43 results on '"Phuongmai Nguyen"'
Search Results
2. Supplementary Figures 1-6, Methods and Materials from CTCFL/BORIS Is a Methylation-Independent DNA-Binding Protein That Preferentially Binds to the Paternal H19 Differentially Methylated Region
- Author
-
David Gius, Andrew P. Feinberg, Mitsuo Oshimura, Hiroyuki Kugoh, Richard S. Lee, Krish Patel, Lunching Sun, Kheem S. Bisht, Hengmi Cui, and Phuongmai Nguyen
- Abstract
Supplementary Figures 1-6, Methods and Materials from CTCFL/BORIS Is a Methylation-Independent DNA-Binding Protein That Preferentially Binds to the Paternal H19 Differentially Methylated Region
- Published
- 2023
3. Data from DNA Methyltransferase 1 and 3B Activate BAG-1 Expression via Recruitment of CTCFL/BORIS and Modulation of Promoter Histone Methylation
- Author
-
David Gius, Andrew P. Feinberg, Jane B. Trepel, Sunmin Lee, Hengmi Cui, Wenqiang Yu, C. Matthew Bradbury, Allen S. Ho, Gil Bar-Sela, Kheem S. Bisht, Phuongmai Nguyen, Lei Huang, and Lunching Sun
- Abstract
In a previous genomic analysis, using somatic methyltransferase (DNMT) knockout cells, we showed that hypomethylation decreased the expression of as many genes as were observed to increase, suggesting a previously unknown mechanism for epigenetic regulation. To address this idea, the expression of the BAG family genes was used as a model. These genes were used because their expression was decreased in DNMT1−/−, DNMT3B−/−, and double knockout cells and increased in DNMT1-overexpressing and DNMT3B-overexpressing cells. Chromatin immunoprecipitation analysis of the BAG-1 promoter in DNMT1-overexpressing or DNMT3B-overexpressing cells showed a permissive dimethyl-H3-K4/dimethyl-H3-K9 chromatin status associated with DNA-binding of CTCFL/BORIS, as well as increased BAG-1 expression. In contrast, a nonpermissive dimethyl-H3-K4/dimethyl-H3-K9 chromatin status was associated with CTCF DNA-binding and decreased BAG-1 expression in the single and double DNMT knockout cells. BORIS short hairpin RNA knockdown decreased both promoter DNA-binding, as well as BAG-1 expression, and changed the dimethyl-H3-K4/dimethyl-H3-K9 ratio to that characteristic of a nonpermissive chromatin state. These results suggest that DNMT1 and DNMT3B regulate BAG-1 expression via insulator protein DNA-binding and chromatin dynamics by regulating histone dimethylation. [Cancer Res 2008;68(8):2726–35]
- Published
- 2023
4. SIRT3 interacts with the daf-16 homolog FOXO3a in the Mitochondria, as well as increases FOXO3a Dependent Gene expression
- Author
-
Kristi Muldoon Jacobs, J. Daniel Pennington, Kheem S. Bisht, Nukhet Aykin-Burns, Hyun-Seok Kim, Mark Mishra, Lunching Sun, Phuongmai Nguyen, Bong-Hyun Ahn, Jaime Leclerc, Chu-Xia Deng, Douglas R. Spitz, David Gius
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Cellular longevity is a complex process relevant to age-related diseases including but not limited to chronic illness such as diabetes and metabolic syndromes. Two gene families have been shown to play a role in the genetic regulation of longevity; the Sirtuin and FOXO families. It is also established that nuclear Sirtuins interact with and under specific cellular conditions regulate the activity of FOXO gene family proteins. Thus, we hypothesize that a mitochondrial Sirtuin (SIRT3) might also interact with and regulate the activity of the FOXO proteins. To address this we used HCT116 cells overexpressing either wild-type or a catalytically inactive dominant negative SIRT3. For the first time we establish that FOXO3a is also a mitochondrial protein and forms a physical interaction with SIRT3 in mitochondria. Overexpression of a wild-type SIRT3 gene increase FOXO3a DNA-binding activity as well as FOXO3a dependent gene expression. Biochemical analysis of HCT116 cells over expressing the deacetylation mutant, as compared to wild-type SIRT3 gene, demonstrated an overall oxidized intracellular environment, as monitored by increase in intracellular superoxide and oxidized glutathione levels. As such, we propose that SIRT3 and FOXO3a comprise a potential mitochondrial signaling cascade response pathway.
- Published
- 2008
5. Sirt2 Regulates Radiation-Induced Injury
- Author
-
DeeDee Smart, Sudhanshu Shukla, Phuongmai Nguyen, Gopal Abbineni, and Ryan Liu
- Subjects
Programmed cell death ,Radiation ,DNA damage ,DNA repair ,Cell ,Biophysics ,Biology ,medicine.disease_cause ,SIRT2 ,Article ,030218 nuclear medicine & medical imaging ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Knockout mouse ,medicine ,Radiology, Nuclear Medicine and imaging ,Progenitor cell ,Carcinogenesis - Abstract
Nguyen, P., Shukla, S., Liu, R., Abbineni, G. and Smart, D. K. Sirt2 Regulates Radiation-Induced Injury. Radiat. Res. 191, 398–412 (2019).Sirtuin 2 (SIRT2) plays a major role in aging, carcinogenesis and neurodegeneration. While it has been shown that SIRT2 is a mediator of stress-induced cell death, the mechanism remains unclear. In this study, we report the role of SIRT2 in mediating radiation-induced cell death and DNA damage using mouse embryonic fibroblasts (MEFs), progenitor cells and tissues from Sirt2 wild-type and genomic knockout mice, and human tumor and primary cell lines as models. The presence of Sirt2 in cells and tissues significantly enhanced the cell's sensitivity to radiation-induced cytotoxicity by delaying the dispersion of radiation-induced γ-H2AX and 53BP1 foci. This enhanced cellular radiosensitivity correlated with reduced expression of pro-survival and DNA repair proteins, and decreased DNA repair capacities involving both homologous repair and non-homologous end joining DNA repair mechanisms compared to those in Sirt2 knockout (KO) and knockdown (KD) phenotypes. Together, these data suggest SIRT2 plays a critical role in mediating the radiation-induced DNA damage response, thus regulating radiation-induced cell death and survival.
- Published
- 2019
6. Radiation-Induced Alteration of the Brain Proteome: Understanding the Role of the Sirtuin 2 Deacetylase in a Murine Model
- Author
-
Uma Shankavaram, Sudhanshu Shukla, Bruce A. Stanley, Phuongmai Nguyen, and DeeDee Smart
- Subjects
Proteome ,Synaptosomal-Associated Protein 25 ,brain ,Central nervous system ,tau Proteins ,Bioinformatics ,SIRT2 ,Biochemistry ,Article ,Myelin oligodendrocyte glycoprotein ,Mice ,Sirtuin 2 ,neurotoxicity ,medicine ,Animals ,Humans ,Dynamin I ,Brain Chemistry ,Mice, Knockout ,biology ,Gene Expression Profiling ,Neurodegeneration ,neurodegeneration ,Neurotoxicity ,Molecular Sequence Annotation ,Neurodegenerative Diseases ,General Chemistry ,medicine.disease ,radiation ,Disease Models, Animal ,medicine.anatomical_structure ,iTRAQ ,Gene Expression Regulation ,Gamma Rays ,Sirtuin ,Cancer research ,biology.protein ,Myelin-Oligodendrocyte Glycoprotein ,Metabolic Networks and Pathways ,Isobaric tag for relative and absolute quantitation - Abstract
Whole brain radiotherapy (WBRT) produces unwanted sequelae, albeit via unknown mechanisms. A deacetylase expressed in the central nervous system, Sirtuin 2 (SIRT2), has been linked to neurodegeneration. Therefore, we sought to challenge the notion that a single disease pathway is responsible for radiation-induced brain injury in Sirt2 wild-type (WT) and knockout (KO) mice at the proteomic level. We utilized isobaric tag for relative and absolute quantitation to analyze brain homogenates from Sirt2 WT and KO mice with and without WBRT. Selected proteins were independently verified, followed by ingenuity pathway analysis. Canonical pathways for Huntington's, Parkinson's, and Alzheimer's were acutely affected by radiation within 72 h of treatment. Although loss of Sirt2 preferentially affected both Huntington's and Parkinson's pathways, WBRT most significantly affected Huntington's-related proteins in the absence of Sirt2. Identical protein expression patterns were identified in Mog following WBRT in both Sirt2 WT and KO mice, revealing a proteomic radiation signature; however, long-term radiation effects were found to be associated with altered levels of a small number of key neurodegeneration-related proteins, identified as Mapt, Mog, Snap25, and Dnm1. Together, these data demonstrate the principle that the presence of Sirt2 can have significant effects on the brain proteome and its response to ionizing radiation.
- Published
- 2015
7. SIRT3 Is a Mitochondria-Localized Tumor Suppressor Required for Maintenance of Mitochondrial Integrity and Metabolism during Stress
- Author
-
Douglas R. Spitz, Nukhet Aykin-Burns, Hyun-Seok Kim, Chu-Xia Deng, Kheem S. Bisht, Athanassios Vassilopoulos, Kristi Muldoon-Jacobs, Krish Patel, Kjerstin M. Owens, Sarki A. Abdulkadir, Phuongmai Nguyen, Keshav K. Singh, David Gius, Seong Hoon Park, Özkan Özden, Jason E. Savage, Riet van der Meer, and J. Daniel Pennington
- Subjects
Genome instability ,Aging ,Cancer Research ,SIRT3 ,PROTEINS ,Breast Neoplasms ,CELLCYCLE ,Mitochondrion ,Biology ,Superoxide dismutase ,Mice ,chemistry.chemical_compound ,Stress, Physiological ,Superoxides ,Sirtuin 3 ,Animals ,Humans ,Genes, Tumor Suppressor ,Mice, Knockout ,Oncogene ,Superoxide ,Cell Biology ,Fibroblasts ,Cell cycle ,Immunohistochemistry ,Molecular biology ,Mitochondria ,Cell biology ,Oxidative Stress ,Cell Transformation, Neoplastic ,Oncology ,chemistry ,Sirtuin ,biology.protein ,Female - Abstract
The sirtuin gene family (SIRT) is hypothesized to regulate the aging process and play a role in cellular repair. This work demonstrates that SIRT3(-/-) mouse embryonic fibroblasts (MEFs) exhibit abnormal mitochondrial physiology as well as increases in stress-induced superoxide levels and genomic instability. Expression of a single oncogene (Myc or Ras) in SIRT3(-/-) MEFs results in in vitro transformation and altered intracellular metabolism. Superoxide dismutase prevents transformation by a single oncogene in SIRT3(-/-) MEFs and reverses the tumor-permissive phenotype as well as stress-induced genomic instability. In addition, SIRT3(-/-) mice develop ER/PR-positive mammary tumors. Finally, human breast and other human cancer specimens exhibit reduced SIRT3 levels. These results identify SIRT3 as a genomically expressed, mitochondria-localized tumor suppressor.
