1
|
Yao W, Hu X, Wang X. Crossing epigenetic frontiers: the intersection of novel histone modifications and diseases. Signal Transduct Target Ther 2024; 9:232. [PMID: 39278916 PMCID: PMC11403012 DOI: 10.1038/s41392-024-01918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/30/2024] [Indexed: 09/18/2024] Open
Abstract
Histone post-translational modifications (HPTMs), as one of the core mechanisms of epigenetic regulation, are garnering increasing attention due to their close association with the onset and progression of diseases and their potential as targeted therapeutic agents. Advances in high-throughput molecular tools and the abundance of bioinformatics data have led to the discovery of novel HPTMs which similarly affect gene expression, metabolism, and chromatin structure. Furthermore, a growing body of research has demonstrated that novel histone modifications also play crucial roles in the development and progression of various diseases, including various cancers, cardiovascular diseases, infectious diseases, psychiatric disorders, and reproductive system diseases. This review defines nine novel histone modifications: lactylation, citrullination, crotonylation, succinylation, SUMOylation, propionylation, butyrylation, 2-hydroxyisobutyrylation, and 2-hydroxybutyrylation. It comprehensively introduces the modification processes of these nine novel HPTMs, their roles in transcription, replication, DNA repair and recombination, metabolism, and chromatin structure, as well as their involvement in promoting the occurrence and development of various diseases and their clinical applications as therapeutic targets and potential biomarkers. Moreover, this review provides a detailed overview of novel HPTM inhibitors targeting various targets and their emerging strategies in the treatment of multiple diseases while offering insights into their future development prospects and challenges. Additionally, we briefly introduce novel epigenetic research techniques and their applications in the field of novel HPTM research.
Collapse
Affiliation(s)
- Weiyi Yao
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xinting Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
| |
Collapse
|
2
|
Dubey SK, Dubey R, Prajapati SC, Jung K, Mohan K, Liu X, Roney J, Tian W, Abney J, Giarmarco MM, Hernandez AG, Liu J, Kleinman ME. Histone deficiency and hypoacetylation in the aging retinal pigment epithelium. Aging Cell 2024; 23:e14108. [PMID: 38408164 PMCID: PMC11113634 DOI: 10.1111/acel.14108] [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: 11/08/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
Histones serve as a major carrier of epigenetic information in the form of post-translational modifications which are vital for controlling gene expression, maintaining cell identity, and ensuring proper cellular function. Loss of histones in the aging genome can drastically impact the epigenetic landscape of the cell leading to altered chromatin structure and changes in gene expression profiles. In this study, we investigated the impact of age-related changes on histone levels and histone acetylation in the retinal pigment epithelium (RPE) and retina of mice. We observed a global reduction of histones H1, H2A, H2B, H3, and H4 in aged RPE/choroid but not in the neural retina. Transcriptomic analyses revealed significant downregulation of histones in aged RPE/choroid including crucial elements of the histone locus body (HLB) complex involved in histone pre-mRNA processing. Knockdown of HINFP, a key HLB component, in human RPE cells induced histone loss, senescence, and the upregulation of senescence-associated secretory phenotype (SASP) markers. Replicative senescence and chronological aging in human RPE cells similarly resulted in progressive histone loss and acquisition of the SASP. Immunostaining of human retina sections revealed histone loss in RPE with age. Acetyl-histone profiling in aged mouse RPE/choroid revealed a specific molecular signature with loss of global acetyl-histone levels, including H3K14ac, H3K56ac, and H4K16ac marks. These findings strongly demonstrate histone loss as a unique feature of RPE aging and provide critical insights into the potential mechanisms linking histone dynamics, cellular senescence, and aging.
