1
|
Villamor-Payà M, Sanchiz-Calvo M, Smak J, Pais L, Sud M, Shankavaram U, Lovgren AK, Austin-Tse C, Ganesh VS, Gay M, Vilaseca M, Arauz-Garofalo G, Palenzuela L, VanNoy G, O’Donnell-Luria A, Stracker TH. De novo TLK1 and MDM1 mutations in a patient with a neurodevelopmental disorder and immunodeficiency. iScience 2024; 27:109984. [PMID: 38868186 PMCID: PMC11166698 DOI: 10.1016/j.isci.2024.109984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/08/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
The Tousled-like kinases 1 and 2 (TLK1/TLK2) regulate DNA replication, repair and chromatin maintenance. TLK2 variants underlie the neurodevelopmental disorder (NDD) 'Intellectual Disability, Autosomal Dominant 57' (MRD57), characterized by intellectual disability and microcephaly. Several TLK1 variants have been reported in NDDs but their functional significance is unknown. A male patient presenting with ID, seizures, global developmental delay, hypothyroidism, and primary immunodeficiency was determined to have a heterozygous TLK1 variant (c.1435C>G, p.Q479E), as well as a mutation in MDM1 (c.1197dupT, p.K400∗). Cells expressing TLK1 p.Q479E exhibited reduced cytokine responses and elevated DNA damage, but not increased radiation sensitivity or DNA repair defects. The TLK1 p.Q479E variant impaired kinase activity but not proximal protein interactions. Our study provides the first functional characterization of NDD-associated TLK1 variants and suggests that, such as TLK2, TLK1 variants may impact development in multiple tissues and should be considered in the diagnosis of rare NDDs.
Collapse
Affiliation(s)
- Marina Villamor-Payà
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- National Cancer Institute, Center for Cancer Research, Radiation Oncology Branch, Bethesda, MD 20892, USA
| | - María Sanchiz-Calvo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Jordann Smak
- National Cancer Institute, Center for Cancer Research, Radiation Oncology Branch, Bethesda, MD 20892, USA
| | - Lynn Pais
- Division of Genetics & Genomics, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Malika Sud
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Uma Shankavaram
- National Cancer Institute, Center for Cancer Research, Radiation Oncology Branch, Bethesda, MD 20892, USA
| | - Alysia Kern Lovgren
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christina Austin-Tse
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Vijay S. Ganesh
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Marina Gay
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Marta Vilaseca
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Gianluca Arauz-Garofalo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Lluís Palenzuela
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Grace VanNoy
- Division of Genetics & Genomics, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anne O’Donnell-Luria
- Division of Genetics & Genomics, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Travis H. Stracker
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- National Cancer Institute, Center for Cancer Research, Radiation Oncology Branch, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Streptonigrin at low concentration promotes heterochromatin formation. Sci Rep 2020; 10:3478. [PMID: 32103104 PMCID: PMC7044429 DOI: 10.1038/s41598-020-60469-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/04/2020] [Indexed: 11/26/2022] Open
Abstract
Heterochromatin is essential for regulating global gene transcription and protecting genome stability, and may play a role in tumor suppression. Drugs promoting heterochromatin are potential cancer therapeutics but very few are known. In order to identify drugs that can promote heterochromatin, we used a cell-based method and screened NCI drug libraries consisting of oncology drugs and natural compounds. Since heterochromatin is originally defined as intensely stained chromatin in the nucleus, we estimated heterochromatin contents of cells treated with different drugs by quantifying the fluorescence intensity of nuclei stained with Hoechst DNA dye. We used HeLa cells and screened 231 FDA-approved oncology and natural substance drugs included in two NCI drug libraries representing a variety of chemical structures. Among these drugs, streptonigrin most prominently caused an increase in Hoechst-stained nuclear fluorescence intensity. We further show that streptonigrin treated cells exhibit compacted DNA foci in the nucleus that co-localize with Heterochromatin Protein 1 alpha (HP1α), and exhibit an increase in total levels of the heterochromatin mark, H3K9me3. Interestingly, we found that streptonigrin promotes heterochromatin at a concentration as low as one nanomolar, and at this concentration there were no detectable effects on cell proliferation or viability. Finally, in line with a previous report, we found that streptonigrin inhibits STAT3 phosphorylation, raising the possibility that non-canonical STAT function may contribute to the effects of streptonigrin on heterochromatin. These results suggest that, at low concentrations, streptonigrin may primarily enhance heterochromatin formation with little toxic effects on cells, and therefore might be a good candidate for epigenetic cancer therapy.
Collapse
|
3
|
Dutta P, Zhang L, Zhang H, Peng Q, Montgrain PR, Wang Y, Song Y, Li J, Li WX. Unphosphorylated STAT3 in heterochromatin formation and tumor suppression in lung cancer. BMC Cancer 2020; 20:145. [PMID: 32087696 PMCID: PMC7036253 DOI: 10.1186/s12885-020-6649-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Aberrant JAK/STAT activation has been detected in many types of human cancers. The role of JAK/STAT activation in cancer has been mostly attributed to direct transcriptional regulation of target genes by phosphorylated STAT (pSTAT), while the unphosphorylated STAT (uSTAT) is believed to be dormant and reside in the cytoplasm. However, several studies have shown that uSTATs can be found in the nucleus. In addition, it has been shown that tissue-specific loss of STAT3 or STAT5 in mice promotes cancer growth in certain tissues, and thus these STAT proteins can act as tumor suppressors. However, no unifying mechanism has been shown for the tumor suppressor function of STATs to date. We have previously demonstrated a non-canonical mode of JAK/STAT signaling for Drosophila STAT and human STAT5A, where a fraction of uSTAT is in the nucleus and associated with Heterochromatin Protein 1 (HP1); STAT activation (by phosphorylation) causes its dispersal, leading to HP1 delocalization and heterochromatin loss. METHODS We used a combination of imaging, cell biological assays, and mouse xenografts to investigate the role of STAT3 in lung cancer development. RESULTS We found that uSTAT3 has a function in promoting heterochromatin formation in lung cancer cells, suppressing cell proliferation in vitro, and suppressing tumor growth in mouse xenografts. CONCLUSIONS Thus, uSTAT3 possesses noncanonical function in promoting heterochromatin formation, and the tumor suppressor function of STAT3 is likely attributable to the heterochromatin-promoting activity of uSTAT3 in the non-canonical JAK/STAT pathway.
Collapse
Affiliation(s)
- Pranabananda Dutta
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Lin Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Huijun Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qin Peng
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Phillippe R Montgrain
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, CA92037, USA
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yuanlin Song
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinghong Li
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Willis X Li
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
4
|
Zimmermann MT, Williams MM, Klee EW, Lomberk GA, Urrutia R. Modeling post-translational modifications and cancer-associated mutations that impact the heterochromatin protein 1α-importin α heterodimers. Proteins 2019; 87:904-916. [PMID: 31152607 PMCID: PMC6790107 DOI: 10.1002/prot.25752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/27/2019] [Indexed: 12/27/2022]
Abstract
Heterochromatin protein 1α (HP1α) is a protein that mediates cancer-associated processes in the cell nucleus. Proteomic experiments, reported here, demonstrate that HP1α complexes with importin α (IMPα), a protein necessary for its nuclear transport. This data is congruent with Simple Linear Motif (SLiM) analyses that identify an IMPα-binding motif within the linker that joins the two globular domains of this protein. Using molecular modeling and dynamics simulations, we develop a model of the IMPα-HP1α complex and investigate the impact of phosphorylation and genomic variants on their interaction. We demonstrate that phosphorylation of the HP1α linker likely regulates its association with IMPα, which has implications for HP1α access to the nucleus, where it functions. Cancer-associated genomic variants do not abolish the interaction of HP1α but instead lead to rearrangements where the variant proteins maintain interaction with IMPα, but with less specificity. Combined, this new mechanistic insight bears biochemical, cell biological, and biomedical relevance.
