3701
|
Progesterone induction of the 11beta-hydroxysteroid dehydrogenase type 2 promoter in breast cancer cells involves coordinated recruitment of STAT5A and progesterone receptor to a distal enhancer and polymerase tracking. Mol Cell Biol 2008; 28:3830-49. [PMID: 18378698 DOI: 10.1128/mcb.01217-07] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Steroid hormone receptors regulate gene expression, interacting with target DNA sequences but also activating cytoplasmic signaling pathways. Using the human 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) gene as a model, we have investigated the contributions of both effects on a human progesterone-responsive promoter in breast cancer cells. Chromatin immunoprecipitation has identified two different mechanisms of hormone-induced progesterone receptor (PR) recruitment to the 11beta-HSD2 promoter: (i) direct PR binding to DNA at the proximal promoter, abrogated when PR contains a mutated DNA binding domain (DBD), and (ii) STAT5A (signal transducer and activator of transcription 5A)-mediated recruitment of PR to an upstream distal region, impaired by AG490, a JAK/STAT pathway inhibitor. The JAK/STAT inhibitor, as well as expression of dominant-negative STAT5A, impairs hormone induction of 11beta-HSD2. On the other hand, the DBD-mutated PR fully supports 11beta-HSD2 expression. These results, along with data from a deletion analysis, indicate that the distal region is crucial for hormone regulation of 11beta-HSD2. We show active RNA polymerase II tracking from the distal region upon PR and STAT5A binding, concomitant with synthesis of noncoding, hormone-dependent RNAs, suggesting that this region works as a hormone-dependent transcriptional enhancer. In conclusion, coordination of PR transcriptional effects and cytoplasmic signaling activation, in particular the JAK/STAT pathway, are critical in regulating progestin-induced endogenous 11beta-HSD2 gene expression in breast cancer cells. This is not unique to this promoter, as AG490 also alters the expression of other progesterone-regulated genes.
Collapse
|
3702
|
Approaches to comparative sequence analysis: towards a functional view of vertebrate genomes. Nat Rev Genet 2008; 9:303-13. [PMID: 18347593 DOI: 10.1038/nrg2185] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The comparison of genomic sequences is now a common approach to identifying and characterizing functional regions in vertebrate genomes. However, for theoretical reasons and because of practical issues, the generation of these data sets is non-trivial and can have many pitfalls. We are currently seeing an explosion of comparative sequence data, the benefits and limitations of which need to be disseminated to the scientific community. This Review provides a critical overview of the different types of sequence data that are available for analysis and of contemporary comparative sequence analysis methods, highlighting both their strengths and limitations. Approaches to determining the biological significance of constrained sequence are also explored.
Collapse
|
3703
|
Abstract
Alternative splicing is a well-characterized mechanism by which multiple transcripts are generated from a single mRNA precursor. By allowing production of several protein isoforms from one pre-mRNA, alternative splicing contributes to proteomic diversity. But what do we know about the origin of this mechanism? Do the same evolutionary forces apply to alternatively and constitutively splice exons? Do similar forces act on all types of alternative splicing? Are the products generated by alternative splicing functional? Why is "improper" recognition of exons and introns allowed by the splicing machinery? In this review, we summarize the current knowledge regarding these issues from an evolutionary perspective.
Collapse
Affiliation(s)
- Eddo Kim
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, Israel
| | | | | |
Collapse
|
3704
|
Construction and characterization of a normalized yeast two-hybrid library derived from a human protein-coding clone collection. Biotechniques 2008; 44:265-73. [PMID: 18330356 DOI: 10.2144/000112674] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The nuclear yeast two-hybrid (Y2H) system is the most widely used technology for detecting interactions between proteins. A common approach is to screen specific test proteins (baits) against large compilations of randomly cloned proteins (prey libraries). For eukaryotic organisms, libraries have traditionally been generated using messenger RNA (mRNA) extracted from various tissues and cells. Here we present a library construction strategy made possible by ongoing public efforts to establish collections of full-length protein encoding clones. Our approach generates libraries that are essentially normalized and contain both randomly fragmented as well as full-length inserts. We refer to this type of protein-coding clone-derived library as random and full-length (RAFL) Y2H library. The library described here is based on clones from the Mammalian Gene Collection, but our strategy is compatible with the use of any protein-coding clone collection from any organism in any vector and does not require inserts to be devoid of untranslated regions. We tested our prototype human RAFL library against a set of baits that had previously been searched against multiple cDNA libraries. These Y2H searches yielded a combination of novel as well as expected interactions, indicating that the RAFL library constitutes a valuable complement to Y2H cDNA libraries.
Collapse
|
3705
|
Abstract
Despite much interest in regulatory evolution, how promoters have evolved remains poorly studied, mainly owing to paucity of data on promoter regions. Using a new set of high-quality experimentally determined core promoters of the human genome, we conducted a comparative analysis of 2,492 human and rhesus macaque promoters and their neighboring nearly neutral regions. We found that the core promoters have an average rate of nucleotide substitution substantially higher than that at 4-fold degenerate sites and only slightly lower than that for the assumed neutral controls of neighboring noncoding regions, suggesting that core promoters are subject to very weak selective constraints. Interestingly, we identified 24 core promoters (at false discovery rate = 50%) that have evolved at an accelerated rate compared with the neutral controls, suggesting that they may have undergone positive selection. The inferred positively selected genes show strong bias in molecular function. We also used population genetic approaches to examine the evolution of core promoters in human populations and found evidence of positive selection at some loci. Taken together, our results suggest that positive selection has played a substantial role in the evolution of transcriptional regulation in primates.
Collapse
Affiliation(s)
- Han Liang
- Department of Ecology and Evolution, University of Chicago, USA
| | | | | |
Collapse
|
3706
|
Abstract
For several decades, only a limited number of noncoding RNAs, such as ribosomal and transfer RNA, have been studied in any depth. In recent years, additional species of noncoding RNAs have increasingly been discovered. Of these, small RNA species attract particular interest because of their essential roles in processes such as RNA silencing and modifications. Detailed analyses revealed several pathways associated with the function of small RNAs. Although these pathways show evolutional conservation, there are substantial differences. Advanced technologies to profile RNAs have accelerated the field further resulting in the discovery of an increasing number of novel species, suggesting that we are only just beginning to appreciate the complexity of small RNAs and their functions. Here, we review recent progress in novel small RNA exploration, including discovered small RNA species, their pathways, and devised technologies.
Collapse
|
3707
|
Ramsey SA, Klemm SL, Zak DE, Kennedy KA, Thorsson V, Li B, Gilchrist M, Gold ES, Johnson CD, Litvak V, Navarro G, Roach JC, Rosenberger CM, Rust AG, Yudkovsky N, Aderem A, Shmulevich I. Uncovering a macrophage transcriptional program by integrating evidence from motif scanning and expression dynamics. PLoS Comput Biol 2008; 4:e1000021. [PMID: 18369420 PMCID: PMC2265556 DOI: 10.1371/journal.pcbi.1000021] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Accepted: 02/04/2008] [Indexed: 01/04/2023] Open
Abstract
Macrophages are versatile immune cells that can detect a variety of pathogen-associated molecular patterns through their Toll-like receptors (TLRs). In response to microbial challenge, the TLR-stimulated macrophage undergoes an activation program controlled by a dynamically inducible transcriptional regulatory network. Mapping a complex mammalian transcriptional network poses significant challenges and requires the integration of multiple experimental data types. In this work, we inferred a transcriptional network underlying TLR-stimulated murine macrophage activation. Microarray-based expression profiling and transcription factor binding site motif scanning were used to infer a network of associations between transcription factor genes and clusters of co-expressed target genes. The time-lagged correlation was used to analyze temporal expression data in order to identify potential causal influences in the network. A novel statistical test was developed to assess the significance of the time-lagged correlation. Several associations in the resulting inferred network were validated using targeted ChIP-on-chip experiments. The network incorporates known regulators and gives insight into the transcriptional control of macrophage activation. Our analysis identified a novel regulator (TGIF1) that may have a role in macrophage activation. Macrophages play a vital role in host defense against infection by recognizing pathogens through pattern recognition receptors, such as the Toll-like receptors (TLRs), and mounting an immune response. Stimulation of TLRs initiates a complex transcriptional program in which induced transcription factor genes dynamically regulate downstream genes. Microarray-based transcriptional profiling has proved useful for mapping such transcriptional programs in simpler model organisms; however, mammalian systems present difficulties such as post-translational regulation of transcription factors, combinatorial gene regulation, and a paucity of available gene-knockout expression data. Additional evidence sources, such as DNA sequence-based identification of transcription factor binding sites, are needed. In this work, we computationally inferred a transcriptional network for TLR-stimulated murine macrophages. Our approach combined sequence scanning with time-course expression data in a probabilistic framework. Expression data were analyzed using the time-lagged correlation. A novel, unbiased method was developed to assess the significance of the time-lagged correlation. The inferred network of associations between transcription factor genes and co-expressed gene clusters was validated with targeted ChIP-on-chip experiments, and yielded insights into the macrophage activation program, including a potential novel regulator. Our general approach could be used to analyze other complex mammalian systems for which time-course expression data are available.
