Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Biol Chem ; 287(14): 10885-93, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22334690

ABSTRACT

Survivin is an oncogenic protein that is highly expressed in breast cancer and has a dual function that is dependent on its subcellular localization. In the cytosol, survivin blocks programmed cell death by inactivating caspase proteins; however, in the nucleus it facilitates cell division by regulating chromosomal movement and cytokinesis. In prior work, we showed that survivin is acetylated by CREB-binding protein (CBP), which restricts its localization to the nuclear compartment and thereby inhibits its anti-apoptotic function. Here, we identify histone deacetylase 6 (HDAC6) as responsible for abrogating CBP-mediated survivin acetylation in the estrogen receptor (ER)-positive breast cancer cell line, MCF-7. HDAC6 directly binds survivin, an interaction that is enhanced by CBP. In quiescent breast cancer cells in culture and in malignant tissue sections from ER+ breast tumors, HDAC6 localizes to a perinuclear region of the cell, undergoing transport to the nucleus following CBP activation where it then deacetylates survivin. Genetically modified mouse embryonic fibroblasts that lack mhdac6 localize survivin predominantly to the nuclear compartment, whereas wild-type mouse embryonic fibroblasts localize survivin to distinct cytoplasmic structures. Together, these data imply that HDAC6 deacetylates survivin to regulate its nuclear export, a feature that may provide a novel target for patients with ER+ breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cell Nucleus/metabolism , Histone Deacetylases/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Repressor Proteins/metabolism , Acetylation , Active Transport, Cell Nucleus , Animals , CREB-Binding Protein/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , Estrogens/metabolism , Histone Deacetylase 6 , Histone Deacetylases/chemistry , Humans , Inhibitor of Apoptosis Proteins/chemistry , Intracellular Space/metabolism , Karyopherins/metabolism , Lysine , Mice , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/chemistry , Survivin , Exportin 1 Protein
2.
J Biol Chem ; 285(46): 36129-37, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20826784

ABSTRACT

The multiple functions of the oncofetal protein survivin are dependent on its selective expression patterns within immunochemically distinct subcellular pools. The mechanism by which survivin localizes to these compartments, however, is only partly understood. Here we show that nuclear accumulation of survivin is promoted by CREB-binding protein (CBP)-dependent acetylation on lysine 129 (129K, Lys-129). We demonstrate a mechanism by which survivin acetylation at this position results in its homodimerization, while deacetylation promotes the formation of survivin monomers that heterodimerize with CRM1 and facilitate its nuclear export. Using proteomic analysis, we identified the oncogenic transcription factor STAT3 as a binding partner of nuclear survivin. We show that acetylated survivin binds to the N-terminal transcriptional activation domain of the STAT3 dimer and represses STAT3 transactivation of target gene promoters. Using multiplex PCR and DNA sequencing, we identified a single-nucleotide polymorphism (A → G) at Lys-129 that exists as a homozygous mutation in a neuroblastoma cell line and corresponds with a defect in survivin nuclear localization. Our results demonstrate that the dynamic equilibrium between survivin acetylation and deacetylation at amino acid 129 determines its interaction with CRM1, its subsequent subcellular localization, and its ability to inhibit STAT3 transactivation, providing a potential route for therapeutic intervention in STAT3-dependent tumors.


Subject(s)
Cell Nucleus/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , STAT3 Transcription Factor/metabolism , Acetylation , Active Transport, Cell Nucleus , CREB-Binding Protein/metabolism , Cell Line, Tumor , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Inhibitor of Apoptosis Proteins , Karyopherins/metabolism , Lysine/genetics , Lysine/metabolism , Microscopy, Fluorescence , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Mutation , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Polymorphism, Single Nucleotide , Protein Binding , Protein Multimerization , Receptors, Cytoplasmic and Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/genetics , Survivin , Transcriptional Activation , Exportin 1 Protein
3.
BMC Mol Biol ; 11: 66, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20807437

