51
|
Gajan A, Barnes VL, Liu M, Saha N, Pile LA. The histone demethylase dKDM5/LID interacts with the SIN3 histone deacetylase complex and shares functional similarities with SIN3. Epigenetics Chromatin 2016; 9:4. [PMID: 26848313 PMCID: PMC4740996 DOI: 10.1186/s13072-016-0053-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 01/14/2016] [Indexed: 01/01/2023] Open
Abstract
Background Regulation of gene expression by histone-modifying enzymes is essential to control cell fate decisions and developmental processes. Two histone-modifying enzymes, RPD3, a deacetylase, and dKDM5/LID, a demethylase, are present in a single complex, coordinated through the SIN3 scaffold protein. While the SIN3 complex has been demonstrated to have functional histone deacetylase activity, the role of the demethylase dKDM5/LID as part of the complex has not been investigated. Results Here, we analyzed the developmental and transcriptional activities of dKDM5/LID in relation to SIN3. Knockdown of either Sin3A or lid resulted in decreased cell proliferation in S2 cells and wing imaginal discs. Conditional knockdown of either Sin3A or lid resulted in flies that displayed wing developmental defects. Interestingly, overexpression of dKDM5/LID rescued the wing developmental defect due to reduced levels of SIN3 in female flies, indicating a major role for dKDM5/LID in cooperation with SIN3 during development. Together, these observed phenotypes strongly suggest that dKDM5/LID as part of the SIN3 complex can impact previously uncharacterized transcriptional networks. Transcriptome analysis revealed that SIN3 and dKDM5/LID regulate many common genes. While several genes implicated in cell cycle and wing developmental pathways were affected upon altering the level of these chromatin factors, a significant affect was also observed on genes required to mount an effective stress response. Further, under conditions of induced oxidative stress, reduction of SIN3 and/or dKDM5/LID altered the expression of a greater number of genes involved in cell cycle-related processes relative to normal conditions. This highlights an important role for SIN3 and dKDM5/LID proteins to maintain proper progression through the cell cycle in environments of cellular stress. Further, we find that target genes are bound by both SIN3 and dKDM5/LID, however, histone acetylation, not methylation, plays a predominant role in gene regulation by the SIN3 complex. Conclusions We have provided genetic evidence to demonstrate functional cooperation between the histone demethylase dKDM5/LID and SIN3. Biochemical and transcriptome data further support functional links between these proteins. Together, the data provide a solid framework for analyzing the gene regulatory pathways through which SIN3 and dKDM5/LID control diverse biological processes in the organism. Electronic supplementary material The online version of this article (doi:10.1186/s13072-016-0053-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ambikai Gajan
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| | - Valerie L Barnes
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| | - Mengying Liu
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| | - Nirmalya Saha
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| | - Lori A Pile
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| |
Collapse
|
52
|
Varier RA, Carrillo de Santa Pau E, van der Groep P, Lindeboom RGH, Matarese F, Mensinga A, Smits AH, Edupuganti RR, Baltissen MP, Jansen PWTC, Ter Hoeve N, van Weely DR, Poser I, van Diest PJ, Stunnenberg HG, Vermeulen M. Recruitment of the Mammalian Histone-modifying EMSY Complex to Target Genes Is Regulated by ZNF131. J Biol Chem 2016; 291:7313-24. [PMID: 26841866 DOI: 10.1074/jbc.m115.701227] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Indexed: 11/06/2022] Open
Abstract
Recent work from others and us revealed interactions between the Sin3/HDAC complex, the H3K4me3 demethylase KDM5A, GATAD1, and EMSY. Here, we characterize the EMSY/KDM5A/SIN3B complex in detail by quantitative interaction proteomics and ChIP-sequencing. We identify a novel substoichiometric interactor of the complex, transcription factor ZNF131, which recruits EMSY to a large number of active, H3K4me3 marked promoters. Interestingly, using an EMSY knock-out line and subsequent rescue experiments, we show that EMSY is in most cases positively correlated with transcriptional activity of its target genes and stimulates cell proliferation. Finally, by immunohistochemical staining of primary breast tissue microarrays we find that EMSY/KDM5A/SIN3B complex subunits are frequently overexpressed in primary breast cancer cases in a correlative manner. Taken together, these data open venues for exploring the possibility that sporadic breast cancer patients with EMSY amplification might benefit from epigenetic combination therapy targeting both the KDM5A demethylase and histone deacetylases.
Collapse
Affiliation(s)
- Radhika A Varier
- From the Department of Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584CG Utrecht, The Netherlands
| | - Enrique Carrillo de Santa Pau
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands
| | - Petra van der Groep
- Department of Pathology, University Medical Center Utrecht Heidelberglaan 100, 3584CX Utrecht, The Netherlands, and
| | - Rik G H Lindeboom
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands
| | - Filomena Matarese
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands
| | - Anneloes Mensinga
- From the Department of Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584CG Utrecht, The Netherlands
| | - Arne H Smits
- From the Department of Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584CG Utrecht, The Netherlands, Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands
| | - Raghu Ram Edupuganti
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands
| | - Marijke P Baltissen
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands
| | - Pascal W T C Jansen
- From the Department of Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584CG Utrecht, The Netherlands, Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands
| | - Natalie Ter Hoeve
- Department of Pathology, University Medical Center Utrecht Heidelberglaan 100, 3584CX Utrecht, The Netherlands, and
| | - Danny R van Weely
- From the Department of Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584CG Utrecht, The Netherlands
| | - Ina Poser
- Max Planck Institute of Molecular Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht Heidelberglaan 100, 3584CX Utrecht, The Netherlands, and
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands
| | - Michiel Vermeulen
- From the Department of Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584CG Utrecht, The Netherlands, Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 30, 6525GA Nijmegen, The Netherlands,
| |
Collapse
|
53
|
Muñoz-Soriano V, Santos D, Durupt FC, Casani S, Paricio N. Scabrous overexpression in the eye affects R3/R4 cell fate specification and inhibits notch signaling. Dev Dyn 2015; 245:166-74. [PMID: 26505171 DOI: 10.1002/dvdy.24362] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 10/07/2015] [Accepted: 10/23/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Planar cell polarity (PCP) in the Drosophila eye is generated when immature ommatidial preclusters acquire opposite chirality in the dorsal and ventral halves of the eye imaginal disc and rotate 90 ° toward the equator. The scabrous (sca) gene is involved in R8 differentiation and in the correct spacing of ommatidial clusters in eye imaginal discs, but it was also suggested to be required during ommatidial rotation. However, no clear relationships between sca and other genes involved in the process were established. RESULTS To explore the role of Sca in PCP establishment, we performed an RNAi-based modifier genetic screen using the rough eye phenotype of sca-overexpressing flies. We found that sca overexpression mainly affects R3/R4 cell specification as it was reported in Notch mutants. Of the 86 modifiers identified in the screen, genes encoding components of Notch signaling and proteins involved in intracellular transport were of particular interest. CONCLUSIONS These and other results obtained with a reporter line of Notch activity indicate that sca overexpression antagonizes Notch signaling in the Drosophila eye, and are inconsistent with Sca being an ommatidial rotation-specific factor. We also found that microtubule motors and other proteins involved in intracellular transport are related with Sca function.
Collapse
Affiliation(s)
- Verónica Muñoz-Soriano
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, Burjasot, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, Burjasot, Spain
| | - Diego Santos
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, Burjasot, Spain
| | - Fabrice C Durupt
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, Burjasot, Spain
| | - Sandra Casani
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, Burjasot, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, Burjasot, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, Burjasot, Spain
| |
Collapse
|
54
|
Ellis K, Friedman C, Yedvobnick B. Drosophila domino Exhibits Genetic Interactions with a Wide Spectrum of Chromatin Protein-Encoding Loci. PLoS One 2015; 10:e0142635. [PMID: 26555684 PMCID: PMC4640824 DOI: 10.1371/journal.pone.0142635] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/23/2015] [Indexed: 11/18/2022] Open
Abstract
The Drosophila domino gene encodes protein of the SWI2/SNF2 family that has widespread roles in transcription, replication, recombination and DNA repair. Here, the potential relationship of Domino protein to other chromatin-associated proteins has been investigated through a genetic interaction analysis. We scored for genetic modification of a domino wing margin phenotype through coexpression of RNAi directed against a set of previously characterized and more newly characterized chromatin-encoding loci. A set of other SWI2/SNF2 loci were also assayed for interaction with domino. Our results show that the majority of tested loci exhibit synergistic enhancement or suppression of the domino wing phenotype. Therefore, depression in domino function sensitizes the wing margin to alterations in the activity of numerous chromatin components. In several cases the genetic interactions are associated with changes in the level of cell death measured across the dorsal-ventral margin of the wing imaginal disc. These results highlight the broad realms of action of many chromatin proteins and suggest significant overlap with Domino function in fundamental cell processes, including cell proliferation, cell death and cell signaling.
