss-of-function mutations in the catalytic domain of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) or have reduced PTEN expression through loss of heterozygosity and/or epigenetic silencing mechanisms. However, specific phenotypic effects of PTEN inactivation in human cancer cells remain poorly defined without a direct causal connection between the loss of PTEN function and the development or progression of cancer. To evaluate the biological and clinical relevance of reduced or deleted PTEN expression, a novel in vitro model system was generated using human somatic cell knockout technologies. Targeted homologous recombination allowed for a single and double allelic deletion, which resulted in reduced and deleted PTEN expression, respectively. We determined that heterozygous loss of PTEN in the nontumorigenic human mammary epithelial cell line MCF-10A was sufficient for activation of the phosphoinositide 3-kinase/AKT and mitogen-activated protein kinase pathways, whereas the homozygous absence of PTEN expression led to a further increased activation of both pathways. The deletion of PTEN was able to confer growth factor–independent proliferation, which was confirmed by the resistance of the PTEN−/− MCF-10A cells to small-molecule inhibitors of the epidermal growth factor receptor. However, neither heterozygous nor homozygous loss of PTEN expression was sufficient to promote anchorage-independent growth, but the loss of PTEN did confer apoptotic resistance to cell rounding and matrix detachment. Finally, MCF-10A cells with the reduction or loss of PTEN showed increased susceptibility to the chemotherapeutic drug doxorubicin but not paclitaxel. [Cancer Res 2009;69(21):8275–83]
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Table 2 from Molecular Target Class Is Predictive of In vitro Response Profile
- Author
-
Richard Wooster, Mary Ann Hardwicke, Kurt Auger, Ronald Wegrzyn, Christopher Moy, Elizabeth McNeil, Stephen Eastman, Yuan H. Wen, Junping Jing, Yan Y. Degenhardt, Kurtis E. Bachman, and Joel Greshock
- Abstract
Supplementary Table 2 from Molecular Target Class Is Predictive of In vitro Response Profile
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 1 from MCT-1 Protein Interacts with the Cap Complex and Modulates Messenger RNA Translational Profiles
- Author
-
Ronald B. Gartenhaus, Huili He, Kurtis E. Bachman, Michele Vitolo, Krystyna Mazan-Mamczarz, Suvobroto Nandi, Bo Shi, and Line S. Reinert
- Abstract
Supplementary Figure 1 from MCT-1 Protein Interacts with the Cap Complex and Modulates Messenger RNA Translational Profiles
- Published
- 2023
- Full Text
- View/download PDF
19. A mechanism of T cell dependent selection of antigen engaged Germinal Center B cells.
- Author
-
Vinod Krishna and Kurtis E Bachman
- Subjects
Medicine ,Science - Abstract
A model of B cell affinity selection is proposed, and an explanation of peripheral tolerance mechanisms through antibody repertoire editing is presented. We show that affinity discrimination between B cells is driven by a competition between obtaining T cell help and removal of B cells from the light zone, either through apoptosis or by a return to the dark zone of germinal centers. We demonstrate that this mechanism also allows for the negative selection of self reactive B cells and maintenance of B cell tolerance during the Germinal Center reaction. Finally, we demonstrate that clonal expansion upon return to the Germinal Center dark zone amplifies differences in the antigen affinity of B cells that survive the light zone.
- Published
- 2018
- Full Text
- View/download PDF
20. Gut-Restricted Selective Cyclooxygenase-2 (COX-2) Inhibitors for Chemoprevention of Colorectal Cancer
- Author
-
Shannon Dallas, Thomas Wilde, Avijit Ghosh, Jianyao Wang, Yawei Dong, Gaochao Tian, Daohong Liao, Xueliang Li, Lawrence Szewczuk, Peter King, David C. Evans, Yifan Shi, Kurtis E. Bachman, Javier Suarez, Beth Pietrak, James P. Edwards, Carlo Sensenhauser, Hao Chen, Vineet Pande, Peter J. Connolly, Karen DiLoreto, William G Bonnette, Shefali Patel, and Zhuming Zhang
- Subjects
Male ,Adenoma ,Cell Survival ,Colorectal cancer ,Antineoplastic Agents ,Disease ,Etoricoxib ,Mice ,Structure-Activity Relationship ,In vivo ,Drug Discovery ,medicine ,Animals ,Humans ,Cell Proliferation ,Cyclooxygenase 2 Inhibitors ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Chemistry ,medicine.disease ,Mice, Inbred C57BL ,Celecoxib ,Cyclooxygenase 2 ,biology.protein ,Cancer research ,Chemoprotective ,Molecular Medicine ,Cyclooxygenase ,Drug Screening Assays, Antitumor ,Colorectal Neoplasms ,Ex vivo ,medicine.drug - Abstract
Selective cyclooxygenase (COX)-2 inhibitors have been extensively studied for colorectal cancer (CRC) chemoprevention. Celecoxib has been reported to reduce the incidence of colorectal adenomas and CRC but is also associated with an increased risk of cardiovascular events. Here, we report a series of gut-restricted, selective COX-2 inhibitors characterized by high colonic exposure and minimized systemic exposure. By establishing acute ex vivo 18F-FDG uptake attenuation as an efficacy proxy, we identified a subset of analogues that demonstrated statistically significant in vivo dose-dependent inhibition of adenoma progression and survival extension in an APCmin/+ mouse model. However, in vitro-in vivo correlation analysis showed their chemoprotective effects were driven by residual systemic COX-2 inhibition, rationalizing their less than expected efficacies and highlighting the challenges associated with COX-2-mediated CRC disease chemoprevention.
- Published
- 2021
- Full Text
- View/download PDF
21. Integration of the Transcriptome and Genome-Wide Landscape of BRD2 and BRD4 Binding Motifs Identifies Key Superenhancer Genes and Reveals the Mechanism of Bet Inhibitor Action in Rheumatoid Arthritis Synovial Fibroblasts
- Author
-
Xuefeng Yin, Alice M. Walsh, Qingxuan Song, Loui Madakamutil, Vinod Krishna, Kurtis E. Bachman, David Pocalyko, Sunil Nagpal, and Ian Anderson
- Subjects
Chromatin binding ,Immunology ,Biology ,Bromodomain ,Chromatin ,Cell biology ,Gene expression profiling ,Transcriptome ,BET inhibitor ,03 medical and health sciences ,0302 clinical medicine ,Cistrome ,Immunology and Allergy ,Transcription factor ,030215 immunology - Abstract
Fibroblast-like synoviocytes (FLS), one of the main cell types of the rheumatoid arthritis (RA) synovium, possess phenotypic and molecular characteristics of transformed cells. JQ1, an inhibitor of the bromodomain and extra terminal domain family that includes BRD2, BRD3, BRD4, and BRDt, has shown efficacy in models of arthritis. We demonstrate that the active isomer of JQ1 but not its inactive isomer inhibits IL-1β–induced RA-FLS activation and proliferation. To understand the mechanism of JQ1 action, we subjected JQ1-treated RA-FLS to transcriptional profiling and determined BRD2 and BRD4 cistromes by identifying their global chromatin binding sites. In addition, assay for transposable accessible chromatin by high throughput sequencing was employed to identify open and closed regions of chromatin in JQ1-treated RA-FLS. Through an integrated analysis of expression profiling, Brd2/Brd4 cistrome data, and changes in chromatin accessibility, we found that JQ1 inhibited key BRD2/BRD4 superenhancer genes, downregulated multiple crucial inflammatory pathways, and altered the genome-wide occupancy of critical transcription factors involved in inflammatory signaling. Our results suggest a pleiotropic effect of JQ1 on pathways that have shown to be individually efficacious in RA (in vitro, in vivo, and/or in humans) and provide a strong rationale for targeting BRD2/BRD4 for disease treatment and interception.
- Published
- 2021
- Full Text
- View/download PDF
22. Abstract A012: Transcription factor expression repertoire basis for epigenetic and transcriptional subtypes of colorectal cancers
- Author
-
Yuba R. Bhandari, Vinod Krishna, Rachael Powers, Sehej Parmar, Sara-Jayne Thursby, Ekta Gupta, Ozlem Kulak, Prashanth Gokare, Joke Reumers, Liesbeth Van Wesenbeeck, Kurtis E. Bachman, Stephen B. Baylin, and Hariharan Easwaran
- Subjects
Cancer Research ,Oncology - Abstract
Colorectal cancers (CRCs) form a heterogenous group classified into epigenetic and transcriptional subtypes. The basis for the epigenetic subtypes, exemplified by varying degrees of promoter DNA hypermethylation, and its relation to the transcriptional subtypes is not well understood. We now link cancer-specific transcription factor (TF) expression dysregulation to methylation alterations near TF-binding sites at promoter and enhancer regions in CRCs and their pre-malignant precursor lesions to provide mechanistic insights into the origins and evolution of the CRC molecular subtypes. A gradient of TF-expression changes forms a basis for the subtypes of abnormal DNA methylation, termed CpG-island promoter DNA methylation phenotypes (CIMP), in CRCs and other cancers. CIMP is tightly correlated with cancer-specific hypermethylation at enhancers, which we term CpG-enhancer methylation phenotype (CEMP). CIMP/CEMP coordination appears to be driven by augmented downregulation of TFs with common binding sites at the hypermethylated enhancers and promoters. The dysregulated expression of TFs related to CIMP/CEMP subtypes occurs early during CRC development, detectable in pre-malignant adenomas. TF-based profiling further identifies patients with worse overall survival. Importantly, altered expression of these TFs discriminates the transcriptome-based consensus molecular subtypes (CMS), thus providing a common basis for CIMP and CMS subtypes. Citation Format: Yuba R. Bhandari, Vinod Krishna, Rachael Powers, Sehej Parmar, Sara-Jayne Thursby, Ekta Gupta, Ozlem Kulak, Prashanth Gokare, Joke Reumers, Liesbeth Van Wesenbeeck, Kurtis E. Bachman, Stephen B. Baylin, Hariharan Easwaran. Transcription factor expression repertoire basis for epigenetic and transcriptional subtypes of colorectal cancers. [abstract]. In: Proceedings of the AACR Special Conference: Cancer Epigenomics; 2022 Oct 6-8; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2022;82(23 Suppl_2):Abstract nr A012.
