1
|
Wang X, Kulik K, Wan TC, Lough JW, Auchampach JA. Histone H2A.Z Deacetylation and Dedifferentiation in Infarcted/Tip60-depleted Cardiomyocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.11.575312. [PMID: 38260622 PMCID: PMC10802610 DOI: 10.1101/2024.01.11.575312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Myocardial infarction (MI) results in the loss of billions of cardiomyocytes (CMs), resulting in cardiac dysfunction. To re-muscularize injured myocardium, new CMs must be generated via renewed proliferation of surviving CMs. Approaches to induce proliferation of CMs after injury have been insufficient. Toward this end we are targeting the acetyltransferase Tip60, encoded by the Kat5 gene, based on the rationale that its pleiotropic functions combine to block CM proliferation at multiple checkpoints. We previously demonstrated that genetic depletion of Tip60 in a mouse model after MI reduces scarring, retains cardiac function, and activates the CM cell-cycle, although it remains unclear whether this culminates in the generation of daughter CMs. In order for pre-existing CMs in the adult heart to undergo proliferation, it has become accepted that they must first dedifferentiate, a process highlighted by loss of maturity, epithelial to mesenchymal transitioning (EMT), and reversion from fatty acid oxidation to glycolytic metabolism, accompanied by softening of the myocardial extracellular matrix (ECM). Based on recently published findings that Tip60 induces and maintains the differentiated state of hematopoietic stem cells and neurons via site-specific acetylation of the histone variant H2A.Z, we assessed levels of acetylated H2A.Z and dedifferentiation markers after depleting Tip60 in CMs post-MI. We report that genetic depletion of Tip60 from CMs after MI results in the near obliteration of acetylated H2A.Z in CM nuclei, accompanied by the altered expression of genes indicative of EMT induction, ECM softening, decreased fatty acid oxidation, and depressed expression of genes that regulate the TCA cycle. In accord with the possibility that site-specific acetylation of H2A.Z maintains adult CMs in a mature state of differentiation, CUT&Tag revealed enrichment of H2A.Zac K4/K7 in genetic motifs and in GO terms respectively associated with CM transcription factor binding and muscle development/differentiation. Along with our previous findings, these results support the notion that Tip60 has multiple targets in CMs that combine to maintain the differentiated state and prevent proliferation.
Collapse
|
2
|
Liu C, Gui Z, An C, Sun F, Gao X, Ge S. STUB1 is acetylated by KAT5 and alleviates myocardial ischemia-reperfusion injury through LATS2-YAP-β-catenin axis. Commun Biol 2024; 7:396. [PMID: 38561411 PMCID: PMC10985082 DOI: 10.1038/s42003-024-06086-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is involved in the pathogenesis of multiple cardiovascular diseases. This study elucidated the biological function of lysine acetyltransferase 5 (KAT5) in cardiomyocyte pyroptosis during MIRI. Oxygen-glucose deprivation/reoxygenation and left anterior descending coronary artery ligation were used to establish MIRI models. Here we show, KAT5 and STIP1 homology and U-box-containing protein 1 (STUB1) were downregulated, while large tumor suppressor kinase 2 (LATS2) was upregulated in MIRI models. KAT5/STUB1 overexpression or LATS2 silencing repressed cardiomyocyte pyroptosis. Mechanistically, KAT5 promoted STUB1 transcription via acetylation modulation, and subsequently caused ubiquitination and degradation of LATS2, which activated YAP/β-catenin pathway. Notably, the inhibitory effect of STUB1 overexpression on cardiomyocyte pyroptosis was abolished by LATS2 overexpression or KAT5 depletion. Our findings suggest that KAT5 overexpression inhibits NLRP3-mediated cardiomyocyte pyroptosis to relieve MIRI through modulation of STUB1/LATS2/YAP/β-catenin axis, providing a potential therapeutic target for MIRI.