Collapse
Affiliation(s)
- Michael T. Zimmermann
- Bioinformatics Research and Development Laboratory, and Precision Medicine Simulation Unit, Genomic Science and Precision Medicine Center (GSPMC)Medical College of WisconsinMilwaukeeWisconsin
- Clinical and Translational Sciences InstituteMedical College of WisconsinMilwaukeeWisconsin
| | - Monique M. Williams
- Department of BiochemistryMayo ClinicRochesterMinnesota
- Division of Biomedical Statistics and InformaticsMayo ClinicRochesterMinnesota
| | - Eric W. Klee
- Department of BiochemistryMayo ClinicRochesterMinnesota
- Division of Biomedical Statistics and InformaticsMayo ClinicRochesterMinnesota
| | - Gwen A. Lomberk
- Division of Research, Department of SurgeryMedical College of WisconsinMilwaukeeWisconsin
- Department of Pharmacology and ToxicologyMedical College of WisconsinMilwaukeeWisconsin
- Genomic Science and Precision Medicine Center (GSPMC)Medical College of WisconsinMilwaukeeWisconsin
| | - Raul Urrutia
- Division of Research, Department of SurgeryMedical College of WisconsinMilwaukeeWisconsin
- Genomic Science and Precision Medicine Center (GSPMC)Medical College of WisconsinMilwaukeeWisconsin
- Department of BiochemistryMedical College of WisconsinMilwaukeeWisconsin
| |
Collapse
|
5
|
PHF2 histone demethylase prevents DNA damage and genome instability by controlling cell cycle progression of neural progenitors. Proc Natl Acad Sci U S A 2019; 116:19464-19473. [PMID: 31488723 DOI: 10.1073/pnas.1903188116] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Histone H3 lysine 9 methylation (H3K9me) is essential for cellular homeostasis; however, its contribution to development is not well established. Here, we demonstrate that the H3K9me2 demethylase PHF2 is essential for neural progenitor proliferation in vitro and for early neurogenesis in the chicken spinal cord. Using genome-wide analyses and biochemical assays we show that PHF2 controls the expression of critical cell cycle progression genes, particularly those related to DNA replication, by keeping low levels of H3K9me3 at promoters. Accordingly, PHF2 depletion induces R-loop accumulation that leads to extensive DNA damage and cell cycle arrest. These data reveal a role of PHF2 as a guarantor of genome stability that allows proper expansion of neural progenitors during development.
Collapse
|
6
|
Roworth AP, Carr SM, Liu G, Barczak W, Miller RL, Munro S, Kanapin A, Samsonova A, La Thangue NB. Arginine methylation expands the regulatory mechanisms and extends the genomic landscape under E2F control. SCIENCE ADVANCES 2019; 5:eaaw4640. [PMID: 31249870 PMCID: PMC6594773 DOI: 10.1126/sciadv.aaw4640] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/22/2019] [Indexed: 06/09/2023]
Abstract
E2F is a family of master transcription regulators involved in mediating diverse cell fates. Here, we show that residue-specific arginine methylation (meR) by PRMT5 enables E2F1 to regulate many genes at the level of alternative RNA splicing, rather than through its classical transcription-based mechanism. The p100/TSN tudor domain protein reads the meR mark on chromatin-bound E2F1, allowing snRNA components of the splicing machinery to assemble with E2F1. A large set of RNAs including spliced variants associate with E2F1 by virtue of the methyl mark. By focusing on the deSUMOylase SENP7 gene, which we identified as an E2F target gene, we establish that alternative splicing is functionally important for E2F1 activity. Our results reveal an unexpected consequence of arginine methylation, where reader-writer interplay widens the mechanism of control by E2F1, from transcription factor to regulator of alternative RNA splicing, thereby extending the genomic landscape under E2F1 control.
Collapse
Affiliation(s)
- Alice Poppy Roworth
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Simon Mark Carr
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Geng Liu
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Wojciech Barczak
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Rebecca Louise Miller
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Shonagh Munro
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Alexander Kanapin
- Institute of Translational Biomedicine, St. Petersburg University, St. Petersburg 199034, Russia
| | - Anastasia Samsonova
- Institute of Translational Biomedicine, St. Petersburg University, St. Petersburg 199034, Russia
| | - Nicholas B. La Thangue
- Laboratory of Cancer Biology, Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK
| |
Collapse
|
7
|
López-Ozuna VM, Hachim IY, Hachim MY, Lebrun JJ, Ali S. Prolactin modulates TNBC aggressive phenotype limiting tumorigenesis. Endocr Relat Cancer 2019; 26:321-337. [PMID: 30640712 DOI: 10.1530/erc-18-0523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/14/2019] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) accounts for ~20% of all breast cancer cases. The management of TNBC represents a challenge due to its aggressive phenotype, heterogeneity and lack of targeted therapy. Loss of cell differentiation and enrichment with breast cancer stem-like cells (BCSC) are features of TNBC contributing to its aggressive nature. Here, we found that treatment of TNBC cells with PRL significantly depletes the highly tumorigenic BCSC subpopulations CD44+/CD24- and ALDH+ and differentiates them to the least tumorigenic CD44-/CD24- and ALDH- phenotype with limited tumorsphere formation and self-renewal capacities. Importantly, we found PRL to induce a heterochromatin phenotype marked by histone H3 lysine 9 trimethylation (H3K9me3) and accompanied by ultra-structural cellular architecture associated with differentiation and senescence rendering the cells refractory to growth signals. Crucially, we found PRL to mediate these effects in vivo in a pre-clinical animal xenograft of TNBC controlling tumor growth. These results reveal that the lactogenic hormone PRL may exert its anti-tumorigenic effects on TNBC through cellular reprogramming indicative of differentiation resulting in the depletion of BCSCs and restricting tumorigenesis.
Collapse
Affiliation(s)
- Vanessa M López-Ozuna
- Department of Medicine, Cancer Research Program, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| | - Ibrahim Y Hachim
- Department of Medicine, Cancer Research Program, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| | - Mahmood Y Hachim
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Jean-Jacques Lebrun
- Department of Medicine, Cancer Research Program, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| | - Suhad Ali
- Department of Medicine, Cancer Research Program, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| |
Collapse
|
8
|
Lin FM, Kumar S, Ren J, Karami S, Bahnassy S, Li Y, Zheng X, Wang J, Bawa-Khalfe T. SUMOylation of HP1α supports association with ncRNA to define responsiveness of breast cancer cells to chemotherapy. Oncotarget 2017; 7:30336-49. [PMID: 27107417 PMCID: PMC5058684 DOI: 10.18632/oncotarget.8733] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/03/2016] [Indexed: 01/19/2023] Open
Abstract
Epigenetic reprogramming allows cancer cells to bypass normal checkpoints and potentiate aberrant proliferation. Several chromatin regulators are subject to reversible SUMO-modification but little is known about how SUMOylation of chromatin-remodelers modulates the cancer epigenome. Recently, we demonstrated that SUMO-protease SENP7L is upregulated in aggressive BCa and maintains hypoSUMOylated heterochromatin protein 1-α (HP1α). Canonical models define HP1α as a "reader" of repressive H3K9m3 marks that supports constitutive heterochromatin. It is unclear how SUMOylation affects HP1α function in BCa cells. This report shows HP1α SUMO-dynamics are closely regulated in a complex with SENP7L and SUMO-E3 Polycomb-2 (PC2/CBX4). This complex accumulates at H3K9m3 sites, hypoSUMOylates HP1α and PC2, and reduces PC2's SUMO-E3 activity. HyperSUMO conditions cause complex dissociation, SUMOylation of PC2 and HP1α, and recruitment of SUMOylated HP1α to multiple DNA-repair genes including Rad51C. SUMOylated HP1α's enrichment at euchromatin requires chromatin-bound non-coding RNA (ncRNA), reduces Rad51C protein, and increases DNA-breaks in BCa cells. Hence, HP1α SUMOylation and consistently low SENP7L increase efficacy of DNA-damaging chemotherapeutic agents. BCa patients on chemotherapy that express low SENP7L exhibit greater survival rates than patients with high SENP7L. Collectively, these studies suggest that SUMOylated HP1α is a critical epigenetic-regulator of DNA-repair in BCa that could define chemotherapy responsiveness.