- Published
- 2010
8. Inhibition of Tumor Cell Motility by the Interferon-inducible GTPase MxA
- Author
-
W. Marston Linehan, J. Frederic Mushinski, Chand Khanna, Lisa M. Stevens, Sunmin Lee, Michel Andre Horisberger, Jane B. Trepel, Min-Jung Lee, Eun Joo Chung, Yeong Sang Kim, and Phuongmai Nguyen
- Subjects
Myxovirus Resistance Proteins ,Time Factors ,Motility ,Biology ,Microtubules ,Biochemistry ,TMPRSS2 ,Metastasis ,Small Molecule Libraries ,Mice ,Molecular Basis of Cell and Developmental Biology ,Cell Movement ,GTP-Binding Proteins ,Tubulin ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Molecular Biology ,Cell Proliferation ,Regulation of gene expression ,Differential display ,Genome, Human ,Cell growth ,Melanoma ,Liver Neoplasms ,Interferon-alpha ,Cell Biology ,medicine.disease ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Amino Acid Substitution ,Cell culture ,Mutation ,Cancer research ,Mutant Proteins ,Drug Screening Assays, Antitumor ,Protein Binding - Abstract
To identify pathways controlling prostate cancer metastasis we performed differential display analysis of the human prostate carcinoma cell line PC-3 and its highly metastatic derivative PC-3M. This revealed that a 78-kDa interferon-inducible GTPase, MxA, was expressed in PC-3 but not in PC-3M cells. The gene encoding MxA, MX1, is located in the region of chromosome 21 deleted as a consequence of fusion of TMPRSS2 and ERG, which has been associated with aggressive, invasive prostate cancer. Stable exogenous MxA expression inhibited in vitro motility and invasiveness of PC-3M cells. In vivo exogenous MxA expression decreased the number of hepatic metastases following intrasplenic injection. Exogenous MxA also reduced motility and invasiveness of highly metastatic LOX melanoma cells. A mutation in MxA that inactivated its GTPase reversed inhibition of motility and invasion in both tumor cell lines. Co-immunoprecipitation studies demonstrated that MxA associated with tubulin, but the GTPase-inactivating mutation blocked this association. Because MxA is a highly inducible gene, an MxA-targeted drug discovery screen was initiated by placing the MxA promoter upstream of a luciferase reporter. Examination of the NCI diversity set of small molecules revealed three hits that activated the promoter. In PC-3M cells, these drugs induced MxA protein and inhibited motility. These data demonstrate that MxA inhibits tumor cell motility and invasion, and that MxA expression can be induced by small molecules, potentially offering a new approach to the prevention and treatment of metastasis.
- Published
- 2009
9. BAT3 and SET1A Form a Complex with CTCFL/BORIS To Modulate H3K4 Histone Dimethylation and Gene Expression
- Author
-
Gil Bar-Sela, Hengmi Cui, David Gius, Lunching Sun, Kheem S. Bisht, Phuongmai Nguyen, Elise C. Kohn, and Andrew P. Feinberg
- Subjects
RNA, Untranslated ,Methylation ,Histones ,Proto-Oncogene Proteins c-myc ,RNA interference ,Chlorocebus aethiops ,Gene expression ,Animals ,Humans ,RNA, Small Interfering ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Transcription factor ,Regulation of gene expression ,Gene knockdown ,biology ,BRCA1 Protein ,Lysine ,Nuclear Proteins ,Proteins ,Histone-Lysine N-Methyltransferase ,Articles ,Cell Biology ,HCT116 Cells ,Molecular biology ,Chromatin ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Histone ,COS Cells ,biology.protein ,RNA, Long Noncoding ,Molecular Chaperones ,Protein Binding ,Transcription Factors - Abstract
Chromatin status is characterized in part by covalent posttranslational modifications of histones that regulate chromatin dynamics and direct gene expression. BORIS (brother of the regulator of imprinted sites) is an insulator DNA-binding protein that is thought to play a role in chromatin organization and gene expression. BORIS is a cancer-germ line gene; these are genes normally present in male germ cells (testis) that are also expressed in cancer cell lines as well as primary tumors. This work identifies SET1A, an H3K4 methyltransferase, and BAT3, a cochaperone recruiter, as binding partners for BORIS, and these proteins bind to the upstream promoter regions of two well-characterized procarcinogenic genes, Myc and BRCA1. RNA interference (RNAi) knockdown of BAT3, as well as SET1A, decreased Myc and BRCA1 gene expression but did not affect the binding properties of BORIS, but RNAi knockdown of BORIS prevented the assembly of BAT3 and SET1A at the Myc and BRCA1 promoters. Finally, chromatin analysis suggested that BORIS and BAT3 exert their effects on gene expression by recruiting proteins such as SET1A that are linked to changes in H3K4 dimethylation. Thus, we propose that BORIS acts as a platform upon which BAT3 and SET1A assemble and exert effects upon chromatin structure and gene expression.
- Published
- 2008
10. DNA Methyltransferase 1 and 3B Activate BAG-1 Expression via Recruitment of CTCFL/BORIS and Modulation of Promoter Histone Methylation
- Author
-
David Gius, Jane B. Trepel, Lunching Sun, Andrew P. Feinberg, Phuongmai Nguyen, Lei Huang, Kheem S. Bisht, Wenqiang Yu, Sunmin Lee, C. Matthew Bradbury, Allen S. Ho, Gil Bar-Sela, and Hengmi Cui
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Cancer Research ,Immunoblotting ,Transfection ,Polymerase Chain Reaction ,Article ,Small hairpin RNA ,Genes, Reporter ,Cell Line, Tumor ,Histone methylation ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,Protein Methyltransferases ,RNA, Neoplasm ,Epigenetics ,Promoter Regions, Genetic ,DNA Primers ,biology ,Histone-Lysine N-Methyltransferase ,Molecular biology ,Chromatin ,DNA-Binding Proteins ,Histone ,Oncology ,CTCF ,Histone methyltransferase ,Colonic Neoplasms ,embryonic structures ,Histone Methyltransferases ,biology.protein ,Chromatin immunoprecipitation ,Plasmids ,Transcription Factors - Abstract
In a previous genomic analysis, using somatic methyltransferase (DNMT) knockout cells, we showed that hypomethylation decreased the expression of as many genes as were observed to increase, suggesting a previously unknown mechanism for epigenetic regulation. To address this idea, the expression of the BAG family genes was used as a model. These genes were used because their expression was decreased in DNMT1−/−, DNMT3B−/−, and double knockout cells and increased in DNMT1-overexpressing and DNMT3B-overexpressing cells. Chromatin immunoprecipitation analysis of the BAG-1 promoter in DNMT1-overexpressing or DNMT3B-overexpressing cells showed a permissive dimethyl-H3-K4/dimethyl-H3-K9 chromatin status associated with DNA-binding of CTCFL/BORIS, as well as increased BAG-1 expression. In contrast, a nonpermissive dimethyl-H3-K4/dimethyl-H3-K9 chromatin status was associated with CTCF DNA-binding and decreased BAG-1 expression in the single and double DNMT knockout cells. BORIS short hairpin RNA knockdown decreased both promoter DNA-binding, as well as BAG-1 expression, and changed the dimethyl-H3-K4/dimethyl-H3-K9 ratio to that characteristic of a nonpermissive chromatin state. These results suggest that DNMT1 and DNMT3B regulate BAG-1 expression via insulator protein DNA-binding and chromatin dynamics by regulating histone dimethylation. [Cancer Res 2008;68(8):2726–35]
- Published
- 2008
11. SIRT3 interacts with the daf-16 homolog FOXO3a in the Mitochondria, as well as increases FOXO3a Dependent Gene expression
- Author
-
Jaime Leclerc, Mark Mishra, Lunching Sun, Nukhet Aykin-Burns, Kheem S. Bisht, Phuongmai Nguyen, J. Daniel Pennington, Douglas R. Spitz, David Gius, Hyun-Seok Kim, Bong Hyun Ahn, Chu-Xia Deng, and Kristi Muldoon Jacobs
- Subjects
Chromatin Immunoprecipitation ,SIRT3 ,Mutant ,Gene Expression ,Mitochondrion ,Transfection ,Applied Microbiology and Biotechnology ,Mitochondrial Proteins ,Superoxides ,Sirtuin 3 ,Chlorocebus aethiops ,Gene expression ,Daf-16 ,Animals ,Humans ,Sirtuins ,Gene family ,FOXO3a ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,HSPA9 ,Glutathione Disulfide ,biology ,Superoxide Dismutase ,Forkhead Box Protein O3 ,Superoxide ,Forkhead Transcription Factors ,Cell Biology ,HCT116 Cells ,Mitochondria ,Biochemistry ,COS Cells ,Sirtuin ,biology.protein ,Protein Binding ,Research Paper ,Developmental Biology - Abstract
Cellular longevity is a complex process relevant to age-related diseases including but not limited to chronic illness such as diabetes and metabolic syndromes. Two gene families have been shown to play a role in the genetic regulation of longevity; the Sirtuin and FOXO families. It is also established that nuclear Sirtuins interact with and under specific cellular conditions regulate the activity of FOXO gene family proteins. Thus, we hypothesize that a mitochondrial Sirtuin (SIRT3) might also interact with and regulate the activity of the FOXO proteins. To address this we used HCT116 cells overexpressing either wild-type or a catalytically inactive dominant negative SIRT3. For the first time we establish that FOXO3a is also a mitochondrial protein and forms a physical interaction with SIRT3 in mitochondria. Overexpression of a wild-type SIRT3 gene increase FOXO3a DNA-binding activity as well as FOXO3a dependent gene expression. Biochemical analysis of HCT116 cells over expressing the deacetylation mutant, as compared to wild-type SIRT3 gene, demonstrated an overall oxidized intracellular environment, as monitored by increase in intracellular superoxide and oxidized glutathione levels. As such, we propose that SIRT3 and FOXO3a comprise a potential mitochondrial signaling cascade response pathway.
