Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Oncogene ; 35(22): 2881-92, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26411365

ABSTRACT

Brain metastases (BM) are a devastating consequence of breast cancer. BM occur more frequently in patients with estrogen receptor-negative (ER-) breast cancer subtypes; HER2 overexpressing (HER2+) tumors and triple-negative (TN) (ER-, progesterone receptor-negative (PR-) and normal HER2) tumors. Young age is an independent risk factor for the development of BM, thus we speculated that higher circulating estrogens in young, pre-menopausal women could exert paracrine effects through the highly estrogen-responsive brain microenvironment. Using a TN experimental metastases model, we demonstrate that ovariectomy decreased the frequency of magnetic resonance imaging-detectable lesions by 56% as compared with estrogen supplementation, and that the combination of ovariectomy and letrozole further reduced the frequency of large lesions to 14.4% of the estrogen control. Human BM expressed 4.2-48.4% ER+ stromal area, particularly ER+ astrocytes. In vitro, E2-treated astrocytes increased proliferation, migration and invasion of 231BR-EGFP cells in an ER-dependent manner. E2 upregulated epidermal growth factor receptor (EGFR) ligands Egf, Ereg and Tgfa mRNA and protein levels in astrocytes, and activated EGFR in brain metastatic cells. Co-culture of 231BR-EGFP cells with E2-treated astrocytes led to the upregulation of the metastatic mediator S100 Calcium-binding protein A4 (S100A4) (1.78-fold, P<0.05). Exogenous EGF increased S100A4 mRNA levels in 231BR-EGFP cells (1.40±0.02-fold, P<0.01 compared with vehicle control) and an EGFR/HER2 inhibitor blocked this effect, suggesting that S100A4 is a downstream effector of EGFR activation. Short hairpin RNA-mediated S100A4 silencing in 231BR-EGFP cells decreased their migration and invasion in response to E2-CM, abolished their increased proliferation in co-cultures with E2-treated astrocytes and decreased brain metastatic colonization. Thus, S100A4 is one effector of the paracrine action of E2 in brain metastatic cells. These studies provide a novel mechanism by which estrogens, acting through ER+ astrocytes in the brain microenvironment, can promote BM of TN breast cancers, and suggests existing endocrine agents may provide some clinical benefit towards reducing and managing BM.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/secondary , Estrogens/metabolism , Paracrine Communication , Triple Negative Breast Neoplasms/pathology , Animals , Astrocytes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Transformation, Neoplastic , ErbB Receptors/metabolism , Estradiol/pharmacology , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Paracrine Communication/drug effects
2.
Cell Death Differ ; 22(8): 1341-52, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25591736

ABSTRACT

Lung cancer is the number one cancer killer, and metastasis is the main cause of high mortality in lung cancer patients. However, mechanisms underlying the development of lung cancer metastasis remain unknown. Using genome-wide transcriptional analysis in an experimental metastasis model, we identified laminin γ2 (LAMC2), an epithelial basement membrane protein, to be significantly upregulated in lung adenocarcinoma metastatic cells. Elevated LAMC2 increased traction force, migration, and invasion of lung adenocarcinoma cells accompanied by the induction of epithelial-mesenchymal transition (EMT). LAMC2 knockdown decreased traction force, migration, and invasion accompanied by EMT reduction in vitro, and attenuated metastasis in mice. LAMC2 promoted migration and invasion via EMT that was integrin ß1- and ZEB1-dependent. High LAMC2 was significantly correlated with the mesenchymal marker vimentin expression in lung adenocarcinomas, and with higher risk of recurrence or death in patients with lung adenocarcinoma. We suggest that LAMC2 promotes metastasis in lung adenocarcinoma via EMT and may be a potential therapeutic target.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Laminin/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Laminin/genetics , Mice
3.
Cell Death Discov ; 1: 15053, 2015.
Article in English | MEDLINE | ID: mdl-28179987