Collapse
Affiliation(s)
- Sushil K. Dubey
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Rashmi Dubey
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Subhash C. Prajapati
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Kyungsik Jung
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Kabhilan Mohan
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Xinan Liu
- Department of Computer ScienceUniversity of KentuckyLexingtonKentuckyUSA
| | - Jacob Roney
- Department of Ophthalmology and Visual SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - Wenjian Tian
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Jennifer Abney
- Department of Ophthalmology and Visual SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | | | - Alvaro G. Hernandez
- Roy J. Carver Biotechnology CenterUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Jinze Liu
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Mark E. Kleinman
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| |
Collapse
|
3
|
Sikder S, Arunkumar G, Melters DP, Dalal Y. Breaking the aging epigenetic barrier. Front Cell Dev Biol 2022; 10:943519. [PMID: 35966762 PMCID: PMC9366916 DOI: 10.3389/fcell.2022.943519] [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: 05/13/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Aging is an inexorable event occurring universally for all organisms characterized by the progressive loss of cell function. However, less is known about the key events occurring inside the nucleus in the process of aging. The advent of chromosome capture techniques and extensive modern sequencing technologies have illuminated a rather dynamic structure of chromatin inside the nucleus. As cells advance along their life cycle, chromatin condensation states alter which leads to a different epigenetic landscape, correlated with modified gene expression. The exact factors mediating these changes in the chromatin structure and function remain elusive in the context of aging cells. The accumulation of DNA damage, reactive oxygen species and loss of genomic integrity as cells cease to divide can contribute to a tumor stimulating environment. In this review, we focus on genomic and epigenomic changes occurring in an aged cell which can contribute to age-related tumor formation.
Collapse
Affiliation(s)
| | | | | | - Yamini Dalal
- Chromatin Structure and Epigenetic Mechanisms, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, NCI, NIH, Bethesda, MD, United States
| |
Collapse
|
4
|
Kamimura R, Uchida D, Kanno SI, Shiraishi R, Hyodo T, Sawatani Y, Shimura M, Hasegawa T, Tsubura-Okubo M, Yaguchi E, Komiyama Y, Fukumoto C, Izumi S, Fujita A, Wakui T, Kawamata H. Identification of Binding Proteins for TSC22D1 Family Proteins Using Mass Spectrometry. Int J Mol Sci 2021; 22:ijms222010913. [PMID: 34681573 PMCID: PMC8536140 DOI: 10.3390/ijms222010913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 11/29/2022] Open
Abstract
TSC-22 (TGF-β stimulated clone-22) has been reported to induce differentiation, growth inhibition, and apoptosis in various cells. TSC-22 is a member of a family in which many proteins are produced from four different family genes. TSC-22 (corresponding to TSC22D1-2) is composed of 144 amino acids translated from a short variant mRNA of the TSC22D1 gene. In this study, we attempted to determine the intracellular localizations of the TSC22D1 family proteins (TSC22D1-1, TSC-22 (TSC22D1-2), and TSC22(86) (TSC22D1-3)) and identify the binding proteins for TSC22D1 family proteins by mass spectrometry. We determined that TSC22D1-1 was mostly localized in the nucleus, TSC-22 (TSC22D1-2) was localized in the cytoplasm, mainly in the mitochondria and translocated from the cytoplasm to the nucleus after DNA damage, and TSC22(86) (TSC22D1-3) was localized in both the cytoplasm and nucleus. We identified multiple candidates of binding proteins for TSC22D1 family proteins in in vitro pull-down assays and in vivo binding assays. Histone H1 bound to TSC-22 (TSC22D1-2) or TSC22(86) (TSC22D1-3) in the nucleus. Guanine nucleotide-binding protein-like 3 (GNL3), which is also known as nucleostemin, bound to TSC-22 (TSC22D1-2) in the nucleus. Further investigation of the interaction of the candidate binding proteins with TSC22D1 family proteins would clarify the biological roles of TSC22D1 family proteins in several cell systems.