Collapse
Affiliation(s)
- Stephen A. Ramsey
- Institute for Systems Biology, Seattle, Washington, United States of America
- * E-mail: (SR); (AA); (IS)
| | - Sandy L. Klemm
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Daniel E. Zak
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Kathleen A. Kennedy
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Vesteinn Thorsson
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Bin Li
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Mark Gilchrist
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Elizabeth S. Gold
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Carrie D. Johnson
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Vladimir Litvak
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Garnet Navarro
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Jared C. Roach
- Institute for Systems Biology, Seattle, Washington, United States of America
| | | | - Alistair G. Rust
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Natalya Yudkovsky
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Alan Aderem
- Institute for Systems Biology, Seattle, Washington, United States of America
- * E-mail: (SR); (AA); (IS)
| | - Ilya Shmulevich
- Institute for Systems Biology, Seattle, Washington, United States of America
- * E-mail: (SR); (AA); (IS)
| |
Collapse
|
3708
|
Thorsen K, Sørensen KD, Brems-Eskildsen AS, Modin C, Gaustadnes M, Hein AMK, Kruhøffer M, Laurberg S, Borre M, Wang K, Brunak S, Krainer AR, Tørring N, Dyrskjøt L, Andersen CL, Orntoft TF. Alternative splicing in colon, bladder, and prostate cancer identified by exon array analysis. Mol Cell Proteomics 2008; 7:1214-24. [PMID: 18353764 DOI: 10.1074/mcp.m700590-mcp200] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Alternative splicing enhances proteome diversity and modulates cancer-associated proteins. To identify tissue- and tumor-specific alternative splicing, we used the GeneChip Human Exon 1.0 ST Array to measure whole-genome exon expression in 102 normal and cancer tissue samples of different stages from colon, urinary bladder, and prostate. We identified 2069 candidate alternative splicing events between normal tissue samples from colon, bladder, and prostate and selected 15 splicing events for RT-PCR validation, 10 of which were successfully validated by RT-PCR and sequencing. Furthermore 23, 19, and 18 candidate tumor-specific splicing alterations in colon, bladder, and prostate, respectively, were selected for RT-PCR validation on an independent set of 81 normal and tumor tissue samples. In total, seven genes with tumor-specific splice variants were identified (ACTN1, CALD1, COL6A3, LRRFIP2, PIK4CB, TPM1, and VCL). The validated tumor-specific splicing alterations were highly consistent, enabling clear separation of normal and cancer samples and in some cases even of different tumor stages. A subset of the tumor-specific splicing alterations (ACTN1, CALD1, and VCL) was found in all three organs and may represent general cancer-related splicing events. In silico protein predictions suggest that the identified cancer-specific splice variants encode proteins with potentially altered functions, indicating that they may be involved in pathogenesis and hence represent novel therapeutic targets. In conclusion, we identified and validated alternative splicing between normal tissue samples from colon, bladder, and prostate in addition to cancer-specific splicing events in colon, bladder, and prostate cancer that may have diagnostic and prognostic implications.
Collapse
Affiliation(s)
- Kasper Thorsen
- Molecular Diagnostic Laboratory, Department of Clinical Biochemistry, Aarhus University Hospital, Skejby, DK-8200 Aarhus N, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3709
|
Replogle K, Arnold AP, Ball GF, Band M, Bensch S, Brenowitz EA, Dong S, Drnevich J, Ferris M, George JM, Gong G, Hasselquist D, Hernandez AG, Kim R, Lewin HA, Liu L, Lovell PV, Mello CV, Naurin S, Rodriguez-Zas S, Thimmapuram J, Wade J, Clayton DF. The Songbird Neurogenomics (SoNG) Initiative: community-based tools and strategies for study of brain gene function and evolution. BMC Genomics 2008; 9:131. [PMID: 18366674 PMCID: PMC2329646 DOI: 10.1186/1471-2164-9-131] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 03/18/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Songbirds hold great promise for biomedical, environmental and evolutionary research. A complete draft sequence of the zebra finch genome is imminent, yet a need remains for application of genomic resources within a research community traditionally focused on ethology and neurobiological methods. In response, we developed a core set of genomic tools and a novel collaborative strategy to probe gene expression in diverse songbird species and natural contexts. RESULTS We end-sequenced cDNAs from zebra finch brain and incorporated additional sequences from community sources into a database of 86,784 high quality reads. These assembled into 31,658 non-redundant contigs and singletons, which we annotated via BLAST search of chicken and human databases. The results are publicly available in the ESTIMA:Songbird database. We produced a spotted cDNA microarray with 20,160 addresses representing 17,214 non-redundant products of an estimated 11,500-15,000 genes, validating it by analysis of immediate-early gene (zenk) gene activation following song exposure and by demonstrating effective cross hybridization to genomic DNAs of other songbird species in the Passerida Parvorder. Our assembly was also used in the design of the "Lund-zfa" Affymetrix array representing approximately 22,000 non-redundant sequences. When the two arrays were hybridized to cDNAs from the same set of male and female zebra finch brain samples, both arrays detected a common set of regulated transcripts with a Pearson correlation coefficient of 0.895. To stimulate use of these resources by the songbird research community and to maintain consistent technical standards, we devised a "Community Collaboration" mechanism whereby individual birdsong researchers develop experiments and provide tissues, but a single individual in the community is responsible for all RNA extractions, labelling and microarray hybridizations. CONCLUSION Immediately, these results set the foundation for a coordinated set of 25 planned experiments by 16 research groups probing fundamental links between genome, brain, evolution and behavior in songbirds. Energetic application of genomic resources to research using songbirds should help illuminate how complex neural and behavioral traits emerge and evolve.
Collapse
Affiliation(s)
- Kirstin Replogle
- Cell & Developmental Biology, Univ. of Illinois, Urbana, IL, USA
- Institute for Genomic Biology, Univ. of Illinois, Urbana, IL, USA
| | | | - Gregory F Ball
- Psychological & Brain Sci., Johns Hopkins Univ., Baltimore, MD, USA
| | - Mark Band
- W.M. Keck Center for Comparative & Functional Genomics, Univ. of Illinois, Urbana, IL, USA
| | | | - Eliot A Brenowitz
- Psychology, Biology, and Bloedel Hearing Research Center, Univ. of Washington, Seattle, WA, USA
| | - Shu Dong
- Cell & Developmental Biology, Univ. of Illinois, Urbana, IL, USA
| | - Jenny Drnevich
- W.M. Keck Center for Comparative & Functional Genomics, Univ. of Illinois, Urbana, IL, USA
| | | | - Julia M George
- Mol. & Integrative Physiology, Univ. of Illinois, Urbana, IL, USA
| | - George Gong
- W.M. Keck Center for Comparative & Functional Genomics, Univ. of Illinois, Urbana, IL, USA
| | | | - Alvaro G Hernandez
- W.M. Keck Center for Comparative & Functional Genomics, Univ. of Illinois, Urbana, IL, USA
| | - Ryan Kim
- W.M. Keck Center for Comparative & Functional Genomics, Univ. of Illinois, Urbana, IL, USA
| | - Harris A Lewin
- Institute for Genomic Biology, Univ. of Illinois, Urbana, IL, USA
- Animal Sciences, Univ. of Illinois, Urbana, IL, USA
| | - Lei Liu
- W.M. Keck Center for Comparative & Functional Genomics, Univ. of Illinois, Urbana, IL, USA
| | - Peter V Lovell
- Neurological Sci. Inst., Oregon Hlth. Sci. Univ., Beaverton, OR, USA
| | - Claudio V Mello
- Neurological Sci. Inst., Oregon Hlth. Sci. Univ., Beaverton, OR, USA
| | - Sara Naurin
- Animal Ecology, Lund University, S-223 62 Lund, Sweden
| | | | - Jyothi Thimmapuram
- W.M. Keck Center for Comparative & Functional Genomics, Univ. of Illinois, Urbana, IL, USA
| | - Juli Wade
- Psychology, Zoology & Neuroscience, Michigan State Univ., East Lansing, MI, USA
| | - David F Clayton
- Cell & Developmental Biology, Univ. of Illinois, Urbana, IL, USA
- Institute for Genomic Biology, Univ. of Illinois, Urbana, IL, USA
- Neuroscience Program, Univ. of Illinois, Urbana, IL, USA
| |
Collapse
|
3710
|
Faulkner GJ, Forrest ARR, Chalk AM, Schroder K, Hayashizaki Y, Carninci P, Hume DA, Grimmond SM. A rescue strategy for multimapping short sequence tags refines surveys of transcriptional activity by CAGE. Genomics 2008; 91:281-8. [PMID: 18178374 DOI: 10.1016/j.ygeno.2007.11.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 11/07/2007] [Accepted: 11/07/2007] [Indexed: 10/22/2022]
Abstract
Cap analysis gene expression (CAGE) is a high-throughput, tag-based method designed to survey the 5' end of capped full-length cDNAs. CAGE has previously been used to define global transcription start site usage and monitor gene activity in mammals. A drawback of the CAGE approach thus far has been the removal of as many as 40% of CAGE sequence tags due to their mapping to multiple genomic locations. Here, we address the origins of multimap tags and present a novel strategy to assign CAGE tags to their most likely source promoter region. When this approach was applied to the FANTOM3 CAGE libraries, the percentage of protein-coding mouse transcriptional frameworks detected by CAGE improved from 42.9 to 57.8% (an increase of 5516 frameworks) with no reduction in CAGE to microarray correlation. These results suggest that the multimap tags produced by high-throughput, short sequence tag-based approaches can be rescued to augment greatly the transcriptome coverage provided by single-map tags alone.
Collapse
Affiliation(s)
- Geoffrey J Faulkner
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
3711
|
Ravasi T, Wells CA, Hume DA. Systems biology of transcription control in macrophages. Bioessays 2008; 29:1215-26. [PMID: 18008376 DOI: 10.1002/bies.20683] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The study of the mammalian immune system offers many advantages to systems biologists. The cellular components of the mammalian immune system are experimentally tractable; they can be isolated or differentiated from in vivo and ex vivo sources and have an essential role in health and disease. For these reasons, the major effectors cells of the innate immune system, macrophages, have been a particular focus in international genome and transcriptome consortia. Genome-scale analysis of the transcriptome, and transcription initiation has enabled the construction of predictive models of transcription control in macrophages that identify the points of control (the major nodes of networks) and the ways in which they interact.