Collapse
Affiliation(s)
- Feng-Ming Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Santosh Kumar
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Jing Ren
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Samaneh Karami
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Shaymaa Bahnassy
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Yue Li
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiaofeng Zheng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tasneem Bawa-Khalfe
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| |
Collapse
|
9
|
Ma Y, Buttitta L. Chromatin organization changes during the establishment and maintenance of the postmitotic state. Epigenetics Chromatin 2017; 10:53. [PMID: 29126440 PMCID: PMC5681785 DOI: 10.1186/s13072-017-0159-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023] Open
Abstract
Background Genome organization changes during development as cells differentiate. Chromatin motion becomes increasingly constrained and heterochromatin clusters as cells become restricted in their developmental potential. These changes coincide with slowing of the cell cycle, which can also influence chromatin organization and dynamics. Terminal differentiation is often coupled with permanent exit from the cell cycle, and existing data suggest a close relationship between a repressive chromatin structure and silencing of the cell cycle in postmitotic cells. Heterochromatin clustering could also contribute to stable gene repression to maintain terminal differentiation or cell cycle exit, but whether clustering is initiated by differentiation, cell cycle changes, or both is unclear. Here we examine the relationship between chromatin organization, terminal differentiation and cell cycle exit. Results We focused our studies on the Drosophila wing, where epithelial cells transition from active proliferation to a postmitotic state in a temporally controlled manner. We find there are two stages of G0 in this tissue, a flexible G0 period where cells can be induced to reenter the cell cycle under specific genetic manipulations and a state we call “robust,” where cells become strongly refractory to cell cycle reentry. Compromising the flexible G0 by driving ectopic expression of cell cycle activators causes a global disruption of the clustering of heterochromatin-associated histone modifications such as H3K27 trimethylation and H3K9 trimethylation, as well as their associated repressors, Polycomb and heterochromatin protein 1 (HP1). However, this disruption is reversible. When cells enter a robust G0 state, even in the presence of ectopic cell cycle activity, clustering of heterochromatin-associated modifications is restored. If cell cycle exit is bypassed, cells in the wing continue to terminally differentiate, but heterochromatin clustering is severely disrupted. Heterochromatin-dependent gene silencing does not appear to be required for cell cycle exit, as compromising the H3K27 methyltransferase Enhancer of zeste, and/or HP1 cannot prevent the robust cell cycle exit, even in the face of normally oncogenic cell cycle activities. Conclusions Heterochromatin clustering during terminal differentiation is a consequence of cell cycle exit, rather than differentiation. Compromising heterochromatin-dependent gene silencing does not disrupt cell cycle exit. Electronic supplementary material The online version of this article (10.1186/s13072-017-0159-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yiqin Ma
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Laura Buttitta
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
10
|
Moyon S, Liang J, Casaccia P. Epigenetics in NG2 glia cells. Brain Res 2016; 1638:183-198. [PMID: 26092401 PMCID: PMC4683112 DOI: 10.1016/j.brainres.2015.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/11/2015] [Accepted: 06/02/2015] [Indexed: 12/16/2022]
Abstract
The interplay of transcription and epigenetic marks is essential for oligodendrocyte progenitor cell (OPC) proliferation and differentiation during development. Here, we review the recent advances in this field and highlight mechanisms of transcriptional repression and activation involved in OPC proliferation, differentiation and plasticity. We also describe how dysregulation of these epigenetic events may affect demyelinating disorders, and consider potential ways to manipulate NG2 cell behavior through modulation of the epigenome. This article is part of a Special Issue entitled SI:NG2-glia(Invited only).
Collapse
Affiliation(s)
- Sarah Moyon
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jialiang Liang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Patrizia Casaccia
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
11
|
Yang M, Yuan ZM. A novel role of PRR14 in the regulation of skeletal myogenesis. Cell Death Dis 2015; 6:e1734. [PMID: 25906157 PMCID: PMC4650536 DOI: 10.1038/cddis.2015.103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/11/2015] [Accepted: 03/13/2015] [Indexed: 12/11/2022]
Abstract
Dysregulation of genes involved in organizing and maintaining nuclear structures, such as SYNE1, SYNE2, TREM43, EMD and LMNA is frequently associated with diverse diseases termed laminopathies, which often affect the muscle tissue. The PRR14 protein was recently reported to tether heterochromatin to nuclear lamina but its function remains largely unknown. Here, we present several lines of evidence demonstrating a critical role of PRR14 in regulation of myoblast differentiation. We found that Prr14 expression was upregulated during skeletal myogenesis. Knockdown of Prr14 impeded, whereas overexpression of PRR14 enhanced C2C12 differentiation. The pro-myogenesis activity of PRR14 seemed to correlate with its ability to support cell survival and to maintain the stability and structure of lamin A/C. In addition, PRR14 stimulated the activity of MyoD via binding to heterochromatin protein 1 alpha (HP1α). The results altogether support a model in which PRR14 promotes skeletal myogenesis via supporting nuclear lamina structure and enhancing the activity of MyoD.
Collapse
Affiliation(s)
- M Yang
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Z-M Yuan
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
12
|
Oyama K, El-Nachef D, Zhang Y, Sdek P, MacLellan WR. Epigenetic regulation of cardiac myocyte differentiation. Front Genet 2014; 5:375. [PMID: 25408700 PMCID: PMC4219506 DOI: 10.3389/fgene.2014.00375] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 10/07/2014] [Indexed: 12/04/2022] Open
Abstract
Cardiac myocytes (CMs) proliferate robustly during fetal life but withdraw permanently from the cell cycle soon after birth and undergo terminal differentiation. This cell cycle exit is associated with the upregulation of a host of adult cardiac-specific genes. The vast majority of adult CMs (ACMs) do not reenter cell cycle even if subjected to mitogenic stimuli. The basis for this irreversible cell cycle exit is related to the stable silencing of cell cycle genes specifically involved in the progression of G2/M transition and cytokinesis. Studies have begun to clarify the molecular basis for this stable gene repression and have identified epigenetic and chromatin structural changes in this process. In this review, we summarize the current understanding of epigenetic regulation of CM cell cycle and cardiac-specific gene expression with a focus on histone modifications and the role of retinoblastoma family members.
Collapse
Affiliation(s)
- Kyohei Oyama
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington Seattle, WA, USA
| | - Danny El-Nachef
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington Seattle, WA, USA
| | - Yiqiang Zhang
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington Seattle, WA, USA
| | - Patima Sdek
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington Seattle, WA, USA
| | - W Robb MacLellan
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington Seattle, WA, USA
| |
Collapse
|
13
|
Rampal R, Levine RL. A primer on genomic and epigenomic alterations in the myeloproliferative neoplasms. Best Pract Res Clin Haematol 2014; 27:83-93. [DOI: 10.1016/j.beha.2014.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/11/2014] [Indexed: 01/13/2023]
|
14
|
A novel activator of CBP/p300 acetyltransferases promotes neurogenesis and extends memory duration in adult mice. J Neurosci 2013; 33:10698-712. [PMID: 23804093 DOI: 10.1523/jneurosci.5772-12.2013] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although the brain functions of specific acetyltransferases such as the CREB-binding protein (CBP) and p300 have been well documented using mutant transgenic mice models, studies based on their direct pharmacological activation are still missing due to the lack of cell-permeable activators. Here we present a small-molecule (TTK21) activator of the histone acetyltransferases CBP/p300, which, when conjugated to glucose-based carbon nanosphere (CSP), passed the blood-brain barrier, induced no toxicity, and reached different parts of the brain. After intraperitoneal administration in mice, CSP-TTK21 significantly acetylated histones in the hippocampus and frontal cortex. Remarkably, CSP-TTK21 treatment promoted the formation of long and highly branched doublecortin-positive neurons in the subgranular zone of the dentate gyrus and reduced BrdU incorporation, suggesting that CBP/p300 activation favors maturation and differentiation of adult neuronal progenitors. In addition, mRNA levels of the neuroD1 differentiation marker and BDNF, a neurotrophin required for the terminal differentiation of newly generated neurons, were both increased in the hippocampus concomitantly with an enrichment of acetylated-histone on their proximal promoter. Finally, we found that CBP/p300 activation during a spatial training, while not improving retention of a recent memory, resulted in a significant extension of memory duration. This report is the first evidence for CBP/p300-mediated histone acetylation in the brain by an activator molecule, which has beneficial implications for the brain functions of adult neurogenesis and long-term memory. We propose that direct stimulation of acetyltransferase function could be useful in terms of therapeutic options for brain diseases.
Collapse
|
15
|
Zeng A, Li YQ, Wang C, Han XS, Li G, Wang JY, Li DS, Qin YW, Shi Y, Brewer G, Jing Q. Heterochromatin protein 1 promotes self-renewal and triggers regenerative proliferation in adult stem cells. ACTA ACUST UNITED AC 2013; 201:409-25. [PMID: 23629965 PMCID: PMC3639387 DOI: 10.1083/jcb.201207172] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adult stem cells (ASCs) capable of self-renewal and differentiation confer the potential of tissues to regenerate damaged parts. Epigenetic regulation is essential for driving cell fate decisions by rapidly and reversibly modulating gene expression programs. However, it remains unclear how epigenetic factors elicit ASC-driven regeneration. In this paper, we report that an RNA interference screen against 205 chromatin regulators identified 12 proteins essential for ASC function and regeneration in planarians. Surprisingly, the HP1-like protein SMED-HP1-1 (HP1-1) specifically marked self-renewing, pluripotent ASCs, and HP1-1 depletion abrogated self-renewal and promoted differentiation. Upon injury, HP1-1 expression increased and elicited increased ASC expression of Mcm5 through functional association with the FACT (facilitates chromatin transcription) complex, which consequently triggered proliferation of ASCs and initiated blastema formation. Our observations uncover an epigenetic network underlying ASC regulation in planarians and reveal that an HP1 protein is a key chromatin factor controlling stem cell function. These results provide important insights into how epigenetic mechanisms orchestrate stem cell responses during tissue regeneration.
