Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 19(8): 1649-1659, 2020 08.
Article in English | MEDLINE | ID: mdl-32404408

ABSTRACT

Multiple myeloma is a hematologic cancer that disrupts normal bone marrow function and has multiple lines of therapeutic options, but is incurable as patients ultimately relapse. We developed a novel antibody-drug conjugate (ADC) targeting CS-1, a protein that is highly expressed on multiple myeloma tumor cells. The anti-CS-1 mAb specifically bound to cells expressing CS-1 and, when conjugated to a cytotoxic pyrrolobenzodiazepine payload, reduced the viability of multiple myeloma cell lines in vitro In mouse models of multiple myeloma, a single administration of the CS-1 ADC caused durable regressions in disseminated models and complete regression in a subcutaneous model. In an exploratory study in cynomolgus monkeys, the CS-1 ADC demonstrated a half-life of 3 to 6 days; however, no highest nonseverely toxic dose was achieved, as bone marrow toxicity was dose limiting. Bone marrow from dosed monkeys showed reductions in progenitor cells as compared with normal marrow. In vitro cell killing assays demonstrated that the CS-1 ADC substantially reduced the number of progenitor cells in healthy bone marrow, leading us to identify previously unreported CS-1 expression on a small population of progenitor cells in the myeloid-erythroid lineage. This finding suggests that bone marrow toxicity is the result of both on-target and off-target killing by the ADC.


Subject(s)
Antibodies, Monoclonal/chemistry , Antineoplastic Agents/pharmacology , Benzodiazepines/chemistry , Immunoconjugates/pharmacology , Membrane Proteins/antagonists & inhibitors , Microfilament Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Pyrroles/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cell Proliferation , Drug Evaluation, Preclinical , Female , Humans , Immunoconjugates/chemistry , Macaca fascicularis , Membrane Proteins/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Microfilament Proteins/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Oncotarget ; 8(39): 65090-65099, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29029414

ABSTRACT

ADAM17 (a disintegrin and metalloproteinase 17)/TACE (TNFα converting enzyme) has emerged as a potential therapeutic target in colorectal cancer (CRC) and other cancers, due in part to its role in regulating various tumor cell surface proteins and growth factors and cytokines in the tumor microenvironment. The emergence of MEDI3622, a highly potent and specific antibody-based ADAM17 inhibitor, has allowed testing of the concept that targeting ADAM17 may be an important new therapeutic approach for CRC patients. We demonstrate that MEDI3622 is highly efficacious on tumor growth in multiple human CRC PDX models, resulting in improved survival of animals bearing tumor xenografts. MEDI3622 was further found to impact Notch pathway activity and tumor-initiating cells. The promising preclinical activity seen here supports further clinical investigation of this treatment approach to improve therapeutic outcome for patients diagnosed with metastatic CRC, including patients with KRAS-mutant tumors for whom other therapeutic options are currently limited.

4.
Pancreas ; 42(5): 861-70, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23462327

ABSTRACT

OBJECTIVE: Pancreatic cancer is a leading cancer type and its molecular pathology is poorly understood. The only potentially curative therapeutic option available is complete surgical resection; however, this is inadequate as most of the patients are diagnosed at an advanced or metastatic stage. Tumor-initiating cells (TICs) constitute a subpopulation of cells within a solid tumor that sustain tumor growth, metastasis, and chemo/radioresistance. Within pancreatic cancer, TICs have been identified based on the expression of specific cell surface markers. METHODS: We use a sphere formation assay to enrich putative TICs and use human serum as a driver of differentiation. We demonstrate by using specific blocking reagents that we can inhibit the differentiation process and maintain TIC-associated markers and genes. RESULTS: We can induce differentiation of pancreatospheres with the addition of human serum, and we identified vitronectin as an inducer of differentiation. We inhibit differentiation by human serum using an arginine-glycine-aspartate-specific peptide, which is Cilengitide; hence, demonstrating this differentiation is mediated via specific integrin receptors. CONCLUSIONS: Overall, our studies further the definition of pancreatic TICs and provide further insight into both the maintenance and differentiation of this lethal population.