- Published
- 2008
12. Distinct Effects of Ionizing Radiation on In vivo Murine Kidney and Brain Normal Tissue Gene Expression
- Author
-
J. Daniel Pennington, Kheem S. Bisht, Weiling Zhao, Tony J. C. Wang, Gil Bar-Sela, C. Matthew Bradbury, Lei Huang, David Gius, Michael E. Robbins, Phuongmai Nguyen, Mark Mishra, Rania T. Awwad, Eric Y. Chuang, Lunching Sun, and Zhijun Chen
- Subjects
Integrins ,Protein Folding ,Cancer Research ,Candidate gene ,Cell type ,Pathology ,medicine.medical_specialty ,Programmed cell death ,Central nervous system ,Biology ,Kidney ,Mice ,Radiation, Ionizing ,Gene expression ,medicine ,Animals ,Lung ,Gene ,Microarray analysis techniques ,Cell Cycle ,Brain ,Cell biology ,Protein Transport ,Metabolism ,medicine.anatomical_structure ,Gene Expression Regulation ,Oncology - Abstract
Purpose: There is a growing awareness that radiation-induced normal tissue injury in late-responding organs, such as the brain, kidney, and lung, involves complex and dynamic responses between multiple cell types that not only lead to targeted cell death but also acute and chronic alterations in cell function. The specific genes involved in the acute and chronic responses of these late-responding normal tissues remain ill defined; understanding these changes is critical to understanding the mechanism of organ damage. As such, the aim of the present study was to identify candidate genes involved in the development of radiation injury in the murine kidney and brain using microarray analysis. Experimental Design: A multimodality experimental approach combined with a comprehensive expression analysis was done to determine changes in normal murine tissue gene expression at 8 and 24 hours after irradiation. Results: A comparison of the gene expression patterns in normal mouse kidney and brain was strikingly different. This observation was surprising because it has been long assumed that the changes in irradiation-induced gene expression in normal tissues are preprogrammed genetic changes that are not affected by tissue-specific origin. Conclusions: This study shows the potential of microarray analysis to identify gene expression changes in irradiated normal tissue cells and suggests how normal cells respond to the damaging effects of ionizing radiation is complex and markedly different in cells of differing origin.
- Published
- 2006
13. Thioredoxin reductase as a novel molecular target for cancer therapy
- Author
-
Douglas R. Spitz, David Gius, Dee Dee K. Smart, Phuongmai Nguyen, and Rania T. Awwad
- Subjects
Thioredoxin Reductase 1 ,Cancer Research ,Thioredoxin-Disulfide Reductase ,Free Radicals ,Cell Survival ,Thioredoxin reductase ,Mutant ,Cell ,Antineoplastic Agents ,Biology ,Thioredoxins ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Gene ,Cell Proliferation ,Cell cycle ,Cell biology ,Oxidative Stress ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,Apoptosis ,Cell culture ,Thioredoxin ,Signal Transduction - Abstract
Tumor cell proliferation, de-differentiation, and progression depend on a complex combination of altered cell cycle regulation, excessive growth factor pathway activation, and decreased apoptosis. The understanding of these complex mechanisms should lead to the identification of potential targets for therapeutic intervention. Redox-sensitive signaling factors also regulate multiple cellular processes including proliferation, cell cycle, and pro-survival signaling cascades, suggesting their potential as molecular targets for anticancer agents. These observations suggest that redox-sensitive signaling factors may be potential novel molecular markers. We hypothesized that thioredoxin reductase-1 (TR), a component of several redox-regulated pathways, may represent a potential molecular target candidate in response to agents that induce oxidative stress. There have been numerous biological studies over the last decade investigating the cell biological, biochemical, and genetic properties of TR both in culture and in in vivo models. In addition, using a series of permanent cell lines that express either a wild-type TR or a dominant mutant TR gene or a chemical agent that inhibits TR we demonstrated that TR meets most criteria that would identify a molecular target. Based on these results we believe TR is a potential molecular target and discuss potential clinical possibilities.
- Published
- 2006
14. Chromosomal protein HMGN1 modulates the expression of N-cadherin
- Author
-
Michael Bustin, Yaffa R. Rubinstein, Yehudit Birger, Katherine L. West, Phuongmai Nguyen, Sunmin Lee, Jane B. Trepel, Takashi Furusawa, Yuri V. Postnikov, and Jae-Hwan Lim
- Subjects
HMGN1 ,medicine.diagnostic_test ,Gene targeting ,Cell Biology ,Biology ,Hmg protein ,Biochemistry ,Molecular biology ,Chromatin ,Cell biology ,Western blot ,Gene expression ,medicine ,biology.protein ,Nuclear protein ,Molecular Biology ,Gene - Abstract
HMGN1 is a nuclear protein that binds to nucleosomes and alters the accessibility of regulatory factors to their chromatin targets. To elucidate its biological function and identify specific HMGN1 target genes, we generated Hmgn1–/– mice. DNA microarray analysis of Hmgn1+/+ and Hmgn1–/– embryonic fibroblasts identified N-cadherin as a potential HMGN1 gene target. RT-PCR and western blot analysis confirmed a linkage between HMGN1 expression and N-cadherin levels. In both transformed and primary mouse embryonic fibroblasts (MEFs), HMGN1 acted as negative regulator of N-cadherin expression. Likewise, the N-cadherin levels in early embryos of Hmgn1–/– mice were higher than those of their Hmgn1+/+ littermates. Loss of HMGN1 increased the adhesiveness, motility and aggregation potential of Hmgn1–/– MEFs, a phenotype consistent with increased levels of N-cadherin protein. Re-expression of wild-type HMGN1, but not of the mutant HMGN1 protein that does not bind to chromatin, in Hmgn1–/– MEFs, decreased the levels of N-cadherin and restored the Hmgn1+/+ phenotype. These studies demonstrate a role for HMGN1 in the regulation of specific gene expression. We suggest that in MEFs, and during early mouse development, the interaction of HMGN1 with chromatin down-regulates the expression of N-cadherin.
- Published
- 2005
15. The epigenome as a molecular marker and target
- Author
-
Dee Dee K. Smart, Lunching Sun, Kheem S. Bisht, Phuongmai Nguyen, Allen S. Ho, Rania T. Awwad, David Gius, C. Matthew Bradbury, and Lei Huang
- Subjects
Gene expression profiling ,Regulation of gene expression ,Genetics ,Cancer Research ,Oncology ,Mechanism (biology) ,DNA methylation ,Gene expression ,Methylation ,Epigenome ,Biology ,Gene ,Cell biology - Abstract
Tumor cell proliferation, de-differentiation, and progression depend on a complex combination of altered cell cycle regulation, excessive growth factor pathway activation, and decreased apoptosis. The understanding of these complex mechanisms should lead to the identification of potential molecular markers, targets, and molecular profiles that should eventually expand and improve therapeutic intervention. It now appears clear that methylation plays a central role in transformation, both in vitro and in vivo. However, the exact targets and mechanism(s) are not yet fully understood. This is partly due to the significant number of genes altered by changes in intracellular methyltransferase activity and the chemical agents used to modulate gene expression. The complex nature of methylation's role in regulating gene expression suggests that in addition to investigating individual genes, researchers should develop more comprehensive methods to examine gene expression patterns and their predictive value as this will likely be necessary in the future. If methylation plays a role in transformation, then it seems logical that genes regulating intracellular methylation status may be used as molecular markers to profile tumors by any new methods currently being developed. Perhaps more noteworthy is that DNMT genes may be found to be novel molecular targets for new factor-specific anticancer agents. This idea will be addressed.
- Published
- 2005
16. Abrogation of p21 Expression by Flavopiridol Enhances Depsipeptide-Mediated Apoptosis in Malignant Pleural Mesothelioma Cells
- Author
-
Dao M. Nguyen, Wilson S. Tsai, Jane B. Trepel, William D. Schrump, David S. Schrump, Phuongmai Nguyen, and G. Aaron Chen
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Mesothelioma ,Cancer Research ,Cell Survival ,Pleural Neoplasms ,Apoptosis ,Biology ,Protein kinase C signaling ,chemistry.chemical_compound ,Piperidines ,Cell Line, Tumor ,Cyclins ,Humans ,Cytotoxic T cell ,Calphostin ,Enzyme Inhibitors ,Flavonoids ,Depsipeptide ,Molecular biology ,Calphostin C ,Oncology ,chemistry ,Cell culture ,Cancer research ,Growth inhibition ,Cell Division - Abstract
Purpose: Recent insights regarding the pathogenesis of malignant pleural mesothelioma (MPM) provide new opportunities for targeted molecular therapies for this highly lethal disease. The present study was undertaken to examine the effects of the histone deacetylase inhibitor, Depsipeptide (DP) FK228, in conjunction with the cyclin-dependent kinase inhibitor, Flavopiridol (FLA), in cultured MPM cells. Experimental Design: Proliferation and apoptosis in drug-treated, virally transduced, or control cells were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and Apo-bromodeoxyuridine techniques. Western blot and ELISA techniques were used to examine signal transduction and cell cycle-related protein levels in MPM cells exposed to DP and/or FLA in the presence or absence of calphostin, phorbol-12,13-dibutyrate, 5,6-dichloro-1-β-d-ribofuranosyl-benzimidazole, or adenoviral p21 transduction. Results: DP (1–50 ng/ml × 6 h) or FLA (100–200 nm × 72 h) alone, mediated low-level, dose-dependent growth inhibition in MPM cells. In contrast, sequential DP/FLA treatment mediated marked growth inhibition and apoptosis in these cell lines. The cytotoxic effects of DP/FLA were considerably less pronounced in cultured normal cells. The proapoptotic effects of DP/FLA treatment coincided with inhibition of DP-mediated induction of p21 by FLA. Overexpression of p21 by adenoviral gene transfer techniques rendered MPM cells refractory to the cytotoxic effects of this treatment regimen. In p21 reporter assays, promoter activation by DP was antagonized by FLA. The magnitude of inhibition of DP-mediated p21 induction by FLA exceeded that observed with the pTEFb antagonist 5,6-dichloro-1-β-d-ribofuranosyl-benzimidazole. Calphostin C abrogated p21 induction mediated by DP and enhanced DP-mediated apoptosis in a manner comparable with FLA in MPM cells; in contrast, phorbol-12,13-dibutyrate blocked FLA-mediated inhibition of p21 induction by DP and markedly protected these cells from the apoptotic effects of sequential DP/FLA. Conclusions: FLA abrogates DP-mediated induction of p21 expression, in part, via inhibition of protein kinase C signaling and markedly potentiates the cytotoxic effects of DP in MPM cells.