ABSTRACT

Ovarian cancer is a deadly disease characterized by primary and acquired resistance to chemotherapy. We previously associated NF-κB signaling with poor survival in ovarian cancer, and functionally demonstrated this pathway as mediating proliferation, invasion and metastasis. We aimed to identify cooperating pathways in NF-κB-dependent ovarian cancer cells, using genome-wide RNA interference as a loss-of-function screen for key regulators of cell survival with IKKß inhibition. Functional genomic screen for interactions with NF-κB in ovarian cancer showed that cells depleted of Caspase8 died better with IKKß inhibition. Overall, low Caspase8 was associated with shorter overall survival in three independent gene expression data sets of ovarian cancers. Conversely, Caspase8 expression was markedly highest in ovarian cancer subtypes characterized by strong T-cell infiltration and better overall prognosis, suggesting that Caspase8 expression increased chemotherapy-induced cell death. We investigated the effects of Caspase8 depletion on apoptosis and necroptosis of TNFα-stimulated ovarian cancer cell lines. Inhibition of NF-κB in ovarian cancer cells switched the effects of TNFα signaling from proliferation to death. Although Caspase8-high cancer cells died by apoptosis, Caspase8 depletion downregulated NF-κB signaling, stabilized RIPK1 and promoted necroptotic cell death. Blockage of NF-κB signaling and depletion of cIAP with SMAC-mimetic further rendered these cells susceptible to killing by necroptosis. These findings have implications for anticancer strategies to improve outcome for women with low Caspase8-expressing ovarian cancer.

4.
Proteomics ; 1(10): 1205-15, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11721633

ABSTRACT

Proteomic based approaches are beginning to be utilized to study the natural history and treatment of breast cancer. A variety of proteomics approaches are under study, and are summarized herein. Two-dimensional gel electrophoresis (2D-PAGE) is still the foundation of most proteomics studies. We present an analysis of 2D-PAGE studies reported to date in breast cancer, including those examining normal/tumor differences and selected populations of breast cells. Newer technologies such as laser capture microdissection and highly sensitive mass spectrometry methods are currently being used together to identify greater numbers of lower abundance proteins that are differentially expressed between defined cell populations. Novel technologies still in developmental phases will enable identification of validated targets in small biopsy specimens, including high density protein arrays, antibody arrays and lysate arrays. Surface-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) analysis enables the high throughput characterization of lysates from very few tumor cells and may be best suited for clinical biomarker studies. We present SELDI-TOF data herein to show the accuracy of the method in a small cohort of breast tumors, as well as its potential discriminatory capability. Such technologies are expected to supplement our armamentarium of mRNA-based assays, and provide critical information on protein levels and post-translational modifications.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Profiling , Neoplasm Proteins/analysis , Proteome/analysis , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Dissection/methods , Electrophoresis, Gel, Two-Dimensional/methods , Female , Humans , Lasers , Mass Screening/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
6.
Cancer Res ; 61(5): 2320-7, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280805

ABSTRACT

We hypothesize that elevation of Nm23-H1 expression in micrometastatic breast cancer cells may inhibit their metastatic colonization and further invasion, and induce differentiation, thus resulting in a clinical benefit. The current study investigated the possible contribution of DNA methylation to the regulation of Nm23-H1 expression, based on the observation that two CpG islands are present in its promoter. 5-Aza-2'-deoxycytidine (5-Aza-CdR), a DNA methylation inhibitor, increased the Nm23-H1 expression of 5 of 11 human breast carcinoma cell lines in vitro, including 3 of 3 metastatically competent lines. Increased Nm23-H1 expression was accompanied by a reduction in motility in vitro, with minimal effect on proliferation. Both increased Nm23-H1 expression and decreased motility were observed using low (75 nM) concentrations of 5-Aza-CdR. Array analysis of MDA-MB-231 breast carcinoma cells treated with 5-Aza-CdR confirmed the elevation of nm23-H1 mRNA, whereas relatively few other genes exhibited altered expression. Bisulfite sequencing of the two CpG islands in a panel of cell lines and in 20 infiltrating ductal carcinomas revealed that one island (-3090 bp to -3922 bp) exhibited infrequent differential methylation. The data indicate that DNA methylation inhibitors can directly or indirectly cause both elevation of Nm23-H1 expression and decreased function in one aspect of metastasis, motility.