Collapse
Affiliation(s)
- An Zeng
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, 200025 Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mahajan K, Mahajan NP. WEE1 tyrosine kinase, a novel epigenetic modifier. Trends Genet 2013; 29:394-402. [PMID: 23537585 DOI: 10.1016/j.tig.2013.02.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/26/2012] [Accepted: 02/25/2013] [Indexed: 11/30/2022]
Abstract
The cell cycle requires cells to duplicate their chromatin, DNA, and histones, while retaining a subset of epigenetic marks, in a highly coordinated manner. The WEE1 kinase was identified as an important regulator during S phase, preventing entry into mitosis until DNA replication has been completed. Interestingly, WEE1 has also emerged as a key player in regulating histone synthesis. It phosphorylates histone H2B at tyrosine 37 in the nucleosomes found upstream of the histone gene cluster, and this suppresses histone transcription in late S phase. These observations highlight a dual role for WEE1 as both a mitotic gatekeeper and a surveyor of chromatin synthesis, providing a direct link between epigenetics and cell-cycle progression. Importantly, this link has implications for the design of novel epigenetic inhibitors targeting cancers that display elevated expression of this kinase.
Collapse
Affiliation(s)
- Kiran Mahajan
- Drug Discovery Department, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | | |
Collapse
|
17
|
Sdek P, Oyama K, Angelis E, Chan SS, Schenke-Layland K, MacLellan WR. Epigenetic regulation of myogenic gene expression by heterochromatin protein 1 alpha. PLoS One 2013; 8:e58319. [PMID: 23505487 PMCID: PMC3594309 DOI: 10.1371/journal.pone.0058319] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 02/02/2013] [Indexed: 12/17/2022] Open
Abstract
Heterochromatin protein 1 (HP1) is an essential heterochromatin-associated protein typically involved in the epigenetic regulation of gene silencing. However, recent reports have demonstrated that HP1 can also activate gene expression in certain contexts including differentiation. To explore the role of each of the three mammalian HP1 family members (α, β and γ) in skeletal muscle, their expression was individually disrupted in differentiating skeletal myocytes. Among the three isoforms of HP1, HP1α was specifically required for myogenic gene expression in myoblasts only. Knockdown of HP1α led to a defect in transcription of skeletal muscle-specific genes including Lbx1, MyoD and myogenin. HP1α binds to the genomic region of myogenic genes and depletion of HP1α results in a paradoxical increase in histone H3 lysine 9 trimethylation (H3K9me3) at these sites. JHDM3A, a H3K9 demethylase also binds to myogenic gene's genomic regions in myoblasts in a HP1α-dependent manner. JHDM3A interacts with HP1α and knockdown of JHDM3A in myoblasts recapitulates the decreased myogenic gene transcription seen with HP1α depletion. These results propose a novel mechanism for HP1α-dependent gene activation by interacting with the demethylase JHDM3A and that HP1α is required for maintenance of myogenic gene expression in myoblasts.
Collapse
Affiliation(s)
- Patima Sdek
- Departments of Medicine/Cardiology, Center for Cardiovascular Biology, Institute for Stem Cell Research, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kyohei Oyama
- Departments of Medicine/Cardiology, Center for Cardiovascular Biology, Institute for Stem Cell Research, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ekaterini Angelis
- Departments of Medicine/Cardiology, Center for Cardiovascular Biology, Institute for Stem Cell Research, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Shing S. Chan
- Departments of Medicine/Cardiology, Center for Cardiovascular Biology, Institute for Stem Cell Research, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Katja Schenke-Layland
- Departments of Medicine/Cardiology, Center for Cardiovascular Biology, Institute for Stem Cell Research, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - W. Robb MacLellan
- Departments of Medicine/Cardiology, Center for Cardiovascular Biology, Institute for Stem Cell Research, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
18
|
Milosevic JD, Kralovics R. Genetic and epigenetic alterations of myeloproliferative disorders. Int J Hematol 2012; 97:183-97. [PMID: 23233154 DOI: 10.1007/s12185-012-1235-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 11/27/2012] [Accepted: 11/27/2012] [Indexed: 01/07/2023]
Abstract
The classical BCR-ABL negative myeloproliferative neoplasms (MPN) polycythemia vera, essential thrombocythemia, and primary myelofibrosis are clonal hematopoietic disorders characterized by excessive production of terminally differentiated myeloid cells. In MPN patients, the disease can progress to secondary myelofibrosis or acute myeloid leukemia. Clonal hematopoiesis, disease phenotype, and progression are caused by somatically acquired genetic lesions of genes involved in cytokine signaling, RNA splicing, as well as epigenetic regulation. This review provides an overview of point mutations and cytogenetic lesions associated with MPN and addresses the role of these somatic lesions in MPN disease progression.
Collapse
Affiliation(s)
- Jelena D Milosevic
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT25.3, 1090 Vienna, Austria
| | | |
Collapse
|
19
|
Differential expression of SUMO-specific protease 7 variants regulates epithelial-mesenchymal transition. Proc Natl Acad Sci U S A 2012; 109:17466-71. [PMID: 23045645 DOI: 10.1073/pnas.1209378109] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Two Sentrin/small ubiquitin-like modifier (SUMO)-specific protease 7 (SENP7) variants are naturally expressed in breast epithelia. Breast cancer (BCa) onset down-regulates the short SENP7 splice variant (SENP7S) and enhances the long transcript (SENP7L). Here, we show that SENP7L induction promotes gene expression profiles that favor aberrant proliferation and initiate epithelial-mesenchymal transition (EMT). SENP7L exhibits an interaction domain for the epigenetic remodeler heterochromatin protein 1 α (HP1α) and isopeptidase activity against SUMO-modified HP1α. Loss of this interaction domain, as observed with SENP7S, favors HP1α SUMOylation. SUMOylated HP1α is enriched at E2F-responsive and mesenchymal gene promoters, silences transcription of these genes, and promotes cellular senescence. Elevated SENP7L renders HP1α hypo-SUMOylated, which relieves transcriptional repression of the same genes and concurrently decreases transcription of epithelial-promoting genes via an HP1α-independent mechanism. Consequently, SENP7L levels correlate with EMT, motility, and invasiveness of BCa cells. Stable knockdown of elevated SENP7L levels lessens the dissemination of highly metastatic BCa cells to the lungs from primary implantation sites in in vivo studies. Thus, differential splicing of the SENP7 regulates either tumor suppression or progression.
Collapse
|
20
|
Anand S, Huntly BJP. Disordered signaling in myeloproliferative neoplasms. Hematol Oncol Clin North Am 2012; 26:1017-35. [PMID: 23009935 DOI: 10.1016/j.hoc.2012.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The human myeloproliferative neoplasms (MPN) have long been associated with abnormal responses to cytokines and activation of signaling pathways, although the exact molecular mechanisms underlying these observations were unknown. This situation altered with the discovery of the JAK2 V617F, which presaged the ongoing description of further mutations predicted to activate canonical signaling pathways in MPN. This article covers the nature of these mutations and summarizes functional experiments in model systems and in human MPN cells to define the signaling pathways altered and how these drive and determine the MPN cellular phenotype. Also discussed are recently described, novel noncanonical signaling pathways to chromatin predicted to alter gene transcription more directly and to also contribute to the MPN phenotype.
