1
|
Hu M, Fan Z. Role and mechanisms of histone methylation in osteogenic/odontogenic differentiation of dental mesenchymal stem cells. Int J Oral Sci 2025; 17:24. [PMID: 40133254 PMCID: PMC11937254 DOI: 10.1038/s41368-025-00353-z] [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: 08/26/2024] [Revised: 01/01/2025] [Accepted: 02/11/2025] [Indexed: 03/27/2025] Open
Abstract
Dental mesenchymal stem cells (DMSCs) are pivotal for tooth development and periodontal tissue health and play an important role in tissue engineering and regenerative medicine because of their multidirectional differentiation potential and self-renewal ability. The cellular microenvironment regulates the fate of stem cells and can be modified using various optimization techniques. These methods can influence the cellular microenvironment, activate disparate signaling pathways, and induce different biological effects. "Epigenetic regulation" refers to the process of influencing gene expression and regulating cell fate without altering DNA sequences, such as histone methylation. Histone methylation modifications regulate pivotal transcription factors governing DMSCs differentiation into osteo-/odontogenic lineages. The most important sites of histone methylation in tooth organization were found to be H3K4, H3K9, and H3K27. Histone methylation affects gene expression and regulates stem cell differentiation by maintaining a delicate balance between major trimethylation sites, generating distinct chromatin structures associated with specific downstream transcriptional states. Several crucial signaling pathways associated with osteogenic differentiation are susceptible to modulation via histone methylation modifications. A deeper understanding of the regulatory mechanisms governing histone methylation modifications in osteo-/odontogenic differentiation and immune-inflammatory responses of DMSCs will facilitate further investigation of the epigenetic regulation of histone methylation in DMSC-mediated tissue regeneration and inflammation. Here is a concise overview of the pivotal functions of epigenetic histone methylation at H3K4, H3K9, and H3K27 in the regulation of osteo-/odontogenic differentiation and renewal of DMSCs in both non-inflammatory and inflammatory microenvironments. This review summarizes the current research on these processes in the context of tissue regeneration and therapeutic interventions.
Collapse
Affiliation(s)
- Meijun Hu
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Zhipeng Fan
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China.
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Zhang J, Liu H, Liu Y, Luo E, Liu S. Unlocking the potential of histone modification in regulating bone metabolism. Biochimie 2024; 227:286-298. [PMID: 39154977 DOI: 10.1016/j.biochi.2024.08.004] [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: 05/23/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/20/2024]
Abstract
Bone metabolism plays a crucial role in maintaining normal bone tissue homeostasis and function. Imbalances between bone formation and resorption can lead to osteoporosis, osteoarthritis, and other bone diseases. The dynamic and complex process of bone remodeling is driven by various factors, including epigenetics. Histone modification, one of the most important and well-studied components of epigenetic regulation, has emerged as a promising area of research in bone metabolism. Different histone proteins and modification sites exert diverse effects on osteogenesis and osteoclastogenesis. In this review, we summarize recent progress in understanding histone modifications in bone metabolism, including specific modification sites and potential regulatory enzymes. Comprehensive knowledge of histone modifications in bone metabolism could reveal new therapeutic targets and treatment strategies for bone diseases.