- Published
- 2022
- Full Text
- View/download PDF
23. DFMO and 5-Azacytidine Increase M1 Macrophages in the Tumor Microenvironment of Murine Ovarian Cancer
- Author
-
Meredith L. Stone, Kurtis E. Bachman, Cynthia A. Zahnow, Stephen B. Baylin, Jackson R. Foley, Matthew Dunworth, Cassandra E. Holbert, Karla R. Wiehagen, Robert A. Casero, Meghan Travers, and Stephen M. Brown
- Subjects
0301 basic medicine ,Cancer Research ,Eflornithine ,Combination therapy ,Macrophage polarization ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,Antineoplastic Combined Chemotherapy Protocols ,Polyamines ,Tumor Cells, Cultured ,Tumor Microenvironment ,medicine ,Animals ,Ovarian Neoplasms ,Tumor microenvironment ,biology ,business.industry ,Macrophages ,medicine.disease ,Immunity, Innate ,Cystadenocarcinoma, Serous ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Azacitidine ,Cancer research ,biology.protein ,Female ,Antibody ,Ovarian cancer ,business ,Epigenetic therapy ,CD8 ,medicine.drug - Abstract
Although ovarian cancer has a low incidence rate, it remains the most deadly gynecologic malignancy. Previous work has demonstrated that the DNMTi 5-Azacytidine (5AZA-C) activates type I interferon signaling to increase IFNγ+ T cells and natural killer (NK) cells and reduce the percentage of macrophages in the tumor microenvironment. To improve the efficacy of epigenetic therapy, we hypothesized that the addition of α-difluoromethylornithine (DFMO), an ornithine decarboxylase inhibitor, may further decrease immunosuppressive cell populations improving outcome. We tested this hypothesis in an immunocompetent mouse model for ovarian cancer and found that in vivo, 5AZA-C and DFMO, either alone or in combination, significantly increased survival, decreased tumor burden, and caused recruitment of activated (IFNγ+) CD4+ T cells, CD8+ T cells, and NK cells. The combination therapy had a striking increase in survival when compared with single-agent treatment, despite a smaller difference in recruited lymphocytes. Instead, combination therapy led to a significant decrease in immunosuppressive cells such as M2 polarized macrophages and an increase in tumor-killing M1 macrophages. In this model, depletion of macrophages with a CSF1R-blocking antibody reduced the efficacy of 5AZA-C + DFMO treatment and resulted in fewer M1 macrophages in the tumor microenvironment. These observations suggest our novel combination therapy modifies macrophage polarization in the tumor microenvironment, recruiting M1 macrophages and prolonging survival. Significance: Combined epigenetic and polyamine-reducing therapy stimulates M1 macrophage polarization in the tumor microenvironment of an ovarian cancer mouse model, resulting in decreased tumor burden and prolonged survival.
- Published
- 2019
- Full Text
- View/download PDF
24. Abstract 2107: Transcription factor repertoire basis for epigenetic and consensus molecular subtypes of colorectal cancer
- Author
-
Steve Baylin, Hariharan Easwaran, Vinod Krishna, Kurtis E. Bachman, Sehej Parmar, Yuba R. Bhandari, and Rachael Powers
- Subjects
Cancer Research ,Oncology ,Colorectal cancer ,Repertoire ,medicine ,Computational biology ,Epigenetics ,Biology ,medicine.disease ,Transcription factor ,digestive system diseases - Abstract
Colorectal cancer (CRC) is classified into distinct subtypes based on genetic, epigenetic and/or transcriptional subtypes. The underlying mechanisms that drive these subtypes are unknown. By integrating whole genome DNA methylation, gene expression, chromatin patterns, and enhancer elements in CRCs, we show that expression of large numbers of transcriptional factors (TFs) is deregulated in CRCs and these expression patterns drive epigenetic and transcriptional subtypes of CRCs. The first key result emerging is a new understanding of important DNA methylation patterns central to classifying CRCs. Of these, different degrees of cancer associated promoter methylation have been recognized that suppress expression, and/or inducibility of important tumor suppressor genes. Stratified by high to low degrees of hypermethylation, this pattern is termed CpG-island promoter DNA methylation phenotypes or CIMP. We now tightly link CIMP to a cancer-specific hypermethylation at enhancer regulatory elements we term, CpG- enhancer methylation phenotype (CEMP). CIMP and CEMP are driven by decreased expression of multiple TFs with binding sites in these regulatory elements, and the involved TFs are frequently identical between the two regions. In contrast, a different set of TF factors are downregulated in non-CIMP tumors, again in conjunction with methylation patterns linking promoter and enhancer target sites for the involved TFs. Further, enhancer regulatory elements that are hypomethylated in cancer relative to normal tissue are associated with increased expression of another distinct set of TFs with target sites in the corresponding enhancers. Importantly, the TF expression patterns, and the above CIMP, CEMP, and hypomethylation patterns, are established early in the pre-malignant adenomas. Importantly, pan-cancer analyses showed that the above CIMP and CEMP subtypes occur across important cancer types in addition to CRC, again in parallel with marked downregulation of TFs that have target sites in the methylated regulatory elements. All of the above newly defined patterns also provide a basis for the transcriptome-based consensus molecular subtypes (CMS) of CRC. Overall, our studies may provide key understanding of how cancers, and their initiation and progression, are diseases which at the end of the day depend on abnormal patterns of TF expression, and how these relate to methylation alterations at target enhancer and promoter regions. Citation Format: Yuba Bhandari, Rachael Powers, Sehej Parmar, Vinod Krishna, Kurtis E. Bachman, Steve Baylin, Hariharan Easwaran. Transcription factor repertoire basis for epigenetic and consensus molecular subtypes of colorectal cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2107.
- Published
- 2021
- Full Text
- View/download PDF
25. Abstract CT236: A phase 1b, multicenter, randomized, blinded, placebo-controlled study to evaluate the efficacy of guselkumab in subjects with familial adenomatous polyposis
- Author
-
Daniel A. Sussman, David S. Weinberg, Kelly Raybold, Bryson W. Katona, Jean-Christophe Saurin, Carol A. Burke, Edward F. Attiyeh, Evelien Dekker, Hong Xie, Eduardo Vilar-Sanchez, Thomas J. Prior, Gary V. Borzillo, Douglas L. Riegert-Johnson, Xavier Llor, Peter P. Stanich, Jeffrey R. Infante, Niloy Jewel Samadder, Philippe Grandval, Kurtis E. Bachman, William M. Grady, Marcia Cruz Correa, Devanand Joseph, and Michael Smith
- Subjects
Cancer Research ,medicine.medical_specialty ,Guselkumab ,Oncology ,business.industry ,Internal medicine ,medicine ,Placebo-controlled study ,business ,medicine.disease ,Gastroenterology ,Familial adenomatous polyposis - Abstract
Background: Familial adenomatous polyposis (FAP) is the most common hereditary polyposis syndrome. It is an autosomal dominant inherited disorder characterized by the early onset of hundreds to thousands of adenomatous polyps throughout the colon. If left untreated, nearly all individuals with this syndrome develop colorectal cancer (CRC) by the third decade of life. Prophylactic colectomy is the standard of care, but individuals remain at risk for malignant transformation of duodenal polyps, rectal polyps for those who have undergone rectal-sparing surgeries, and ileal pouch polyps for those with ileal pouch-anal anastomoses. Multiple studies with both nonselective and selective cyclooxygenase inhibitors (such as sulindac or celecoxib) have shown that anti-inflammatory agents may prevent the formation and inhibit the growth of colorectal adenomatous polyps. However, toxicities associated with these agents and their limited efficacy have prevented their further development. Therefore, there is a high unmet need for novel treatment options to reduce polyp burden, delay or eliminate the need for colectomy and recurrent rectal surgery, and intercept the development of adenocarcinomas in individuals with FAP. Polyps from individuals with FAP display inflammatory features associated with the activation of the IL-23/IL-17/JAK/STAT3 pathway. This inflammation is thought to contribute to further carcinogenesis, culminating in tumor development. Specifically, IL-23 is linked to tumor growth and progression in CRC, and adenomas with high-grade dysplasia showed elevated levels of IL-17A and pSTAT3. Guselkumab, a human monoclonal antibody directed against the p19 subunit of IL-23, specifically targets IL-23 and inhibits its interaction with the IL-23 receptor. Pre-clinical models suggest that inhibition of IL-23 signaling will result in less inflammation and reduce tumor development. Methods: This randomized, blind, placebo-controlled study will evaluate the safety and efficacy of guselkumab in adults with FAP (genetic or clinical diagnosis) who have already undergone colectomy. Polyps with a sum of diameters ≥10 mm in the rectum or pouch are required. Subjects will be randomized equally to one of three study arms: 100 mg, 300 mg, or placebo given subcutaneously every 4 weeks for 6 doses. The primary efficacy endpoint is percentage change from baseline in rectal/pouch polyp burden after 24 weeks. Secondary efficacy endpoints include duodenal polyp burden change and changes in InSiGHT and Spigelman staging. Exploratory translational research objectives will explore changes in RNA expression profiles, epigenomic profiles, cytokine levels, and the microbiome. Exclusion criteria include prior IL-23 targeted therapies and any polyps >1 cm that cannot be removed. While participating, subjects are required to stop any other FAP-directed drug therapy except for aspirin. As of January 2019, 18% of the planned 72 subjects have been enrolled. ClinicalTrials.gov Identifier: NCT03649971. Citation Format: Eduardo Vilar-Sanchez, Carol Burke, Marcia R. Cruz Correa, Evelien Dekker, William M. Grady, Philippe Grandval, Bryson W. Katona, Xavier Llor, Douglas L. Riegert-Johnson, Jean-Christophe Saurin, Peter Stanich, Daniel A. Sussman, David Weinberg, Edward F. Attiyeh, Devanand Joseph, Kelly Raybold, Gary V. Borzillo, Thomas J. Prior, Michael Smith, Hong Xie, Kurtis E. Bachman, Jeffrey R. Infante, Niloy Jewel Samadder. A phase 1b, multicenter, randomized, blinded, placebo-controlled study to evaluate the efficacy of guselkumab in subjects with familial adenomatous polyposis [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 CT236.