Collapse
Affiliation(s)
- Can Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, P.R. China
| | - Zhongxuan Gui
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, P.R. China
| | - Cheng An
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, P.R. China
| | - Fei Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, P.R. China
| | - Xiaotian Gao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, P.R. China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, P.R. China.
| |
Collapse
|
3
|
Lin CY, Chang YM, Tseng HY, Shih YL, Yeh HH, Liao YR, Tang HH, Hsu CL, Chen CC, Yan YT, Kao CF. Epigenetic regulator RNF20 underlies temporal hierarchy of gene expression to regulate postnatal cardiomyocyte polarization. Cell Rep 2023; 42:113416. [PMID: 37967007 DOI: 10.1016/j.celrep.2023.113416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 09/19/2023] [Accepted: 10/25/2023] [Indexed: 11/17/2023] Open
Abstract
Differentiated cardiomyocytes (CMs) must undergo diverse morphological and functional changes during postnatal development. However, the mechanisms underlying initiation and coordination of these changes remain unclear. Here, we delineate an integrated, time-ordered transcriptional network that begins with expression of genes for cell-cell connections and leads to a sequence of structural, cell-cycle, functional, and metabolic transitions in mouse postnatal hearts. Depletion of histone H2B ubiquitin ligase RNF20 disrupts this gene network and impairs CM polarization. Subsequently, assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis confirmed that RNF20 contributes to chromatin accessibility in this context. As such, RNF20 is likely to facilitate binding of transcription factors at the promoters of genes involved in cell-cell connections and actin organization, which are crucial for CM polarization and functional integration. These results suggest that CM polarization is one of the earliest events during postnatal heart development and provide insights into how RNF20 regulates CM polarity and the postnatal gene program.
Collapse
Affiliation(s)
- Chia-Yeh Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsin-Yi Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yen-Ling Shih
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsiao-Hui Yeh
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - You-Rou Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Han-Hsuan Tang
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chia-Ling Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| |
Collapse
|
4
|
Shibahara D, Akanuma N, Kobayashi IS, Heo E, Ando M, Fujii M, Jiang F, Prin PN, Pan G, Wong K, Costa DB, Bararia D, Tenen DG, Watanabe H, Kobayashi SS. TIP60 is required for tumorigenesis in non-small cell lung cancer. Cancer Sci 2023; 114:2400-2413. [PMID: 36916958 PMCID: PMC10236639 DOI: 10.1111/cas.15785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Histone modifications play crucial roles in transcriptional activation, and aberrant epigenetic changes are associated with oncogenesis. Lysine (K) acetyltransferases 5 (TIP60, also known as KAT5) is reportedly implicated in cancer development and maintenance, although its function in lung cancer remains controversial. Here we demonstrate that TIP60 knockdown in non-small cell lung cancer cell lines decreased tumor cell growth, migration, and invasion. Furthermore, analysis of a mouse lung cancer model with lung-specific conditional Tip60 knockout revealed suppressed tumor formation relative to controls, but no apparent effects on normal lung homeostasis. RNA-seq and ChIP-seq analyses of inducible TIP60 knockdown H1975 cells relative to controls revealed transglutaminase enzyme (TGM5) as downstream of TIP60. Investigation of a connectivity map database identified several candidate compounds that decrease TIP60 mRNA, one that suppressed tumor growth in cell culture and in vivo. In addition, TH1834, a TIP60 acetyltransferase inhibitor, showed comparable antitumor effects in cell culture and in vivo. Taken together, suppression of TIP60 activity shows tumor-specific efficacy against lung cancer, with no overt effect on normal tissues. Our work suggests that targeting TIP60 could be a promising approach to treating lung cancer.