ABSTRACT

BACKGROUND: Postnatal expansion of the pancreatic ß-cell mass is required to maintain glucose homeostasis immediately after birth. This ß-cell expansion is regulated by multiple growth factors, including glucose, insulin, insulin-like growth factor (IGF-1) and epidermal growth factor (EGF). These mitogens signal through several downstream pathways (AKT, ERK, STAT3, and JNK) to regulate the survival and proliferation of ß-cells. Survivin, an oncofetal protein with both pro-proliferative and anti-apoptotic properties, is a known transcriptional target of both IGF-1 and EGF in cancer cells. Here, we analyzed the effects of the ß-cell mitogens IGF-1 and EGF on survivin regulation in the established pancreatic ß-cell model cell lines, MIN6 and INS-1 and in primary mouse islets. RESULTS: In pancreatic ß-cells, treatment with glucose, insulin, or EGF increased survivin protein levels at early time points. By contrast, no significant effects on survivin were observed following IGF-1 treatment. EGF-stimulated increases in survivin protein were abrogated in the presence of downstream inhibitors of the Raf-1/MEK/ERK pathway. EGF had no significant effect on survivin transcription however it prolonged the half-life of the survivin protein and stabilized survivin protein levels by inhibiting surviving ubiquitination. CONCLUSIONS: This study defines a novel mechanism of survivin regulation by EGF through the Raf-1/MEK/ERK pathway in pancreatic ß-cells, via prolongation of survivin protein half-life and inhibition of the ubiquitin-mediated proteasomal degradation pathway. This mechanism may be important for regulating ß-cell expansion after birth.


Subject(s)
Epidermal Growth Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Animals , Cell Line , Enzyme Activation , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Glucose/metabolism , Humans , Inhibitor of Apoptosis Proteins/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-raf/genetics , Rats , Repressor Proteins/genetics , Survivin , Ubiquitination
5.
Oncogene ; 24(12): 1994-2007, 2005 Mar 17.
Article in English | MEDLINE | ID: mdl-15688031

ABSTRACT

Survivin is an inhibitor of apoptosis protein that also plays critical roles in regulating the cell cycle and mitosis. Its prominent expression in essentially all human malignancies, and low or absent expression in most normal tissues, suggests that it would be an ideal target for cancer-directed therapy. Impeding development of safe and effective survivin antagonists for clinical use is a lack of understanding of the molecular mechanisms by which survivin differentially affects apoptosis and cell division, in normal and malignant cells. We show that the diverse functional roles of survivin can be explained, in part, by its heterodimerization with survivin splice variants in tumor cells. Survivin and survivin-DeltaEx3 interact within the mitochondria where they may inhibit mitochondrial-dependent apoptosis. If the expression of all survivin forms is eliminated by siRNA transfections, cells undergo both apoptosis and defective cell division. Overall, we provide new insights suggesting that targeting specific survivin isoforms, rather than survivin alone, may selectively and effectively destroy tumor cells. These findings are likely to have a significant impact in the design of biologic agents for clinical therapy.


Subject(s)
Alternative Splicing , Cell Death/genetics , Cell Division/genetics , Microtubule-Associated Proteins/genetics , Apoptosis/genetics , Brain Neoplasms , Cell Line, Tumor , Cerebellar Neoplasms , Genetic Variation , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Medulloblastoma , Mitochondria/genetics , Mitochondria/metabolism , Neoplasm Proteins , Polymerase Chain Reaction , Survivin , Transfection
6.
NPJ Genom Med ; 1: 16016, 2016.
Article in English | MEDLINE | ID: mdl-29263815

ABSTRACT

Targeted next-generation sequencing (NGS) identified a novel loss of function mutation in GARS, a gene linked to Charcot-Marie-Tooth disease (CMT), in a paediatric acute lymphoblastic leukaemia patient with severe chemotherapy-induced peripheral neuropathy (CIPN) due to vincristine. The patient was clinically asymptomatic, and lacked a family history of neuropathy. The effect of the mutation was modelled in a zebrafish knockdown system that recapitulated the symptoms of the patient both prior to and after treatment with vincristine. Confocal microscopy of pre- and post-synaptic markers revealed that the GARS knockdown results in changes to peripheral motor neurons, acetylcholine receptors and their co-localisation in neuromuscular junctions (NMJs), whereas a sensitive and reproducible stimulus-response assay demonstrated that the changes correlating with the GARS mutation in themselves fail to produce peripheral neuropathy symptoms. However, with vincristine treatment the GARS knockdown exacerbates decreased stimulus response and NMJ lesions. We propose that there is substantial benefit in the use of a targeted NGS screen of cancer patients who are to be treated with microtubule targeting agents for deleterious mutations in CMT linked genes, and for the screening in zebrafish of reagents that might inhibit CIPN.

