1
|
Liu H, Earley B, Mendoza A, Hunt P, Teng S, Schneider DL, Kornfeld K. A single high-zinc activation enhancer can control two genes oriented head-to-head in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2025:jkaf089. [PMID: 40408183 DOI: 10.1093/g3journal/jkaf089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/04/2025] [Indexed: 05/25/2025]
Abstract
Enhancers play critical roles in gene expression, but a full understanding of their complex functions has yet to be defined. The cellular response to excess zinc levels in Caenorhabditis elegans requires the HIZR-1 transcription factor, which binds the high-zinc activation (HZA) enhancer in the promoters of multiple target genes. Cadmium hijacks the excess zinc response by binding and activating HIZR-1. By analyzing the genome-wide transcriptional response to excess zinc and cadmium, we identified two positions in the genome where head-to-head oriented genes are both induced by metals. In both examples, a single predicted HZA enhancer is positioned between the two translational start sites. We hypothesized that a single enhancer can control both head-to-head genes, an arrangement that has not been extensively characterized. To test this hypothesis, we used CRISPR genome editing to precisely delete the HZAmT enhancer positioned between mtl-2 and T08G5.1; in this mutant, both head-to-head genes display severely reduced zinc-activated transcription, whereas zinc-activated transcription of more distant genes was not strongly affected. Deleting the HZAcF enhancer positioned between cdr-1 and F35E8.10 caused both head-to-head genes to display reduced cadmium-activated transcription, whereas cadmium-activated transcription of more distant genes was not strongly affected. These studies rigorously document that a single HZA enhancer can control two head-to-head genes, advancing our understanding of the diverse functions of enhancers.
Collapse
Affiliation(s)
- Hanwenheng Liu
- Department of Developmental Biology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Brian Earley
- Department of Developmental Biology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Adelita Mendoza
- Department of Developmental Biology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Patrick Hunt
- Department of Developmental Biology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Sean Teng
- Department of Developmental Biology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Daniel Luke Schneider
- Department of Developmental Biology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kerry Kornfeld
- Department of Developmental Biology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
2
|
Liu H, Earley B, Mendoza A, Hunt P, Teng S, Schneider DL, Kornfeld K. A single high-zinc activation enhancer can control two genes orientated head-to-head in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.19.624376. [PMID: 40196504 PMCID: PMC11974713 DOI: 10.1101/2024.11.19.624376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Enhancers play critical roles in gene expression, but a full understanding of their complex functions has yet to be defined. The cellular response to excess zinc levels in C. elegans requires the HIZR-1 transcription factor, which binds the high-zinc activation (HZA) enhancer in the promoters of multiple target genes. Cadmium hijacks the excess zinc response by binding and activating HIZR-1. By analyzing the genome-wide transcriptional response to excess zinc and cadmium, we identified two positions in the genome where head-to-head oriented genes are both induced by metals. In both examples, a single predicted HZA enhancer is positioned between the two translational start sites. We hypothesized that a single enhancer can control both head-to-head genes, an arrangement that has not been extensively characterized. To test this hypothesis, we used CRISPR genome editing to precisely delete the HZAmT enhancer positioned between mtl-2 and T08G5.1; in this mutant, both head-to-head genes display severely reduced zinc-activated transcription, whereas zinc-activated transcription of more distant genes was not strongly affected. Deleting the HZAcF enhancer positioned between cdr-1 and F35E8.10 caused both head-to-head genes to display reduced cadmium-activated transcription, whereas cadmium-activated transcription of more distant genes was not strongly affected. These studies rigorously document that a single HZA enhancer can control two head-to-head genes, advancing our understanding of the diverse functions of enhancers.
Collapse
Affiliation(s)
- Hanwenheng Liu
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63130, USA
| | - Brian Earley
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63130, USA
- Current address: Blackfoot High School, 870 S. Fisher Avenue, Blackfoot, Idaho 83221, USA
| | - Adelita Mendoza
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63130, USA
- Current address: University of Colorado at Boulder, 3415 Colorado Ave, Boulder, CO 80303, USA
| | - Patrick Hunt
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63130, USA
| | - Sean Teng
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63130, USA
- Current address: Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | - Daniel L. Schneider
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63130, USA
| | - Kerry Kornfeld
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63130, USA
| |
Collapse
|
3
|
Nojima T, Proudfoot NJ. Mechanisms of lncRNA biogenesis as revealed by nascent transcriptomics. Nat Rev Mol Cell Biol 2022; 23:389-406. [DOI: 10.1038/s41580-021-00447-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 12/14/2022]
|
4
|
Law WD, Fogarty EA, Vester A, Antonellis A. A genome-wide assessment of conserved SNP alleles reveals a panel of regulatory SNPs relevant to the peripheral nerve. BMC Genomics 2018; 19:311. [PMID: 29716548 PMCID: PMC5930951 DOI: 10.1186/s12864-018-4692-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 04/17/2018] [Indexed: 12/29/2022] Open
Abstract
Background Identifying functional non-coding variation is critical for defining the genetic contributions to human disease. While single-nucleotide polymorphisms (SNPs) within cis-acting transcriptional regulatory elements have been implicated in disease pathogenesis, not all cell types have been assessed and functional validations have been limited. In particular, the cells of the peripheral nervous system have been excluded from genome-wide efforts to link non-coding SNPs to altered gene function. Addressing this gap is essential for defining the genetic architecture of diseases that affect the peripheral nerve. We developed a computational pipeline to identify SNPs that affect regulatory function (rSNPs) and evaluated our predictions on a set of 144 regions in Schwann cells, motor neurons, and muscle cells. Results We identified 28 regions that display regulatory activity in at least one cell type and 13 SNPs that affect regulatory function. We then tailored our pipeline to one peripheral nerve cell type by incorporating SOX10 ChIP-Seq data; SOX10 is essential for Schwann cells. We prioritized 22 putative SOX10 response elements harboring a SNP and rapidly validated two rSNPs. We then selected one of these elements for further characterization to assess the biological relevance of our approach. Deletion of the element from the genome of cultured Schwann cells—followed by differential gene expression studies—revealed Tubb2b as a candidate target gene. Studying the enhancer in developing mouse embryos revealed activity in SOX10-positive cells including the dorsal root ganglia and melanoblasts. Conclusions Our efforts provide insight into the utility of employing strict conservation for rSNP discovery. This strategy, combined with functional analyses, can yield candidate target genes. In support of this, our efforts suggest that investigating the role of Tubb2b in SOX10-positive cells may reveal novel biology within these cell populations. Electronic supplementary material The online version of this article (10.1186/s12864-018-4692-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William D Law
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Elizabeth A Fogarty
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Aimée Vester
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anthony Antonellis
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA. .,Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Neurology, University of Michigan Medical School, 3710A Medical Sciences II, 1241 E. Catherine St. SPC 5618, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
5
|
Reporter Analyses Reveal Redundant Enhancers that Confer Robustness on Cis-Regulatory Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018. [PMID: 29542081 DOI: 10.1007/978-981-10-7545-2_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
Reporter analyses of Hox1 and Brachyury (Bra) genes have revealed examples of redundant enhancers that provide regulatory robustness. Retinoic acid (RA) activates through an RA-response element the transcription of Hox1 in the nerve cord of the ascidian Ciona intestinalis. We also found a weak RA-independent neural enhancer within the second intron of Hox1. The Hox1 gene in the larvacean Oikopleura dioica is also expressed in the nerve cord. The O. dioica genome, however, does not contain the RA receptor-encoding gene, and the expression of Hox1 has become independent of RA. We have found that the upstream sequence of the O. dioica Hox1 was able to activate reporter gene expression in the nerve cord of the C. intestinalis embryo, suggesting that an RA-independent regulatory system in the nerve cord might be common in larvaceans and ascidians. This RA-independent redundant regulatory system may have facilitated the Oikopleura ancestor losing RA signaling without an apparent impact on Hox1 expression domains. On the other hand, vertebrate Bra is expressed in the ventral mesoderm and notochord, whereas its ascidian ortholog is exclusively expressed in the notochord. Fibroblast growth factor (FGF) induces Bra in the ventral mesoderm in vertebrates, whereas it induces Bra in the notochord in ascidians. Disruption of the FGF signal does not completely silence Bra expression in ascidians, suggesting that FGF-dependent and independent enhancers might comprise a redundant regulatory system in ascidians. The existence of redundant enhancers, therefore, provides regulatory robustness that may facilitate the acquisition of new expression domains.
