28 results on '"Elenbaas B"'
Search Results
2. The MDM2 oncoprotein binds specifically to RNA through its RING finger domain
- Author
-
Elenbaas B, Matthias Dobbelstein, Roth J, Shenk T, and Aj, Levine
- Subjects
Ribosomal Proteins ,Base Sequence ,Molecular Sequence Data ,RNA, Ribosomal, 5S ,Nuclear Proteins ,Proto-Oncogene Proteins c-mdm2 ,Zinc Fingers ,Neoplasm Proteins ,Proto-Oncogene Proteins ,Poly G ,Humans ,Nucleic Acid Conformation ,Point Mutation ,RNA ,Amino Acid Sequence ,Research Article ,Sequence Deletion - Abstract
BACKGROUND: The cellular mdm2 gene has transforming activity when overexpressed and is amplified in a variety of human tumors. At least part of the transforming ability of the MDM2 protein is due to binding and inactivating the p53 tumor suppressor protein. Additionally, this protein forms a complex in vivo with the L5 ribosomal protein and its associated 5S ribosomal RNA and may be part of a ribosomal complex. MATERIALS AND METHODS: A RNA homopolymer binding assay and a SELEX procedure have been used to characterize the RNA-binding activity of MDM2. RESULTS: The MDM2 protein binds efficiently to the homopolyribonucleotide poly(G) but not to other homopolyribonucleotides. This binding is independent of the interaction of MDM2 with the L5 protein, which occurs through the central acidic domain of MDM2. An RNA SELEX procedure was performed to identify specific RNA ligands that bind with high affinity to the human MDM2 (HDM2) protein. After 10 rounds of selection and amplification, a subset of RNA molecules that bound efficiently to HDM2 was isolated from a randomized pool. Sequencing of these selected ligands revealed that a small number of sequence motifs were selected. The specific RNA binding occurs through the RING finger domain of the protein. Furthermore, a single amino acid substitution in the RING finger domain, G446S, completely abolishes the specific RNA binding. CONCLUSIONS: These observations, showing that MDM2 binds the L5/5S ribosomal ribonucleoprotein particle and can also bind to specific RNA sequences or structures, suggest a role for MDM2 in translational regulation in a cell.
- Published
- 1996
3. 105 BIIB024, a potent pan-Raf kinase inhibitor for melanoma and solid tumors
- Author
-
Elenbaas, B., primary, Singh, L., additional, Boccia, A., additional, Cullen, P., additional, Peng, H., additional, Rohde, E., additional, Raimundo, B., additional, Kumaravel, G., additional, and Joseph, I., additional
- Published
- 2010
- Full Text
- View/download PDF
4. NAVOCEANO survey 2010 vision and reality….How close are they?
- Author
-
Reed, B, primary, Elenbaas, B, additional, Smith, G, additional, McCosh, L, additional, Wahl, R, additional, Duvieilh, T, additional, and Wagstaff, M, additional
- Published
- 2010
- Full Text
- View/download PDF
5. Rules governing the creation of human tumor cells
- Author
-
Hahn, W.C., primary, Elenbaas, B., additional, Stewart, S., additional, Watnick, R., additional, Yang, J., additional, Mani, S.A., additional, and Weinberg, R.A., additional
- Published
- 2001
- Full Text
- View/download PDF
6. The carboxyl-terminal domain of the p53 protein regulates sequence-specific DNA binding through its nonspecific nucleic acid-binding activity.
- Author
-
Bayle, J H, primary, Elenbaas, B, additional, and Levine, A J, additional
- Published
- 1995
- Full Text
- View/download PDF
7. Alternatively spliced forms in the carboxy-terminal domain of the p53 protein regulate its ability to promote annealing of complementary single strands of nucleic acids
- Author
-
Wu, L, primary, Bayle, J H, additional, Elenbaas, B, additional, Pavletich, N P, additional, and Levine, A J, additional
- Published
- 1995
- Full Text
- View/download PDF
8. The ribosomal L5 protein is associated with mdm-2 and mdm-2-p53 complexes
- Author
-
Marechal, V, primary, Elenbaas, B, additional, Piette, J, additional, Nicolas, J C, additional, and Levine, A J, additional
- Published
- 1994
- Full Text
- View/download PDF
9. Several hydrophobic amino acids in the p53 amino-terminal domain are required for transcriptional activation, binding to mdm-2 and the adenovirus 5 E1B 55-kD protein.
- Author
-
Lin, J, primary, Chen, J, additional, Elenbaas, B, additional, and Levine, A J, additional
- Published
- 1994
- Full Text
- View/download PDF
10. Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells.
- Author
-
Elenbaas, B, Spirio, L, Koerner, F, Fleming, M D, Zimonjic, D B, Donaher, J L, Popescu, N C, Hahn, W C, and Weinberg, R A
- Abstract
A number of genetic mutations have been identified in human breast cancers, yet the specific combinations of mutations required in concert to form breast carcinoma cells remain unknown. One approach to identifying the genetic and biochemical alterations required for this process involves the transformation of primary human mammary epithelial cells (HMECs) to carcinoma cells through the introduction of specific genes. Here we show that introduction of three genes encoding the SV40 large-T antigen, the telomerase catalytic subunit, and an H-Ras oncoprotein into primary HMECs results in cells that form tumors when transplanted subcutaneously or into the mammary glands of immunocompromised mice. The tumorigenicity of these transformed cells was dependent on the level of ras oncogene expression. Interestingly, transformation of HMECs but not two other human cell types was associated with amplifications of the c-myc oncogene, which occurred during the in vitro growth of the cells. Tumors derived from the transformed HMECs were poorly differentiated carcinomas that infiltrated through adjacent tissue. When these cells were injected subcutaneously, tumors formed in only half of the injections and with an average latency of 7.5 weeks. Mixing the epithelial tumor cells with Matrigel or primary human mammary fibroblasts substantially increased the efficiency of tumor formation and decreased the latency of tumor formation, demonstrating a significant influence of the stromal microenvironment on tumorigenicity. Thus, these observations establish an experimental system for elucidating both the genetic and cell biological requirements for the development of breast cancer.
- Published
- 2001
11. Essential function of Wnt-4 in mammary gland development downstream of progesterone signaling.
- Author
-
Brisken, C, Heineman, A, Chavarria, T, Elenbaas, B, Tan, J, Dey, S K, McMahon, J A, McMahon, A P, and Weinberg, R A
- Abstract
Female reproductive hormones control mammary gland morphogenesis. In the absence of the progesterone receptor (PR) from the mammary epithelium, ductal side-branching fails to occur. We can overcome this defect by ectopic expression of the protooncogene Wnt-1. Transplantation of mammary epithelia from Wnt-4(-)/(-) mice shows that Wnt-4 has an essential role in side-branching early in pregnancy. PR and Wnt-4 mRNAs colocalize to the luminal compartment of the ductal epithelium. Progesterone induces Wnt-4 in mammary epithelial cells and is required for increased Wnt-4 expression during pregnancy. Thus, Wnt signaling is essential in mediating progesterone function during mammary gland morphogenesis.
- Published
- 2000
12. Essential function of Wnt-4 in mammary gland development downstream of progesterone signaling
- Author
-
Brisken, C., Heineman, A., Chavarria, T., Elenbaas, B., Tan, J., Dey, S. K., McMahon, J. A., McMahon, A. P., and Weinberg, R. A.
- Abstract
Female reproductive hormones control mammary gland morphogenesis. In the absence of the progesterone receptor (PR) from the mammary epithelium, ductal side-branching fails to occur. We can overcome this defect by ectopic expression of the protooncogene Wnt-1. Transplantation of mammary epithelia from Wnt-4(-)/(-) mice shows that Wnt-4 has an essential role in side-branching early in pregnancy. PR and Wnt-4 mRNAs colocalize to the luminal compartment of the ductal epithelium. Progesterone induces Wnt-4 in mammary epithelial cells and is required for increased Wnt-4 expression during pregnancy. Thus, Wnt signaling is essential in mediating progesterone function during mammary gland morphogenesis.
13. Author Correction: ATR inhibition augments the efficacy of lurbinectedin in small-cell lung cancer.
- Author
-
Schultz CW, Zhang Y, Elmeskini R, Zimmermann A, Fu H, Murai Y, Wangsa D, Kumar S, Takahashi N, Atkinson D, Saha LK, Lee CF, Elenbaas B, Desai P, Sebastian R, Sharma AK, Abel M, Schroeder B, Krishnamurthy M, Bassel LL, Kumar R, Roper N, Aladjem M, Zenke FT, Ohler ZW, Pommier Y, and Thomas A
- Published
- 2024
- Full Text
- View/download PDF
14. Relationship among DDR gene mutations, TMB and PD-L1 in solid tumour genomes identified using clinically actionable biomarker assays.
- Author
-
Wang D, Elenbaas B, Murugesan K, Shah K, Montesion M, Gounaris I, Scheuenpflug J, Locatelli G, and Feng Z
- Abstract
The DNA damage response (DDR) pathway regulates DNA repair and cell survival, and inactivating mutations in DDR genes can increase tumour mutational burden (TMB), a predictive biomarker of treatment benefit from anti-PD-1/PD-L1 immunotherapies. However, a better understanding of the relationship among specific DDR mutations, TMB and PD-L1 expression is needed to improve translational strategies. Here, we determined genomic alteration frequencies in selected DDR genes that are clinically actionable biomarkers and investigated their association with TMB and PD-L1 in bladder, colorectal, non-small cell lung, ovarian and prostate cancers using the FoundationInsights
® web portal. Our results not only confirm known associations, such as mismatch repair and POLE gene mutations with high TMB, but also identify significant associations between mutations in the SWI/SNF chromatin remodelling genes ARID1A and SMARCA4 and high TMB in multiple tumour types. Mutations in the ATR gene were associated with high TMB in colorectal and prostate cancers; however, associations between individual DDR mutations and high PD-L1 expression were uncommon and tumour-type specific. Finally, we found that high TMB and high PD-L1 expression were poorly associated, emphasising their independence as predictive biomarkers for immune checkpoint inhibitor use., (© 2023. Nature Publishing Group UK.)- Published
- 2023
- Full Text
- View/download PDF
15. p70S6K/Akt dual inhibitor DIACC3010 is efficacious in preclinical models of gastric cancer alone and in combination with trastuzumab.
- Author
-
Fukuoka S, Koga Y, Yamauchi M, Koganemaru S, Yasunaga M, Shitara K, Doi T, Yoshino T, Kuronita T, Elenbaas B, Wahra P, Zhang H, Crowley L, Jenkins MH, Clark A, and Kojima T
- Subjects
- Animals, Mice, Humans, Ribosomal Protein S6 Kinases, 70-kDa, Proto-Oncogene Proteins c-akt, Phosphatidylinositol 3-Kinases, Protein Kinase Inhibitors, Angiogenesis Inhibitors, Disease Models, Animal, Stomach Neoplasms drug therapy
- Abstract
The PI3K-Akt-mTOR (PAM) pathway is implicated in tumor progression in many tumor types, including metastatic gastric cancer (GC). The initial promise of PAM inhibitors has been unrealized in the clinic, presumably due, in part, to the up-regulation of Akt signaling that occurs when the pathway is inhibited. Here we present that DIACC3010 (formerly M2698), an inhibitor of two nodes in the PAM pathway, p70S6K and Akt 1/3, blocks the pathway in in vitro and in vivo preclinical models of GC while providing a mechanism that inhibits signaling from subsequent Akt up-regulation. Utilizing GC cell lines and xenograft models, we identified potential markers of DIACC3010-sensitivity in Her2-negative tumors, i.e., PIK3CA mutations, low basal pERK, and a group of differentially expressed genes (DEGs). The combination of DIACC3010 and trastuzumab was evaluated in Her2-positive cell lines and models. Potential biomarkers for the synergistic efficacy of the combination of DIACC3010 + trastuzumab also included DEGs as well as a lack of up-regulation of pERK. Of 27 GC patient-derived xenograft (PDX) models tested in BALB/c nu/nu mice, 59% were sensitive to DIACC3010 + trastuzumab. Of the 21 HER2-negative PDX models, DIACC3010 significantly inhibited the growth of 38%. Altogether, these results provide a path forward to validate the potential biomarkers of DIACC3010 sensitivity in GC and support clinical evaluation of DIACC3010 monotherapy and combination with trastuzumab in patients with HER2- negative and positive advanced GCs, respectively., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
16. ATR inhibition augments the efficacy of lurbinectedin in small-cell lung cancer.
- Author
-
Schultz CW, Zhang Y, Elmeskini R, Zimmermann A, Fu H, Murai Y, Wangsa D, Kumar S, Takahashi N, Atkinson D, Saha LK, Lee CF, Elenbaas B, Desai P, Sebastian R, Sharma AK, Abel M, Schroeder B, Krishnamurthy M, Kumar R, Roper N, Aladjem M, Zenke FT, Ohler ZW, Pommier Y, and Thomas A
- Subjects
- Humans, Neoplasm Recurrence, Local, Ataxia Telangiectasia Mutated Proteins metabolism, Small Cell Lung Carcinoma drug therapy, Lung Neoplasms pathology, Carcinoma, Non-Small-Cell Lung drug therapy
- Abstract
Small-cell lung cancer (SCLC) is the most lethal type of lung cancer. Specifically, MYC-driven non-neuroendocrine SCLC is particularly resistant to standard therapies. Lurbinectedin was recently approved for the treatment of relapsed SCLC, but combinatorial approaches are needed to increase the depth and duration of responses to lurbinectedin. Using high-throughput screens, we found inhibitors of ataxia telangiectasia mutated and rad3 related (ATR) as the most effective agents for augmenting lurbinectedin efficacy. First-in-class ATR inhibitor berzosertib synergized with lurbinectedin in multiple SCLC cell lines, organoid, and in vivo models. Mechanistically, ATR inhibition abrogated S-phase arrest induced by lurbinectedin and forced cell cycle progression causing mitotic catastrophe and cell death. High CDKN1A/p21 expression was associated with decreased synergy due to G1 arrest, while increased levels of ERCC5/XPG were predictive of increased combination efficacy. Importantly, MYC-driven non-neuroendocrine tumors which are resistant to first-line therapies show reduced CDKN1A/p21 expression and increased ERCC5/XPG indicating they are primed for response to lurbinectedin-berzosertib combination. The combination is being assessed in a clinical trial NCT04802174., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.)
- Published
- 2023
- Full Text
- View/download PDF
17. Selective Inhibition of ATM-dependent Double-strand Break Repair and Checkpoint Control Synergistically Enhances the Efficacy of ATR Inhibitors.
- Author
-
Turchick A, Zimmermann A, Chiu LY, Dahmen H, Elenbaas B, Zenke FT, Blaukat A, and Vassilev LT
- Subjects
- Humans, Ataxia Telangiectasia Mutated Proteins, DNA Repair, Cell Cycle Proteins metabolism, Protein Kinase Inhibitors pharmacology, DNA Damage, Checkpoint Kinase 1 genetics, Tumor Suppressor Protein p53 genetics, Ataxia Telangiectasia
- Abstract
Ataxia telangiectasia and Rad3-related protein (ATR) kinase regulate a key cell regulatory node for maintaining genomic integrity by preventing replication fork collapse. ATR inhibition has been shown to increase replication stress resulting in DNA double-strand breaks (DSBs) and cancer cell death, and several inhibitors are under clinical investigation for cancer therapy. However, activation of cell-cycle checkpoints controlled by ataxia telangiectasia-mutated (ATM) kinase could minimize the lethal consequences of ATR inhibition and protect cancer cells. Here, we investigate ATR-ATM functional relationship and potential therapeutic implications. In cancer cells with functional ATM and p53 signaling, selective suppression of ATR catalytic activity by M6620 induced G1-phase arrest to prevent S-phase entry with unrepaired DSBs. The selective ATM inhibitors, M3541 and M4076, suppressed both ATM-dependent cell-cycle checkpoints, and DSB repair lowered the p53 protective barrier and extended the life of ATR inhibitor-induced DSBs. Combination treatment amplified the fraction of cells with structural chromosomal defects and enhanced cancer cell death. ATM inhibitor synergistically potentiated the ATR inhibitor efficacy in cancer cells in vitro and increased ATR inhibitor efficacy in vivo at doses that did not show overt toxicities. Furthermore, a combination study in 26 patient-derived xenograft models of triple-negative breast cancer with the newer generation ATR inhibitor M4344 and ATM inhibitor M4076 demonstrated substantial improvement in efficacy and survival compared with single-agent M4344, suggesting a novel and potentially broad combination approach to cancer therapy., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
18. Therapeutic targeting of ATR yields durable regressions in small cell lung cancers with high replication stress.
- Author
-
Thomas A, Takahashi N, Rajapakse VN, Zhang X, Sun Y, Ceribelli M, Wilson KM, Zhang Y, Beck E, Sciuto L, Nichols S, Elenbaas B, Puc J, Dahmen H, Zimmermann A, Varonin J, Schultz CW, Kim S, Shimellis H, Desai P, Klumpp-Thomas C, Chen L, Travers J, McKnight C, Michael S, Itkin Z, Lee S, Yuno A, Lee MJ, Redon CE, Kindrick JD, Peer CJ, Wei JS, Aladjem MI, Figg WD, Steinberg SM, Trepel JB, Zenke FT, Pommier Y, Khan J, and Thomas CJ
- Subjects
- Aged, Antineoplastic Agents pharmacology, Ataxia Telangiectasia Mutated Proteins genetics, DNA Replication drug effects, DNA Topoisomerases, Type I genetics, Genomic Instability genetics, Humans, Lung Neoplasms metabolism, Middle Aged, Neoplasm Recurrence, Local metabolism, Protein Kinase Inhibitors pharmacology, Signal Transduction drug effects, Small Cell Lung Carcinoma metabolism, Ataxia Telangiectasia Mutated Proteins metabolism, Isoxazoles pharmacology, Lung Neoplasms drug therapy, Neoplasm Recurrence, Local drug therapy, Pyrazines pharmacology, Small Cell Lung Carcinoma drug therapy
- Abstract
Small cell neuroendocrine cancers (SCNCs) are recalcitrant cancers arising from diverse primary sites that lack effective treatments. Using chemical genetic screens, we identified inhibition of ataxia telangiectasia and rad3 related (ATR), the primary activator of the replication stress response, and topoisomerase I (TOP1), nuclear enzyme that suppresses genomic instability, as synergistically cytotoxic in small cell lung cancer (SCLC). In a proof-of-concept study, we combined M6620 (berzosertib), first-in-class ATR inhibitor, and TOP1 inhibitor topotecan in patients with relapsed SCNCs. Objective response rate among patients with SCLC was 36% (9/25), achieving the primary efficacy endpoint. Durable tumor regressions were observed in patients with platinum-resistant SCNCs, typically fatal within weeks of recurrence. SCNCs with high neuroendocrine differentiation, characterized by enhanced replication stress, were more likely to respond. These findings highlight replication stress as a potentially transformative vulnerability of SCNCs, paving the way for rational patient selection in these cancers, now treated as a single disease., Competing Interests: Declaration of interests H.D., A.Z., and F.T.Z. are employees of Merck KGaA, Darmstadt, Germany. B.E. is an employee of the EMD Serono Research & Development Institute Inc., Billerica, MA, USA; a business of Merck KGaA, Darmstadt, Germany. J.P. was an employee of the EMD Serono Research & Development Institute Inc., Billerica, MA, USA at the time of study. A.T., and Y.P. report research funding to the institution from the following entities: EMD Serono Research & Development Institute Inc., Billerica, MA, USA, AstraZeneca, Tarveda Therapeutics, and Prolynx Inc., (Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
19. MEK inhibition leads to BRCA2 downregulation and sensitization to DNA damaging agents in pancreas and ovarian cancer models.
- Author
-
Vena F, Jia R, Esfandiari A, Garcia-Gomez JJ, Rodriguez-Justo M, Ma J, Syed S, Crowley L, Elenbaas B, Goodstal S, Hartley JA, and Hochhauser D
- Abstract
Targeting the DNA damage response (DDR) in tumors with defective DNA repair is a clinically successful strategy. The RAS/RAF/MEK/ERK signalling pathway is frequently deregulated in human cancers. In this study, we explored the effects of MEK inhibition on the homologous recombination pathway and explored the potential for combination therapy of MEK inhibitors with DDR inhibitors and a hypoxia-activated prodrug. We studied effects of combining pimasertib, a selective allosteric inhibitor of MEK1/2, with olaparib, a small molecule inhibitor of poly (adenosine diphosphate [ADP]-ribose) polymerases (PARP), and with the hypoxia-activated prodrug evofosfamide in ovarian and pancreatic cancer cell lines. Apoptosis was assessed by Caspase 3/7 assay and protein expression was detected by immunoblotting. DNA damage response was monitored with γH2AX and RAD51 immunofluorescence staining. In vivo antitumor activity of pimasertib with evofosfamide were assessed in pancreatic cancer xenografts. We found that BRCA2 protein expression was downregulated following pimasertib treatment under hypoxic conditions. This translated into reduced homologous recombination repair demonstrated by levels of RAD51 foci. MEK inhibition was sufficient to induce formation of γH2AX foci, suggesting that inhibition of this pathway would impair DNA repair. When combined with olaparib or evofosfamide, pimasertib treatment enhanced DNA damage and increased apoptosis. The combination of pimasertib with evofosfamide demonstrated increased anti-tumor activity in BRCA wild-type Mia-PaCa-2 xenograft model, but not in the BRCA mutated BxPC3 model. Our data suggest that targeted MEK inhibition leads to impaired homologous recombination DNA damage repair and increased PARP inhibition sensitivity in BRCA-2 proficient cancers., Competing Interests: CONFLICTS OF INTEREST This research was funded by an educational grant from EMD Serono Research and Development Institute.
- Published
- 2018
- Full Text
- View/download PDF
20. Structure-based design of low-nanomolar PIM kinase inhibitors.
- Author
-
Ishchenko A, Zhang L, Le Brazidec JY, Fan J, Chong JH, Hingway A, Raditsis A, Singh L, Elenbaas B, Hong VS, Marcotte D, Silvian L, Enyedy I, and Chao J
- Subjects
- Binding Sites, Crystallography, X-Ray, Hydrogen Bonding, Molecular Dynamics Simulation, Protein Binding, Protein Isoforms antagonists & inhibitors, Protein Isoforms metabolism, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors metabolism, Protein Structure, Tertiary, Proto-Oncogene Proteins c-pim-1 metabolism, Static Electricity, Structure-Activity Relationship, Drug Design, Protein Kinase Inhibitors chemistry, Proto-Oncogene Proteins c-pim-1 antagonists & inhibitors
- Abstract
PIM kinases are implicated in variety of cancers by promoting cell survival and proliferation and are targets of interest for therapeutic intervention. We have identified a low-nanomolar pan-PIM inhibitor (PIM1/2/3 potency 5:14:2nM) using structure based modeling. The crystal structure of this compound with PIM1 confirmed the predicted binding mode and protein-ligand interactions except those in the acidic ribose pocket. We show the SAR suggesting the importance of having a hydrogen bond donor in this pocket for inhibiting PIM2; however, this interaction is not important for inhibiting PIM1 or PIM3. In addition, we report the discovery of a new class of PIM inhibitors by using computational de novo design tool implemented in MOE software (Chemical Computing Group). These inhibitors have a different interaction profile., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
21. Identification of SFRP1 as a candidate mediator of stromal-to-epithelial signaling in prostate cancer.
- Author
-
Joesting MS, Perrin S, Elenbaas B, Fawell SE, Rubin JS, Franco OE, Hayward SW, Cunha GR, and Marker PC
- Subjects
- Animals, Apoptosis genetics, Cell Growth Processes genetics, Disease Progression, Epithelial Cells metabolism, Epithelial Cells pathology, Fibroblasts metabolism, Fibroblasts pathology, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Intercellular Signaling Peptides and Proteins biosynthesis, Male, Membrane Proteins biosynthesis, Mice, Prostatic Neoplasms metabolism, Signal Transduction genetics, Stromal Cells metabolism, Stromal Cells pathology, Transfection, Tumor Cells, Cultured, Wnt Proteins biosynthesis, Wnt Proteins genetics, beta Catenin biosynthesis, beta Catenin genetics, Intercellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology
- Abstract
Genetic changes in epithelial cells initiate the development of prostatic adenocarcinomas. As nascent tumors grow and undergo progression, epithelial tumor cells are intimately associated with stromal cells. Stromal cells within the tumor microenvironment acquire new properties, including the capacity to promote phenotypic and genetic progression in adjacent epithelial cells. Affymetrix microarrays were used to identify 119 genes differentially expressed between normal-derived and carcinoma-derived prostatic stromal cells. These included 31 genes encoding extracellular proteins that may act as stromal-to-epithelial paracrine signals. Further investigation of one of these genes, secreted frizzled related protein 1 (SFRP1), revealed that its expression parallels prostatic growth with high expression during prostatic development, low expression in the adult prostate, and elevated expression in prostatic tumor stroma. In addition, as prostatic epithelial cells progressed to a tumorigenic state under the influence of tumor stroma, SFRP1 became overexpressed in the progressed epithelial cells. To further understand the roles of SFRP1 in the prostate, we tested the affects of increased SFRP1 levels on prostatic tissues and cells. Treatment of developing prostates with SFRP1 in culture led to increased organ growth. Treatment of a human prostatic epithelial cell line with SFRP1 led to increased proliferation, decreased apoptosis, and decreased signaling through the Wnt/beta-catenin pathway in vitro and increased proliferation in vivo. These data suggest that overexpression of SFRP1 by prostatic tumor stroma may account for the previously reported capacity of prostatic tumor stroma to provide a pro-proliferative paracrine signal to adjacent epithelial cells.
- Published
- 2005
- Full Text
- View/download PDF
22. Immortalization and transformation of primary human airway epithelial cells by gene transfer.
- Author
-
Lundberg AS, Randell SH, Stewart SA, Elenbaas B, Hartwell KA, Brooks MW, Fleming MD, Olsen JC, Miller SW, Weinberg RA, and Hahn WC
- Subjects
- Blotting, Western, Cell Differentiation, Cell Division, Cell Line, Transformed, Cells, Cultured, DNA-Binding Proteins, Genes, ras genetics, Genetic Vectors, Humans, Simian virus 40 genetics, Telomerase genetics, Telomerase metabolism, Time Factors, Bronchi metabolism, Bronchi pathology, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Epithelial Cells metabolism, Epithelial Cells pathology, Transduction, Genetic
- Abstract
One critical step in the development of a cancerous cell is its acquisition of an unlimited replicative lifespan, the process termed immortalization. Experimental model systems designed to study cellular transformation ex vivo have relied to date on the in vitro selection of a subpopulation of cells that have become immortalized through treatment with chemical or physical mutagens and the selection of rare clonal variants. In this study, we describe the direct immortalization of primary human airway epithelial cells through the successive introduction of the Simian Virus 40 Early Region and the telomerase catalytic subunit hTERT. Cells immortalized in this way are now responsive to malignant transformation by an introduced H-ras or K-ras oncogene. These immortalized human airway epithelial cells, which have been created through the stepwise introduction of genetic alterations, provide a novel experimental model system with which to study further the biology of the airway epithelial cell and to dissect the molecular basis of lung cancer pathogenesis.
- Published
- 2002
- Full Text
- View/download PDF
23. Enumeration of the simian virus 40 early region elements necessary for human cell transformation.
- Author
-
Hahn WC, Dessain SK, Brooks MW, King JE, Elenbaas B, Sabatini DM, DeCaprio JA, and Weinberg RA
- Subjects
- Antigens, Viral, Tumor genetics, Cell Division, Cell Line, Cellular Senescence, DNA-Binding Proteins, Fibroblasts, Humans, Oncogene Protein p21(ras) genetics, Oncogene Protein p21(ras) metabolism, Retinoblastoma Protein antagonists & inhibitors, Retinoblastoma Protein metabolism, Signal Transduction, Simian virus 40 genetics, Telomerase genetics, Telomerase metabolism, Time Factors, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 metabolism, Antigens, Viral, Tumor metabolism, Cell Transformation, Neoplastic, Simian virus 40 physiology
- Abstract
While it is clear that cancer arises from the accumulation of genetic mutations that endow the malignant cell with the properties of uncontrolled growth and proliferation, the precise combinations of mutations that program human tumor cell growth remain unknown. The study of the transforming proteins derived from DNA tumor viruses in experimental models of transformation has provided fundamental insights into the process of cell transformation. We recently reported that coexpression of the simian virus 40 (SV40) early region (ER), the gene encoding the telomerase catalytic subunit (hTERT), and an oncogenic allele of the H-ras gene in normal human fibroblast, kidney epithelial, and mammary epithelial cells converted these cells to a tumorigenic state. Here we show that the SV40 ER contributes to tumorigenic transformation in the presence of hTERT and oncogenic H-ras by perturbing three intracellular pathways through the actions of the SV40 large T antigen (LT) and the SV40 small t antigen (ST). LT simultaneously disables the retinoblastoma (pRB) and p53 tumor suppressor pathways; however, complete transformation of human cells requires the additional perturbation of protein phosphatase 2A by ST. Expression of ST in this setting stimulates cell proliferation, permits anchorage-independent growth, and confers increased resistance to nutrient deprivation. Taken together, these observations define the elements of the SV40 ER required for the transformation of human cells and begin to delineate a set of intracellular pathways whose disruption, in aggregate, appears to be necessary to generate tumorigenic human cells.
- Published
- 2002
- Full Text
- View/download PDF
24. Heterotypic signaling between epithelial tumor cells and fibroblasts in carcinoma formation.
- Author
-
Elenbaas B and Weinberg RA
- Subjects
- Animals, Breast Neoplasms etiology, Breast Neoplasms metabolism, Carcinoma blood supply, Carcinoma metabolism, Cell Division, Extracellular Matrix Proteins metabolism, Female, Growth Substances biosynthesis, Humans, Matrix Metalloproteinases metabolism, Mice, Neovascularization, Pathologic, Paracrine Communication, Stromal Cells metabolism, Carcinoma etiology, Fibroblasts metabolism
- Abstract
Tumors arise from cells that have sustained genetic mutations resulting in deregulation of several of their normal growth-controlling mechanisms. Much of the research concerning the origins of cancer has focused on the genetic mutations within tumor cells, treating tumorigenesis as a cell-autonomous process governed by the genes carried by the tumor cells themselves. However, it is increasingly apparent that the stromal microenvironment in which the tumor cells develop profoundly influences many steps of tumor progression. In various experimental tumor models, the microenvironment affects the efficiency of tumor formation, the rate of tumor growth, the extent of invasiveness, and the ability of tumor cells to metastasize. In carcinomas, the influences of the microenvironment are mediated, in large part, by paracrine signaling between epithelial tumor cells and neighboring stromal fibroblasts. In this review, we summarize recent advances in understanding the paracrine signaling interactions between epithelial cancer cells and associated fibroblasts and examine the effects of these bidirectional interactions on various aspects of carcinoma formation. We note, however, that paracrine signaling between other cell types within the carcinomas, such as endothelial cells and inflammatory cells, may play equally important roles in tumor formation and we will refer to these heterotypic interactions where relevant., (Copyright 2001 Academic Press.)
- Published
- 2001
- Full Text
- View/download PDF
25. Conservation of structural domains and biochemical activities of the MDM2 protein from Xenopus laevis.
- Author
-
Marechal V, Elenbaas B, Taneyhill L, Piette J, Mechali M, Nicolas JC, Levine AJ, and Moreau J
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cloning, Molecular, Gene Expression Regulation, Developmental, Humans, Mice, Molecular Sequence Data, Protein Binding, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-mdm2, Ribosomal Proteins metabolism, Sequence Alignment, Sequence Homology, Amino Acid, Tumor Suppressor Protein p53 metabolism, Xenopus Proteins, Xenopus laevis embryology, Nuclear Proteins, Proto-Oncogene Proteins chemistry, Xenopus laevis genetics
- Abstract
The Mdm2 gene is the best known cellular regulator of p53 tumor suppressor activity. We report here the cloning and characterization of Xdm2, its homolog in Xenopus laevis. Human, mouse and Xenopus MDM2 proteins are more than 65% identical in several regions which are likely to be important for the biological activities of MDM2. Region I is sufficient for binding p53 and inhibiting its G1 arrest and apoptosis functions. Region II contains most of a central acidic region required for interaction with the L5 ribosomal protein and a putative C4 zinc finger. Region III is nearly identical from Xenopus to human and comprises the RING finger domain. We show that this structural conservation is associated with the conservation of three biochemical activities of MDM2; binding to the p53 and L5 proteins and specifically to RNA. Lastly, Xdm2 expression during early development is mainly restricted from the oocyte stage I/II to the blastula stage and is possibly independent of transcriptional activation by p53. These data as well as the utilization of Xenopus laevis to investigate the roles of MDM2 and p53 during early embryogenesis are discussed.
- Published
- 1997
- Full Text
- View/download PDF
26. Structure of the MDM2 oncoprotein bound to the p53 tumor suppressor transactivation domain.
- Author
-
Kussie PH, Gorina S, Marechal V, Elenbaas B, Moreau J, Levine AJ, and Pavletich NP
- Subjects
- Binding Sites, Crystallization, Crystallography, X-Ray, Hydrogen Bonding, Models, Molecular, Protein Binding, Protein Folding, Protein Structure, Secondary, Protein Structure, Tertiary, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-mdm2, Transcription Factors chemistry, Transcription Factors metabolism, Tumor Suppressor Protein p53 metabolism, Nuclear Proteins, Protein Conformation, Proto-Oncogene Proteins chemistry, Transcriptional Activation, Tumor Suppressor Protein p53 chemistry
- Abstract
The MDM2 oncoprotein is a cellular inhibitor of the p53 tumor suppressor in that it can bind the transactivation domain of p53 and downregulate its ability to activate transcription. In certain cancers, MDM2 amplification is a common event and contributes to the inactivation of p53. The crystal structure of the 109-residue amino-terminal domain of MDM2 bound to a 15-residue transactivation domain peptide of p53 revealed that MDM2 has a deep hydrophobic cleft on which the p53 peptide binds as an amphipathic alpha helix. The interface relies on the steric complementarity between the MDM2 cleft and the hydrophobic face of the p53 alpha helix and, in particular, on a triad of p53 amino acids-Phe19, Trp23, and Leu26-which insert deep into the MDM2 cleft. These same p53 residues are also involved in transactivation, supporting the hypothesis that MDM2 inactivates p53 by concealing its transactivation domain. The structure also suggests that the amphipathic alpha helix may be a common structural motif in the binding of a diverse family of transactivation factors to the TATA-binding protein-associated factors.
- Published
- 1996
- Full Text
- View/download PDF
27. p53 and its 14 kDa C-terminal domain recognize primary DNA damage in the form of insertion/deletion mismatches.
- Author
-
Lee S, Elenbaas B, Levine A, and Griffith J
- Subjects
- Base Composition, Base Sequence, Binding Sites, DNA isolation & purification, DNA ultrastructure, Electrophoresis, Polyacrylamide Gel, Humans, Microscopy, Electron, Molecular Sequence Data, Oligodeoxyribonucleotides, Peptide Fragments isolation & purification, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Recombinant Proteins ultrastructure, Templates, Genetic, Tumor Suppressor Protein p53 isolation & purification, Tumor Suppressor Protein p53 ultrastructure, DNA metabolism, DNA Damage, Mutagenesis, Insertional, Peptide Fragments metabolism, Sequence Deletion, Tumor Suppressor Protein p53 metabolism
- Abstract
Insertion/deletion (IDL) mismatches in DNA are lesions consisting of extra bases on one strand. Here, the binding of p53 and its 14 kDa C-terminal domain to DNAs containing one or three 3-cytosine IDL mismatches was examined. Electron microscopy showed that both p53 forms bound predominantly as tetramers at the lesions while single-stranded binding proteins did not bind. Gel retardation assays showed that p53 formed highly stable complexes when the DNA contained the IDL mismatches, but only unstable complexes when the DNA lacked lesions (but did contain free ends). The highly stable complexes had a half-life of > 2 hr, suggesting that upon encountering lesions, p53 may recruit other proteins to the site, providing a signal for DNA damage.
- Published
- 1995
- Full Text
- View/download PDF
28. Two upstream elements activate transcription of a major histocompatibility complex class I gene in vitro.
- Author
-
Driggers PH, Elenbaas BA, An JB, Lee IJ, and Ozato K
- Subjects
- Animals, Base Sequence, Binding Sites genetics, Cell Line, DNA-Binding Proteins genetics, Humans, Methylation, Mice, Molecular Sequence Data, NFI Transcription Factors, Nuclear Proteins, Transcription Factors genetics, Transfection genetics, Y-Box-Binding Protein 1, CCAAT-Enhancer-Binding Proteins, Enhancer Elements, Genetic genetics, Genes, MHC Class I genetics, Promoter Regions, Genetic genetics, Transcription, Genetic genetics
- Abstract
Expression of major histocompatibility complex (MHC) class I genes exhibits unique tissue and developmental specificity. In an effort to study molecular mechanisms of MHC class I gene regulation, an in vitro transcription system has been established. In B cell nuclear extracts a template DNA containing the mouse H-2Ld promoter sequence accurately directed RNA polymerase II-dependent transcription of a G-free cassette. A conserved class I regulatory complex previously shown to moderately enhance promoter activity in vivo enhanced transcription in vitro by 2-3 fold. Much of this enhancement was accounted for by a 40 bp fragment within the complex, which was capable of activating a basal H-2Ld promoter in either orientation. Farther downstream, another element called site B was identified, which independently activated MHC class I transcription in vitro by 2-4 fold. Site B bound a specific nuclear factor(s) through an NF-1 binding site but not through a neighboring CCAAT site. The functional significance of site B in vivo was demonstrated in transfection experiments in which site B enhanced MHC class I promoter activity to a degree comparable to that seen in vitro. With the identification of the two upstream activators, MHC class I genes may serve as a model to study roles of sequence-specific DNA-binding proteins in transcription in vitro.
- Published
- 1992
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.