Collapse
Affiliation(s)
- Ryouta Kamimura
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Daisuke Uchida
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon 791-0295, Ehime, Japan;
| | - Shin-ichiro Kanno
- Division of Dynamic Proteome, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-machi, Sendai 980-8575, Aobaku, Japan;
| | - Ryo Shiraishi
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Toshiki Hyodo
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Yuta Sawatani
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
- Section of Dentistry, Oral and Maxillofacial Surgery, Kamitsuga General Hospital, 1-1033 Shimoda-machi, Kanuma 322-8550, Tochigi, Japan
| | - Michiko Shimura
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
- Section of Dentistry and Oral and Maxillofacial Surgery, Sano Kosei General Hospital, 1728 Horigomecho, Sano 327-8511, Tochigi, Japan
| | - Tomonori Hasegawa
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Maki Tsubura-Okubo
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
- Section of Dentistry and Oral and Maxillofacial Surgery, Sano Kosei General Hospital, 1728 Horigomecho, Sano 327-8511, Tochigi, Japan
| | - Erika Yaguchi
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Yuske Komiyama
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Chonji Fukumoto
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Sayaka Izumi
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Atsushi Fujita
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Takahiro Wakui
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Hitoshi Kawamata
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
- Correspondence: ; Tel.: +81-282-87-2130; Fax: +81-282-86-1681
| |
Collapse
|
5
|
Saha A, Dalal Y. A glitch in the snitch: the role of linker histone H1 in shaping the epigenome in normal and diseased cells. Open Biol 2021; 11:210124. [PMID: 34343462 PMCID: PMC8331230 DOI: 10.1098/rsob.210124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Histone H1s or the linker histones are a family of dynamic chromatin compacting proteins that are essential for higher-order chromatin organization. These highly positively charged proteins were previously thought to function solely as repressors of transcription. However, over the last decade, there is a growing interest in understanding this multi-protein family, finding that not all variants act as repressors. Indeed, the H1 family members appear to have distinct affinities for chromatin and may potentially affect distinct functions. This would suggest a more nuanced contribution of H1 to chromatin organization. The advent of new technologies to probe H1 dynamics in vivo, combined with powerful computational biology, and in vitro imaging tools have greatly enhanced our knowledge of the mechanisms by which H1 interacts with chromatin. This family of proteins can be metaphorically compared to the Golden Snitch from the Harry Potter series, buzzing on and off several regions of the chromatin, in combat with competing transcription factors and chromatin remodellers, thereby critical to the epigenetic endgame on short and long temporal scales in the life of the nucleus. Here, we summarize recent efforts spanning structural, computational, genomic and genetic experiments which examine the linker histone as an unseen architect of chromatin fibre in normal and diseased cells and explore unanswered fundamental questions in the field.
Collapse
Affiliation(s)
- Ankita Saha
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yamini Dalal
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Abstract
As the main protein components of chromatin, histones play central roles in gene regulation as spools of winding DNA. Histones are subject to various modifications, including phosphorylation, acetylation, glycosylation, methylation, ubiquitination and citrullination, which affect gene transcription. Histone citrullination, a posttranscriptional modification catalyzed by peptidyl arginine deiminase (PAD) enzymes, is involved in human carcinogenesis. In this study, we highlighted the functions of histone citrullination in physiological regulation and tumors. Additionally, because histone citrullination involves forming neutrophil extracellular traps (NETs), the relationship between NETs and tumors was illustrated. Finally, the clinical application of histone citrullination and PAD inhibitors was discussed.
Collapse
Affiliation(s)
- Dongwei Zhu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212013, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yue Zhang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212013, China.
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212013, China.
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
7
|
Histone H1 Post-Translational Modifications: Update and Future Perspectives. Int J Mol Sci 2020; 21:ijms21165941. [PMID: 32824860 PMCID: PMC7460583 DOI: 10.3390/ijms21165941] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022] Open
Abstract
Histone H1 is the most variable histone and its role at the epigenetic level is less characterized than that of core histones. In vertebrates, H1 is a multigene family, which can encode up to 11 subtypes. The H1 subtype composition is different among cell types during the cell cycle and differentiation. Mass spectrometry-based proteomics has added a new layer of complexity with the identification of a large number of post-translational modifications (PTMs) in H1. In this review, we summarize histone H1 PTMs from lower eukaryotes to humans, with a particular focus on mammalian PTMs. Special emphasis is made on PTMs, whose molecular function has been described. Post-translational modifications in H1 have been associated with the regulation of chromatin structure during the cell cycle as well as transcriptional activation, DNA damage response, and cellular differentiation. Additionally, PTMs in histone H1 that have been linked to diseases such as cancer, autoimmune disorders, and viral infection are examined. Future perspectives and challenges in the profiling of histone H1 PTMs are also discussed.