7.
Mol Cancer Ther ; 13(3): 675-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24431073

ABSTRACT

Inhibition of XPO1 (CRM1)-mediated nuclear export of multiple tumor suppressor proteins has been proposed as a novel cancer therapeutic strategy to turn off oncogenic signals and enhance tumor suppression. Survivin is a multifunctional protein with oncogenic properties when expressed in the cytoplasm that requires the XPO1-RanGTP complex for its nuclear export. We investigated the antitumor mechanisms of the drug-like selective inhibitors of nuclear export (SINE) XPO1 antagonists KPT-185, KPT-251 KPT-276, and KPT-330 in estrogen receptor-positive and triple-negative breast cancer (TNBC) cell lines and xenograft models of human breast tumors. KPT compounds significantly inhibited breast cancer cell growth and induced tumor cell death, both in vitro and in vivo. These drugs initially promoted survivin accumulation within tumor cell nuclei. However, their major in vitro effect was to decrease survivin cytoplasmic protein levels, correlating with the onset of apoptosis. XPO1 inhibition repressed Survivin transcription by inhibiting CREB-binding protein-mediated STAT3 acetylation, and blocking STAT3 binding to the Survivin promoter. In addition, caspase-3 was activated to cleave survivin, rendering it unavailable to bind X-linked inhibitor of apoptosis protein and block the caspase cascade. Collectively, these data demonstrate that XPO1 inhibition by SINE compounds represses STAT3 transactivation to block the selective oncogenic properties of survivin and supports their clinical use in TNBC.


Subject(s)
Inhibitor of Apoptosis Proteins/genetics , Karyopherins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , STAT3 Transcription Factor/genetics , Triple Negative Breast Neoplasms/genetics , Active Transport, Cell Nucleus/genetics , Apoptosis/drug effects , Caspase 3 , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Female , Genetic Therapy , Humans , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Survivin , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy , Exportin 1 Protein
8.
PLoS One ; 8(12): e82513, 2013.
Article in English | MEDLINE | ID: mdl-24349301

ABSTRACT

Neuroblastoma is an embryonal tumor of childhood with a heterogenous clinical presentation that reflects differences in activation of complex biological signaling pathways. Protein phosphorylation is a key component of cellular signal transduction and plays a critical role in processes that control cancer cell growth and survival. We used shotgun LC/MS to compare phosphorylation between a human MYCN amplified neuroblastoma cell line (NB10), modeling a resistant tumor, and a human neural precursor cell line (NPC), modeling a normal baseline neural crest cell. 2181 unique phosphorylation sites representing 1171 proteins and 2598 phosphopeptides were found. Protein kinases accounted for 6% of the proteome, with a predominance of tyrosine kinases, supporting their prominent role in oncogenic signaling pathways. Highly abundant receptor tyrosine kinase (RTK) phosphopeptides in the NB10 cell line relative to the NPC cell line included RET, insulin-like growth factor 1 receptor/insulin receptor (IGF-1R/IR), and fibroblast growth factor receptor 1 (FGFR1). Multiple phosphorylated peptides from downstream mediators of the PI3K/AKT/mTOR and RAS pathways were also highly abundant in NB10 relative to NPC. Our analysis highlights the importance of RET, IGF-1R/IR and FGFR1 as RTKs in neuroblastoma and suggests a methodology that can be used to identify potential novel biological therapeutic targets. Furthermore, application of this previously unexploited technology in the clinic opens the possibility of providing a new wide-scale molecular signature to assess disease progression and prognosis.


Subject(s)
Neuroblastoma/metabolism , Phosphoproteins/metabolism , Proteomics , Proto-Oncogene Proteins c-ret/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Cell Line, Tumor , Cell Survival/drug effects , Humans , MAP Kinase Signaling System , Molecular Sequence Annotation , Neural Stem Cells/metabolism , Neuroblastoma/genetics , Phosphatidylinositol 3-Kinases , Phosphoproteins/genetics , Protein Interaction Maps , Protein Kinase Inhibitors/pharmacology , Proteome , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, Insulin/antagonists & inhibitors , raf Kinases/metabolism
9.
Int J Biochem Mol Biol ; 3(2): 137-51, 2012.
Article in English | MEDLINE | ID: mdl-22773955

ABSTRACT

CRM1 (Chromosomal Maintenance 1, also known as Exportin 1) is the major mammalian export protein that facilitates the transport of large macromolecules including RNA and protein across the nuclear membrane to the cytoplasm. The gene encoding CRM1 was originally identified in yeast as required to maintain higher order chromosome structure. In mammalian cells, CRM1 was found to bind several nuclear pore proteins hence its role in nuclear-cytosolic transport was discovered. In addition to nuclear-cytosolic transport, CRM1 also plays a role in centrosome duplication and spindle assembly, especially in response to DNA damage. The crystal structure of CRM1 suggests a complex protein that binds the Ran protein bound to GTP, allowing for a conformational change that facilitates binding to different cargo proteins through a nuclear export signal (NES). Included in the cadre of cargo are multiple tumor suppressor and oncoproteins as p53, BRCA1, Survivin, NPM, and APC, which function in the nucleus to regulate transcription or aid in chromosomal assembly and movement. An imbalance in the cytosolic level of these proteins has been observed in cancer cells, resulting in either inactivation (tumor suppressor) or an excess of anti-apoptotic activity (oncoprotein). Thus, the concept of inhibiting CRM1 has been explored as a potential therapeutic intervention. Indeed, inhibition of CRM1 by a variety of small molecules that interfere with cargo-NES binding results in cancer cell death. Whether all of these proteins together are responsible for this phenotype or whether specific proteins are required for this effect is unclear at this time.