- Published
- 2020
- Full Text
- View/download PDF
26. Reply to Haffner et al.: DNA hypomethylation renders tumors more immunogenic
- Author
-
Vipul Bhargava, Lauren Murphy, Ie Ming Shih, Karla R. Wiehagen, Meredith L. Stone, Reiner Strick, Huili Li, Michael J. Topper, Tian Li Wang, Chien Fu Hung, Stephen B. Baylin, Kurtis E. Bachman, Pamela L. Strissel, Katherine B. Chiappinelli, Cynthia A. Zahnow, Meghan Travers, Dimitrios Mathios, Glenn S. Cowley, and Michael Lim
- Subjects
0301 basic medicine ,Tumor microenvironment ,Multidisciplinary ,Chemistry ,medicine.drug_class ,Histone deacetylase inhibitor ,medicine.disease ,DNA methyltransferase ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer cell ,medicine ,Cancer research ,Ovarian cancer ,Givinostat ,DNA hypomethylation - Abstract
In the letter by Haffner et al. (1), they report that seminoma cell-intrinsic DNA hypomethylation is associated with endogenous retroviral expression, an IFN response, and lymphocytic infiltration. Their data complement and support our recent therapeutic study in a mouse model of ovarian cancer (2) and support our observations that low doses of the DNA methyltransferase (DNMT) inhibitor azacytidine (AZA), in combination with the histone deacetylase inhibitor, givinostat, activate type 1 IFN signaling in ovarian (2) and lung (3) cancer cells to increase numbers and activation of immune cells in the tumor microenvironment and to increase sensitivity to the … [↵][1]5To whom correspondence may be addressed. Email: zahnoci{at}jhmi.edu or sbaylin{at}jhmi.edu. [1]: #xref-corresp-1-1
- Published
- 2018
- Full Text
- View/download PDF
27. Epigenetic therapy activates type I interferon signaling in murine ovarian cancer to reduce immunosuppression and tumor burden
- Author
-
Lauren Murphy, Meghan Travers, Stephen B. Baylin, Dimitrios Mathios, Glenn S. Cowley, Meredith L. Stone, Kurtis E. Bachman, Karla R. Wiehagen, Huili Li, Tian Li Wang, Michael Lim, Katherine B. Chiappinelli, Ie Ming Shih, Cynthia A. Zahnow, Vipul Bhargava, Chien Fu Hung, Pamela L. Strissel, Reiner Strick, and Michael J. Topper
- Subjects
0301 basic medicine ,Tumor microenvironment ,Multidisciplinary ,business.industry ,medicine.medical_treatment ,Immunosuppression ,medicine.disease ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,Immune system ,PNAS Plus ,Interferon ,Cancer research ,Medicine ,business ,Ovarian cancer ,CD8 ,Epigenetic therapy ,medicine.drug - Abstract
Ovarian cancer is the most lethal of all gynecological cancers, and there is an urgent unmet need to develop new therapies. Epithelial ovarian cancer (EOC) is characterized by an immune suppressive microenvironment, and response of ovarian cancers to immune therapies has thus far been disappointing. We now find, in a mouse model of EOC, that clinically relevant doses of DNA methyltransferase and histone deacetylase inhibitors (DNMTi and HDACi, respectively) reduce the immune suppressive microenvironment through type I IFN signaling and improve response to immune checkpoint therapy. These data indicate that the type I IFN response is required for effective in vivo antitumorigenic actions of the DNMTi 5-azacytidine (AZA). Through type I IFN signaling, AZA increases the numbers of CD45+ immune cells and the percentage of active CD8+ T and natural killer (NK) cells in the tumor microenvironment, while reducing tumor burden and extending survival. AZA also increases viral defense gene expression in both tumor and immune cells, and reduces the percentage of macrophages and myeloid-derived suppressor cells in the tumor microenvironment. The addition of an HDACi to AZA enhances the modulation of the immune microenvironment, specifically increasing T and NK cell activation and reducing macrophages over AZA treatment alone, while further increasing the survival of the mice. Finally, a triple combination of DNMTi/HDACi plus the immune checkpoint inhibitor α-PD-1 provides the best antitumor effect and longest overall survival, and may be an attractive candidate for future clinical trials in ovarian cancer.
- Published
- 2017
- Full Text
- View/download PDF
28. TIM-3 Engagement Promotes Effector Memory T Cell Differentiation of Human Antigen-Specific CD8 T Cells by Activating mTORC1
- Author
-
Nina Chi Sabins, Michelle Kinder, Sandra Santulli-Marotto, Raluca Verona, Richard Carter, Kurtis E. Bachman, Olesya Chornoguz, Vipul Bhargava, Fred Kaplan, Shixue Shen, and Karen Leander
- Subjects
0301 basic medicine ,T cell ,Immunology ,T-Cell Antigen Receptor Specificity ,Biology ,CD8-Positive T-Lymphocytes ,Mechanistic Target of Rapamycin Complex 1 ,Lymphocyte Activation ,Immune Regulation ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,MART-1 Antigen ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,IL-2 receptor ,Antigens ,Phosphorylation ,Antigen-presenting cell ,Hepatitis A Virus Cellular Receptor 2 ,Cells, Cultured ,Lymphokines ,ZAP70 ,Gene Expression Profiling ,CD28 ,Antibodies, Monoclonal ,Natural killer T cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,ras Proteins ,Immunologic Memory ,Protein Processing, Post-Translational ,Cell Division ,030215 immunology - Abstract
T cell expression of TIM-3 following Ag encounter has been associated with a continuum of functional states ranging from effector memory T cells to exhaustion. We have designed an in vitro culture system to specifically address the impact of anti–TIM-3/TIM-3 engagement on human Ag-specific CD8 T cells during a normal response to Ag and found that anti–TIM-3 treatment enhances T cell function. In our in vitro T cell culture system, MART1-specific CD8 T cells were expanded from healthy donors using artificial APCs. To ensure that the T cells were the only source of TIM-3, cells were rechallenged with peptide-loaded artificial APCs in the presence of anti–TIM-3 Ab. In these conditions, anti–TIM-3 treatment promotes generation of effector T cells as shown by acquisition of an activated phenotype, increased cytokine production, enhanced proliferation, and a transcription program associated with T cell differentiation. Activation of mTORC1 has been previously demonstrated to enhance CD8 T cell effector function and differentiation. Anti–TIM-3 drives CD8 T cell differentiation through activation of the mTORC1 as evidenced by increased levels of phosphorylated S6 protein and rhebl1 transcript. Altogether these findings suggest that anti–TIM-3, together with Ag, drives differentiation in favor of effector T cells via the activation of mTOR pathway. To our knowledge, this is the first report demonstrating that TIM-3 engagement during Ag stimulation directly influences T cell differentiation through mTORC1.
- Published
- 2017
29. Lapatinib inhibits the growth of esophageal squamous cell carcinoma and synergistically interacts with 5-fluorouracil in patient-derived xenograft models
- Author
-
Bang-Ce Ye, Wenmin Hou, Kurtis E. Bachman, Xuehua Zhu, Xia Qin, Li Liu, Crystal Ying Qin Zang, Hui Wang, Maogui Fei, Joel Greshock, Ping Liu, Pingkuan Zhang, and Hanlim Moon
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Esophageal Neoplasms ,Organoplatinum Compounds ,Receptor, ErbB-2 ,medicine.drug_class ,Mice, Nude ,Antineoplastic Agents ,Lapatinib ,Tyrosine-kinase inhibitor ,Mice ,Random Allocation ,Cell Line, Tumor ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,Phosphorylation ,skin and connective tissue diseases ,neoplasms ,Cell Proliferation ,Oncogene ,biology ,Cancer ,Drug Synergism ,General Medicine ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,Oxaliplatin ,ErbB Receptors ,Carcinoma, Squamous Cell ,Quinazolines ,Cancer research ,biology.protein ,Esophageal Squamous Cell Carcinoma ,Fluorouracil ,Transcriptome ,Tyrosine kinase ,medicine.drug - Abstract
Lapatinib is a dual tyrosine kinase inhibitor of epidermal growth factor receptor (EGFR) and human EGFR-2 (HER2) tyrosine kinase domains. To explore the potential utility of lapatinib for the treatment of esophageal squamous cell carcinoma (ESCC), we examined the expression profiles of EGFR and HER2 in tumor tissues and in paired adjacent non-neoplastic tissues from patients with ESCC. We evaluated the antitumor effects of lapatinib alone or in combination with oxaliplatin or 5-fluorouracil (5-FU) on a panel of primary ESCC cells in vitro with various levels of EGFR and HER2 expression. The in vivo effect of lapatinib alone or in combination with oxaliplatin or 5-FU was evaluated using a primary ESCC xenograft model. EGFR was overexpressed in 80.9% (76/94) of the ESCC samples, while 24.5% (23/94) of the samples overexpressed HER2. EGFR and HER2 co-overexpression was detected in 22.3% of samples (21/94). In vitro, the primary ESCC cells were more sensitive to lapatinib combined with 5-FU or oxaliplatin than to lapatinib alone. Lapatinib in combination with 5-FU had more potent antitumor effects in the primary ESCC xenograft model, and markedly reduced the phosphorylation of EGFR and HER2, compared with lapatinib alone or in combination with oxaliplatin. These data indicate that lapatinib has activity in EGFR- and/or HER2-expressing ESCC primary cells, and that lapatinib in combination with 5-FU may be a promising treatment strategy for patients with ESCC.