Subject(s)
Cell Differentiation/drug effects , Neoplastic Stem Cells/drug effects , Snake Venoms/pharmacology , Vitronectin/pharmacology , Biomarkers, Tumor/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin alphaVbeta3/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptors, Vitronectin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serum/physiology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured
5.
Genomics ; 99(2): 108-17, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22206861

ABSTRACT

TICs are characterized by their ability to self-renew, differentiate and initiate tumor formation. miRNAs are small noncoding RNAs that bind to mRNAs resulting in regulation of gene expression and biological functions. The role of miRNAs and TICs in cancer progression led us to hypothesize that miRNAs may regulate genes involved in TIC maintenance. Using whole genome miRNA and mRNA expression profiling of TICs from primary prostate cancer cells, we identified a set of up-regulated miRNAs and a set of genes down-regulated in PSs. Inhibition of these miRNAs results in a decrease of prostatosphere formation and an increase in target gene expression. This study uses genome-wide miRNA profiling to analyze expression in TICs. We connect aberrant miRNA expression and deregulated gene expression in TICs. These findings can contribute to a better understanding of the molecular mechanisms governing TIC development/maintenance and the role that miRNAs have in the fundamental biology of TICs.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Spheroids, Cellular/metabolism , Cell Proliferation , Gene Expression Profiling , Humans , Male , MicroRNAs/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured , Up-Regulation
6.
Int J Cancer ; 128(8): 1946-54, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-20568112

ABSTRACT

The BMI1 oncogene promotes prostate cancer (PC) progression. High B-cell-specific Moloney murine leukemia virus integration site 1 (BMI1) expression predicts poor prognosis in PC patients. Recent evidence suggests that BMI1 may also play a role in docetaxel chemoresistance. However, mechanisms and clinical significance of BMI1-related chemoresistance have not been investigated. For this purpose, BMI1 was silenced in 2 PC cell lines (LNCaP and DU 145). Cell proliferation and apoptosis after docetaxel treatment were measured. Guanine oxidation was assessed by in-cell western. Global gene expression analysis was performed on BMI1 silenced cells. Oncomine database was used to compare in vitro data with gene expression in PC samples. BMI1 silencing had no effect on cell proliferation but significantly enhanced docetaxel-induced antitumor activity. Gene expression analysis demonstrated that BMI1 silencing downregulates a set of antioxidant genes. Docetaxel treatment increased guanine oxidation, whereas the antioxidant N-acetyl cysteine rescued docetaxel-induced cell death. Examination of clinical datasets revealed a positive correlation of BMI1 and antioxidant gene expression. BMI1-controlled antioxidant genes were predictive of poor prognosis in PC patients. In conclusion, BMI1 enhances antioxidant response, thereby allowing PC survival after docetaxel-based chemotherapy. BMI1-controlled antioxidant genes are overexpressed in aggressive PC and should be tested as predictors of chemotherapy failure.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Drug Resistance, Neoplasm/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Taxoids/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Cycle , Cell Proliferation , Docetaxel , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Male , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 1 , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
7.
Stem Cells ; 28(3): 390-8, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-19998373

ABSTRACT

There is mounting evidence that tumors are initiated by a rare subset of cells called cancer stem cells (CSCs). CSCs are generally quiescent, self-renew, form tumors at low numbers, and give rise to the heterogeneous cell types found within a tumor. CSCs isolated from multiple tumor types differentiate both in vivo and in vitro when cultured in serum, yet the factors responsible for their differentiation have not yet been identified. Here we show that vitronectin is the component of human serum driving stem cell differentiation through an integrin alpha V beta 3-dependent mechanism. CSCs cultured on vitronectin result in downregulation of stem cell genes, modulation of differentiation markers, and loss of beta-catenin nuclear localization. Blocking integrin alpha V beta 3 inhibits differentiation and subsequently tumor formation. Thus, CSCs must be engaged by one or more extracellular signals to differentiate and initiate tumor formation, defining a new axis for future novel therapies aimed at both the extrinsic and intracellular pathways.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Vitronectin/metabolism , Animals , Biomarkers/analysis , Biomarkers/metabolism , Blood Proteins/metabolism , Blood Proteins/pharmacology , Breast Neoplasms/chemically induced , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Carcinoma/chemically induced , Carcinoma/metabolism , Carcinoma/physiopathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Chromatography, Liquid , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/metabolism , Male , Mass Spectrometry , Mice , Neoplasms/chemically induced , Neoplasms/physiopathology , Neoplastic Stem Cells/drug effects , Nuclear Localization Signals/drug effects , Nuclear Localization Signals/metabolism , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/physiopathology , Vitronectin/pharmacology , beta Catenin/drug effects , beta Catenin/metabolism
8.
Clin Exp Metastasis ; 26(5): 433-46, 2009.
Article in English | MEDLINE | ID: mdl-19221883