Collapse
|
6
|
Enhancer adoption caused by genomic insertion elicits interdigital Shh expression and syndactyly in mouse. Proc Natl Acad Sci U S A 2017; 115:1021-1026. [PMID: 29255029 PMCID: PMC5798340 DOI: 10.1073/pnas.1713339115] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In this study, we reexamined an old mouse mutant named Hammer toe (Hm), which arose spontaneously almost a half century ago and exhibits a limb phenotype with webbing. We revealed that a 150-kb noncoding genomic fragment that was originally located in chromosome 14 has been inserted into a genomic region proximal to Sonic hedgehog (Shh), located in chromosome 5. This inserted fragment possesses enhancer activity to induce Shh expression in the interdigital regions in Hm, which in turn down-regulates bone morphogenetic protein signaling and eventually results in syndactyly and web formation. Since the donor fragment residing in chromosome 14 has enhancer activity to induce interdigital gene expression, the Hm mutation appears to be an archetypal case of enhancer adoption. Acquisition of new cis-regulatory elements (CREs) can cause alteration of developmental gene regulation and may introduce morphological novelty in evolution. Although structural variation in the genome generated by chromosomal rearrangement is one possible source of new CREs, only a few examples are known, except for cases of retrotransposition. In this study, we show the acquisition of novel regulatory sequences as a result of large genomic insertion in the spontaneous mouse mutation Hammer toe (Hm). Hm mice exhibit syndactyly with webbing, due to suppression of interdigital cell death in limb development. We reveal that, in the Hm genome, a 150-kb noncoding DNA fragment from chromosome 14 is inserted into the region upstream of the Sonic hedgehog (Shh) promoter in chromosome 5. Phenotyping of mouse embryos with a series of CRISPR/Cas9-aided partial deletion of the 150-kb insert clearly indicated that two different regions are necessary for the syndactyly phenotype of Hm. We found that each of the two regions contains at least one enhancer for interdigital regulation. These results show that a set of enhancers brought by the large genomic insertion elicits the interdigital Shh expression and the Hm phenotype. Transcriptome analysis indicates that ectopic expression of Shh up-regulates Chordin (Chrd) that antagonizes bone morphogenetic protein signaling in the interdigital region. Indeed, Chrd-overexpressing transgenic mice recapitulated syndactyly with webbing. Thus, the Hm mutation provides an insight into enhancer acquisition as a source of creation of novel gene regulation.
Collapse
|
7
|
Posner M, Murray KL, McDonald MS, Eighinger H, Andrew B, Drossman A, Haley Z, Nussbaum J, David LL, Lampi KJ. The zebrafish as a model system for analyzing mammalian and native α-crystallin promoter function. PeerJ 2017; 5:e4093. [PMID: 29201567 PMCID: PMC5708185 DOI: 10.7717/peerj.4093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 11/04/2017] [Indexed: 12/24/2022] Open
Abstract
Previous studies have used the zebrafish to investigate the biology of lens crystallin proteins and their roles in development and disease. However, little is known about zebrafish α-crystallin promoter function, how it compares to that of mammals, or whether mammalian α-crystallin promoter activity can be assessed using zebrafish embryos. We injected a variety of α-crystallin promoter fragments from each species combined with the coding sequence for green fluorescent protein (GFP) into zebrafish zygotes to determine the resulting spatiotemporal expression patterns in the developing embryo. We also measured mRNA levels and protein abundance for all three zebrafish α-crystallins. Our data showed that mouse and zebrafish αA-crystallin promoters generated similar GFP expression in the lens, but with earlier onset when using mouse promoters. Expression was also found in notochord and skeletal muscle in a smaller percentage of embryos. Mouse αB-crystallin promoter fragments drove GFP expression primarily in zebrafish skeletal muscle, with less common expression in notochord, lens, heart and in extraocular regions of the eye. A short fragment containing only a lens-specific enhancer region increased lens and notochord GFP expression while decreasing muscle expression, suggesting that the influence of mouse promoter control regions carries over into zebrafish embryos. The two paralogous zebrafish αB-crystallin promoters produced subtly different expression profiles, with the aBa promoter driving expression equally in notochord and skeletal muscle while the αBb promoter resulted primarily in skeletal muscle expression. Messenger RNA for zebrafish αA increased between 1 and 2 days post fertilization (dpf), αBa increased between 4 and 5 dpf, but αBb remained at baseline levels through 5 dpf. Parallel reaction monitoring (PRM) mass spectrometry was used to detect αA, aBa, and αBb peptides in digests of zebrafish embryos. In whole embryos, αA-crystallin was first detected by 2 dpf, peaked in abundance by 4–5 dpf, and was localized to the eye. αBa was detected in whole embryo at nearly constant levels from 1–6 dpf, was also localized primarily to the eye, and its abundance in extraocular tissues decreased from 4–7 dpf. In contrast, due to its low abundance, no αBb protein could be detected in whole embryo, or dissected eye and extraocular tissues. Our results show that mammalian α-crystallin promoters can be efficiently screened in zebrafish embryos and that their controlling regions are well conserved. An ontogenetic shift in zebrafish aBa-crystallin promoter activity provides an interesting system for examining the evolution and control of tissue specificity. Future studies that combine these promoter based approaches with the expanding ability to engineer the zebrafish genome via techniques such as CRISPR/Cas9 will allow the manipulation of protein expression to test hypotheses about lens crystallin function and its relation to lens biology and disease.
Collapse
Affiliation(s)
- Mason Posner
- Department of Biology/Toxicology, Ashland University, Ashland, OH, United States of America
| | - Kelly L Murray
- Department of Biology/Toxicology, Ashland University, Ashland, OH, United States of America
| | - Matthew S McDonald
- Department of Biology/Toxicology, Ashland University, Ashland, OH, United States of America
| | - Hayden Eighinger
- Department of Biology/Toxicology, Ashland University, Ashland, OH, United States of America
| | - Brandon Andrew
- Department of Biology/Toxicology, Ashland University, Ashland, OH, United States of America
| | - Amy Drossman
- Department of Biology/Toxicology, Ashland University, Ashland, OH, United States of America
| | - Zachary Haley
- Department of Biology/Toxicology, Ashland University, Ashland, OH, United States of America
| | - Justin Nussbaum
- Department of Biology, Lakeland Community College, Kirtland, OH, United States of America
| | - Larry L David
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR, United States of America
| | - Kirsten J Lampi
- Department of Integrative Biosciences, Oregon Health and Science University, Portland, OR, United States of America
| |
Collapse
|
8
|
Zhao W, Siegel D, Biton A, Tonqueze OL, Zaitlen N, Ahituv N, Erle DJ. CRISPR-Cas9-mediated functional dissection of 3'-UTRs. Nucleic Acids Res 2017; 45:10800-10810. [PMID: 28985357 PMCID: PMC5737544 DOI: 10.1093/nar/gkx675] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/25/2017] [Indexed: 11/21/2022] Open
Abstract
Many studies using reporter assays have demonstrated that 3′ untranslated regions (3′-UTRs) regulate gene expression by controlling mRNA stability and translation. Due to intrinsic limitations of heterologous reporter assays, we sought to develop a gene editing approach to investigate the regulatory activity of 3′-UTRs in their native context. We initially used dual-CRISPR (clustered, regularly interspaced, short palindromic repeats)-Cas9 targeting to delete DNA regions corresponding to nine chemokine 3′-UTRs that destabilized mRNA in a reporter assay. Targeting six chemokine 3′-UTRs increased chemokine mRNA levels as expected. However, targeting CXCL1, CXCL6 and CXCL8 3′-UTRs unexpectedly led to substantial mRNA decreases. Metabolic labeling assays showed that targeting these three 3′-UTRs increased mRNA stability, as predicted by the reporter assay, while also markedly decreasing transcription, demonstrating an unexpected role for 3′-UTR sequences in transcriptional regulation. We further show that CRISPR–Cas9 targeting of specific 3′-UTR elements can be used for modulating gene expression and for highly parallel localization of active 3′-UTR elements in the native context. Our work demonstrates the duality and complexity of 3′-UTR sequences in regulation of gene expression and provides a useful approach for modulating gene expression and for functional annotation of 3′-UTRs in the native context.