- Published
- 2004
17. Regulation of leukemic cell adhesion, proliferation, and survival by β-catenin
- Author
-
Pierre A. Henkart, Jeffrey S. Rubin, Jin Woo Kim, Jung Sik Kim, Donald P. Bottaro, Eun Joo Chung, Jane B. Trepel, Lilian Soon, Ho Jung Oh, Sunmin Lee, Judith E. Karp, Sang Gu Hwang, Su Jae Lee, Phuongmai Nguyen, and P. Bonvini
- Subjects
Transcriptional Activation ,Cell Survival ,Immunology ,Apoptosis ,Biology ,Lymphocyte Activation ,Biochemistry ,Jurkat cells ,Cell–cell interaction ,Enhancer binding ,Cell Adhesion ,Tumor Cells, Cultured ,Humans ,Cell adhesion ,Cytoskeleton ,beta Catenin ,Leukemia ,Myeloid leukemia ,Cell Biology ,Hematology ,Cell biology ,Cytoskeletal Proteins ,Haematopoiesis ,Catenin ,Trans-Activators ,Cell Division ,Signal Transduction ,K562 cells - Abstract
In epithelial cells β-catenin plays a critical role as a component of the cell-cell adhesion apparatus and as a coactivator of the TCF/LEF (T-cell transcription factor/lymphoid enhancer binding factor) family of transcription factors. Deregulation of β-catenin has been implicated in the malignant transformation of cells of epithelial origin. However, a function for β-catenin in hematologic malignancies has not been reported. β-Catenin is not detectable in normal peripheral blood T cells but is expressed in T–acute lymphoblastic leukemia cells and other tumor lines of hematopoietic origin and in primary lymphoid and myeloid leukemia cells. β-Catenin function was examined in Jurkat T–acute lymphoblastic leukemia cells. Overexpression of dominant-negative β-catenin or dominant-negative TCF reduced β-catenin nuclear signaling and inhibited Jurkat proliferation and clonogenicity. Similarly, these constructs inhibited proliferation of K562 and HUT-102 cells. Reduction of β-catenin expression with β-catenin antisense down-regulated adhesion of Jurkat cells in response to phytohemagglutinin. Incubation of Jurkat cells with anti-Fas induced caspase-dependent limited proteolysis of β-catenin N- and C-terminal regions and rapid redistribution of β-catenin to the detergent-insoluble cytoskeleton, concomitant with a marked decline in nuclear β-catenin signaling. Fas-mediated apoptosis was potentiated by inhibition of β-catenin nuclear signaling. The data suggest that β-catenin can play a significant role in promoting leukemic cell proliferation, adhesion, and survival.
- Published
- 2002
18. SIRT2 Interacts with β-catenin to Inhibit Wnt Signaling Output in Response to Radiation-Induced Stress
- Author
-
Jane B. Trepel, DeeDee Smart, Sunmin Lee, Dominique Lorang-Leins, and Phuongmai Nguyen
- Subjects
Cancer Research ,Beta-catenin ,Cellular homeostasis ,SIRT2 ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins c-myc ,Mice ,Sirtuin 2 ,medicine ,Animals ,Humans ,Cyclin D1 ,Molecular Biology ,Wnt Signaling Pathway ,beta Catenin ,Cell Proliferation ,Radiation ,biology ,Wnt signaling pathway ,LRP5 ,Fibroblasts ,Cell biology ,Gene Expression Regulation, Neoplastic ,Oxidative Stress ,Oncology ,Matrix Metalloproteinase 9 ,Catenin ,Cancer research ,biology.protein ,Carcinogenesis ,Cell aging ,Protein Binding ,Signal Transduction - Abstract
Wnt signaling is critical to maintaining cellular homeostasis via regulation of cell division, mitigation of cell stress, and degradation. Aberrations in Wnt signaling contribute to carcinogenesis and metastasis, whereas sirtuins have purported roles in carcinogenesis, aging, and neurodegeneration. Therefore, the hypothesis that sirtuin 2 (SIRT2) directly interacts with β-catenin and whether this interaction alters the expression of Wnt target genes to produce an altered cellular phenotype was tested. Coimmunoprecipitation studies, using mouse embryonic fibroblasts (MEF) from Sirt2 wild-type and genomic knockout mice, demonstrate that β-catenin directly binds SIRT2. Moreover, this interaction increases in response to oxidative stress induced by ionizing radiation. In addition, this association inhibits the expression of important Wnt target genes such as survivin (BIRC5) , cyclin D1 (CCND1) , and c-myc (MYC) . In Sirt2 null MEFs, an upregulation of matrix metalloproteinase 9 (MMP9) and decreased E-cadherin (CDH1) expression is observed that produces increased cellular migration and invasion. Together, these data demonstrate that SIRT2, a tumor suppressor lost in multiple cancers, inhibits the Wnt signaling pathway in nonmalignant cells by binding to β-catenin and that SIRT2 plays a critical role in the response to oxidative stress from radiation. Implications: Disruption of the SIRT2–β-catenin interaction represents an endogenous therapeutic target to prevent transformation and preserve the integrity of aging cells against exogenous stressors such as reactive oxygen species. Mol Cancer Res; 12(9); 1244–53. ©2014 AACR . This article is featured in Highlights of This Issue, [p. 1193][1] [1]: /lookup/volpage/12/1193?iss=9
- Published
- 2014
19. Regulation of actin cytoskeleton in lymphocytes: PKC-? disrupts IL-3-induced membrane ruffles downstream of Rac1
- Author
-
Mikhail V. Blagosklonny, Peter Acs, Peter M. Blumberg, Susan H. Garfield, Jane B. Trepel, Richard P. Nordan, I. A. Alexandrov, J. Frederic Mushinski, Phuongmai Nguyen, and Larisa Y. Romanova
- Subjects
RHOA ,biology ,Physiology ,Clinical Biochemistry ,Cell ,RAC1 ,Cell Biology ,CDC42 ,Actin cytoskeleton ,Cell biology ,medicine.anatomical_structure ,Cell culture ,medicine ,biology.protein ,Actin ,Protein kinase C - Abstract
In the murine pre-B lymphoid cell line Baf3, the presence of IL-3 is required for the formation of membrane ruffles that intensely stain for actin and are responsible for the elongated cell phenotype. Withdrawal of IL-3 dissolves ruffled protrusions and converts the cell phenotype to round. Flow cytometric analysis of the cell shape showed that an inactive analog of Rac1 but not inactive RhoA or inactive cdc42 rounds the cells in the presence of IL-3. Constitutively activated Rac1 restores the elongated cell phenotype to IL-3-starved cells. We conclude that the activity of Rac1 is necessary and sufficient for the IL-3-induced assembly of membrane ruffles. Similar to the IL-3 withdrawal, phorbol 12-myristate 13-acetate (PMA) dissolves actin-formed membrane ruffles and rounds the cells in the presence of IL-3. Flow cytometric analysis of the cell shape demonstrated that in the presence of IL-3 the PMA-induced cell rounding cannot be abolished by constitutively active Rac1 but can be imitated by inactive Rac1. These data indicate that PMA disrupts the IL-3 pathway downstream of Rac1. Cells rounded by PMA return to the elongated phenotype concomitantly with PKC depletion. PMA-induced cell rounding can be reversed by the PKC-specific inhibitor GF109203X. Experiments with overexpression in Baf3 of individual PKC isoforms and a dominant negative PKC-delta indicate that activation of PKC-delta but not other PKC isoforms is responsible for disruption of membrane ruffles.
- Published
- 1999
20. The Benzoquinone Ansamycin Geldanamycin Stimulates Proteolytic Degradation of Focal Adhesion Kinase
- Author
-
Len Neckers, Theodor W. Schulte, Phuongmai Nguyen, Hans-Joachim Ochel, and Jane B. Trepel
- Subjects
Proteasome Endopeptidase Complex ,Lactams, Macrocyclic ,Endocrinology, Diabetes and Metabolism ,PTK2 ,Biology ,Biochemistry ,Focal adhesion ,Mice ,chemistry.chemical_compound ,Endocrinology ,Multienzyme Complexes ,Enzyme Stability ,Benzoquinones ,Tumor Cells, Cultured ,Genetics ,Animals ,RNA, Messenger ,Enzyme Inhibitors ,Molecular Biology ,PTK2B ,Hydrolysis ,Ansamycin ,Quinones ,3T3 Cells ,Protein-Tyrosine Kinases ,Geldanamycin ,Hsp90 ,Cysteine Endopeptidases ,Rifabutin ,chemistry ,Focal Adhesion Kinase 1 ,Focal Adhesion Protein-Tyrosine Kinases ,Cancer research ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Cell Adhesion Molecules ,Tyrosine kinase - Abstract
FAK is a nonreceptor tyrosine kinase involved in adhesion-mediated signal transduction whose level of expression is related to the invasiveness of malignant tumors. In seeking strategies to downregulate FAK, we treated various cell lines in vitro with the benzoquinone ansamycin geldanamycin (GA) which was previously described as a tyrosine kinase inhibitor, but recently has been shown to exert its effects by interfering with the chaperone function of members of the hsp90 family of heat-shock proteins. We evaluated the effects of benzoquinone ansamycins on FAK steady-state protein level and FAK half-life in breast and prostate carcinoma, Ewing's sarcoma, and 3T3 fibroblasts. Our data demonstrate that GA stimulates the proteolytic degradation of FAK in all cell lines examined and markedly reduces the half-life of newly synthesized FAK protein without significantly altering the level of FAK mRNA. These data demonstrate FAK to be another tyrosine kinase sensitive to the destabilizing effects of benzoquinone ansamycins and further show that small molecule-mediated pharmacologic modulation of FAK protein level is a feasible approach to the interdiction of FAK function.