ABSTRACT

Development of metastasis is a leading cause of cancer-induced death. Acquisition of an invasive tumor cell phenotype suggests loss of cell adhesion and basement membrane breakdown during a process termed epithelial-to-mesenchymal transition (EMT). Recently, cancer stem cells (CSC) were discovered to mediate solid tumor initiation and progression. Prostate CSCs are a subpopulation of CD44(+) cells within the tumor that give rise to differentiated tumor cells and also self-renew. Using both primary and established prostate cancer cell lines, we tested the assumption that CSCs are more invasive. The ability of unsorted cells and CD44-positive and -negative subpopulations to undergo Matrigel invasion and EMT was evaluated, and the gene expression profiles of these cells were analyzed by microarray and a subset confirmed using QRT-PCR. Our data reveal that a subpopulation of CD44(+) CSC-like cells invade Matrigel through an EMT, while in contrast, CD44(-) cells are non-invasive. Furthermore, the genomic profile of the invasive cells closely resembles that of CD44(+)CD24(-) prostate CSCs and shows evidence for increased Hedgehog signaling. Finally, invasive cells from DU145 and primary prostate cancer cells are more tumorigenic in NOD/SCID mice compared with non-invasive cells. Our data strongly suggest that basement membrane invasion, an early and necessary step in metastasis development, is mediated by these potential cancer stem cells.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , CD24 Antigen/biosynthesis , Epithelium/pathology , Genomics , Humans , Hyaluronan Receptors/biosynthesis , Male , Mesoderm/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Stem Cells/metabolism
9.
Clin Cancer Res ; 14(19): 6073-80, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829485

ABSTRACT

PURPOSE: Advanced prostate cancer is first treated with androgen deprivation therapy. However, tumors become resistant to and grow despite castrate levels of testosterone. Growth and proliferation of CRPC is mediated by gain-of-function changes in the AR and AR reactivation. Expression of manganese superoxide dismutase (SOD2), which regulates cellular ROS, is markedly down-regulated in CRPC when compared with hormone-responsive tumors. EXPERIMENTAL DESIGN: Here, we knocked down SOD2 expression in AR-expressing LNCaP prostate cancer cells and determined gene expression changes, transcription factor binding, and AR transcription activity in SOD2 knockdown cells. RESULTS: SOD2 knockdown results in an increase in ROS. Gene expression changes induced by SOD2 knockdown results in the up-regulation of genes that are also androgen responsive and 46% of genes up-regulated 2-fold by the androgen ligand R1881 are also up-regulated to the same extent with SOD2 knockdown. The induction of many of these genes with SOD2 knockdown, such as VEGFA and FKBP5, is reversible with the antioxidant N-acetylcysteine, suggesting that this mechanism is directly linked to ROS. Furthermore, an array for transcription factor DNA-binding activity shows that SOD2 knockdown induces DNA binding by several transcription factors, including AR. SOD2 knockdown-induced AR activation was confirmed by electrophoretic mobility shift assay and luciferase activity, and both were readily reversible with N-acetylcysteine. CONCLUSIONS: These findings show that down-regulation of SOD2 induces AR activity in a ROS-dependent manner, and suggest that there may be a role for antioxidant therapy in CRPC.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Silencing , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Superoxide Dismutase/genetics , Androgen Antagonists/therapeutic use , Androgens , Antioxidants/metabolism , Castration , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Ligands , Male , Reactive Oxygen Species , Tacrolimus Binding Proteins/metabolism , Testosterone/pharmacology , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Blood ; 109(9): 3953-62, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17192397