Collapse
Affiliation(s)
- Wenxue Zhao
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - David Siegel
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - Anne Biton
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA.,Centre de Bioinformatique, Biostatistique et Biologie Intégrative, C3BI, USR 3756 Institut Pasteur et CNRS, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Olivier Le Tonqueze
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - Noah Zaitlen
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, Institute for Human Genetics, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - David J Erle
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| |
Collapse
|
9
|
Tam KT, Chan PK, Zhang W, Law PP, Tian Z, Fung Chan GC, Philipsen S, Festenstein R, Tan-Un KC. Identification of a novel distal regulatory element of the human Neuroglobin gene by the chromosome conformation capture approach. Nucleic Acids Res 2017; 45:115-126. [PMID: 27651453 PMCID: PMC5224503 DOI: 10.1093/nar/gkw820] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 12/24/2022] Open
Abstract
Neuroglobin (NGB) is predominantly expressed in the brain and retina. Studies suggest that NGB exerts protective effects to neuronal cells and is implicated in reducing the severity of stroke and Alzheimer's disease. However, little is known about the mechanisms which regulate the cell type-specific expression of the gene. In this study, we hypothesized that distal regulatory elements (DREs) are involved in optimal expression of the NGB gene. By chromosome conformation capture we identified two novel DREs located -70 kb upstream and +100 kb downstream from the NGB gene. ENCODE database showed the presence of DNaseI hypersensitive and transcription factors binding sites in these regions. Further analyses using luciferase reporters and chromatin immunoprecipitation suggested that the -70 kb region upstream of the NGB gene contained a neuronal-specific enhancer and GATA transcription factor binding sites. Knockdown of GATA-2 caused NGB expression to drop dramatically, indicating GATA-2 as an essential transcription factor for the activation of NGB expression. The crucial role of the DRE in NGB expression activation was further confirmed by the drop in NGB level after CRISPR-mediated deletion of the DRE. Taken together, we show that the NGB gene is regulated by a cell type-specific loop formed between its promoter and the novel DRE.
Collapse
MESH Headings
- Binding Sites
- CRISPR-Cas Systems
- Cell Line, Tumor
- Chromosomes, Human, Pair 14/chemistry
- Deoxyribonuclease I/genetics
- Deoxyribonuclease I/metabolism
- GATA2 Transcription Factor/genetics
- GATA2 Transcription Factor/metabolism
- Gene Editing
- Gene Expression Regulation
- Genes, Reporter
- Globins/antagonists & inhibitors
- Globins/genetics
- Globins/metabolism
- HeLa Cells
- Humans
- K562 Cells
- Luciferases/genetics
- Luciferases/metabolism
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuroglobin
- Neurons/cytology
- Neurons/metabolism
- Organ Specificity
- Protein Binding
- RNA, Guide, CRISPR-Cas Systems/genetics
- RNA, Guide, CRISPR-Cas Systems/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Regulatory Elements, Transcriptional
- Signal Transduction
Collapse
Affiliation(s)
- Kin Tung Tam
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
| | - Ping Kei Chan
- Gene Control Mechanisms and Disease Group, Department of Medicine, Division of Brain Sciences and MRC Clinical Sciences Centre, Imperial College School of Medicine, London W12 0NN, United Kingdom
| | - Wei Zhang
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
| | - Pui Pik Law
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
- Gene Control Mechanisms and Disease Group, Department of Medicine, Division of Brain Sciences and MRC Clinical Sciences Centre, Imperial College School of Medicine, London W12 0NN, United Kingdom
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
| | - Zhipeng Tian
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
- School of Professional and Continuing Education (HKU SPACE), The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
| | - Godfrey Chi Fung Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Richard Festenstein
- Gene Control Mechanisms and Disease Group, Department of Medicine, Division of Brain Sciences and MRC Clinical Sciences Centre, Imperial College School of Medicine, London W12 0NN, United Kingdom
| | - Kian Cheng Tan-Un
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
- School of Professional and Continuing Education (HKU SPACE), The University of Hong Kong, Pokfulam Road, Hong Kong S.A.R., China
| |
Collapse
|
10
|
Skvortsova YV, Kondratieva SA, Zinovyeva MV, Nikolaev LG, Azhikina TL, Gainetdinov IV. Intragenic Locus in Human PIWIL2 Gene Shares Promoter and Enhancer Functions. PLoS One 2016; 11:e0156454. [PMID: 27248499 PMCID: PMC4889060 DOI: 10.1371/journal.pone.0156454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/13/2016] [Indexed: 01/18/2023] Open
Abstract
Recently, more evidence supporting common nature of promoters and enhancers has been accumulated. In this work, we present data on chromatin modifications and non-polyadenylated transcription characteristic for enhancers as well as results of in vitro luciferase reporter assays suggesting that PIWIL2 alternative promoter in exon 7 also functions as an enhancer for gene PHYHIP located 60Kb upstream. This finding of an intragenic enhancer serving as a promoter for a shorter protein isoform implies broader impact on understanding enhancer-promoter networks in regulation of gene expression.
Collapse
Affiliation(s)
- Yulia V Skvortsova
- Department of Genomics and Postgenomic Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sofia A Kondratieva
- Department of Genomics and Postgenomic Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Marina V Zinovyeva
- Department of Genomics and Postgenomic Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Lev G Nikolaev
- Department of Genomics and Postgenomic Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Tatyana L Azhikina
- Department of Genomics and Postgenomic Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ildar V Gainetdinov
- Department of Genomics and Postgenomic Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
11
|
Toft DJ, Fuller M, Schipma M, Chen F, Cryns VL, Layden BT. αB-crystallin and HspB2 deficiency is protective from diet-induced glucose intolerance. GENOMICS DATA 2016; 9:10-7. [PMID: 27330996 PMCID: PMC4909821 DOI: 10.1016/j.gdata.2016.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/29/2022]
Abstract
Emerging evidence suggests molecular chaperones have a role in the pathogenesis of obesity and diabetes. As αB-crystallin and HspB2 are molecular chaperones and data suggests their expression is elevated in the skeletal muscle of diabetic and obese animals, we sought to determine if αB-crystallin and HspB2 collectively play a functional role in the metabolic phenotype of diet-induced obesity. Using αB-crystallin/HspB2 knockout and littermate wild-type controls, it was observed that mice on the high fat diet gained more weight as compared to the normal chow group and genotype did not impact this weight gain. To test if the genotype and/or diet influenced glucose homeostasis, intraperitoneal glucose challenge was performed. While similar on normal chow diet, wild-type mice on the high fat diet exhibited higher glucose levels during the glucose challenge compared to the αB-crystallin/HspB2 knockout mice. Although wild-type mice had higher glucose levels, insulin levels were similar for both genotypes. Insulin tolerance testing revealed that αB-crystallin/HspB2 knockout mice were more sensitive to insulin, leading to lower glucose levels over time, which is indicative of a difference in insulin sensitivity between the genotypes on a high fat diet. Transcriptome analyses of skeletal muscle in αB-crystallin/HspB2 knockout and wild-type mice on a normal or high fat diet revealed reductions in cytokine pathway genes in αB-crystallin/HspB2 knockout mice, which may contribute to their improved insulin sensitivity. Collectively, these data reveal that αB-crystallin/HspB2 plays a role in development of insulin resistance during a high fat diet challenge.
Collapse
Affiliation(s)
- Daniel J Toft
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Miles Fuller
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Matthew Schipma
- Next Generation Sequencing Core, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Feng Chen
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Vincent L Cryns
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Brian T Layden
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| |
Collapse
|
12
|
Abstract
αB-crystallin is a widely expressed member of the small heat shock protein family that protects cells from stress by its dual function as a molecular chaperone to preserve proteostasis and as a cell death antagonist that negatively regulates components of the conserved apoptotic cell death machinery. Deregulated expression of αB-crystallin occurs in a broad array of solid tumors and has been linked to tumor progression and poor clinical outcomes. This review will focus on new insights into the molecular mechanisms by which oncogenes, oxidative stress, matrix detachment and other tumor microenvironmental stressors deregulate αB-crystallin expression. We will also review accumulating evidence pointing to an essential role for αB-crystallin in the multi-step metastatic cascade whereby tumor cells colonize distant organs by circumventing a multitude of barriers to cell migration and survival. Finally, we will evaluate emerging strategies to therapeutically target αB-crystallin and/or interacting proteins to selectively activate apoptosis and/or derail the metastatic cascade in an effort to improve outcomes for patients with metastatic disease.