Collapse
Affiliation(s)
- Shubha Anand
- Department of Haematology, Cambridge Institute of Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | | |
Collapse
|
21
|
Meister P, Schott S, Bedet C, Xiao Y, Rohner S, Bodennec S, Hudry B, Molin L, Solari F, Gasser SM, Palladino F. Caenorhabditis elegans Heterochromatin protein 1 (HPL-2) links developmental plasticity, longevity and lipid metabolism. Genome Biol 2011; 12:R123. [PMID: 22185090 PMCID: PMC3334618 DOI: 10.1186/gb-2011-12-12-r123] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 11/30/2011] [Accepted: 12/20/2011] [Indexed: 01/23/2023] Open
Abstract
Background Heterochromatin protein 1 (HP1) family proteins have a well-characterized role in heterochromatin packaging and gene regulation. Their function in organismal development, however, is less well understood. Here we used genome-wide expression profiling to assess novel functions of the Caenorhabditis elegans HP1 homolog HPL-2 at specific developmental stages. Results We show that HPL-2 regulates the expression of germline genes, extracellular matrix components and genes involved in lipid metabolism. Comparison of our expression data with HPL-2 ChIP-on-chip profiles reveals that a significant number of genes up- and down-regulated in the absence of HPL-2 are bound by HPL-2. Germline genes are specifically up-regulated in hpl-2 mutants, consistent with the function of HPL-2 as a repressor of ectopic germ cell fate. In addition, microarray results and phenotypic analysis suggest that HPL-2 regulates the dauer developmental decision, a striking example of phenotypic plasticity in which environmental conditions determine developmental fate. HPL-2 acts in dauer at least partly through modulation of daf-2/IIS and TGF-β signaling pathways, major determinants of the dauer program. hpl-2 mutants also show increased longevity and altered lipid metabolism, hallmarks of the long-lived, stress resistant dauers. Conclusions Our results suggest that the worm HP1 homologue HPL-2 may coordinately regulate dauer diapause, longevity and lipid metabolism, three processes dependent on developmental input and environmental conditions. Our findings are of general interest as a paradigm of how chromatin factors can both stabilize development by buffering environmental variation, and guide the organism through remodeling events that require plasticity of cell fate regulation.
Collapse
Affiliation(s)
- Peter Meister
- Laboratory of Molecular and Cellular Biology, CNRS, Université de Lyon, Ecole Normale Supérieure, Lyon Cedex 07, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Sdek P, Zhao P, Wang Y, Huang CJ, Ko CY, Butler PC, Weiss JN, Maclellan WR. Rb and p130 control cell cycle gene silencing to maintain the postmitotic phenotype in cardiac myocytes. ACTA ACUST UNITED AC 2011; 194:407-23. [PMID: 21825075 PMCID: PMC3153646 DOI: 10.1083/jcb.201012049] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The mammalian heart loses its regenerative potential soon after birth. Adult cardiac myocytes (ACMs) permanently exit the cell cycle, and E2F-dependent genes are stably silenced, although the underlying mechanism is unclear. Heterochromatin, which silences genes in many biological contexts, accumulates with cardiac differentiation. H3K9me3, a histone methylation characteristic of heterochromatin, also increases in ACMs and at E2F-dependent promoters. We hypothesize that genes relevant for cardiac proliferation are targeted to heterochromatin by retinoblastoma (Rb) family members interacting with E2F transcription factors and recruiting heterochromatin protein 1 (HP1) proteins. To test this hypothesis, we created cardiac-specific Rb and p130 inducible double knockout (IDKO) mice. IDKO ACMs showed a decrease in total heterochromatin, and cell cycle genes were derepressed, leading to proliferation of ACMs. Although Rb/p130 deficiency had no effect on total H3K9me3 levels, recruitment of HP1-γ to promoters was lost. Depleting HP1-γ up-regulated proliferation-promoting genes in ACMs. Thus, Rb and p130 have overlapping roles in maintaining the postmitotic state of ACMs through their interaction with HP1-γ to direct heterochromatin formation and silencing of proliferation-promoting genes.
Collapse
Affiliation(s)
- Patima Sdek
- Cardiovascular Research Laboratory, Department of Medicine and Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Reuther GW. Recurring mutations in myeloproliferative neoplasms alter epigenetic regulation of gene expression. Am J Cancer Res 2011; 1:752-762. [PMID: 22016825 PMCID: PMC3195930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 05/25/2011] [Indexed: 05/31/2023] Open
Abstract
The prevalence of activating JAK2 mutations in myeloproliferative neoplasms (MPNs) suggests that aberrant gene expression due to deregulated signaling of the JAK2/STAT pathway plays an important role in the etiology of these diseases. While likely true, recent work has uncovered some fascinating new insights into both the function of mutationally-activated JAK2 as well as other mutated gene products in MPNs, and how these mutations may affect gene expression. In addition to being a cytoplasmic tyrosine kinase that relays signals from cytokine receptors, activated JAK2 can also function in the nucleus where it phosphorylates histones and deregulates binding of the transcriptional repressor HP1α. In addition, MPN-associated JAK2 mutants phosphorylate PRMT5 and inhibit its histone methyltransferase activity. Thus, in addition to the classical JAK/STAT pathway, JAK2 activating mutations in MPNs may deregulate gene expression by altering epigenetic mechanisms. Studies aimed at identifying the biochemical ramifications of other recurring MPN mutations also suggest deregulated epigenetic modifications may be important in MPN formation. Mutant TET2, as well as IDH1/2, impairs the hydroxylation of methylcytosine, thus affecting DNA methylation. Likewise, mutations in EZH2, a histone methyl transferase, ASXL1, which functions in chromatin modifier complexes, and the DNA methyltransferase DNMT3A, appear to inactivate the functions of these proteins toward regulating the epigenetic state of genes. Thus, it is likely that the control of gene expression by epigenetic mechanisms plays an important role in MPNs, since multiple recurring mutations in MPNs alter normal epigenetic regulatory mechanisms.
Collapse
Affiliation(s)
- Gary W Reuther
- Department of Molecular Oncology, Moffitt Cancer Center and Research Institute USA
| |
Collapse
|
24
|
Tell R, Rivera CA, Eskra J, Taglia LN, Blunier A, Wang QT, Benya RV. Gastrin-releasing peptide signaling alters colon cancer invasiveness via heterochromatin protein 1Hsβ. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:672-8. [PMID: 21281799 DOI: 10.1016/j.ajpath.2010.10.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 09/09/2010] [Accepted: 10/01/2010] [Indexed: 01/08/2023]
Abstract
Epithelial cells lining the adult colon do not normally express gastrin-releasing peptide (GRP) or its receptor (GRPR). In contrast, GRP/GRPR can be aberrantly expressed in colon cancer where they are associated with improved patient survival rates. However, the mechanism of action whereby these proteins mediate their beneficial effects is not known. Heterochromatin protein 1 is an epigenetic modifier of gene transcription for which three different isoforms exist in humans: HP1(Hsα), HP1(Hsβ), and HP1(Hsγ). In breast cancer and melanoma, respectively, HP1(Hsα) and HP1(Hsβ) have been shown to modulate the aggressiveness of tumor cells in vivo. In contrast, the role of HP1 in colon cancer has not been elucidated, and a mechanism of regulating the expression of any HP1 isoform in any context has not yet been identified. In this article we demonstrate that abrogating GRP/GRPR signaling specifically down-regulates HP1(Hsβ) expression and that inhibiting GRPR signaling, or ablating HP1(Hsβ) expression, increases colon cancer cell invasiveness in vitro. These findings identify for the first time a signaling pathway regulating heterochromatin protein expression and suggest a mechanism whereby aberrantly expressed GRPR might alter the outcome of patients with colorectal cancer.
Collapse
Affiliation(s)
- Robert Tell
- Departments of Medicine and Biological Sciences, UIC Cancer Center, University of Illinois at Chicago, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Affiliation(s)
- Prim B Singh
- Division of Immunoepigenetics, Department of Immunology and Cell Biology, Research Center Borstel, D-23845 Borstel, Germany.
| | | |
Collapse
|
26
|
Mosch K, Franz H, Soeroes S, Singh PB, Fischle W. HP1 recruits activity-dependent neuroprotective protein to H3K9me3 marked pericentromeric heterochromatin for silencing of major satellite repeats. PLoS One 2011; 6:e15894. [PMID: 21267468 PMCID: PMC3022755 DOI: 10.1371/journal.pone.0015894] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 11/29/2010] [Indexed: 11/19/2022] Open
Abstract
H3 lysine 9 trimethylation (H3K9me3) is a histone posttranslational modification (PTM) that has emerged as hallmark of pericentromeric heterochromatin. This constitutive chromatin domain is composed of repetitive DNA elements, whose transcription is differentially regulated. Mammalian cells contain three HP1 proteins, HP1α, HP1β and HP1γ These have been shown to bind to H3K9me3 and are thought to mediate the effects of this histone PTM. However, the mechanisms of HP1 chromatin regulation and the exact functional role at pericentromeric heterochromatin are still unclear. Here, we identify activity-dependent neuroprotective protein (ADNP) as an H3K9me3 associated factor. We show that ADNP does not bind H3K9me3 directly, but that interaction is mediated by all three HP1 isoforms in vitro. However, in cells ADNP localization to areas of pericentromeric heterochromatin is only dependent on HP1α and HP1β. Besides a PGVLL sequence patch we uncovered an ARKS motif within the ADNP homeodomain involved in HP1 dependent H3K9me3 association and localization to pericentromeric heterochromatin. While knockdown of ADNP had no effect on HP1 distribution and heterochromatic histone and DNA modifications, we found ADNP silencing major satellite repeats. Our results identify a novel factor in the translation of H3K9me3 at pericentromeric heterochromatin that regulates transcription.