- Published
- 2013
- Full Text
- View/download PDF
30. Loss and reduced expression of PTEN correlate with advanced-stage gastric carcinoma
- Author
-
Wenmin Hou, Xuehua Zhu, Maogui Fei, Kurtis E. Bachman, Joel Greshock, Jiuhong Kang, Crystal Ying Qin, and Xia Qin
- Subjects
phosphatase and tensin homolog ,Cancer Research ,Pathology ,medicine.medical_specialty ,Tissue microarray ,Oncogene ,biology ,tissue microarray ,Cancer ,General Medicine ,Articles ,medicine.disease ,Molecular medicine ,receiver operator characteristic curve ,advanced gastric carcinoma ,Immunology and Microbiology (miscellaneous) ,immunohistochemistry ,Cancer research ,Carcinoma ,medicine ,biology.protein ,Tensin ,PTEN ,Immunohistochemistry - Abstract
Phosphatase and tensin homolog (PTEN) is a tumor suppressor involved in multiple cell processes. To investigate the role of PTEN in the development of gastric carcinoma, we determined the expression pattern of PTEN in primary gastric carcinoma and in paired adjacent non-neoplastic tissue. We also determined the correlation of PTEN expression with clinicopathological characteristics and patient survival. Overall, 159 gastric carcinomas and 151 paired adjacent non-neoplastic tissues were used in the present study. PTEN expression was determined using tissue microarrays and immunohistochemistry. The clinical sensitivity and specificity of PTEN expression were calculated using receiver operator characteristic curves. Results showed that the loss of cytoplasmic PTEN was significantly more frequent in carcinoma tissue compared with adjacent non-neoplastic tissue (62 vs. 5%, respectively; P
- Published
- 2012
31. Sensitivity of Cancer Cells to Plk1 Inhibitor GSK461364A Is Associated with Loss of p53 Function and Chromosome Instability
- Author
-
Maureen R. Bleam, Christopher Moy, Sylvie Laquerre, Mark Richter, Jeffrey R. Jackson, Scott Powers, Wendy S. Halsey, Richard Wooster, Yan Degenhardt, Kurtis E. Bachman, Barbara L. Weber, Nancy Liu-Sullivan, Junping Jing, Ashley M. Hughes, Aidan G. Gilmartin, Joel Greshock, and Xi-Ping Zhang
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Cancer Research ,Time Factors ,Cell Survival ,Immunoblotting ,Mitosis ,Cell Cycle Proteins ,Thiophenes ,Protein Serine-Threonine Kinases ,Biology ,PLK1 ,Polyploidy ,Inhibitory Concentration 50 ,RNA interference ,Cell Line, Tumor ,Chromosomal Instability ,Neoplasms ,Proto-Oncogene Proteins ,Chromosome instability ,medicine ,Humans ,Gene silencing ,Cell Proliferation ,Reverse Transcriptase Polymerase Chain Reaction ,Cell growth ,Cancer ,medicine.disease ,Oncology ,Mutation ,Cancer cell ,Cancer research ,RNA Interference ,Tumor Suppressor Protein p53 - Abstract
Polo-like kinases are a family of serine threonine kinases that are critical regulators of cell cycle progression and DNA damage response. Predictive biomarkers for the Plk1-selective inhibitor GSK461364A were identified by comparing the genomics and genetics of a panel of human cancer cell lines with their response to a drug washout followed by an outgrowth assay. In this assay, cell lines that have lost p53 expression or carry mutations in the TP53 gene tended to be more sensitive to GSK461364A. These more sensitive cell lines also had increased levels of chromosome instability, a characteristic associated with loss of p53 function. Further mechanistic studies showed that p53 wild-type (WT) and not mutant cells can activate a postmitotic tetraploidy checkpoint and arrest at pseudo-G1 state after GSK461364A treatment. RNA silencing of WT p53 increased the antiproliferative activity of GSK461364A. Furthermore, silencing of p53 or p21/CDKN1A weakened the tetraploidy checkpoint in cells that survived mitotic arrest and mitotic slippage. As many cancer therapies tend to be more effective in p53 WT patients, the higher sensitivity of p53-deficient tumors toward GSK461364A could potentially offer an opportunity to treat tumors that are refractory to other chemotherapies as well as early line therapy for these genotypes. Mol Cancer Ther; 9(7); 2079–89. ©2010 AACR.
- Published
- 2010
- Full Text
- View/download PDF
32. Catalogue, cause, complexity and cure; the many uses of cancer genome sequence
- Author
-
Kurtis E. Bachman and Richard Wooster
- Subjects
Genetics ,Genome, Human ,Somatic cell ,Cancer ,Sequence Analysis, DNA ,Disease ,Biology ,medicine.disease ,Genome ,DNA sequencing ,Germline mutation ,Neoplasms ,Databases, Genetic ,Mutation ,medicine ,Humans ,Genetic Predisposition to Disease ,Gene ,Developmental Biology ,Sequence (medicine) - Abstract
DNA sequence and bioinformatics technology have enabled the analysis of the cancer genome, revealing the vast genetic complexity of this disease. The patterns of somatic mutations are a rich archaeological record of the insults received by the genome that have added to our understanding of the mutagenic process. However, very few frequently mutated genes have been identified with the majority of somatic mutational events occurring infrequently. These infrequent mutations, however, have been shown to effect well-defined biological pathways that are critical in driving the development and progression of human tumours, for example the MAPK and PI3K pathways. Current cancer sequencing studies are now providing somatic mutation data for distinct tumour types and subtypes, leading to the identification of disease-specific alterations and potential therapeutic targets.
- Published
- 2010
- Full Text
- View/download PDF
33. Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells
- Author
-
Khola Tahir, Kurtis E. Bachman, Ben Ho Park, Todd Waldman, Stuart S. Martin, Marta Szmacinski, Michele Vitolo, Michele B. Weiss, and David J. Weber
- Subjects
Cancer Research ,Paclitaxel ,Morpholines ,Blotting, Western ,Apoptosis ,Biology ,Article ,Loss of heterozygosity ,Phosphatidylinositol 3-Kinases ,Nitriles ,Butadienes ,Humans ,Tensin ,PTEN ,Anoikis ,RNA, Messenger ,Epidermal growth factor receptor ,Enzyme Inhibitors ,Mammary Glands, Human ,Protein kinase B ,Cell Proliferation ,Antibiotics, Antineoplastic ,Integrases ,Reverse Transcriptase Polymerase Chain Reaction ,Cell growth ,PTEN Phosphohydrolase ,Epithelial Cells ,Flow Cytometry ,Antineoplastic Agents, Phytogenic ,Cell Transformation, Neoplastic ,Oncology ,Chromones ,Doxorubicin ,Cancer research ,biology.protein ,Female ,Mitogen-Activated Protein Kinases ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Many cancers, including breast cancer, harbor loss-of-function mutations in the catalytic domain of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) or have reduced PTEN expression through loss of heterozygosity and/or epigenetic silencing mechanisms. However, specific phenotypic effects of PTEN inactivation in human cancer cells remain poorly defined without a direct causal connection between the loss of PTEN function and the development or progression of cancer. To evaluate the biological and clinical relevance of reduced or deleted PTEN expression, a novel in vitro model system was generated using human somatic cell knockout technologies. Targeted homologous recombination allowed for a single and double allelic deletion, which resulted in reduced and deleted PTEN expression, respectively. We determined that heterozygous loss of PTEN in the nontumorigenic human mammary epithelial cell line MCF-10A was sufficient for activation of the phosphoinositide 3-kinase/AKT and mitogen-activated protein kinase pathways, whereas the homozygous absence of PTEN expression led to a further increased activation of both pathways. The deletion of PTEN was able to confer growth factor–independent proliferation, which was confirmed by the resistance of the PTEN−/− MCF-10A cells to small-molecule inhibitors of the epidermal growth factor receptor. However, neither heterozygous nor homozygous loss of PTEN expression was sufficient to promote anchorage-independent growth, but the loss of PTEN did confer apoptotic resistance to cell rounding and matrix detachment. Finally, MCF-10A cells with the reduction or loss of PTEN showed increased susceptibility to the chemotherapeutic drug doxorubicin but not paclitaxel. [Cancer Res 2009;69(21):8275–83]
- Published
- 2009
- Full Text
- View/download PDF
34. Knockin of mutant PIK3CA activates multiple oncogenic pathways
- Author
-
D. Marc Rosen, Michaela J. Higgins, Joseph P. Garay, David Cosgrove, Grace M. Wang, Josh Lauring, Morassa Mohseni, Kurtis E. Bachman, Michele B. Weiss, Ben Ho Park, John P. Gustin, Yuko Konishi, Samuel R. Denmeade, Abde M. Abukhdeir, Hiroyuki Konishi, Akina Tamaki, Michele Vitolo, and Bedri Karakas
- Subjects
Class I Phosphatidylinositol 3-Kinases ,Transplantation, Heterologous ,Mutant ,Mice, Nude ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Glycogen Synthase Kinase 3 ,Mice ,Phosphatidylinositol 3-Kinases ,Cyclin D1 ,Nude mouse ,Epidermal growth factor ,Cell Line, Tumor ,Gene knockin ,medicine ,Animals ,Humans ,Gene Knock-In Techniques ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Mammary Glands, Human ,neoplasms ,PI3K/AKT/mTOR pathway ,Mutation ,Glycogen Synthase Kinase 3 beta ,Multidisciplinary ,TOR Serine-Threonine Kinases ,Gene targeting ,Oncogenes ,Biological Sciences ,biology.organism_classification ,Cancer research ,Colorectal Neoplasms ,Protein Kinases ,Proto-Oncogene Proteins c-akt - Abstract
The phosphatidylinositol 3-kinase subunit PIK3CA is frequently mutated in human cancers. Here we used gene targeting to “knock in” PIK3CA mutations into human breast epithelial cells to identify new therapeutic targets associated with oncogenic PIK3CA. Mutant PIK3CA knockin cells were capable of epidermal growth factor and mTOR-independent cell proliferation that was associated with AKT, ERK, and GSK3β phosphorylation. Paradoxically, the GSK3β inhibitors lithium chloride and SB216763 selectively decreased the proliferation of human breast and colorectal cancer cell lines with oncogenic PIK3CA mutations and led to a decrease in the GSK3β target gene CYCLIN D1 . Oral treatment with lithium preferentially inhibited the growth of nude mouse xenografts of HCT-116 colon cancer cells with mutant PIK3CA compared with isogenic HCT-116 knockout cells containing only wild-type PIK3CA . Our findings suggest GSK3β is an important effector of mutant PIK3CA , and that lithium, an FDA-approved therapy for bipolar disorders, has selective antineoplastic properties against cancers that harbor these mutations.