Collapse
|
13
|
Liu S, Yan B, Lai W, Chen L, Xiao D, Xi S, Jiang Y, Dong X, An J, Chen X, Cao Y, Tao Y. As a novel p53 direct target, bidirectional gene HspB2/αB-crystallin regulates the ROS level and Warburg effect. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:592-603. [PMID: 24859470 DOI: 10.1016/j.bbagrm.2014.05.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 05/13/2014] [Accepted: 05/15/2014] [Indexed: 02/02/2023]
Abstract
Many mammalian genes are composed of bidirectional gene pairs with the two genes separated by less than 1.0kb. The transcriptional regulation and function of these bidirectional genes remain largely unclear. Here, we report that bidirectional gene pair HspB2/αB-crystallin, both of which are members of the small heat shock protein gene family, is a novel direct target gene of p53. Two potential binding sites of p53 are present in the intergenic region of HspB2/αB-crystallin. p53 up-regulated the bidirectional promoter activities of HspB2/αB-crystallin. Actinomycin D (ActD), an activator of p53, induces the promoter and protein activities of HspB2/αB-crystallin. p53 binds to two p53 binding sites in the intergenic region of HspB2/αB-crystallin in vitro and in vivo. Moreover, the products of bidirectional gene pair HspB2/αB-crystallin regulate glucose metabolism, intracellular reactive oxygen species (ROS) level and the Warburg effect by affecting metabolic genes, including the synthesis of cytochrome c oxidase 2 (SCO2), hexokinase II (HK2), and TP53-induced glycolysis and apoptosis regulator (TIGAR). The ROS level and the Warburg effect are affected after the depletion of p53, HspB2 and αB-crystallin respectively. Finally, we show that both HspB2 and αB-crystallin are linked with human renal carcinogenesis. These findings provide novel insights into the role of p53 as a regulator of bidirectional gene pair HspB2/αB-crystallin-mediated ROS and the Warburg effect.
Collapse
Affiliation(s)
- Shuang Liu
- Center for Medicine Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Cancer Research Institute, Central South University, Changsha, Hunan 410078, China; Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Bin Yan
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, China; Center for Molecular Imaging, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan 410078, China; Key Laboratory of Carcinogenesis, Ministry of Health, Hunan 410078, China
| | - Weiwei Lai
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, China; Center for Molecular Imaging, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan 410078, China; Key Laboratory of Carcinogenesis, Ministry of Health, Hunan 410078, China
| | - Ling Chen
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, China; Center for Molecular Imaging, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan 410078, China; Key Laboratory of Carcinogenesis, Ministry of Health, Hunan 410078, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Sichuan Xi
- Thoracic Oncology Section, Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892 USA
| | - Yiqun Jiang
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, China; Center for Molecular Imaging, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan 410078, China; Key Laboratory of Carcinogenesis, Ministry of Health, Hunan 410078, China
| | - Xin Dong
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, China; Center for Molecular Imaging, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan 410078, China; Key Laboratory of Carcinogenesis, Ministry of Health, Hunan 410078, China
| | - Jing An
- State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 4010078, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ya Cao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, China; Center for Molecular Imaging, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan 410078, China; Key Laboratory of Carcinogenesis, Ministry of Health, Hunan 410078, China
| | - Yongguang Tao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, China; Center for Molecular Imaging, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan 410078, China; Key Laboratory of Carcinogenesis, Ministry of Health, Hunan 410078, China.
| |
Collapse
|
14
|
Gupta D, Harvey SAK, Kenchegowda D, Swamynathan S, Swamynathan SK. Regulation of mouse lens maturation and gene expression by Krüppel-like factor 4. Exp Eye Res 2013; 116:205-18. [PMID: 24076321 DOI: 10.1016/j.exer.2013.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/29/2013] [Accepted: 09/12/2013] [Indexed: 02/01/2023]
Abstract
Conditional disruption of Klf4 in the surface ectoderm-derived tissues of the eye results in defective cornea, conjunctiva and the lens. This report describes the effects of disruption of Klf4 in the lens in greater detail. Expression of Klf4, first detected in the embryonic day-12 (E12) mouse lens, peaked at E16 and was decreased in later stages. Early embryonic disruption of Klf4 resulted in a smaller lens with cortical vacuolation and nuclear opacity. Microarray comparison of Klf4CN and WT lens transcriptomes revealed fewer changes in the E16.5 (59 increases, 20 decreases of >1.5-fold) than the PN56 Klf4CN lens (239 increases, 182 decreases of >2-fold). Klf4-target genes in the lens were distinct from those previously identified in the cornea, suggesting disparate functions for Klf4 in these functionally related tissues. Transcripts encoding different crystallins were down-regulated in the Klf4CN lens. Shsp/αB-crystallin promoter activity was stimulated upon co-transfection with pCI-Klf4. Mitochondrial density was significantly higher in the Klf4CN lens epithelial cells, consistent with mitochondrial dysfunction being the most significantly affected pathway within the PN56 Klf4CN lens. The Klf4CN lens contained elevated levels of Alox12 and Alox15 transcripts, less reduced glutathione (GSH) and more oxidized glutathione (GSSG) than the WT, suggesting that it is oxidatively stressed. Although the expression of 2087 genes was modulated during WT lens maturation, transcripts encoding crystallins were abundant at E16.5 and remained stable at PN56. Among the 1065 genes whose expression increased during WT lens maturation, there were 104 Klf4-target genes (9.8%) with decreased expression in the PN56 Klf4CN lens. Taken together, these results demonstrate that Klf4 expression is developmentally regulated in the mouse lens, where it controls the expression of genes associated with lens maturation and redox homeostasis.
Collapse
Affiliation(s)
- Divya Gupta
- Department of Ophthalmology, Eye and Ear Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
15
|
Chen RAJ, Down TA, Stempor P, Chen QB, Egelhofer TA, Hillier LW, Jeffers TE, Ahringer J. The landscape of RNA polymerase II transcription initiation in C. elegans reveals promoter and enhancer architectures. Genome Res 2013; 23:1339-47. [PMID: 23550086 PMCID: PMC3730107 DOI: 10.1101/gr.153668.112] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RNA polymerase transcription initiation sites are largely unknown in Caenorhabditis elegans. The initial 5′ end of most protein-coding transcripts is removed by trans-splicing, and noncoding initiation sites have not been investigated. We characterized the landscape of RNA Pol II transcription initiation, identifying 73,500 distinct clusters of initiation. Bidirectional transcription is frequent, with a peak of transcriptional pairing at 120 bp. We assign transcription initiation sites to 7691 protein-coding genes and find that they display features typical of eukaryotic promoters. Strikingly, the majority of initiation events occur in regions with enhancer-like chromatin signatures. Based on the overlap of transcription initiation clusters with mapped transcription factor binding sites, we define 2361 transcribed intergenic enhancers. Remarkably, productive transcription elongation across these enhancers is predominantly in the same orientation as that of the nearest downstream gene. Directed elongation from an upstream enhancer toward a downstream gene could potentially deliver RNA polymerase II to a proximal promoter, or alternatively might function directly as a distal promoter. Our results provide a new resource to investigate transcription regulation in metazoans.
Collapse
Affiliation(s)
- Ron A-J Chen
- The Gurdon Institute, and Department of Genetics, University of Cambridge, Cambridge CB3 0DH, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
16
|
de Thonel A, Le Mouël A, Mezger V. Transcriptional regulation of small HSP-HSF1 and beyond. Int J Biochem Cell Biol 2012; 44:1593-612. [PMID: 22750029 DOI: 10.1016/j.biocel.2012.06.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 06/07/2012] [Accepted: 06/08/2012] [Indexed: 12/16/2022]
Abstract
The members of the small heat shock protein (sHSP) family are molecular chaperones that play major roles in development, stress responses, and diseases, and have been envisioned as targets for therapy, particularly in cancer. The molecular mechanisms that regulate their transcription, in normal, stress, or pathological conditions, are characterized by extreme complexity and subtlety. Although historically linked to the heat shock transcription factors (HSFs), the stress-induced or developmental expression of the diverse members, including HSPB1/Hsp27/Hsp25, αA-crystallin/HSPB4, and αB-crystallin/HSPB5, relies on the combinatory effects of many transcription factors. Coupled with remarkably different cis-element architectures in the sHsp regulatory regions, they confer to each member its developmental expression or stress-inducibility. For example, multiple regulatory pathways coordinate the spatio-temporal expression of mouse αA-, αB-crystallin, and Hsp25 genes during lens development, through the action of master genes, like the large Maf family proteins and Pax6, but also HSF4. The inducibility of Hsp27 and αB-crystallin transcription by various stresses is exerted by HSF-dependent mechanisms, by which concomitant induction of Hsp27 and αB-crystallin expression is observed. In contrast, HSF-independent pathways can lead to αB-crystallin expression, but not to Hsp27 induction. Not surprisingly, deregulation of the expression of sHSP is associated with various pathologies, including cancer, neurodegenerative, or cardiac diseases. However, many questions remain to be addressed, and further elucidation of the developmental mechanisms of sHsp gene transcription might help to unravel the tissue- and stage-specific functions of this fascinating class of proteins, which might prove to be crucial for future therapeutic strategies. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.