Collapse
Affiliation(s)
- Daisuke Shibahara
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Naoki Akanuma
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
- Department of PathologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ikei S. Kobayashi
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Eunyoung Heo
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
- Department of Internal MedicineSMG‐SNU Boramae Medical CenterSeoulSouth Korea
| | - Mariko Ando
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Masanori Fujii
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Feng Jiang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Department of Genetics and Genomic SciencesTisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - P. Nicholas Prin
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Gilbert Pan
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Kwok‐Kin Wong
- Perlmutter Cancer CenterNYU Langone Medical CenterNew YorkNew YorkUSA
| | - Daniel B. Costa
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Deepak Bararia
- Harvard Stem Cell Institute, Harvard Medical SchoolBostonMassachusettsUSA
| | - Daniel G. Tenen
- Harvard Stem Cell Institute, Harvard Medical SchoolBostonMassachusettsUSA
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - Hideo Watanabe
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Department of Genetics and Genomic SciencesTisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Susumu S. Kobayashi
- Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
- Harvard Stem Cell Institute, Harvard Medical SchoolBostonMassachusettsUSA
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| |
Collapse
|
5
|
Pollina EA, Gilliam DT, Landau AT, Lin C, Pajarillo N, Davis CP, Harmin DA, Yap EL, Vogel IR, Griffith EC, Nagy MA, Ling E, Duffy EE, Sabatini BL, Weitz CJ, Greenberg ME. A NPAS4-NuA4 complex couples synaptic activity to DNA repair. Nature 2023; 614:732-741. [PMID: 36792830 PMCID: PMC9946837 DOI: 10.1038/s41586-023-05711-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 01/05/2023] [Indexed: 02/17/2023]
Abstract
Neuronal activity is crucial for adaptive circuit remodelling but poses an inherent risk to the stability of the genome across the long lifespan of postmitotic neurons1-5. Whether neurons have acquired specialized genome protection mechanisms that enable them to withstand decades of potentially damaging stimuli during periods of heightened activity is unknown. Here we identify an activity-dependent DNA repair mechanism in which a new form of the NuA4-TIP60 chromatin modifier assembles in activated neurons around the inducible, neuronal-specific transcription factor NPAS4. We purify this complex from the brain and demonstrate its functions in eliciting activity-dependent changes to neuronal transcriptomes and circuitry. By characterizing the landscape of activity-induced DNA double-strand breaks in the brain, we show that NPAS4-NuA4 binds to recurrently damaged regulatory elements and recruits additional DNA repair machinery to stimulate their repair. Gene regulatory elements bound by NPAS4-NuA4 are partially protected against age-dependent accumulation of somatic mutations. Impaired NPAS4-NuA4 signalling leads to a cascade of cellular defects, including dysregulated activity-dependent transcriptional responses, loss of control over neuronal inhibition and genome instability, which all culminate to reduce organismal lifespan. In addition, mutations in several components of the NuA4 complex are reported to lead to neurodevelopmental and autism spectrum disorders. Together, these findings identify a neuronal-specific complex that couples neuronal activity directly to genome preservation, the disruption of which may contribute to developmental disorders, neurodegeneration and ageing.
Collapse
Affiliation(s)
- Elizabeth A Pollina
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel T Gilliam
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Andrew T Landau
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Cindy Lin
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Naomi Pajarillo
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | | - David A Harmin
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ee-Lynn Yap
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ian R Vogel
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Eric C Griffith
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - M Aurel Nagy
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Emi Ling
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Erin E Duffy
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Charles J Weitz
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
6
|
Shen F, Zhuang S. Histone Acetylation and Modifiers in Renal Fibrosis. Front Pharmacol 2022; 13:760308. [PMID: 35559244 PMCID: PMC9086452 DOI: 10.3389/fphar.2022.760308] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 04/04/2022] [Indexed: 12/23/2022] Open
Abstract
Histones are the most abundant proteins bound to DNA in eukaryotic cells and frequently subjected to post-modifications such as acetylation, methylation, phosphorylation and ubiquitination. Many studies have shown that histone modifications, especially histone acetylation, play an important role in the development and progression of renal fibrosis. Histone acetylation is regulated by three families of proteins, including histone acetyltransferases (HATs), histone deacetylases (HDACs) and bromodomain and extraterminal (BET) proteins. These acetylation modifiers are involved in a variety of pathophysiological processes leading to the development of renal fibrosis, including partial epithelial-mesenchymal transition, renal fibroblast activation, inflammatory response, and the expression of pro-fibrosis factors. In this review, we summarize the role and regulatory mechanisms of HATs, HDACs and BET proteins in renal fibrosis and provide evidence for targeting these modifiers to treat various chronic fibrotic kidney diseases in animal models.