Collapse
Affiliation(s)
- Jiayuan Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
3
|
Rummukainen P, Tarkkonen K, Al Majidi R, Puolakkainen T, Nieminen-Pihala V, Valensisi C, Saastamoinen L, Hawkins D, Heino TJ, Ivaska KK, Kiviranta R. The complex role of Rcor2: Regulates mesenchymal stromal cell differentiation in vitro but is dispensable in vivo. Bone 2024; 187:117180. [PMID: 38944098 DOI: 10.1016/j.bone.2024.117180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Recent research has revealed several important pathways of epigenetic regulation leading to transcriptional changes in bone cells. Rest Corepressor 2 (Rcor2) is a coregulator of Lysine-specific histone demethylase 1 (Lsd1), a demethylase linked to osteoblast activity, hematopoietic stem cell differentiation and malignancy of different neoplasms. However, the role of Rcor2 in osteoblast differentiation has not yet been examined in detail. We have previously shown that Rcor2 is highly expressed in mesenchymal stromal cells (MSC) and particularly in the osteoblastic lineage. The role of Rcor2 in osteoblastic differentiation in vitro was further characterized and we demonstrate here that lentiviral silencing of Rcor2 in MC3T3-E1 cells led to a decrease in osteoblast differentiation. This was indicated by decreased alkaline phosphatase and von Kossa stainings as well as by decreased expression of several osteoblast-related marker genes. RNA-sequencing of the Rcor2-downregulated MC3T3-E1 cells showed decreased repression of Rcor2 target genes, as well as significant upregulation of majority of the differentially expressed genes. While the heterozygous, global loss of Rcor2 in vivo did not lead to a detectable bone phenotype, conditional deletion of Rcor2 in limb-bud mesenchymal cells led to a moderate decrease in cortical bone volume. These findings were not accentuated by challenging bone formation by ovariectomy or tibial fracture. Furthermore, a global deletion of Rcor2 led to decreased white adipose tissue in vivo and decreased the capacity of primary cells to differentiate into adipocytes in vitro. The conditional deletion of Rcor2 led to decreased adiposity in fracture callus. Taken together, these results suggest that epigenetic regulation of mesenchymal stromal cell differentiation is mediated by Rcor2, which could thus play an important role in defining the MSC fate.
Collapse
Affiliation(s)
- Petri Rummukainen
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland.
| | - Kati Tarkkonen
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland
| | - Rana Al Majidi
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland
| | - Tero Puolakkainen
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland
| | - Vappu Nieminen-Pihala
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland
| | - Cristina Valensisi
- Division of Medical Genetics, Department of Medicine, University of Washington, United States of America, Division of Medical Genetics Health Sciences Building, Rm K253 Box 357720, Seattle, WA 98195-7720
| | - Lauri Saastamoinen
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland
| | - David Hawkins
- Division of Medical Genetics, Department of Medicine, University of Washington, United States of America, Division of Medical Genetics Health Sciences Building, Rm K253 Box 357720, Seattle, WA 98195-7720
| | - Terhi J Heino
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland
| | - Kaisa K Ivaska
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland
| | - Riku Kiviranta
- Institute of Biomedicine, University of Turku, Turku, Faculty of Medicine, FI-20014, Finland; Department of Endocrinology, Turku University Hospital, PO Box 52 20521, Turku, Finland
| |
Collapse
|
4
|
Dashti P, Lewallen EA, Gordon JAR, Montecino MA, Davie JR, Stein GS, van Leeuwen JPTM, van der Eerden BCJ, van Wijnen AJ. Epigenetic regulators controlling osteogenic lineage commitment and bone formation. Bone 2024; 181:117043. [PMID: 38341164 DOI: 10.1016/j.bone.2024.117043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/08/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
Bone formation and homeostasis are controlled by environmental factors and endocrine regulatory cues that initiate intracellular signaling pathways capable of modulating gene expression in the nucleus. Bone-related gene expression is controlled by nucleosome-based chromatin architecture that limits the accessibility of lineage-specific gene regulatory DNA sequences and sequence-specific transcription factors. From a developmental perspective, bone-specific gene expression must be suppressed during the early stages of embryogenesis to prevent the premature mineralization of skeletal elements during fetal growth in utero. Hence, bone formation is initially inhibited by gene suppressive epigenetic regulators, while other epigenetic regulators actively support osteoblast differentiation. Prominent epigenetic regulators that stimulate or attenuate osteogenesis include lysine methyl transferases (e.g., EZH2, SMYD2, SUV420H2), lysine deacetylases (e.g., HDAC1, HDAC3, HDAC4, HDAC7, SIRT1, SIRT3), arginine methyl transferases (e.g., PRMT1, PRMT4/CARM1, PRMT5), dioxygenases (e.g., TET2), bromodomain proteins (e.g., BRD2, BRD4) and chromodomain proteins (e.g., CBX1, CBX2, CBX5). This narrative review provides a broad overview of the covalent modifications of DNA and histone proteins that involve hundreds of enzymes that add, read, or delete these epigenetic modifications that are relevant for self-renewal and differentiation of mesenchymal stem cells, skeletal stem cells and osteoblasts during osteogenesis.