Collapse
Affiliation(s)
- Kaitlyn Ellis
- Biology Department, Emory University, Atlanta, Georgia, United States of America
| | - Chloe Friedman
- Biology Department, Emory University, Atlanta, Georgia, United States of America
| | - Barry Yedvobnick
- Biology Department, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
55
|
Disruption of Methionine Metabolism in Drosophila melanogaster Impacts Histone Methylation and Results in Loss of Viability. G3-GENES GENOMES GENETICS 2015; 6:121-32. [PMID: 26546310 PMCID: PMC4704710 DOI: 10.1534/g3.115.024273] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Histone methylation levels, which are determined by the action of both histone demethylases and methyltransferases, impact multiple biological processes by affecting gene expression activity. Methionine metabolism generates the major methyl donor S-adenosylmethionine (SAM) for histone methylation. The functions of methionine metabolic enzymes in regulating biological processes as well as the interaction between the methionine pathway and histone methylation, however, are still not fully understood. Here, we report that reduced levels of some enzymes involved in methionine metabolism and histone demethylases lead to lethality as well as wing development and cell proliferation defects in Drosophila melanogaster. Additionally, disruption of methionine metabolism can directly affect histone methylation levels. Reduction of little imaginal discs (LID) histone demethylase, but not lysine-specific demethylase 2 (KDM2) demethylase, is able to counter the effects on histone methylation due to reduction of SAM synthetase (SAM-S). Taken together, these results reveal an essential role of key enzymes that control methionine metabolism and histone methylation. Additionally, these findings are an indication of a strong connection between metabolism and epigenetics.
Collapse
|
56
|
Doyen CM, Chalkley GE, Voets O, Bezstarosti K, Demmers JA, Moshkin YM, Verrijzer CP. A Testis-Specific Chaperone and the Chromatin Remodeler ISWI Mediate Repackaging of the Paternal Genome. Cell Rep 2015; 13:1310-1318. [PMID: 26549447 DOI: 10.1016/j.celrep.2015.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 08/12/2015] [Accepted: 10/03/2015] [Indexed: 11/19/2022] Open
Abstract
During spermatogenesis, the paternal genome is repackaged into a non-nucleosomal, highly compacted chromatin structure. Bioinformatic analysis revealed that Drosophila sperm chromatin proteins are characterized by a motif related to the high-mobility group (HMG) box, which we termed male-specific transcript (MST)-HMG box. MST77F is a MST-HMG-box protein that forms an essential component of sperm chromatin. The deposition of MST77F onto the paternal genome requires the chaperone function of tNAP, a testis-specific NAP protein. MST77F, in turn, enables the stable incorporation of MST35Ba and MST35Bb into sperm chromatin. Following MST-HMG-box protein deposition, the ATP-dependent chromatin remodeler ISWI mediates the appropriate organization of sperm chromatin. Conversely, at fertilization, maternal ISWI targets the paternal genome and drives its repackaging into de-condensed nucleosomal chromatin. Failure of this transition in ISWI mutant embryos is followed by mitotic defects, aneuploidy, and haploid embryonic divisions. Thus, ISWI enables bi-directional transitions between two fundamentally different forms of chromatin.
Collapse
Affiliation(s)
- Cécile M Doyen
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands
| | - Gillian E Chalkley
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands
| | - Olaf Voets
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands
| | - Karel Bezstarosti
- Proteomics Centre, Erasmus University Medical Center, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands
| | - Jeroen A Demmers
- Proteomics Centre, Erasmus University Medical Center, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands
| | - Yuri M Moshkin
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands
| | - C Peter Verrijzer
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands.
| |
Collapse
|
57
|
The Drosophila histone demethylase dKDM5/LID regulates hematopoietic development. Dev Biol 2015; 405:260-8. [DOI: 10.1016/j.ydbio.2015.07.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 06/12/2015] [Accepted: 07/12/2015] [Indexed: 01/08/2023]
|
58
|
Barnes VL, Bhat A, Unnikrishnan A, Heydari AR, Arking R, Pile LA. SIN3 is critical for stress resistance and modulates adult lifespan. Aging (Albany NY) 2015; 6:645-60. [PMID: 25133314 PMCID: PMC4169859 DOI: 10.18632/aging.100684] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Coordinate control of gene activity is critical for fitness and longevity of an organism. The SIN3 histone deacetylase (HDAC) complex functions as a transcriptional repressor of many genes. SIN3-regulated genes include those that encode proteins affecting multiple aspects of mitochondrial function, such as energy production and stress responsiveness, important for health maintenance. Here we used Drosophila melanogaster as a model organism to examine the role of SIN3 in the regulation of fitness and longevity. Adult flies with RNA interference (RNAi) induced knockdown expression of Sin3A have reduced climbing ability; an activity that likely requires fully functional mitochondria. Additionally, compared to wild type, adult Sin3A knockdown flies were more sensitive to oxidative stress. Interestingly, media supplementation with the antioxidant glutathione largely restored fly tolerance to oxidative stress. Although Sin3A knockdown flies exhibited decreased longevity compared to wild type, no significant changes in expression of many well-categorized aging genes were observed. We found, however, that Sin3A knockdown corresponded to a significant reduction in expression of genes encoding proteins involved in the de novo synthesis of glutathione. Taken together, the data support a model whereby SIN3 regulates a gene expression program required for proper mitochondrial function and effective stress response during adulthood.
Collapse
Affiliation(s)
- Valerie L Barnes
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, 48202, USA
| | - Abhineeth Bhat
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, 48202, USA
| | - Archana Unnikrishnan
- Department of Nutrition and Food Science Wayne State University, Detroit, Michigan, 48202,USA
| | - Ahmad R Heydari
- Department of Nutrition and Food Science Wayne State University, Detroit, Michigan, 48202,USA
| | - Robert Arking
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, 48202, USA
| | - Lori A Pile
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, 48202, USA
| |
Collapse
|
59
|
Schwanbeck R. The role of epigenetic mechanisms in Notch signaling during development. J Cell Physiol 2015; 230:969-81. [PMID: 25336183 DOI: 10.1002/jcp.24851] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 10/16/2014] [Indexed: 12/16/2022]
Abstract
The Notch pathway is a highly conserved cell-cell communication pathway in metazoan involved in numerous processes during embryogenesis, development, and adult organisms. Ligand-receptor interaction of Notch components on adjacent cells facilitates controlled sequential proteolytic cleavage resulting in the nuclear translocation of the intracellular domain of Notch (NICD). There it binds to the Notch effector protein RBP-J, displaces a corepressor complex and enables the induction of target genes by recruitment of coactivators in a cell-context dependent manner. Both, the gene-specific repression and the context dependent activation require an intense communication with the underlying chromatin of the regulatory regions. Since the epigenetic landscape determines the function of the genome, processes like cell fate decision, differentiation, and self-renewal depend on chromatin structure and its remodeling during development. In this review, structural features enabling the Notch pathway to read these epigenetic marks by proteins interacting with RBP-J/Notch will be discussed. Furthermore, mechanisms of the Notch pathway to write and erase chromatin marks like histone acetylation and methylation are depicted as well as ATP-dependent chromatin remodeling during the activation of target genes. An additional fine-tuning of transcriptional regulation upon Notch activation seems to be controlled by the commitment of miRNAs. Since cells within an organism have to react to environmental changes, and developmental and differentiation cues in a proper manner, different signaling pathways have to crosstalk to each other. The chromatin status may represent one major platform to integrate these different pathways including the canonical Notch signaling.
Collapse
Affiliation(s)
- Ralf Schwanbeck
- Institute of Biochemistry, Medical Faculty, University of Kiel, Kiel, Germany
| |
Collapse
|
60
|
Yu Z, Liu J, Deng WM, Jiao R. Histone chaperone CAF-1: essential roles in multi-cellular organism development. Cell Mol Life Sci 2015; 72:327-37. [PMID: 25292338 PMCID: PMC11114026 DOI: 10.1007/s00018-014-1748-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/16/2014] [Accepted: 09/29/2014] [Indexed: 01/01/2023]
Abstract
More and more studies have shown chromatin remodelers and histone modifiers play essential roles in regulating developmental patterns by organizing specific chromosomal architecture to establish programmed transcriptional profiles, with implications that histone chaperones execute a coordinating role in these processes. Chromatin assembly factor-1 (CAF-1), an evolutionarily conserved three-subunit protein complex, was identified as a histone chaperone coupled with DNA replication and repair in cultured mammalian cells and yeasts. Interestingly, recent findings indicate CAF-1 may have important regulatory roles during development by interacting with specific transcription factors and epigenetic regulators. In this review, we focus on the essential roles of CAF-1 in regulating heterochromatin organization, asymmetric cell division, and specific signal transduction through epigenetic modulations of the chromatin. In the end, we aim at providing a current image of facets of CAF-1 as a histone chaperone to orchestrate cell proliferation and differentiation during multi-cellular organism development.