Collapse
Affiliation(s)
- Timothy Ravasi
- Scripps NeuroAIDS Preclinical Studies Centre and Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
3712
|
Davuluri RV, Suzuki Y, Sugano S, Plass C, Huang THM. The functional consequences of alternative promoter use in mammalian genomes. Trends Genet 2008; 24:167-77. [PMID: 18329129 DOI: 10.1016/j.tig.2008.01.008] [Citation(s) in RCA: 284] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Revised: 01/17/2008] [Accepted: 01/18/2008] [Indexed: 12/19/2022]
Abstract
We are beginning to appreciate the increasing complexity of mammalian gene structure. A phenomenon that adds an important dimension to this complexity is the use of alternative gene promoters that drive widespread cell type, tissue type or developmental gene regulation. Recent annotations of the human genome suggest that almost one half of the protein-coding genes contain alternative promoters, including those of many disease-associated genes. Aberrant use of one promoter over another has been found to be associated with various diseases, including cancer. Here we discuss the functional consequences of use and misuse of alternative promoters in normal and disease genomes and review the molecular mechanisms regulating alternative promoter use in mammalian genomes.
Collapse
Affiliation(s)
- Ramana V Davuluri
- Human Cancer Genetics Program, Comprehensive Cancer Center, Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | |
Collapse
|
3713
|
Abstract
In order to describe a cell at molecular level, a notion of a “gene” is neither necessary nor helpful. It is sufficient to consider the molecules (i.e., chromosomes, transcripts, proteins) and their interactions to describe cellular processes. The downside of the resulting high resolution is that it becomes very tedious to address features on the organismal and phenotypic levels with a language based on molecular terms. Looking for the missing link between biological disciplines dealing with different levels of biological organization, we suggest to return to the original intent behind the term “gene”. To this end, we propose to investigate whether a useful notion of “gene” can be constructed based on an underlying notion of function, and whether this can serve as the necessary link and embed the various distinct gene concepts of biological (sub)disciplines in a coherent theoretical framework. In reply to the Genon Theory recently put forward by Klaus Scherrer and Jürgen Jost in this journal, we shall discuss a general approach to assess a gene definition that should then be tested for its expressiveness and potential cross-disciplinary relevance.
Collapse
Affiliation(s)
- Sonja J Prohaska
- Santa Fe Institute, 1399 Hyde Park Rd., Santa Fe, NM, 87501, USA.
| | | |
Collapse
|
3714
|
Mellor J, Dudek P, Clynes D. A glimpse into the epigenetic landscape of gene regulation. Curr Opin Genet Dev 2008; 18:116-22. [PMID: 18295475 DOI: 10.1016/j.gde.2007.12.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Accepted: 12/18/2007] [Indexed: 01/19/2023]
Abstract
Post-translational modifications to histone proteins and methylation of DNA comprise the epigenome of a cell. The epigenome, which changes through development, controls access to our genes. Recent advances in DNA sequencing technology has led to genome-wide distribution data for a limited number of histone modifications in mammalian stem cells and some differentiated lineages. These studies reveal predictive correlations between histone modifications, different classes of gene and chromosomal features. Moreover, this glimpse into our epigenome challenges current ideas about regulation of gene expression. Many genes in stem cells are poised for expression with initiated RNA polymerase II at the promoter. This state is maintained by an epigenetic mark through multiple lineages until the gene is expressed.
Collapse
Affiliation(s)
- Jane Mellor
- Division of Molecular Genetics, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| | | | | |
Collapse
|
3715
|
Talmud PJ, Cooper JA, Palmen J, Lovering R, Drenos F, Hingorani AD, Humphries SE. Chromosome 9p21.3 coronary heart disease locus genotype and prospective risk of CHD in healthy middle-aged men. Clin Chem 2008; 54:467-74. [PMID: 18250146 DOI: 10.1373/clinchem.2007.095489] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND We investigated whether chromosome 9p21.3 single-nucleotide polymorphisms (SNPs), identified in coronary heart disease (CHD) genome-wide association scans, added significantly to the predictive utility for CHD of conventional risk factors (CRF) in the Framingham risk score (FRS) algorithm. METHODS In the Northwick Park Heart Study II of 2742 men (270 CHD events occurring during a 15-year prospective study), rs10757274 A>G [mean frequency G = 0.48 (95% CI 0.47-0.50)] was genotyped. Using the area under the ROC curve (A(ROC)) and the likelihood ratio (LR) statistic, we assessed the discriminatory performance of the FRS based on CRFs with and without genotype. RESULTS rs10757274 A>G was associated with incident CHD, with an effect size as reported previously [hazard ratio in GG vs AA men of 1.60 (95% CI 1.12-2.28)], independent of CRFs and family history of CHD. Although the A(ROC) for CRFs alone [0.62 (95% CI 0.58-0.66)] did not increase significantly (P = 0.14) when rs10757274 A>G genotype was added [0.64 (95% CI 0.60-0.68)], including genotype gave better fit (LR P = 0.01) and including rs10757274 moved 369 men (13.5% of the total) into more accurate risk categories. To model polygenic effects, 10 hypothetical, randomly assigned gene variants, with similar effect size and frequencies were added. Two variants made significant A(ROC) improvements to the FRS prediction (P = 0.01), whereas further variants had smaller incremental effects (final A(ROC) = 0.71, P <0.001 vs CRFs; LR vs CRFs P <0.0001). CONCLUSIONS Although overall, rs10757274 did not add substantially to the usefulness of the FRS for predicting future events, it did improve reclassification of CHD risk, and thus may have clinical utility.
Collapse
Affiliation(s)
- Philippa J Talmud
- Centre for Cardiovascular Genetics, Department of Medicine, University College London, London, UK
| | | | | | | | | | | | | |
Collapse
|
3716
|
Rada-Iglesias A, Ameur A, Kapranov P, Enroth S, Komorowski J, Gingeras TR, Wadelius C. Whole-genome maps of USF1 and USF2 binding and histone H3 acetylation reveal new aspects of promoter structure and candidate genes for common human disorders. Genome Res 2008; 18:380-92. [PMID: 18230803 PMCID: PMC2259102 DOI: 10.1101/gr.6880908] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Accepted: 12/11/2007] [Indexed: 12/15/2022]
Abstract
Transcription factors and histone modifications are crucial regulators of gene expression that mutually influence each other. We present the DNA binding profiles of upstream stimulatory factors 1 and 2 (USF1, USF2) and acetylated histone H3 (H3ac) in a liver cell line for the whole human genome using ChIP-chip at a resolution of 35 base pairs. We determined that these three proteins bind mostly in proximity of protein coding genes transcription start sites (TSSs), and their bindings are positively correlated with gene expression levels. Based on the spatial and functional relationship between USFs and H3ac at protein coding gene promoters, we found similar promoter architecture for known genes and the novel and less-characterized transcripts human mRNAs and spliced ESTs. Furthermore, our analysis revealed a previously underestimated abundance of genes in a bidirectional conformation, where USFs are bound in between TSSs. After taking into account this promoter conformation, the results indicate that H3ac is mainly located downstream of TSS, and it is at this genomic location where it positively correlates with gene expression. Finally, USF1, which is associated to familial combined hyperlipidemia, was found to bind and potentially regulate nuclear mitochondrial genes as well as genes for lipid and cholesterol metabolism, frequently in collaboration with GA binding protein transcription factor alpha (GABPA, nuclear respiratory factor 2 [NRF-2]). This expands our understanding about the transcriptional control of metabolic processes and its alteration in metabolic disorders.
Collapse
Affiliation(s)
- Alvaro Rada-Iglesias
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-75185 Uppsala, Sweden
| | - Adam Ameur
- Linnaeus Centre for Bioinformatics, Uppsala University, SE-75185 Uppsala, Sweden
| | | | - Stefan Enroth
- Linnaeus Centre for Bioinformatics, Uppsala University, SE-75185 Uppsala, Sweden
| | - Jan Komorowski
- Linnaeus Centre for Bioinformatics, Uppsala University, SE-75185 Uppsala, Sweden
- Interdisciplinary Centre for Mathematical and Computational Modelling, Warsaw University, 02-106 Warsaw, Poland
| | | | - Claes Wadelius
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-75185 Uppsala, Sweden
| |
Collapse
|
3717
|
Johnson DS, Li W, Gordon DB, Bhattacharjee A, Curry B, Ghosh J, Brizuela L, Carroll JS, Brown M, Flicek P, Koch CM, Dunham I, Bieda M, Xu X, Farnham PJ, Kapranov P, Nix DA, Gingeras TR, Zhang X, Holster H, Jiang N, Green RD, Song JS, McCuine SA, Anton E, Nguyen L, Trinklein ND, Ye Z, Ching K, Hawkins D, Ren B, Scacheri PC, Rozowsky J, Karpikov A, Euskirchen G, Weissman S, Gerstein M, Snyder M, Yang A, Moqtaderi Z, Hirsch H, Shulha HP, Fu Y, Weng Z, Struhl K, Myers RM, Lieb JD, Liu XS. Systematic evaluation of variability in ChIP-chip experiments using predefined DNA targets. Genome Res 2008; 18:393-403. [PMID: 18258921 PMCID: PMC2259103 DOI: 10.1101/gr.7080508] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 12/12/2007] [Indexed: 02/07/2023]
Abstract
The most widely used method for detecting genome-wide protein-DNA interactions is chromatin immunoprecipitation on tiling microarrays, commonly known as ChIP-chip. Here, we conducted the first objective analysis of tiling array platforms, amplification procedures, and signal detection algorithms in a simulated ChIP-chip experiment. Mixtures of human genomic DNA and "spike-ins" comprised of nearly 100 human sequences at various concentrations were hybridized to four tiling array platforms by eight independent groups. Blind to the number of spike-ins, their locations, and the range of concentrations, each group made predictions of the spike-in locations. We found that microarray platform choice is not the primary determinant of overall performance. In fact, variation in performance between labs, protocols, and algorithms within the same array platform was greater than the variation in performance between array platforms. However, each array platform had unique performance characteristics that varied with tiling resolution and the number of replicates, which have implications for cost versus detection power. Long oligonucleotide arrays were slightly more sensitive at detecting very low enrichment. On all platforms, simple sequence repeats and genome redundancy tended to result in false positives. LM-PCR and WGA, the most popular sample amplification techniques, reproduced relative enrichment levels with high fidelity. Performance among signal detection algorithms was heavily dependent on array platform. The spike-in DNA samples and the data presented here provide a stable benchmark against which future ChIP platforms, protocol improvements, and analysis methods can be evaluated.