Collapse
Affiliation(s)
- Parisa Dashti
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Eric A Lewallen
- Department of Biological Sciences, Hampton University, Hampton, VA, USA
| | | | - Martin A Montecino
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad Andres Bello, Santiago, Chile; Millennium Institute Center for Genome Regulation (CRG), Santiago, Chile
| | - James R Davie
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada; CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, Manitoba R3E 0V9, Canada.
| | - Gary S Stein
- Department of Biochemistry, University of Vermont, Burlington, VT, USA
| | | | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands.
| | - Andre J van Wijnen
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Biochemistry, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
5
|
Karakatsanis NM, Hamey JJ, Wilkins MR. Taking Me away: the function of phosphorylation on histone lysine demethylases. Trends Biochem Sci 2024; 49:257-276. [PMID: 38233282 DOI: 10.1016/j.tibs.2023.12.004] [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: 07/27/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 01/19/2024]
Abstract
Histone lysine demethylases (KDMs) regulate eukaryotic gene transcription by catalysing the removal of methyl groups from histone proteins. These enzymes are intricately regulated by the kinase signalling system in response to internal and external stimuli. Here, we review the mechanisms by which kinase-mediated phosphorylation influence human histone KDM function. These include the changing of histone KDM subcellular localisation or chromatin binding, the altering of protein half-life, changes to histone KDM complex formation that result in histone demethylation, non-histone demethylation or demethylase-independent effects, and effects on histone KDM complex dissociation. We also explore the structural context of phospho-sites on histone KDMs and evaluate how this relates to function.
Collapse
Affiliation(s)
- Nicola M Karakatsanis
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, Australia
| | - Joshua J Hamey
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, Australia
| | - Marc R Wilkins
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, Australia.
| |
Collapse
|
6
|
Xia SL, Ma ZY, Wang B, Gao F, Guo SY, Chen XH. Icariin promotes the proliferation and osteogenic differentiation of bone-derived mesenchymal stem cells in patients with osteoporosis and T2DM by upregulating GLI-1. J Orthop Surg Res 2023; 18:500. [PMID: 37454090 DOI: 10.1186/s13018-023-03998-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND The function of mesenchymal stem cells (MSCs) from patients with osteoporosis (OP) is impaired and worsens in patients with type 2 diabetes mellitus (T2DM). Icariin (ICA) is the major active flavonoid glucoside isolated from traditional Chinese herbal Epimedium pubescens, and confirmed able to improve bone mass of OP patients. OBJECTIVE To investigate the effect of ICA on the proliferation and osteogenic differentiation of bone-derived MSCs (BMSCs) from patients with OP and T2DM and uncover the potential mechanism. METHODS BMSCs were treated with ICA, and proliferation and osteogenic potency were evaluated using the 2,5-diphenyl-2H-tetrazolium bromide (MTT) assay and detection of osteogenic markers (ALP, RUNX2, SPP1, COL1A1, and mineralized nodules) was performed. RNA sequencing and bioinformatic analysis were performed to identify differentially expressed genes (DEGs) after ICA treatment and screen proliferation- and osteogenic differentiation-related processes. Gene gain and loss were performed to confirm the role of the key candidate gene. RESULTS ICA significantly promoted the proliferation and osteogenic differentiation of BMSCs. A total of 173 DEGs were identified after ICA treatment. Six DEGs (GLI-1, IGF2, BMP6, WNT5A, PTHLH, and MAPK14) enriched in both proliferation- and osteogenic differentiation-related processes were screened; GLI-1 had the highest validated |log2FC| value. Overexpression of GLI-1 enhanced the proliferation and osteogenic differentiation of BMSCs, and knockdown of GLI-1 weakened the positive effect of ICA on BMSCs. CONCLUSION ICA promoted the proliferation and osteogenic differentiation of impaired BMSCs by upregulating GLI-1.