Collapse
Affiliation(s)
- Zhongsheng Yu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, The Chinese Academy of Sciences, Datun Road 15, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100080 China
| | - Jiyong Liu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, The Chinese Academy of Sciences, Datun Road 15, Beijing, 100101 China
- Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Dongfengxi Road 195, Guangzhou, 510182 China
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295 USA
| | - Renjie Jiao
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, The Chinese Academy of Sciences, Datun Road 15, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100080 China
- Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Dongfengxi Road 195, Guangzhou, 510182 China
| |
Collapse
|
61
|
EMSY promoted the growth and migration of ovarian cancer cells. Tumour Biol 2014; 36:3085-92. [DOI: 10.1007/s13277-014-2944-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/04/2014] [Indexed: 01/08/2023] Open
|
62
|
Auer JS, Nagel AC, Schulz A, Wahl V, Preiss A. MAPK-dependent phosphorylation modulates the activity of Suppressor of Hairless in Drosophila. Cell Signal 2014; 27:115-24. [PMID: 25452105 DOI: 10.1016/j.cellsig.2014.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 10/16/2014] [Indexed: 02/08/2023]
Abstract
Cell differentiation strictly depends on the epidermal growth factor receptor (EGFR)- and Notch-signalling pathways, which are closely intertwined. Here we address the molecular cross talk at the level of Suppressor of Hairless [Su(H)]. The Drosophila transcription factor Su(H) mediates Notch signalling at the DNA level: in the presence of signalling input Su(H) assembles an activator complex on Notch target genes and a repressor complex in its absence. Su(H) contains a highly conserved mitogen activated protein kinase (MAPK) target sequence. Here we provide evidence that Su(H) is phosphorylated in response to MAPK activity. Mutation of the Su(H) MAPK-site modulated the Notch signalling output: whereas a phospho-deficient Su(H)(MAPK-ko) isoform provoked a stronger Notch signalling activity, a phospho-mimetic Su(H)(MAPK-ac) mutant resulted in its attenuation. In vivo assays in Drosophila cell culture as well as in flies support the idea that Su(H) phosphorylation affects the dynamics of repressor or activator complex formation or the transition from the one into the other complex. In summary, the phosphorylation of Su(H) attenuates Notch signalling in vivo in several developmental settings. Consequently, a decrease of EGFR signal causes an increase of Notch signalling intensity. Hence, the antagonistic relationship between EGFR- and Notch-signalling pathways may involve a direct modification of Su(H) by MAPK in several developmental contexts of fly development. The high sequence conservation of the MAPK target site in the mammalian Su(H) homologues supports the idea that EGFR signalling impacts on Notch activity in a similar way in humans as well.
Collapse
Affiliation(s)
- Jasmin S Auer
- Institut für Genetik (240A), Universität Hohenheim, Garbenstr. 30 Stuttgart, 70599, Germany
| | - Anja C Nagel
- Institut für Genetik (240A), Universität Hohenheim, Garbenstr. 30 Stuttgart, 70599, Germany
| | - Adriana Schulz
- Institut für Genetik (240A), Universität Hohenheim, Garbenstr. 30 Stuttgart, 70599, Germany
| | - Vanessa Wahl
- Institut für Genetik (240A), Universität Hohenheim, Garbenstr. 30 Stuttgart, 70599, Germany
| | - Anette Preiss
- Institut für Genetik (240A), Universität Hohenheim, Garbenstr. 30 Stuttgart, 70599, Germany.
| |
Collapse
|
63
|
Nishibuchi G, Shibata Y, Hayakawa T, Hayakawa N, Ohtani Y, Sinmyozu K, Tagami H, Nakayama JI. Physical and functional interactions between the histone H3K4 demethylase KDM5A and the nucleosome remodeling and deacetylase (NuRD) complex. J Biol Chem 2014; 289:28956-70. [PMID: 25190814 DOI: 10.1074/jbc.m114.573725] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Histone H3K4 methylation has been linked to transcriptional activation. KDM5A (also known as RBP2 or JARID1A), a member of the KDM5 protein family, is an H3K4 demethylase, previously implicated in the regulation of transcription and differentiation. Here, we show that KDM5A is physically and functionally associated with two histone deacetylase complexes. Immunoaffinity purification of KDM5A confirmed a previously described association with the SIN3B-containing histone deacetylase complex and revealed an association with the nucleosome remodeling and deacetylase (NuRD) complex. Sucrose density gradient and sequential immunoprecipitation analyses further confirmed the stable association of KDM5A with these two histone deacetylase complexes. KDM5A depletion led to changes in the expression of hundreds of genes, two-thirds of which were also controlled by CHD4, the NuRD catalytic subunit. Gene ontology analysis confirmed that the genes commonly regulated by both KDM5A and CHD4 were categorized as developmentally regulated genes. ChIP analyses suggested that CHD4 modulates H3K4 methylation levels at the promoter and coding regions of target genes. We further demonstrated that the Caenorhabditis elegans homologues of KDM5 and CHD4 function in the same pathway during vulva development. These results suggest that KDM5A and the NuRD complex cooperatively function to control developmentally regulated genes.
Collapse
Affiliation(s)
- Gohei Nishibuchi
- From the Graduate School of Natural Sciences, Nagoya City University, Nagoya 467-8501
| | - Yukimasa Shibata
- the Department of Bioscience, Graduate School of Science and Technology, Kwansei-Gakuin University, Sanda, Hyogo 669-1337, and
| | | | | | | | - Kaori Sinmyozu
- Proteomics Support Unit, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Hideaki Tagami
- From the Graduate School of Natural Sciences, Nagoya City University, Nagoya 467-8501
| | - Jun-ichi Nakayama
- From the Graduate School of Natural Sciences, Nagoya City University, Nagoya 467-8501, the Laboratory for Chromatin Dynamics and
| |
Collapse
|
64
|
Nie X, Wang H, Li J, Holec S, Berger F. The HIRA complex that deposits the histone H3.3 is conserved in Arabidopsis and facilitates transcriptional dynamics. Biol Open 2014; 3:794-802. [PMID: 25086063 PMCID: PMC4163656 DOI: 10.1242/bio.20148680] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In animals, replication-independent incorporation of nucleosomes containing the histone variant H3.3 enables global reprogramming of histone modifications and transcriptional profiles. H3.3 enrichment over gene bodies correlates with gene transcription in animals and plants. In animals, H3.3 is deposited into chromatin by specific protein complexes, including the HIRA complex. H3.3 variants evolved independently and acquired similar properties in animals and plants, questioning how the H3.3 deposition machinery evolved in plants and what are its biological functions. We performed phylogenetic analyses in the plant kingdom and identified in Arabidopsis all orthologs of human genes encoding members of the HIRA complex. Genetic analyses, biochemical data and protein localisation suggest that these proteins form a complex able to interact with H3.3 in Arabidopsis in a manner similar to that described in mammals. In contrast to animals, where HIRA is required for fertilization and early development, loss of function of HIRA in Arabidopsis causes mild phenotypes in the adult plant and does not perturb sexual reproduction and embryogenesis. Rather, HIRA function is required for transcriptional reprogramming during dedifferentiation of plant cells that precedes vegetative propagation and for the appropriate transcription of genes responsive to biotic and abiotic factors. We conclude that the molecular function of the HIRA complex is conserved between plants and animals. Yet plants diversified HIRA functions to enable asexual reproduction and responsiveness to the environment in response to the plant sessile lifestyle.
Collapse
Affiliation(s)
- Xin Nie
- Temasek Lifesciences Laboratory, 1 Research Link, National University of Singapore, 117604 Singapore Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543 Singapore
| | - Haifeng Wang
- Temasek Lifesciences Laboratory, 1 Research Link, National University of Singapore, 117604 Singapore Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543 Singapore
| | - Jing Li
- Temasek Lifesciences Laboratory, 1 Research Link, National University of Singapore, 117604 Singapore Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543 Singapore
| | - Sarah Holec
- Temasek Lifesciences Laboratory, 1 Research Link, National University of Singapore, 117604 Singapore
| | - Frédéric Berger
- Temasek Lifesciences Laboratory, 1 Research Link, National University of Singapore, 117604 Singapore Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543 Singapore
| |
Collapse
|
65
|
Xie G, Yu Z, Jia D, Jiao R, Deng WM. E(y)1/TAF9 mediates the transcriptional output of Notch signaling in Drosophila. J Cell Sci 2014; 127:3830-9. [PMID: 25015288 DOI: 10.1242/jcs.154583] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Transcriptional activation of Notch signaling targets requires the formation of a ternary complex that involves the intracellular domain of the Notch receptor (NICD), DNA-binding protein Suppressor of Hairless [Su(H), RPBJ in mammals] and coactivator Mastermind (Mam). Here, we report that E(y)1/TAF9, a component of the transcription factor TFIID complex, interacts specifically with the NICD-Su(H)-Mam complex to facilitate the transcriptional output of Notch signaling. We identified E(y)1/TAF9 in a large-scale in vivo RNA interference (RNAi) screen for genes that are involved in a Notch-dependent mitotic-to-endocycle transition in Drosophila follicle cells. Knockdown of e(y)1/TAF9 displayed Notch-mutant-like phenotypes and defects in target gene and activity reporter expression in both the follicle cells and wing imaginal discs. Epistatic analyses in these two tissues indicated that E(y)1/TAF9 functions downstream of Notch cleavage. Biochemical studies in S2 cells demonstrated that E(y)1/TAF9 physically interacts with the transcriptional effectors of Notch signaling Su(H) and NICD. Taken together, our data suggest that the association of the NICD-Su(H)-Mastermind complex with E(y)1/TAF9 in response to Notch activation recruits the transcription initiation complex to induce Notch target genes, coupling Notch signaling with the transcription machinery.