- Published
- 1999
21. Inhibition of the 3-Hydroxy-3-methylglutaryl-coenzyme A Reductase Pathway Induces p53-independent Transcriptional Regulation of p21 in Human Prostate Carcinoma Cells
- Author
-
Seong-Jin Kim, Farzaneh Pirnia, Jane B. Trepel, Xiao-Fan Wang, Yung Hyun Choi, Won-Ki Kang, Mahn Joon Ha, Jeen Lee, Phuongmai Nguyen, and Su Jae Lee
- Subjects
biology ,Kinase ,Cyclin-dependent kinase 2 ,Retinoblastoma protein ,E2F1 Transcription Factor ,Cell Biology ,Cell cycle ,Biochemistry ,Molecular biology ,biology.protein ,Transcriptional regulation ,Molecular Biology ,E2F Transcription Factors ,Transcription factor - Abstract
Progression through the cell cycle is controlled by the induction of cyclins and the activation of cognate cyclin-dependent kinases. The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor lovastatin induces growth arrest and cell death in certain cancer cell types. We have pursued the mechanism of growth arrest in PC-3-M cells, a p53-null human prostate carcinoma cell line. Lovastatin treatment increased protein and mRNA levels of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1), increased binding of p21 with Cdk2, markedly inhibited cyclin E- and Cdk2-associated phosphorylation of histone H1 or GST-retinoblastoma protein, enhanced binding of the retinoblastoma protein to the transcription factor E2F-1 in vivo, and induced the activation of a p21 promoter reporter construct. By using p21 promoter deletion constructs, the lovastatin-responsive element was mapped to a region between -93 and -64 relative to the transcription start site. Promoter mutation analysis indicated that the lovastatin-responsive site coincided with the previously identified transforming growth factor-beta-responsive element. These data indicate that in human prostate carcinoma cells an inhibitor of the HMG-CoA reductase pathway can circumvent the loss of wild-type p53 function and induce critical downstream regulatory events leading to transcriptional activation of p21.
- Published
- 1998
22. In vivo degradation of N-myc in neuroblastoma cells is mediated by the 26S proteasome
- Author
-
P. Bonvini, Leonard M. Neckers, Jane B. Trepel, and Phuongmai Nguyen
- Subjects
Proteasome Endopeptidase Complex ,Cancer Research ,Proteolysis ,Lactacystin ,Protein polyubiquitination ,Cysteine Proteinase Inhibitors ,Biology ,Substrate Specificity ,Proto-Oncogene Proteins c-myc ,Neuroblastoma ,chemistry.chemical_compound ,Ubiquitin ,In vivo ,Tumor Cells, Cultured ,Genetics ,medicine ,Humans ,Ubiquitins ,Molecular Biology ,medicine.diagnostic_test ,Calpain ,Molecular biology ,Acetylcysteine ,chemistry ,Proteasome ,biology.protein ,Lysosomes ,N-Myc ,Peptide Hydrolases - Abstract
N-myc is a short-lived transcription factor, frequently amplified in human neuroblastomas. The ubiquitin-proteasome system is involved in the degradation of many short-lived cellular proteins and previous studies have shown that ubiquitin-dependent proteolysis is implicated in the turn-over of N-myc in vitro. However, calpain has also been implicated in N-myc degradation in vitro. Here we report that, in vivo, N-myc is a sensitive substrate for the 26S proteasome in N-myc amplified neuroblastoma cells. We observed that inhibition of the 26S proteasome with two inhibitors, ALLnL and lactacystin, led to an elevation of the N-myc protein steady-state and increased N-myc protein polyubiquitination, as revealed by ubiquitin Western blotting. Pulse-chase experiments have shown that the increased N-myc levels resulted from stabilization of the protein. In contrast treatment with several calpain and cathepsin inhibitors failed to block N-myc degradation in vivo. Furthermore, fluorescence microscopy of ALLnL-treated cells localized N-myc exclusively to the nuclear compartment, suggesting the absence of a requirement for transport to the cytoplasm prior to degradation.
- Published
- 1998
23. Regulation of Cyclin D1 by Calpain Protease
- Author
-
Yung Hyun Choi, Ming-Lei Wu, Phuongmai Nguyen, Jeen Lee, Joung Soon Jang, Su Jae Lee, Emiko Takano, Jane B. Trepel, Pierre A. Henkart, and Masatoshi Maki
- Subjects
Male ,Cyclin D ,Lactacystin ,Cyclin A ,Cyclin B ,Cysteine Proteinase Inhibitors ,Biochemistry ,Mice ,chemistry.chemical_compound ,Cyclin D1 ,Tumor Cells, Cultured ,Animals ,Humans ,Cyclin B1 ,Molecular Biology ,Cyclin ,Protein Synthesis Inhibitors ,biology ,Calpain ,Prostatic Neoplasms ,3T3 Cells ,Cell Biology ,Acetylcysteine ,chemistry ,Dactinomycin ,biology.protein ,Cancer research ,Cyclin A2 ,Half-Life - Abstract
Cyclin D1, a critical positive regulator of G1 progression, has been implicated in the pathogenesis of certain cancers. Regulation of cyclin D1 occurs at the transcriptional and posttranscriptional level. Here we present evidence that cyclin D1 levels are regulated at the posttranscriptional level by the Ca2+-activated protease calpain. Serum starvation of NIH 3T3 cells resulted in rapid loss of cyclin D1 protein that was completely reversible by calpain inhibitors. Actinomycin D and lovastatin induced rapid loss of cyclin D1 in prostate and breast cancer cells that was reversible by calpain inhibitors and not by phenylmethylsulfonyl fluoride, caspase inhibitors, or lactacystin, a specific inhibitor of the 26 S proteasome. Treatment of intact NIH 3T3, prostate, and breast cancer cells with a calpain inhibitor dramatically increased the half-life of cyclin D1 protein. Addition of purified calpain to PC-3-M lysates resulted in Ca2+-dependent cyclin D1 degradation. Transient expression of the calpain inhibitor calpastatin increased cyclin D1 protein in serum-starved NIH 3T3 cells. Cyclins A, E, and B1 have been reported to be regulated by proteasome-associated proteolysis. The data presented here implicate calpain in cyclin D1 posttranslational regulation.
- Published
- 1997
24. Destabilization of Raf-1 by Geldanamycin Leads to Disruption of the Raf-1–MEK–Mitogen-Activated Protein Kinase Signalling Pathway
- Author
-
Mikhail V. Blagosklonny, Leonard M. Neckers, J F Johnston, J F Mushinski, B P Monia, Phuongmai Nguyen, Jane B. Trepel, L Romanova, and T W Schulte
- Subjects
Transcriptional Activation ,MAPK/ERK pathway ,Lactams, Macrocyclic ,MAP Kinase Kinase Kinase 1 ,Protein Serine-Threonine Kinases ,Biology ,Mitogen-activated protein kinase kinase ,Transfection ,MAP2K7 ,Jurkat Cells ,Mice ,Genes, Reporter ,Proto-Oncogene Proteins ,Benzoquinones ,Animals ,Humans ,ASK1 ,HSP90 Heat-Shock Proteins ,c-Raf ,Enzyme Inhibitors ,Molecular Biology ,Protein Kinase C ,Protein kinase C ,MAP kinase kinase kinase ,Quinones ,3T3 Cells ,DNA ,Cell Biology ,Oligonucleotides, Antisense ,Protein-Tyrosine Kinases ,Molecular biology ,Recombinant Proteins ,Rats ,Cell biology ,Proto-Oncogene Proteins c-raf ,Calcium-Calmodulin-Dependent Protein Kinases ,Tetradecanoylphorbol Acetate ,Cell Division ,Research Article ,Signal Transduction - Abstract
The serine/threonine kinase Raf-1 functions downstream of Rats in a signal transduction cascade which transmits mitogenic stimuli from the plasma membrane to the nucleus. Raf-1 integrates signals coming from extracellular factors and, in turn, activates its substrate, MEK kinase. MEK activates mitogen-activated protein kinase (MAPK), which phosphorylates other kinases as well as transcription factors. Raf-1 exists in a complex with HSP90 and other proteins. The benzoquinone ansamycin geldanamycin (GA) binds to HSP90 and disrupts the Raf-1-HSP90 multimolecular complex, leading to destabilization of Raf-1. In this study, we examined whether Raf-1 destabilization is sufficient to block the Raf-1-MEK-MAPK signalling pathway and whether GA specifically inactivates the Raf-1 component of this pathway. Using the model system of NIH 3T3 cells stimulated with phorbol 12-myristate 13-acetate (PMA), we show that GA does not affect the ability of protein kinase C alpha to be activated by phorbol esters, but it does block activation of MEK and MAPK. Further, GA does not decrease the activity of constitutively active MEK in transiently transfected cells. Finally, disruption of the Raf-1-MEK-MAPK signalling pathway by GA prevents both the PMA-induced proliferative response and PMA-induced activation of a MAPK-sensitive nuclear transcription factor. Thus, we demonstrate that interaction between HSP90 and Raf-1 is a sine qua non for Raf stability and function as a signal transducer and that the effects observed cannot be attributed to a general impairment of protein kinase function.