Collapse
Affiliation(s)
- Sheng-Li Xia
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Zi-Yuan Ma
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Bin Wang
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Feng Gao
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Sheng-Yang Guo
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Xu-Han Chen
- Zhoupu Community Health Service Center, 163 Shenmei East Road, Pudong New Area, Shanghai, 201318, China.
| |
Collapse
|
7
|
Gopinathan G, Luan X, Diekwisch TGH. Epigenetic Repression of RUNX2 and OSX Promoters Controls the Nonmineralized State of the Periodontal Ligament. Genes (Basel) 2023; 14:201. [PMID: 36672941 PMCID: PMC9858805 DOI: 10.3390/genes14010201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
The nonmineralized state of the mammalian periodontal ligament is one of the hallmarks of vertebrate evolution as it provides resilient and nontraumatic tooth anchorage for effective predation. Here we sought to determine how the chromatin state of key mineralization gene promoters contributes to the nonmineralized periodontal ligament in the midst of fully mineralized alveolar bone and cementum anchor tissues. In developing mouse periodontal tissues, RUNX2 was localized to alveolar bone-lining cells, while OSX was localized throughout the periodontal ligament's soft tissue. Matching RT-PCR amplification data and western blot comparisons demonstrated that the expression of RUNX2 and OSX bone mineralization transcription factors was at least 2.5-fold elevated in alveolar bone osteoblasts versus periodontal ligament fibroblasts. ChIP enrichment data along the RUNX2 and OSX promoters revealed increased H3K4me3 marks in alveolar bone osteoblasts, while H3K9me3 and H3K27me3 marks were elevated in periodontal ligament fibroblasts. In support of an epigenetic mechanism responsible for the inhibition of mineralization gene expression in periodontal progenitors, histone methylation inhibitors DZNep and Chaetocin reactivated RUNX2 and OSX expression in periodontal progenitors and increased alkaline phosphatase and Alizarin Red, while the in vivo application of DZNep in rat maxillae resulted in aberrant mineralization in the periodontal ligament and a narrowing of the nonmineralized periodontal space. Together, these studies demonstrate that the nonmineralized state of the mammalian periodontal ligament is controlled by an epigenetic regulation of the RUNX2 and OSX key mineralization gene promoters.
Collapse
Affiliation(s)
- Gokul Gopinathan
- Center for Craniofacial Research and Diagnosis, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Xianghong Luan
- Center for Craniofacial Research and Diagnosis, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Thomas G. H. Diekwisch
- Department of Oral and Craniofacial Sciences, University of Rochester School of Medicine and Dentistry, 625 Elmwood Avenue, Rochester, NY 14620, USA
| |
Collapse
|
8
|
Chen Q, Sinha KM, de Crombrugghe B, Krahe R. Osteoblast-Specific Overexpression of Nucleolar Protein NO66/RIOX1 in Mouse Embryos Leads to Osteoporosis in Adult Mice. J Osteoporos 2023; 2023:8998556. [PMID: 36660551 PMCID: PMC9845042 DOI: 10.1155/2023/8998556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
In previous study, we showed that nucleolar protein 66 (NO66) is a chromatin modifier and negatively regulates Osterix activity as well as mesenchymal progenitor differentiation. Genetic ablation of the NO66 (RIOX1) gene in cells of the Prx1-expressing mesenchymal lineage leads to acceleration of osteochondrogenic differentiation and a larger skeleton in adult mice, whereas mesenchyme-specific overexpression of NO66 inhibits osteochondrogenesis resulting in dwarfism and osteopenia. However, the impact of NO66 overexpression in cells of the osteoblast lineage in vivo remains largely undefined. Here, we generated osteoblast-specific transgenic mice overexpressing a FLAG-tagged NO66 transgene driven by the 2.3 kB alpha-1type I collagen (Col1a1) promoter. We found that overexpression of NO66 in cells of the osteoblast lineage did not cause overt defects in developmental bones but led to osteoporosis in the long bones of adult mice. This includes decreased bone volume (BV), bone volume density (bone volume/total volume, BV/TV), and bone mineral density (BMD) in cancellous compartment of long bones, along with the accumulation of fatty droplets in bone marrow. Ex vivo culture of the bone marrow mesenchymal stem/stromal cells (BMSCs) from adult Col1a1-NO66 transgenic mice showed an increase in adipogenesis and a decrease in osteogenesis. Taken together, these data demonstrate a crucial role for NO66 in adult bone formation and homeostasis. Our Col1a1-NO66 transgenic mice provide a novel animal model for the mechanistic and therapeutic study of NO66 in osteoporosis.