- Published
- 2009
- Full Text
- View/download PDF
35. Knock-in of Mutant K-ras in Nontumorigenic Human Epithelial Cells as a New Model for Studying K-ras–Mediated Transformation
- Author
-
Bedri Karakas, Abde M. Abukhdeir, Hiroyuki Konishi, Eike Gallmeier, Josh Lauring, Kurtis E. Bachman, Joseph P. Garay, John P. Gustin, Yuko Konishi, and Ben Ho Park
- Subjects
Cancer Research ,Somatic cell ,Transgene ,Mutant ,Breast Neoplasms ,Biology ,medicine.disease_cause ,Cell Line ,Gene knockin ,medicine ,Humans ,Breast ,Transgenes ,Phosphorylation ,Telomerase ,Alleles ,Mitogen-Activated Protein Kinase Kinases ,Mutation ,Kinase ,Epithelial Cells ,Molecular biology ,Cell Transformation, Neoplastic ,Genes, ras ,Oncology ,Cell culture ,Cancer cell ,ras Proteins ,Tumor Suppressor Protein p53 ,Proto-Oncogene Proteins c-akt - Abstract
The oncogenic function of mutant ras in mammalian cells has been extensively investigated using multiple human and animal models. These systems include overexpression of exogenous mutant ras transgenes, conditionally expressed knock-in mouse models, and somatic cell knockout of mutant and wild-type ras genes in human cancer cell lines. However, phenotypic discrepancies between knock-in mice and transgenic mutant ras overexpression prompted us to evaluate the consequences of targeted knock-in of an oncogenic K-ras mutation in the nontumorigenic human breast epithelial cell line MCF-10A and hTERT-immortalized human mammary epithelial cells. Our results show several significant differences between mutant K-ras knock-in cells versus their transgene counterparts, including limited phosphorylation of the downstream molecules extracellular signal-regulated kinase and AKT, minor proliferative capacity in the absence of an exogenous growth factor, and the inability to form colonies in semisolid medium. Analysis of 16 cancer cell lines carrying mutant K-ras genes indicated that 50% of cancer cells harbor nonoverexpressed heterozygous K-ras mutations similar to the expression seen in our knock-in cell lines. Thus, this system serves as a new model for elucidating the oncogenic contribution of mutant K-ras as expressed in a large fraction of human cancer cells. [Cancer Res 2007;67(18):8460–7]
- Published
- 2007
- Full Text
- View/download PDF
36. p21 gene knock down does not identify genetic effectors seen with gene knock out
- Author
-
Michele Vitolo, Kurtis E. Bachman, Ben Ho Park, Hiroyuki Konishi, Bedri Karakas, Abde M. Abukhdeir, Ashani T. Weeraratna, and John P. Gustin
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Pharmacology ,Genetics ,Cancer Research ,biology ,Effector ,Somatic cell ,Phenotype ,Article ,Oncology ,Transforming Growth Factor beta ,RNA interference ,Cyclin-dependent kinase ,Cell Line, Tumor ,Gene expression ,biology.protein ,Humans ,Molecular Medicine ,RNA Interference ,RNA, Small Interfering ,Gene ,Gene knockout - Abstract
RNA interference (RNAi) has become a popular tool for analyzing gene function in cancer research. The feasibility of using RNAi in cellular and animal models as an alternative to conventional gene knock out approaches has been demonstrated. Although these studies show that RNAi can recapitulate phenotypes seen in knock out animals and their derived cell lines, a systematic study rigorously comparing downstream effector genes between RNAi and gene knock out has not been performed. Here we present data contrasting the phenotypic and genotypic changes that occur with either stable knock down via RNAi of the cyclin dependent kinase inhibitor p21 versus its somatic cell knock out counterpart in the human mammary epithelial cell line MCF-10A. Our results demonstrate that p21 knock down clones display a growth proliferative response upon exposure to Transforming Growth Factor-Beta Type 1 (TGFbeta) similar to p21 knock out clones. However, gene expression profiles were significantly different in p21 knock down cells versus p21 knock out clones. Importantly p21 knock down clones did not display increased gene expression of interleukin-1alpha (IL-1alpha), a critical effector of this growth response previously validated in p21 knock out cells. We conclude that gene knock out can yield additional vital information that may be missed with gene knock down strategies.
- Published
- 2007
- Full Text
- View/download PDF
37. The RUNX2 transcription factor cooperates with the YES-associated protein, YAP65, to promote cell transformation
- Author
-
Antonino Passaniti, Keli J. Renoud, Ian E. Anglin, Kurtis E. Bachman, Michele Vitolo, Ronald B. Gartenhaus, and William M. Mahoney
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,musculoskeletal diseases ,Cancer Research ,Core Binding Factor Alpha 1 Subunit ,Biology ,Models, Biological ,Cell Line ,Mice ,stomatognathic system ,Cell Line, Tumor ,Coactivator ,Animals ,Humans ,Promoter Regions, Genetic ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Pharmacology ,Cell growth ,musculoskeletal, neural, and ocular physiology ,Runt ,YAP-Signaling Proteins ,Cell cycle ,Phosphoproteins ,musculoskeletal system ,Gene Expression Regulation, Neoplastic ,RUNX2 ,Cell Transformation, Neoplastic ,Oncology ,embryonic structures ,TAF2 ,NIH 3T3 Cells ,Cancer research ,Bone maturation ,Molecular Medicine ,RNA Interference ,Transcription Factors - Abstract
The Runt box domain DNA-binding transcription factors (RUNX) play key roles in hematopoietic, bone, and gastric development. These factors regulate angiogenesis and tumorigenic events, functioning as either activators or repressors of target genes. Although RUNX2 is an essential bone maturation factor, it has also been found to promote transformation in vivo and cell proliferation in vitro, perhaps by associating with specific coactivators or corepressors. Adenoviral-mediated overexpression of dominant negative RUNX2 or specific reduction of RUNX2 with RNA-interference inhibits cell proliferation. To determine whether RUNX2 also plays a role in cell transformation, RUNX2 interactions with the coactivator Yes-associated protein (YAP65) were examined. RUNX2 associated with YAP65 via a proline-rich segment in the C-terminal domain (PPPY) and coexpression of RUNX2 and YAP65 significantly increased foci formation and anchorage-independent growth relative to each factor alone. However, in contrast to wild-type RUNX2, a mutant RUNX2(P409A), which does not bind YAP65, did not cooperate with YAP65 to promote anchorage-independent growth. RUNX2 is a strong repressor of the cyclin-dependent kinase inhibitor p21(CIP1), which is known to mediate cell transformation. Overexpression of YAP65 prevented RUNX2-dependent downregulation of p21(CIP1) protein expression while promoting cell transformation. The RUNX2(P409A) mutant retained the ability to bind DNA and repress the p21(CIP1) promoter as shown by DNA precipitation and luciferase-reporter assays, respectively, but was not able to relieve repression of the p21(CIP1) promoter. Therefore, these results reveal a novel function of the RUNX2 and YAP65 interaction in oncogenic transformation that may be mediated by modulation of p21(CIP1) protein expression.
- Published
- 2007
- Full Text
- View/download PDF
38. Persistent mismatch repair deficiency following targeted correction of hMLH1
- Author
-
Michele Vitolo, Giancarlo Marra, Ben Ho Park, Kurtis E. Bachman, Katja Baerenfaller, and M B Weiss
- Subjects
Genetics ,Cancer Research ,Base Sequence ,DNA Repair ,Base Pair Mismatch ,Genetic enhancement ,Blotting, Western ,Nuclear Proteins ,Gene targeting ,Biology ,digestive system diseases ,Stop codon ,Exon ,Cell Line, Tumor ,Cancer cell ,Humans ,Molecular Medicine ,DNA mismatch repair ,Carrier Proteins ,MutL Protein Homolog 1 ,Homologous recombination ,Molecular Biology ,Gene ,Adaptor Proteins, Signal Transducing ,DNA Primers - Abstract
The use of gene therapy to correct mutated or lost gene function for the treatment of human cancers has been an active, yet problematic area of biomedical research. Many technical difficulties, including efficient tissue-specific delivery, integration site specificity and general toxicity, are being addressed. Little is known, however, about the genetic and phenotypic stability that accompanies a successful gene-specific targeting event in a cancer cell. This question was addressed following the creation of a colon cancer cell line in which a mutated hMLH1 gene was corrected via targeted homologous recombination. This correction resulted in the expression of wild-type hMLH1 protein, restoration of the hPMS2 protein and mismatch repair (MMR) proficiency. One of two hMLH1-corrected clones, however, was found to retain defects in MMR activity. These cells continued to express the corrected hMLH1 protein, but had lost expression of another MMR protein, hMSH6. DNA sequence analysis of the hMSH6 gene revealed biallelic expansions of a cytosine repeat region in exon 5 that result in frameshifts leading to premature stop codons. These findings suggest that, similar to acquired drug resistance, the presence of genetically heterogeneous cancer cell populations or acquisition of compensatory mutations can result in 'resistance' to gene replacement therapy.
- Published
- 2006
- Full Text
- View/download PDF
39. The Consensus Coding Sequences of Human Breast and Colorectal Cancers
- Author
-
Tobias Sjöblom, Siân Jones, Laura D. Wood, D. Williams Parsons, Jimmy Lin, Thomas D. Barber, Diana Mandelker, Rebecca J. Leary, Janine Ptak, Natalie Silliman, Steve Szabo, Phillip Buckhaults, Christopher Farrell, Paul Meeh, Sanford D. Markowitz, Joseph Willis, Dawn Dawson, James K. V. Willson, Adi F. Gazdar, James Hartigan, Leo Wu, Changsheng Liu, Giovanni Parmigiani, Ben Ho Park, Kurtis E. Bachman, Nickolas Papadopoulos, Bert Vogelstein, Kenneth W. Kinzler, and Victor E. Velculescu
- Subjects
Male ,Colorectal cancer ,Sequence analysis ,Breast Neoplasms ,Biology ,Polymerase Chain Reaction ,Genome ,Breast cancer ,Cell Line, Tumor ,Consensus Sequence ,medicine ,Consensus sequence ,Humans ,Neoplasm ,Gene ,Multidisciplinary ,Genome, Human ,Computational Biology ,Sequence Analysis, DNA ,medicine.disease ,Amino Acid Substitution ,Mutation ,Immunology ,Cancer research ,Female ,Human genome ,Colorectal Neoplasms ,Databases, Nucleic Acid ,Genes, Neoplasm - Abstract
The elucidation of the human genome sequence has made it possible to identify genetic alterations in cancers in unprecedented detail. To begin a systematic analysis of such alterations, we determined the sequence of well-annotated human protein-coding genes in two common tumor types. Analysis of 13,023 genes in 11 breast and 11 colorectal cancers revealed that individual tumors accumulate an average of ∼90 mutant genes but that only a subset of these contribute to the neoplastic process. Using stringent criteria to delineate this subset, we identified 189 genes (average of 11 per tumor) that were mutated at significant frequency. The vast majority of these genes were not known to be genetically altered in tumors and are predicted to affect a wide range of cellular functions, including transcription, adhesion, and invasion. These data define the genetic landscape of two human cancer types, provide new targets for diagnostic and therapeutic intervention, and open fertile avenues for basic research in tumor biology.