Subject(s)
Azacitidine/analogs & derivatives , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Cell Movement/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic/physiology , Genes, Tumor Suppressor , Monomeric GTP-Binding Proteins/genetics , Nucleoside-Diphosphate Kinase , Transcription Factors/genetics , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , CpG Islands/genetics , DNA Methylation/drug effects , Decitabine , Humans , Monomeric GTP-Binding Proteins/biosynthesis , NM23 Nucleoside Diphosphate Kinases , Neoplasm Metastasis , Promoter Regions, Genetic/genetics , Transcription Factors/biosynthesis , Transfection , Tumor Cells, Cultured
8.
Cancer Res ; 60(10): 2611-5, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10825131

ABSTRACT

The role of cyclin D1 overexpression in human breast premalignancy was investigated using immortal, nontumorigenic MCF-10A cells. Previous work documented that cyclin D1 overexpression promoted in vitro anchorage-independent colonization. We now report that the colonization of MCF-10A cyclin D1 transfectants was preferentially inhibited by gamma-radiation and specific classes of apoptosis inducers [Apo-2 ligand (Apo-2L), but not tumor necrosis factor alpha]. Antibody inhibition studies and semiquantitative PCR indicated that radiation inhibition of colonization was partially mediated via the Apo2L/TRAIL pathway. The apoptotic removal of cyclin D1-overexpressing, colonization-competent premalignant breast cells by Apo2L/TRAIL or other biologicals may represent a novel approach to the prevention of breast cancer.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Cyclin D1/biosynthesis , Gamma Rays/therapeutic use , Membrane Glycoproteins/metabolism , Precancerous Conditions/pathology , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism , Apoptosis Regulatory Proteins , Breast Neoplasms/radiotherapy , Carcinoma, Intraductal, Noninfiltrating/radiotherapy , Cell Division/radiation effects , Female , Humans , Ligands , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Cells, Cultured
9.
Breast Cancer Res Treat ; 59(1): 27-39, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10752677

ABSTRACT

Cyclin D1 is frequently overexpressed in human breast ductal carcinoma in situ (DCIS) specimens, which confer a high risk for the development of infiltrating ductal carcinoma. If causally involved in the genesis of human breast malignancy, cyclin D1 may represent an interesting target for chemopreventive approaches, as it sits at the junction of many growth factor and hormonal pathways. We have used the MCF-10A human breast cell line, derived from a mastectomy containing a low risk premalignant lesion, as a model system. Three cyclin D1 transfectants exhibited physiologically relevant levels of transgene overexpression, but no coordinate overexpression of other cell cycle related genes. Proliferation assays, flow cytometry, and cdk enzymatic assays of anchorage-dependent proliferation indicated only a minimal and transient effect of cyclin D1. In contrast, cyclin D1 overexpression significantly stimulated anchorage-independent colonization in soft agar or methylcellulose, accompanied by greater Gl-S progression. The cdk4 activity of the control- and cyclin D1 transfectants in colonization assays was comparable, but the cdk2 activity was higher in the latter. Injection of control- and cyclin D1 transfected MCF-10A cells in matrigel into nude mice failed to produce tumors within 1.5 years. The data suggest that cyclin D1 overexpression is an early feature of breast neoplastic progression, and can contribute to cancer development through the promotion of colonization.


Subject(s)
Breast Neoplasms/metabolism , CDC2-CDC28 Kinases , Cyclin D1/biosynthesis , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , Precancerous Conditions/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Breast Neoplasms/pathology , Cyclin D1/genetics , Cyclin-Dependent Kinase 2 , Female , Humans , Mice , Mice, Nude , Precancerous Conditions/physiopathology , Risk Factors , Transforming Growth Factors/pharmacology , Tumor Cells, Cultured
11.
Breast Cancer Res ; 2(6): 408-16, 2000.
Article in English | MEDLINE | ID: mdl-11250734

ABSTRACT

The present is an overview of recent data that describes the genetic underpinnings of the suppression of cancer metastasis. Despite the explosion of new information about the genetics of cancer, only six human genes have thus far been shown to suppress metastasis functionally. Not all have been shown to be functional in breast carcinoma. Several additional genes inhibit various steps of the metastatic cascade, but do not necessarily block metastasis when tested using in vivo assays. The implications of this are discussed. Two recently discovered metastasis suppressor genes block proliferation of tumor cells at a secondary site, offering a new target for therapeutic intervention.