Collapse
Affiliation(s)
- Fengchen Shen
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
7
|
Auchampach J, Han L, Huang GN, Kühn B, Lough JW, O'Meara CC, Payumo AY, Rosenthal NA, Sucov HM, Yutzey KE, Patterson M. Measuring cardiomyocyte cell-cycle activity and proliferation in the age of heart regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H579-H596. [PMID: 35179974 PMCID: PMC8934681 DOI: 10.1152/ajpheart.00666.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 12/14/2022]
Abstract
During the past two decades, the field of mammalian myocardial regeneration has grown dramatically, and with this expanded interest comes increasing claims of experimental manipulations that mediate bona fide proliferation of cardiomyocytes. Too often, however, insufficient evidence or improper controls are provided to support claims that cardiomyocytes have definitively proliferated, a process that should be strictly defined as the generation of two de novo functional cardiomyocytes from one original cardiomyocyte. Throughout the literature, one finds inconsistent levels of experimental rigor applied, and frequently the specific data supplied as evidence of cardiomyocyte proliferation simply indicate cell-cycle activation or DNA synthesis, which do not necessarily lead to the generation of new cardiomyocytes. In this review, we highlight potential problems and limitations faced when characterizing cardiomyocyte proliferation in the mammalian heart, and summarize tools and experimental standards, which should be used to support claims of proliferation-based remuscularization. In the end, definitive establishment of de novo cardiomyogenesis can be difficult to prove; therefore, rigorous experimental strategies should be used for such claims.
Collapse
Affiliation(s)
- John Auchampach
- Department of Pharmacology and Toxicology and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lu Han
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
- Division of Pediatric Cardiology, Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, California
| | - Bernhard Kühn
- Division of Cardiology, Pediatric Institute for Heart Regeneration and Therapeutics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania
- McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania
| | - John W Lough
- Department of Cell Biology Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Caitlin C O'Meara
- Department of Physiology and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alexander Y Payumo
- Department of Biological Sciences, San José State University, San Jose, California
| | - Nadia A Rosenthal
- The Jackson Laboratory, Bar Harbor, Maine
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
- National Heart and Lung Institute, Imperial College of London, London, United Kingdom
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Katherine E Yutzey
- The Heart Institute, Cincinnati Children's Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Michaela Patterson
- Department of Cell Biology Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
8
|
Wang X, Wan TC, Lauth A, Purdy AL, Kulik KR, Patterson M, Lough JW, Auchampach JA. Conditional depletion of the acetyltransferase Tip60 protects against the damaging effects of myocardial infarction. J Mol Cell Cardiol 2022; 163:9-19. [PMID: 34610340 PMCID: PMC8816866 DOI: 10.1016/j.yjmcc.2021.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 02/03/2023]
Abstract
Injury from myocardial infarction (MI) and consequent post-MI remodeling is accompanied by massive loss of cardiomyocytes (CM), a cell type critical for contractile function that is for all practical purposes non-regenerable due to its profound state of proliferative senescence. Identification of factors that limit CM survival and/or constrain CM renewal provides potential therapeutic targets. Tip60, a pan-acetyltransferase encoded by the Kat5 gene, has been reported to activate apoptosis as well as multiple anti-proliferative pathways in non-cardiac cells; however, its role in CMs, wherein it is abundantly expressed, remains unknown. Here, using mice containing floxed Kat5 alleles and a tamoxifen-activated Myh6-MerCreMer recombinase transgene, we report that conditional depletion of Tip60 in CMs three days after MI induced by permanent coronary artery ligation greatly improves functional recovery for up to 28 days. This is accompanied by diminished scarring, activation of cell-cycle transit markers in CMs within the infarct border and remote zones, reduced expression of cell-cycle inhibitors pAtm and p27, and reduced apoptosis in the remote regions. These findings implicate Tip60 as a novel, multifactorial target for limiting the damaging effects of ischemic heart disease.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Tina C. Wan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Amelia Lauth