10.
Int J Biochem Mol Biol ; 3(1): 28-35, 2012.
Article in English | MEDLINE | ID: mdl-22509478

ABSTRACT

Neuroblastoma is the most common solid tumor of infancy, accounting for 15% of all cancer cell deaths in children. Expression of the anti-apoptotic protein survivin in these tumors correlates with poor prognostic features and resistance to therapy. The mammalian target of rapamycin (mTOR) protein is being explored as a potential therapeutic target in patients with this disease. The objective of this study was to test the hypothesis that rapamycin regulates survivin expression and function in neuroblastoma cells. To explore this hypothesis, we treated two different neuroblastoma lines (NB7, NB8) and a well-characterized control lung cancer cell line, A549, with varying doses of rapamycin (0.1-10µM) for serial time points (2-48 hours). RNA and protein expression levels were then evaluated by quantitative RT-PCR and western blotting, respectively. Cell proliferation and apoptosis were assayed by WST-1 and Annexin V. The results showed a rapamycin-dependent increase in survivin mRNA and protein levels in the neuroblastoma cell lines in a dose- and time-dependent fashion, while a decrease in these levels was observed in control cells. Rapamycin inhibited cell proliferation in both A549 and neuroblastoma cells however neuroblastoma cells had less apoptosis than A549 cells (9% vs. 20%). In summary, our results indicate that rapamycin induces expression of the anti-apoptotic protein survivin in neuroblastoma cells which may protect these cells from programmed cell death. Induction of survivin by rapamycin could therefore be a potential mechanism of neuroblastoma tumor cell resistance and rapamycin may not be an effective therapeutic agent for these tumors.

11.
Hum Pathol ; 43(6): 865-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22055399

ABSTRACT

Global gene expression profiling studies led to the recent classification of breast cancer into 4 distinct molecular subtypes including luminal, human epidermal growth factor receptor 2 enriched, basal like, and unclassified. Here, we used immunohistochemistry to evaluate expression of the antiapoptotic protein Survivin and its recently described acetylated form, Survivin acetyl129, in normal breast tissue and in 226 primary breast tumors of different molecular subtypes. Correlation of Survivin expression with molecular markers and its impact on patient outcomes were analyzed. Eighty-four percent of basal-like tumors expressed high levels of total Survivin, whereas 52% of luminal tumors expressed high levels of acetylated Survivin (P < .001). Overall survival (91%) for tumors expressing low levels of total Survivin was better than that for tumors expressing high levels of total Survivin (72%, P = .02), whereas the reverse was true for tumors expressing acetylated Survivin. In hierarchical cluster analysis, total Survivin clustered with basal marker expression, whereas acetylated Survivin clustered with luminal marker expression. In multivariate analysis, high total Survivin expression was an independent predictor of worse overall survival in patients with breast cancer (relative risk, 11; P < .01). These data indicate that high levels of total Survivin predict poor outcome in patients with grade 3 invasive ductal carcinoma and correlate directly with a basal-like phenotype. In contrast, high expression of the acetylated form of the protein associates with a favorable outcome and preferentially correlates with luminal-type tumors. Survivin likely has different functions in distinct breast cancer subtypes, and diagnostic strategies that incorporate immunohistochemical markers that detect both Survivin forms may help better strategize patient risk and direct therapy.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Inhibitor of Apoptosis Proteins/biosynthesis , Acetylation , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Female , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins/genetics , Kaplan-Meier Estimate , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Survivin , Tissue Array Analysis
12.
Diabetes ; 57(10): 2718-27, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18599523