Subject(s)
Antigens, CD , Breast Neoplasms/pathology , Carcinoma/secondary , Genes, Tumor Suppressor , MAP Kinase Kinase 4 , Membrane Glycoproteins , Neoplasm Metastasis/genetics , Nucleoside-Diphosphate Kinase , Proto-Oncogene Proteins , Breast Neoplasms/genetics , Cadherins/genetics , Cadherins/physiology , Carcinoma/genetics , Carcinoma/pathology , Cell Adhesion/genetics , Disease Progression , Female , Forecasting , Humans , Kangai-1 Protein , Kisspeptins , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/physiology , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/physiology , NM23 Nucleoside Diphosphate Kinases , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Proteins/genetics , Proteins/physiology , Repressor Proteins , Transcription Factors/genetics , Transcription Factors/physiology , Tumor Suppressor Proteins
12.
J Bioenerg Biomembr ; 32(3): 301-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11768314

ABSTRACT

Tumor metastasis is the leading cause of death in cancer patients. From a series of tumor cohort studies, low expression of Nm23/NDP kinase has been correlated with poor patient prognosis and survival, lymph node infiltration, and histopathological indicators of high metastatic potential in a number of cancer types, including mammary and ovarian carcinomas and melanoma. In other tumor types, no correlation has been established. Transfection of Nm23/NDP kinase cDNA into highly metastatic breast, melanoma, prostrate and squamous cell carcinomas, and colon adenocarcinoma cells significantly reduced the metastatic competency of the cells in vivo. In culture, cell motility, invasion, and colonization were inhibited, whereas tumorigenicity and cellular proliferation were not affected, indicating that Nm23/NDP kinase acts as a metastasis suppressor.


Subject(s)
Monomeric GTP-Binding Proteins/physiology , Neoplasms/enzymology , Nucleoside-Diphosphate Kinase/physiology , Transcription Factors/physiology , Gene Expression Profiling , Humans , Loss of Heterozygosity , Monomeric GTP-Binding Proteins/genetics , Mutagenesis , NM23 Nucleoside Diphosphate Kinases , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/therapy , Nucleoside-Diphosphate Kinase/genetics , Transcription Factors/genetics , Transfection
13.
Biochim Biophys Acta ; 1449(2): 157-68, 1999 Mar 08.
Article in English | MEDLINE | ID: mdl-10082974

ABSTRACT

NM23 (NDP kinase) modulates the gating of muscarinic K+ channels by agonists through a mechanism distinct from GTP regeneration. To better define the function of NM23 in this pathway and to identify sites in NM23 that are important for its role in muscarinic K+ channel function, we utilized MDA-MB-435 human breast carcinoma cells that express low levels of NM23-H1. M2 muscarinic receptors and GIRK1/GIRK4 channel subunits were co-expressed in cells stably transfected with vector only (control), wild-type NM23-H1, or several NM23-H1 mutants. Lysates from all cell lines tested exhibit comparable nucleoside diphosphate (NDP) kinase activity. Whole cell patch clamp recordings revealed a substantial reduction of the acute desensitization of muscarinic K+ currents in cells overexpressing NM23-H1. The mutants NM23-H1P96S and NM23-H1S44A resembled wild-type NM23-H1 in their ability to reduce desensitization. In contrast, mutants NM23-H1S120G and NM23-H1S120A completely abolished the effect of NM23-H1 on desensitization of muscarinic K+ currents. Furthermore, NM23-H1S120G potentiated acute desensitization, indicating that this mutant retains the ability to interact with the muscarinic pathway, but has properties antithetical to those of the wild-type protein. We conclude that NM23 acts as a suppressor of the processes leading to the desensitization of muscarinic K+ currents, and that Ser-120 is essential for its actions.


Subject(s)
Monomeric GTP-Binding Proteins , Nucleoside-Diphosphate Kinase , Potassium Channels/drug effects , Receptors, Muscarinic/drug effects , Transcription Factors/pharmacology , Humans , Mutation , NM23 Nucleoside Diphosphate Kinases , Patch-Clamp Techniques , Potassium Channels/chemistry , Potassium Channels/metabolism , Receptors, Muscarinic/metabolism , Serine/chemistry , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured
15.
Biochem Soc Symp ; 63: 261-71, 1998.
Article in English | MEDLINE | ID: mdl-9513729