Collapse
|
17
|
Sorotokina AN, Chernov IP, Stukacheva EA, Nikolaev LG, Sverdlov ED. [Enhancer activity of DNA fragments from FXYD5-COX7A region of human chromosome 19]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2011; 37:244-52. [PMID: 21721257 DOI: 10.1134/s1068162011020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The enhancer activity of four previously identified within the one megabase region of human chromosome 19 DNA fragments was investigated. All four fragments had similar tissue-specific profile--maximum of enhancer activity was observed in HEK293 and minimum in HeLa cells. Enhancers obtained had pronounced specificity toward cytomegalovirus promoter compared with SV40 promoter. Functional dissection of one of the fragments (enhancer 14) demonstrated that only its inner 127 b.p. part possessed enhancer activity. The negative regulators, i.e. silencers or insulators are probably located in flanking regions of enhancer 14 and limit its effect on promoter. At the same time, enhancer activity of enhancer 14 depends on its orientation relative to promoter that isn't typical to majority of enhancer elements. Inner 127 b.p. fragment contains 11 transcription factor binding sites; 8 of them are factors that take part in immune system regulation. Enhancer 14 is located 500 b.p. upstream of transcription start site of TYROBP (DAP12) gene that codes for of T-killer cells activator protein and possibly functions as tissue-specific enhancer for this gene.
Collapse
|
18
|
Bonello GB, Pham MH, Begum K, Sigala J, Sataranatarajan K, Mummidi S. An evolutionarily conserved TNF-alpha-responsive enhancer in the far upstream region of human CCL2 locus influences its gene expression. THE JOURNAL OF IMMUNOLOGY 2011; 186:7025-38. [PMID: 21551367 DOI: 10.4049/jimmunol.0900643] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Comparative cross-species genomic analysis has served as a powerful tool to discover novel noncoding regulatory regions that influence gene expression in several cytokine loci. In this study, we have identified several evolutionarily conserved regions (ECRs) that are shared between human, rhesus monkey, dog, and horse and that are upstream of the promoter regions that have been previously shown to play a role in regulating CCL2 gene expression. Of these, an ECR that was ~16.5 kb (-16.5 ECR) upstream of its coding sequence contained a highly conserved NF-κB site. The region encompassing the -16.5 ECR conferred TNF-α responsiveness to homologous and heterologous promoters. In vivo footprinting demonstrated that specific nucleotide residues in the -16.5 ECR were protected or became hypersensitive after TNF-α treatment. The footprinted regions were found to bind NF-κB subunits in vitro and in vivo. Mutation/deletion of the conserved NF-κB binding site in the -16.5 ECR led to loss of TNF-α responsiveness. After TNF-α stimulation, the -16.5 ECR showed increased sensitivity to nuclease digestion and loss of histone signatures that are characteristic of a repressive chromatin. Chromosome conformation capture assays indicated that -16.5 ECR physically interacts with the CCL2 proximal promoter after TNF-α stimulation. Taken together, these results suggest that the -16.5 ECR may play a critical role in the regulation of CCL2.
Collapse
Affiliation(s)
- Grégory B Bonello
- Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | | | | | | | | | | |
Collapse
|
19
|
Liu S, Piatigorsky J. Regulation of mouse small heat shock protein αb-crystallin gene by aryl hydrocarbon receptor. PLoS One 2011; 6:e17904. [PMID: 21494593 PMCID: PMC3073930 DOI: 10.1371/journal.pone.0017904] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 02/16/2011] [Indexed: 12/16/2022] Open
Abstract
The stress-inducible small heat shock protein (shsp)/αB-crystallin gene is expressed highly in the lens and moderately in other tissues. Here we provide evidence that it is a target gene of the aryl hydrocarbon receptor (AhR) transcription factor. A sequence (−329/−323, CATGCGA) similar to the consensus xenobiotic responsive element (XRE), called here XRE-like, is present in the αBE2 region of αB-crystallin enhancer and can bind AhR in vitro and in vivo. αB-crystallin protein levels were reduced in retina, lens, cornea, heart, skeletal muscle and cultured muscle fibroblasts of AhR−/− mice; αB-crystallin mRNA levels were reduced in the eye, heart and skeletal muscle of AhR−/− mice. Increased AhR stimulated αB-crystallin expression in transfection experiments conducted in conjunction with the aryl hydrocarbon receptor nuclear translocator (ARNT) and decreased AhR reduced αB-crystallin expression. AhR effect on aB-crystallin promoter activity was cell-dependent in transfection experiments. AhR up-regulated αB-crystallin promoter activity in transfected HeLa, NIH3T3 and COS-7 cells in the absence of exogenously added ligand (TCDD), but had no effect on the αB-crystallin promoter in C2C12, CV-1 or Hepa-1 cells with or without TCDD. TCDD enhanced AhR-stimulated αB-crystallin promoter activity in transfected αTN4 cells. AhR could bind to an XRE-like site in the αB-crystallin enhancer in vitro and in vivo. Finally, site-specific mutagenesis experiments showed that the XRE-like motif was necessary for both basal and maximal AhR-induction of αB-crystallin promoter activity. Our data strongly suggest that AhR is a regulator of αB-crystallin gene expression and provide new avenues of research for the mechanism of tissue-specific αB-crystallin gene regulation under normal and physiologically stressed conditions.
Collapse
Affiliation(s)
- Shuang Liu
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Laboratory of Experimental Immunology, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
- * E-mail: (JP); (SL)
| | - Joram Piatigorsky
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JP); (SL)
| |
Collapse
|
20
|
The small heat shock protein HspB2 is a novel anti-apoptotic protein that inhibits apical caspase activation in the extrinsic apoptotic pathway. Breast Cancer Res Treat 2010; 124:307-15. [PMID: 20087649 DOI: 10.1007/s10549-010-0735-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 01/06/2010] [Indexed: 01/19/2023]
Abstract
Members of the conserved small heat shock protein (sHSP) family, such as αB-crystallin and Hsp27, are constitutively expressed in diverse malignancies and have been linked to several hallmark features of cancer including apoptosis resistance. In contrast, the sHSP HspB2/MKBP, which shares an intergenic promoter with αB-crystallin, was discovered as a chaperone of the myotonic dystrophy protein kinase and has not been previously implicated in apoptosis regulation. Here we describe a new function for HspB2 as a novel inhibitor of apical caspase activation in the extrinsic apoptotic pathway. Specifically, we demonstrate that HspB2 is expressed in a subset of human breast cancer cell lines and that ectopic expression of HspB2 in breast cancer cells confers resistance to apoptosis induced by both TRAIL and TNF-α. We also show that HspB2 inhibits the extrinsic apoptotic pathway by suppressing apical caspases-8 and 10 activation, thereby blocking downstream apoptotic events, such as Bid cleavage and caspase-3 activation. Consistent with these in vitro effects, HspB2 attenuates the anti-tumor activity of TRAIL in an orthotopic xenograft model of breast cancer. Collectively, our results reveal a novel function of HspB2 as an anti-apoptotic protein that negatively regulates apical caspase activation in the extrinsic apoptotic pathway.