Collapse
Affiliation(s)
- David S. Johnson
- Department of Genetics, Stanford University Medical Center, Stanford, California 94305, USA
| | - Wei Li
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | | | | - Bo Curry
- Agilent Technologies, Inc., Santa Clara, California 95051, USA
| | - Jayati Ghosh
- Agilent Technologies, Inc., Santa Clara, California 95051, USA
| | | | - Jason S. Carroll
- Cancer Research UK, Cambridge Research Institute, Cambridge, CB2 0RE, United Kingdom
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Paul Flicek
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - Christoph M. Koch
- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - Ian Dunham
- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - Mark Bieda
- Department of Pharmacology and the Genome Center, University of California–Davis, Davis, California 95616, USA
| | - Xiaoqin Xu
- Department of Pharmacology and the Genome Center, University of California–Davis, Davis, California 95616, USA
| | - Peggy J. Farnham
- Department of Pharmacology and the Genome Center, University of California–Davis, Davis, California 95616, USA
| | | | - David A. Nix
- HCI Bio Informatics, Huntsman Cancer Institute, Salt Lake City, Utah 84112, USA
| | | | - Xinmin Zhang
- Roche NimbleGen, Inc., Madison, Wisconsin 53719, USA
| | | | - Nan Jiang
- Roche NimbleGen, Inc., Madison, Wisconsin 53719, USA
| | | | - Jun S. Song
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | | - Elizabeth Anton
- Department of Genetics, Stanford University Medical Center, Stanford, California 94305, USA
| | - Loan Nguyen
- Department of Genetics, Stanford University Medical Center, Stanford, California 94305, USA
| | | | - Zhen Ye
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, California 92093-0653, USA
| | - Keith Ching
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, California 92093-0653, USA
| | - David Hawkins
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, California 92093-0653, USA
| | - Bing Ren
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, California 92093-0653, USA
| | - Peter C. Scacheri
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Joel Rozowsky
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
| | - Alexander Karpikov
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
| | - Ghia Euskirchen
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520, USA
| | - Sherman Weissman
- Department of Genetics, Yale University, New Haven, Connecticut 06520, USA
| | - Mark Gerstein
- Department of Genetics, Yale University, New Haven, Connecticut 06520, USA
| | - Michael Snyder
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520, USA
| | - Annie Yang
- Genentech Inc., South San Francisco, California 94080-4990, USA
| | - Zarmik Moqtaderi
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115-5730, USA
| | - Heather Hirsch
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115-5730, USA
| | - Hennady P. Shulha
- Biomedical Engineering Department, Boston University, Boston, Massachusetts 02215, USA
| | - Yutao Fu
- Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| | - Zhiping Weng
- Biomedical Engineering Department, Boston University, Boston, Massachusetts 02215, USA
- Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| | - Kevin Struhl
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115-5730, USA
| | - Richard M. Myers
- Department of Genetics, Stanford University Medical Center, Stanford, California 94305, USA
| | - Jason D. Lieb
- Department of Biology and Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-3280, USA
| | - X. Shirley Liu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
3718
|
Martínez-Frías ML, Rodríguez-Pinilla E. Problem of using cases with genetic anomalies as a reference group in case-control studies on drug use and birth defects. BIRTH DEFECTS RESEARCH. PART A, CLINICAL AND MOLECULAR TERATOLOGY 2008; 82:173-175. [PMID: 18186122 DOI: 10.1002/bdra.20424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
|
3719
|
Abstract
Over two metres of DNA is packaged into each nucleus in the human body in a manner that still allows for gene regulation. This remarkable feat is accomplished by the wrapping of DNA around histone proteins in repeating units of nucleosomes to form a structure known as chromatin. This chromatin structure is subject to various modifications that have profound influences on gene expression. Recently developed techniques to study chromatin modifications at a genome-wide scale are now allowing researchers to probe the complex components that make up epigenomes. Here we review genome-wide approaches to studying epigenomic structure and the exciting findings that have been obtained using these technologies.
Collapse
Affiliation(s)
- Dustin E Schones
- Laboratory of Molecular Immunology, The National Heart, Lung and Blood Institute, National Institutes of Health, Building 10, Room 7B05, 9,000 Rockville Pike, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
3720
|
Heinäniemi M, Uski JO, Degenhardt T, Carlberg C. Meta-analysis of primary target genes of peroxisome proliferator-activated receptors. Genome Biol 2008; 8:R147. [PMID: 17650321 PMCID: PMC2323243 DOI: 10.1186/gb-2007-8-7-r147] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Revised: 07/02/2007] [Accepted: 07/25/2007] [Indexed: 12/20/2022] Open
Abstract
A combined experimental and in silico approach identifies Peroxisome Proliferator Activated Receptor (PPAR) binding sites and six novel target genes in the human genome. Background Peroxisome proliferator-activated receptors (PPARs) are known for their critical role in the development of diseases, such as obesity, cardiovascular disease, type 2 diabetes and cancer. Here, an in silico screening method is presented, which incorporates experiment- and informatics-derived evidence, such as DNA-binding data of PPAR subtypes to a panel of PPAR response elements (PPREs), PPRE location relative to the transcription start site (TSS) and PPRE conservation across multiple species, for more reliable prediction of PPREs. Results In vitro binding and in vivo functionality evidence agrees with in silico predictions, validating the approach. The experimental analysis of 30 putative PPREs in eight validated PPAR target genes indicates that each gene contains at least one functional, strong PPRE that occurs without positional bias relative to the TSS. An extended analysis of the cross-species conservation of PPREs reveals limited conservation of PPRE patterns, although PPAR target genes typically contain strong or multiple medium strength PPREs. Human chromosome 19 was screened using this method, with validation of six novel PPAR target genes. Conclusion An in silico screening approach is presented, which allows increased sensitivity of PPAR binding site and target gene detection.
Collapse
Affiliation(s)
- Merja Heinäniemi
- Department of Biochemistry, University of Kuopio, FIN-70211 Kuopio, Finland
- Life Sciences Research Unit, University of Luxembourg, L-1511 Luxembourg
| | - J Oskari Uski
- Department of Biochemistry, University of Kuopio, FIN-70211 Kuopio, Finland
| | - Tatjana Degenhardt
- Department of Biochemistry, University of Kuopio, FIN-70211 Kuopio, Finland
| | - Carsten Carlberg
- Department of Biochemistry, University of Kuopio, FIN-70211 Kuopio, Finland
- Life Sciences Research Unit, University of Luxembourg, L-1511 Luxembourg
| |
Collapse
|
3721
|
Vijg J. The role of DNA damage and repair in aging: new approaches to an old problem. Mech Ageing Dev 2008; 129:498-502. [PMID: 18394680 DOI: 10.1016/j.mad.2008.02.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 02/22/2008] [Accepted: 02/22/2008] [Indexed: 11/30/2022]
Abstract
DNA damage and mutations have been implicated as key causal events in the biological process of aging. In this context, it has been hypothesized that the complex of genome maintenance systems acts as a longevity assurance system by signaling and repairing damage or removing cells that are beyond repair. In the past, various approaches have been taken to clarify the importance of preserving genome integrity for healthy aging. Here I will briefly review these approaches in the context of the progress made in improving our understanding of the interrelationship between DNA damage, genome maintenance and mutations.
Collapse
Affiliation(s)
- Jan Vijg
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA.
| |
Collapse
|
3722
|
Seng KC, Seng CK. The success of the genome-wide association approach: a brief story of a long struggle. Eur J Hum Genet 2008; 16:554-64. [PMID: 18285837 DOI: 10.1038/ejhg.2008.12] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The genome-wide association approach has been the most powerful and efficient study design thus far in identifying genetic variants that are associated with complex human diseases. This approach became feasible as the result of several key advancements in genetic knowledge, genotyping technologies, statistical analysis algorithms and the availability of large collections of cases and controls. With all these necessary tools in hand, many genome-wide association studies were recently completed, and many more studies which will explore the genetic basis of various complex diseases and quantitative traits are soon to come. This approach has started to reap the fruits of its labor over the past several months. Publications of genome-wide association studies in several complex diseases such as inflammatory bowel disease, type-2 diabetes, breast cancer and prostate cancer have been abundant in the first half of this year. The aims of this review are firstly, to provide a timely summary for most of the genome-wide association studies that have been published until June/July 2007 and secondly, to evaluate to what extent these results have been validated in subsequent replication studies.