- Published
- 1996
25. Genistein-stimulated adherence of prostate cancer cells is associated with the binding of focal adhesion kinase to beta-l-integrin
- Author
-
Edward Kyle, Phuongmai Nguyen, Len Neckers, Raymond C. Bergan, Jane B. Trepel, and Christian Ingui
- Subjects
Cancer Research ,biology ,Kinase ,Cell ,Integrin ,Genistein ,General Medicine ,medicine.disease ,Focal adhesion ,chemistry.chemical_compound ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,chemistry ,Cell culture ,Cancer research ,medicine ,biology.protein ,Cell adhesion - Abstract
The isoflavinoid genistein is a protein-tyrosine kinase inhibitor which has been identified as a putative cancer prevention agent. Its consumption is associated with a low incidence of clinical metastatic prostate cancer in the face of a sustained high incidence of organ-confined prostate cancer. We therefore undertook studies to examine genistein's effect upon cell adhesion as one possible mechanism by which it could be acting as an antimetastatic agent. A morphogenic analysis revealed that genistein caused cell flattening in a variety of cell lines: PC3-M, PC3, and DU-145 prostate carcinoma cells, as well as MCF-7 breast carcinoma cells. Mechanistic studies focused on the highly metastatic PC3-M cell line, and revealed that cell flattening was accompanied by an increase in cell adhesion. Further investigations demonstrated that focal adhesion kinase (FAK) accumulated in areas of focal cell attachment, and that this accumulation occurred only when cells were actively undergoing genistein-mediated morphologic change. Concurrent formation of a complex between the cell attachment molecule, beta-1-integrin, and FAK was shown to occur, and to correlate with transient activation of FAK activity. Genistein is presented as a novel investigative tool for use in the study of molecular events involved in the process of cell adhesion.
- Published
- 1996
26. CTCFL/BORIS Is a Methylation-Independent DNA-Binding Protein That Preferentially Binds to the Paternal H19 Differentially Methylated Region
- Author
-
Hengmi Cui, David Gius, Lunching Sun, Mitsuo Oshimura, Richard S. Lee, Hiroyuki Kugoh, Kheem S. Bisht, Phuongmai Nguyen, Krish Patel, and Andrew P. Feinberg
- Subjects
Male ,Cancer Research ,RNA, Untranslated ,Biology ,Insulator (genetics) ,DNA-binding protein ,Methylation ,Article ,Fathers ,Mice ,Cell Line, Tumor ,Animals ,Humans ,Transgenes ,Regulation of gene expression ,Zinc finger ,DNA Methylation ,Molecular biology ,Chromatin ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Oncology ,CTCF ,DNA methylation ,Female ,RNA, Long Noncoding ,Chromatin immunoprecipitation - Abstract
The CTCF paralog BORIS (brother of the regulator of imprinted sites) is an insulator DNA-binding protein thought to play a role in chromatin organization and gene expression. Under normal physiologic conditions, BORIS is predominantly expressed during embryonic male germ cell development; however, it is also expressed in tumors and tumor cell lines and, as such, has been classified as a cancer-germline or cancer-testis gene. It has been suggested that BORIS may be a pro-proliferative factor, whereas CTCF favors antiproliferation. BORIS and CTCF share similar zinc finger DNA-binding domains and seem to bind to identical target sequences. Thus, one critical question is the mechanism governing the DNA-binding specificity of these two proteins when both are present in tumor cells. Chromatin immunoprecipitation (ChIP) in HCT116 cells and their hypermethylated variant showed that BORIS binds to methylated DNA sequences, whereas CTCF binds to unmethylated DNA. Electromobility shift assays, using both whole-cell extracts and in vitro translated CTCF and BORIS protein, and methylation-specific ChIP PCR showed that BORIS is a methylation-independent DNA-binding protein. Finally, experiments in murine hybrid cells containing either the maternal or paternal human chromosome 11 showed that BORIS preferentially binds to the methylated paternal H19 differentially methylated region, suggesting a mechanism in which the affinity of CTCF for the unmethylated maternal allele directs the DNA binding of BORIS toward the paternal allele. [Cancer Res 2008;68(14):5546–51]
- Published
- 2008
27. DNMT1 as a molecular target in a multimodality-resistant phenotype in tumor cells
- Author
-
Aradhana Kaushal, Lunching Sun, Kheem S. Bisht, Kristi Muldoon-Jacobs, David Gius, Mark Mishra, Lei Huang, Rania T. Awwad, J. Daniel Pennington, Phuongmai Nguyen, C. Matthew Bradbury, and Stephanie Markovina
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Cancer Research ,Somatic cell ,Antineoplastic Agents ,Biology ,Cell Line, Tumor ,Neoplasms ,Gene expression ,medicine ,Animals ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,Cytotoxicity ,Molecular Biology ,Etoposide ,Cisplatin ,Hydrogen Peroxide ,Molecular biology ,Phenotype ,Rats ,Oxidative Stress ,Oncology ,Cell culture ,Drug Resistance, Neoplasm ,DNMT1 ,Cancer research ,Azacitidine ,Drug Screening Assays, Antitumor ,Proto-Oncogene Proteins c-fos ,medicine.drug - Abstract
We have previously shown that hydrogen peroxide–resistant permanent (OC-14) cells are resistant to the cytotoxicity of several exogenous oxidative and anticancer agents including H2O2, etoposide, and cisplatin; and we refer to this process as an oxidative multimodality-resistant phenotype (MMRP). Furthermore, OC-14 cells contain increased activator protein 1 activity, and inhibition of activator protein 1 reversed the MMRP. In this study, we show that permanent Rat-1 cell lines genetically altered to overexpress c-Fos also displayed a similar MMRP to H2O2, etoposide, and cisplatin as OC-14 cells. Gene expression analysis of the OC-14 cells and c-Fos–overexpressing cells showed increased DNMT1 expression. Where OC-14 and c-Fos–overexpressing cells were exposed to 5-aza-2′-deoxycytidine, which inhibits DNMT activity, a significant but incomplete reversal of the MMRP was observed. Thus, it seems logical to suggest that DNMT1 might be at least one target in the MMRP. Rat-1 cells genetically altered to overexpress DNMT1 were also shown to be resistant to the cytotoxicity of H2O2, etoposide, and cisplatin. Finally, somatic HCT116 knockout cells that do not express either DNMT1 (DNMT1−/−) or DNMT3B (DNMT3B−/−) were shown to be more sensitive to the cytotoxicity of H2O2, etoposide, and cisplatin compared with control HCT116 cells. This work is the first example of a role for the epigenome in tumor cell resistance to the cytotoxicity of exogenous oxidative (H2O2) or systemic (etoposide and cisplatin) agents and highlights a potential role for DNMT1 as a potential molecular target in cancer therapy. (Mol Cancer Res 2008;6(2):243–9)
- Published
- 2008
28. Chromosomal protein HMGN1 modulates the expression of N-cadherin
- Author
-
Yaffa R, Rubinstein, Takashi, Furusawa, Jae-Hwan, Lim, Yuri V, Postnikov, Katherine L, West, Yehudit, Birger, Sunmin, Lee, Phuongmai, Nguyen, Jane B, Trepel, and Michael, Bustin
- Subjects
Mice, Knockout ,Reverse Transcriptase Polymerase Chain Reaction ,Blotting, Western ,Down-Regulation ,Gene Expression Regulation, Developmental ,Fibroblasts ,Cadherins ,Embryo, Mammalian ,Chromatin ,Chromosomes ,Article ,Mice ,Cell Movement ,Gene Targeting ,Mutation ,Cell Adhesion ,Animals ,Cell Adhesion Molecules ,Cells, Cultured ,HMGN1 Protein ,beta Catenin ,Cell Line, Transformed ,Protein Binding - Abstract
HMGN1 is a nuclear protein that binds to nucleosomes and alters the accessibility of regulatory factors to their chromatin targets. To elucidate its biological function and identify specific HMGN1 target genes, we generated Hmgn1-/- mice. DNA microarray analysis of Hmgn1+/+ and Hmgn1-/- embryonic fibroblasts identified N-cadherin as a potential HMGN1 gene target. RT-PCR and western blot analysis confirmed a linkage between HMGN1 expression and N-cadherin levels. In both transformed and primary mouse embryonic fibroblasts (MEFs), HMGN1 acted as negative regulator of N-cadherin expression. Likewise, the N-cadherin levels in early embryos of Hmgn1-/- mice were higher than those of their Hmgn1+/+ littermates. Loss of HMGN1 increased the adhesiveness, motility and aggregation potential of Hmgn1-/- MEFs, a phenotype consistent with increased levels of N-cadherin protein. Re-expression of wild-type HMGN1, but not of the mutant HMGN1 protein that does not bind to chromatin, in Hmgn1-/- MEFs, decreased the levels of N-cadherin and restored the Hmgn1+/+ phenotype. These studies demonstrate a role for HMGN1 in the regulation of specific gene expression. We suggest that in MEFs, and during early mouse development, the interaction of HMGN1 with chromatin down-regulates the expression of N-cadherin.
- Published
- 2005
29. Redox-sensitive signaling factors as a novel molecular targets for cancer therapy
- Author
-
Kheem S. Bisht, David Gius, DeeDee Smart, Lunching Sun, Phuongmai Nguyen, J. Daniel Pennington, and Tony J. C. Wang
- Subjects
Cancer Research ,Programmed cell death ,Antineoplastic Agents ,Biology ,medicine.disease_cause ,Neoplasms ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Enzyme Inhibitors ,Autocrine signalling ,Cell damage ,Pharmacology ,Drug discovery ,medicine.disease ,Cell biology ,Oxidative Stress ,Infectious Diseases ,Oncology ,Drug Resistance, Neoplasm ,Thioredoxin ,Signal transduction ,Oxidation-Reduction ,Intracellular ,Oxidative stress ,Signal Transduction - Abstract
Tumor cells undergoing proliferation, de-differentiation and progression depend on a complex set of respiratory pathways to generate the necessary energy. The metabolites from these pathways produce significant oxidative stress and must be buffered to prevent permanent cell damage and cell death. It is now clear that, in order to cope with and defend against the detrimental effects of oxidative stress, a series of redox-sensitive, pro-survival signaling pathways and factors regulate a complex intracellular redox buffering network. This review develops the hypothesis that tumor cells use these redox-sensitive, pro-survival signaling pathways and factors - up-regulated due to increased tumor cell respiration - to evade the damaging and cytotoxic effects of specific anticancer agents. It further suggests that redox-sensitive, signaling factors may be potential novel targets for drug discovery.