Collapse
Affiliation(s)
- Kerstin Mosch
- Laboratory of Chromatin Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Henriette Franz
- Laboratory of Chromatin Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Szabolcs Soeroes
- Laboratory of Chromatin Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Prim B. Singh
- Division of Immunoepigenetics, Research Center Borstel, Borstel, Germany
| | - Wolfgang Fischle
- Laboratory of Chromatin Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- * E-mail:
| |
Collapse
|
27
|
Bousiges O, Vasconcelos APD, Neidl R, Cosquer B, Herbeaux K, Panteleeva I, Loeffler JP, Cassel JC, Boutillier AL. Spatial memory consolidation is associated with induction of several lysine-acetyltransferase (histone acetyltransferase) expression levels and H2B/H4 acetylation-dependent transcriptional events in the rat hippocampus. Neuropsychopharmacology 2010; 35:2521-37. [PMID: 20811339 PMCID: PMC3055563 DOI: 10.1038/npp.2010.117] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 07/08/2010] [Accepted: 07/08/2010] [Indexed: 12/12/2022]
Abstract
Numerous genetic studies have shown that the CREB-binding protein (CBP) is an essential component of long-term memory formation, through its histone acetyltransferase (HAT) function. E1A-binding protein p300 and p300/CBP-associated factor (PCAF) have also recently been involved in memory formation. By contrast, only a few studies have reported on acetylation modifications during memory formation, and it remains unclear as to how the system is regulated during this dynamic phase. We investigated acetylation-dependent events and the expression profiles of these HATs during a hippocampus-dependent task taxing spatial reference memory in the Morris water maze. We found a specific increase in H2B and H4 acetylation in the rat dorsal hippocampus, while spatial memory was being consolidated. This increase correlated with the degree of specific acetylated histones enrichment on some memory/plasticity-related gene promoters. Overall, a global increase in HAT activity was measured during this memory consolidation phase, together with a global increase of CBP, p300, and PCAF expression. Interestingly, these regulations were altered in a model of hippocampal denervation disrupting spatial memory consolidation, making it impossible for the hippocampus to recruit the CBP pathway (CBP regulation and acetylated-H2B-dependent transcription). CBP has long been thought to be present in limited concentrations in the cells. These results show, for the first time, that CBP, p300, and PCAF are dynamically modulated during the establishment of a spatial memory and are likely to contribute to the induction of a specific epigenetic tagging of the genome for hippocampus-dependent (spatial) memory consolidation. These findings suggest the use of HAT-activating molecules in new therapeutic strategies of pathological aging, Alzheimer's disease, and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Olivier Bousiges
- Inserm, U692, Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Strasbourg, France
- Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Faculté de Médecine, Université de Strasbourg, UMRS692, Strasbourg, France
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Anne Pereira de Vasconcelos
- LINC FRE3289, Institut Fédératif de Recherche IFR 37, Université de Strasbourg-CNRS, GDR CNRS 2905, Strasbourg, France
| | - Romain Neidl
- Inserm, U692, Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Strasbourg, France
- Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Faculté de Médecine, Université de Strasbourg, UMRS692, Strasbourg, France
| | - Brigitte Cosquer
- LINC FRE3289, Institut Fédératif de Recherche IFR 37, Université de Strasbourg-CNRS, GDR CNRS 2905, Strasbourg, France
| | - Karine Herbeaux
- LINC FRE3289, Institut Fédératif de Recherche IFR 37, Université de Strasbourg-CNRS, GDR CNRS 2905, Strasbourg, France
| | - Irina Panteleeva
- Inserm, U692, Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Strasbourg, France
| | - Jean-Philippe Loeffler
- Inserm, U692, Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Strasbourg, France
- Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Faculté de Médecine, Université de Strasbourg, UMRS692, Strasbourg, France
| | - Jean-Christophe Cassel
- LINC FRE3289, Institut Fédératif de Recherche IFR 37, Université de Strasbourg-CNRS, GDR CNRS 2905, Strasbourg, France
| | - Anne-Laurence Boutillier
- Inserm, U692, Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Strasbourg, France
- Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Faculté de Médecine, Université de Strasbourg, UMRS692, Strasbourg, France
| |
Collapse
|
28
|
Grant J, Verrill C, Coustham V, Arneodo A, Palladino F, Monier K, Khalil A. Perinuclear distribution of heterochromatin in developing C. elegans embryos. Chromosome Res 2010; 18:873-85. [PMID: 21116703 DOI: 10.1007/s10577-010-9175-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 11/16/2010] [Indexed: 10/18/2022]
Abstract
Specific nuclear domains are nonrandomly positioned within the nuclear space, and this preferential positioning has been shown to play an important role in genome activity and stability. Well-known examples include the organization of repetitive DNA in telomere clusters or in the chromocenter of Drosophila and mammalian cells, which may provide a means to control the availability of general repressors, such as the heterochromatin protein 1 (HP1). We have specifically characterized the intranuclear positioning of in vivo fluorescence of the Caenorhabditis elegans HP1 homologue HPL-2 as a marker for heterochromatin domains in developing embryos. For this purpose, the wavelet transform modulus maxima (WTMM) segmentation method was generalized and adapted to segment the small embryonic cell nuclei in three dimensions. The implementation of a radial distribution algorithm revealed a preferential perinuclear positioning of HPL-2 fluorescence in wild-type embryos compared with the diffuse and homogeneous nuclear fluorescence observed in the lin-13 mutants. For all other genotypes analyzed, the quantitative analysis highlighted various degrees of preferential HPL-2 positioning at the nuclear periphery, which directly correlates with the number of HPL-2 foci previously counted on 2D projections. Using a probabilistic 3D cell nuclear model, we found that any two nuclei having the same number of foci, but with a different 3D probabilistic positioning scheme, can have significantly different counts in the 2D maximum projection, thus showing the deceptive limitations of using techniques of 2D maximum projection foci counts. By this approach, a strong perinuclear positioning of HPL-2 foci was brought into light upon inactivation of conserved chromatin-associated proteins, including the HAT cofactor TRAPP.
Collapse
Affiliation(s)
- Jeremy Grant
- Department of Mathematics and Statistics, University of Maine, Orono, ME 04469, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Bartkova J, Moudry P, Hodny Z, Lukas J, Rajpert-De Meyts E, Bartek J. Heterochromatin marks HP1γ, HP1α and H3K9me3, and DNA damage response activation in human testis development and germ cell tumours. ACTA ACUST UNITED AC 2010; 34:e103-13. [DOI: 10.1111/j.1365-2605.2010.01096.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
30
|
Swiss VA, Casaccia P. Cell-context specific role of the E2F/Rb pathway in development and disease. Glia 2010; 58:377-90. [PMID: 19795505 DOI: 10.1002/glia.20933] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Development of the central nervous system (CNS) requires the generation of neuronal and glial cell subtypes in appropriate numbers, and this demands the careful coordination of cell-cycle exit, survival, and differentiation. The E2F/Rb pathway is critical for cell-cycle regulation and also modulates survival and differentiation of distinct cell types in the developing and adult CNS. In this review, we first present the specific temporal patterns of expression of the E2F and Rb family members during CNS development and then discuss the genetic ablation of single or multiple members of these two families. Overall, the available data suggest a time-dependent and cell-context specific role of E2F and Rb family members in the developing and adult CNS.