Collapse
Affiliation(s)
- Ku Chee Seng
- Center for Molecular Epidemiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | | |
Collapse
|
3723
|
Kininis M, Kraus WL. A global view of transcriptional regulation by nuclear receptors: gene expression, factor localization, and DNA sequence analysis. NUCLEAR RECEPTOR SIGNALING 2008; 6:e005. [PMID: 18301785 PMCID: PMC2254333 DOI: 10.1621/nrs.06005] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 02/07/2008] [Indexed: 01/22/2023]
Abstract
Recent genomic analyses of transcription factor binding, histone modification, and gene expression have provided a global view of transcriptional regulation by nuclear receptors (NRs) that complements an existing large body of literature on gene-specific studies. The picture emerging from these genomic studies indicates that NRs bind at promoter-proximal and promoter-distal enhancers in conjunction with other transcription factors (e.g., activator protein-1, Sp1 and FOXA1). This binding promotes the recruitment of coregulators that mediate the posttranslational modification of histones at promoters and enhancers. Ultimately, signaling through liganded NRs stimulates changes in the occupancy of RNA polymerase II (Pol II) or the activation of preloaded Pol II at target promoters. Chromosomal looping and/or Pol II tracking may underlie promoter-enhancer communication. Interestingly, the direct target genes of NR signaling represent a limited subset of all the genes regulated by NR ligands, with the rest being regulated through secondary effects. As suggested by previous gene-specific analyses, NR-mediated outcomes are highly cell type- and promoter-specific, highlighting the complexity of transcriptional regulation by NRs and the value of genomic analyses for identifying commonly shared patterns. Overall, NRs share common themes in their patterns of localization and transcriptional regulation across mammalian genomes. In this review, we provide an overview of recent advances in the understanding of NR-mediated transcription garnered from genomic analyses of gene expression, factor localization, and target DNA sequences.
Collapse
Affiliation(s)
- Miltiadis Kininis
- Department of Molecular Biology and Genetics, Graduate Field of Genetics and Development, Cornell University, Ithaca, New York, USA
| | | |
Collapse
|
3724
|
Abstract
The Latin word "facultas" literally means "opportunity." Facultative heterochromatin (fHC) then designates genomic regions in the nucleus of a eukaryotic cell that have the opportunity to adopt open or compact conformations within temporal and spatial contexts. This review focuses on the molecular and functional aspects of fHC that distinguish it from constitutive heterochromatin (cHC) and euchromatin (EC) and discusses various concepts regarding the regulation of fHC structure. We begin by revisiting the historical developments that gave rise to our current appreciation of fHC.
Collapse
Affiliation(s)
- Patrick Trojer
- Howard Hughes Medical Institute, New York University Medical School, 522 First Avenue, New York, NY 10016, USA
| | | |
Collapse
|
3725
|
Parelho V, Hadjur S, Spivakov M, Leleu M, Sauer S, Gregson HC, Jarmuz A, Canzonetta C, Webster Z, Nesterova T, Cobb BS, Yokomori K, Dillon N, Aragon L, Fisher AG, Merkenschlager M. Cohesins functionally associate with CTCF on mammalian chromosome arms. Cell 2008; 132:422-33. [PMID: 18237772 DOI: 10.1016/j.cell.2008.01.011] [Citation(s) in RCA: 700] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 12/04/2007] [Accepted: 01/04/2008] [Indexed: 01/26/2023]
Abstract
Cohesins mediate sister chromatid cohesion, which is essential for chromosome segregation and postreplicative DNA repair. In addition, cohesins appear to regulate gene expression and enhancer-promoter interactions. These noncanonical functions remained unexplained because knowledge of cohesin-binding sites and functional interactors in metazoans was lacking. We show that the distribution of cohesins on mammalian chromosome arms is not driven by transcriptional activity, in contrast to S. cerevisiae. Instead, mammalian cohesins occupy a subset of DNase I hypersensitive sites, many of which contain sequence motifs resembling the consensus for CTCF, a DNA-binding protein with enhancer blocking function and boundary-element activity. We find cohesins at most CTCF sites and show that CTCF is required for cohesin localization to these sites. Recruitment by CTCF suggests a rationale for noncanonical cohesin functions and, because CTCF binding is sensitive to DNA methylation, allows cohesin positioning to integrate DNA sequence and epigenetic state.
Collapse
Affiliation(s)
- Vania Parelho
- Lymphocyte Development Group, MRC Clinical Sciences Centre, Imperial College London, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3726
|
Williams RBH, Chan EKF, Cowley MJ, Little PFR. The influence of genetic variation on gene expression. Genome Res 2008; 17:1707-16. [PMID: 18063559 DOI: 10.1101/gr.6981507] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The view that changes to the control of gene expression rather than alterations to protein sequence are central to the evolution of organisms has become something of a truism in molecular biology. In reality, the direct evidence for this is limited, and only recently have we had the ability to look more globally at how genetic variation influences gene expression, focusing upon inter-individual variation in gene expression and using microarrays to test for differences in mRNA levels. Here, we review the scope of these experimental analyses, what they are designed to tell us about genetic variation, and what are their limitations from both a technical and a conceptual viewpoint. We conclude that while we are starting to understand the impact of this class of genetic variation upon steady-state mRNA levels, we are still far from identifying the potential phenotypic and evolutionary outcomes.
Collapse
Affiliation(s)
- Rohan B H Williams
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Randwick, NSW 2052, Australia
| | | | | | | |
Collapse
|
3727
|
Bradley RG, Binder EB, Epstein MP, Tang Y, Nair HP, Liu W, Gillespie CF, Berg T, Evces M, Newport DJ, Stowe ZN, Heim CM, Nemeroff CB, Schwartz A, Cubells JF, Ressler KJ. Influence of child abuse on adult depression: moderation by the corticotropin-releasing hormone receptor gene. ARCHIVES OF GENERAL PSYCHIATRY 2008; 65:190-200. [PMID: 18250257 PMCID: PMC2443704 DOI: 10.1001/archgenpsychiatry.2007.26] [Citation(s) in RCA: 447] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
CONTEXT Genetic inheritance and developmental life stress both contribute to major depressive disorder in adults. Child abuse and trauma alter the endogenous stress response, principally corticotropin-releasing hormone and its downstream effectors, suggesting that a gene x environment interaction at this locus may be important in depression. OBJECTIVE To examine whether the effects of child abuse on adult depressive symptoms are moderated by genetic polymorphisms within the corticotropin-releasing hormone type 1 receptor (CRHR1) gene. DESIGN Association study examining gene x environment interactions between genetic polymorphisms at the CRHR1 locus and measures of child abuse on adult depressive symptoms. SETTING General medical clinics of a large, public, urban hospital and Emory University, Atlanta, Georgia. PARTICIPANTS The primary participant population was 97.4% African American, of low socioeconomic status, and with high rates of lifetime trauma (n = 422). A supportive independent sample (n = 199) was distinct both ethnically (87.7% Caucasian) and socioeconomically (less impoverished). MAIN OUTCOME MEASURES Beck Depression Inventory scores and history of major depressive disorder by the Structured Clinical Interview for DSM-IV Axis I Disorders. RESULTS Fifteen single-nucleotide polymorphisms spanning 57 kilobases of the CRHR1 gene were examined. We found significant gene x environment interactions with multiple individual single-nucleotide polymorphisms (eg, rs110402, P = .008) as well as with a common haplotype spanning intron 1 (P < .001). Specific CRHR1 polymorphisms appeared to moderate the effect of child abuse on the risk for adult depressive symptoms. These protective effects were supported with similar findings in a second independent sample (n = 199). CONCLUSIONS These data support the corticotropin-releasing hormone hypothesis of depression and suggest that a gene x environment interaction is important for the expression of depressive symptoms in adults with CRHR1 risk or protective alleles who have a history of child abuse.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Alleles
- Bipolar Disorder/diagnosis
- Bipolar Disorder/genetics
- Bipolar Disorder/psychology
- Black People/genetics
- Black People/psychology
- Child
- Child Abuse/diagnosis
- Child Abuse/psychology
- Chromosomes, Human, Pair 16
- Cross-Cultural Comparison
- Depressive Disorder, Major/diagnosis
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/psychology
- Female
- Gene Frequency/genetics
- Genetic Predisposition to Disease/genetics
- Genetics, Population
- Genotype
- Haplotypes
- Humans
- Linkage Disequilibrium
- Male
- Mental Disorders/diagnosis
- Mental Disorders/genetics
- Mental Disorders/psychology
- Middle Aged
- Phenotype
- Polymorphism, Single Nucleotide/genetics
- Receptors, Corticotropin-Releasing Hormone/genetics
- Risk Factors
- Social Environment
- Socioeconomic Factors
- White People/genetics
- White People/psychology
- Black or African American
- CRF Receptor, Type 1
Collapse
|
3728
|
Li XY, MacArthur S, Bourgon R, Nix D, Pollard DA, Iyer VN, Hechmer A, Simirenko L, Stapleton M, Hendriks CLL, Chu HC, Ogawa N, Inwood W, Sementchenko V, Beaton A, Weiszmann R, Celniker SE, Knowles DW, Gingeras T, Speed TP, Eisen MB, Biggin MD. Transcription factors bind thousands of active and inactive regions in the Drosophila blastoderm. PLoS Biol 2008; 6:e27. [PMID: 18271625 PMCID: PMC2235902 DOI: 10.1371/journal.pbio.0060027] [Citation(s) in RCA: 364] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Accepted: 12/19/2007] [Indexed: 01/22/2023] Open
Abstract
Identifying the genomic regions bound by sequence-specific regulatory factors is central both to deciphering the complex DNA cis-regulatory code that controls transcription in metazoans and to determining the range of genes that shape animal morphogenesis. We used whole-genome tiling arrays to map sequences bound in Drosophila melanogaster embryos by the six maternal and gap transcription factors that initiate anterior-posterior patterning. We find that these sequence-specific DNA binding proteins bind with quantitatively different specificities to highly overlapping sets of several thousand genomic regions in blastoderm embryos. Specific high- and moderate-affinity in vitro recognition sequences for each factor are enriched in bound regions. This enrichment, however, is not sufficient to explain the pattern of binding in vivo and varies in a context-dependent manner, demonstrating that higher-order rules must govern targeting of transcription factors. The more highly bound regions include all of the over 40 well-characterized enhancers known to respond to these factors as well as several hundred putative new cis-regulatory modules clustered near developmental regulators and other genes with patterned expression at this stage of embryogenesis. The new targets include most of the microRNAs (miRNAs) transcribed in the blastoderm, as well as all major zygotically transcribed dorsal-ventral patterning genes, whose expression we show to be quantitatively modulated by anterior-posterior factors. In addition to these highly bound regions, there are several thousand regions that are reproducibly bound at lower levels. However, these poorly bound regions are, collectively, far more distant from genes transcribed in the blastoderm than highly bound regions; are preferentially found in protein-coding sequences; and are less conserved than highly bound regions. Together these observations suggest that many of these poorly bound regions are not involved in early-embryonic transcriptional regulation, and a significant proportion may be nonfunctional. Surprisingly, for five of the six factors, their recognition sites are not unambiguously more constrained evolutionarily than the immediate flanking DNA, even in more highly bound and presumably functional regions, indicating that comparative DNA sequence analysis is limited in its ability to identify functional transcription factor targets.