Collapse
Affiliation(s)
- Qin Chen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Krishna M. Sinha
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Benoit de Crombrugghe
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ralf Krahe
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
9
|
Rummukainen P, Tarkkonen K, Dudakovic A, Al-Majidi R, Nieminen-Pihala V, Valensisi C, Hawkins RD, van Wijnen AJ, Kiviranta R. Lysine-Specific Demethylase 1 (LSD1) epigenetically controls osteoblast differentiation. PLoS One 2022; 17:e0265027. [PMID: 35255108 PMCID: PMC8901060 DOI: 10.1371/journal.pone.0265027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/18/2022] [Indexed: 02/03/2023] Open
Abstract
Epigenetic mechanisms regulate osteogenic lineage differentiation of mesenchymal stromal cells. Histone methylation is controlled by multiple lysine demethylases and is an important step in controlling local chromatin structure and gene expression. Here, we show that the lysine-specific histone demethylase Kdm1A/Lsd1 is abundantly expressed in osteoblasts and that its suppression impairs osteoblast differentiation and bone nodule formation in vitro. Although Lsd1 knockdown did not affect global H3K4 methylation levels, genome-wide ChIP-Seq analysis revealed high levels of Lsd1 at gene promoters and its binding was associated with di- and tri-methylation of histone 3 at lysine 4 (H3K4me2 and H3K4me3). Lsd1 binding sites in osteoblastic cells were enriched for the Runx2 consensus motif suggesting a functional link between the two proteins. Importantly, inhibition of Lsd1 activity decreased osteoblast activity in vivo. In support, mesenchymal-targeted knockdown of Lsd1 led to decreased osteoblast activity and disrupted primary spongiosa ossification and reorganization in vivo. Together, our studies demonstrate that Lsd1 occupies Runx2-binding cites at H3K4me2 and H3K4me3 and its activity is required for proper bone formation.
Collapse
Affiliation(s)
| | - Kati Tarkkonen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, United States of America
| | - Rana Al-Majidi
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Cristina Valensisi
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - R. David Hawkins
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Andre J. van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, United States of America
- Department of Biochemistry, University of Vermont, Burlington, VT, United States of America
- * E-mail: (AJW); (RK)
| | - Riku Kiviranta
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
- * E-mail: (AJW); (RK)
| |
Collapse
|
10
|
Ma Q, Song C, Yin B, Shi Y, Ye L. The role of Trithorax family regulating osteogenic and Chondrogenic differentiation in mesenchymal stem cells. Cell Prolif 2022; 55:e13233. [PMID: 35481717 PMCID: PMC9136489 DOI: 10.1111/cpr.13233] [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: 01/12/2022] [Revised: 03/17/2022] [Accepted: 03/28/2022] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) hold great promise and clinical efficacy in bone/cartilage regeneration. With a deeper understanding of stem cell biology over the past decade, epigenetics stands out as one of the most promising ways to control MSCs differentiation. Trithorax group (TrxG) proteins, including the COMPASS family, ASH1L, CBP/p300 as histone modifying factors, and the SWI/SNF complexes as chromatin remodelers, play an important role in gene expression regulation during the process of stem cell differentiation. This review summarises the components and functions of TrxG complexes. We provide an overview of the regulation mechanisms of TrxG in MSCs osteogenic and chondrogenic differentiation, and discuss the prospects of epigenetic regulation mediated by TrxG in bone and cartilage regeneration.