Collapse
Affiliation(s)
- Victoria A Swiss
- Department of Neuroscience and Genetics and Genomics, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | |
Collapse
|
31
|
Talluri S, Isaac CE, Ahmad M, Henley SA, Francis SM, Martens AL, Bremner R, Dick FA. A G1 checkpoint mediated by the retinoblastoma protein that is dispensable in terminal differentiation but essential for senescence. Mol Cell Biol 2010; 30:948-60. [PMID: 20008551 PMCID: PMC2815577 DOI: 10.1128/mcb.01168-09] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 09/23/2009] [Accepted: 12/07/2009] [Indexed: 11/20/2022] Open
Abstract
Terminally differentiated cell types are needed to live and function in a postmitotic state for a lifetime. Cellular senescence is another type of permanent arrest that blocks the proliferation of cells in response to genotoxic stress. Here we show that the retinoblastoma protein (pRB) uses a mechanism to block DNA replication in senescence that is distinct from its role in permanent cell cycle exit associated with terminal differentiation. Our work demonstrates that a subtle mutation in pRB that cripples its ability to interact with chromatin regulators impairs heterochromatinization and repression of E2F-responsive promoters during senescence. In contrast, terminally differentiated nerve and muscle cells bearing the same mutation fully exit the cell cycle and block E2F-responsive gene expression by a different mechanism. Remarkably, this reveals that pRB recruits chromatin regulators primarily to engage a stress-responsive G(1) arrest program.
Collapse
Affiliation(s)
- Srikanth Talluri
- London Regional Cancer Program, Children's Health Research Institute, Department of Biochemistry, University of Western Ontario, London, Ontario, Canada, Genetics and Development Division, Toronto Western Research Institute, Department of Ophthalmology and Visual Science, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Christian E. Isaac
- London Regional Cancer Program, Children's Health Research Institute, Department of Biochemistry, University of Western Ontario, London, Ontario, Canada, Genetics and Development Division, Toronto Western Research Institute, Department of Ophthalmology and Visual Science, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad Ahmad
- London Regional Cancer Program, Children's Health Research Institute, Department of Biochemistry, University of Western Ontario, London, Ontario, Canada, Genetics and Development Division, Toronto Western Research Institute, Department of Ophthalmology and Visual Science, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Shauna A. Henley
- London Regional Cancer Program, Children's Health Research Institute, Department of Biochemistry, University of Western Ontario, London, Ontario, Canada, Genetics and Development Division, Toronto Western Research Institute, Department of Ophthalmology and Visual Science, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Sarah M. Francis
- London Regional Cancer Program, Children's Health Research Institute, Department of Biochemistry, University of Western Ontario, London, Ontario, Canada, Genetics and Development Division, Toronto Western Research Institute, Department of Ophthalmology and Visual Science, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Alison L. Martens
- London Regional Cancer Program, Children's Health Research Institute, Department of Biochemistry, University of Western Ontario, London, Ontario, Canada, Genetics and Development Division, Toronto Western Research Institute, Department of Ophthalmology and Visual Science, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Rod Bremner
- London Regional Cancer Program, Children's Health Research Institute, Department of Biochemistry, University of Western Ontario, London, Ontario, Canada, Genetics and Development Division, Toronto Western Research Institute, Department of Ophthalmology and Visual Science, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Frederick A. Dick
- London Regional Cancer Program, Children's Health Research Institute, Department of Biochemistry, University of Western Ontario, London, Ontario, Canada, Genetics and Development Division, Toronto Western Research Institute, Department of Ophthalmology and Visual Science, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Dawson MA, Bannister AJ, Göttgens B, Foster SD, Bartke T, Green AR, Kouzarides T. JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin. Nature 2009; 461:819-22. [PMID: 19783980 PMCID: PMC3785147 DOI: 10.1038/nature08448] [Citation(s) in RCA: 481] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Accepted: 08/21/2009] [Indexed: 12/12/2022]
Abstract
Activation of Janus kinase 2 (JAK2) by chromosomal translocations or point mutations is a frequent event in haematological malignancies. JAK2 is a non-receptor tyrosine kinase that regulates several cellular processes by inducing cytoplasmic signalling cascades. Here we show that human JAK2 is present in the nucleus of haematopoietic cells and directly phosphorylates Tyr 41 (Y41) on histone H3. Heterochromatin protein 1alpha (HP1alpha), but not HP1beta, specifically binds to this region of H3 through its chromo-shadow domain. Phosphorylation of H3Y41 by JAK2 prevents this binding. Inhibition of JAK2 activity in human leukaemic cells decreases both the expression of the haematopoietic oncogene lmo2 and the phosphorylation of H3Y41 at its promoter, while simultaneously increasing the binding of HP1alpha at the same site. Tauhese results identify a previously unrecognized nuclear role for JAK2 in the phosphorylation of H3Y41 and reveal a direct mechanistic link between two genes, jak2 and lmo2, involved in normal haematopoiesis and leukaemia.
Collapse
Affiliation(s)
- Mark A. Dawson
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge, CB2 0XY, UK
- Addenbrooke’s Hospital, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Andrew J. Bannister
- Gurdon Institute and Department of Pathology, Tennis Court Road, Cambridge, CB2 1QN, UK
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge, CB2 0XY, UK
| | - Samuel D. Foster
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge, CB2 0XY, UK
| | - Till Bartke
- Gurdon Institute and Department of Pathology, Tennis Court Road, Cambridge, CB2 1QN, UK
| | - Anthony R. Green
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge, CB2 0XY, UK
- Addenbrooke’s Hospital, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Tony Kouzarides
- Gurdon Institute and Department of Pathology, Tennis Court Road, Cambridge, CB2 1QN, UK
| |
Collapse
|
33
|
Northcott PA, Nakahara Y, Wu X, Feuk L, Ellison DW, Croul S, Mack S, Kongkham PN, Peacock J, Dubuc A, Ra YS, Zilberberg K, McLeod J, Scherer SW, Sunil Rao J, Eberhart CG, Grajkowska W, Gillespie Y, Lach B, Grundy R, Pollack IF, Hamilton RL, Van Meter T, Carlotti CG, Boop F, Bigner D, Gilbertson RJ, Rutka JT, Taylor MD. Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat Genet 2009; 41:465-72. [PMID: 19270706 PMCID: PMC4454371 DOI: 10.1038/ng.336] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 01/15/2009] [Indexed: 01/20/2023]
Abstract
We used high-resolution SNP genotyping to identify regions of genomic gain and loss in the genomes of 212 medulloblastomas, malignant pediatric brain tumors. We found focal amplifications of 15 known oncogenes and focal deletions of 20 known tumor suppressor genes (TSG), most not previously implicated in medulloblastoma. Notably, we identified previously unknown amplifications and homozygous deletions, including recurrent, mutually exclusive, highly focal genetic events in genes targeting histone lysine methylation, particularly that of histone 3, lysine 9 (H3K9). Post-translational modification of histone proteins is critical for regulation of gene expression, can participate in determination of stem cell fates and has been implicated in carcinogenesis. Consistent with our genetic data, restoration of expression of genes controlling H3K9 methylation greatly diminishes proliferation of medulloblastoma in vitro. Copy number aberrations of genes with critical roles in writing, reading, removing and blocking the state of histone lysine methylation, particularly at H3K9, suggest that defective control of the histone code contributes to the pathogenesis of medulloblastoma.
Collapse
Affiliation(s)
- Paul A Northcott
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mehler MF. Epigenetic principles and mechanisms underlying nervous system functions in health and disease. Prog Neurobiol 2008; 86:305-41. [PMID: 18940229 PMCID: PMC2636693 DOI: 10.1016/j.pneurobio.2008.10.001] [Citation(s) in RCA: 197] [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: 07/24/2008] [Revised: 10/03/2008] [Accepted: 10/09/2008] [Indexed: 01/11/2023]
Abstract
Epigenetics and epigenomic medicine encompass a new science of brain and behavior that are already providing unique insights into the mechanisms underlying brain development, evolution, neuronal and network plasticity and homeostasis, senescence, the etiology of diverse neurological diseases and neural regenerative processes. Epigenetic mechanisms include DNA methylation, histone modifications, nucleosome repositioning, higher order chromatin remodeling, non-coding RNAs, and RNA and DNA editing. RNA is centrally involved in directing these processes, implying that the transcriptional state of the cell is the primary determinant of epigenetic memory. This transcriptional state can be modified not only by internal and external cues affecting gene expression and post-transcriptional processing, but also by RNA and DNA editing through activity-dependent intracellular transport and modulation of RNAs and RNA regulatory supercomplexes, and through trans-neuronal and systemic trafficking of functional RNA subclasses. These integrated processes promote dynamic reorganization of nuclear architecture and the genomic landscape to modulate functional gene and neural networks with complex temporal and spatial trajectories. Epigenetics represents the long sought after molecular interface mediating gene-environmental interactions during critical periods throughout the lifecycle. The discipline of environmental epigenomics has begun to identify combinatorial profiles of environmental stressors modulating the latency, initiation and progression of specific neurological disorders, and more selective disease biomarkers and graded molecular responses to emerging therapeutic interventions. Pharmacoepigenomic therapies will promote accelerated recovery of impaired and seemingly irrevocably lost cognitive, behavioral, sensorimotor functions through epigenetic reprogramming of endogenous regional neural stem cell fate decisions, targeted tissue remodeling and restoration of neural network integrity, plasticity and connectivity.