ABSTRACT

The nm23 genes were discovered on the basis of their reduced expression by highly metastatic cell lines. This trend was confirmed in cohorts of several types of human carcinomas and melanomas. Several transfection studies have demonstrated the suppressive effect of nm23 overexpression on the metastatic aggressiveness of melanoma and breast carcinoma cells in vivo. These transfection experiments have also demonstrated an effect of nm23 overexpression on cellular functions involved in the metastatic phenotype, such as cell motility, and point to a regulatory role for Nm23 proteins in cellular signalling pathways. Nm23 homologues from various species are also involved in normal tissue development and differentiation. Transfection of nm23-H1 into breast cancer cells provided a functional demonstration of the involvement of this gene in the differentiation of mammary epithelial cells. However, the molecular mechanism of these biological effects remains unknown. Several biochemical activities have been reported for Nm23, including NDP kinase activity, serine autophosphorylation and protein-histidine kinase activity. To define the possible significance of these biochemical activities, we carried out site-directed mutagenesis of the relevant codons of nm23-H1 cDNA and studied the effects upon transfection into MDA-MB-435 human breast carcinoma cells. We have also used Nm23 expression as a molecular marker to identify novel compounds that are active against the most aggressive tumour cells. This approach revealed that none of the standard agents currently in clinical use is preferentially active against the most aggressive tumour cells, and allowed us to identify new compounds that are preferentially inhibitory towards low-Nm23-expressing breast carcinoma and melanoma cell lines. This analysis also revealed a significant correlation between Nm23 levels and sensitivity of the tumour cells to alkylating agents. A functional implication of Nm23 proteins in this phenomenon was demonstrated after transfection of nm23 cDNAs into melanoma and breast and ovarian carcinoma cells.


Subject(s)
Monomeric GTP-Binding Proteins , Neoplasm Metastasis/genetics , Nucleoside-Diphosphate Kinase , Transcription Factors/genetics , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Cell Differentiation , Growth , Humans , Melanoma/genetics , Molecular Sequence Data , NM23 Nucleoside Diphosphate Kinases , Transcription Factors/chemistry , Transcription Factors/physiology , Transfection
16.
Cancer Epidemiol Biomarkers Prev ; 7(1): 29-35, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9456240

ABSTRACT

The development of prevention strategies for breast cancer will require a molecular map of carcinogenesis. We have investigated gene expression patterns in premalignant and early carcinomatous human breast lesions that confer to the patient varying risks for developing invasive breast cancer. The relative expression levels of one of the retinoid receptors, retinoid X receptor (RXR), was determined by in situ hybridization to 58 biopsy specimens; RXR mRNA grain density over each lesion was compared to that over the normal ductal/lobular units in each section. Overexpression of RXR mRNA was observed in 66% of noncomedo ductal carcinoma in situ (DCIS), which confer a >8-fold increase in breast cancer risk, and 88% of comedo DCIS lesions, which are associated with a yet higher risk. In contrast, only 8% of lesions that confer little or no increase in breast cancer risk overexpressed RXR mRNA (P = 0.0008). Limited in situ hybridization data using retinoic acid receptor (RAR) riboprobes showed overexpression of RAR alpha, but not RAR beta or -gamma, in only a modest percentage (36%) of cases, suggesting that all members of the retinoid receptor superfamily are not similarly regulated. Immunohistochemistry performed on 52 DCIS specimens for alpha, beta, and gamma isoforms of RXR confirmed its overexpression at the protein level and implicate RXR alpha as the predominant overexpressed form. The data indicate that RXR overexpression is associated with an increased risk for the development of invasive breast cancer in human breast lesions and suggest the hypothesis that it is causally involved in breast oncogenesis. The implications for retinoid chemoprevention are discussed.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/pathology , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Breast Neoplasms/genetics , Carcinoma in Situ/genetics , Carcinoma, Ductal, Breast/genetics , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization , In Vitro Techniques , Neoplasm Invasiveness , Retinoid X Receptors , Retinoids/therapeutic use , Risk , Up-Regulation
17.
Breast Cancer Res Treat ; 52(1-3): 17-28, 1998.
Article in English | MEDLINE | ID: mdl-10066069

ABSTRACT

Cyclins are regulatory subunits for cyclin dependent kinases in the coordination of the cell cycle. Cyclins can also serve non-cell cycle functions, such as the transactivation of estrogen receptor by cyclin D. Evidence for the participation of the G1 cyclins D and E in breast cancer is summarized, including transgenic and knockout mice, transfections, and expression patterns in cohort studies. Overexpression of cyclin D has been reported in ductal carcinoma in situ (DCIS), and similar overexpression of cyclin E is suggested. Strategies to reduce cyclin expression are discussed as potential prevention efforts.