Collapse
|
21
|
Bosman JD, Yehiely F, Evans JR, Cryns VL. Regulation of alphaB-crystallin gene expression by the transcription factor Ets1 in breast cancer. Breast Cancer Res Treat 2010; 119:63-70. [PMID: 19205872 PMCID: PMC2791186 DOI: 10.1007/s10549-009-0330-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Accepted: 01/20/2009] [Indexed: 11/26/2022]
Abstract
Recent studies indicate that the small heat shock protein alphaB-crystallin is expressed in poor prognosis basal-like breast tumors and likely contributes to their aggressive phenotype. However, the mechanisms underlying the deregulated expression of alphaB-crystallin in basal-like tumors are poorly understood. Using a bioinformatics approach, we identified a putative DNA binding motif in the human alphaB-crystallin promoter for the proto-oncogene Ets1, a member of the ETS transcription factor family that bind to DNA at palindromic ETS-binding sites (EBS). Here we demonstrate that ectopic expression of Ets1 activates the alphaB-crystallin promoter by an EBS-dependent mechanism and increases alphaB-crystallin protein levels, while silencing Ets1 reduces alphaB-crystallin promoter activity and protein levels. Chromatin immunoprecipitation analyses showed that endogenous Ets1 binds to the alphaB-crystallin promoter in basal-like breast cancer cells in vivo. Interrogation of publically available gene expression data revealed that Ets1 is expressed in human basal-like breast tumors and is associated with poor survival. Collectively, our results point to a previously unrecognized link between the oncogenic transcription factor Ets1 and alphaB-crystallin in basal-like breast cancer.
Collapse
Affiliation(s)
- Joshua D Bosman
- Cell Death Regulation Laboratory, Departments of Medicine and Cell and Molecular Biology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
22
|
Induction of the small heat shock protein alphaB-crystallin by genotoxic stress is mediated by p53 and p73. Breast Cancer Res Treat 2009; 122:159-68. [PMID: 19777343 DOI: 10.1007/s10549-009-0542-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 09/02/2009] [Indexed: 12/14/2022]
Abstract
The small heat shock protein alphaB-crystallin is a molecular chaperone that is induced by stress and protects cells by inhibiting protein aggregation and apoptosis. To identify novel transcriptional regulators of the alphaB-crystallin gene, we examined the alphaB-crystallin promoter for conserved transcription factor DNA-binding elements and identified a putative response element for the p53 tumor suppressor protein. Ectopic expression of wild-type p53 induced alphaB-crystallin mRNA and protein with delayed kinetics compared to p21. Additionally, the induction of alphaB-crystallin by genotoxic stress was inhibited by siRNAs targeting p53. Although the p53-dependent transactivation of an alphaB-crystallin promoter luciferase reporter required the putative p53RE, chromatin immunoprecipitation failed to detect p53 binding to the alphaB-crystallin promoter. These results suggested an indirect mechanism of transactivation involving p53 family members p63 or p73. DeltaNp73 was dramatically induced by p53 in a TAp73-dependent manner, and silencing p73 suppressed the transcriptional activation of alphaB-crystallin by p53. Moreover, ectopic expression of DeltaNp73alpha (but not other p73 isoforms) increased alphaB-crystallin mRNA levels in the absence of p53. Collectively, our results link the molecular chaperone alphaB-crystallin to the cellular genotoxic stress response via a novel mechanism of transcriptional regulation by p53 and p73.
Collapse
|
23
|
Whittaker R, Glassy MS, Gude N, Sussman MA, Gottlieb RA, Glembotski CC. Kinetics of the translocation and phosphorylation of alphaB-crystallin in mouse heart mitochondria during ex vivo ischemia. Am J Physiol Heart Circ Physiol 2009; 296:H1633-42. [PMID: 19252088 DOI: 10.1152/ajpheart.01227.2008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
alphaB-crystallin (alphaBC) is a small heat shock protein expressed at high levels in the myocardium where it protects from ischemia-reperfusion damage. Ischemia-reperfusion activates p38 MAP kinase, leading to the phosphorylation of alphaBC on serine 59 (P-alphaBC-S59), enhancing its ability to protect myocardial cells from damage. In the heart, ischemia-reperfusion also causes the translocation of alphaBC from the cytosol to other cellular locations, one of which was recently shown to be mitochondria. However, it is not known whether alphaBC translocates to mitochondria during ischemia-reperfusion, nor is it known whether alphaBC phosphorylation takes place before or after translocation. In the present study, analyses of mitochondrial fractions isolated from mouse hearts subjected to various times of ex vivo ischemia-reperfusion showed that alphaBC translocation to mitochondria was maximal after 20 min of ischemia and then declined steadily during reperfusion. Phosphorylation of mitochondrial alphaBC was maximal after 30 min of ischemia, suggesting that at least in part it occurred after alphaBC association with mitochondria. Consistent with this was the finding that translocation of activated p38 to mitochondria was maximal after only 10 min of ischemia. The overexpression of alphaBC-AAE, which mimics alphaBC phosphorylated on serine 59, has been shown to stabilize mitochondrial membrane potential and to inhibit apoptosis. In the present study, infection of neonatal rat cardiac myocytes with adenovirus-encoded alphaBC-AAE decreased peroxide-induced mitochondrial cytochrome c release. These results suggest that during ischemia alphaBC translocates to mitochondria, where it is phosphorylated and contributes to modulating mitochondrial damage upon reperfusion.
Collapse
Affiliation(s)
- R Whittaker
- SDSU Heart Institute and the Dept. of Biology, San Diego State Univ., San Diego CA 92182. )
| | | | | | | | | | | |
Collapse
|
24
|
Hu Z, Yang B, Lu W, Zhou W, Zeng L, Li T, Wang X. HSPB2/MKBP, a novel and unique member of the small heat-shock protein family. J Neurosci Res 2008; 86:2125-33. [PMID: 18615620 DOI: 10.1002/jnr.21682] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Although proteins belonging to the sHSP superfamily are diverse in sequence and size, most share characteristic features, including 1) a small molecular mass of 12-43 kDa, 2) a conserved alpha-crystallin domain of 80-100 residues, 3) formation of large oligomers, 4) a dynamic quaternary structure, and 5) induction by stress conditions and chaperone activity in suppressing protein aggregation. HSPB2/MKBP (myotonic dystrophy kinase-bind-protein) retains the structural motif of the alpha-crystallin family of HSPs but shows a unique nature compared with canonical family members, characterized by gene allocation, specific binding partners in skeletal muscle, and unique stress responsiveness. MKBP may be involved in the pathogenesis of myotonic dystrophy and contribute to the neuropathology in both Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis, Dutch type.
Collapse
Affiliation(s)
- Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Woolfe A, Elgar G. Organization of conserved elements near key developmental regulators in vertebrate genomes. ADVANCES IN GENETICS 2008; 61:307-38. [PMID: 18282512 DOI: 10.1016/s0065-2660(07)00012-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sequence conservation has traditionally been used as a means to target functional regions of complex genomes. In addition to its use in identifying coding regions of genes, the recent availability of whole genome data for a number of vertebrates has permitted high-resolution analyses of the noncoding "dark matter" of the genome. This has resulted in the identification of a large number of highly conserved sequence elements that appear to be preserved in all bony vertebrates. Further positional analysis of these conserved noncoding elements (CNEs) in the genome demonstrates that they cluster around genes involved in developmental regulation. This chapter describes the identification and characterization of these elements, with particular reference to their composition and organization.
Collapse
Affiliation(s)
- Adam Woolfe
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
| | | |
Collapse
|
26
|
Benjamin IJ, Guo Y, Srinivasan S, Boudina S, Taylor RP, Rajasekaran NS, Gottlieb R, Wawrousek EF, Abel ED, Bolli R. CRYAB and HSPB2 deficiency alters cardiac metabolism and paradoxically confers protection against myocardial ischemia in aging mice. Am J Physiol Heart Circ Physiol 2007; 293:H3201-9. [PMID: 17873008 PMCID: PMC3683980 DOI: 10.1152/ajpheart.01363.2006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The abundantly expressed small molecular weight proteins, CRYAB and HSPB2, have been implicated in cardioprotection ex vivo. However, the biological roles of CRYAB/HSPB2 coexpression for either ischemic preconditioning and/or protection in situ remain poorly defined. Wild-type (WT) and age-matched ( approximately 5-9 mo) CRYAB/HSPB2 double knockout (DKO) mice were subjected either to 30 min of coronary occlusion and 24 h of reperfusion in situ or preconditioned with a 4-min coronary occlusion/4-min reperfusion x 6, before similar ischemic challenge (ischemic preconditioning). Additionally, WT and DKO mice were subjected to 30 min of global ischemia in isolated hearts ex vivo. All experimental groups were assessed for area at risk and infarct size. Mitochondrial respiration was analyzed in isolated permeabilized cardiac skinned fibers. As a result, DKO mice modestly altered heat shock protein expression. Surprisingly, infarct size in situ was reduced by 35% in hearts of DKO compared with WT mice (38.8 +/- 17.9 vs. 59.8 +/- 10.6% area at risk, P < 0.05). In DKO mice, ischemic preconditioning was additive to its infarct-sparing phenotype. Similarly, infarct size after ischemia and reperfusion ex vivo was decreased and the production of superoxide and creatine kinase release was decreased in DKO compared with WT mice (P < 0.05). In permeabilized fibers, ADP-stimulated respiration rates were modestly reduced and calcium-dependent ATP synthesis was abrogated in DKO compared with WT mice. In conclusion, contrary to expectation, our findings demonstrate that CRYAB and HSPB2 deficiency induces profound adaptations that are related to 1) a reduction in calcium-dependent metabolism/respiration, including ATP production, and 2) decreased superoxide production during reperfusion. We discuss the implications of these disparate results in the context of phenotypic responses reported for CRYAB/HSPB2-deficient mice to different ischemic challenges.