Collapse
Affiliation(s)
- Gengqiang Xie
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295, USA
| | - Zhongsheng Yu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China
| | - Dongyu Jia
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295, USA
| | - Renjie Jiao
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295, USA
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, FL 32304-4295, USA
| |
Collapse
|
66
|
Human-Chromatin-Related Protein Interactions Identify a Demethylase Complex Required for Chromosome Segregation. Cell Rep 2014; 8:297-310. [DOI: 10.1016/j.celrep.2014.05.050] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 04/24/2014] [Accepted: 05/27/2014] [Indexed: 01/14/2023] Open
|
67
|
Im JS, Keaton M, Lee KY, Kumar P, Park J, Dutta A. ATR checkpoint kinase and CRL1βTRCP collaborate to degrade ASF1a and thus repress genes overlapping with clusters of stalled replication forks. Genes Dev 2014; 28:875-87. [PMID: 24700029 PMCID: PMC4003279 DOI: 10.1101/gad.239194.114] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Many chemotherapeutic agents, such as doxorubicin (DOX), interfere with DNA replication. Here, Dutta and colleagues show that DOX treatment produces clusters of stalled replication forks and transcriptional repression of neighboring genes. An ATR-dependent checkpoint pathway that down-regulates histone chaperone ASF1a is shown to repress genes overlapping with stalled replication forks. Furthermore, ASF1a-depleted cancer cells are more sensitive to DOX, suggesting that the loss of this histone chaperone, as seen in several cancers, could be a personalized tumor marker for sensitivity to DOX. Many agents used for chemotherapy, such as doxorubicin, interfere with DNA replication, but the effect of this interference on transcription is largely unknown. Here we show that doxorubicin induces the firing of dense clusters of neoreplication origins that lead to clusters of stalled replication forks in gene-rich parts of the genome, particularly on expressed genes. Genes that overlap with these clusters of stalled forks are actively dechromatinized, unwound, and repressed by an ATR-dependent checkpoint pathway. The ATR checkpoint pathway causes a histone chaperone normally associated with the replication fork, ASF1a, to degrade through a CRL1βTRCP-dependent ubiquitination/proteasome pathway, leading to the localized dechromatinization and gene repression. Therefore, a globally active checkpoint pathway interacts with local clusters of stalled forks to specifically repress genes in the vicinity of the stalled forks, providing a new mechanism of action of chemotherapy drugs like doxorubicin. Finally, ASF1a-depleted cancer cells are more sensitive to doxorubicin, suggesting that the 7%–10% of prostate adenocarcinomas and adenoid cystic carcinomas reported to have homozygous deletion or significant underexpression of ASF1a should be tested for high sensitivity to doxorubicin.
Collapse
Affiliation(s)
- Jun-Sub Im
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | |
Collapse
|
68
|
Hou J, Wang Z, Yang L, Guo X, Yang G. The function of EMSY in cancer development. Tumour Biol 2014; 35:5061-6. [PMID: 24609898 DOI: 10.1007/s13277-013-1584-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022] Open
Abstract
EMSY was first reported to bind BRCA2 and to inactivate the function of BRCA2, leading to the development of sporadic breast and ovarian cancers. The function of EMSY may also be involved in DNA damage repair, genomic instability, and chromatin remolding. Recent studies have shown that amplification of EMSY was also associated with other cancers such as prostate and pancreatic cancers and linked to tumor phenotypes and clinical outcomes. By reviewing literatures published since 2003, here, we have summarized the recent advances of EMSY in cancer development.
Collapse
Affiliation(s)
- Jing Hou
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | | | | | | | | |
Collapse
|
69
|
Viré E, Curtis C, Davalos V, Git A, Robson S, Villanueva A, Vidal A, Barbieri I, Aparicio S, Esteller M, Caldas C, Kouzarides T. The breast cancer oncogene EMSY represses transcription of antimetastatic microRNA miR-31. Mol Cell 2014; 53:806-18. [PMID: 24582497 PMCID: PMC3988886 DOI: 10.1016/j.molcel.2014.01.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 11/11/2013] [Accepted: 01/23/2014] [Indexed: 11/16/2022]
Abstract
Amplification of the EMSY gene in sporadic breast and ovarian cancers is a poor prognostic indicator. Although EMSY has been linked to transcriptional silencing, its mechanism of action is unknown. Here, we report that EMSY acts as an oncogene, causing the transformation of cells in vitro and potentiating tumor formation and metastatic features in vivo. We identify an inverse correlation between EMSY amplification and miR-31 expression, an antimetastatic microRNA, in the METABRIC cohort of human breast samples. Re-expression of miR-31 profoundly reduced cell migration, invasion, and colony-formation abilities of cells overexpressing EMSY or haboring EMSY amplification. We show that EMSY is recruited to the miR-31 promoter by the DNA binding factor ETS-1, and it represses miR-31 transcription by delivering the H3K4me3 demethylase JARID1b/PLU-1/KDM5B. Altogether, these results suggest a pathway underlying the role of EMSY in breast cancer and uncover potential diagnostic and therapeutic targets in sporadic breast cancer.
Collapse
Affiliation(s)
- Emmanuelle Viré
- Gurdon Institute and Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Christina Curtis
- Department of Oncology and Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, CB2 0RE, UK
| | - Veronica Davalos
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08907, Spain
| | - Anna Git
- Department of Oncology and Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, CB2 0RE, UK
| | - Samuel Robson
- Gurdon Institute and Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Alberto Villanueva
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08907, Spain
| | - August Vidal
- Department of Pathological Anatomy, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08907, Spain
| | | | - Samuel Aparicio
- Department of Molecular Oncology, British Columbia Cancer Agency, 675 West 10(th) Avenue, V5Z 1L3 Vancouver, Canada
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08907, Spain
| | - Carlos Caldas
- Department of Oncology and Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, CB2 0RE, UK; Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge University Hospital, NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK; Cambridge Experimental Cancer Medicine Centre (ECMC), Cambridge CB2 0RE, UK
| | - Tony Kouzarides
- Gurdon Institute and Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK.
| |
Collapse
|
70
|
Garrote AM, Redondo P, Montoya G, Muñoz IG. Purification, crystallization and preliminary X-ray diffraction analysis of the kinase domain of human tousled-like kinase 2. Acta Crystallogr F Struct Biol Commun 2014; 70:354-7. [PMID: 24598926 PMCID: PMC3944701 DOI: 10.1107/s2053230x14002581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/04/2014] [Indexed: 11/10/2022] Open
Abstract
Tousled-like kinases (TLKs) are an evolutionarily conserved family of serine/threonine protein kinases involved in chromatin dynamics, including DNA replication and repair, transcription and chromosome segregation. The two members of the family reported in humans, namely TLK1 and TLK2, localize to the cell nucleus and are capable of forming homo- or hetero-oligomers by themselves. To characterize the role of TLK2, its C-terminal kinase domain was cloned and overexpressed in Escherichia coli followed by purification to homogeneity. Crystallization experiments in the presence of ATP-γ-S yielded crystals suitable for X-ray diffraction analysis belonging to two different space groups: tetragonal I4122 and cubic P213. The latter produced the best diffracting crystal (3.4 Å resolution using synchrotron radiation), with unit-cell parameters a = b = c = 126.05 Å, α = β = γ = 90°. The asymmetric unit contained one protein molecule, with a Matthews coefficient of 4.59 Å(3) Da(-1) and a solvent content of 73.23%.
Collapse
Affiliation(s)
- Ana M. Garrote
- Macromolecular Crystallography Group, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Pilar Redondo
- Macromolecular Crystallography Group, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Guillermo Montoya
- Macromolecular Crystallography Group, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Structural Biology Group, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Inés G. Muñoz
- Macromolecular Crystallography Group, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
71
|
Sanidas I, Polytarchou C, Hatziapostolou M, Ezell SA, Kottakis F, Hu L, Guo A, Xie J, Comb MJ, Iliopoulos D, Tsichlis PN. Phosphoproteomics screen reveals akt isoform-specific signals linking RNA processing to lung cancer. Mol Cell 2014; 53:577-90. [PMID: 24462114 DOI: 10.1016/j.molcel.2013.12.018] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/02/2013] [Accepted: 12/18/2013] [Indexed: 01/09/2023]
Abstract
The three Akt isoforms are functionally distinct. Here we show that their phosphoproteomes also differ, suggesting that their functional differences are due to differences in target specificity. One of the top cellular functions differentially regulated by Akt isoforms is RNA processing. IWS1, an RNA processing regulator, is phosphorylated by Akt3 and Akt1 at Ser720/Thr721. The latter is required for the recruitment of SETD2 to the RNA Pol II complex. SETD2 trimethylates histone H3 at K36 during transcription, creating a docking site for MRG15 and PTB. H3K36me3-bound MRG15 and PTB regulate FGFR-2 splicing, which controls tumor growth and invasiveness downstream of IWS1 phosphorylation. Twenty-one of the twenty-four non-small-cell-lung carcinomas we analyzed express IWS1. More importantly, the stoichiometry of IWS1 phosphorylation in these tumors correlates with the FGFR-2 splicing pattern and with Akt phosphorylation and Akt3 expression. These data identify an Akt isoform-dependent regulatory mechanism for RNA processing and demonstrate its role in lung cancer.