Collapse
Affiliation(s)
- Qingge Ma
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenghao Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bei Yin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Chakraborty S, Sinha S, Sengupta A. Emerging trends in chromatin remodeler plasticity in mesenchymal stromal cell function. FASEB J 2020; 35:e21234. [PMID: 33337557 DOI: 10.1096/fj.202002232r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022]
Abstract
Emerging evidences highlight importance of epigenetic regulation and their integration with transcriptional and cell signaling machinery in determining tissue resident adult pluripotent mesenchymal stem/stromal cell (MSC) activity, lineage commitment, and multicellular development. Histone modifying enzymes and large multi-subunit chromatin remodeling complexes and their cell type-specific plasticity remain the central defining features of gene regulation and establishment of tissue identity. Modulation of transcription factor expression gradient ex vivo and concomitant flexibility of higher order chromatin architecture in response to signaling cues are exciting approaches to regulate MSC activity and tissue rejuvenation. Being an important constituent of the adult bone marrow microenvironment/niche, pathophysiological perturbation in MSC homeostasis also causes impaired hematopoietic stem/progenitor cell function in a non-cell autonomous mechanism. In addition, pluripotent MSCs can function as immune regulatory cells, and they reside at the crossroad of innate and adaptive immune response pathways. Research in the past few years suggest that MSCs/stromal fibroblasts significantly contribute to the establishment of immunosuppressive microenvironment in shaping antitumor immunity. Therefore, it is important to understand mesenchymal stromal epigenome and transcriptional regulation to leverage its applications in regenerative medicine, epigenetic memory-guided trained immunity, immune-metabolic rewiring, and precision immune reprogramming. In this review, we highlight the latest developments and prospects in chromatin biology in determining MSC function in the context of lineage commitment and immunomodulation.
Collapse
Affiliation(s)
- Sayan Chakraborty
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| | - Sayantani Sinha
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| | - Amitava Sengupta
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| |
Collapse
|
12
|
Liu Q, Li M, Wang S, Xiao Z, Xiong Y, Wang G. Recent Advances of Osterix Transcription Factor in Osteoblast Differentiation and Bone Formation. Front Cell Dev Biol 2020; 8:601224. [PMID: 33384998 PMCID: PMC7769847 DOI: 10.3389/fcell.2020.601224] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
With increasing life expectations, more and more patients suffer from fractures either induced by intensive sports or other bone-related diseases. The balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption is the basis for maintaining bone health. Osterix (Osx) has long been known to be an essential transcription factor for the osteoblast differentiation and bone mineralization. Emerging evidence suggests that Osx not only plays an important role in intramembranous bone formation, but also affects endochondral ossification by participating in the terminal cartilage differentiation. Given its essentiality in skeletal development and bone formation, Osx has become a new research hotspot in recent years. In this review, we focus on the progress of Osx's function and its regulation in osteoblast differentiation and bone mass. And the potential role of Osx in developing new therapeutic strategies for osteolytic diseases was discussed.
Collapse
Affiliation(s)
- Qian Liu
- Key Laboratory of Brain and Neuroendocrine Diseases, College of Hunan Province, Hunan University of Medicine, Huaihua, China
- Biomedical Research Center, Hunan University of Medicine, Huaihua, China
| | - Mao Li
- Biomedical Research Center, Hunan University of Medicine, Huaihua, China
| | - Shiyi Wang
- XiangYa School of Medicine, Central South University, Changsha, China
| | - Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yuanyuan Xiong
- Key Laboratory of Brain and Neuroendocrine Diseases, College of Hunan Province, Hunan University of Medicine, Huaihua, China
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guangwei Wang
- Key Laboratory of Brain and Neuroendocrine Diseases, College of Hunan Province, Hunan University of Medicine, Huaihua, China
- Biomedical Research Center, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Although many signalling pathways have been discovered to be essential in mesenchymal stem/stromal (MSC) differentiation, it has become increasingly clear in recent years that epigenetic regulation of gene transcription is a vital component of lineage determination, encompassing diet, lifestyle and parental influences on bone, fat and cartilage development. RECENT FINDINGS This review discusses how specific enzymes that modify histone methylation and acetylation or DNA methylation orchestrate the differentiation programs in lineage determination of MSC and the epigenetic changes that facilitate development of bone related diseases such as osteoporosis. The review also describes how environmental factors such as mechanical loading influence the epigenetic signatures of MSC, and how the use of chemical agents or small peptides can regulate epigenetic drift in MSC populations during ageing and disease. Epigenetic regulation of MSC lineage commitment is controlled through changes in enzyme activity, which modifies DNA and histone residues leading to alterations in chromatin structure. The co-ordinated epigenetic regulation of transcriptional activation and repression act to mediate skeletal tissue homeostasis, where deregulation of this process can lead to bone loss during ageing or osteoporosis.