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Alexandra L. Purdy
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Katherine R. Kulik
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Michaela Patterson
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - John W. Lough
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - John A. Auchampach
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
9
|
Wang X, Lupton C, Lauth A, Wan TC, Foster P, Patterson M, Auchampach JA, Lough JW. Evidence that the acetyltransferase Tip60 induces the DNA damage response and cell-cycle arrest in neonatal cardiomyocytes. J Mol Cell Cardiol 2021; 155:88-98. [PMID: 33609538 PMCID: PMC8154663 DOI: 10.1016/j.yjmcc.2021.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 12/19/2022]
Abstract
Tip60, a pan-acetyltransferase encoded by the Kat5 gene, is enriched in the myocardium; however, its function in the heart is unknown. In cancer cells, Tip60 acetylates Atm (Ataxia-telangiectasia mutated), enabling its auto-phosphorylation (pAtm), which activates the DNA damage response (DDR). It was recently reported that activation of pAtm at the time of birth induces the DDR in cardiomyocytes (CMs), resulting in proliferative senescence. We therefore hypothesized that Tip60 initiates this process, and that depletion of Tip60 accordingly diminishes the DDR while extending the duration of CM cell-cycle activation. To test this hypothesis, an experimental model was used wherein a Myh6-driven Cre-recombinase transgene was activated on postnatal day 0 (P0) to recombine floxed Kat5 alleles and induce Tip60 depletion in neonatal CMs, without causing pathogenesis. Depletion of Tip60 resulted in reduced numbers of pAtm-positive CMs during the neonatal period, which correlated with reduced numbers of pH2A.X-positive CMs and decreased expression of genes encoding markers of the DDR as well as inflammation. This was accompanied by decreased expression of the cell-cycle inhibitors Meis1 and p27, activation of the cell-cycle in CMs, reduced CM size, and increased numbers of mononuclear/diploid CMs. Increased expression of fetal markers suggested that Tip60 depletion promotes a fetal-like proliferative state. Finally, infarction of Tip60-depleted hearts at P7 revealed improved cardiac function at P39 accompanied by reduced fibrosis, increased CM cell-cycle activation, and reduced apoptosis in the remote zone. These findings indicate that, among its pleiotropic functions, Tip60 induces the DDR in CMs, contributing to proliferative senescence.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Pharmacology and Toxicology and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Carri Lupton
- Department of Cell Biology, Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Amelia Lauth
- Department of Cell Biology, Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Tina C Wan
- Department of Pharmacology and Toxicology and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Parker Foster
- Department of Cell Biology, Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - John A Auchampach
- Department of Pharmacology and Toxicology and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America.
| | - John W Lough
- Department of Cell Biology, Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America.
| |
Collapse
|
10
|
Mucke HA. Patent highlights October-November 2020. Pharm Pat Anal 2021; 10:51-58. [PMID: 33594903 DOI: 10.4155/ppa-2021-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 01/29/2021] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
11
|