Collapse
|
8
|
Staneva D, Georgieva M, Miloshev G. Kluyveromyces lactis genome harbours a functional linker histone encoding gene. FEMS Yeast Res 2016; 16:fow034. [PMID: 27189369 DOI: 10.1093/femsyr/fow034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2016] [Indexed: 11/14/2022] Open
Abstract
Linker histones are essential components of chromatin in eukaryotes. Through interactions with linker DNA and nucleosomes they facilitate folding and maintenance of higher-order chromatin structures and thus delicately modulate gene activity. The necessity of linker histones in lower eukaryotes appears controversial and dubious. Genomic data have shown that Schizosaccharomyces pombe does not possess genes encoding linker histones while Kluyveromyces lactis has been reported to have a pseudogene. Regarding this controversy, we have provided the first direct experimental evidence for the existence of a functional linker histone gene, KlLH1, in K. lactis genome. Sequencing of KlLH1 from both genomic DNA and copy DNA confirmed the presence of an intact open reading frame. Transcription and splicing of the KlLH1 sequence as well as translation of its mRNA have been studied. In silico analysis revealed homology of KlLH1p to the histone H1/H5 protein family with predicted three domain structure characteristic for the linker histones of higher eukaryotes. This strongly proves that the yeast K. lactis does indeed possess a functional linker histone gene thus entailing the evolutionary preservation and significance of linker histones. The nucleotide sequences of KlLH1 are deposited in the GenBank under accession numbers KT826576, KT826577 and KT826578.
Collapse
Affiliation(s)
- Dessislava Staneva
- Laboratory of Yeast Molecular Genetics, Institute of Molecular Biology, Bulgarian Academy of Sciences, 'Acad. Roumen Tsanev', Sofia 1113, Bulgaria
| | - Milena Georgieva
- Laboratory of Yeast Molecular Genetics, Institute of Molecular Biology, Bulgarian Academy of Sciences, 'Acad. Roumen Tsanev', Sofia 1113, Bulgaria
| | - George Miloshev
- Laboratory of Yeast Molecular Genetics, Institute of Molecular Biology, Bulgarian Academy of Sciences, 'Acad. Roumen Tsanev', Sofia 1113, Bulgaria
| |
Collapse
|
9
|
Kofman AE, Huszar JM, Payne CJ. Transcriptional analysis of histone deacetylase family members reveal similarities between differentiating and aging spermatogonial stem cells. Stem Cell Rev Rep 2013; 9:59-64. [PMID: 22729928 DOI: 10.1007/s12015-012-9392-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The differentiation of adult stem cells involves extensive chromatin remodeling, mediated in part by the gene products of histone deacetylase (HDAC) family members. While the transcriptional downregulation of HDACs can impede stem cell self-renewal in certain contexts, it may also promote stem cell maintenance under other circumstances. In self-renewing, differentiating, and aging spermatogonial stem cells (SSCs), the gene expression dynamics of HDACs have not yet been characterized. To gain further insight with these studies, we analyzed the transcriptional profiles of six HDAC family members, previously identified to be the most highly expressed in self-renewing SSCs, during stem cell differentiation and aging. Here we discovered that in both differentiating and aging SSCs the expression of Sirt4 increases, while the expression of Hdac2, Hdac6, and Sirt1 decreases. When SSCs are exposed to the lifespan-enhancing drug rapamycin in vivo, the resultant HDAC gene expression patterns are opposite of those seen in the differentiating and aging SSCs, with increased Hdac2, Hdac6, and Sirt1 and decreased Hdac8, Hdac9, and Sirt4. Our findings suggest that HDACs important for stem cell maintenance and oxidative capacity are downregulated as adult stem cells differentiate or age. These results provide important insights into the epigenetic regulation of stem cell differentiation and aging in mammals.