- Published
- 2006
- Full Text
- View/download PDF
40. Physiologic estrogen receptor alpha signaling in non-tumorigenic human mammary epithelial cells
- Author
-
Keith Brenner, Judith Keen, Brian G. Blair, Kurtis E. Bachman, Michele Vitolo, Nancy E. Davidson, Abde M. Abukhdeir, Joselin Lim, Yi Huang, Hiroyuki Konishi, Ben Ho Park, Vanita Sahasranaman, and Bedri Karakas
- Subjects
Cancer Research ,medicine.medical_specialty ,Time Factors ,Estrogen receptor ,Biology ,Response Elements ,Epidermal growth factor ,Cell Line, Tumor ,Internal medicine ,medicine ,Humans ,Breast ,Phosphorylation ,skin and connective tissue diseases ,Estrogen receptor beta ,Cell Proliferation ,Estradiol ,Fulvestrant ,Estrogen Receptor alpha ,GATA3 ,Epithelial Cells ,Estrogens ,Sequence Analysis, DNA ,Endocrinology ,Gene Expression Regulation ,Oncology ,Selective estrogen receptor modulator ,Cancer research ,Precancerous Conditions ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists ,Tamoxifen ,Signal Transduction ,medicine.drug - Abstract
Currently, a number of breast cancer cell lines exist that serve as models for both estrogen receptor alpha (ERalpha) positive and ERalpha negative disease. Models are also available for pre-neoplastic breast epithelial cells that do not express ERalpha; however, there are no ideal systems for studying pre-neoplastic cells that are ERalpha positive. This has been largely due to the inability to establish an estrogen growth stimulated, non-tumorigenic breast epithelial cell line, as most human breast epithelial cells engineered to overexpress ERalpha have been found to be growth inhibited by estrogens. We have developed independently derived clones from the non-cancerous MCF-10A human breast cell line that express ERalpha and are growth stimulated by 17-beta-estradiol (E2) in the absence of epidermal growth factor (EGF), a cytokine normally required for MCF-10A cell proliferation. This effect is blocked by the selective estrogen receptor modulator (SERM), Tamoxifen and the selective estrogen receptor downregulator, ICI 182,780 (Faslodex, Fulvestrant). Exposure of these cells to EGF and E2 results in a growth inhibitory phenotype similar to previous reports. These data present a reconciling explanation for the previously described paradoxical effects of ERalpha overexpression, and provide a model for examining the carcinogenic effects of estrogens in non-tumorigenic human breast epithelial cells.
- Published
- 2006
- Full Text
- View/download PDF
41. Mutation of the PIK3CA oncogene in human cancers
- Author
-
Bedri Karakas, Kurtis E. Bachman, and Ben Ho Park
- Subjects
Cancer Research ,Class I Phosphatidylinositol 3-Kinases ,p110α ,Reviews ,Phosphatidylinositol 3-Kinases ,P110α ,Biology ,medicine.disease_cause ,PI3K ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Germline mutation ,Neoplasms ,medicine ,Humans ,Phosphatidylinositol ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Genetics ,0303 health sciences ,Mutation ,Cancer ,PIK3CA ,medicine.disease ,3. Good health ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,somatic mutations ,Carcinogenesis ,Signal Transduction - Abstract
It is now well established that cancer is a genetic disease and that somatic mutations of oncogenes and tumour suppressor genes are the initiators of the carcinogenic process. The phosphatidylinositol 3-kinase signalling pathway has previously been implicated in tumorigenesis, and evidence over the past year suggests a pivotal role for the phosphatidylinositol 3-kinase catalytic subunit, PIK3CA, in human cancers. In this review, we analyse recent reports describing PIK3CA mutations in a variety of human malignancies, and discuss their possible implications for diagnosis and therapy.
- Published
- 2006
- Full Text
- View/download PDF
42. De novo CpG Island Methylation in Human Cancer Cells
- Author
-
Kornel E. Schuebel, Kurtis E. Bachman, Ray Whay Chiu Yen, Hiromu Suzuki, Angela H. Ting, Ina Rhee, Kam Wing Jair, and Stephen B. Baylin
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Cancer Research ,Methyltransferase ,DNMT3B ,environment and public health ,DNA methyltransferase ,chemistry.chemical_compound ,Tumor Cells, Cultured ,Animals ,Humans ,Medicine ,DNA (Cytosine-5-)-Methyltransferases ,Gene ,Genetics ,urogenital system ,business.industry ,Methylation ,DNA Methylation ,Oncology ,CpG site ,chemistry ,Colonic Neoplasms ,embryonic structures ,DNA methylation ,CpG Islands ,Drosophila ,Genetic Engineering ,business ,DNA - Abstract
A major obstacle toward understanding how patterns of abnormal mammalian cytosine DNA methylation are established is the difficulty in quantitating the de novo methylation activities of DNA methyltransferases (DNMT) thought to catalyze these reactions. Here, we describe a novel method, using native human CpG island substrates from genes that frequently become hypermethylated in cancer, which generates robust activity for measuring de novo CpG methylation. We then survey colon cancer cells with genetically engineered deficiencies in different DNMTs and find that the major activity against these substrates in extracts of these cells is DNMT1, with minor contribution from DNMT 3b and none from DNMT3a, the only known bona fide de novo methyltransferases. The activity of DNMT1 against unmethylated CpG rich DNA was further tested by introducing CpG island substrates and DNMT1 into Drosophila melanogaster cells. The exogenous DNMT1 methylates the integrated mammalian CpG islands but not the Drosophila DNA. Additionally, in human cancer cells lacking DNMT1 and DNMT3b and having nearly absent genomic methylation, gene-specific de novo methylation can be initiated by reintroduction of DNMT1. Our studies provide a new assay for de novo activity of DNMTs and data suggesting a potential role for DNMT1 in the initiation of promoter CpG island hypermethylation in human cancer cells. (Cancer Res 2006; 66(2): 682-92)
- Published
- 2006
- Full Text
- View/download PDF
43. Duel nature of TGF-?? signaling: tumor suppressor vs. tumor promoter
- Author
-
Kurtis E. Bachman and Ben Ho Park
- Subjects
Cancer Research ,Growth factor ,medicine.medical_treatment ,Epithelial Cells ,SMAD ,Biology ,Endoglin ,Cell biology ,Paracrine signalling ,Cell Transformation, Neoplastic ,Phenotype ,Cytokine ,Oncology ,Transforming Growth Factor beta ,Neoplasms ,TGF beta signaling pathway ,medicine ,Cytokines ,Humans ,Genes, Tumor Suppressor ,Signal transduction ,Promoter Regions, Genetic ,Receptors, Transforming Growth Factor beta ,Signal Transduction ,Transforming growth factor - Abstract
Transforming growth factor beta type I (TGF-beta) is a ubiquitous cytokine that is well known for its ability to inhibit epithelial cell proliferation. Somatic mutations abrogating the TGF-beta signal transduction pathway are found in many gastrointestinal cancers, confirming its importance as a tumor suppressor. In contrast, many nongastrointestinal epithelial malignancies lack these somatic alterations, yet these cancers still acquire resistance to the growth-inhibitory effects of TGF-beta. In many instances, this resistance is part of a signaling switch whereby TGF-beta loses its growth inhibitory effects and is then used by the epithelial cell in a growth-promoting fashion. The mechanisms that underlie this change in the phenotypic growth response to TGF-beta are now being elucidated. This review focuses on recent advances in understanding the dual nature of the TGF-beta pathway as it relates to human carcinogenesis.Elucidating the molecular basis that enables epithelial cells to change from a growth-suppressive to growth-stimulatory phenotype on TGF-beta exposure is an area of active research. Besides enhancing cancer cell growth, TGF-beta is also thought to promote a malignant cell's ability to metastasize by mediating changes in the cytoskeletal architecture, known as an epithelial-to-mesenchymal transition. This process enables a cancer cell to invade and spread to distal sites. Strong evidence has now emerged suggesting that the ability of a cell to use TGF-beta as a growth-promoting/invasive cytokine is a result of a number of different cellular and nuclear factors, including the absence or disruption of cyclin-dependent kinase inhibitors. This imbalance in cell cycle regulators may be the key element that dictates a cell's response to TGF-beta as growth-inhibitory versus growth-stimulatory, thus explaining the dual nature of TGF-beta signaling.Current studies are beginning to shed light on the mechanisms that allow some nongastrointestinal epithelial cancers to evade the growth inhibitory effects of TGF-beta while simultaneously using this cytokine for growth advantage. By dissecting this phenotypic switch during tumor development, important genes, proteins, and pathways that are involved with TGF-beta signaling continue to be discovered. Knowledge of how premalignant cells and tumor cells respond to the growth promoting effects of TGF-beta and the genes that regulate this process will aid in the development of novel therapeutics and treatment strategies.