Collapse
Affiliation(s)
- Dimitrios Cakouros
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| |
Collapse
|
14
|
Zhang L, Chen Q, Chen Z, Wang Y, Gamboa JL, Ikizler TA, Garibotto G, Mitch WE. Mechanisms Regulating Muscle Protein Synthesis in CKD. J Am Soc Nephrol 2020; 31:2573-2587. [PMID: 32764136 DOI: 10.1681/asn.2019121277] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/21/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND CKD induces loss of muscle proteins partly by suppressing muscle protein synthesis. Muscles of mice with CKD have increased expression of nucleolar protein 66 (NO66), as do muscle biopsy specimens from patients with CKD or those undergoing hemodialysis. Inflammation stimulates NO66 expression and changes in NF-κB mediate the response. METHODS Subtotal nephrectomy created a mouse model of CKD with BUN >80 mg/dl. Crossing NO66flox/flox with MCK-Cre mice bred muscle-specific NO66 (MCK-NO66) knockout mice. Experiments assessed the effect of removing NO66. RESULTS Muscle-specific NO66 knockout in mice blocks CKD-induced loss of muscle mass and improves protein synthesis. NO66 suppression of ribosomal biogenesis via demethylase activity is the mechanism behind these responses. In muscle cells, expression of NO66, but not of demethylase-dead mutant NO66, decreased H3K4me3 and H3K36me3 and suppressed pre-rRNA expression. Knocking out NO66 increased the enrichment of H3K4me3 and H3K36me3 on ribosomal DNA. In primary muscle cells and in muscles of mice without NO66, ribosomal RNA, pre-rRNA, and protein synthesis all increased. CONCLUSIONS CKD suppresses muscle protein synthesis via epigenetic mechanisms that NO66 mediates. Blocking NO66 could suggest strategies that counter CKD-induced abnormal muscle protein catabolism.
Collapse
Affiliation(s)
- Liping Zhang
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Qin Chen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zihong Chen
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Ying Wang
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Jorge L Gamboa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Talat Alp Ikizler
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Giacomo Garibotto
- Nephrology Division, Department of Internal Medicine, Genoa University, Scientific Hospitalization and Treatment Institute Policlinico San Martino Hospital, Genoa, Italy
| | - William E Mitch
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
15
|
Kim I, Park JW. Hypoxia-driven epigenetic regulation in cancer progression: A focus on histone methylation and its modifying enzymes. Cancer Lett 2020; 489:41-49. [PMID: 32522693 DOI: 10.1016/j.canlet.2020.05.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/28/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
Abstract
The mechanism underlying hypoxia-driven chromatin remodeling is a long-lasting question. For the last two decades, this question has been resolved in part. It is now widely agreed that hypoxia dynamically changes the methylation status of histones to control gene expression. Hypoxia-inducible factor (HIF) plays a central role in cellular responses to hypoxia through transcriptional activation of numerous genes. At least in part, the hypoxic regulation of histone methylation is attributed to the HIF-mediated expression of histone modifying enzymes. Protein hydroxylation and histone demethylation have emerged as the oxygen sensing processes because they are catalyzed by a family of 2-oxoglutarate (2OG)-dependent dioxygenases whose activities depend upon the ambient oxygen level. Recently, it has been extensively investigated that the 2OG dioxygenases oxygen-dependently regulate histone methylation. Nowadays, the hypoxic change in the histone methylation status is regarded as an important event to drive malignant behaviors of cancer cells. In this review, we introduced and summarized the cellular processes that govern hypoxia-driven regulation of histone methylation in the context of cancer biology. We also discussed the emerging roles of histone methyltransferases and demethylases in epigenetic response to hypoxia.