ABSTRACT

OBJECTIVE: Diabetes results from a deficiency of functional beta-cells due to both an increase in beta-cell death and an inhibition of beta-cell replication. The molecular mechanisms responsible for these effects in susceptible individuals are mostly unknown. The objective of this study was to determine whether a gene critical for cell division and cell survival in cancer cells, survivin, might also be important for beta-cells. RESEARCH DESIGN AND METHODS: We generated mice harboring a conditional deletion of survivin in pancreatic endocrine cells using mice with a Pax-6-Cre transgene promoter construct driving tissue-specific expression of Cre-recombinase in these cells. We performed metabolic studies and immunohistochemical analyses to determine the effects of a mono- and biallelic deletion of survivin. RESULTS: Selective deletion of survivin in pancreatic endocrine cells in the mouse had no discernible effects during embryogenesis but was associated with striking decreases in beta-cell number after birth, leading to hyperglycemia and early-onset diabetes by 4 weeks of age. Serum insulin levels were significantly decreased in animals lacking endocrine cell survivin, with relative stability of other hormones. Exogenous expression of survivin in mature beta-cells lacking endogenous survivin completely rescued the hyperglycemic phenotype and the decrease in beta-cell mass, confirming the specificity of the survivin effect in these cells. CONCLUSIONS: Our findings implicate survivin in the maintenance of beta-cell mass through both replication and antiapoptotic mechanisms. Given the widespread involvement of survivin in cancer, a novel role for survivin may well be exploited in beta-cell regulation in diseased states, such as diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Microtubule-Associated Proteins/physiology , Animals , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Gene Deletion , Glucagon/metabolism , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Insulin/metabolism , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Repressor Proteins , Somatostatin/metabolism , Survivin
13.
Blood ; 109(4): 1479-89, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17038538

ABSTRACT

The identification of alternative splice variants of Survivin that possess distinct functions from those originally identified for the main Survivin isoform has greatly increased the complexity of our understanding of the role of Survivin in different cells. Previous functional studies of the Survivin splice variants have been performed almost exclusively in cancer cells. However, Survivin has increasingly been implicated in other normal physiologic and pathophysiologic processes, including angiogenesis. In this study, we dissect the involvement of Survivin DeltaEx3 in angiogenesis. We show by confocal microscopy that a pool of endothelial Survivin DeltaEx3 is localized to membrane ruffles. We also demonstrate that Survivin DeltaEx3 is the Survivin splice variant responsible for modulating angiogenesis in vitro, in tube formation assays, and in vivo, in an in vivo angiogenesis assay. Our data indicate that Survivin DeltaEx3 may regulate angiogenesis via several mechanisms including cell invasion, migration, and Rac1 activation. Our findings identify a novel pathway regulating angiogenesis through Survivin DeltaEx3 and a novel mechanism for Rac1 activation during angiogenesis. In conclusion, our results provide new insights into the regulation of endothelial cell homeostasis and angiogenesis by the Survivin proteins.


Subject(s)
Alternative Splicing , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neovascularization, Physiologic , Cell Movement , Cells, Cultured , Humans , Inhibitor of Apoptosis Proteins , Protein Isoforms , Sequence Deletion , Survivin , rac1 GTP-Binding Protein/metabolism
14.
J Biol Chem ; 279(39): 40511-20, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15271987

ABSTRACT

Survivin is a putative oncogene that is aberrantly expressed in cancer cells. It has been hypothesized to play a central role in cancer progression and resistance to therapy in diverse tumor types. Although some of the transcriptional processes regulating its expression have been established, the diversity of genes that may be controlling the levels of its expression in both normal cells as well as in cancer cells has not been fully explored. The most common genetically mutated pathways in human malignancies are the p53 tumor suppressor pathway and the RB/E2F pathway. Both of these pathways, when intact, provide essential checkpoints in the maintenance of normal cell growth and protect the cell from DNA damage. Using non-transformed embryonic fibroblasts, we provide evidence of a molecular link between the regulation of survivin transcription and the RB/E2F family of proteins. We demonstrate that both pRB and p130 can interact with the survivin promoter and can repress survivin transcription. We also show that the E2F activators (E2F1, E2F2, and E2F3) can bind to the survivin promoter and induce survivin transcription. Genetically modified cells that harbor deletions in various members of the RB/E2F family confirm our data from the wild-type cells. Our findings implicate several members of the RB/E2F pathway in an intricate mechanism of survivin gene regulation that, when genetically altered during the process of tumorigenesis, may function within cancer cells to aberrantly alter survivin levels and enhance tumor progression.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation , Microtubule-Associated Proteins/biosynthesis , Retinoblastoma Protein/physiology , Transcription Factors/physiology , Adenoviridae/genetics , Animals , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Line , Cells, Cultured , Chromatin/metabolism , DNA Damage , Disease Progression , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F3 Transcription Factor , Fibroblasts/metabolism , Gene Deletion , Genes, Reporter , Humans , Inhibitor of Apoptosis Proteins , Mice , Microtubule-Associated Proteins/metabolism , Mutation , Neoplasm Proteins , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survivin , Time Factors , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL