801
|
Raver-Shapira N, Marciano E, Meiri E, Spector Y, Rosenfeld N, Moskovits N, Bentwich Z, Oren M. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell 2007; 26:731-43. [PMID: 17540598 DOI: 10.1016/j.molcel.2007.05.017] [Citation(s) in RCA: 1030] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 05/05/2007] [Accepted: 05/17/2007] [Indexed: 02/07/2023]
Abstract
p53 is a potent tumor suppressor, whose biological effects are largely due to its function as a transcriptional regulator. Here we report that, in addition to regulating the expression of hundreds of protein-coding genes, p53 also modulates the levels of microRNAs (miRNAs). Specifically, p53 can induce expression of microRNA-34a (miR-34a) in cultured cells as well as in irradiated mice, by binding to a perfect p53 binding site located within the gene that gives rise to miR-34a. Processing of the primary transcript into mature miR-34a involves the excision of a 30 kb intron. Notably, inactivation of miR-34a strongly attenuates p53-mediated apoptosis in cells exposed to genotoxic stress, whereas overexpression of miR-34a mildly increases apoptosis. Hence, miR-34a is a direct proapoptotic transcriptional target of p53 that can mediate some of p53's biological effects. Perturbation of miR-34a expression, as occurs in some human cancers, may thus contribute to tumorigenesis by attenuating p53-dependent apoptosis.
Collapse
Affiliation(s)
- Nina Raver-Shapira
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | |
Collapse
|
802
|
Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH, Feldmann G, Yamakuchi M, Ferlito M, Lowenstein CJ, Arking DE, Beer MA, Maitra A, Mendell JT. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell 2007; 26:745-52. [PMID: 17540599 PMCID: PMC1939978 DOI: 10.1016/j.molcel.2007.05.010] [Citation(s) in RCA: 1571] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 04/27/2007] [Accepted: 05/10/2007] [Indexed: 12/12/2022]
Abstract
The p53 tumor suppressor protein is a critical regulator of the cellular response to cancer-initiating insults such as genotoxic stress. In this report, we demonstrate that microRNAs (miRNAs) are important components of the p53 transcriptional network. Global miRNA expression analyses identified a cohort of miRNAs that exhibit p53-dependent upregulation following DNA damage. One such miRNA, miR-34a, is commonly deleted in human cancers and, as shown here, frequently absent in pancreatic cancer cells. Characterization of the miR-34a primary transcript and promoter demonstrates that this miRNA is directly transactivated by p53. Expression of miR-34a causes dramatic reprogramming of gene expression and promotes apoptosis. Much like the known set of p53-regulated genes, miR-34a-responsive genes are highly enriched for those that regulate cell-cycle progression, apoptosis, DNA repair, and angiogenesis. Therefore, it is likely that an important function of miR-34a is the modulation and fine-tuning of the gene expression program initiated by p53.
Collapse
Affiliation(s)
- Tsung-Cheng Chang
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Erik A. Wentzel
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Oliver A. Kent
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Kalyani Ramachandran
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Michael Mullendore
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Kwang Hyuck Lee
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Georg Feldmann
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Munekazu Yamakuchi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Marcella Ferlito
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Charles J. Lowenstein
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Dan E. Arking
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Michael A. Beer
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Anirban Maitra
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Joshua T. Mendell
- The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
803
|
Jariwala U, Prescott J, Jia L, Barski A, Pregizer S, Cogan JP, Arasheben A, Tilley WD, Scher HI, Gerald WL, Buchanan G, Coetzee GA, Frenkel B. Identification of novel androgen receptor target genes in prostate cancer. Mol Cancer 2007; 6:39. [PMID: 17553165 PMCID: PMC1904239 DOI: 10.1186/1476-4598-6-39] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Accepted: 06/06/2007] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The androgen receptor (AR) plays critical roles in both androgen-dependent and castrate-resistant prostate cancer (PCa). However, little is known about AR target genes that mediate the receptor's roles in disease progression. RESULTS Using Chromatin Immunoprecipitation (ChIP) Display, we discovered 19 novel loci occupied by the AR in castrate resistant C4-2B PCa cells. Only four of the 19 AR-occupied regions were within 10-kb 5'-flanking regulatory sequences. Three were located up to 4-kb 3' of the nearest gene, eight were intragenic and four were in gene deserts. Whereas the AR occupied the same loci in C4-2B (castrate resistant) and LNCaP (androgen-dependent) PCa cells, differences between the two cell lines were observed in the response of nearby genes to androgens. Among the genes strongly stimulated by DHT in C4-2B cells--D-dopachrome tautomerase (DDT), Protein kinase C delta (PRKCD), Glutathione S- transferase theta 2 (GSTT2), Transient receptor potential cation channel subfamily V member 3 (TRPV3), and Pyrroline-5-carboxylate reductase 1 (PYCR1)--most were less strongly or hardly stimulated in LNCaP cells. Another AR target gene, ornithine aminotransferase (OAT), was AR-stimulated in a ligand-independent manner, since it was repressed by AR siRNA knockdown, but not stimulated by DHT. We also present evidence for in vivo AR-mediated regulation of several genes identified by ChIP Display. For example, PRKCD and PYCR1, which may contribute to PCa cell growth and survival, are expressed in PCa biopsies from primary tumors before and after ablation and in metastatic lesions in a manner consistent with AR-mediated stimulation. CONCLUSION AR genomic occupancy is similar between LNCaP and C4-2B cells and is not biased towards 5' gene flanking sequences. The AR transcriptionally regulates less than half the genes nearby AR-occupied regions, usually but not always, in a ligand-dependent manner. Most are stimulated and a few are repressed. In general, response is stronger in C4-2B compared to LNCaP cells. Some of the genes near AR-occupied regions appear to be regulated by the AR in vivo as evidenced by their expression levels in prostate cancer tumors of various stages. Several AR target genes discovered in the present study, for example PRKCD and PYCR1, may open avenues in PCa research and aid the development of new approaches for disease management.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Androgens
- Binding Sites
- Cell Adhesion Molecules/biosynthesis
- Cell Adhesion Molecules/genetics
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/metabolism
- Chromosomes, Human/drug effects
- Chromosomes, Human/metabolism
- Dihydrotestosterone/pharmacology
- Extracellular Matrix Proteins/biosynthesis
- Extracellular Matrix Proteins/genetics
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Glutathione Transferase/biosynthesis
- Glutathione Transferase/genetics
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Male
- Mucin-6
- Mucins/biosynthesis
- Mucins/genetics
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Nuclear Proteins/biosynthesis
- Nuclear Proteins/genetics
- Oligonucleotide Array Sequence Analysis
- Ornithine-Oxo-Acid Transaminase/biosynthesis
- Ornithine-Oxo-Acid Transaminase/genetics
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Protein Kinase C-delta/biosynthesis
- Protein Kinase C-delta/genetics
- Pyrroline Carboxylate Reductases/biosynthesis
- Pyrroline Carboxylate Reductases/genetics
- Receptors, Androgen/genetics
- Receptors, Androgen/physiology
- TRPV Cation Channels/biosynthesis
- TRPV Cation Channels/genetics
- Transcription, Genetic
- delta-1-Pyrroline-5-Carboxylate Reductase
Collapse
Affiliation(s)
- Unnati Jariwala
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Jennifer Prescott
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Li Jia
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Artem Barski
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Steve Pregizer
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Jon P Cogan
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Armin Arasheben
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide/Hanson Institute, Adelaide, Australia
| | - Howard I Scher
- Genitourinary Oncology Service, Division of Solid Tumor Oncology, Memorial Sloan-Kettering Cancer Center, Department of Medicine, Joan and Sanford I. Weill College of Medicine, New York, NY, USA
| | - William L Gerald
- Genitourinary Oncology Service, Division of Solid Tumor Oncology, Memorial Sloan-Kettering Cancer Center, Department of Medicine, Joan and Sanford I. Weill College of Medicine, New York, NY, USA
| | - Grant Buchanan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide/Hanson Institute, Adelaide, Australia
| | - Gerhard A Coetzee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Baruch Frenkel
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, USA
| |
Collapse
|
804
|
Sandelin A, Carninci P, Lenhard B, Ponjavic J, Hayashizaki Y, Hume DA. Mammalian RNA polymerase II core promoters: insights from genome-wide studies. Nat Rev Genet 2007; 8:424-36. [PMID: 17486122 DOI: 10.1038/nrg2026] [Citation(s) in RCA: 379] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The identification and characterization of mammalian core promoters and transcription start sites is a prerequisite to understanding how RNA polymerase II transcription is controlled. New experimental technologies have enabled genome-wide discovery and characterization of core promoters, revealing that most mammalian genes do not conform to the simple model in which a TATA box directs transcription from a single defined nucleotide position. In fact, most genes have multiple promoters, within which there are multiple start sites, and alternative promoter usage generates diversity and complexity in the mammalian transcriptome and proteome. Promoters can be described by their start site usage distribution, which is coupled to the occurrence of cis-regulatory elements, gene function and evolutionary constraints. A comprehensive survey of mammalian promoters is a major step towards describing and understanding transcriptional control networks.
Collapse
Affiliation(s)
- Albin Sandelin
- Genome Exploration Research Group (Genome Network Project Core Group), RIKEN Genomic Sciences Center (GSC), RIKEN Yokohama Institute, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | | | | | | | | | | |
Collapse
|
805
|
Smith J, Wardle F, Loose M, Stanley E, Patient R. Germ layer induction in ESC--following the vertebrate roadmap. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2007; Chapter 1:Unit 1D.1. [PMID: 18785165 DOI: 10.1002/9780470151808.sc01d01s1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Controlled differentiation of pluripotential cells takes place routinely and with great success in developing vertebrate embryos. It therefore makes sense to take note of how this is achieved and use this knowledge to control the differentiation of embryonic stem cells (ESCs). An added advantage is that the differentiated cells resulting from this process in embryos have proven functionality and longevity. This unit reviews what is known about the embryonic signals that drive differentiation in one of the most informative of the vertebrate animal models of development, the amphibian Xenopus laevis. It summarizes their identities and the extent to which their activities are dose-dependent. The unit details what is known about the transcription factor responses to these signals, describing the networks of interactions that they generate. It then discusses the target genes of these transcription factors, the effectors of the differentiated state. Finally, how these same developmental programs operate during germ layer formation in the context of ESC differentiation is summarized.
Collapse
Affiliation(s)
- Jim Smith
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|
806
|
Brynczka C, Labhart P, Merrick BA. NGF-mediated transcriptional targets of p53 in PC12 neuronal differentiation. BMC Genomics 2007; 8:139. [PMID: 17540029 PMCID: PMC1894799 DOI: 10.1186/1471-2164-8-139] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Accepted: 05/31/2007] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND p53 is recognized as a critical regulator of the cell cycle and apoptosis. Mounting evidence also suggests a role for p53 in differentiation of cells including neuronal precursors. We studied the transcriptional role of p53 during nerve growth factor-induced differentiation of the PC12 line into neuron-like cells. We hypothesized that p53 contributed to PC12 differentiation through the regulation of gene targets distinct from its known transcriptional targets for apoptosis or DNA repair. RESULTS Using a genome-wide chromatin immunoprecipitation cloning technique, we identified and validated 14 novel p53-regulated genes following NGF treatment. The data show p53 protein was transcriptionally activated and contributed to NGF-mediated neurite outgrowth during differentiation of PC12 cells. Furthermore, we describe stimulus-specific regulation of a subset of these target genes by p53. The most salient differentiation-relevant target genes included wnt7b involved in dendritic extension and the tfcp2l4/grhl3 grainyhead homolog implicated in ectodermal development. Additional targets included brk, sdk2, sesn3, txnl2, dusp5, pon3, lect1, pkcbpb15 and other genes. CONCLUSION Within the PC12 neuronal context, putative p53-occupied genomic loci spanned the entire Rattus norvegicus genome upon NGF treatment. We conclude that receptor-mediated p53 transcriptional activity is involved in PC12 differentiation and may suggest a contributory role for p53 in neuronal development.
Collapse
Affiliation(s)
- Christopher Brynczka
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
- Department of Environmental and Molecular Toxicology, North Carolina State University, Raleigh, North Carolina 27606, USA
| | - Paul Labhart
- Genpathway, Inc., San Diego, California 92121, USA
| | - B Alex Merrick
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
807
|
Johnson DS, Mortazavi A, Myers RM, Wold B. Genome-wide mapping of in vivo protein-DNA interactions. Science 2007; 316:1497-502. [PMID: 17540862 DOI: 10.1126/science.1141319] [Citation(s) in RCA: 2047] [Impact Index Per Article: 113.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In vivo protein-DNA interactions connect each transcription factor with its direct targets to form a gene network scaffold. To map these protein-DNA interactions comprehensively across entire mammalian genomes, we developed a large-scale chromatin immunoprecipitation assay (ChIPSeq) based on direct ultrahigh-throughput DNA sequencing. This sequence census method was then used to map in vivo binding of the neuron-restrictive silencer factor (NRSF; also known as REST, for repressor element-1 silencing transcription factor) to 1946 locations in the human genome. The data display sharp resolution of binding position [+/-50 base pairs (bp)], which facilitated our finding motifs and allowed us to identify noncanonical NRSF-binding motifs. These ChIPSeq data also have high sensitivity and specificity [ROC (receiver operator characteristic) area >/= 0.96] and statistical confidence (P <10(-4)), properties that were important for inferring new candidate interactions. These include key transcription factors in the gene network that regulates pancreatic islet cell development.
Collapse
Affiliation(s)
- David S Johnson
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305-5120, USA
| | | | | | | |
Collapse
|
808
|
Chen N, Mah A, Blacque OE, Chu J, Phgora K, Bakhoum MW, Hunt Newbury CR, Khattra J, Chan S, Go A, Efimenko E, Johnsen R, Phirke P, Swoboda P, Marra M, Moerman DG, Leroux MR, Baillie DL, Stein LD. Identification of ciliary and ciliopathy genes in Caenorhabditis elegans through comparative genomics. Genome Biol 2007; 7:R126. [PMID: 17187676 PMCID: PMC1794439 DOI: 10.1186/gb-2006-7-12-r126] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 10/20/2006] [Accepted: 12/22/2006] [Indexed: 01/05/2023] Open
Abstract
Comparative genomic analysis of three nematode species identifies 93 genes that encode putative components of the ciliated neurons in C. elegans and are subject to the same regulatory control. Background The recent availability of genome sequences of multiple related Caenorhabditis species has made it possible to identify, using comparative genomics, similarly transcribed genes in Caenorhabditis elegans and its sister species. Taking this approach, we have identified numerous novel ciliary genes in C. elegans, some of which may be orthologs of unidentified human ciliopathy genes. Results By screening for genes possessing canonical X-box sequences in promoters of three Caenorhabditis species, namely C. elegans, C. briggsae and C. remanei, we identified 93 genes (including known X-box regulated genes) that encode putative components of ciliated neurons in C. elegans and are subject to the same regulatory control. For many of these genes, restricted anatomical expression in ciliated cells was confirmed, and control of transcription by the ciliogenic DAF-19 RFX transcription factor was demonstrated by comparative transcriptional profiling of different tissue types and of daf-19(+) and daf-19(-) animals. Finally, we demonstrate that the dye-filling defect of dyf-5(mn400) animals, which is indicative of compromised exposure of cilia to the environment, is caused by a nonsense mutation in the serine/threonine protein kinase gene M04C9.5. Conclusion Our comparative genomics-based predictions may be useful for identifying genes involved in human ciliopathies, including Bardet-Biedl Syndrome (BBS), since the C. elegans orthologs of known human BBS genes contain X-box motifs and are required for normal dye filling in C. elegans ciliated neurons.
Collapse
Affiliation(s)
- Nansheng Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
| | - Allan Mah
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
| | - Oliver E Blacque
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
- School of Biomolecular and Biomedical Sciences, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jeffrey Chu
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
| | - Kiran Phgora
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
| | - Mathieu W Bakhoum
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
| | - C Rebecca Hunt Newbury
- Department of Zoology, University of British Columbia, West Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | - Jaswinder Khattra
- Department of Zoology, University of British Columbia, West Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | - Susanna Chan
- Department of Zoology, University of British Columbia, West Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | - Anne Go
- Department of Zoology, University of British Columbia, West Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | - Evgeni Efimenko
- Karolinska Institute, Department of Biosciences and Nutrition, Södertörn University College, School of Life Sciences, S-14189 Huddinge, Sweden
| | - Robert Johnsen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
| | - Prasad Phirke
- Karolinska Institute, Department of Biosciences and Nutrition, Södertörn University College, School of Life Sciences, S-14189 Huddinge, Sweden
| | - Peter Swoboda
- Karolinska Institute, Department of Biosciences and Nutrition, Södertörn University College, School of Life Sciences, S-14189 Huddinge, Sweden
| | - Marco Marra
- British Columbia Cancer Agency, Genome Sciences Centre, Vancouver, British Columbia, Canada V5Z 4S6
| | - Donald G Moerman
- Department of Zoology, University of British Columbia, West Mall, Vancouver, British Columbia, Canada V6T 1Z4
| | - Michel R Leroux
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
| | - David L Baillie
- Department of Molecular Biology and Biochemistry, Simon Fraser University, University Drive, Burnaby, British Columbia, Canada V5A 1S6
| | - Lincoln D Stein
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
809
|
Magrini R, Russo D, Fronza G, Inga A, Menichini P. The kinetics of p53-binding and histone acetylation at target promoters do not strictly correlate with gene expression after UV damage. J Cell Biochem 2007; 100:1276-87. [PMID: 17063487 DOI: 10.1002/jcb.21122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We have addressed the correlation between sequence-specific DNA binding by the tumor suppressor p53 and transactivation of various target genes, in the context of UV irradiation responses. In A549 cells (p53WT), p53 occupancy at the p21, mdm2, and puma promoters increased significantly after UV irradiation. In contrast, p21 mRNA levels did not change, mdm2 mRNA decreased and both p21 and mdm2 proteins were downregulated shortly after UV. At later times, higher p53 occupancy correlated with enhanced expression of these two genes both at mRNA and protein levels. In the p53 mutant cell lines LX1 (R273H) and SKMes1 (R280K), no significant p53-binding was detected at the gene targets analyzed. Accordingly, p21 and mdm2 proteins were not upregulated after UV irradiation. The kinetics of histone acetylation did not strictly correlate with gene expression. In fact, high levels of acetylated H3 (AcH3) and, particularly, acetylated H4 (AcH4) histones were found shortly after UV irradiation on p21 and mdm2 promoters. At the later time point, when transactivation was detected, acetylation levels decreased significantly although remaining higher than basal levels. Our results indicate that p53 transcription-dependent and -independent responses are activated with different kinetics after UV, possibly relating to the repair of UV-induced DNA damage. Based on the histone acetylation pattern we hypothesize that the DNA repair function of p53, associated to global genome repair and foci of DNA damage, may be relevant for all p53-binding sites, including those where occupancy by p53 is also associated to transcriptional modulation.
Collapse
Affiliation(s)
- Roberta Magrini
- Molecular Mutagenesis Unit, Department of Translational Oncology, National Cancer Research Institute (IST), Largo R. Benzi, 10, 16132 Genova, Italy
| | | | | | | | | |
Collapse
|
810
|
Sequence variations in DNA repair gene XPC is associated with lung cancer risk in a Chinese population: a case-control study. BMC Cancer 2007; 7:81. [PMID: 17498315 PMCID: PMC1884164 DOI: 10.1186/1471-2407-7-81] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 05/13/2007] [Indexed: 11/10/2022] Open
Abstract
Background The nucleotide excision repair (NER) protein, xeroderma pigmentosum C (XPC), participates in recognizing DNA lesions and initiating DNA repair in response to DNA damage. Because mutations in XPC cause a high risk of cancer in XP patients, we hypothesized that inherited sequence variations in XPC may alter DNA repair and thus susceptibility to cancer. Methods In this hospital-based case-control study, we investigated five XPC tagging, common single nucleotide polymorphisms (tagging SNPs) in 1,010 patients with newly diagnosed lung cancer and 1,011 matched cancer free controls in a Chinese population. Results In individual tagging SNP analysis, we found that rs3731055AG+AA variant genotypes were associated with a significantly decreased risk of lung adenocarcinoma [adjusted odds ratio (OR), 0.71; 95% confidence interval (CI), 0.56–0.90] but an increased risk of small cell carcinomas [adjusted OR, 1.79; 95% CI, 1.05–3.07]. Furthermore, we found that haplotype ACCCA was associated with a decreased risk of lung adenocarcinoma [OR, 0.78; 95% CI, 0.62–0.97] but an increased risk of small cell carcinomas [OR, 1.68; 95% CI, 1.04–2.71], which reflected the presence of rs3731055A allele in this haplotype. Further stratified analysis revealed that the protective effect of rs3731055AG+AA on risk of lung adenocarcinoma was more evident among young subjects (age ≤ 60) and never smokers. Conclusion These results suggest that inherited sequence variations in XPC may modulate risk of lung cancer, especially lung adenocarcinoma, in Chinese populations. However, these findings need to be verified in larger confirmatory studies with more comprehensively selected tagging SNPs.
Collapse
|
811
|
Kim TH, Abdullaev ZK, Smith AD, Ching KA, Loukinov DI, Green RD, Zhang MQ, Lobanenkov VV, Ren B. Analysis of the vertebrate insulator protein CTCF-binding sites in the human genome. Cell 2007; 128:1231-45. [PMID: 17382889 PMCID: PMC2572726 DOI: 10.1016/j.cell.2006.12.048] [Citation(s) in RCA: 811] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 11/23/2006] [Accepted: 12/28/2006] [Indexed: 12/31/2022]
Abstract
Insulator elements affect gene expression by preventing the spread of heterochromatin and restricting transcriptional enhancers from activation of unrelated promoters. In vertebrates, insulator's function requires association with the CCCTC-binding factor (CTCF), a protein that recognizes long and diverse nucleotide sequences. While insulators are critical in gene regulation, only a few have been reported. Here, we describe 13,804 CTCF-binding sites in potential insulators of the human genome, discovered experimentally in primary human fibroblasts. Most of these sequences are located far from the transcriptional start sites, with their distribution strongly correlated with genes. The majority of them fit to a consensus motif highly conserved and suitable for predicting possible insulators driven by CTCF in other vertebrate genomes. In addition, CTCF localization is largely invariant across different cell types. Our results provide a resource for investigating insulator function and possible other general and evolutionarily conserved activities of CTCF sites.
Collapse
Affiliation(s)
- Tae Hoon Kim
- Ludwig Institute for Cancer Research, 9500 Gilman Drive, La Jolla, CA 92093-0653
- To whom correspondence should be addressed,
,
| | - Ziedulla K. Abdullaev
- National Institutes of Allergy and Infectious Disease, 5640 Fishers Lane, Rockville, MD 20852
| | - Andrew D. Smith
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724
| | - Keith A. Ching
- Ludwig Institute for Cancer Research, 9500 Gilman Drive, La Jolla, CA 92093-0653
| | - Dmitri I. Loukinov
- National Institutes of Allergy and Infectious Disease, 5640 Fishers Lane, Rockville, MD 20852
| | | | - Michael Q. Zhang
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724
| | - Victor V. Lobanenkov
- National Institutes of Allergy and Infectious Disease, 5640 Fishers Lane, Rockville, MD 20852
| | - Bing Ren
- Ludwig Institute for Cancer Research, 9500 Gilman Drive, La Jolla, CA 92093-0653
- To whom correspondence should be addressed,
,
| |
Collapse
|
812
|
Kapranov P, Willingham AT, Gingeras TR. Genome-wide transcription and the implications for genomic organization. Nat Rev Genet 2007; 8:413-23. [PMID: 17486121 DOI: 10.1038/nrg2083] [Citation(s) in RCA: 547] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent evidence of genome-wide transcription in several species indicates that the amount of transcription that occurs cannot be entirely accounted for by current sets of genome-wide annotations. Evidence indicates that most of both strands of the human genome might be transcribed, implying extensive overlap of transcriptional units and regulatory elements. These observations suggest that genomic architecture is not colinear, but is instead interleaved and modular, and that the same genomic sequences are multifunctional: that is, used for multiple independently regulated transcripts and as regulatory regions. What are the implications and consequences of such an interleaved genomic architecture in terms of increased information content, transcriptional complexity, evolution and disease states?
Collapse
Affiliation(s)
- Philipp Kapranov
- Affymetrix, Inc., 3420 Central Expressway, Santa Clara, California 95051, USA
| | | | | |
Collapse
|
813
|
Menendez D, Inga A, Jordan JJ, Resnick MA. Changing the p53 master regulatory network: ELEMENTary, my dear Mr Watson. Oncogene 2007; 26:2191-201. [PMID: 17401428 DOI: 10.1038/sj.onc.1210277] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The p53 master regulatory network provides for the stress-responsive direct control of a vast number of genes in humans that can be grouped into several biological categories including cell-cycle control, apoptosis and DNA repair. Similar to other sequence-specific master regulators, there is a matrix of key components, which provide for variation within the p53 master regulatory network that include p53 itself, target response element sequences (REs) that provide for p53 regulation of target genes, chromatin, accessory proteins and transcription machinery. Changes in any of these can impact the expression of individual genes, groups of genes and the eventual biological responses. The many REs represent the core of the master regulatory network. Since defects or altered expression of p53 are associated with over 50% of all cancers and greater than 90% of p53 mutations are in the sequence-specific DNA-binding domain, it is important to understand the relationship between wild-type or mutant p53 proteins and the target response elements. In the words of the legendary detective Sherlock Holmes, it is 'Elementary, my dear Mr. Watson'.
Collapse
Affiliation(s)
- D Menendez
- Laboratory of Molecular Genetics, Chromosome Stability Section, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | | | | | | |
Collapse
|
814
|
Bensaad K, Vousden KH. p53: new roles in metabolism. Trends Cell Biol 2007; 17:286-91. [PMID: 17481900 DOI: 10.1016/j.tcb.2007.04.004] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 03/07/2007] [Accepted: 04/23/2007] [Indexed: 11/22/2022]
Abstract
Virtually all cancers show metabolic changes that result in upregulation of glycolysis and glucose consumption. Although discovered in the 1920s, how this glycolytic switch happens, and whether it is a cause or a consequence of the malignant process, has remained a matter of debate. The p53 tumor suppressor gene, discovered some 30 years ago, has a key role in preventing cancer development. Recent discoveries revealing new functions for p53 in the regulation of glucose metabolism and oxidative stress have brought together these two venerable fields of cancer biology. These activities of p53 appear to be key in tumor suppression, and shed some light on the pathways that underlie the metabolic changes in cancer cells.
Collapse
Affiliation(s)
- Karim Bensaad
- The Beatson Institute for Cancer Research, Glasgow, UK
| | | |
Collapse
|
815
|
Zhang X, Diao S, Rao Q, Xing H, Liu H, Liao X, Wang M, Wang J. Identification of a Novel Isoform of iASPP and its Interaction with p53. J Mol Biol 2007; 368:1162-71. [PMID: 17391696 DOI: 10.1016/j.jmb.2007.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 02/28/2007] [Accepted: 03/01/2007] [Indexed: 01/07/2023]
Abstract
iASPP is an inhibitory member of ASPP (apoptosis stimulating protein of p53, or Ankyrin repeats, SH3 domain and proline-rich region contain Protein) family. As reported previously, it at least includes two isoforms, one is iASPP/RAI (351 amino acids, aa) and the other is iASPP (828 aa).Here, we identified a novel open reading frame of human iASPP, which encodes a 407 aa protein and highly matches with the C terminus of iASPP (828 aa, CAI60219). Hereafter, iASPP (407 aa) will be referred to as iASPP-SV (iASPP splice variant). In further study, we found that iASPP-SV is a nuclear protein, and is capable of binding to p53 in vivo. Moreover, overexpression of iASPP-SV can inhibit the transcriptional activity of p53 on the promoters of both Bax and p21.
Collapse
Affiliation(s)
- Xinwei Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300020, China
| | | | | | | | | | | | | | | |
Collapse
|
816
|
Sandmann T, Girardot C, Brehme M, Tongprasit W, Stolc V, Furlong EE. A core transcriptional network for early mesoderm development in Drosophila melanogaster. Genes Dev 2007; 21:436-49. [PMID: 17322403 PMCID: PMC1804332 DOI: 10.1101/gad.1509007] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Embryogenesis is controlled by large gene-regulatory networks, which generate spatially and temporally refined patterns of gene expression. Here, we report the characteristics of the regulatory network orchestrating early mesodermal development in the fruitfly Drosophila, where the transcription factor Twist is both necessary and sufficient to drive development. Through the integration of chromatin immunoprecipitation followed by microarray analysis (ChIP-on-chip) experiments during discrete time periods with computational approaches, we identified >2000 Twist-bound cis-regulatory modules (CRMs) and almost 500 direct target genes. Unexpectedly, Twist regulates an almost complete cassette of genes required for cell proliferation in addition to genes essential for morophogenesis and cell migration. Twist targets almost 25% of all annotated Drosophila transcription factors, which may represent the entire set of regulators necessary for the early development of this system. By combining in vivo binding data from Twist, Mef2, Tinman, and Dorsal we have constructed an initial transcriptional network of early mesoderm development. The network topology reveals extensive combinatorial binding, feed-forward regulation, and complex logical outputs as prevalent features. In addition to binary activation and repression, we suggest that Twist binds to almost all mesodermal CRMs to provide the competence to integrate inputs from more specialized transcription factors.
Collapse
Affiliation(s)
- Thomas Sandmann
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Charles Girardot
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Marc Brehme
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Waraporn Tongprasit
- Genome Research Facility, NASA Ames Research Center, Moffet Field, California 94035, USA
| | - Viktor Stolc
- Genome Research Facility, NASA Ames Research Center, Moffet Field, California 94035, USA
| | - Eileen E.M. Furlong
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
- Corresponding author.E-MAIL ; FAX 49-6221-387166
| |
Collapse
|
817
|
Abstract
The p53 tumour suppressor protein can efficiently inhibit tumour development. This activity reflects its ability to induce a number of different responses, including cell cycle arrest and apoptosis. Recent studies have revealed some interesting insights into how the choice of response to p53 is regulated, highlighting a correlation between the activation of cell cycle arrest and survival with the ability of p53 to reduce oxidative stress and protect cells from genotoxic damage. Understanding the molecular mechanisms that determine which response is selected may allow us to modulate these pathways so that therapeutic reactivation of p53 favours apoptotic cell death in tumour cells, but a reversible--and therefore far less toxic--induction of cell cycle arrest in normal cells.
Collapse
Affiliation(s)
- Karen H Vousden
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| |
Collapse
|
818
|
Lin CY, Vega VB, Thomsen JS, Zhang T, Kong SL, Xie M, Chiu KP, Lipovich L, Barnett DH, Stossi F, Yeo A, George J, Kuznetsov VA, Lee YK, Charn TH, Palanisamy N, Miller LD, Cheung E, Katzenellenbogen BS, Ruan Y, Bourque G, Wei CL, Liu ET. Whole-genome cartography of estrogen receptor alpha binding sites. PLoS Genet 2007; 3:e87. [PMID: 17542648 PMCID: PMC1885282 DOI: 10.1371/journal.pgen.0030087] [Citation(s) in RCA: 357] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Accepted: 04/17/2007] [Indexed: 11/19/2022] Open
Abstract
Using a chromatin immunoprecipitation-paired end diTag cloning and sequencing strategy, we mapped estrogen receptor α (ERα) binding sites in MCF-7 breast cancer cells. We identified 1,234 high confidence binding clusters of which 94% are projected to be bona fide ERα binding regions. Only 5% of the mapped estrogen receptor binding sites are located within 5 kb upstream of the transcriptional start sites of adjacent genes, regions containing the proximal promoters, whereas vast majority of the sites are mapped to intronic or distal locations (>5 kb from 5′ and 3′ ends of adjacent transcript), suggesting transcriptional regulatory mechanisms over significant physical distances. Of all the identified sites, 71% harbored putative full estrogen response elements (EREs), 25% bore ERE half sites, and only 4% had no recognizable ERE sequences. Genes in the vicinity of ERα binding sites were enriched for regulation by estradiol in MCF-7 cells, and their expression profiles in patient samples segregate ERα-positive from ERα-negative breast tumors. The expression dynamics of the genes adjacent to ERα binding sites suggest a direct induction of gene expression through binding to ERE-like sequences, whereas transcriptional repression by ERα appears to be through indirect mechanisms. Our analysis also indicates a number of candidate transcription factor binding sites adjacent to occupied EREs at frequencies much greater than by chance, including the previously reported FOXA1 sites, and demonstrate the potential involvement of one such putative adjacent factor, Sp1, in the global regulation of ERα target genes. Unexpectedly, we found that only 22%–24% of the bona fide human ERα binding sites were overlapping conserved regions in whole genome vertebrate alignments, which suggest limited conservation of functional binding sites. Taken together, this genome-scale analysis suggests complex but definable rules governing ERα binding and gene regulation. Estrogen receptors (ERs) play key roles in facilitating the transcriptional effects of hormone functions in target tissues. To obtain a genome-wide view of ERα binding sites, we applied chromatin immunoprecipitation coupled with a cloning and sequencing strategy using chromatin immunoprecipitation pair end-tagging technology to map ERα binding sites in MCF-7 human breast cancer cells. We identified 1,234 high quality ERα binding sites in the human genome and demonstrated that the binding sites are frequently adjacent to genes significantly associated with breast cancer disease status and outcome. The mapping results also revealed that ERα can influence gene expression across distances of up to 100 kilobases or more, that genes that are induced or repressed utilize sites in different regions relative to the transcript (suggesting different mechanisms of action), and that ERα binding sites are only modestly conserved in evolution. Using computational approaches, we identified potential interactions with other transcription factor binding sites adjacent to the ERα binding elements. Taken together, these findings suggest complex but definable rules governing ERα binding and gene regulation and provide a valuable dataset for mapping the precise control nodes for one of the most important nuclear hormone receptors in breast cancer biology.
Collapse
Affiliation(s)
- Chin-Yo Lin
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | | | - Jane S Thomsen
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | - Tao Zhang
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | - Say Li Kong
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | - Min Xie
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | - Kuo Ping Chiu
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | | | - Daniel H Barnett
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Fabio Stossi
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Ailing Yeo
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Joshy George
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | | | - Yew Kok Lee
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | - Tze Howe Charn
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | | | - Lance D Miller
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | - Edwin Cheung
- Genome Institute of Singapore, Singapore, Republic of Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Benita S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yijun Ruan
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | | | - Chia-Lin Wei
- Genome Institute of Singapore, Singapore, Republic of Singapore
| | - Edison T Liu
- Genome Institute of Singapore, Singapore, Republic of Singapore
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
819
|
Ma B, Pan Y, Zheng J, Levine AJ, Nussinov R. Sequence analysis of p53 response-elements suggests multiple binding modes of the p53 tetramer to DNA targets. Nucleic Acids Res 2007; 35:2986-3001. [PMID: 17439973 PMCID: PMC1888811 DOI: 10.1093/nar/gkm192] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2006] [Revised: 03/11/2007] [Accepted: 03/17/2007] [Indexed: 01/02/2023] Open
Abstract
The p53 tetramer recognizes specifically a 20-bp DNA element. Here, we examined symmetries encoded in p53 response elements (p53REs). We analyzed base inversion correlations within the half-site, as well as in the full-site palindrome. We found that p53REs are not only direct repeats of half-sites; rather, two p53 half-sites couple to form a higher order 20 bp palindrome. The palindrome couplings between the half-sites are stronger for the human than for the mouse genome. The full-site palindrome and half-site palindrome are controlled by insertions between the two half-sites. The most notable feature is that the full-site palindrome with coupling between quarter-sites one and four (H14 coupling) dominates the p53REs without insertions. The most frequently observed insertion in human p53REs of 3 bp enhances the half-site palindrome. The statistical frequencies of the coupling between the half-sites in the human genome correlate with grouped experimental p53 affinities with p53REs. Examination of known p53REs indicates the H14 couplings are stronger for positive regulation than for negatively regulated p53REs, with repressors having the lowest H14 couplings. We propose that the palindromic sequence couplings may encode such potential preferred multiple binding modes of the p53 tetramer to DNA.
Collapse
Affiliation(s)
- Buyong Ma
- Basic Research Program, SAIC-Frederick Inc., Center for Cancer Research Nanobiology Program, NCI-Frederick, Frederick, MD 21702, USA.
| | | | | | | | | |
Collapse
|
820
|
Agarwal SK, Impey S, McWeeney S, Scacheri PC, Collins FS, Goodman RH, Spiegel AM, Marx SJ. Distribution of menin-occupied regions in chromatin specifies a broad role of menin in transcriptional regulation. Neoplasia 2007; 9:101-7. [PMID: 17356705 PMCID: PMC1813935 DOI: 10.1593/neo.06706] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Revised: 12/27/2006] [Accepted: 12/29/2006] [Indexed: 11/18/2022] Open
Abstract
Menin is the protein product of the MEN1 tumor-suppressor gene; one allele of MEN1 is inactivated in the germ line of patients with "multiple endocrine neoplasia type 1" (MEN1) cancer syndrome. Menin interacts with several proteins involved in transcriptional regulation. RNA expression analyses have identified several menin-regulated genes that could represent proximal or distal interaction sites for menin. This report presents a substantial and unbiased sampling of menin-occupied chromatin regions using Serial Analysis of Chromatin Occupancy; this method combines chromatin immuno-precipitation with Serial Analysis of Gene Expression. Hundreds of menin-occupied genomic sites were identified in promoter regions (32% of menin-occupied loci), near the 3' end of genes (14%), or inside genes (21%), extending other data about menin recruitments to many sites of transcriptional activity. A large number of menin-occupied sites (33%) were located outside known gene regions. Additional annotation of the human genome could help in identifying genes at these loci, or these might be gene-free regions of the genome where menin occupancy could play some structural or regulatory role. Menin occupancy at many intragenic positions distant from the core promoter reveals an unexpected type of menin target region at many loci in the genome. These unbiased data also suggest that menin could play a broad role in transcriptional regulation.
Collapse
Affiliation(s)
- Sunita K Agarwal
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1802, USA.
| | | | | | | | | | | | | | | |
Collapse
|
821
|
Lefort K, Mandinova A, Ostano P, Kolev V, Calpini V, Kolfschoten I, Devgan V, Lieb J, Raffoul W, Hohl D, Neel V, Garlick J, Chiorino G, Dotto GP. Notch1 is a p53 target gene involved in human keratinocyte tumor suppression through negative regulation of ROCK1/2 and MRCKalpha kinases. Genes Dev 2007; 21:562-77. [PMID: 17344417 PMCID: PMC1820898 DOI: 10.1101/gad.1484707] [Citation(s) in RCA: 255] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Little is known about the regulation and function of the Notch1 gene in negative control of human tumors. Here we show that Notch1 gene expression and activity are substantially down-modulated in keratinocyte cancer cell lines and tumors, with expression of this gene being under p53 control in these cells. Genetic suppression of Notch signaling in primary human keratinocytes is sufficient, together with activated ras, to cause aggressive squamous cell carcinoma formation. Similar tumor-promoting effects are also caused by in vivo treatment of mice, grafted with keratinocytes expressing oncogenic ras alone, with a pharmacological inhibitor of endogenous Notch signaling. These effects are linked with a lesser commitment of keratinocytes to differentiation, an expansion of stem cell populations, and a mechanism involving up-regulation of ROCK1/2 and MRCKalpha kinases, two key effectors of small Rho GTPases previously implicated in neoplastic progression. Thus, the Notch1 gene is a p53 target with a role in human tumor suppression through negative regulation of Rho effectors.
Collapse
Affiliation(s)
- Karine Lefort
- Department of Biochemistry, University of Lausanne, Epalinges CH-1066, Switzerland
| | - Anna Mandinova
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Paola Ostano
- Laboratory of Cancer Pharmacogenomics, Fondo “Edo Tempia,” Biella 13900, Italy
| | - Vihren Kolev
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Valerie Calpini
- Department of Biochemistry, University of Lausanne, Epalinges CH-1066, Switzerland
| | - Ingrid Kolfschoten
- Department of Biochemistry, University of Lausanne, Epalinges CH-1066, Switzerland
| | - Vikram Devgan
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Jocelyn Lieb
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Wassim Raffoul
- Department of Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne CH-1011, Switzerland
| | - Daniel Hohl
- Department of Dermatology, Centre Hospitalier Universitaire Vaudois, Lausanne CH-1011, Switzerland
| | - Victor Neel
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Jonathan Garlick
- Division of Cancer Biology and Tissue Engineering, Tufts University Dental School, Boston, Massachusetts 02111, USA
| | - Giovanna Chiorino
- Laboratory of Cancer Pharmacogenomics, Fondo “Edo Tempia,” Biella 13900, Italy
| | - G. Paolo Dotto
- Department of Biochemistry, University of Lausanne, Epalinges CH-1066, Switzerland
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
- Corresponding author.E-MAIL ; FAX 41-21-692-5705
| |
Collapse
|
822
|
Kerley-Hamilton JS, Pike AM, Hutchinson JA, Freemantle SJ, Spinella MJ. The direct p53 target gene, FLJ11259/DRAM, is a member of a novel family of transmembrane proteins. BIOCHIMICA ET BIOPHYSICA ACTA 2007; 1769:209-19. [PMID: 17397945 PMCID: PMC1936437 DOI: 10.1016/j.bbaexp.2007.02.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 02/08/2007] [Accepted: 02/08/2007] [Indexed: 10/23/2022]
Abstract
The tumor suppressor p53 regulates diverse biological processes primarily via activation of downstream target genes. Even though many p53 target genes have been described, the precise mechanisms of p53 biological actions are uncertain. In previous work we identified by microarray analysis a candidate p53 target gene, FLJ11259/DRAM. In this report we have identified three uncharacterized human proteins with sequence homology to FLJ11259, suggesting that FLJ11259 is a member of a novel family of proteins with six transmembrane domains. Several lines of investigation confirm FLJ11259 is a direct p53 target gene. p53 siRNA prevented cisplatin-mediated up-regulation of FLJ11259 in NT2/D1 cells. Likewise in HCT116 p53+/+ cells and MCF10A cells, FLJ11259 is induced by cisplatin treatment but to a much lesser extent in isogenic p53-suppressed cells. A functional p53 response element was identified 22.3 kb upstream of the first coding exon of FLJ11259 and is shown to be active in reporter assays. In addition, chromatin immunoprecipitation assays indicate that p53 binds directly to this element in vivo and that binding is enhanced following cisplatin treatment. Confocal microscopy showed that an FLJ-GFP fusion protein localizes mainly in a punctate pattern in the cytoplasm. Overexpression studies in Cos-7, Saos2, and NT2/D1 cells suggest that FLJ11259 is associated with increased clonal survival. In summary, we have identified FLJ11259/DRAM as a p53-inducible member of a novel family of transmembrane proteins. FLJ11259/DRAM may be an important modulator of p53 responses in diverse tumor types.
Collapse
|
823
|
Abstract
Pluripotent stem cells, similar to more restricted stem cells, are able to both self-renew and generate differentiated progeny. Although this dual functionality has been much studied, the search for molecular signatures of 'stemness' and pluripotency is only now beginning to gather momentum. While the focus of much of this work has been on the transcriptional features of embryonic stem cells, recent studies have indicated the importance of unique epigenetic profiles that keep key developmental genes 'poised' in a repressed but activatable state. Determining how these epigenetic features relate to the transcriptional signatures of ES cells, and whether they are also important in other types of stem cell, is a key challenge for the future.
Collapse
Affiliation(s)
- Mikhail Spivakov
- Lymphocyte Development Group, MRC Clinical Sciences Centre, Imperial College School of Medicine, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | | |
Collapse
|
824
|
Kruse JJCM, Svensson JP, Huigsloot M, Giphart-Gassler M, Schoonen WGEJ, Polman JEM, Jean Horbach G, van de Water B, Vrieling H. A portrait of cisplatin-induced transcriptional changes in mouse embryonic stem cells reveals a dominant p53-like response. Mutat Res 2007; 617:58-70. [PMID: 17327130 DOI: 10.1016/j.mrfmmm.2006.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 11/09/2006] [Accepted: 12/15/2006] [Indexed: 05/14/2023]
Abstract
Accumulation of damage in undifferentiated cells may threaten homeostasis and regenerative capacity. Remarkably, p53 has been suggested to be transcriptionally inactive in these cells. To gain insight in the kinetics and interplay of the predominant transcriptional responses of DNA damage signalling pathways in undifferentiated cells, mouse embryonic stem cells were exposed to cisplatin at four different time points (2, 4, 8 and 24h) and concentrations (1, 2, 5 and 10 microM). RNA was isolated and subjected to genome-wide expression profiling. Up to one fourth of the tested genes could be identified as being differentially expressed (false discovery rate=10%) after the cisplatin treatment. Clustering of the expression changes showed a strong time dependency. To investigate the relationship between affected genes, a gene set analysis method was used. Functionally related gene sets were defined using gene ontologies or transcription factor binding sites and were tested for overrepresentation within the differentially expressed genes. A variety of gene sets were clearly enriched among which 'apoptosis' and 'cell cycle' were the most pronounced. Furthermore, there was a strong enrichment of genes with a p53-binding motif. The involvement of the 'cell cycle' and 'apoptosis' gene sets in the cisplatin response was detected at concentrations and time points where the respective biological assays were still negative. The results reveal novel insights into the mechanisms which maintain the genomic integrity in undifferentiated cells. Additionally the results illustrate that gene set analysis of genome-wide expression changes provides a sensitive instrument to detect cellular stress responses to DNA damage.
Collapse
|
825
|
Rahman-Roblick R, Johannes Roblick U, Hellman U, Conrotto P, Liu T, Becker S, Hirschberg D, Jörnvall H, Auer G, Wiman KG. p53 targets identified by protein expression profiling. Proc Natl Acad Sci U S A 2007; 104:5401-6. [PMID: 17372198 PMCID: PMC1828709 DOI: 10.1073/pnas.0700794104] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
p53 triggers cell cycle arrest and apoptosis through transcriptional regulation of specific target genes. We have investigated the effect of p53 activation on the proteome using 2D gel electrophoresis analysis of mitomycin C-treated HCT116 colon carcinoma cells carrying wild-type p53. Approximately 5,800 protein spots were separated in overlapping narrow-pH-range gel strips, and 115 protein spots showed significant expression changes upon p53 activation. The identity of 55 protein spots was obtained by mass spectrometry. The majority of the identified proteins have no previous connection to p53. The proteins fall into different functional categories, such as mRNA processing, translation, redox regulation, and apoptosis, consistent with the idea that p53 regulates multiple cellular pathways. p53-dependent regulation of five of the up-regulated proteins, eIF5A, hnRNP C1/C2, hnRNP K, lamin A/C, and Nm23-H1, and two of the down-regulated proteins, Prx II and TrpRS, was examined in further detail. Analysis of mRNA expression levels demonstrated both transcription-dependent and transcription-independent regulation among the identified targets. Thus, this study reveals protein targets of p53 and highlights the role of transcription-independent effects for the p53-induced biological response.
Collapse
Affiliation(s)
- Rubaiyat Rahman-Roblick
- *Department of Oncology–Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Uwe Johannes Roblick
- *Department of Oncology–Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
- Department of Surgery, University of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany; and
| | - Ulf Hellman
- Ludwig Institute for Cancer Research, Box 595, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Paolo Conrotto
- Ludwig Institute for Cancer Research, Box 595, Uppsala University, SE-751 24 Uppsala, Sweden
- Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule Zürich, Wolfgang-Pauli Strasse 10, 8093 Zürich, Switzerland
| | - Tao Liu
- *Department of Oncology–Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Susanne Becker
- *Department of Oncology–Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Daniel Hirschberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Hans Jörnvall
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Gert Auer
- *Department of Oncology–Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Klas G. Wiman
- *Department of Oncology–Pathology, Cancer Center Karolinska, Karolinska Institutet, SE-171 76 Stockholm, Sweden
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
826
|
Yugawa T, Handa K, Narisawa-Saito M, Ohno SI, Fujita M, Kiyono T. Regulation of Notch1 gene expression by p53 in epithelial cells. Mol Cell Biol 2007; 27:3732-42. [PMID: 17353266 PMCID: PMC1899981 DOI: 10.1128/mcb.02119-06] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The E6 protein of cervical cancer-associated human papillomaviruses (HPVs) is known to suppress keratinocyte differentiation through unidentified mechanisms. Notch1 is a determinant of keratinocyte differentiation and functions as a tumor suppressor in mammalian epidermis. Here, we report that the Notch1 gene is a novel target of p53 and can be down-regulated by E6 through p53 degradation in normal human epithelial cells. Thus, inactivation of p53 by E6 or short-hairpin RNA (shRNA) resulted in reduced Notch1 expression at the transcription level, and a p53-responsive element could be identified in the Notch1 promoter. The expression of E6, p53 shRNA, or Notch1 shRNA suppressed both spontaneous keratinocyte differentiation in culture and its induction upon DNA damage. Furthermore, the induction of Notch1 and differentiation makers as well as thickening of the epidermal layer upon UV irradiation was observed in wild-type but not in p53-deficient mouse skin. Together, our findings not only demonstrate a novel link between p53 and Notch1 in keratinocyte differentiation upon genotoxic stress but also suggest a novel tumor suppressor mechanism of p53 in the development of squamous cell carcinomas, including HPV-induced tumors.
Collapse
Affiliation(s)
- Takashi Yugawa
- Virology Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | |
Collapse
|
827
|
Sbisà E, Catalano D, Grillo G, Licciulli F, Turi A, Liuni S, Pesole G, De Grassi A, Caratozzolo MF, D'Erchia AM, Navarro B, Tullo A, Saccone C, Gisel A. p53FamTaG: a database resource of human p53, p63 and p73 direct target genes combining in silico prediction and microarray data. BMC Bioinformatics 2007; 8 Suppl 1:S20. [PMID: 17430565 PMCID: PMC1885850 DOI: 10.1186/1471-2105-8-s1-s20] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background The p53 gene family consists of the three genes p53, p63 and p73, which have polyhedral non-overlapping functions in pivotal cellular processes such as DNA synthesis and repair, growth arrest, apoptosis, genome stability, angiogenesis, development and differentiation. These genes encode sequence-specific nuclear transcription factors that recognise the same responsive element (RE) in their target genes. Their inactivation or aberrant expression may determine tumour progression or developmental disease. The discovery of several protein isoforms with antagonistic roles, which are produced by the expression of different promoters and alternative splicing, widened the complexity of the scenario of the transcriptional network of the p53 family members. Therefore, the identification of the genes transactivated by p53 family members is crucial to understand the specific role for each gene in cell cycle regulation. We have combined a genome-wide computational search of p53 family REs and microarray analysis to identify new direct target genes. The huge amount of biological data produced has generated a critical need for bioinformatic tools able to manage and integrate such data and facilitate their retrieval and analysis. Description We have developed the p53FamTaG database (p53 FAMily TArget Genes), a modular relational database, which contains p53 family direct target genes selected in the human genome searching for the presence of the REs and the expression profile of these target genes obtained by microarray experiments. p53FamTaG database also contains annotations of publicly available databases and links to other experimental data. The genome-wide computational search of the REs was performed using PatSearch, a pattern-matching program implemented in the DNAfan tool. These data were integrated with the microarray results we produced from the overexpression of different isoforms of p53, p63 and p73 stably transfected in isogenic cell lines, allowing the comparative study of the transcriptional activity of all the proteins in the same cellular background. p53FamTaG database is available free at Conclusion p53FamTaG represents a unique integrated resource of human direct p53 family target genes that is extensively annotated and provides the users with an efficient query/retrieval system which displays the results of our microarray experiments and allows the export of RE sequences. The database was developed for supporting and integrating high-throughput in silico and experimental analyses and represents an important reference source of knowledge for research groups involved in the field of oncogenesis, apoptosis and cell cycle regulation.
Collapse
Affiliation(s)
- Elisabetta Sbisà
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| | - Domenico Catalano
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| | - Giorgio Grillo
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| | - Flavio Licciulli
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| | - Antonio Turi
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| | - Sabino Liuni
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| | - Graziano Pesole
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
- Dipartimento di Biochimica e Biologia Molecolare, "Ernesto Quagliariello", Università degli Studi di Bari, Via Orabona, 4, 70126 Bari, Italy
| | - Anna De Grassi
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
- Dipartimento di Biochimica e Biologia Molecolare, "Ernesto Quagliariello", Università degli Studi di Bari, Via Orabona, 4, 70126 Bari, Italy
| | - Mariano Francesco Caratozzolo
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
- Dipartimento di Biochimica e Biologia Molecolare, "Ernesto Quagliariello", Università degli Studi di Bari, Via Orabona, 4, 70126 Bari, Italy
| | - Anna Maria D'Erchia
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
- Dipartimento di Biochimica e Biologia Molecolare, "Ernesto Quagliariello", Università degli Studi di Bari, Via Orabona, 4, 70126 Bari, Italy
| | - Beatriz Navarro
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| | - Apollonia Tullo
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| | - Cecilia Saccone
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
- Dipartimento di Biochimica e Biologia Molecolare, "Ernesto Quagliariello", Università degli Studi di Bari, Via Orabona, 4, 70126 Bari, Italy
| | - Andreas Gisel
- Istituto di Tecnologie Biomediche-Sede di Bari, CNR, Via Amendola, 122/D 70126 Bari, Italy
| |
Collapse
|
828
|
Wang ZX, Teh CHL, Kueh JLL, Lufkin T, Robson P, Stanton LW. Oct4 and Sox2 directly regulate expression of another pluripotency transcription factor, Zfp206, in embryonic stem cells. J Biol Chem 2007; 282:12822-30. [PMID: 17344211 DOI: 10.1074/jbc.m611814200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It is well known that Oct4 and Sox2 play an important role in the maintenance of embryonic stem cell pluripotency. These transcription factors bind to regulatory regions within hundreds of target genes to control their expression. Zfp206 is a recently characterized transcription factor that has a role in maintaining stem cell pluripotency. We have demonstrated here that Zfp206 is a direct downstream target of Oct4 and Sox2. Two composite sox-oct binding sites have been identified within the first intron of Zfp206. We have demonstrated binding of Oct4 and Sox2 to this region. In addition, we have shown that Oct4 or Sox2 alone can activate transcription via one of these sox-oct elements, although the presence of both Oct4 and Sox2 gave rise to a synergistic effect. These studies extend our understanding of the transcriptional network that operates to regulate the differentiation potential of embryonic stem cells.
Collapse
Affiliation(s)
- Zheng-Xu Wang
- Department of Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore 138672
| | | | | | | | | | | |
Collapse
|
829
|
Li L, Liang Y, Bass RL. GAPWM: a genetic algorithm method for optimizing a position weight matrix. Bioinformatics 2007; 23:1188-94. [PMID: 17341493 DOI: 10.1093/bioinformatics/btm080] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
MOTIVATION Position weight matrices (PMWs) are simple models commonly used in motif-finding algorithms to identify short functional elements, such as cis-regulatory motifs, on genes. When few experimentally verified motifs are available, estimation of the PWM may be poor. The resultant PWM may not reliably discriminate a true motif from a false one. While experimentally identifying such motifs remains time-consuming and expensive, low-resolution binding data from techniques such as ChIP-on-chip and ChIP-PET have become available. We propose a novel but simple method to improve a poorly estimated PWM using ChIP data. METHODOLOGY Starting from an existing PWM, a set of ChIP sequences, and a set of background sequences, our method, GAPWM, derives an improved PWM via a genetic algorithm that maximizes the area under the receiver operating characteristic (ROC) curve. GAPWM can easily incorporate prior information such as base conservation. We tested our method on two PMWs (Oct4/Sox2 and p53) using three recently published ChIP data sets (human Oct4, mouse Oct4 and human p53). RESULTS GAPWM substantially increased the sensitivity/specificity of a poorly estimated PWM and further improved the quality of a good PWM. Furthermore, it still functioned when the starting PWM contained a major error. The ROC performance of GAPWM compared favorably with that of MEME and others. With increasing availability of ChIP data, our method provides an alternative for obtaining high-quality PWMs for genome-wide identification of transcription factor binding sites. AVAILABILITY The C source code and all data used in this report are available at http://dir.niehs.nih.gov/dirbb/gapwm. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Leping Li
- Biostatistics Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA.
| | | | | |
Collapse
|
830
|
Garg M, Chaurasiya D, Rana R, Jagadish N, Kanojia D, Dudha N, Kamran N, Salhan S, Bhatnagar A, Suri S, Gupta A, Suri A. Sperm-associated antigen 9, a novel cancer testis antigen, is a potential target for immunotherapy in epithelial ovarian cancer. Clin Cancer Res 2007; 13:1421-1428. [PMID: 17332284 DOI: 10.1158/1078-0432.ccr-06-2340] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE Cancer testis antigens are a group of tumor antigens with gene expression restricted to male germ cells in the testis and in various cancerous tissues. Recently, we reported a novel testis-specific sperm-associated antigen 9 (SPAG9) gene, a new member of the c-Jun NH(2)-terminal kinase-interacting protein family, having functional role in sperm-egg fusion and mitogen-activated protein kinase signaling pathway. National Center for Biotechnology Information Blast searches revealed SPAG9 nucleotide sequence similarities with expressed sequence tags of various cancerous tissues. In an effort to examine the clinical utility of SPAG9, we investigated the SPAG9 mRNA and protein expression in epithelial ovarian cancer (EOC). Humoral immune response to SPAG9 was also evaluated in EOC patients. EXPERIMENTAL DESIGN We determined the expression profile of SPAG9 transcript by reverse transcription-PCR and RNA in situ hybridization and SPAG9 protein expression by immunohistochemistry in EOC specimens and human ovarian cancer cell lines. Using ELISA and Western blotting, we analyzed specific antibodies for SPAG9 in sera from patients with EOC. RESULTS SPAG9 mRNA and protein expression was detected in 90% of EOC tissues and in all three human ovarian cancer cell lines. Specific SPAG9 antibodies were detected in 67% of EOC patients and not in sera from healthy individuals. CONCLUSIONS Our findings indicate that SPAG9 is highly expressed in EOC and immunogenic in patients. Humoral immune response against SPAG9 in early stages of EOC suggests its important role in early diagnostics. These results collectively suggest that SPAG9, a novel member of cancer testis antigen family, could be a potential target for the development of diagnostic and therapeutic methods in EOC.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Antibodies, Neoplasm/blood
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor/analysis
- Blotting, Western
- Cell Line, Tumor
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Gene Expression
- Gene Expression Profiling
- Humans
- Immunohistochemistry
- Immunotherapy
- In Situ Hybridization
- Neoplasms, Glandular and Epithelial/blood
- Neoplasms, Glandular and Epithelial/immunology
- Neoplasms, Glandular and Epithelial/metabolism
- Ovarian Neoplasms/blood
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/metabolism
- RNA, Messenger/analysis
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Manoj Garg
- Genes and Proteins Laboratory, National Institute of Immunology, New Delhi, India
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
831
|
Wang P, Yu J, Zhang L. The nuclear function of p53 is required for PUMA-mediated apoptosis induced by DNA damage. Proc Natl Acad Sci U S A 2007; 104:4054-9. [PMID: 17360476 PMCID: PMC1820707 DOI: 10.1073/pnas.0700020104] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The tumor suppressor p53 can induce apoptosis by activating gene expression in the nucleus, or by directly permeabilizing mitochondria in the cytoplasm. It has been shown that PUMA, a downstream target of p53 and a BH3-only Bcl-2 family member, plays an essential role in apoptosis induced by both nuclear and cytoplasmic p53. To understand how PUMA does so, we used homologous recombination to delete the binding sites of p53 in the promoter of PUMA in human colorectal cancer cells. As a result, the induction of PUMA and apoptosis in response to p53 and DNA-damaging agents were abrogated. Transcription coactivator recruitment and histone modifications in the PUMA promoter were suppressed. However, induction of PUMA and apoptosis in response to non-DNA-damaging stimuli were unaffected. These results indicate that the binding of nuclear p53 to the specific sites within the PUMA promoter is essential for its ability to induce apoptosis and is likely to be required for its tumor suppressive capacity.
Collapse
Affiliation(s)
- Peng Wang
- Departments of Pharmacology and Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213
| | - Jian Yu
- Departments of Pharmacology and Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213
| | - Lin Zhang
- Departments of Pharmacology and Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
832
|
Tavtigian SV, Pierotti MA, Børresen-Dale AL. International Agency for Research on Cancer workshop on 'Expression array analyses in breast cancer taxonomy'. Breast Cancer Res 2007; 8:303. [PMID: 17096863 PMCID: PMC1797037 DOI: 10.1186/bcr1609] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In May 2006, a workshop on Expression array analyses in breast cancer taxonomy was held at the International Agency for Research on Cancer (IARC). The workshop covered an array of topics from the validity of the currently defined breast tumor subtypes and other expression profile-based signatures to the technical limitations of expression analysis and the types of platforms on which these omics results will eventually reach clinical practice. Overall, the workshop participants believed firmly that tumor taxonomy is likely to yield improved prognostic and predictive markers. Even so, further standardization and validation are required before clinical trials are set in motion.
Collapse
Affiliation(s)
- Sean V Tavtigian
- Genetic Susceptibility Group, Genetics and Epidemiology Cluster, International Agency for Research on Cancer World Health Organization 150 Cours Albert-Thomas, 69372 Lyon Cedex 08, France
| | - Marco A Pierotti
- Scientific Direction, Fondazione IRCCS Istituto Nazionale dei Tumori Via Venezian, 120133 Milan, Italy
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Rikshospitalet-Radiumhospitalet Medical Center, Oslo, Norway
| |
Collapse
|
833
|
Tu Y, Wu W, Wu T, Cao Z, Wilkins R, Toh BH, Cooper ME, Chai Z. Antiproliferative autoantigen CDA1 transcriptionally up-regulates p21(Waf1/Cip1) by activating p53 and MEK/ERK1/2 MAPK pathways. J Biol Chem 2007; 282:11722-31. [PMID: 17317670 DOI: 10.1074/jbc.m609623200] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We previously reported that overexpression of cell division autoantigen 1 (CDA1) in HeLa cells arrests cell growth and inhibits DNA synthesis at S-phase. Here we show that CDA1-induced arrest of cell growth is accompanied by increases in protein and mRNA levels of the cyclin-dependent kinase (Cdk) inhibitor protein, p21(Waf1/Cip1) (p21). Both p21 induction and cell growth arrest are reversed when CDA1 expression is inhibited. CDA1 also increases p53 protein, but not its mRNA, in a time- and dose-dependent manner. MDM2, a ubiquitin ligase regulating p53 degradation, is inactivated by CDA1, suggesting that p53 protein accumulation is due to decreased protein degradation. Knockdown of p53, using siRNA targeting two sites of p53 mRNA, abrogates transcriptional induction of p21 by CDA1. Deletion of the p53 responsive element in the distal region of p21 promoter attenuates promoter activity in response to CDA1. DNA damage caused by camptothecin treatment increases mRNA and protein levels of CDA1, accompanied by induction of p53. The DNA damage-induced p53 induction is markedly attenuated by CDA1 knockdown. CDA1 induces phosphorylation of ERK1/2(p44/42), an activity blocked by PD98059 and U0126, inhibitors of the upstream kinase MEK1/2. The MEK inhibitors also block induction of p21 mRNA and abrogate p21 promoter activity stimulated by CDA1. Cell cycle kinases, Cdk1, -2, -4, and -6 are inhibited by CDA1 overexpression. We conclude that CDA1 induces p53- and MEK/ERK1/2 MAPK-dependent expression of p21 by acting through the p53 responsive element in the p21 promoter and that this contributes to its antiproliferative activity.
Collapse
Affiliation(s)
- Yugang Tu
- Diabetes and Metabolism Division, Baker Heart Research Institute, Melbourne, Victoria 3004, Australia
| | | | | | | | | | | | | | | |
Collapse
|
834
|
Yochum GS, McWeeney S, Rajaraman V, Cleland R, Peters S, Goodman RH. Serial analysis of chromatin occupancy identifies beta-catenin target genes in colorectal carcinoma cells. Proc Natl Acad Sci U S A 2007; 104:3324-9. [PMID: 17360646 PMCID: PMC1805576 DOI: 10.1073/pnas.0611576104] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Most instances of colorectal cancer are due to abnormalities in the Wnt signaling pathway, resulting in nuclear accumulation of beta-catenin. beta-Catenin activates transcription of target genes primarily by associating with the T cell factor/lymphoid enhancer-binding factor (TCF/Lef) family of transcription factors. In this report, we use serial analysis of chromatin occupancy (SACO) to identify 412 high-confidence beta-catenin targets in HCT116 colorectal carcinoma cells. Of these targets, 84% contained a consensus TCF motif and were occupied by TCF4 in vivo. Examination of the flanking 5-bp residues in each consensus revealed motif-specific enrichment at neighboring sites. beta-Catenin binding was localized to the 5' promoters, internal regions, and 3' UTRs of protein-coding genes. Furthermore, 15 components of the canonical Wnt pathway were identified as beta-catenin target genes, suggesting that feed-forward and feedback mechanisms exist to modulate the Wnt signal in colon cancer cells.
Collapse
Affiliation(s)
- Gregory S. Yochum
- *Vollum Institute
- To whom correspondence may be addressed at:
Vollum Institute, L-474, Oregon Health & Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239. E-mail: or
| | - Shannon McWeeney
- Division of Biostatistics, Department of Public Health and Preventative Medicine, and
- Oregon Health & Science University Cancer Institute, Oregon Health & Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239
| | - Veena Rajaraman
- Oregon Health & Science University Cancer Institute, Oregon Health & Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239
| | | | | | - Richard H. Goodman
- *Vollum Institute
- To whom correspondence may be addressed at:
Vollum Institute, L-474, Oregon Health & Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239. E-mail: or
| |
Collapse
|
835
|
Gustafsdottir SM, Schlingemann J, Rada-Iglesias A, Schallmeiner E, Kamali-Moghaddam M, Wadelius C, Landegren U. In vitro analysis of DNA-protein interactions by proximity ligation. Proc Natl Acad Sci U S A 2007; 104:3067-72. [PMID: 17360610 PMCID: PMC1805562 DOI: 10.1073/pnas.0611229104] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Protein-binding DNA sequence elements encode a variety of regulated functions of genomes. Information about such elements is currently in a state of rapid growth, but improved methods are required to characterize the sequence specificity of DNA-binding proteins. We have established an in vitro method for specific and sensitive solution-phase analysis of interactions between proteins and nucleic acids in nuclear extracts, based on the proximity ligation assay. The reagent consumption is very low, and the excellent sensitivity of the assay enables analysis of as few as 1-10 cells. We show that our results are highly reproducible, quantitative, and in good agreement with both EMSA and predictions obtained by using a motif finding software. This assay can be a valuable tool to characterize in-depth the sequence specificity of DNA-binding proteins and to evaluate effects of polymorphisms in known transcription factor binding sites.
Collapse
Affiliation(s)
- Sigrun M Gustafsdottir
- Rudbeck Laboratory, Department of Genetics and Pathology, Uppsala University, Dag Hammarskjöldsväg 20, SE-75185 Uppsala, Sweden.
| | | | | | | | | | | | | |
Collapse
|
836
|
Abstract
The tumor suppressor protein p53 is mutated in over half of human cancers. Despite 25 years of study, the complex regulation of this protein remains unclear. After serendipitously detecting RNA binding by p53 in the yeast three-hybrid system (Y3H), we are exploring the specificity and function of this interaction. Electrophoretic mobility shift assays show that full-length p53 binds equally to RNAs that are strongly distinguished in the Y3H. RNA binding blocks sequence-specific DNA binding by p53. The C-terminus of p53 is necessary and sufficient for strong RNA interaction in vitro. Mouse and human C-terminal p53 peptides have different affinities for RNA, and an acetylated human p53 C-terminal peptide does not bind RNA. Circular dichroism spectroscopy of p53 peptides shows that RNA binding does not induce a structural change in the p53 C-terminal peptide, and C-terminal peptides do not detectably affect the structure of RNA. These results demonstrate that p53 binds RNA with little sequence specificity, RNA binding has the potential to regulate DNA binding, and RNA-p53 interactions can be regulated by acetylation of the p53 C-terminus.
Collapse
Affiliation(s)
- Kasandra J-L Riley
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
837
|
Mattia M, Gottifredi V, McKinney K, Prives C. p53-Dependent p21 mRNA elongation is impaired when DNA replication is stalled. Mol Cell Biol 2007; 27:1309-20. [PMID: 17158927 PMCID: PMC1800727 DOI: 10.1128/mcb.01520-06] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 10/05/2006] [Accepted: 11/30/2006] [Indexed: 11/20/2022] Open
Abstract
We have previously reported that when DNA replication is blocked in some human cell lines, p53 is impaired in its ability to induce a subset of its key target genes, including p21(WAF1/CIP1). Here, we investigated the reason for this impairment by comparing the effects of two agents, hydroxyurea (HU), which arrests cells in early S phase and impairs induction of p21, and daunorubicin, which causes a G(2) block and leads to robust activation of p21 by p53. HU treatment was shown to inhibit p21 mRNA transcription rather than alter its mRNA stability. Nevertheless, chromatin immunoprecipitation assays revealed that HU impacts neither p53 binding nor acetylation of histones H3 and H4 within the p21 promoter. Furthermore, recruitment of the TFIID/TATA-binding protein complex and the large subunit of RNA polymerase II (RNA Pol II) are equivalent after HU and daunorubicin treatments. Relative to daunorubicin treatment, however, transcription elongation of the p21 gene is significantly impaired in cells treated with HU, as evidenced by reduced occupancy of RNA Pol II at regions downstream of the start site. Likewise, in the p21 downstream region after administration of HU, there is less of a specifically phosphorylated form of RNA Pol II (Pol II-C-terminal domain serine 2P) which occurs only when the polymerase is elongating RNA. We propose that while the DNA replication checkpoint is unlikely to regulate the assembly of a p21 promoter initiation complex, it signals to one or more factors involved in the process of transcriptional elongation.
Collapse
Affiliation(s)
- Melissa Mattia
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | | | | |
Collapse
|
838
|
Ohki R, Kawase T, Ohta T, Ichikawa H, Taya Y. Dissecting functional roles of p53 N-terminal transactivation domains by microarray expression analysis. Cancer Sci 2007; 98:189-200. [PMID: 17233836 PMCID: PMC11159457 DOI: 10.1111/j.1349-7006.2006.00375.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The p53 protein exerts its tumor suppressive function mainly by acting as a transcription activator. Two transactivation domains (TADs) located at the amino-terminus of p53 are required for transcription activation, and the activity of TADs is tightly regulated by post-translational modifications, such as phosphorylation. We attempted to dissect the functions of the two TADs and phosphorylation within the TADs by analyzing p53 target genes induced by full-length p53 (FL-p53), N-terminally deleted p53 isoform lacking the first TAD (Delta1stTAD) and p53 carrying point mutations at all serine residues within the two TADs (TAD-S/A). By performing a comprehensive survey by employing microarray expression analysis, the induction of target genes by FL-p53, Delta1stTAD and TAD-S/A was analyzed. All p53s showed different target gene induction patterns, suggesting the importance of the two TADs and phosphorylation within the TADs in target gene induction. Although Delta1stTAD showed a marked decrease in the ability to induce genes induced by FL-p53, Delta1stTAD induced many apoptosis-related genes that were not induced by FL-p53, suggesting the roles of these Delta1stTAD-induced genes in Delta1stTAD-dependent apoptosis. Approximately 80% of genes induced by FL-p53 were not induced by TAD-S/A, including 29 previously reported p53 target genes such as Hdm2 and Bax, emphasizing the importance of phosphorylation within the TADs. These results demonstrate the significance of the regulation and differential roles of the N-terminal TADs in p53 transcriptional activity.
Collapse
Affiliation(s)
- Rieko Ohki
- Radiobiology Division, National Cancer Center Research Institute, Tsujiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | |
Collapse
|
839
|
Yan J, Jiang J, Lim CA, Wu Q, Ng HH, Chin KC. BLIMP1 regulates cell growth through repression of p53 transcription. Proc Natl Acad Sci U S A 2007; 104:1841-6. [PMID: 17264218 PMCID: PMC1784014 DOI: 10.1073/pnas.0605562104] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tight regulation of p53 is essential for maintaining normal cell growth. Here we report that BLIMP1 acts in an autoregulatory feedback loop that controls p53 activity through repression of p53 transcription. p53 binds to and positively regulates BLIMP1, which encodes for a known B cell transcriptional repressor. Knockdown of BLIMP1 by siRNA results in both apoptosis and growth arrest in human colon cancer cells and cell-cycle arrest in primary human fibroblasts. Interestingly, the levels of both p53 mRNA and protein are substantially increased after BLIMP1 depletion, which is accompanied by the induction of p53 target genes. Importantly, the apoptosis induced by BLIMP1 depletion in HCT116 cells is largely abrogated in cells lacking p53 or in cells depleted in p53 by siRNA. We further demonstrate that BLIMP1 binds to the p53 promoter and represses p53 transcription, and this provides a mechanistic explanation for the induction of p53 response in cells depleted of BLIMP1. Hence, suppression of p53 transcription is a crucial function of endogenous BLIMP1 and is essential for normal cell growth.
Collapse
Affiliation(s)
- Junli Yan
- *Gene Expression Laboratory, Genome Institute of Singapore, Singapore 138672
| | - Jianming Jiang
- Laboratory of Immunology and Virology and
- *Gene Expression Laboratory, Genome Institute of Singapore, Singapore 138672
- Department of Biological Sciences, National University of Singapore, Singapore 117543; and
| | - Ching Aeng Lim
- *Gene Expression Laboratory, Genome Institute of Singapore, Singapore 138672
| | - Qiang Wu
- *Gene Expression Laboratory, Genome Institute of Singapore, Singapore 138672
| | - Huck-Hui Ng
- *Gene Expression Laboratory, Genome Institute of Singapore, Singapore 138672
- Department of Biological Sciences, National University of Singapore, Singapore 117543; and
- To whom correspondence may be addressed at:
Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672. E-mail:
or
| | - Keh-Chuang Chin
- Laboratory of Immunology and Virology and
- Singapore Immunology Network, Immunos, Singapore 138672
- To whom correspondence may be addressed at:
Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672. E-mail:
or
| |
Collapse
|
840
|
Yang A, Zhu Z, Kapranov P, McKeon F, Church GM, Gingeras TR, Struhl K. Relationships between p63 binding, DNA sequence, transcription activity, and biological function in human cells. Mol Cell 2007; 24:593-602. [PMID: 17188034 DOI: 10.1016/j.molcel.2006.10.018] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2006] [Revised: 08/08/2006] [Accepted: 10/12/2006] [Indexed: 01/10/2023]
Abstract
Using tiled microarrays covering the entire human genome, we identify approximately 5800 target sites for p63, a p53 homolog essential for stratified epithelial development. p63 targets are enriched for genes involved in cell adhesion, proliferation, death, and signaling pathways. The quality of the derived DNA sequence motif for p63 targets correlates with binding strength binding in vivo, but only a small minority of motifs in the genome is bound by p63. Conversely, many p63 targets have motif scores expected for random genomic regions. Thus, p63 binding in vivo is highly selective and often requires additional factors beyond the simple protein-DNA interaction. There is a significant, but complex, relationship between p63 target sites and p63-responsive genes, with DeltaNp63 isoforms being linked to transcriptional activation. Many p63 binding regions are evolutionarily conserved and/or associated with sequence motifs for other transcription factors, suggesting that a substantial portion of p63 sites is biologically relevant.
Collapse
Affiliation(s)
- Annie Yang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
841
|
Menendez D, Inga A, Snipe J, Krysiak O, Schönfelder G, Resnick MA. A single-nucleotide polymorphism in a half-binding site creates p53 and estrogen receptor control of vascular endothelial growth factor receptor 1. Mol Cell Biol 2007; 27:2590-600. [PMID: 17242190 PMCID: PMC1899907 DOI: 10.1128/mcb.01742-06] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interactions between master regulatory pathways provide higher-order controls for cellular regulation. Recently, we reported a C-->T single-nucleotide polymorphism (SNP) in the vascular endothelial growth factor receptor 1 (VEGFR-1/Flt1) promoter that merges human VEGF and p53 pathways. This finding suggested a new layer in environmental controls of a pathway relevant to several diseases. The Flt1-T SNP created what appeared to be a half-site p53 target response element (RE). The absence of information about p53 gene responsiveness mediated by half-site REs led us to address how it influences Flt1 expression. We now identify a second regulatory sequence comprising a partial RE for estrogen receptors (ERs) upstream of the p53 binding site. Surprisingly, this provides for synergistic stimulation of transcription specifically at the Flt1-T allele through the combined action of ligand-bound ER and stress-induced p53. In addition to demonstrating direct control of Flt1 expression by ER and p53 proteins acting as sequence-specific transcription factors at half-site REs, we establish a new interaction between three master regulatory pathways, p53, ER, and VEGF. The mechanism of joint regulation through half-sites is likely relevant to transcriptional control of other targets and expands the number of genes that may be directly controlled in master regulatory networks.
Collapse
Affiliation(s)
- Daniel Menendez
- Laboratory of Molecular Genetics MD3-01, National Institute of Environmental Health Sciences, 111 Alexander Drive, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | |
Collapse
|
842
|
Hudson ME, Snyder M. High-throughput methods of regulatory element discovery. Biotechniques 2007; 41:673, 675, 677 passim. [PMID: 17191608 DOI: 10.2144/000112322] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
With the number of organisms whose genomes have been sequenced, a vast amount of information concerning the genetic structure of an organism's genome has been collected. However, effective experiment means to study how this information is accessed have only recently been developed. In this review, three basic methods for identifying regions of protein-DNA interaction will be introduced. The first two, chromatin immunoprecipitation (ChIP)-chip and ChIP-PET (for paired-end ditag), rely on the enrichment provided by chromosomal immunoprecipitation to interrogate the genomic sequence for the interaction sites of a protein of interest. In contrast, protein microarrays allow the identification of DNA binding protein that interacts with a DNA sequence of interest. These complementary methods of exploring protein-DNA interactions will increase our fundamental knowledge of how the information contained within the genome sequence is accessed and processed.
Collapse
|
843
|
Parker DS, Blauwkamp T, Cadigan KM. Wnt/β‐catenin‐mediated transcriptional regulation. WNT SIGNALING IN EMBRYONIC DEVELOPMENT 2007. [DOI: 10.1016/s1574-3349(06)17001-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
844
|
p53, BRCA1 and breast Cancer chemoresistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 608:70-86. [PMID: 17993233 DOI: 10.1007/978-0-387-74039-3_5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tumor suppressor genes p53 and BRCA1 are involved in hereditary as well as sporadic breast cancer development and therapeutic responses. While p53 mutations contribute to resistance to chemo- and radiotherapy, BRCA1 dysfunction leads to enhanced sensitivity to DNA damaging therapeutic agents. The biochemical pathways used by p53 and BRCA1 for signaling tumor suppression involve some cross-talk including repression of BRCA1 transcription by p53 and altered selectivity of p53-dependent gene activation by BRCA1. In this chapter we review clinical and preclinical data implicating p53 and BRCA1 in breast cancer chemosensitivity. We discuss the known signaling pathways downstream of p53 or BRCA1 that contribute to their modulation of therapeutic responses, and we discuss the implications of p53 or BRCA1 mutation in therapeutic design.
Collapse
|
845
|
Golubovskaya VM, Cance WG. Focal adhesion kinase and p53 signaling in cancer cells. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 263:103-53. [PMID: 17725966 DOI: 10.1016/s0074-7696(07)63003-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The progression of human cancer is characterized by a process of tumor cell motility, invasion, and metastasis to distant sites, requiring the cancer cells to be able to survive the apoptotic pressures of anchorage-independent conditions. One of the critical tyrosine kinases linked to these processes of tumor invasion and survival is the focal adhesion kinase (FAK). FAK was first isolated from human tumors, and FAK mRNA was found to be upregulated in invasive and metastatic human breast and colon cancer samples. Recently, the FAK promoter was cloned, and it has been found to contain p53-binding sites. p53 inhibits FAK transcription, and recent data show direct binding of FAK and p53 proteins in vitro and in vivo. The structure of FAK and p53, proteins interacting with FAK, and the role of FAK in tumorigenesis and FAK-p53-related therapy are reviewed. This review focuses on FAK signal transduction pathways, particularly on FAK and p53 signaling, revealing a new paradigm in cell biology, linking signaling from the extracellular matrix to the nucleus.
Collapse
Affiliation(s)
- Vita M Golubovskaya
- Department of Surgery, University of Florida School of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | | |
Collapse
|
846
|
Pregizer S, Barski A, Gersbach CA, García AJ, Frenkel B. Identification of novel Runx2 targets in osteoblasts: Cell type-specific BMP-dependent regulation of Tram2. J Cell Biochem 2007; 102:1458-71. [PMID: 17486635 DOI: 10.1002/jcb.21366] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Runx2 is an osteoblast master transcription factor and a target for bone morphogenetic protein (BMP) signaling, but our knowledge of events downstream of Runx2 is limited. In this study, we used ChIP Display to discover seven novel genomic regions occupied by Runx2 in living MC3T3-E1 osteoblastic cells. Six of these regions are found within or up to 1-kb away from annotated genes, but only two are found within 5'-gene flanking sequences. One of the newly identified Runx2 target genes is Tram2, whose product facilitates proper folding of type I collagen. We demonstrate that Tram2 mRNA is suppressed in non-osteoblasts when Runx2 is over-expressed, and that this suppression is alleviated upon treatment with BMP-2. Moreover, we show that BMP-induced Runx2 expression in the C3H10T1/2, ST2, C2C12, and MC3T3-E1 cell lines coincides with an increase in Tram2 mRNA levels. Thus, Runx2 may regulate Tram2 expression in a BMP-dependent manner, and Tram2 may participate in the overall osteogenic function of Runx2. Among the other Runx2 target genes discovered in this study are Lnx2, an intracellular scaffolding protein that may play a role in Notch signaling, and Tnfrsf12a, a Tumor Necrosis Factor receptor family member that influences both osteoblast and osteoclast differentiation. Expanding our knowledge of Runx2 target genes, and manipulation of these genes, are warranted to better understand the regulation of osteoblast function and to provide opportunities for the development of new bone anabolics.
Collapse
Affiliation(s)
- Steven Pregizer
- Department of Biochemistry & Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | |
Collapse
|
847
|
Abstract
Several technologies that emerged in the post-genomic era have been particularly useful in dissecting the molecular mechanisms of complex biological processes through the systems approach. Here, we review how three of these technologies, namely transcriptional profiling, large-scale RNA interference (RNAi) and genome-wide location analysis of protein-DNA interactions, have been used in the study of ageing in metazoans. We also highlight recent developments of these three technologies and how these developments are applicable to ageing research.
Collapse
Affiliation(s)
- Ching-Aeng Lim
- Gene Regulation Laboratory, Genome Institute of Singapore, Singapore 138672.
| | | |
Collapse
|
848
|
Johung K, Goodwin EC, DiMaio D. Human papillomavirus E7 repression in cervical carcinoma cells initiates a transcriptional cascade driven by the retinoblastoma family, resulting in senescence. J Virol 2006; 81:2102-16. [PMID: 17182682 PMCID: PMC1865941 DOI: 10.1128/jvi.02348-06] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
This work demonstrates a central role for the retinoblastoma (Rb) family in driving the transcriptional program of induced and replicative senescence. HeLa cervical carcinoma cells rapidly undergo senescence when the human papillomavirus (HPV) type 18 E7 gene in these cells is repressed by the bovine papillomavirus (BPV) E2 protein. This senescence response requires the endogenous Rb pathway but not the p53 pathway. Microarray analysis 6 days after BPV E2 introduction into HeLa cells identified 224 cellular genes induced by E7 repression and 354 repressed genes. Many repressed genes were involved in cell cycle progression, and numerous induced genes encoded lysosomal proteins. These gene expression changes were blocked by constitutive expression of the wild-type HPV16 E7 or adenovirus E1A gene, but not by E7 or E1A mutants defective for Rb binding. Short hairpin RNAs targeting the Rb family also inhibited these gene expression changes and blocked senescence. Therefore, surprisingly, the transcriptional response to BPV E2 expression was entirely dependent on E7 repression and activation of the Rb family, and the BPV E2 protein did not directly affect the expression of cellular genes. Activation of the Rb family repressed E2F-responsive genes and stimulated transcriptional activators, thereby mobilizing multiple signals, such as repression of B-MYB and DEK, that were independently sufficient to induce senescence. There was extensive overlap between the transcriptional profiles of senescent, late-passage primary human fibroblasts and senescent cervical carcinoma cells, suggesting that this Rb family-mediated transcriptional cascade also plays a central role in replicative senescence.
Collapse
Affiliation(s)
- Kimberly Johung
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | |
Collapse
|
849
|
Lokshin M, Li Y, Gaiddon C, Prives C. p53 and p73 display common and distinct requirements for sequence specific binding to DNA. Nucleic Acids Res 2006; 35:340-52. [PMID: 17170001 PMCID: PMC1802567 DOI: 10.1093/nar/gkl1047] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Revised: 11/03/2006] [Accepted: 11/03/2006] [Indexed: 12/11/2022] Open
Abstract
Although p53 and p73 share considerable homology in their DNA-binding domains, there have been few studies examining their relative interactions with DNA as purified proteins. Comparing p53 and p73beta proteins, our data show that zinc chelation by EDTA is significantly more detrimental to the ability of p73beta than of p53 to bind DNA, most likely due to the greater effect that the loss of zinc has on the conformation of the DNA-binding domain of p73. Furthermore, prebinding to DNA strongly protects p73beta but not p53 from chelation by EDTA suggesting that DNA renders the core domain of p73 less accessible to its environment. Further exploring these biochemical differences, a five-base sub-sequence was identified in the p53 consensus binding site that confers a greater DNA-binding stability on p73beta than on full-length p53 in vitro. Surprisingly, p53 lacking its C-terminal non-specific DNA-binding domain (p53Delta30) demonstrates the same sequence discrimination as does p73beta. In vivo, both p53 and p73beta exhibit higher transactivation of a reporter with a binding site containing this sub-sequence, suggesting that lower in vitro dissociation translates to higher in vivo transactivation of sub-sequence-containing sites.
Collapse
Affiliation(s)
- Maria Lokshin
- Department of Biological Sciences, Columbia UniversityNew York, NY 10027, USA
- U692 INSERM-Universite Louis Pasteur, Signalisations Moleculaires et Neurodegenerescence11 rue Human, 67085 Strasbourg, France
| | - Yingchun Li
- Department of Biological Sciences, Columbia UniversityNew York, NY 10027, USA
- U692 INSERM-Universite Louis Pasteur, Signalisations Moleculaires et Neurodegenerescence11 rue Human, 67085 Strasbourg, France
| | - Christian Gaiddon
- U692 INSERM-Universite Louis Pasteur, Signalisations Moleculaires et Neurodegenerescence11 rue Human, 67085 Strasbourg, France
| | - Carol Prives
- To whom correspondence should be addressed. Tel: +1 212 854 2557; Fax: +1 212 865 8246;
| |
Collapse
|
850
|
Khattra J, Delaney AD, Zhao Y, Siddiqui A, Asano J, McDonald H, Pandoh P, Dhalla N, Prabhu AL, Ma K, Lee S, Ally A, Tam A, Sa D, Rogers S, Charest D, Stott J, Zuyderduyn S, Varhol R, Eaves C, Jones S, Holt R, Hirst M, Hoodless PA, Marra MA. Large-scale production of SAGE libraries from microdissected tissues, flow-sorted cells, and cell lines. Genome Res 2006; 17:108-16. [PMID: 17135571 PMCID: PMC1716260 DOI: 10.1101/gr.5488207] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We describe the details of a serial analysis of gene expression (SAGE) library construction and analysis platform that has enabled the generation of >298 high-quality SAGE libraries and >30 million SAGE tags primarily from sub-microgram amounts of total RNA purified from samples acquired by microdissection. Several RNA isolation methods were used to handle the diversity of samples processed, and various measures were applied to minimize ditag PCR carryover contamination. Modifications in the SAGE protocol resulted in improved cloning and DNA sequencing efficiencies. Bioinformatic measures to automatically assess DNA sequencing results were implemented to analyze the integrity of ditag structure, linker or cross-species ditag contamination, and yield of high-quality tags per sequence read. Our analysis of singleton tag errors resulted in a method for correcting such errors to statistically determine tag accuracy. From the libraries generated, we produced an essentially complete mapping of reliable 21-base-pair tags to the mouse reference genome sequence for a meta-library of approximately 5 million tags. Our analyses led us to reject the commonly held notion that duplicate ditags are artifacts. Rather than the usual practice of discarding such tags, we conclude that they should be retained to avoid introducing bias into the results and thereby maintain the quantitative nature of the data, which is a major theoretical advantage of SAGE as a tool for global transcriptional profiling.
Collapse
Affiliation(s)
- Jaswinder Khattra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Allen D. Delaney
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Yongjun Zhao
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Asim Siddiqui
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Jennifer Asano
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Helen McDonald
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Pawan Pandoh
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Noreen Dhalla
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Anna-liisa Prabhu
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Kevin Ma
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Stephanie Lee
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Adrian Ally
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Angela Tam
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Danne Sa
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Sean Rogers
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - David Charest
- Genome British Columbia, Vancouver, British Columbia V5Z 1C6, Canada
| | - Jeff Stott
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Scott Zuyderduyn
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Richard Varhol
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Connie Eaves
- Terry Fox Laboratory, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Steven Jones
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Robert Holt
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Martin Hirst
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Pamela A. Hoodless
- Terry Fox Laboratory, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
- Corresponding author.E-mail ; fax (604) 877-6085
| |
Collapse
|