Wang X, Lauth A, Wan TC, Lough JW, Auchampach JA. Myh6-driven Cre recombinase activates the DNA damage response and the cell cycle in the myocardium in the absence of loxP sites. Dis Model Mech 2020; 13:dmm046375. [PMID: 33106234 PMCID: PMC7758623 DOI: 10.1242/dmm.046375] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/02/2020] [Indexed: 12/20/2022] Open
Abstract
Regeneration of muscle in the damaged myocardium is a major objective of cardiovascular research, for which purpose many investigators utilize mice containing transgenes encoding Cre recombinase to recombine loxP-flanked target genes. An unfortunate side effect of the Cre-loxP model is the propensity of Cre recombinase to inflict off-target DNA damage, which has been documented in various eukaryotic cell types including cardiomyocytes (CMs). In the heart, reported effects of Cre recombinase include contractile dysfunction, fibrosis, cellular infiltration and induction of the DNA damage response (DDR). During experiments on adult mice containing a widely used Myh6-merCremer transgene, the protein product of which is activated by tamoxifen, we observed large, transient, off-target effects of merCremer, some of which have not previously been reported. On Day 3 after the first of three daily tamoxifen injections, immunofluorescent microscopy of heart sections revealed that the presence of merCremer protein in myonuclei was nearly uniform, thereafter diminishing to near extinction by Day 6; during this time, cardiac function was depressed as determined by echocardiography. On Day 5, peaks of apoptosis and expression of DDR-regulatory genes were observed, highlighted by >25-fold increased expression of Brca1 Concomitantly, the expression of genes encoding cyclin-A2, cyclin-B2 and cyclin-dependent kinase 1, which regulate the G2/S cell-cycle transition, were dramatically increased (>50- to 100-fold). Importantly, immunofluorescent staining revealed that this was accompanied by peaks in Ki67, 5'-bromodeoxyuridine and phosphohistone H3 labeling in non-CMs, as well as CMs. We further document that tamoxifen-induced activation of merCremer exacerbates cardiac dysfunction following myocardial infarction. These findings, when considered in the context of previous reports, indicate that the presence of merCremer in the nucleus induces DNA damage and unscheduled cell-cycle activation. Although these effects are transient, the inclusion of appropriate controls, coupled with an awareness of the defects caused by Cre recombinase, are required to avoid misinterpreting results when using Cre-loxP models for cardiac regeneration studies.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amelia Lauth
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tina C Wan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John W Lough
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John A Auchampach
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
12
|
Peng L, Qian M, Liu Z, Tang X, Sun J, Jiang Y, Sun S, Cao X, Pang Q, Liu B. Deacetylase-independent function of SIRT6 couples GATA4 transcription factor and epigenetic activation against cardiomyocyte apoptosis. Nucleic Acids Res 2020; 48:4992-5005. [PMID: 32239217 PMCID: PMC7229816 DOI: 10.1093/nar/gkaa214] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/21/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
SIRT6 deacetylase activity improves stress resistance via gene silencing and genome maintenance. Here, we reveal a deacetylase-independent function of SIRT6, which promotes anti-apoptotic gene expression via the transcription factor GATA4. SIRT6 recruits TIP60 acetyltransferase to acetylate GATA4 at K328/330, thus enhancing its chromatin binding capacity. In turn, GATA4 inhibits the deacetylase activity of SIRT6, thus ensuring the local chromatin accessibility via TIP60-promoted H3K9 acetylation. Significantly, the treatment of doxorubicin (DOX), an anti-cancer chemotherapeutic, impairs the SIRT6-TIP60-GATA4 trimeric complex, blocking GATA4 acetylation and causing cardiomyocyte apoptosis. While GATA4 hyperacetylation-mimic retains the protective effect against DOX, the hypoacetylation-mimic loses such ability. Thus, the data reveal a novel SIRT6-TIP60-GATA4 axis, which promotes the anti-apoptotic pathway to prevent DOX toxicity. Targeting the trimeric complex constitutes a new strategy to improve the safety of DOX chemotherapy in clinical application.