- Published
- 2005
- Full Text
- View/download PDF
44. The Methyl-CpG Binding Protein MBD1 Interacts with the p150 Subunit of Chromatin Assembly Factor 1
- Author
-
Michael R. Rountree, Stephen B. Baylin, Brian E. Reese, and Kurtis E. Bachman
- Subjects
Chromosomal Proteins, Non-Histone ,Macromolecular Substances ,DNA methyltransferase ,Chromodomain ,Mice ,Non-histone protein ,Heterochromatin ,Animals ,Humans ,Chromatin Assembly Factor-1 ,Molecular Biology ,Cells, Cultured ,Transcriptional Regulation ,Cell Nucleus ,Mammals ,Genetics ,Binding Sites ,biology ,3T3 Cells ,Cell Biology ,DNA Methylation ,Mi-2/NuRD complex ,Chromatin ,DNA-Binding Proteins ,Repressor Proteins ,Histone ,Chromobox Protein Homolog 5 ,Multiprotein Complexes ,biology.protein ,CpG Islands ,Heterochromatin protein 1 ,Carrier Proteins ,Protein Binding ,Transcription Factors - Abstract
The methylation of cytosines in promoter regions containing CpG islands leads to the transcriptional inactivation of the downstream coding sequence in vertebrate cells (3, 34). This epigenetic regulation of gene expression has been shown to play a role in the silencing of imprinted alleles, genes on the inactive X-chromosome, and a number of tumor suppressor genes (3, 4). It is thought that this method of transcriptional silencing can be mediated through the recruitment of chromatin-modifying factors, such as histone deacetylases (HDACs), to the methylated promoters (6, 27, 34, 44). The subsequent altering of the local chromatin structure can lead to gene silencing through the formation of a heritable heterochromatin state (5). A family of proteins known as methyl-CpG binding proteins (MBD1 to -4), is thought to play an important role in methylation-mediated transcriptional silencing (6, 27, 29, 44). MeCP2 was the first member of this family to be characterized (20). It contains a methyl-CpG binding domain (MBD) and a transcriptional-repression domain (TRD), which facilitates an interaction with, and targets the Sin3A/HDAC complex to, methylated DNA (27). Like MeCP2, MBD1 to -3 have been shown to be potent transcriptional repressors (6, 13, 29, 30, 45, 46). MBD4 is a DNA glycosylase which repairs G:T mismatches (14). Each member of this family, with the exception of MBD3, forms complexes with methylated DNA in mammalian cells, and all but MBD1 and MBD4 have been placed in known chromatin-remodeling complexes (6, 13, 14, 29, 30, 45, 46). MBD2 is a component of the MeCP1 repressor complex and directly interacts with the Sin3A/HDAC members within this complex (6, 30). MBD3 has been shown to be a member of the Mi-2/NuRD complex (44, 46). MBD1 (formally, PCM1) is the least characterized of the original methyl-CpG binding proteins. Deletion studies have demonstrated that MBD1 contains an N-terminal MBD that is required for binding to methylated DNA and a C-terminal TRD that mediates transcriptional repression through an association with HDAC activity (7, 11, 12, 29). The exact mechanism through which the TRD facilitates transcriptional repression, or how MBD1 itself initiates transcriptional silencing, is not completely understood, since MBD1 has not been placed in a known repressor complex. Unlike the other methyl-CpG binding proteins, which act as transcriptional repressors at methylated promoters, full-length MBD1 has been shown to repress unmethylated promoters in vitro (11, 12). This repression activity appears to be dependent on three cysteine-rich motifs, known as CXXC domains, similar to the CXXC domains seen in DNA methyltransferase 1 (DNMT1) and the mammalian trithorax protein, HRX1 (7, 11, 12). MBD1 is the only methyl-CpG binding protein to contain these domains. Alternative splicing within the C terminus and CXXC domains gives rise to five isoforms of MBD1 (12). These alternatively spliced variants show differences in both methylation-dependent and -independent repression (11, 12). It is now apparent that particular chromatin states play critical roles in the transcriptional activity of the cell. One of the key players in the determination and inheritance of such chromatin states is chromatin assembly factor 1 (CAF-1) (25, 37, 38). CAF-1 is a complex of three subunits, p150, p60, and p48, and is responsible for the assembly of nucleosomes onto newly replicated DNA (16). The CAF-1 complex initiates nucleosome assembly following replication by recruiting acetylated histones H3 and H4 to the DNA (43). The p150 subunit is associated with the replication-coupled assembly activity and is known to interact with PCNA, linking its activity to DNA replication and DNA repair (22, 23, 36). In Saccharomyces cerevisiae, CAF-1 is responsible for the inheritance of epigenetically determined chromatin states and subsequent gene silencing. Deletion mutants of the three CAF-1 subunits are defective in the stable inheritance of gene silencing at the mating-type loci and at telomeres (9, 10, 17, 24). The p150 subunit associates with transcriptionally silent heterochromatin through its interaction with HP1 proteins (25). In addition, recent evidence suggests that CAF-1 may also play a key role in methylation-mediated gene silencing. Expression of a truncated form of the p150 subunit of CAF-1, a fragment lacking a portion of the N terminus, was capable of relieving the repression of a methylated reporter gene without disrupting the promoter methylation (42). In the present study, we show a direct interaction between MBD1 and the p150 subunit of CAF-1. We have determined that the C terminus of CAF-1 p150 and the MBD of MBD1 are essential for this interaction and that these two proteins form a complex in mammalian cells with HP1α. Furthermore, we show in nuclear localization studies the potential importance of the CAF-1 p150 subunit in the association of MBD1 with heterochromatin. This interaction now places MBD1 in a known chromatin assembly and structuring complex that is associated with transcriptional repression.
- Published
- 2003
- Full Text
- View/download PDF
45. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells
- Author
-
Christoph Lengauer, Stephen B. Baylin, Ina Rhee, Ray Whay Chiu Yen, Hengmi Cui, Andrew P. Feinberg, Kurtis E. Bachman, Bert Vogelstein, Ben Ho Park, Kenneth W. Kinzler, Kornel E. Schuebel, and Kam Wing Jair
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Molecular Sequence Data ,Biology ,DNA methyltransferase ,Epigenetics of physical exercise ,Insulin-Like Growth Factor II ,Neoplasms ,Histone methylation ,Tumor Cells, Cultured ,Humans ,Genes, Tumor Suppressor ,Amino Acid Sequence ,DNA (Cytosine-5-)-Methyltransferases ,Gene Silencing ,Cancer epigenetics ,Epigenetics ,Promoter Regions, Genetic ,RNA-Directed DNA Methylation ,Cyclin-Dependent Kinase Inhibitor p16 ,Tissue Inhibitor of Metalloproteinase-3 ,Multidisciplinary ,DNA, Neoplasm ,DNA Methylation ,Gene Expression Regulation, Neoplastic ,Histone methyltransferase ,Gene Targeting ,DNA methylation ,Cancer research - Abstract
Inactivation of tumour suppressor genes is central to the development of all common forms of human cancer. This inactivation often results from epigenetic silencing associated with hypermethylation rather than intragenic mutations. In human cells, the mechanisms underlying locus-specific or global methylation patterns remain unclear. The prototypic DNA methyltransferase, Dnmt1, accounts for most methylation in mouse cells, but human cancer cells lacking DNMT1 retain significant genomic methylation and associated gene silencing. We disrupted the human DNMT3b gene in a colorectal cancer cell line. This deletion reduced global DNA methylation by less than 3%. Surprisingly, however, genetic disruption of both DNMT1 and DNMT3b nearly eliminated methyltransferase activity, and reduced genomic DNA methylation by greater than 95%. These marked changes resulted in demethylation of repeated sequences, loss of insulin-like growth factor II (IGF2) imprinting, abrogation of silencing of the tumour suppressor gene p16INK4a, and growth suppression. Here we demonstrate that two enzymes cooperatively maintain DNA methylation and gene silencing in human cancer cells, and provide compelling evidence that such methylation is essential for optimal neoplastic proliferation.
- Published
- 2002
- Full Text
- View/download PDF
46. Dnmt3a and Dnmt3b Are Transcriptional Repressors That Exhibit Unique Localization Properties to Heterochromatin
- Author
-
Stephen B. Baylin, Michael R. Rountree, and Kurtis E. Bachman
- Subjects
X-linked Nuclear Protein ,Transcription, Genetic ,Euchromatin ,Heterochromatin ,DNMT3B ,Biology ,Biochemistry ,DNA-binding protein ,Histone Deacetylases ,DNA Methyltransferase 3A ,Mice ,Animals ,DNA (Cytosine-5-)-Methyltransferases ,Molecular Biology ,EZH2 ,DNA Helicases ,Nuclear Proteins ,3T3 Cells ,Cell Biology ,Cell cycle ,Molecular biology ,Chromatin ,DNA-Binding Proteins ,Repressor Proteins ,embryonic structures ,Heterochromatin protein 1 ,Transcription Factors - Abstract
We demonstrate that the recently identified DNA methyltransferases, Dnmt3a and Dnmt3b, like DNMT1, repress transcription in a methylation-independent manner. Dnmt3a and Dnmt3b repress transcription primarily through a plant homeodomain-like motif that is shared with the ATRX protein but is not present in DNMT1. Unlike DNMT1, which localizes to replication foci during S-phase in murine embryonic fibroblasts, Dnmt3a co-localizes with heterochromatin protein 1 alpha (HP1 alpha) and methyl-CpG binding proteins throughout the cell cycle to late-replicating pericentromeric heterochromatin. In contrast to Dnmt3a, Dnmt3b remained diffuse in the nucleus of embryonic fibroblasts at all cell cycle stages. However, Dnmt3a and Dnmt3b co-localize to these pericentromeric heterochromatin regions in murine embryonic stem cells. This finding is important to the fact that mutations in DNMT3B are found in the developmental syndrome, ICF (immunodeficiency, centromeric heterochromatin instability, and facial anomalies), which involves extensive loss of methylation from pericentromeric regions. The localization of Dnmt3a and Dnmt3b was unaffected in Dnmt1 null embryonic stem cells, which lose the majority of methylation at pericentromeric major satellite repeats, suggesting that these enzymes are not dependent upon preexisting methylation for their targeting. DNMT1 is then positioned to reestablish transcriptionally repressive chromatin as cells replicate, while Dnmt3a and Dnmt3b may help to establish such chromatin in late S-phase and maintain this repressive heterochromatin throughout the cell cycle in a developmentally and/or cell type manner.