- Published
- 2005
30. CL100 expression is down-regulated in advanced epithelial ovarian cancer and its re-expression decreases its malignant potential
- Author
-
Yung Hyun Choi, Ichiro Kinoshita, Paul Dent, Nelly Auersperg, Ramon G Manzano, Silvestre Vicent, Mark Dayton, Jane B. Trepel, Luis M. Montuenga, Michael J. Birrer, Ginger J. Gardner, and Phuongmai Nguyen
- Subjects
Cancer Research ,medicine.medical_specialty ,endocrine system diseases ,Tumor suppressor gene ,Blotting, Western ,Motility ,Down-Regulation ,Mice, Nude ,Cell Cycle Proteins ,Biology ,Cell morphology ,Polymerase Chain Reaction ,Immediate-Early Proteins ,Immunoenzyme Techniques ,Mice ,Cell Movement ,Internal medicine ,Protein Phosphatase 1 ,Genetics ,medicine ,Cell Adhesion ,Phosphoprotein Phosphatases ,Tumor Cells, Cultured ,Animals ,Humans ,Cyclin D1 ,Luciferases ,Molecular Biology ,DNA Primers ,Ovarian Neoplasms ,Oncogene ,Genetic transfer ,Cancer ,Cell Differentiation ,Dual Specificity Phosphatase 1 ,Epithelial Cells ,medicine.disease ,Blotting, Northern ,Phosphoric Monoester Hydrolases ,Cystadenocarcinoma, Serous ,Adenocarcinoma, Papillary ,Endocrinology ,Cell culture ,Cancer research ,RNA ,Female ,Protein Tyrosine Phosphatases ,Ovarian cancer - Abstract
Although early stage ovarian cancer can be eAectively treated with surgery and chemotherapy, the majority of cases present with advanced disease, which remains essentially incurable. Unfortunately, little is known about the genes important for the development and progression of this disease. In this study, the expression of 68 phosphatases was determined in immortalized ovarian epithelial cells (IOSE) and compared to ovarian cancer cell lines. CL100, a dual specificity phosphatase, displayed 10‐25-fold higher expression in normal compared to malignant ovarian cell lines. Immunohistochemical staining of normal ovaries and 68 ovarian cancer specimens confirmed this diAerential expression. Re-expression of CL100 in ovarian cancer cells decreased adherent and nonadherent cell growth and induced phenotypic changes including loss of filopodia and lamellipodia with an associated decrease in cell motility. Induced expression of CL100 in ovarian cancer cells suppressed intraperitoneal tumor growth in nude mice. These results show for the first time that CL100 expression is altered in human ovarian cancer, that CL100 expression changes cell morphology and motility, and that it suppresses intraperitoneal growth of human ovarian epithelial cancer. These data suggest that down-regulation of CL100 may play a role in the progression of human ovarian cancer. Oncogene (2002) 21, 4435‐4447. doi:10.1038/sj.onc. 1205542
- Published
- 2001
31. The hsp90-related protein TRAP1 is a mitochondrial protein with distinct functional properties
- Author
-
David O. Toft, David B. Donner, Barbara A.L. Owen, Phuongmai Nguyen, Sara J. Felts, and Jane B. Trepel
- Subjects
Molecular Sequence Data ,Fluorescent Antibody Technique ,Biology ,Mitochondrion ,Biochemistry ,Hsp90 inhibitor ,Cell Line ,chemistry.chemical_compound ,polycyclic compounds ,Animals ,Drosophila Proteins ,Humans ,Amino Acid Sequence ,HSP90 Heat-Shock Proteins ,Receptor ,Molecular Biology ,Cloning ,Cell Biology ,Geldanamycin ,Hsp90 ,Radicicol ,Mitochondria ,chemistry ,Chaperone (protein) ,biology.protein ,Drosophila ,Sequence Alignment - Abstract
The hsp90 family of molecular chaperones was expanded recently due to the cloning of TRAP1 and hsp75 by yeast two-hybrid screens. Careful analysis of the human TRAP1 and hsp75 sequences revealed that they are identical, and we have cloned a similar protein from Drosophila. Immunofluorescence data show that human TRAP1 is localized to mitochondria. This mitochondrial localization is supported by the existence of mitochondrial localization sequences in the amino termini of both the human and Drosophila proteins. Due to the striking homology of TRAP1 to hsp90, we tested the ability of TRAP1 to function as an hsp90-like chaperone. TRAP1 did not form stable complexes with the classic hsp90 co-chaperones p23 and Hop (p60). Consistent with these observations, TRAP1 had no effect on the hsp90-dependent reconstitution of hormone binding to the progesterone receptor in vitro, nor could it substitute for hsp90 to promote maturation of the receptor to its hormone-binding state. However, TRAP1 is sufficiently conserved with hsp90 such that it bound ATP, and this binding was sensitive to the hsp90 inhibitor geldanamycin. In addition, TRAP1 exhibited ATPase activity that was inhibited by both geldanamycin and radicicol. Thus, TRAP1 has functions that are distinct from those of hsp90.
- Published
- 2000
32. Regulation of BRCA1 by protein degradation
- Author
-
Mikhail V. Blagosklonny, Phuongmai Nguyen, Won G. An, Giovanni Melillo, Jane B. Trepel, and Leonard M. Neckers
- Subjects
Cancer Research ,Proteases ,Proteasome Endopeptidase Complex ,endocrine system diseases ,Leupeptins ,medicine.medical_treatment ,Proteolysis ,Lactacystin ,Down-Regulation ,Receptors, Cytoplasmic and Nuclear ,Protein degradation ,Karyopherins ,chemistry.chemical_compound ,Multienzyme Complexes ,Endopeptidases ,Genetics ,medicine ,Tumor Cells, Cultured ,Humans ,RNA, Messenger ,skin and connective tissue diseases ,Molecular Biology ,Cell Nucleus ,Protease ,biology ,medicine.diagnostic_test ,BRCA1 Protein ,Calpain ,Hydrolysis ,Cell biology ,Cysteine Endopeptidases ,Biochemistry ,chemistry ,Proteasome ,Gene Expression Regulation ,Proteasome inhibitor ,biology.protein ,Dactinomycin ,Carrier Proteins ,medicine.drug ,Half-Life - Abstract
BRCA1, a tumor suppressor protein implicated in hereditary forms of breast and ovarian cancer, is transcriptionally regulated in a proliferation-dependent manner. In this study, we demonstrate a substantial role for proteolysis in regulating the BRCA1 steady-state protein level in several cell lines. N-acetyl-leu-leu-norleucinal (ALLN), an inhibitor of the proteasome, calpain, and cathepsins, caused BRCA1 protein to accumulate in the nucleus of several human breast, prostate, and melanoma cell lines which express low or undetectable basal levels of BRCA1 protein, but not in cells with high basal expression of BRCA1. Protease inhibition did not increase BRCA1 synthesis, nor change its mRNA level, but it dramatically prolonged the protein's half-life. In contrast to ALLN, lactacystin and PS341, two specific proteasome inhibitors, as well as calpastatin peptide and PD150606, two selective calpain inhibitors, had no effect on BRCA1 stability, whereas ALLM, an effective calpain and cathepsin inhibitor but weak proteasome inhibitor, did stimulate accumulation of BRCA1. Moreover, three inhibitors of acidic cysteine proteases, chloroquine, ammonium chloride and bafilomycin, were as effective as ALLN. These results demonstrate that degradation by a cathepsin-like protease in fine balance with BRCA1 transcription is responsible for maintaining the low steady-state level of BRCA1 protein seen in many cancer cells.
- Published
- 1999
33. Posttranslational regulation of the retinoblastoma gene family member p107 by calpain protease
- Author
-
Jeen Lee, Ming-Lei Wu, Sang Gu Hwang, Masatoshi Maki, Joung Soon Jang, Su Jae Lee, Emiko Takano, Pierre A. Henkart, Phuongmai Nguyen, Jane B. Trepel, and Yung Hyun Choi
- Subjects
Cancer Research ,Leupeptins ,medicine.medical_treatment ,Lactacystin ,Retinoblastoma-Like Protein p107 ,Cyclin B ,Cysteine Proteinase Inhibitors ,Retinoblastoma Protein ,Amino Acid Chloromethyl Ketones ,chemistry.chemical_compound ,Genetics ,medicine ,Tumor Cells, Cultured ,Humans ,Post-translational regulation ,Lovastatin ,Cyclin B1 ,Molecular Biology ,Calpastatin ,Protease ,Expression vector ,biology ,Calpain ,Retinoblastoma protein ,Nuclear Proteins ,Dipeptides ,Ketones ,Cell biology ,Acetylcysteine ,Biochemistry ,chemistry ,Proteasome ,Bucladesine ,embryonic structures ,biology.protein ,Fluorouracil ,biological phenomena, cell phenomena, and immunity ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Protein Processing, Post-Translational - Abstract
The retinoblastoma protein plays a critical role in regulating the G1/S transition. Less is known about the function and regulation of the homologous pocket protein p107. Here we present evidence for the posttranslational regulation of p107 by the Ca2+-activated protease calpain. Three negative growth regulators, the HMG-CoA reductase inhibitor lovastatin, the antimetabolite 5-fluorouracil, and the cyclic nucleotide dibutyryl cAMP were found to induce cell type-specific loss of p107 protein which was reversible by the calpain inhibitor leucyl-leucyl-norleucinal but not by the serine protease inhibitor phenylmethylsulfonylfluoride, caspase inhibitors, or lactacystin, a specific inhibitor of the 26S proteasome. Purified calpain induced Ca2+-dependent p107 degradation in cell lysates. Transient expression of the specific calpain inhibitor calpastatin blocked the loss of p107 protein in lovastatin-treated cells, and the half-life of p107 was markedly lengthened in lovastatian-treated cells stably transfected with a calpastatin expression vector versus cells transfected with vector alone. The data presented here demonstrate down-regulation of p107 protein in response to various antiproliferative signals, and implicate calpain in p107 posttranslational regulation.