Collapse
Affiliation(s)
- Linyuan Peng
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Minxian Qian
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Zuojun Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Xiaolong Tang
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Jie Sun
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Yue Jiang
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Shimin Sun
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo 255049, China
| | - Xinyue Cao
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China
| | - Qiuxiang Pang
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo 255049, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China.,Carson International Cancer Center, Shenzhen University Health Science Center, Shenzhen 518055, China.,Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
13
|
Yang Y, Yang G, Yu L, Lin L, Liu L, Fang M, Xu Y. An Interplay Between MRTF-A and the Histone Acetyltransferase TIP60 Mediates Hypoxia-Reoxygenation Induced iNOS Transcription in Macrophages. Front Cell Dev Biol 2020; 8:484. [PMID: 32626711 PMCID: PMC7315810 DOI: 10.3389/fcell.2020.00484] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 05/22/2020] [Indexed: 01/23/2023] Open
Abstract
Cardiac ischemia-reperfusion injury (IRI) represents a major pathophysiological event associated with permanent loss of heart function. Several inter-dependent processes contribute to cardiac IRI that include accumulation of reactive oxygen species (ROS), aberrant inflammatory response, and depletion of energy supply. Inducible nitric oxide synthase (iNOS) is a pro-inflammatory mediator and a major catalyst of ROS generation. In the present study we investigated the epigenetic mechanism whereby iNOS transcription is up-regulated in macrophages in the context of cardiac IRI. We report that germline deletion or systemic inhibition of myocardin-related transcription factor A (MRTF-A) in mice attenuated up-regulation of iNOS following cardiac IRI in the heart. In cultured macrophages, depletion or inhibition of MRTF-A suppressed iNOS induction by hypoxia-reoxygenation (HR). In contrast, MRTF-A over-expression potentiated activation of the iNOS promoter by HR. MRTF-A directly binds to the iNOS promoter in response to HR stimulation. MRTF-A binding to the iNOS promoter was synonymous with active histone modifications including trimethylated H3K4, acetylated H3K9, H3K27, and H4K16. Further analysis revealed that MRTF-A interacted with H4K16 acetyltransferase TIP60 to synergistically activate iNOS transcription. TIP60 depletion or inhibition achieved equivalent effects as MRTF-A depletion/inhibition in terms of iNOS repression. Of interest, TIP60 appeared to form a crosstalk with the H3K4 trimethyltransferase complex to promote iNOS trans-activation. In conclusion, we data suggest that the MRTF-A-TIP60 axis may play a critical role in iNOS transcription in macrophages and as such be considered as a potential target for the intervention of cardiac IRI.
Collapse
Affiliation(s)
- Yuyu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China.,Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Guang Yang
- Department of Pathology, Soochow Municipal Hospital Affiliated with Nanjing Medical University, Soochow, China
| | - Liming Yu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Disease, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ling Lin
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Li Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Disease, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Mingming Fang
- Center for Experimental Medicine, Jiangsu Health Vocational College, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yong Xu
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Disease, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Lam B, Roudier E. Considering the Role of Murine Double Minute 2 in the Cardiovascular System? Front Cell Dev Biol 2020; 7:320. [PMID: 31921839 PMCID: PMC6916148 DOI: 10.3389/fcell.2019.00320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/21/2019] [Indexed: 01/26/2023] Open
Abstract
The E3 ubiquitin ligase Murine double minute 2 (MDM2) is the main negative regulator of the tumor protein p53 (TP53). Extensive studies over more than two decades have confirmed MDM2 oncogenic role through mechanisms both TP53-dependent and TP53-independent oncogenic function. These studies have contributed to designate MDM2 as a therapeutic target of choice for cancer treatment and the number of patents for MDM2 antagonists has increased immensely over the last years. However, the question of the physiological functions of MDM2 has not been fully resolved yet, particularly when expressed and regulated physiologically in healthy tissue. Cardiovascular complications are almost an inescapable side-effect of anti-cancer therapies. While several MDM2 antagonists are entering phase I, II and even III of clinical trials, this review proposes to bring awareness on the physiological role of MDM2 in the cardiovascular system.