Collapse
Affiliation(s)
- Mark F Mehler
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Rose F. Kennedy Center 401, 1410 Pelham Parkway South, Bronx, NY 10461, USA.
| |
Collapse
|
35
|
Takanashi M, Oikawa K, Fujita K, Kudo M, Kinoshita M, Kuroda M. Heterochromatin protein 1gamma epigenetically regulates cell differentiation and exhibits potential as a therapeutic target for various types of cancers. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 174:309-16. [PMID: 19056850 DOI: 10.2353/ajpath.2009.080148] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Heterochromatin protein 1 (HP1) is a chromosomal protein that participates in both chromatin packaging and gene silencing. Three HP1 isoforms (alpha, beta, and gamma) occur in mammals, but their functional differences are still incompletely understood. In this study, we found that HP1gamma levels are decreased during adipocyte differentiation, whereas HP1alpha and beta levels are expressed constitutively during adipogenesis in cultured preadipocyte cells. In addition, ectopic overexpression of HP1gamma inhibited adipogenesis. Furthermore, we did not detect any HP1gamma protein in the differentiated cells of various normal human tissues. These results suggest that the loss of HP1gamma is required for cell differentiation to occur. On the other hand, the methylation levels of lysine 20 (K20) on histone H4 showed a significant correlation with HP1gamma expression in both these preadipocyte cells and normal tissue samples. However, all cancer tissues examined were positive for HP1gamma but were often negative for trimethylated histone H4 K20. Thus, a dissociation of the correlation between HP1gamma expression and histone H4 K20 trimethylation may reflect the malfunction of epigenetic control. Finally, suppression of HP1gamma expression restrained cell growth in various cancer-derived cell lines, suggesting that HP1gamma may be an effective target for gene therapy against various human cancers. Taken together, our results demonstrate the novel function of HP1gamma in the epigenetic regulation of both cell differentiation and cancer development.
Collapse
|
36
|
Aucott R, Bullwinkel J, Yu Y, Shi W, Billur M, Brown JP, Menzel U, Kioussis D, Wang G, Reisert I, Weimer J, Pandita RK, Sharma GG, Pandita TK, Fundele R, Singh PB. HP1-beta is required for development of the cerebral neocortex and neuromuscular junctions. J Cell Biol 2008; 183:597-606. [PMID: 19015315 PMCID: PMC2582898 DOI: 10.1083/jcb.200804041] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Accepted: 10/09/2008] [Indexed: 11/22/2022] Open
Abstract
HP1 proteins are thought to be modulators of chromatin organization in all mammals, yet their exact physiological function remains unknown. In a first attempt to elucidate the function of these proteins in vivo, we disrupted the murine Cbx1 gene, which encodes the HP1-beta isotype, and show that the Cbx1(-/-) -null mutation leads to perinatal lethality. The newborn mice succumbed to acute respiratory failure, whose likely cause is the defective development of neuromuscular junctions within the endplate of the diaphragm. We also observe aberrant cerebral cortex development in Cbx1(-/-) mutant brains, which have reduced proliferation of neuronal precursors, widespread cell death, and edema. In vitro cultures of neurospheres from Cbx1(-/-) mutant brains reveal a dramatic genomic instability. Our results demonstrate that HP1 proteins are not functionally redundant and that they are likely to regulate lineage-specific changes in heterochromatin organization.
Collapse
Affiliation(s)
- Rebecca Aucott
- Division of Immunoepigenetics, Department of Immunology and Cell Biology, Research Center Borstel, Borstel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Remaud S, Audibert A, Gho M. S-phase favours notch cell responsiveness in the Drosophila bristle lineage. PLoS One 2008; 3:e3646. [PMID: 18985153 PMCID: PMC2574411 DOI: 10.1371/journal.pone.0003646] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Accepted: 10/15/2008] [Indexed: 11/18/2022] Open
Abstract
We have studied cell sensitivity to Notch pathway signalling throughout the cell cycle. As model system, we used the Drosophila bristle lineage where at each division N plays a crucial role in fate determination. Using in vivo imaging, we followed this lineage and activated the N-pathway at different moments of the secondary precursor cell cycle. We show that cells are more susceptible to respond to N-signalling during the S-phase. Thus, the period of heightened sensitivity coincided with the period of the S-phase. More importantly, modifications of S-phase temporality induced corresponding changes in the period of the cell's reactivity to N-activation. Moreover, S-phase abolition was correlated with a decrease in the expression of tramtrack, a downstream N-target gene. Finally, N cell responsiveness was modified after changes in chromatin packaging. We suggest that high-order chromatin structures associated with the S-phase create favourable conditions that increase the efficiency of the transcriptional machinery with respect to N-target genes.
Collapse
Affiliation(s)
- Sylvie Remaud
- Université Pierre et Marie Curie, UMR 7622, Paris, France
- CNRS, UMR 7622, Paris, France
| | - Agnès Audibert
- Université Pierre et Marie Curie, UMR 7622, Paris, France
- CNRS, UMR 7622, Paris, France
| | - Michel Gho
- Université Pierre et Marie Curie, UMR 7622, Paris, France
- CNRS, UMR 7622, Paris, France
- * E-mail:
| |
Collapse
|
38
|
Yahi H, Fritsch L, Philipot O, Guasconi V, Souidi M, Robin P, Polesskaya A, Losson R, Harel-Bellan A, Ait-Si-Ali S. Differential cooperation between heterochromatin protein HP1 isoforms and MyoD in myoblasts. J Biol Chem 2008; 283:23692-700. [PMID: 18599480 DOI: 10.1074/jbc.m802647200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mechanisms of transcriptional repression are important during cell differentiation. Mammalian heterochromatin protein 1 isoforms HP1alpha, HP1beta, and HP1gamma play important roles in the regulation of chromatin structure and function. We explored the possibility of different roles for the three HP1 isoforms in an integrated system, skeletal muscle terminal differentiation. In this system, terminal differentiation is initiated by the transcription factor MyoD, whose target genes remain mainly silent until myoblasts are induced to differentiate. Here we show that HP1alpha and HP1beta isoforms, but not HP1gamma, interact with MyoD in myoblasts. This interaction is direct, as shown using recombinant proteins in vitro. A gene reporter assay revealed that HP1alpha and HP1beta, but not HP1gamma, inhibit MyoD transcriptional activity, suggesting a model in which MyoD could serve as a bridge between nucleosomes and chromatin-binding proteins such as HDACs and HP1. Chromatin immunoprecipitation assays show a preferential recruitment of HP1 proteins on MyoD target genes in proliferating myoblasts. Finally, modulation of HP1 protein level impairs MyoD target gene expression and muscle terminal differentiation. Together, our data show a nonconventional interaction between HP1 and a tissue-specific transcription factor, MyoD. In addition, they strongly suggest that HP1 isoforms play important roles during muscle terminal differentiation in an isoform-dependent manner.
Collapse
Affiliation(s)
- Hakima Yahi
- Institut André Lwoff, CNRS, FRE 2944, 7 rue Guy Moquet, Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Vandromme M, Chailleux C, Escaffit F, Trouche D. Binding of the Retinoblastoma Protein Is Not the Determinant for Stable Repression of Some E2F-Regulated Promoters in Muscle Cells. Mol Cancer Res 2008; 6:418-25. [DOI: 10.1158/1541-7786.mcr-07-0381] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Buttitta LA, Edgar BA. Mechanisms controlling cell cycle exit upon terminal differentiation. Curr Opin Cell Biol 2007; 19:697-704. [PMID: 18035529 PMCID: PMC2700000 DOI: 10.1016/j.ceb.2007.10.004] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 10/05/2007] [Indexed: 11/24/2022]
Abstract
Coordinating terminal differentiation with permanent exit from the cell cycle is crucial for proper organogenesis, yet how the cell cycle is blocked in differentiated tissues remains unclear. Important roles for retinoblastoma family proteins and Cyclin-dependent kinase inhibitors have been delineated, but in many cases it remains unclear what triggers cell cycle exit. This review focuses on describing recent advances in deciphering how terminal differentiation and exit from the cell cycle are coordinated.
Collapse
Affiliation(s)
- Laura A. Buttitta
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109. Fax: 206-667-3308
| | - Bruce A. Edgar
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109. Fax: 206-667-3308
| |
Collapse
|