Collapse
Affiliation(s)
- Amber E Kofman
- Human Molecular Genetics Program, Children's Memorial Research Center, Chicago, IL 60614, USA
| | | | | |
Collapse
|
10
|
Popova EY, Grigoryev SA, Fan Y, Skoultchi AI, Zhang SS, Barnstable CJ. Developmentally regulated linker histone H1c promotes heterochromatin condensation and mediates structural integrity of rod photoreceptors in mouse retina. J Biol Chem 2013; 288:17895-907. [PMID: 23645681 DOI: 10.1074/jbc.m113.452144] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mature rod photoreceptor cells contain very small nuclei with tightly condensed heterochromatin. We observed that during mouse rod maturation, the nucleosomal repeat length increases from 190 bp at postnatal day 1 to 206 bp in the adult retina. At the same time, the total level of linker histone H1 increased reaching the ratio of 1.3 molecules of total H1 per nucleosome, mostly via a dramatic increase in H1c. Genetic elimination of the histone H1c gene is functionally compensated by other histone variants. However, retinas in H1c/H1e/H1(0) triple knock-outs have photoreceptors with bigger nuclei, decreased heterochromatin area, and notable morphological changes suggesting that the process of chromatin condensation and rod cell structural integrity are partly impaired. In triple knock-outs, nuclear chromatin exposed several epigenetic histone modification marks masked in the wild type chromatin. Dramatic changes in exposure of a repressive chromatin mark, H3K9me2, indicate that during development linker histone plays a role in establishing the facultative heterochromatin territory and architecture in the nucleus. During retina development, the H1c gene and its promoter acquired epigenetic patterns typical of rod-specific genes. Our data suggest that histone H1c gene expression is developmentally up-regulated to promote facultative heterochromatin in mature rod photoreceptors.
Collapse
Affiliation(s)
- Evgenya Y Popova
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | | | | | | | | |
Collapse
|
11
|
Novak A, Amit M, Ziv T, Segev H, Fishman B, Admon A, Itskovitz-Eldor J. Proteomics profiling of human embryonic stem cells in the early differentiation stage. Stem Cell Rev Rep 2012; 8:137-49. [PMID: 21732092 DOI: 10.1007/s12015-011-9286-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The regulatory pathways responsible for maintaining human embryonic stem cells (hESCs) in an undifferentiated state have yet to be elucidated. Since these pathways are thought to be governed by complex protein cues, deciphering the changes that occur in the proteomes of the ESCs during differentiation is important for understanding the expansion and differentiation processes involved. In this study, we present the first quantitative comparison of the hESC protein profile in the undifferentiated and early differentiated states. We used iTRAQ (isobaric tags for relative and absolute quantification) labeling combined with two dimensional capillary chromatography coupled with tandem mass spectrometry (μLC-MS/MS) to achieve comparative proteomics of hESCs at the undifferentiated stage, and at 6, 48, and 72 h after initiation of differentiation. In addition, two dimensional electrophoresis (2-DE) was performed on differentiating hESCs at eleven points of time during the first 72 h of differentiation. The results indicate that during the first 48 h of hESC differentiation, many processes are initiated and are later reversed, including chromatin remodeling, heterochromatin spreading, a decrease in transcription and translation, a decrease in glycolytic proteins and cytoskeleton remodeling, and a decrease in focal and cell adhesion. Only 72 h after differentiation induction did the expression of the homeobox prox1 protein increase, indicating the beginning of developmental processes.