- Published
- 2001
- Full Text
- View/download PDF
47. Aberrant patterns of DNA methylation, chromatin formation and gene expression in cancer
- Author
-
James G. Herman, Manel Esteller, Stephen B. Baylin, Kornel E. Schuebel, Kurtis E. Bachman, and Michael R. Rountree
- Subjects
Transcription, Genetic ,Biology ,Models, Biological ,Epigenetics of physical exercise ,Neoplasms ,Histone methylation ,Genetics ,Animals ,Humans ,Gene Silencing ,Epigenetics ,Cancer epigenetics ,Promoter Regions, Genetic ,Molecular Biology ,RNA-Directed DNA Methylation ,Genetics (clinical) ,Epigenomics ,Pioneer factor ,General Medicine ,DNA Methylation ,Chromatin ,Gene Expression Regulation, Neoplastic ,Mutation ,DNA methylation ,Disease Progression ,Cancer research ,CpG Islands - Abstract
Gene function in cancer can be disrupted either through genetic alterations, which directly mutate or delete genes, or epigenetic alterations, which alter the heritable state of gene expression. The latter events are mediated by formation of transcriptionally repressive chromatin states around gene transcription start sites and an associated gain of methylation in normally unmethylated CpG islands in these regions. The genes affected include over half of the tumor suppressor genes that cause familial cancers when mutated in the germline; the selective advantage for genetic and epigenetic dysfunction in these genes is very similar. The aberrant methylation can begin very early in tumor progression and mediate most of the important pathway abnormalities in cancer including loss of cell cycle control, altered function of transcription factors, altered receptor function, disruption of normal cell-cell and cell-substratum interaction, inactivation of signal transduction pathways, loss of apoptotic signals and genetic instability. The active role of the aberrant methylation in transcriptional silencing of genes is becoming increasingly understood and involves a synergy between the methylation and histone deacetylase (HDAC) activity. This synergy can be mediated directly by HDAC interaction with DNA methylating enzymes and by recruitment through complexes involving methyl-cytosine binding proteins. In the translational arena, the promoter hypermethylation changes hold great promise as DNA tumor markers and their potentially reversible state creates a target for cancer therapeutic strategies involving gene reactivation.
- Published
- 2001
- Full Text
- View/download PDF
48. Analysis of glutamine dependency in non-small cell lung cancer: GLS1 splice variant GAC is essential for cancer cell growth
- Author
-
Richard Wooster, A. Pieter J. van den Heuvel, Kurtis E. Bachman, and Junping Jing
- Subjects
Cancer Research ,Lung Neoplasms ,Glutamine ,Biology ,Glutaminase ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Humans ,Protein Isoforms ,Molecular Targeted Therapy ,Cell Proliferation ,Pharmacology ,Glutaminolysis ,Cell growth ,Cancer ,medicine.disease ,Warburg effect ,Oncology ,Biochemistry ,Cell culture ,Anaerobic glycolysis ,Cancer cell ,Cancer research ,Molecular Medicine ,Glycolysis ,Research Paper - Abstract
One of the hallmarks of cancer is metabolic deregulation. Many tumors display increased glucose uptake and breakdown through the process of aerobic glycolysis, also known as the Warburg effect. Less studied in cancer development and progression is the importance of the glutamine (Gln) pathway, which provides cells with a variety of essential products to sustain cell proliferation, such as ATP and macromolecules for biosynthesis. To this end Gln dependency was assessed in a panel of non-small cell lung cancer lines (NSCLC). Gln was found to be essential for the growth of cells with high rates of glutaminolysis, and after exploring multiple genes in the Gln pathway, GLS1 was found to be the key enzyme associated with this dependence. This dependence was confirmed by observing the rescue of decreased growth by exogenous addition of downstream metabolites of glutaminolysis. Expression of the GLS1 splice variant KGA was found to be decreased in tumors compared with normal lung tissue. Transient knock down of GLS1 splice variants indicated that loss of GAC had the most detrimental effect on cancer cell growth. In conclusion, NSCLC cell lines depend on Gln for glutaminolysis to a varying degree, in which the GLS1 splice variant GAC plays an essential role and is a potential target for cancer metabolism-directed therapy.
- Published
- 2012
49. Combined Phosphatase and Tensin Homolog (PTEN) Loss and Fatty Acid Synthase (FAS) Overexpression Worsens the Prognosis of Chinese Patients with Hepatocellular Carcinoma
- Author
-
Xia Qin, Joel Greshock, Wenmin Hou, Xuehua Zhu, Kurtis E. Bachman, Richard Wooster, Maogui Fei, Jiuhong Kang, and Crystal Ying Qin
- Subjects
Male ,PTEN ,lcsh:Chemistry ,Immunoenzyme Techniques ,FAS ,prognosis ,Chinese ,hepatocellular carcinoma ,Tensin ,lcsh:QH301-705.5 ,Spectroscopy ,Tissue microarray ,Liver Neoplasms ,General Medicine ,Middle Aged ,Prognosis ,Computer Science Applications ,Fatty Acid Synthase, Type I ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Fatty acid synthase ,Lymphatic Metastasis ,Hepatocellular carcinoma ,Immunohistochemistry ,Female ,Adult ,China ,Carcinoma, Hepatocellular ,Biology ,Article ,Catalysis ,Inorganic Chemistry ,Downregulation and upregulation ,Biomarkers, Tumor ,medicine ,Humans ,Neoplasm Invasiveness ,Physical and Theoretical Chemistry ,Molecular Biology ,Survival rate ,Aged ,Neoplasm Staging ,Organic Chemistry ,PTEN Phosphohydrolase ,medicine.disease ,digestive system diseases ,lcsh:Biology (General) ,lcsh:QD1-999 ,biology.protein ,Cancer research ,Neoplasm Grading - Abstract
We aimed to investigate the expression pattern of phosphatase and tensin homolog (PTEN), to evaluate the relationship between PTEN expression and clinicopathological characteristics, including fatty acid synthase (FAS) expression, and to determine the correlations of PTEN and FAS expression with survival in Chinese patients with hepatocellular carcinoma (HCC). The expression patterns of PTEN and FAS were determined using tissue microarrays and immunohistochemistry. The expression of PTEN was compared with the clinicopathological characteristics of HCC, including FAS expression. Receiver operator characteristic curves were used to calculate the clinical sensitivity and specificity of PTEN expression. Kaplan-Meier survival curves were constructed to evaluate the correlations of PTEN loss and FAS overexpression with overall survival. We found that the loss of PTEN expression occurred predominantly in the cytoplasm, while FAS was mainly localized to the cytoplasm. Cytoplasmic and total PTEN expression levels were significantly decreased in HCC compared with adjacent non-neoplastic tissue (both, p < 0.0001). Decreased cytoplasmic and total PTEN expression showed significant clinical sensitivity and specificity for HCC (both, p < 0.0001). Downregulation of PTEN in HCC relative to non-neoplastic tissue was significantly correlated with histological grade (p = 0.043 for histological grades I–II versus grade III). Loss of total PTEN was significantly correlated with FAS overexpression (p = 0.014). Loss of PTEN was also associated with poor prognosis of patients with poorly differentiated HCC (p = 0.049). Moreover, loss of PTEN combined with FAS overexpression was associated with significantly worse prognosis compared with other HCC cases (p = 0.011). Our data indicate that PTEN may serve as a potential diagnostic and prognostic marker of HCC. Upregulating PTEN expression and inhibiting FAS expression may offer a novel therapeutic approach for HCC.
- Published
- 2012
- Full Text
- View/download PDF
50. Detection of tumor PIK3CA status in metastatic breast cancer using peripheral blood
- Author
-
Kathleen M. Murphy, Stacie Jeter, Jane Zorzi, Pedram Argani, Ben Ho Park, Michaela J. Higgins, Leisha A. Emens, Vered Stearns, George R. Oliver, Evan Barnathan, Shannon Slater, Brian G. Blair, Antonio C. Wolff, Leslie Cope, Kurtis E. Bachman, Peng Huang, Frank Diehl, Penny Powers, Danijela Jelovac, Phillip Angenendt, Joel Greshock, John H. Fetting, and Carol D. Riley
- Subjects
Oncology ,Adult ,Genetic Markers ,Cancer Research ,medicine.medical_specialty ,Pathology ,Class I Phosphatidylinositol 3-Kinases ,Concordance ,Breast Neoplasms ,medicine.disease_cause ,Article ,Cohort Studies ,Phosphatidylinositol 3-Kinases ,Breast cancer ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Neoplasm Metastasis ,Prospective cohort study ,neoplasms ,Aged ,Retrospective Studies ,Aged, 80 and over ,Mutation ,business.industry ,Retrospective cohort study ,DNA, Neoplasm ,Middle Aged ,medicine.disease ,Metastatic breast cancer ,Genetic marker ,Female ,business ,Cohort study - Abstract
Purpose: We sought to evaluate the feasibility of detecting PIK3CA mutations in circulating tumor DNA (ctDNA) from plasma of patients with metastatic breast cancer using a novel technique called BEAMing. Experimental Design: In a retrospective analysis, 49 tumor and temporally matched plasma samples from patients with breast cancer were screened for PIK3CA mutations by BEAMing. We then prospectively screened the ctDNA of 60 patients with metastatic breast cancer for PIK3CA mutations by BEAMing and compared the findings with results obtained by screening corresponding archival tumor tissue DNA using both sequencing and BEAMing. Results: The overall frequency of PIK3CA mutations by BEAMing was similar in both patient cohorts (29% and 28.3%, respectively). In the retrospective cohort, the concordance of PIK3CA mutation status by BEAMing between formalin-fixed, paraffin-embedded (FFPE) samples and ctDNA from temporally matched plasma was 100% (34 of 34). In the prospective cohort, the concordance rate among 51 evaluable cases was 72.5% between BEAMing of ctDNA and sequencing of archival tumor tissue DNA. When the same archival tissue DNA was screened by both sequencing and BEAMing for PIK3CA mutations (n = 41 tissue samples), there was 100% concordance in the obtained results. Conclusions: Analysis of plasma-derived ctDNA for the detection of PIK3CA mutations in patients with metastatic breast cancer is feasible. Our results suggest that PIK3CA mutational status can change upon disease recurrence, emphasizing the importance of reassessing PIK3CA status on contemporary (not archival) biospecimens. These results have implications for the development of predictive biomarkers of response to targeted therapies. Clin Cancer Res; 18(12); 3462–9. ©2012 AACR.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.