Collapse
Affiliation(s)
- Ioannis Sanidas
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Christos Polytarchou
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; Division of Digestive Diseases, Center for Systems Biomedicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Maria Hatziapostolou
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; Division of Digestive Diseases, Center for Systems Biomedicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Scott A Ezell
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Filippos Kottakis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Lan Hu
- Department of Biostatistics and Computational Biology, Center for Cancer Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ailan Guo
- Cell Signaling Technology, Danvers, MA 01923, USA
| | - Jianxin Xie
- Cell Signaling Technology, Danvers, MA 01923, USA
| | | | - Dimitrios Iliopoulos
- Division of Digestive Diseases, Center for Systems Biomedicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Philip N Tsichlis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
72
|
|
73
|
Yu Z, Wu H, Chen H, Wang R, Liang X, Liu J, Li C, Deng WM, Jiao R. CAF-1 promotes Notch signaling through epigenetic control of target gene expression during Drosophila development. Development 2013; 140:3635-44. [PMID: 23942516 DOI: 10.1242/dev.094599] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The histone chaperone CAF-1 is known for its role in DNA replication-coupled histone deposition. However, loss of function causes lethality only in higher multicellular organisms such as mice and flies, but not in unicellular organisms such as yeasts, suggesting that CAF-1 has other important functions than histone deposition during animal development. Emerging evidence indicates that CAF-1 also has a role in higher order chromatin organization and heterochromatin-mediated gene expression; it remains unclear whether CAF-1 has a role in specific signaling cascades to promote gene expression during development. Here, we report that knockdown of one of the subunits of Drosophila CAF-1, dCAF-1-p105 (Caf1-105), results in phenotypes that resemble those of, and are augmented synergistically by, mutations of Notch positive regulatory pathway components. Depletion of dCAF-1-p105 leads to abrogation of cut expression and to downregulation of other Notch target genes in wing imaginal discs. dCAF-1-p105 is associated with Suppressor of Hairless [Su(H)] and regulates its binding to the enhancer region of E(spl)mβ. The association of dCAF-1-p105 with Su(H) on chromatin establishes an active local chromatin status for transcription by maintaining a high level of histone H4 acetylation. In response to induced Notch activation, dCAF-1 associates with the Notch intracellular domain to activate the expression of Notch target genes in cultured S2 cells, manifesting the role of dCAF-1 in Notch signaling. Together, our results reveal a novel epigenetic function of dCAF-1 in promoting Notch pathway activity that regulates normal Drosophila development.
Collapse
Affiliation(s)
- Zhongsheng Yu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, The Chinese Academy of Sciences, Datun Road 15, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Affiliation(s)
- Jonathan M. G. Higgins
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mary Herbert
- Newcastle Fertility Centre and Institute for Aging and Health, International Centre for Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
75
|
Moshkin YM, Doyen CM, Kan TW, Chalkley GE, Sap K, Bezstarosti K, Demmers JA, Ozgur Z, van Ijcken WFJ, Verrijzer CP. Histone chaperone NAP1 mediates sister chromatid resolution by counteracting protein phosphatase 2A. PLoS Genet 2013; 9:e1003719. [PMID: 24086141 PMCID: PMC3784504 DOI: 10.1371/journal.pgen.1003719] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 06/26/2013] [Indexed: 12/27/2022] Open
Abstract
Chromosome duplication and transmission into daughter cells requires the precisely orchestrated binding and release of cohesin. We found that the Drosophila histone chaperone NAP1 is required for cohesin release and sister chromatid resolution during mitosis. Genome-wide surveys revealed that NAP1 and cohesin co-localize at multiple genomic loci. Proteomic and biochemical analysis established that NAP1 associates with the full cohesin complex, but it also forms a separate complex with the cohesin subunit stromalin (SA). NAP1 binding to cohesin is cell-cycle regulated and increases during G2/M phase. This causes the dissociation of protein phosphatase 2A (PP2A) from cohesin, increased phosphorylation of SA and cohesin removal in early mitosis. PP2A depletion led to a loss of centromeric cohesion. The distinct mitotic phenotypes caused by the loss of either PP2A or NAP1, were both rescued by their concomitant depletion. We conclude that the balanced antagonism between NAP1 and PP2A controls cohesin dissociation during mitosis.
Collapse
Affiliation(s)
- Yuri M. Moshkin
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cecile M. Doyen
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tsung-Wai Kan
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gillian E. Chalkley
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Karen Sap
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Karel Bezstarosti
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen A. Demmers
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Zeliha Ozgur
- Genomics Centre, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - C. Peter Verrijzer
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
76
|
Doyen CM, Moshkin YM, Chalkley GE, Bezstarosti K, Demmers JAA, Rathke C, Renkawitz-Pohl R, Verrijzer CP. Subunits of the histone chaperone CAF1 also mediate assembly of protamine-based chromatin. Cell Rep 2013; 4:59-65. [PMID: 23810557 DOI: 10.1016/j.celrep.2013.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/25/2013] [Accepted: 06/01/2013] [Indexed: 11/20/2022] Open
Abstract
One of the most dramatic forms of chromatin reorganization occurs during spermatogenesis, when the paternal genome is repackaged from a nucleosomal to a protamine-based structure. We assessed the role of the canonical histone chaperone CAF1 in Drosophila spermatogenesis. In this process, CAF1 does not behave as a complex, but its subunits display distinct chromatin dynamics. During histone-to-protamine replacement, CAF1-p180 dissociates from the DNA while CAF1-p75 binds and stays on as a component of sperm chromatin. Association of CAF1-p75 with the paternal genome depends on CAF1-p180 and protamines. Conversely, CAF1-p75 binds protamines and is required for their incorporation into sperm chromatin. Histone removal, however, occurs independently of CAF1 or protamines. Thus, CAF1-p180 and CAF1-p75 function in a temporal hierarchy during sperm chromatin assembly, with CAF1-p75 acting as a protamine-loading factor. These results show that CAF1 subunits mediate the assembly of two fundamentally different forms of chromatin.
Collapse
Affiliation(s)
- Cécile M Doyen
- Department of Biochemistry, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Attia M, Rachez C, Avner P, Rogner UC. Nucleosome assembly proteins and their interacting proteins in neuronal differentiation. Arch Biochem Biophys 2013; 534:20-6. [DOI: 10.1016/j.abb.2012.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/21/2012] [Accepted: 09/22/2012] [Indexed: 12/21/2022]
|
78
|
Dai Q, Andreu-Agullo C, Insolera R, Wong LC, Shi SH, Lai EC. BEND6 is a nuclear antagonist of Notch signaling during self-renewal of neural stem cells. Development 2013; 140:1892-902. [PMID: 23571214 DOI: 10.1242/dev.087502] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The activity of the Notch pathway revolves around a CSL-class transcription factor, which recruits distinct complexes that activate or repress target gene expression. The co-activator complex is deeply conserved and includes the cleaved Notch intracellular domain (NICD) and Mastermind. By contrast, numerous CSL co-repressor proteins have been identified, and these are mostly different between invertebrate and vertebrate systems. In this study, we demonstrate that mammalian BEND6 is a neural BEN-solo factor that shares many functional attributes with Drosophila Insensitive, a co-repressor for the Drosophila CSL factor. BEND6 binds the mammalian CSL protein CBF1 and antagonizes Notch-dependent target activation. In addition, its association with Notch- and CBF1-regulated enhancers is promoted by CBF1 and antagonized by activated Notch. In utero electroporation experiments showed that ectopic BEND6 inhibited Notch-mediated self-renewal of neocortical neural stem cells and promoted neurogenesis. Conversely, knockdown of BEND6 increased NSC self-renewal in wild-type neocortex, and exhibited genetic interactions with gain and loss of Notch pathway activity. We recapitulated all of these findings in cultured neurospheres, in which overexpression and depletion of BEND6 caused reciprocal effects on neural stem cell renewal and neurogenesis. These data reveal a novel mammalian CSL co-repressor in the nervous system, and show that the Notch-inhibitory activity of certain BEN-solo proteins is conserved between flies and mammals.
Collapse
Affiliation(s)
- Qi Dai
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | | | | | | | | | | |
Collapse
|
79
|
Orsi GA, Algazeery A, Meyer RE, Capri M, Sapey-Triomphe LM, Horard B, Gruffat H, Couble P, Aït-Ahmed O, Loppin B. Drosophila Yemanuclein and HIRA cooperate for de novo assembly of H3.3-containing nucleosomes in the male pronucleus. PLoS Genet 2013; 9:e1003285. [PMID: 23408912 PMCID: PMC3567178 DOI: 10.1371/journal.pgen.1003285] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 12/14/2012] [Indexed: 01/13/2023] Open
Abstract
The differentiation of post-meiotic spermatids in animals is characterized by a unique reorganization of their nuclear architecture and chromatin composition. In many species, the formation of sperm nuclei involves the massive replacement of nucleosomes with protamines, followed by a phase of extreme nuclear compaction. At fertilization, the reconstitution of a nucleosome-based paternal chromatin after the removal of protamines requires the deposition of maternally provided histones before the first round of DNA replication. This process exclusively uses the histone H3 variant H3.3 and constitutes a unique case of genome-wide replication-independent (RI) de novo chromatin assembly. We had previously shown that the histone H3.3 chaperone HIRA plays a central role for paternal chromatin assembly in Drosophila. Although several conserved HIRA-interacting proteins have been identified from yeast to human, their conservation in Drosophila, as well as their actual implication in this highly peculiar RI nucleosome assembly process, is an open question. Here, we show that Yemanuclein (YEM), the Drosophila member of the Hpc2/Ubinuclein family, is essential for histone deposition in the male pronucleus. yem loss of function alleles affect male pronucleus formation in a way remarkably similar to Hira mutants and abolish RI paternal chromatin assembly. In addition, we demonstrate that HIRA and YEM proteins interact and are mutually dependent for their targeting to the decondensing male pronucleus. Finally, we show that the alternative ATRX/XNP-dependent H3.3 deposition pathway is not involved in paternal chromatin assembly, thus underlining the specific implication of the HIRA/YEM complex for this essential step of zygote formation. Chromosome organization relies on a basic functional unit called the nucleosome, in which DNA is wrapped around a core of histone proteins. However, during male gamete formation, the majority of histones are replaced by sperm-specific proteins that are adapted to sexual reproduction but incompatible with the formation of the first zygotic nucleus. These proteins must therefore be replaced by histones upon fertilization, in a replication-independent chromatin assembly process that requires the histone deposition factor HIRA. In this study, we identified the protein Yemanuclein (YEM) as a new partner of HIRA at fertilization. We show that, in eggs laid by yem mutant females, the male pronucleus fails to assemble its nucleosomes, resulting in the loss of paternal chromosomes at the first zygotic division. In addition, we found that YEM and HIRA are mutually dependent to perform chromatin assembly at fertilization, demonstrating that they tightly cooperate in vivo. Finally, we demonstrate that the replication-independent chromatin assembly factor ATRX/XNP is not involved in the assembly of paternal nucleosomes. In conclusion, our results shed new light into critical mechanisms controlling paternal chromosome formation at fertilization.