Collapse
Affiliation(s)
- Ivor J Benjamin
- Center for Cardiovascular Translational Biomedicine, University of Utah, School of Medicine, Salt Lake City, UT, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ward RD, Davis SW, Cho M, Esposito C, Lyons RH, Cheng JF, Rubin EM, Rhodes SJ, Raetzman LT, Smith TPL, Camper SA. Comparative genomics reveals functional transcriptional control sequences in the Prop1 gene. Mamm Genome 2007; 18:521-37. [PMID: 17557180 PMCID: PMC1998882 DOI: 10.1007/s00335-007-9008-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Accepted: 01/26/2007] [Indexed: 11/30/2022]
Abstract
Mutations in PROP1 are a common genetic cause of multiple pituitary hormone deficiency (MPHD). We used a comparative genomics approach to predict the transcriptional regulatory domains of Prop1 and tested them in cell culture and mice. A BAC transgene containing Prop1 completely rescues the Prop1 mutant phenotype, demonstrating that the regulatory elements necessary for proper PROP1 transcription are contained within the BAC. We generated DNA sequences from the PROP1 genes in lemur, pig, and five different primate species. Comparison of these with available human and mouse PROP1 sequences identified three putative regulatory sequences that are highly conserved. These are located in the PROP1 promoter proximal region, within the first intron of PROP1, and downstream of PROP1. Each of the conserved elements elicited orientation-specific enhancer activity in the context of the Drosophila alcohol dehydrogenase minimal promoter in both heterologous and pituitary-derived cells lines. The intronic element is sufficient to confer dorsal expansion of the pituitary expression domain of a transgene, suggesting that this element is important for the normal spatial expression of endogenous Prop1 during pituitary development. This study illustrates the usefulness of a comparative genomics approach in the identification of regulatory elements that may be the site of mutations responsible for some cases of MPHD.
Collapse
Affiliation(s)
- Robert D. Ward
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan USA
- Present Address: Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas USA
| | - Shannon W. Davis
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan USA
| | - MinChul Cho
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan USA
| | - Constance Esposito
- DNA Sequencing Core Facility, University of Michigan, Ann Arbor, Michigan USA
| | - Robert H. Lyons
- DNA Sequencing Core Facility, University of Michigan, Ann Arbor, Michigan USA
| | - Jan-Fang Cheng
- DOE Joint Genome Institute, U.S. Department of Energy, Walnut Creek, California, USA
| | - Edward M. Rubin
- DOE Joint Genome Institute, U.S. Department of Energy, Walnut Creek, California, USA
| | - Simon J. Rhodes
- Department of Biology, Indiana University-Purdue University Indianapolis (IUPUI), Indianapolis, Indiana USA
| | - Lori T. Raetzman
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan USA
- Present Address: Department Molecular and Integrative Physiology, University of Illinois, Urbana, Illinois USA
| | - Timothy P. L. Smith
- U.S. Meat Animal Research Center, U.S. Department of Agriculture, Agricultural Research Service (USDA/ARS), Clay Center, Nebraska, USA
| | - Sally A. Camper
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan USA
- 4909 Buhl Bldg., 1241 E. Catherine St., Ann Arbor, MI 48109-0618 USA
| |
Collapse
|
28
|
Li Y, Hough RB, Piatigorsky J. Tissue-specific activity of the blind mole rat and the two nucleotide-mutated mouse alphaB-crystallin promoter in transgenic mice. Proc Natl Acad Sci U S A 2007; 104:2608-13. [PMID: 17293452 PMCID: PMC1796782 DOI: 10.1073/pnas.0611684104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The alphaB-crystallin and HspB2 genes are located approximately 0.9 kb apart in a head-to-head arrangement in mammals. Previous experiments have shown that a truncated -668/+45 alphaB-crystallin enhancer/promoter fragment from blind mole rats (Spalax ehrenbergi), which have nonfunctional lenses, lacks lens activity and has enhanced muscle activity in transgenic mice. Here we show that the full-length mole rat alphaB-crystallin intergenic region behaves similarly in transgenic mice. A two-nucleotide mutation ((-273)CA-->G) in the mouse alphaB-crystallin enhancer/promoter fragment mimicking the wild-type mole rat sequence functionally converted the mouse promoter fragment to that of the wild-type mole rat promoter when tested in transgenic mice. The reciprocal mutation in the mole rat promoter fragment ((-272)G-->CA) did not affect its activity. Oligonucleotides from the wild-type mouse and mole rat alphaB-crystallin promoter region under study formed distinct complexes with nuclear proteins from cultured cells. The mouse mutant sequence lost binding ability, whereas the mutated mole rat sequence gained the ability to form a complex similar in size to that of the wild-type mouse oligonucleotide. Our data support the idea that blind mole rats' alphaB-crystallin promoter activity was modified during the evolution of subterranean life and shows that tissue-specific promoter activity can be modulated by changing as few as two apparently neutral nucleotides in the mouse alphaB-crystallin enhancer region, implying the importance of the context of regulatory sequences for promoter activity.
Collapse
Affiliation(s)
- Yan Li
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-0704
| | - R. Barry Hough
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-0704
| | - Joram Piatigorsky
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-0704
- *To whom correspondence should be addressed at: Laboratory of Molecular and Developmental Biology, 7 Memorial Drive/Building 7, Room 100, Bethesda, MD 20892-0704. E-mail:
| |
Collapse
|
29
|
Chen PY, Chang WSW, Chou RH, Lai YK, Lin SC, Chi CY, Wu CW. Two non-homologous brain diseases-related genes, SERPINI1 and PDCD10, are tightly linked by an asymmetric bidirectional promoter in an evolutionarily conserved manner. BMC Mol Biol 2007; 8:2. [PMID: 17212813 PMCID: PMC1796892 DOI: 10.1186/1471-2199-8-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Accepted: 01/09/2007] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Despite of the fact that mammalian genomes are far more spacious than prokaryotic genomes, recent nucleotide sequencing data have revealed that many mammalian genes are arranged in a head-to-head orientation and separated by a small intergenic sequence. Extensive studies on some of these neighboring genes, in particular homologous gene pairs, have shown that these genes are often co-expressed in a symmetric manner and regulated by a shared promoter region. Here we report the identification of two non-homologous brain disease-related genes, with one coding for a serine protease inhibitor (SERPINI1) and the other for a programmed cell death-related gene (PDCD10), being tightly linked together by an asymmetric bidirectional promoter in an evolutionarily conserved fashion. This asymmetric bidirectional promoter, in cooperation with some cis-acting elements, is responsible for the co-regulation of the gene expression pattern as well as the tissue specificity of SERPINI1 and PDCD10. RESULTS While SERPINI1 is predominantly expressed in normal brain and down-regulated in brain tumors, PDCD10 is ubiquitously expressed in all normal tissues but its gene transcription becomes aberrant in different types of cancers. By measuring the luciferase activity in various cell lysates, their 851-bp intergenic sequence was shown to be capable of driving the reporter gene expression in either direction. A 175-bp fragment from nt 1 to 175 in the vicinity of PDCD10 was further determined to function as a minimal bidirectional promoter. A critical regulatory fragment, from nt 176-473 outside the minimal promoter in the intergenic region, was identified to contain a strong repressive element for SERPINI1 and an enhancer for PDCD10. These cis-acting elements may exist to help coordinate the expression and regulation of the two flanking genes. CONCLUSION For all non-homologous genes that have been described to be closely adjacent in the mammalian genomes, the intergenic region of the head-to-head PDCD10-SERPINI1 gene pair provides an interesting and informative example of a complex regulatory system that governs the expression of both genes not only through an asymmetric bidirectional promoter, but also through fine-tuned regulations with some cis-acting elements.