Collapse
Affiliation(s)
- Xiao-yong Li
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Stewart MacArthur
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Richard Bourgon
- Statistics Department, University of California Berkeley, Berkeley, California, United States of America
| | - David Nix
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Daniel A Pollard
- Biophysics Graduate Group, University of California Berkeley, Berkeley, California, United States of America
| | - Venky N Iyer
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, United States of America
| | - Aaron Hechmer
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Lisa Simirenko
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Mark Stapleton
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Cris L. Luengo Hendriks
- Biophysics Graduate Group, University of California Berkeley, Berkeley, California, United States of America
| | - Hou Cheng Chu
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Nobuo Ogawa
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - William Inwood
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Victor Sementchenko
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Amy Beaton
- Biophysics Graduate Group, University of California Berkeley, Berkeley, California, United States of America
| | - Richard Weiszmann
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Susan E Celniker
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - David W Knowles
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Tom Gingeras
- Affymetrix, Inc., Santa Clara, California, United States of America
| | - Terence P Speed
- Statistics Department, University of California Berkeley, Berkeley, California, United States of America
| | - Michael B Eisen
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, United States of America
- Center for Integrative Genomics, University of California Berkeley, Berkeley, California, United States of America
- California Institute for Quantitative Biosciences, Berkeley, California, United States of America
| | - Mark D Biggin
- Genomics Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| |
Collapse
|
3729
|
Zichi D, Eaton B, Singer B, Gold L. Proteomics and diagnostics: Let's Get Specific, again. Curr Opin Chem Biol 2008; 12:78-85. [DOI: 10.1016/j.cbpa.2008.01.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Accepted: 01/15/2008] [Indexed: 12/21/2022]
|
3730
|
Boyle AP, Davis S, Shulha HP, Meltzer P, Margulies EH, Weng Z, Furey TS, Crawford GE. High-resolution mapping and characterization of open chromatin across the genome. Cell 2008; 132:311-22. [PMID: 18243105 PMCID: PMC2669738 DOI: 10.1016/j.cell.2007.12.014] [Citation(s) in RCA: 1053] [Impact Index Per Article: 61.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 10/22/2007] [Accepted: 12/04/2007] [Indexed: 12/28/2022]
Abstract
Mapping DNase I hypersensitive (HS) sites is an accurate method of identifying the location of genetic regulatory elements, including promoters, enhancers, silencers, insulators, and locus control regions. We employed high-throughput sequencing and whole-genome tiled array strategies to identify DNase I HS sites within human primary CD4+ T cells. Combining these two technologies, we have created a comprehensive and accurate genome-wide open chromatin map. Surprisingly, only 16%-21% of the identified 94,925 DNase I HS sites are found in promoters or first exons of known genes, but nearly half of the most open sites are in these regions. In conjunction with expression, motif, and chromatin immunoprecipitation data, we find evidence of cell-type-specific characteristics, including the ability to identify transcription start sites and locations of different chromatin marks utilized in these cells. In addition, and unexpectedly, our analyses have uncovered detailed features of nucleosome structure.
Collapse
Affiliation(s)
- Alan P. Boyle
- Institute for Genome Sciences & Policy, Duke University, Durham, NC 27708, USA
| | - Sean Davis
- Center for Cancer Research, National Cancer Institute
| | - Hennady P. Shulha
- Biomedical Engineering Department, Boston University, Boston, MA 02215, USA
| | - Paul Meltzer
- Center for Cancer Research, National Cancer Institute
| | - Elliott H. Margulies
- National Human Genome Research Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhiping Weng
- Biomedical Engineering Department, Boston University, Boston, MA 02215, USA
| | - Terrence S. Furey
- Institute for Genome Sciences & Policy, Duke University, Durham, NC 27708, USA
| | - Gregory E. Crawford
- Institute for Genome Sciences & Policy, Duke University, Durham, NC 27708, USA
| |
Collapse
|
3731
|
Genome-wide B1 retrotransposon binds the transcription factors dioxin receptor and Slug and regulates gene expression in vivo. Proc Natl Acad Sci U S A 2008; 105:1632-7. [PMID: 18223155 DOI: 10.1073/pnas.0708366105] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Alterations in tissue-specific gene expression greatly affect cell function. Transcription factors (TFs) interact with cis-acting binding sites in noncoding enhancer promoter regions. Transposable elements (TEs) are abundant and similarly represented among mammalian genomes. TEs are important in gene regulation, but their function is not well understood. We have characterized a TE containing functional TF-binding sites for the carcinogen-activated dioxin receptor xenobiotic responsive element (XRE) and the epithelial-mesenchymal transition regulator Slug (Slug site). A Mus promoter database was scanned for XREs to predict coregulation with other TFs. We identified an overrepresented (1,398 genes) B1 retrotransposon containing XRE and Slug sites within 35 bp of each other (designated as B1-X35S). This B1-X35S retrotransposon differed from classic B1s by the presence of the Slug site and by its differential nucleotide conservation outside the X35S region. Phylogenetically, B1-X35S appeared recently in evolution, close to the B1-B subfamily. Comparative gene expression in 61 mouse tissues revealed that B1-X35S-containing genes had lower median expression levels than those with canonical B1 TEs, suggesting a repressive role for X35S. Indeed, X35S was functional and able to bind aryl hydrocarbon (dioxin) receptor (AhR) and Slug and, importantly, to repress cis-reporter genes. Moreover, AhR and Slug were recruited to X35S in vivo and repressed the endogenous expression of X35S-containing genes. Our results demonstrate the existence of a widely present B1 subfamily in the mouse. Because AhR and Slug are relevant in tumor development and differentiation, X35S may represent a genome-wide regulatory mechanism and a tool to modulate gene expression.
Collapse
|
3732
|
Abstract
Biologists have until now conceded that bacterial gene transfer to multicellular animals is relatively uncommon in Nature. A new study showing promiscuous insertions of bacterial endosymbiont genes into invertebrate genomes ushers in a shift in this paradigm.
Collapse
Affiliation(s)
- Seth R Bordenstein
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, The Marine Biological Laboratory, Woods Hole, Massachusetts 02543, USA.
| |
Collapse
|
3733
|
Liakakos T, Roukos DH. More controversy than ever - challenges and promises towards personalized treatment of gastric cancer. Ann Surg Oncol 2008; 15:956-60. [PMID: 18214617 PMCID: PMC2266788 DOI: 10.1245/s10434-007-9798-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 12/12/2007] [Indexed: 02/03/2023]
Affiliation(s)
- Theodore Liakakos
- Department of Surgery, Ioannina University School of Medicine, Ioannina University, GR-45110, Ioannina, 45, Greece
| | | |
Collapse
|
3734
|
A novel RNA transcript with antiapoptotic function is silenced in fragile X syndrome. PLoS One 2008; 3:e1486. [PMID: 18213394 PMCID: PMC2194623 DOI: 10.1371/journal.pone.0001486] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Accepted: 12/21/2007] [Indexed: 01/07/2023] Open
Abstract
Several genome-wide transcriptomics efforts have shown that a large percentage of the mammalian genome is transcribed into RNAs, however, only a small percentage (1-2%) of these RNAs is translated into proteins. Currently there is an intense interest in characterizing the function of the different classes of noncoding RNAs and their relevance to human disease. Using genomic approaches we discovered FMR4, a primate-specific noncoding RNA transcript (2.4 kb) that resides upstream and likely shares a bidirectional promoter with FMR1. FMR4 is a product of RNA polymerase II and has a similar half-life to FMR1. The CGG expansion in the 5' UTR of FMR1 appears to affect transcription in both directions as we found FMR4, similar to FMR1, to be silenced in fragile X patients and up-regulated in premutation carriers. Knockdown of FMR4 by several siRNAs did not affect FMR1 expression, nor vice versa, suggesting that FMR4 is not a direct regulatory transcript for FMR1. However, FMR4 markedly affected human cell proliferation in vitro; siRNAs knockdown of FMR4 resulted in alterations in the cell cycle and increased apoptosis, while the overexpression of FMR4 caused an increase in cell proliferation. Collectively, our results demonstrate an antiapoptotic function of FMR4 and provide evidence that a well-studied genomic locus can show unexpected functional complexity. It cannot be excluded that altered FMR4 expression might contribute to aspects of the clinical presentation of fragile X syndrome and/or related disorders.