Collapse
Affiliation(s)
- Guillermo A. Orsi
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR5534, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Ahmed Algazeery
- Institut de Génétique Humaine, CNRS UPR 1142, Montpellier, France
| | - Régis E. Meyer
- Institut de Génétique Humaine, CNRS UPR 1142, Montpellier, France
| | - Michèle Capri
- Institut de Génétique Humaine, CNRS UPR 1142, Montpellier, France
| | - Laure M. Sapey-Triomphe
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR5534, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Béatrice Horard
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR5534, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Henri Gruffat
- Laboratoire de Biologie Moléculaire des Herpesvirus, INSERM U758, Ecole Normale Supérieure de Lyon, France
| | - Pierre Couble
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR5534, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Ounissa Aït-Ahmed
- Institut de Génétique Humaine, CNRS UPR 1142, Montpellier, France
- * E-mail: (OA-A); (BL)
| | - Benjamin Loppin
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR5534, Université Claude Bernard Lyon 1, Villeurbanne, France
- * E-mail: (OA-A); (BL)
| |
Collapse
|
80
|
Ezell SA, Tsichlis PN. Akt1, EMSY, BRCA2 and type I IFN signaling: a novel arm of the IFN response. Transcription 2012; 3:305-9. [PMID: 23117821 DOI: 10.4161/trns.21904] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Interferon-stimulated transcription is thought to occur mainly through the action of the JAK/STAT pathway. However, recent findings revealed an additional PI3K/Akt-dependent pathway, which contributes to the induction of a set of interferon-stimulated genes (ISGs) through the regulation of the transcriptional repressor EMSY.
Collapse
Affiliation(s)
- Scott A Ezell
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA
| | | |
Collapse
|
81
|
Lloret-Llinares M, Pérez-Lluch S, Rossell D, Morán T, Ponsa-Cobas J, Auer H, Corominas M, Azorín F. dKDM5/LID regulates H3K4me3 dynamics at the transcription-start site (TSS) of actively transcribed developmental genes. Nucleic Acids Res 2012; 40:9493-505. [PMID: 22904080 PMCID: PMC3479210 DOI: 10.1093/nar/gks773] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
H3K4me3 is a histone modification that accumulates at the transcription-start site (TSS) of active genes and is known to be important for transcription activation. The way in which H3K4me3 is regulated at TSS and the actual molecular basis of its contribution to transcription remain largely unanswered. To address these questions, we have analyzed the contribution of dKDM5/LID, the main H3K4me3 demethylase in Drosophila, to the regulation of the pattern of H3K4me3. ChIP-seq results show that, at developmental genes, dKDM5/LID localizes at TSS and regulates H3K4me3. dKDM5/LID target genes are highly transcribed and enriched in active RNApol II and H3K36me3, suggesting a positive contribution to transcription. Expression-profiling show that, though weakly, dKDM5/LID target genes are significantly downregulated upon dKDM5/LID depletion. Furthermore, dKDM5/LID depletion results in decreased RNApol II occupancy, particularly by the promoter-proximal Pol lloser5 form. Our results also show that ASH2, an evolutionarily conserved factor that locates at TSS and is required for H3K4me3, binds and positively regulates dKDM5/LID target genes. However, dKDM5/LID and ASH2 do not bind simultaneously and recognize different chromatin states, enriched in H3K4me3 and not, respectively. These results indicate that, at developmental genes, dKDM5/LID and ASH2 coordinately regulate H3K4me3 at TSS and that this dynamic regulation contributes to transcription.
Collapse
Affiliation(s)
- Marta Lloret-Llinares
- Institute of Molecular Biology of Barcelona, CSIC and Institute for Research in Biomedicine, IRB Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Wu CY, Tsai YP, Wu MZ, Teng SC, Wu KJ. Epigenetic reprogramming and post-transcriptional regulation during the epithelial-mesenchymal transition. Trends Genet 2012; 28:454-63. [PMID: 22717049 DOI: 10.1016/j.tig.2012.05.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/18/2012] [Accepted: 05/18/2012] [Indexed: 01/08/2023]
Abstract
The epithelial-mesenchymal transition (EMT) is a developmental process that is important for organ development, metastasis, cancer stemness, and organ fibrosis. The EMT process is regulated by different signaling pathways as well as by various epigenetic and post-transcriptional mechanisms. Here, we review recent progress describing the role of different chromatin modifiers in various signaling events leading to EMT, including hypoxia, transforming growth factor (TGF)-β, Notch, and Wnt. We also discuss post-transcriptional mechanisms, such as RNA alternative splicing and the effects of miRNAs in EMT regulation. Furthermore, we highlight on-going and future work aimed at a detailed understanding of the epigenetic and post-transcriptional mechanisms that regulate EMT. This work will shed new light on the cellular and tumorigenic processes affected by EMT misregulation.
Collapse
Affiliation(s)
- Chung-Yin Wu
- Department of Occupational Medicine, Far Eastern Memorial Hospital, New Taipei City, 220, Taiwan
| | | | | | | | | |
Collapse
|
83
|
Symbiodinium transcriptomes: genome insights into the dinoflagellate symbionts of reef-building corals. PLoS One 2012; 7:e35269. [PMID: 22529998 PMCID: PMC3329448 DOI: 10.1371/journal.pone.0035269] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Accepted: 03/13/2012] [Indexed: 12/20/2022] Open
Abstract
Dinoflagellates are unicellular algae that are ubiquitously abundant in aquatic environments. Species of the genus Symbiodinium form symbiotic relationships with reef-building corals and other marine invertebrates. Despite their ecologic importance, little is known about the genetics of dinoflagellates in general and Symbiodinium in particular. Here, we used 454 sequencing to generate transcriptome data from two Symbiodinium species from different clades (clade A and clade B). With more than 56,000 assembled sequences per species, these data represent the largest transcriptomic resource for dinoflagellates to date. Our results corroborate previous observations that dinoflagellates possess the complete nucleosome machinery. We found a complete set of core histones as well as several H3 variants and H2A.Z in one species. Furthermore, transcriptome analysis points toward a low number of transcription factors in Symbiodinium spp. that also differ in the distribution of DNA-binding domains relative to other eukaryotes. In particular the cold shock domain was predominant among transcription factors. Additionally, we found a high number of antioxidative genes in comparison to non-symbiotic but evolutionary related organisms. These findings might be of relevance in the context of the role that Symbiodinium spp. play as coral symbionts. Our data represent the most comprehensive dinoflagellate EST data set to date. This study provides a comprehensive resource to further analyze the genetic makeup, metabolic capacities, and gene repertoire of Symbiodinium and dinoflagellates. Overall, our findings indicate that Symbiodinium possesses some unique characteristics, in particular the transcriptional regulation in Symbiodinium may differ from the currently known mechanisms of eukaryotic gene regulation.
Collapse
|
84
|
Huang H, Jiao R. Roles of chromatin assembly factor 1 in the epigenetic control of chromatin plasticity. SCIENCE CHINA-LIFE SCIENCES 2012; 55:15-9. [DOI: 10.1007/s11427-012-4269-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 07/30/2011] [Indexed: 10/14/2022]
|
85
|
Giri S, Prasanth SG. Replicating and transcribing on twisted roads of chromatin. Brief Funct Genomics 2012; 11:188-204. [PMID: 22267489 DOI: 10.1093/bfgp/elr047] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chromatin, a complex of DNA and proteins in the eukaryotic cell nucleus governs various cellular processes including DNA replication, DNA repair and transcription. Chromatin architecture and dynamics dictates the timing of cellular events by regulating proteins' accessibility to DNA as well as by acting as a scaffold for protein-protein interactions. Nucleosome, the basic unit of chromatin consists of a histone octamer comprised of (H3-H4)2 tetramer and two H2A-H2B dimers on which 146 bp of DNA is wrapped around ~1.6 times. Chromatin changes brought about by histone modifications, histone-modifying enzymes, chromatin remodeling factors, histone chaperones, histone variants and chromatin dynamics influence the regulation and timing of gene expression. Similarly, the timing of DNA replication is dependent on the chromatin context that in turn dictates origin selection. Further, during the process of DNA replication, not only does an organism's DNA have to be accurately replicated but also the chromatin structure and the epigenetic marks have to be faithfully transmitted to the daughter cells. Active transcription has been shown to repress replication while at the same time it has been shown that when origins are located at promoters, because of enhanced chromatin accessibility, they fire efficiently. In this review, we focus on how chromatin modulates two fundamental processes, DNA replication and transcription.