- Published
- 1999
34. [Untitled]
- Author
-
Tony J. C. Wang, Phuongmai Nguyen, Gil Bar-Sela, L. Huang, J. Pennington, David Gius, and L. Sun
- Subjects
Cancer Research ,Radiation ,Oncology ,business.industry ,CTCF ,Medicine ,Radiology, Nuclear Medicine and imaging ,business ,Transcription factor ,Cell biology - Published
- 2006
35. Post-translational Regulation of Superoxide Dismutase as a Tumor-suppressor Effect of SIRT3
- Author
-
David Gius, Kheem S. Bisht, R. Tao, O. Ozden, and Phuongmai Nguyen
- Subjects
Cancer Research ,Radiation ,SIRT3 ,biology ,business.industry ,law.invention ,Superoxide dismutase ,Oncology ,law ,biology.protein ,Cancer research ,Medicine ,Suppressor ,Radiology, Nuclear Medicine and imaging ,Post-translational regulation ,business - Published
- 2010
36. Abstract 1328: p53 represses microRNAs let-7a and let-7b in response to ionizing radiation
- Author
-
Toren Finkel, Jason E. Savage, Curtis C. Harris, Lui Cao, David Ly, Anthony Saleh, Tu Dan, William DeGraff, Phuongmai Nguyen, David Gius, Benjamin P. Soule, James B. Mitchel, and Nicole L. Simone
- Subjects
Cancer Research ,Radiosensitizer ,DNA repair ,Chemistry ,Cancer ,Transfection ,Cell cycle ,medicine.disease ,Cyclin D1 ,Oncology ,microRNA ,Gene expression ,Cancer research ,medicine - Abstract
Radiation-induced free radicals damage cells resulting in a stress response that involves large scale changes in gene expression including microRNAs (miRNA). Specifically, decreased expression of the let-7 family of miRNA has been reported. let-7 regulates cell cycle and DNA repair proteins such as Ras, cyclin D1, Rad 18 and Rad 51, which can all effect the radiation response. However, the precise role of the let-7 family and the mechanisms regulating this repression has not been elucidated. In this study, we demonstrate a mechanism underlying the let-7a3 and let-7b response to radiation exposure. Using real time RT-PCR we demonstrate that let-7a and b expression decreases by 80-90% in response to 0.5-10 Gy doses of IR in cultured HCT116 colon cancer cells. This effect is also observed by RT-PCR in response to other sources of geneotoxic stress such as etoposide or UV irradiation suggesting that a reduction in let-7 is common among DNA damaging events. We also show that this decrease in expression of let-7a and b is dependent on p53 as the decrease is absent in HCT116 p53 knockout cells. Further analysis of the effect of p53 on let-7a3 and b expression by CHIP showed an association of p53 with a predicted binding site in the host gene promoter. After treatment of mice with radiation a decrease in let-7a and b expression is observed in radio sensitive tissues. However no decrease was observed in tissues collected from p53 KO mice. Transfection of additional let-7a or let-7b into HCT116 cells decreased survival after radiation treatment, but survival is unchanged by increased let-7 in HCT116 p53 knockout cells. These results suggest that p53 can directly repress let-7 expression. Also, let-7 may play an important role in the regulation and fine-tuning of the p53 cell stress response to radiation. Furthermore, therapeutics aimed at inducing over expression of let-7 may be useful as radiation sensitizers. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1328.
- Published
- 2010
37. Radiation-Induced Alteration of the Brain Proteome: Understanding the Role of the Sirtuin 2 Deacetylase in a Murine Model.
- Author
-
Shukla, Sudhanshu, Shankavaram, Uma T., Phuongmai Nguyen, Stanley, Bruce A., and Smart, DeeDee K.
- Published
- 2015
- Full Text
- View/download PDF
38. 66 A SIRT3/FOXO3a/MnSOD signaling model for mitochondrial superoxide levels
- Author
-
Kristi Muldoon Jacobs, J. Daniel Pennington, David Gius, Mark Mishra, and Phuongmai Nguyen
- Subjects
SIRT3 ,Chemistry ,Mitochondrial superoxide ,Molecular Medicine ,Cell Biology ,Molecular Biology ,Cell biology - Published
- 2007
39. 143 The interferon-inducible GTPase MxA is a metastasis suppressor
- Author
-
A. Mendoza, C. Khanna, J. Trepel, S. Lee, M.A. Horrisberger, M.-J. Lee, J.F. Mushinski, E.J. Chung, W.M. Linehan, and Phuongmai Nguyen
- Subjects
Cancer Research ,Oncology ,Cancer research ,Metastasis suppressor ,Biology ,Interferon-inducible GTPase - Published
- 2004
40. BAT3 and SET1A Form a Complex with CTCFL/BORIS To Modulate H3K4 Histone Dimethylation and Gene Expression.
- Author
-
Phuongmai Nguyen, Bar-Sela, Gil, Lunching Sun, Bisht, Kheem S., Hengmi Cui, Kohn, Elise, Feinberg, Andrew P., and Gius, David
- Subjects
- *
CHROMATIN , *GENETIC regulation , *GENE expression , *NUCLEIC acids , *CARRIER proteins - Abstract
Chromatin status is characterized in part by covalent posttranslational modifications of histones that regulate chromatin dynamics and direct gene expression. BORIS (brother of the regulator of imprinted sites) is an insulator DNA-binding protein that is thought to play a role in chromatin organization and gene expression. BORIS is a cancer-germ line gene; these are genes normally present in male germ cells (testis) that are also expressed in cancer cell lines as well as primary tumors. This work identifies SET1A, an H3K4 methyltransferase, and BAT3, a cochaperone recruiter, as binding partners for BORIS, and these proteins bind to the upstream promoter regions of two well-characterized procarcinogenic genes, Myc and BRCA1. RNA interference (RNAi) knockdown of BAT3, as well as SET1A, decreased Myc and BRCA1 gene expression but did not affect the binding properties of BORIS, but RNAi knockdown of BORIS prevented the assembly of BAT3 and SET1A at the Myc and BRCA1 promoters. Finally, chromatin analysis suggested that BORIS and BAT3 exert their effects on gene expression by recruiting proteins such as SET1A that are linked to changes in H3K4 dimethylation. Thus, we propose that BORIS acts as a platform upon which BAT3 and SET1A assemble and exert effects upon chromatin structure and gene expression. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
41. CL100 expression is down-regulated in advanced epithelial ovarian cancer and its re-expression decreases its malignant potential.
- Author
-
Manzano, Ramon G., Montuenga, Luis M., Dayton, Mark, Dent, Paul, Kinoshita, Ichiro, Vicent, Silvestre, Gardner, Ginger J., PhuongMai Nguyen, Chot, Yung-Hyun, Trepel, Jane, Auersperg, Nelly, and Birrer, Michael J.
- Subjects
OVARIAN cancer ,PHOSPHATASES ,ONCOLOGY - Abstract
Presents information on a study which determined the expression of 68 phosphatases in immortalized ovarian cancer cell lines. Information on epithelial ovarian cancer; Materials and methodology of the study; Results and discussion.
- Published
- 2002
- Full Text
- View/download PDF
42. Cyclic AMP induces inhibition of cyclin A expression and growth arrest in human hepatoma cells
- Author
-
Jane B. Trepel, Yung Hyun Choi, Jung Sik Kim, Su Jae Lee, Phuongmai Nguyen, and Jeen Lee
- Subjects
HepG2 ,Cyclin E ,Carcinoma, Hepatocellular ,Time Factors ,Hepatocellular carcinoma ,Cyclin D ,Cyclin A ,Cyclin B ,Down-Regulation ,Gene Expression ,Apoptosis ,Protein Serine-Threonine Kinases ,Cell cycle arrest ,Cyclin-dependent kinase ,Proto-Oncogene Proteins ,CDC2-CDC28 Kinases ,Tumor Cells, Cultured ,Cyclic AMP ,Humans ,RNA, Messenger ,Molecular Biology ,biology ,Chemistry ,Cell Cycle ,Cyclin-Dependent Kinase 2 ,Liver Neoplasms ,Cyclin-dependent kinase 3 ,Cyclin-Dependent Kinase 4 ,Cell Biology ,Cyclin-Dependent Kinases ,Cell biology ,biology.protein ,Cancer research ,Cyclin-dependent kinase complex ,Cyclin A2 - Abstract
Classical cytotoxic therapy has been minimally useful in the treatment of hepatocellular carcinoma. In an effort to develop a new approach to the treatment of this neoplasm, we have investigated the signal transduction pathways regulating the growth of human hepatoma cells. In the data reported here, cyclic AMP (cAMP), a negative growth regulator for many cells of epithelial origin, induced G1 synchronization and apoptosis in the HepG2 human hepatoma cell line. The effects of cAMP on the components of the G1/S transition were analyzed. There was no detectable effect of two different cAMP analogs, 8-bromo cAMP or dibutyryl cAMP on the level of the D-type cyclins, cyclin E, cyclin-dependent kinase 2, cyclin-dependent kinase 4, p53, or the cyclin-dependent kinase inhibitors p21 or p27. In contrast, the cAMP analogs induced a dramatic downregulation of cyclin A protein, cyclin A messenger RNA, and cyclin A-dependent kinase activity. Cyclin A-dependent kinase has been shown to be required for the G1–S transition. Furthermore, cyclin A deregulation has been implicated in the pathogenesis of hepatocellular carcinoma. The data reported here suggest a novel signal transduction-based approach to hepatoma therapy.
- Full Text
- View/download PDF
43. The epigenome as a molecular marker and targetThis article is a US Government work and, as such, is in the public domain in the United States of America.: Implications for cancer.
- Author
-
David Gius, C. Matthew Bradbury, Lunching Sun, Rania T. Awwad, Lei Huang, Dee Dee K. Smart, Kheem S. Bisht, Allen S. Ho, and Phuongmai Nguyen
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.