Collapse
|
3735
|
Kemmer D, Podowski RM, Yusuf D, Brumm J, Cheung W, Wahlestedt C, Lenhard B, Wasserman WW. Gene characterization index: assessing the depth of gene annotation. PLoS One 2008; 3:e1440. [PMID: 18213364 PMCID: PMC2194620 DOI: 10.1371/journal.pone.0001440] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Accepted: 12/16/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We introduce the Gene Characterization Index, a bioinformatics method for scoring the extent to which a protein-encoding gene is functionally described. Inherently a reflection of human perception, the Gene Characterization Index is applied for assessing the characterization status of individual genes, thus serving the advancement of both genome annotation and applied genomics research by rapid and unbiased identification of groups of uncharacterized genes for diverse applications such as directed functional studies and delineation of novel drug targets. METHODOLOGY/PRINCIPAL FINDINGS The scoring procedure is based on a global survey of researchers, who assigned characterization scores from 1 (poor) to 10 (extensive) for a sample of genes based on major online resources. By evaluating the survey as training data, we developed a bioinformatics procedure to assign gene characterization scores to all genes in the human genome. We analyzed snapshots of functional genome annotation over a period of 6 years to assess temporal changes reflected by the increase of the average Gene Characterization Index. Applying the Gene Characterization Index to genes within pharmaceutically relevant classes, we confirmed known drug targets as high-scoring genes and revealed potentially interesting novel targets with low characterization indexes. Removing known drug targets and genes linked to sequence-related patent filings from the entirety of indexed genes, we identified sets of low-scoring genes particularly suited for further experimental investigation. CONCLUSIONS/SIGNIFICANCE The Gene Characterization Index is intended to serve as a tool to the scientific community and granting agencies for focusing resources and efforts on unexplored areas of the genome. The Gene Characterization Index is available from http://cisreg.ca/gci/.
Collapse
Affiliation(s)
- Danielle Kemmer
- Center for Genomics and Bioinformatics, Karolinska Institute, Stockholm, Sweden
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Raf M. Podowski
- Center for Genomics and Bioinformatics, Karolinska Institute, Stockholm, Sweden
| | - Dimas Yusuf
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Jochen Brumm
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- Department of Statistics, University of British Columbia, Vancouver, Canada
| | - Warren Cheung
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Claes Wahlestedt
- Center for Genomics and Bioinformatics, Karolinska Institute, Stockholm, Sweden
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Boris Lenhard
- Center for Genomics and Bioinformatics, Karolinska Institute, Stockholm, Sweden
- Computational Biology Unit, Bergen Center for Computational Science, Sars International Centre for Marine Molecular Biology, Unifob AS, University of Bergen, Bergen, Norway
| | - Wyeth W. Wasserman
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
3736
|
Ge X, Rubinstein WS, Jung YC, Wu Q. Genome-wide analysis of antisense transcription with Affymetrix exon array. BMC Genomics 2008; 9:27. [PMID: 18211689 PMCID: PMC2257944 DOI: 10.1186/1471-2164-9-27] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Accepted: 01/22/2008] [Indexed: 01/11/2023] Open
Abstract
Background A large number of natural antisense transcripts have been identified in human and mouse genomes. Study of their potential functions clearly requires cost-efficient method for expression analysis. Results Here we show that Affymetrix Exon arrays, which were designed to detect conventional transcripts in the sense orientation, can be used to monitor antisense expression across all exonic loci in mammalian genomes. Through modification of the cDNA synthesis protocol, we labeled single-strand cDNA in the reverse orientation as in the standard protocol, thus enabling the detection of antisense transcripts using the same array. Applying this technique to human Jurkat cells, we identified antisense transcription at 2,088 exonic loci of 1,516 UniGene clusters. Many of these antisense transcripts were not observed previously and some were validated by orientation-specific RT-PCR. Conclusion Our results suggest that with a modified protocol Affymetrix human, mouse and rat Exon arrays can be used as a routine method for genome-wide analysis of antisense transcription in these genomes.
Collapse
Affiliation(s)
- Xijin Ge
- Department of Mathematics and Statistics, South Dakota State University, Box 2220, Brookings, SD 57007, USA.
| | | | | | | |
Collapse
|
3737
|
Marino MJ, Knutsen LJS, Williams M. Emerging Opportunities for Antipsychotic Drug Discovery in the Postgenomic Era. J Med Chem 2008; 51:1077-107. [PMID: 18198826 DOI: 10.1021/jm701094q] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Michael J. Marino
- Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380
| | - Lars J. S. Knutsen
- Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380
| | - Michael Williams
- Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380
| |
Collapse
|
3738
|
Roukos DH. Innovative genomic-based model for personalized treatment of gastric cancer: integrating current standards and new technologies. Expert Rev Mol Diagn 2008; 8:29-39. [PMID: 18088228 DOI: 10.1586/14737159.8.1.29] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the era of network biology, understanding the complexity of the signaling pathways network in cancer origin, progression and metastasis will dramatically alter and improve treatment strategies. Prognosis of gastric cancer remains poor. Clinical decisions on treatment are based on tumor-node-metastasis (TNM) staging, but are suboptimal. This perspective review, integrating several concepts, including cancer stem cells, provides a novel treatment model for tailoring the best treatment in individual patients with gastric cancer. Biologic metastatic steps (invasion, angiogenesis, intra/extravasation, colonization and microenvironment at distant organs) are orchestrated by mutated genes. Identifying and profiling these key genes and their interactions with environmental factors such as Helicobacter pylori, driver mutations and interacting signaling pathways using high-throughput technologies (including omics, resequencing, genome-wide associations studies and RNAi) in unbiased studies can lead to the development of both novel biomarkers and targeted agents. A comprehensive bench-to-bedside treatment-guided algorithm is provided for optimum preoperative or postoperative combination of cytotoxic and targeted agents. The protocol can be applied with adequate modification for most solid tumors.
Collapse
Affiliation(s)
- Dimitrios H Roukos
- Surgical Oncology Research Unit, Department of Surgery, Ioannina University School of Medicine, GR 451 10 Ioannina, Greece.
| |
Collapse
|
3739
|
Costa FF. Non-coding RNAs, epigenetics and complexity. Gene 2008; 410:9-17. [PMID: 18226475 DOI: 10.1016/j.gene.2007.12.008] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2007] [Revised: 11/26/2007] [Accepted: 12/07/2007] [Indexed: 01/05/2023]
Abstract
Several aspects of epigenetics are strongly linked to non-coding RNAs, especially small RNAs that can direct the cytosine methylation and histone modifications that are implicated in gene expression regulation in complex organisms. A fundamental characteristic of epigenetics is that the same genome can show alternative phenotypes, which are based in different epigenetic states. Some of the most studied complex epigenetic phenomena including transposon activity and silencing recently exemplified by piRNAs (piwi-interacting RNAs), position effect variegation, X-chromosome inactivation, parental imprinting, and paramutation have direct or indirect participation of an RNA component. Conceivably, most of the non-coding RNAs with no described function yet, are players in epigenetic mechanisms that are still not completely understood. In that regard, RNAs were recently implicated in new mechanisms of genetic information transfer in yeast, plants and mice. In this review article, the hypothesis that non-coding RNAs might be the main component of complex organisms acquired during evolution will be explored. The question of how evolutionary theories have been challenged by these molecules in association with epigenetic mechanisms will also be discussed here.
Collapse
Affiliation(s)
- Fabrício F Costa
- Cancer Biology and Epigenomics Program, Children's Memorial Research Center and Northwestern University's Feinberg School of Medicine, 2300 Children's Plaza, Chicago, IL 60614, USA.
| |
Collapse
|
3740
|
On the relation between promoter divergence and gene expression evolution. Mol Syst Biol 2008; 4:159. [PMID: 18197176 PMCID: PMC2238714 DOI: 10.1038/msb4100198] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Accepted: 11/06/2007] [Indexed: 01/05/2023] Open
Abstract
Recent studies have characterized significant differences in the cis-regulatory sequences of related organisms, but the impact of these differences on gene expression remains largely unexplored. Here, we show that most previously identified differences in transcription factor (TF)-binding sequences of yeasts and mammals have no detectable effect on gene expression, suggesting that compensatory mechanisms allow promoters to rapidly evolve while maintaining a stabilized expression pattern. To examine the impact of changes in cis-regulatory elements in a more controlled setting, we compared the genes induced during mating of three yeast species. This response is governed by a single TF (STE12), and variations in its predicted binding sites can indeed account for about half of the observed expression differences. The remaining unexplained differences are correlated with the increased divergence of the sequences that flank the binding sites and an apparent modulation of chromatin structure. Our analysis emphasizes the flexibility of promoter structure, and highlights the interplay between specific binding sites and general chromatin structure in the control of gene expression.
Collapse
|
3741
|
Lindgren CM, McCarthy MI. Mechanisms of disease: genetic insights into the etiology of type 2 diabetes and obesity. ACTA ACUST UNITED AC 2008; 4:156-63. [PMID: 18212765 DOI: 10.1038/ncpendmet0723] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 11/16/2007] [Indexed: 12/13/2022]
Abstract
Until recently, progress in identification of the genetic variants influencing predisposition to common forms of diabetes and obesity has been slow, a sharp contrast to the large number of genes implicated in rare monogenic forms of both conditions. Recent advances have transformed the situation, however, enabling researchers to undertake well-powered scans able to detect association signals across the entire genome. For type 2 diabetes, the six high-density genome-wide association studies so far performed have extended the number of loci harboring common variants implicated in diabetes susceptibility into double figures. One of these loci, mapping to the fat mass and obesity associated gene (FTO), influences diabetes risk through a primary effect on fat mass, making this the first common variant known to influence weight and individual risk of obesity. These findings offer two main avenues for clinical translation. First, the identification of new pathways involved in disease predisposition-for example, those influencing zinc transport and pancreatic islet regeneration in the case of type 2 diabetes-offers opportunities for development of novel therapeutic and preventative approaches. Second, with continuing efforts to identify additional genetic variants, it may become possible to use patterns of predisposition to tailor individual management of these conditions.