Collapse
Affiliation(s)
- Brian Lam
- Angiogenesis Research Group, School of Kinesiology and Health Sciences, Muscle Health Research Center, Faculty of Health, York University, Toronto, ON, Canada
| | - Emilie Roudier
- Angiogenesis Research Group, School of Kinesiology and Health Sciences, Muscle Health Research Center, Faculty of Health, York University, Toronto, ON, Canada
| |
Collapse
|
15
|
Spearman AD, Ke X, Fu Q, Lane RH, Majnik A. Adverse maternal environment leads to cardiac fibrosis in adult male mice. Birth Defects Res 2019; 110:1551-1555. [PMID: 30576090 DOI: 10.1002/bdr2.1428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/21/2018] [Accepted: 11/02/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cardiac fibrosis is a cardinal feature of multiple types of cardiovascular disease, which lead to heart failure. Multiple studies connect adverse maternal environment (AME) with cardiac fibrosis. AME does not always result in fibrosis, though. An additional "insult", such as an adult Western diet (WD), is frequently necessary. The additive effects of AME and adult WD on cardiac fibrosis is not well-understood. AME can also alter DNA methylation. DNA methyltransferase (DNMT) and ten-eleven translocation (TET) are methylation modifying genes that regulate DNA methylation, but it is unknown if AME changes cardiac gene expression of DNMT and TET. We sought to use a model of AME and adult WD to investigate the development of cardiac fibrosis and cardiac mRNA expression of DNMT and TET genes. METHODS We exposed dams to WD or control diet (CD) 5 weeks before pregnancy and through lactation. We added environmental stressors during the last third of pregnancy to dams on WD to create AME. Dams on CD experienced no added stressors to create control maternal environment (CME). Male offspring were weaned at Postnatal Week 3 (W3) and placed on WD or CD to create four groups: CME-CD, CME-WD, AME-CD, and AME-WD. RESULTS AME-WD increased cardiac fibrosis in adulthood (p < .05), whereas AME-CD and CME-WD did not. TET1-3 and DNMT3a mRNA levels decreased in AME versus CME offspring (p < .01). CONCLUSION AME increases susceptibility to cardiac fibrosis in adult male mice. Early-life changes to TET expression may mediate susceptibility to fibrosis, but further testing is needed.
Collapse
Affiliation(s)
- Andrew D Spearman
- Department of Pediatrics, Division of Cardiology, Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xingrao Ke
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Qi Fu
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Robert H Lane
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Amber Majnik
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
16
|
Henderson J, Distler J, O'Reilly S. The Role of Epigenetic Modifications in Systemic Sclerosis: A Druggable Target. Trends Mol Med 2019; 25:395-411. [PMID: 30858032 DOI: 10.1016/j.molmed.2019.02.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 02/07/2023]
Abstract
Systemic sclerosis (SSc) is a rare autoimmune disorder characterised by skin fibrosis that often also affects internal organs, eventually resulting in mortality. Although management of the symptoms has extended lifespan, patients still suffer from poor quality of life, hence the need for improved therapies. Development of efficacious treatments has been stymied by the unknown aetiology, although recent advancements suggest a potentially key role for epigenetics - the regulation of gene expression by noncoding RNAs and chemical modifications to DNA or DNA-associated proteins. Herein, the evidence implicating epigenetics in the pathogenesis of SSc is discussed with an emphasis on the therapeutic potential this introduces to the field - particularly the repurposing of epigenetic targeting cancer therapeutics and newly emerging miRNA-based strategies.
Collapse
Affiliation(s)
- John Henderson
- Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Tyne and Wear, Newcastle upon Tyne NE2 8ST, UK
| | - Joerg Distler
- Department of Internal Medicine 3, Erlangen University, Erlangen, Germany
| | - Steven O'Reilly
- Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Tyne and Wear, Newcastle upon Tyne NE2 8ST, UK.
| |
Collapse
|
17
|
Phosphorylation of TIP60 Suppresses 53BP1 Localization at DNA Damage Sites. Mol Cell Biol 2018; 39:MCB.00209-18. [PMID: 30297459 DOI: 10.1128/mcb.00209-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/25/2018] [Indexed: 12/20/2022] Open
Abstract
A proper balance between the repair of DNA double-strand breaks (DSBs) by homologous recombination and nonhomologous end joining is critical for maintaining genome integrity and preventing tumorigenesis. This balance is regulated and fine-tuned by a variety of factors, including cell cycle and the chromatin environment. The histone acetyltransferase TIP60 was previously shown to suppress pathological end joining and promote homologous recombination. However, it is unknown how regulatory posttranslational modifications impact TIP60 acetyltransferase activity to influence the outcome of DSB responses. In this study, we report that phosphorylation of TIP60 on serines 90 and 86 is important for limiting the accumulation of the pro-end joining factor 53BP1 at DSBs in S and G2 cell cycle phases. Mutation of these sites disrupts histone acetylation changes in response to DNA damage, BRCA1 localization to DSBs, and poly(ADP-ribose) polymerase (PARP) inhibitor resistance. These findings reveal that phosphorylation directs TIP60-dependent acetylation to promote homologous recombination and maintain genome stability.
Collapse
|