Subject(s)
Breast Neoplasms/chemistry , Cyclins/analysis , Animals , Breast Neoplasms/prevention & control , Cyclin D1/analysis , Cyclin D1/physiology , Cyclin E/analysis , Cyclin E/physiology , Cyclins/genetics , Cyclins/physiology , Female , Humans , Mice , Precancerous Conditions/chemistry , Retinoids/pharmacology , Tamoxifen/pharmacology
18.
Breast Dis ; 10(5-6): 47-50, 1998 Dec.
Article in English | MEDLINE | ID: mdl-15687578

ABSTRACT

The breast cancer advocacy community has impacted the course of basic research in several ways. They have heightened public awareness of research initiatives, and increased funding for basic and other research efforts. Advocates now participate in many study sections, which prioritize research proposals for funding, and scientific oversight committees. This article discusses these changes and how they have worked to date, and future efforts for consideration.

19.
J Biol Chem ; 272(36): 22526-30, 1997 Sep 05.
Article in English | MEDLINE | ID: mdl-9278405

ABSTRACT

Decreased levels of the nm23 gene product have been correlated with increased tumor metastatic potential in a variety of malignancies. At least a subset of the regulatory properties of Nm23 has been proposed to be due to transactivation of the human c-myc oncogene through binding to a homopyrimidine tract 140 base pairs upstream of the transcription start site (termed the CT element or the PuF site). Conventional transcription factors possess DNA binding and transactivation domains; Nm23 fusion proteins were used to address two questions. First, if provided with a well characterized DNA binding domain, does Nm23 possess a transactivation domain capable of stimulating transcription of an appropriate reporter? Second, if provided with a potent transactivation domain, is the DNA binding of Nm23 of sufficient specificity and affinity to direct the fusion protein to a CT-dependent reporter? Since reporter gene expression was not stimulated in either case, we conclude that Nm23 does not directly stimulate transcription through binding to the CT element and that its antimetastatic and other reported functions are likely due to other biochemical activities.


Subject(s)
Monomeric GTP-Binding Proteins , Nucleoside-Diphosphate Kinase , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Transcriptional Activation , Animals , COS Cells , DNA-Binding Proteins , Genes, myc , HeLa Cells , Humans , NM23 Nucleoside Diphosphate Kinases , Promoter Regions, Genetic
20.
Proc Natl Acad Sci U S A ; 94(17): 9000-5, 1997 Aug 19.
Article in English | MEDLINE | ID: mdl-9256424

ABSTRACT

Nm23 genes, which encode nucleoside diphosphate kinases, have been implicated in suppressing tumor metastasis. The motility of human breast carcinoma cells can be suppressed by transfection with wild-type nm23-H1, but not by transfections with two nm23-H1 mutants, nm23-H1(S12OG) and nm23-H1(P96S). Here we report that nm23-H1 can transfer a phosphate from its catalytic histidine to aspartate or glutamate residues on 43-kDa membrane proteins. One of the 43-kDa membrane proteins was not phosphorylated by either nm23-H1(P96S) or nm23-H1(S120G), and another was phosphorylated much more slowly by nm23-H1(P96S) and by nm23-H1(S120G) than by wild-type nm23-H1. Nm23-H1 also can transfer phosphate from its catalytic histidine to histidines on ATP-citrate lyase and succinic thiokinase. The rates of phosphorylation of ATP-citrate lyase by nm23-H1(S120G) and nm23-H1(P96S) were similar to that by wild-type nm23-H1. The rate of phosphorylation of succinic thiokinase by nm23-H1(S120) was similar to that by wild-type nm23-H1, and the rate of phosphorylation of succinic thiokinase by nm23-H1(P96S) was about half that by wild-type nm23-H1. Thus, the transfer of phosphate from nm23-H1 to aspartates or glutamates on other proteins appears to correlate better with the suppression of motility than does the transfer to histidines.


Subject(s)
Cell Movement/genetics , Gene Expression Regulation , Monomeric GTP-Binding Proteins , Nucleoside-Diphosphate Kinase/genetics , Transcription Factors/genetics , Animals , Gene Transfer Techniques , Humans , Liver/enzymology , NM23 Nucleoside Diphosphate Kinases , Nucleoside-Diphosphate Kinase/metabolism , PC12 Cells , Phosphorylation , Rats , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...