Collapse
Affiliation(s)
- Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics at the University of Oxford, Oxford, UK
| | | |
Collapse
|
3742
|
The Environmental Polymorphisms Registry: a DNA resource to study genetic susceptibility loci. Hum Genet 2008; 123:207-14. [PMID: 18193459 DOI: 10.1007/s00439-007-0457-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Accepted: 12/18/2007] [Indexed: 10/22/2022]
Abstract
The National Institute of Environmental Health Sciences is establishing a DNA repository named the Environmental Polymorphisms Registry (EPR). The goal is to recruit 20,000 subjects from the greater Research Triangle Park region of North Carolina and collect a sample of each subject's DNA for genetic study. Personal information is obtained from each EPR subject and linked to their sample in coded form. Once individuals with the genotypes of interest are identified, their samples are decoded, and their names and contact information are given to scientists for follow-up studies in which genotype is important. "Recruit-by-genotype" resources such as the EPR require a transparent consent process and rigorous human subjects protection measures. Unlike the EPR, most US DNA resources are anonymous. Once scientists identify potentially significant genetic variants, they must screen new populations to find individuals with the variants of interest to study. The EPR eliminates this time consuming and expensive step. In designing the EPR, consideration was given to achieving high response rates, minimizing attrition and maximizing usefulness for future research studies. Subjects are recruited from outpatient clinics in area medical centers as well as from the general population to ascertain individuals in diverse states of health. Data are collected on race, ethnicity, gender and age, and are monitored for demographic diversity. As of November 2007, 7,788 individuals have been recruited into the EPR and their DNA samples have been used in numerous genetic studies. EPR subjects have also been solicited for several follow-up studies with high response rates (>90%). The success of the EPR based on the number of subjects recruited and genetic studies underway, suggests that it will be a model for future DNA resources.
Collapse
|
3743
|
Eddy J, Maizels N. Conserved elements with potential to form polymorphic G-quadruplex structures in the first intron of human genes. Nucleic Acids Res 2008; 36:1321-33. [PMID: 18187510 PMCID: PMC2275096 DOI: 10.1093/nar/gkm1138] [Citation(s) in RCA: 232] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
To understand how potential for G-quadruplex formation might influence regulation of gene expression, we examined the 2 kb spanning the transcription start sites (TSS) of the 18 217 human RefSeq genes, distinguishing contributions of template and nontemplate strands. Regions both upstream and downstream of the TSS are G-rich, but the downstream region displays a clear bias toward G-richness on the nontemplate strand. Upstream of the TSS, much of the G-richness and potential for G-quadruplex formation derives from the presence of well-defined canonical regulatory motifs in duplex DNA, including CpG dinucleotides which are sites of regulatory methylation, and motifs recognized by the transcription factor SP1. This challenges the notion that quadruplex formation upstream of the TSS contributes to regulation of gene expression. Downstream of the TSS, G-richness is concentrated in the first intron, and on the nontemplate strand, where polymorphic sequence elements with potential to form G-quadruplex structures and which cannot be accounted for by known regulatory motifs are found in almost 3000 (16%) of the human RefSeq genes, and are conserved through frogs. These elements could in principle be recognized either as DNA or as RNA, providing structural targets for regulation at the level of transcription or RNA processing.
Collapse
Affiliation(s)
- Johanna Eddy
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195-7650, USA
| | | |
Collapse
|
3744
|
Nelson JD, Flanagin S, Kawata Y, Denisenko O, Bomsztyk K. Transcription of laminin gamma1 chain gene in rat mesangial cells: constitutive and inducible RNA polymerase II recruitment and chromatin states. Am J Physiol Renal Physiol 2008; 294:F525-33. [PMID: 18184742 DOI: 10.1152/ajprenal.00299.2007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The laminin gamma1 chain, a critical component of the extracellular matrix, is encoded by the 125-kb-long Lamc1 locus. We profiled RNA polymerase II (Pol II) and histone modifications along the Lamc1 locus to explore transcription of this gene in its native chromatin environment. Treatment with 12-O-tetradecanoylphorbol-13-acetate increased Lamc1 mRNA in rat mesangial cells (RMC). This increase was matched by an increase in Pol II density along the entire length of the Lamc1 locus. In contrast, in the hepatocarcinoma cell line (HTC-IR) an increase in Pol II density was restricted to the promoter and was not followed by mRNA induction. The pattern of histone H3 methylation was similar for both cell types but an increase in H3 lysine 9 acetylation observed at the 5'-end was weaker in HTC-IR cells than in RMC. All of the histone modifications showed spatial patterns where levels differed greatly between the 5'- and 3'-ends of Lamc1. Conversely, at the short, highly induced egr-1 gene the differences in chromatin marks between the 5'- and 3'-ends were much smaller. The results of this study suggest that 1) Lamc1 transcription can be controlled after transcription initiation, to our knowledge, the first time this has been shown in an extracellular matrix gene, and 2) the length of a gene is a factor that can affect the chromatin environment for Pol II elongation.
Collapse
Affiliation(s)
- Joel D Nelson
- Molecular and Cellular Biology Program, University of Washington Medicine Lake Union, Seattle, Washington 98109, USA
| | | | | | | | | |
Collapse
|
3745
|
Schneider R, Grosschedl R. Dynamics and interplay of nuclear architecture, genome organization, and gene expression. Genes Dev 2008; 21:3027-43. [PMID: 18056419 DOI: 10.1101/gad.1604607] [Citation(s) in RCA: 322] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The organization of the genome in the nucleus of a eukaryotic cell is fairly complex and dynamic. Various features of the nuclear architecture, including compartmentalization of molecular machines and the spatial arrangement of genomic sequences, help to carry out and regulate nuclear processes, such as DNA replication, DNA repair, gene transcription, RNA processing, and mRNA transport. Compartmentalized multiprotein complexes undergo extensive modifications or exchange of protein subunits, allowing for an exquisite dynamics of structural components and functional processes of the nucleus. The architecture of the interphase nucleus is linked to the spatial arrangement of genes and gene clusters, the structure of chromatin, and the accessibility of regulatory DNA elements. In this review, we discuss recent studies that have provided exciting insight into the interplay between nuclear architecture, genome organization, and gene expression.
Collapse
|
3746
|
|
3747
|
Wu JQ, Du J, Rozowsky J, Zhang Z, Urban AE, Euskirchen G, Weissman S, Gerstein M, Snyder M. Systematic analysis of transcribed loci in ENCODE regions using RACE sequencing reveals extensive transcription in the human genome. Genome Biol 2008; 9:R3. [PMID: 18173853 PMCID: PMC2395237 DOI: 10.1186/gb-2008-9-1-r3] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 12/06/2007] [Accepted: 01/03/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent studies of the mammalian transcriptome have revealed a large number of additional transcribed regions and extraordinary complexity in transcript diversity. However, there is still much uncertainty regarding precisely what portion of the genome is transcribed, the exact structures of these novel transcripts, and the levels of the transcripts produced. RESULTS We have interrogated the transcribed loci in 420 selected ENCyclopedia Of DNA Elements (ENCODE) regions using rapid amplification of cDNA ends (RACE) sequencing. We analyzed annotated known gene regions, but primarily we focused on novel transcriptionally active regions (TARs), which were previously identified by high-density oligonucleotide tiling arrays and on random regions that were not believed to be transcribed. We found RACE sequencing to be very sensitive and were able to detect low levels of transcripts in specific cell types that were not detectable by microarrays. We also observed many instances of sense-antisense transcripts; further analysis suggests that many of the antisense transcripts (but not all) may be artifacts generated from the reverse transcription reaction. Our results show that the majority of the novel TARs analyzed (60%) are connected to other novel TARs or known exons. Of previously unannotated random regions, 17% were shown to produce overlapping transcripts. Furthermore, it is estimated that 9% of the novel transcripts encode proteins. CONCLUSION We conclude that RACE sequencing is an efficient, sensitive, and highly accurate method for characterization of the transcriptome of specific cell/tissue types. Using this method, it appears that much of the genome is represented in polyA+ RNA. Moreover, a fraction of the novel RNAs can encode protein and are likely to be functional.
Collapse
Affiliation(s)
- Jia Qian Wu
- Molecular, Cellular and Developmental Biology Department, KBT918, Yale University, New Haven, Connecticut 06511, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
3748
|
|
3749
|
Brent MR. Steady progress and recent breakthroughs in the accuracy of automated genome annotation. Nat Rev Genet 2008; 9:62-73. [DOI: 10.1038/nrg2220] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
3750
|
Rajangam AS, Yang H, Teeri TT, Arvestad L. Evolution of a domain conserved in microtubule-associated proteins of eukaryotes. Adv Appl Bioinform Chem 2008; 1:51-69. [PMID: 21918606 PMCID: PMC3169935 DOI: 10.2147/aabc.s3211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The microtubule network, the major organelle of the eukaryotic cytoskeleton, is involved in cell division and differentiation but also with many other cellular functions. In plants, microtubules seem to be involved in the ordered deposition of cellulose microfibrils by a so far unknown mechanism. Microtubule-associated proteins (MAP) typically contain various domains targeting or binding proteins with different functions to microtubules. Here we have investigated a proposed microtubule-targeting domain, TPX2, first identified in the Kinesin-like protein 2 in Xenopus. A TPX2 containing microtubule binding protein, PttMAP20, has been recently identified in poplar tissues undergoing xylogenesis. Furthermore, the herbicide 2,6-dichlorobenzonitrile (DCB), which is a known inhibitor of cellulose synthesis, was shown to bind specifically to PttMAP20. It is thus possible that PttMAP20 may have a role in coupling cellulose biosynthesis and the microtubular networks in poplar secondary cell walls. In order to get more insight into the occurrence, evolution and potential functions of TPX2-containing proteins we have carried out bioinformatic analysis for all genes so far found to encode TPX2 domains with special reference to poplar PttMAP20 and its putative orthologs in other plants.
Collapse
Affiliation(s)
- Alex S Rajangam
- KTH Biotechnology, Swedish Center for Biomimetic Fiber Engineering, AlbaNova, Stockholm, Sweden
| | | | | | | |
Collapse
|