ABSTRACT

Reactive oxygen species are known to be involved in several cellular processes, including cell signaling. SOD2 is a key enzyme in the conversion of reactive oxygen species and has been implicated in a host of disease states, including cancer. Using an integrated, whole-cell approach encompassing epigenetics, genomics, and proteomics, we have defined the role of SOD2 in multiple myeloma. We show that the SOD2 promoter is methylated in several cell lines and there is a correlative decrease in expression. Furthermore, myeloma patient samples have decreased SOD2 expression compared with healthy donors. Overexpression of SOD2 results in decreased proliferation and altered sensitivity to 2-methoxyestradiol-induced DNA damage and apoptosis. Genomic profiling revealed regulation of 65 genes, including genes involved in tumorigenesis, and proteomic analysis identified activation of the JAK/STAT pathway. Analysis of nearly 400 activated transcription factors identified 31 transcription factors with altered DNA binding activity, including XBP1, NFAT, forkhead, and GAS binding sites. Integration of data from our gestalt molecular analysis has defined a role for SOD2 in cellular proliferation, JAK/STAT signaling, and regulation of several transcription factors.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Multiple Myeloma/enzymology , Neoplasm Proteins/biosynthesis , Signal Transduction , Superoxide Dismutase/biosynthesis , 2-Methoxyestradiol , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/genetics , Estradiol/analogs & derivatives , Estradiol/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Multiple Myeloma/genetics , Neoplasm Proteins/genetics , Proteomics , Reactive Oxygen Species , Signal Transduction/drug effects , Signal Transduction/genetics , Superoxide Dismutase/genetics , Tubulin Modulators/pharmacology
11.
Cancer Biol Ther ; 5(9): 1154-60, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16855375

ABSTRACT

Inactivation of the p53 pathway is a common feature of neoplasia. Dysregulation of the p53 pathway has been shown to involve mutations of p53, increased expression of the p53 inhibitor HDM-2, or epigenetic silencing of the p53 promoter. In multiple myeloma, a neoplasia of terminally differentiated B cells, p53 mutations and deletions are relatively rare and occur in late stage disease. Here, we show that the p53 promoter is hypermethylated in several multiple myeloma cell lines in comparison to normal plasma cells. Two cell lines containing mutant p53, Lp-1 and OPM-2, show a methylation pattern that suggests that they contain one methylated and one unmethylated mutant allele. Two other cell lines, KMS-11 and OPM-2, show hypermethylation of p53 with a lack of expression. In all cell lines tested, treatment with a demethylating agents results in higher expression of p53. Furthermore, following increased expression of p53, treatment of the myeloma cell lines with a p53 activating peptide induces apoptosis. Therefore, combinatorial treatment with demethylating agents followed by delivery of a p53 activating peptide may be an effective therapeutic strategy against multiple myeloma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cytidine/analogs & derivatives , Genes, p53/drug effects , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Peptides/pharmacology , Amino Acid Sequence , Apoptosis/genetics , Cell Line, Tumor , Cytidine/genetics , Cytidine/pharmacology , DNA Methylation/drug effects , Drug Synergism , Epigenesis, Genetic/drug effects , Gene Silencing/drug effects , Humans , Molecular Sequence Data , Multiple Myeloma/pathology , Promoter Regions, Genetic , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
12.
Epigenetics ; 1(3): 138-45, 2006.
Article in English | MEDLINE | ID: mdl-17965616

ABSTRACT

Multiple myeloma (MM) has prominent features of karyotypic instability at the earliest stage, leading to extreme genetic abnormalities as the disease progresses. These successive genetic alterations can be attributed, in part, to defects in DNA repair pathways. A possible mechanism of dysregulation of the DNA repair pathway is epigenetic gene silencing. Therefore, we sought to determine the methylation status of enzymes involved in the base excision repair pathway in multiple myeloma cell lines. Here, we report the aberrant DNA methylation of TDG, one of the enzymes involved in base excision repair of damaged DNA, in several multiple myeloma cell lines but not in normal human plasma cells. DNA hypermethylation of TDG in the MM cell lines leads to lower gene expression levels that results in less efficient DNA repair activity in response to hydrogen peroxideinduced DNA damage. Expression of exogenous TDG can functionally compensate for lower repair activities of damaged DNA in the KAS-6/1 myeloma cell line, which has extensive DNA hypermethylation of the TDG promoter. Hypermethylation of DNA damage repair genes in MM cell lines may provide an explanation for the frequent genomic instability, as well as point mutations, that are encountered in MM.