Collapse
Affiliation(s)
- Sumanprava Giri
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, 601S Goodwin Avenue, Urbana, IL 61801, USA
| | | |
Collapse
|
86
|
Targeting Notch signaling for cancer therapeutic intervention. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 65:191-234. [PMID: 22959027 DOI: 10.1016/b978-0-12-397927-8.00007-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Notch signaling pathway is an evolutionarily conserved, intercellular signaling cascade. The Notch proteins are single-pass receptors that are activated upon interaction with the Delta (or Delta-like) and Jagged/Serrate families of membrane-bound ligands. Association of ligand-receptor leads to proteolytic cleavages that liberate the Notch intracellular domain (NICD) from the plasma membrane. The NICD translocates to the nucleus, where it forms a complex with the DNA-binding protein CSL, displacing a histone deacetylase (HDAc)-corepressor (CoR) complex from CSL. Components of a transcriptional complex, such as MAML1 and histone acetyltransferases (HATs), are recruited to the NICD-CSL complex, leading to the transcriptional activation of Notch target genes. The Notch signaling pathway plays a critical role in cell fate decision, tissue patterning, morphogenesis, and is hence regarded as a developmental pathway. However, if this pathway goes awry, it contributes to cellular transformation and tumorigenesis. There is mounting evidence that this pathway is dysregulated in a variety of malignancies, and can behave as either an oncogene or a tumor suppressor depending upon cell context. This chapter highlights the current evidence for aberration of the Notch signaling pathway in a wide range of tumors from hematological cancers, such as leukemia and lymphoma, through to lung, skin, breast, pancreas, colon, prostate, ovarian, brain, and liver tumors. It proposes that the Notch signaling pathway may represent novel target for cancer therapeutic intervention.
Collapse
|
87
|
Abstract
The 'histone code' hypothesis states that chromatin-based regulation of nuclear processes such as transcription is brought about by the combination of distinct modifications (histone marks) at specific loci. Its correct establishment involves chromatin cross-talks, ensuring an ordered and concerted deposition/removal of a particular set of modifications that act together to give the correct transcriptional outcome. Histone methylation on lysine residues can negatively or positively impact on gene transcription, depending on the residue and on its degree of methylation. Thanks to this complexity and given the number of chromatin 'readers' that can recognize methylated lysine residues, histone methylation plays a very special role in specifying the various chromatin states. The recent discovery of histone demethylases, which represent a large family of enzymes often containing histone modification binding modules, sheds new light on cross-talk mechanisms involving methylated residues. In the present review, after a brief overview of the various families of histone demethylases, we describe the different mechanisms by which they participate in chromatin cross-talks and how these mechanisms are integrated to achieve the mutual exclusion or the link between chromatin marks, leading to the establishment of the correct histone code.
Collapse
|
88
|
Wright DE, Wang CY, Kao CF. Flickin' the ubiquitin switch: the role of H2B ubiquitylation in development. Epigenetics 2011; 6:1165-75. [PMID: 21937884 DOI: 10.4161/epi.6.10.17745] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The reversible ubiquitylation of histone H2B has long been implicated in transcriptional activation and gene silencing. However, many questions regarding its regulation and effects on chromatin structure remain unanswered. In addition, while several studies have uncovered an involvement of this modification in the control of certain developmental processes, a more general understanding of its requirement is lacking. Herein, we present a broad overview of the pathways known to be regulated by H2B ubiquitylation, while drawing parallels between findings in disparate organisms, in order to facilitate continued delineation of its spatiotemporal role in development. Finally, we integrate the findings of recent studies into how H2B ubiquitylation affects chromatin, and cast an eye over emerging areas for future research.
Collapse
|
89
|
Cave JW. Selective repression of Notch pathway target gene transcription. Dev Biol 2011; 360:123-31. [PMID: 21963536 DOI: 10.1016/j.ydbio.2011.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 08/28/2011] [Accepted: 09/14/2011] [Indexed: 12/21/2022]
Abstract
The Notch signaling pathway regulates metazoan development, in part, by directly controlling the transcription of target genes. For a given cellular context, however, only subsets of the known target genes are transcribed when the pathway is activated. Thus, there are context-dependent mechanisms that selectively maintain repression of target gene transcription when the Notch pathway is activated. This review focuses on molecular mechanisms that have been recently reported to mediate selective repression of Notch pathway target gene transcription. These mechanisms are essential for generating the complex spatial and temporal expression patterns of Notch target genes during development.
Collapse
Affiliation(s)
- John W Cave
- Dept. of. Neurology and Neuroscience, Weill Cornell Medical College, 785 Mamaroneck Ave., White Plains, NY 10605, USA.
| |
Collapse
|
90
|
Arzate-Mejía RG, Valle-García D, Recillas-Targa F. Signaling epigenetics: Novel insights on cell signaling and epigenetic regulation. IUBMB Life 2011; 63:881-95. [DOI: 10.1002/iub.557] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 07/12/2011] [Indexed: 12/12/2022]
|
91
|
Abstract
Mulligan et al. (2011) show here that the NAD(+)-dependent SIRT1 (H4K16; H1K26) deacetylase acts in concert with the LSD1 (H3K4) demethylase to repress Notch-induced transcription, thus coupling two distinct histone modifications at a key epigenetic switch for Notch target genes.
Collapse
Affiliation(s)
- Katherine A Jones
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037-1099, USA.
| |
Collapse
|
92
|
Kato Y. The multiple roles of Notch signaling during left-right patterning. Cell Mol Life Sci 2011; 68:2555-67. [PMID: 21544546 PMCID: PMC11114802 DOI: 10.1007/s00018-011-0695-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 03/28/2011] [Accepted: 04/19/2011] [Indexed: 10/18/2022]
Abstract
The establishment of left-right (LR) asymmetry is regulated by intricate signaling mechanisms during embryogenesis and this asymmetry is critical for morphogenesis as well as the positioning of internal organs within the organism. Recent progress including elucidation of ion transporters, leftward nodal flow, and regulation of asymmetric gene expression contributes to our understanding of how the breaking of the symmetry is initiated and how this laterality information is subsequently transmitted to the organ primordium. A number of developmental signaling pathways have been implicated in this complex process. In this review, we will focus on the roles of the Notch signaling pathway during development of LR asymmetry. The Notch signaling pathway is a short-range communication system between neighboring cells. While Notch signaling plays essential roles in regulating the morphogenesis of the node and left-specific expression of Nodal in the lateral plate mesoderm, a hallmark gene in LR patterning, Notch signaling also suppresses the expression of Pitx2 that is a direct downstream target of Nodal during later stages of development. This negative activity of Notch signaling towards left-specific activity was recently shown to be inhibited by the B cell lymphoma 6 (BCL6)/BCL6 co-repressor (BcoR) transcriptional repressor complex in a target-specific manner. The complex regulation of Notch-dependent gene expression for LR asymmetry will be highlighted in this review.
Collapse
Affiliation(s)
- Yoichi Kato
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| |
Collapse
|
93
|
Insensitive is a corepressor for Suppressor of Hairless and regulates Notch signalling during neural development. EMBO J 2011; 30:3120-33. [PMID: 21765394 DOI: 10.1038/emboj.2011.218] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 06/08/2011] [Indexed: 11/08/2022] Open
Abstract
The Notch intracellular domain functions as a co-activator for the DNA-binding protein Suppressor of Hairless (Su(H)) to mediate myriad cell fate decisions. Notch pathway activity is balanced by transcriptional repression, mediated by Su(H) in concert with its Drosophila corepressor Hairless. We demonstrate that the Drosophila neural BEN-solo protein Insensitive (Insv) is a nuclear factor that inhibits Notch signalling during multiple peripheral nervous system cell fate decisions. Endogenous Insv was particularly critical when repressor activity of Su(H) was compromised. Reciprocally, ectopic Insv generated several Notch loss-of-function phenotypes, repressed most Notch targets in the E(spl)-C, and opposed Notch-mediated activation of an E(spl)m3-luc reporter. A direct role for Insv in transcriptional repression was indicated by binding of Insv to Su(H), and by strong chromatin immunoprecipitation of endogenous Insv to most E(spl)-C loci. Strikingly, ectopic Insv fully rescued sensory organ precursors in Hairless null clones, indicating that Insv can antagonize Notch independently of Hairless. These data shed first light on the in vivo function for a BEN-solo protein as an Su(H) corepressor in the Notch pathway regulating neural development.