Collapse
Affiliation(s)
- Atara Novak
- Sohnis and Forman Families Center for Stem Cell and Tissue Regeneration Research, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
12
|
Medrzycki M, Zhang Y, McDonald JF, Fan Y. Profiling of linker histone variants in ovarian cancer. FRONT BIOSCI-LANDMRK 2012; 17:396-406. [PMID: 22201751 DOI: 10.2741/3934] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
H1 linker histones play a key role in facilitating higher order chromatin folding. Emerging evidence suggests that H1 and its multiple variants are important epigenetic factors in modulating chromatin function and gene expression. Ovarian cancer is a devastating disease, ranking the fifth leading cause of all women cancer death due to its poor prognosis and difficulty in early diagnosis. Although epigenetic alterations in ovarian cancers are being appreciated in general, the role of H1 has not been explored. Here, using quantitative RT-PCR assays, we systematically examined the expression of 7 H1 genes in 33 human epithelial ovarian tumors. Whereas the expression of H1.3 was markedly increased, the expression of H10, H1.1, H1.4 and H1x were significantly reduced in malignant adenocarcinomas compared with benign adenomas. Strikingly, ovarian adenocarcinomas and adenomas exhibited characteristic expression patterns, and expression profiling of 7 H1 genes in tumor samples discriminated adenocarcinomas vs. adenomas with high accuracy. These findings indicate that the expression of H1 variants is exquisitely regulated and may serve as potential epigenetic biomarkers for ovarian cancer.
Collapse
Affiliation(s)
- Magdalena Medrzycki
- School of Biology, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, GA 30332, USA
| | | | | | | |
Collapse
|
13
|
Chen YJ, Tsai WH, Chen YL, Ko YC, Chou SP, Chen JY, Lin SF. Epstein-Barr virus (EBV) Rta-mediated EBV and Kaposi's sarcoma-associated herpesvirus lytic reactivations in 293 cells. PLoS One 2011; 6:e17809. [PMID: 21423768 PMCID: PMC3053391 DOI: 10.1371/journal.pone.0017809] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 02/11/2011] [Indexed: 12/15/2022] Open
Abstract
Epstein–Barr virus (EBV) Rta belongs to a lytic switch gene family that is evolutionarily conserved in all gamma-herpesviruses. Emerging evidence indicates that cell cycle arrest is a common means by which herpesviral immediate-early protein hijacks the host cell to advance the virus's lytic cycle progression. To examine the role of Rta in cell cycle regulation, we recently established a doxycycline (Dox)-inducible Rta system in 293 cells. In this cell background, inducible Rta modulated the levels of signature G1 arrest proteins, followed by induction of the cellular senescence marker, SA-β-Gal. To delineate the relationship between Rta-induced cell growth arrest and EBV reactivation, recombinant viral genomes were transferred into Rta-inducible 293 cells. Somewhat unexpectedly, we found that Dox-inducible Rta reactivated both EBV and Kaposi's sarcoma-associated herpesvirus (KSHV), to similar efficacy. As a consequence, the Rta-mediated EBV and KSHV lytic replication systems, designated as EREV8 and ERKV, respectively, were homogenous, robust, and concurrent with cell death likely due to permissive lytic replication. In addition, the expression kinetics of EBV lytic genes in Dox-treated EREV8 cells was similar to that of their KSHV counterparts in Dox-induced ERKV cells, suggesting that a common pathway is used to disrupt viral latency in both cell systems. When the time course was compared, cell cycle arrest was achieved between 6 and 48 h, EBV or KSHV reactivation was initiated abruptly at 48 h, and the cellular senescence marker was not detected until 120 h after Dox treatment. These results lead us to hypothesize that in 293 cells, Rta-induced G1 cell cycle arrest could provide (1) an ideal environment for virus reactivation if EBV or KSHV coexists and (2) a preparatory milieu for cell senescence if no viral genome is available. The latter is hypothetical in a transient-lytic situation.