Subject(s)
DNA Methylation , DNA, Neoplasm/genetics , Gene Silencing , Multiple Myeloma/genetics , Thymine DNA Glycosylase/genetics , Cell Line, Tumor , DNA Modification Methylases/genetics , DNA Repair/genetics , Enzyme Inhibitors , Gene Expression Regulation, Neoplastic , Humans , Polymerase Chain Reaction
13.
Cancer Biol Ther ; 4(5): 585-92, 2005 May.
Article in English | MEDLINE | ID: mdl-15908783

ABSTRACT

The generation of reactive oxygen species (ROS) by mitochondrial electron transport chain (ETC) and oxidative phosphorylation activity, has been linked to modifications of multiple molecular processes, including lipid peroxidation, signaling pathway and transcription factor modulation, and oxidative damage to DNA. Oxidative damage by endogenous ROS has been associated with the etiology of various pathological states. There are numerous reports that levels of manganese superoxide dismutase enzyme (MnSOD), an antioxidant enzyme responsible for the attenuation of ROS, are lowered in cancer cells, but the reasons for this reduction are poorly defined. Epigenetic silencing of genes involved in tumor suppression and DNA repair is known to occur in a variety of malignant cell types. Here we report that in the human multiple myeloma cell line KAS 6/1, the SOD-2 gene, encoding manganese superoxide dismutase, is epigenetically silenced as a result of promoter hypermethylation. The DNA methyltransferase inhibitor Zebularine reverses SOD-2 promoter methylation, increasing gene expression and enzyme levels. Infection of KAS 6/1 cells with a recombinant adenovirus carrying the MnSOD cDNA reduced the cell proliferation rate by approximately one-half, confirming the detrimental effects of epigenetic silencing of SOD-2 expression.


Subject(s)
Cell Proliferation , Epigenesis, Genetic , Gene Silencing , Multiple Myeloma/enzymology , Superoxide Dismutase/genetics , Cell Line, Tumor , DNA Methylation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Promoter Regions, Genetic , Superoxide Dismutase/analysis , Superoxide Dismutase/metabolism
14.
J Biol Chem ; 280(6): 4182-7, 2005 Feb 11.
Article in English | MEDLINE | ID: mdl-15550378

ABSTRACT

During tumorigenesis, selective proliferative advantage in certain cell subsets is associated with accumulation of multiple genetic alterations. For instance, multiple myeloma is characterized by frequent karyotypic instability at the earliest stage, progressing to extreme genetic abnormalities as the disease progresses. These successive genetic alterations can be attributed, in part, to defects in DNA repair pathways, perhaps based on epigenetic gene silencing of proteins involved in DNA damage repair. Here we report epigenetic hypermethylation of the hHR23B gene, a key component of the nucleotide excision repair in response to DNA damage, in interleukin-6 (IL-6)-responsive myeloma KAS-6/1 cells. This hypermethylation was significantly abated by Zebularine, a potent demethylating agent, with a consequent increase in the hHR23B mRNA level. Subsequent removal of this drug and supplementation with IL-6 in the culture medium re-established DNA hypermethylation of the hHR23B gene and silencing of mRNA expression levels. The inclination of DNA to be remethylated, at least within the hHR23B gene promoter region, reflects an epigenetic driving force by the cytogenetic/tumorigenic status of KAS-6/1 myeloma. The IL-6 response of KAS-6/1 myeloma also raises a question of whether the proneoplastic growth factor, such as IL-6, supports the epigenetic silencing of important DNA repair genes via promoter hypermethylation during the development of multiple myeloma.


Subject(s)
DNA-Binding Proteins/genetics , Interleukin-6/metabolism , Multiple Myeloma/metabolism , Base Sequence , Cell Line, Tumor , CpG Islands , Culture Media/pharmacology , Cytidine/analogs & derivatives , Cytosine/chemistry , DNA/metabolism , DNA Damage , DNA Methylation , DNA Repair , DNA Repair Enzymes , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Radiation , Down-Regulation , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Reporter , Humans , Karyotyping , Kinetics , Molecular Sequence Data , Plasmids/metabolism , Promoter Regions, Genetic , Pyrimidine Nucleosides/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfites/pharmacology , Transcription, Genetic , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...