Collapse
|
94
|
Wan Y, Chen W, Xing J, Tan J, Li B, Chen H, Lin Z, Chiang JH, Ramsey S. Transcriptome profiling reveals a novel role for trichostatin A in antagonizing histone chaperone Chz1 mediated telomere anti-silencing. FEBS Lett 2011; 585:2519-25. [DOI: 10.1016/j.febslet.2011.06.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Revised: 06/12/2011] [Accepted: 06/20/2011] [Indexed: 01/06/2023]
|
95
|
Maier D, Kurth P, Schulz A, Russell A, Yuan Z, Gruber K, Kovall RA, Preiss A. Structural and functional analysis of the repressor complex in the Notch signaling pathway of Drosophila melanogaster. Mol Biol Cell 2011; 22:3242-52. [PMID: 21737682 PMCID: PMC3164469 DOI: 10.1091/mbc.e11-05-0420] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Despite high conservation of the Notch pathway, its repression appears diverse between organisms. In Drosophila, a high-affinity complex forms between the CSL orthologue Su(H) and Hairless, which is analyzed in great detail in vitro and in vivo. Drosophila Hairless is shown to bind CBF1 and inhibit Notch transcriptional output in mammalian cells. In metazoans, the highly conserved Notch pathway drives cellular specification. On receptor activation, the intracellular domain of Notch assembles a transcriptional activator complex that includes the DNA-binding protein CSL, a composite of human C-promoter binding factor 1, Suppressor of Hairless of Drosophila melanogaster [Su(H)], and lin-12 and Glp-1 phenotype of Caenorhabditis elegans. In the absence of ligand, CSL represses Notch target genes. However, despite the structural similarity of CSL orthologues, repression appears largely diverse between organisms. Here we analyze the Notch repressor complex in Drosophila, consisting of the fly CSL protein, Su(H), and the corepressor Hairless, which recruits general repressor proteins. We show that the C-terminal domain of Su(H) is necessary and sufficient for forming a high-affinity complex with Hairless. Mutations in Su(H) that affect interactions with Notch and Mastermind have no effect on Hairless binding. Nonetheless, we demonstrate that Notch and Hairless compete for CSL in vitro and in cell culture. In addition, we identify a site in Hairless that is crucial for binding Su(H) and subsequently show that this Hairless mutant is strongly impaired, failing to properly assemble the repressor complex in vivo. Finally, we demonstrate Hairless-mediated inhibition of Notch signaling in a cell culture assay, which hints at a potentially similar repression mechanism in mammals that might be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Dieter Maier
- Institut für Genetik, Universität Hohenheim, 70593 Stuttgart, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Di Stefano L, Dyson NJ. The complex roles of histone demethylases in vivo. Cell Cycle 2011; 10:2049-50. [PMID: 21597322 DOI: 10.4161/cc.10.13.15758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
97
|
Chen M, Pereira-Smith OM, Tominaga K. Loss of the chromatin regulator MRG15 limits neural stem/progenitor cell proliferation via increased expression of the p21 Cdk inhibitor. Stem Cell Res 2011; 7:75-88. [PMID: 21621175 PMCID: PMC3130620 DOI: 10.1016/j.scr.2011.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 02/24/2011] [Accepted: 04/15/2011] [Indexed: 01/01/2023] Open
Abstract
Chromatin regulation is crucial for many biological processes such as transcriptional regulation, DNA replication, and DNA damage repair. We have found that it is also important for neural stem/progenitor cell (NSC) function and neurogenesis. Here, we demonstrate that expression of the cyclin-dependent kinase inhibitor p21 is specifically up-regulated in Mrg15 deficient NSCs. Knockdown of p21 expression by p21 shRNA results in restoration of cell proliferation. This indicates that p21 is directly involved in the growth defects observed in Mrg15 deficient NSCs. Activated p53 accumulates in Mrg15 deficient NSCs and this most likely accounts for the up-regulation of p21 expression in the cells. We observed decreased p53 and p21 levels and a concomitant increase in the percentage of BrdU positive cells in Mrg15 null cultures following expression of p53 shRNA. DNA damage foci, as indicated by immunostaining for γH2AX and 53BP1, are detectable in a sub-population of Mrg15 deficient NSC cultures under normal growing conditions and the majority of p21-positive cells are also positive for 53BP1 foci. Furthermore, Mrg15 deficient NSCs exhibit severe defects in DNA damage response following ionizing radiation. Our observations highlight the importance of chromatin regulation and DNA damage response in NSC function and maintenance.
Collapse
Affiliation(s)
- Meizhen Chen
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| | - Olivia M. Pereira-Smith
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| | - Kaoru Tominaga
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| |
Collapse
|
98
|
Abstract
Singapore grouper iridovirus (SGIV), a major pathogen of concern for grouper aquaculture, has a double-stranded DNA (dsDNA) genome with 162 predicted open reading frames, for which a total of 62 SGIV proteins have been identified. One of these, ORF158L, bears no sequence homology to any other known protein. Knockdown of orf158L using antisense morpholino oligonucleotides resulted in a significant decrease in virus yield in grouper embryonic cells. ORF158L was observed in nuclei and virus assembly centers of virus-infected cells. This observation led us to study the structure and function of ORF158L. The crystal structure determined at 2.2-Å resolution reveals that ORF158L partially exhibits a structural resemblance to the histone binding region of antisilencing factor 1 (Asf1), a histone H3/H4 chaperon, despite the fact that there is no significant sequence identity between the two proteins. Interactions of ORF158L with the histone H3/H4 complex and H3 were demonstrated by isothermal titration calorimetry (ITC) experiments. Subsequently, the results of ITC studies on structure-based mutants of ORF158L suggested Arg67 and Ala93 were key residues for histone H3 interactions. Moreover, a combination of approaches of ORF158L knockdown and isobaric tags/mass spectrometry for relative and absolute quantifications (iTRAQ) revealed that ORF158L may be involved in both the regulation and the expression of histone H3 and H3 methylation. Our present studies suggest that ORF158L may function as a histone H3 chaperon, enabling it to control host cellular gene expression and to facilitate viral replication.
Collapse
|
99
|
Mulligan P, Yang F, Di Stefano L, Ji JY, Ouyang J, Nishikawa JL, Toiber D, Kulkarni M, Wang Q, Najafi-Shoushtari SH, Mostoslavsky R, Gygi SP, Gill G, Dyson NJ, Näär AM. A SIRT1-LSD1 corepressor complex regulates Notch target gene expression and development. Mol Cell 2011; 42:689-99. [PMID: 21596603 PMCID: PMC3119599 DOI: 10.1016/j.molcel.2011.04.020] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 03/30/2011] [Accepted: 04/22/2011] [Indexed: 01/28/2023]
Abstract
Epigenetic regulation of gene expression by histone-modifying corepressor complexes is central to normal animal development. The NAD(+)-dependent deacetylase and gene repressor SIRT1 removes histone H4K16 acetylation marks and facilitates heterochromatin formation. However, the mechanistic contribution of SIRT1 to epigenetic regulation at euchromatic loci and whether it acts in concert with other chromatin-modifying activities to control developmental gene expression programs remain unclear. We describe here a SIRT1 corepressor complex containing the histone H3K4 demethylase LSD1/KDM1A and several other LSD1-associated proteins. SIRT1 and LSD1 interact directly and play conserved and concerted roles in H4K16 deacetylation and H3K4 demethylation to repress genes regulated by the Notch signaling pathway. Mutations in Drosophila SIRT1 and LSD1 orthologs result in similar developmental phenotypes and genetically interact with the Notch pathway in Drosophila. These findings offer new insights into conserved mechanisms of epigenetic gene repression and regulation of development by SIRT1 in metazoans.
Collapse
Affiliation(s)
- Peter Mulligan
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Fajun Yang
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Luisa Di Stefano
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
| | - Jun-Yuan Ji
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
| | - Jian Ouyang
- Tufts University School of Medicine, Department of Anatomy and Cellular Biology, Boston, MA 02111
| | - Joy L. Nishikawa
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Debra Toiber
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
| | - Madhura Kulkarni
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | | | - S. Hani Najafi-Shoushtari
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Raul Mostoslavsky
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Grace Gill
- Tufts University School of Medicine, Department of Anatomy and Cellular Biology, Boston, MA 02111
| | - Nicholas J. Dyson
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
| | - Anders M. Näär
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149, 13 Street, Charlestown, MA 02129
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
100
|
Zhu Y, Weng M, Yang Y, Zhang C, Li Z, Shen WH, Dong A. Arabidopsis homologues of the histone chaperone ASF1 are crucial for chromatin replication and cell proliferation in plant development. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2011; 66:443-55. [PMID: 21251110 DOI: 10.1111/j.1365-313x.2011.04504.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Anti-silencing function1 (ASF1) is an evolutionarily conserved histone chaperone. Studies in yeast and animals indicate that ASF1 proteins play important roles in various chromatin-based processes, including gene transcription, DNA replication and repair. While two genes encoding ASF1 homologues, AtASF1A and AtASF1B, are found in the Arabidopsis genome, their function has not been studied. Here we report that both AtASF1A and AtASF1B proteins bind histone H3, and are localized in the cytoplasm and the nucleus. Loss-of-function of either AtASF1A or AtASF1B did not show obvious defects, whereas simultaneous knockdown of both genes in the double mutant Atasf1ab drastically inhibited plant growth and caused abnormal vegetative and reproductive organ development. The Atasf1ab mutant plants exhibit cell number reduction, S-phase delay/arrest, and reduced polyploidy levels. Selective up-regulation of expression of a subset of genes, including those involved in S-phase checkpoints and the CYCB1;1 gene at the G₂-to-M transition, was observed in Atasf1ab. Furthermore, the Atasf1ab-triggered replication fork stalling constitutively activates the DNA damage checkpoint and repair genes, including ATM, ATR, PARP1 and PARP2 as well as several genes of the homologous recombination (HR) pathway but not genes of the non-homologous end joining (NHEJ) pathway. In spite of the activation of repair genes, an increased level of DNA damage was detected in Atasf1ab, suggesting that defects in the mutant largely exceed the available capacity of the repair machinery. Taken together, our study establishes crucial roles for the AtASF1A and AtASF1B genes in chromatin replication, maintenance of genome integrity and cell proliferation during plant development.
Collapse
Affiliation(s)
- Yan Zhu
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | | | | | | | | | | | | |
Collapse
|