Collapse
Affiliation(s)
- Yen-Ju Chen
- College of Medicine, Graduate Institute of Microbiology, National Taiwan University, Taipei, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Wan-Hua Tsai
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Yu-Lian Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Ying-Chieh Ko
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Sheng-Ping Chou
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Jen-Yang Chen
- College of Medicine, Graduate Institute of Microbiology, National Taiwan University, Taipei, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
- * E-mail: (S-FL); (J-YC)
| | - Su-Fang Lin
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County, Taiwan
- * E-mail: (S-FL); (J-YC)
| |
Collapse
|
14
|
Huynh JL, Casaccia P. Defining the chromatin landscape in demyelinating disorders. Neurobiol Dis 2009; 39:47-52. [PMID: 19853663 DOI: 10.1016/j.nbd.2009.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 10/13/2009] [Accepted: 10/14/2009] [Indexed: 01/04/2023] Open
Abstract
An intricate network of epigenetic factors regulates cell differentiation by modulating the chromatin structure and ultimately affecting gene expression. This review describes the chromatin landscape defining oligodendrocyte progenitor differentiation during development and remyelination. We shall discuss the current knowledge regarding modifications of chromatin components during the progression of progenitors into myelinating cells and discuss the potential contribution of histone variants, microRNAs, and DNA methylation. We shall also briefly address how changes to this chromatin landscape can disturb this natural progression and alter the capacity to remyelinate.
Collapse
Affiliation(s)
- Jimmy Long Huynh
- Department of Neuroscience and Genetics and Genomic Sciences, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1065, New York, NY 10029, USA
| | | |
Collapse
|
15
|
Chambery A, Colucci-D’Amato L, Vissers JPC, Scarpella S, Langridge JI, Parente A. Proteomic Profiling of Proliferating and Differentiated Neural mes-c-myc A1 Cell Line from Mouse Embryonic Mesencephalon by LC−MS. J Proteome Res 2008; 8:227-38. [DOI: 10.1021/pr800454n] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Angela Chambery
- Dipartimento di Scienze della Vita, Seconda Università di Napoli, I-81100 Caserta, Italy, Istituto di Genetica e Biofisica “A. Buzzati-Traverso”, Consiglio Nazionale delle Ricerche, 80131-Napoli, Italy, and Waters Corporation, MS Technologies Center, M22 5PP Manchester, United Kingdom
| | - Luca Colucci-D’Amato
- Dipartimento di Scienze della Vita, Seconda Università di Napoli, I-81100 Caserta, Italy, Istituto di Genetica e Biofisica “A. Buzzati-Traverso”, Consiglio Nazionale delle Ricerche, 80131-Napoli, Italy, and Waters Corporation, MS Technologies Center, M22 5PP Manchester, United Kingdom
| | - Johannes P. C. Vissers
- Dipartimento di Scienze della Vita, Seconda Università di Napoli, I-81100 Caserta, Italy, Istituto di Genetica e Biofisica “A. Buzzati-Traverso”, Consiglio Nazionale delle Ricerche, 80131-Napoli, Italy, and Waters Corporation, MS Technologies Center, M22 5PP Manchester, United Kingdom
| | - Simona Scarpella
- Dipartimento di Scienze della Vita, Seconda Università di Napoli, I-81100 Caserta, Italy, Istituto di Genetica e Biofisica “A. Buzzati-Traverso”, Consiglio Nazionale delle Ricerche, 80131-Napoli, Italy, and Waters Corporation, MS Technologies Center, M22 5PP Manchester, United Kingdom
| | - James I. Langridge
- Dipartimento di Scienze della Vita, Seconda Università di Napoli, I-81100 Caserta, Italy, Istituto di Genetica e Biofisica “A. Buzzati-Traverso”, Consiglio Nazionale delle Ricerche, 80131-Napoli, Italy, and Waters Corporation, MS Technologies Center, M22 5PP Manchester, United Kingdom
| | - Augusto Parente
- Dipartimento di Scienze della Vita, Seconda Università di Napoli, I-81100 Caserta, Italy, Istituto di Genetica e Biofisica “A. Buzzati-Traverso”, Consiglio Nazionale delle Ricerche, 80131-Napoli, Italy, and Waters Corporation, MS Technologies Center, M22 5PP Manchester, United Kingdom
| |
Collapse
|