Collapse
Affiliation(s)
- Ping-Yen Chen
- President's Laboratory and Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan, ROC
- Department of Life Sciences, National Tsing Hua University, Hsinchu City 300, Taiwan, ROC
| | - Wun-Shaing W Chang
- President's Laboratory and Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Ruey-Hwang Chou
- President's Laboratory and Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Yiu-Kay Lai
- Department of Life Sciences, National Tsing Hua University, Hsinchu City 300, Taiwan, ROC
- Department of Bioresources, Da-Yeh University, Changhua County 515, Taiwan, ROC
| | - Sheng-Chieh Lin
- President's Laboratory and Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Chia-Yi Chi
- President's Laboratory and Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Cheng-Wen Wu
- President's Laboratory and Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan, ROC
| |
Collapse
|
30
|
McEwen GK, Woolfe A, Goode D, Vavouri T, Callaway H, Elgar G. Ancient duplicated conserved noncoding elements in vertebrates: a genomic and functional analysis. Genome Res 2006; 16:451-65. [PMID: 16533910 PMCID: PMC1457030 DOI: 10.1101/gr.4143406] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Accepted: 01/03/2006] [Indexed: 12/25/2022]
Abstract
Fish-mammal genomic comparisons have proved powerful in identifying conserved noncoding elements likely to be cis-regulatory in nature, and the majority of those tested in vivo have been shown to act as tissue-specific enhancers associated with genes involved in transcriptional regulation of development. Although most of these elements share little sequence identity to each other, a small number are remarkably similar and appear to be the product of duplication events. Here, we searched for duplicated conserved noncoding elements in the human genome, using comparisons with Fugu to select putative cis-regulatory sequences. We identified 124 families of duplicated elements, each containing between two and five members, that are highly conserved within and between vertebrate genomes. In 74% of cases, we were able to assign a specific set of paralogous genes with annotation relating to transcriptional regulation and/or development to each family, thus removing much of the ambiguity in identifying associated genes. We find that duplicate elements have the potential to up-regulate reporter gene expression in a tissue-specific manner and that expression domains often overlap, but are not necessarily identical, between family members. Over two thirds of the families are conserved in duplicate in fish and appear to predate the large-scale duplication events thought to have occurred at the origin of vertebrates. We propose a model whereby gene duplication and the evolution of cis-regulatory elements can be considered in the context of increased morphological diversity and the emergence of the modern vertebrate body plan.
Collapse
Affiliation(s)
- Gayle K. McEwen
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SB, United Kingdom
- MRC Biostatistics Unit, Institute of Public Health, Cambridge CB2 2SR, United Kingdom
| | - Adam Woolfe
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SB, United Kingdom
| | - Debbie Goode
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
| | - Tanya Vavouri
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SB, United Kingdom
| | - Heather Callaway
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
| | - Greg Elgar
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom
| |
Collapse
|
31
|
Cvekl A, Yang Y, Chauhan BK, Cveklova K. Regulation of gene expression by Pax6 in ocular cells: a case of tissue-preferred expression of crystallins in lens. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2005; 48:829-44. [PMID: 15558475 PMCID: PMC2080872 DOI: 10.1387/ijdb.041866ac] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Lens development is an excellent model for genetic and biochemical studies of embryonic induction, cell cycle regulation, cellular differentiation and signal transduction. Differentiation of lens is characterized by lens-preferred expression and accumulation of water-soluble proteins, crystallins. Crystallins are required for light transparency, refraction and maintenance of lens integrity. Here, we review mechanisms of lens-preferred expression of crystallin genes by employing synergism between developmentally regulated DNA-binding transcription factors: Pax6, c-Maf, MafA/L-Maf, MafB, NRL, Sox2, Sox1, RARbeta/RXRbeta, RORalpha, Prox1, Six3, gammaFBP-B and HSF2. These factors are differentially expressed in lens precursor cells, lens epithelium and primary and secondary lens fibers. They exert their function in combination with ubiquitously expressed factors (e.g. AP-1, CREB, pRb, TFIID and USF) and co-activators/chromatin remodeling proteins (e.g. ASC-2 and CBP/p300). A special function belongs to Pax6, a paired domain and homeodomain-containing protein, which is essential for lens formation. Pax6 is expressed in lens progenitor cells before the onset of crystallin expression and it serves as an important regulatory factor required for expression of c-Maf, MafA/L-Maf, Six3, Prox1 and retinoic acid signaling both in lens precursor cells and the developing lens. The roles of these factors are illustrated by promoter studies of mouse alphaA-, alphaB-, gammaF- and guinea pig zeta-crystallins. Pax6 forms functional complexes with a number of transcription factors including the retinoblastoma protein, pRb, MafA, Mitf and Sox2. We present novel data showing that pRb antagonizes Pax6-mediated activation of the alphaA-crystallin promoter likely by inhibiting binding of Pax6 to DNA.
Collapse
Affiliation(s)
- Ales Cvekl
- The Department of Ophthalmology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | | | |
Collapse
|
32
|
Pham DQD, Douglass PL, Chavez CA, Shaffer JJ. Regulation of the ferritin heavy-chain homologue gene in the yellow fever mosquito, Aedes aegypti. INSECT MOLECULAR BIOLOGY 2005; 14:223-36. [PMID: 15926891 DOI: 10.1111/j.1365-2583.2004.00550.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
In the yellow fever mosquito Aedes aegypti, the ferritin heavy-chain homologue (HCH) gene is induced by blood feeding. This suggests that ferritin may serve as a cytotoxic protector against the oxidative challenge of the blood meal and may be essential for the survival of the insect. In this study, various cis-acting elements for the gene were identified and mapped. Transfection assays showed that the strength and activity of a subset of these elements are orientation-dependent. The shift observed for the ferritin HCH cis-acting elements is unique among known ferritin genes. DNase I footprinting data together with Transfac analyses identified a number of putative sites known for their involvement in developmental and cell proliferation processes.
Collapse
Affiliation(s)
- D Q-D Pham
- Department of Biological Sciences, University of Wisconsin-Parkside, Kenosha, WI 53141, USA.
| | | | | | | |
Collapse
|
33
|
Doerwald L, van Rheede T, Dirks RP, Madsen O, Rexwinkel R, van Genesen ST, Martens GJ, de Jong WW, Lubsen NH. Sequence and Functional Conservation of the Intergenic Region Between the Head-to-Head Genes Encoding the Small Heat Shock Proteins αB-Crystallin and HspB2 in the Mammalian Lineage. J Mol Evol 2004; 59:674-86. [PMID: 15693623 DOI: 10.1007/s00239-004-2659-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An unexpected feature of the large mammalian genome is the frequent occurrence of closely linked head-to-head gene pairs. Close apposition of such gene pairs has been suggested to be due to sharing of regulatory elements. We show here that the head-to-head gene pair encoding two small heat shock proteins, alphaB-crystallin and HspB2, is closely linked in all major mammalian clades, suggesting that this close linkage is of selective advantage. Yet alphaB-crystallin is abundantly expressed in lens and muscle and in response to a heat shock, while HspB2 is abundant only in muscle and not upregulated by a heat shock. The intergenic distance between the genes for these two proteins in mammals ranges from 645 bp (platypus) to 1069 bp (opossum), with an average of about 900 bp; in chicken the distance was the same as in duck (1.6 kb). Phylogenetic footprinting and sequence alignment identified a number of conserved sequence elements close to the HspB2 promoter and two farther upstream. All known regulatory elements of the mouse alphaB-crystallin promoter are conserved, except in platypus and birds. The lens-specific region 1 (LSR1) and the heat shock elements (HSEs) lack in birds; in platypus the LSR1 is reduced to a Pax-6 site, while the Pax-6 site in LSR2 and a HSE are absent. Most likely the primordial mammalian alphaB-crystallin promoter had two LSRs and two HSEs. In transfection experiments the platypus alphaB-crystallin promoter retained heat shock responsiveness and lens expression. It also directed lens expression in Xenopus laevis transgenes, as did the HspB2 promoter of rat or blind mole rat. Deletion of the middle of the intergenic region including the upstream enhancer affected the activity of both the rat alphaB-crystallin and the HspB2 promoters, suggesting sharing of the enhancer region by the two promoters.
Collapse
Affiliation(s)
- Linda Doerwald
- Department of Biochemistry, Faculty of Science, University of Nijmegen, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|