Collapse
Affiliation(s)
- Iljin Kim
- Department of Pharmacology, Cancer Research Institute, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong-Wan Park
- Department of Pharmacology, Cancer Research Institute, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Huang D, Hou X, Zhang D, Zhang Q, Yan C. Two novel polysaccharides from rhizomes of Cibotium barometz promote bone formation via activating the BMP2/SMAD1 signaling pathway in MC3T3-E1 cells. Carbohydr Polym 2020; 231:115732. [PMID: 31888819 DOI: 10.1016/j.carbpol.2019.115732] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/19/2019] [Accepted: 12/09/2019] [Indexed: 01/06/2023]
Abstract
Cibotium barometz, an important traditional Chinese medicine, is used in strengthening bones and tendons. We found that C. barometz crude polysaccharides (CB70) could alleviate bone loss and markedly improve the biomechanical properties of OVX rats. Thus, to clarify biological active ingredient(s) of CB70, two homogeneous polysaccharides (CBP70-1-1 and CBP70-1-2) were purified from CB70. A combination of monosaccharide composition, FT-IR, GC-MS and NMR analysis indicated that CBP70-1-1 was composed of →6)-D-Galp-(1→, D-Glcp-(1→, →3,6)-D-Manp-(1→, →4)-D-Glcp-(1→ and →6)-D-Glcp-(1→ with relative molecular weights of 12,724 Da, and CBP70-1-2 was composed of →4)-D-Glcp-(1→, D-Glcp-(1→, →3,6)-D-Manp-(1→, →6)-D-Galp-(1→, →4,6)-D-Glcp-(1→ and →3)-L-Araf-(1→ with relative molecular weights of 3611 Da. Morphological analyses revealed that CBP70-1-1 and CBP70-1-2 appeared as a sheet that were irregular in size and shape, while the surface of CBP70-1-1 was full of sharp protuberances and CBP70-1-2 was smooth. Furthermore, the effects of CBP70-1-1 and CBP70-1-2 on the proliferation, differentiation and mineralization of mouse pre-osteoblastic MC3T3-E1 cells were assessed via CCK-8 assay, alkaline phosphatase activity assay, and alizarin red-based assay, respectively. These results revealed that CBP70-1-1 and CBP70-1-2 significantly promoted the proliferation, differentiation and mineralization of MC3T3-E1 cells, even better than E2. More importantly, quantitative real-time PCR and Western blot analysis indicated that CBP70-1-2 pronouncedly promoted the expression of osteogenic-related marker genes (Runx2, Osx, Ocn and Opn) and proteins (BMP2, RUNX2, OSX and p-SMAD1), which implies that the osteogenic activity of CBP70-1-2 is accomplished mainly by activating the BMP2/SMAD1 signaling pathway. These findings suggest CBP70-1-2 as a potential natural anti-osteoporotic agent for pharmacotherapy.
Collapse
Affiliation(s)
- Dong Huang
- Center for Clinical Precision Medication, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510006, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xin Hou
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Dawei Zhang
- Department of Osteoporosis, Shajing People's Hospital of Bao'an Shenzhen, Shenzhen, 518104, China
| | - Qian Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Chunyan Yan
- Center for Clinical Precision Medication, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510006, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
17
|
Adipose-Derived Stem Cells and Ceiling Culture-Derived Preadipocytes Cultured from Subcutaneous Fat Tissue Differ in Their Epigenetic Characteristics and Osteogenic Potential. Plast Reconstr Surg 2020; 144:644-655. [PMID: 31461020 DOI: 10.1097/prs.0000000000005913] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Adipose-derived stem cells and ceiling culture-derived preadipocytes can be harvested from subcutaneous adipose tissue. Little is known about the epigenetic differences, which may contribute to differences in osteogenic potential, between these cell types. The purpose of this study was to address the osteogenic potential and underlying epigenetic status of adipose-derived stem cells and ceiling culture-derived preadipocytes. METHODS Adipose-derived stem cells and ceiling culture-derived preadipocytes were cultured from abdominal subcutaneous fat tissues of four metabolically healthy, lean female patients. After 7 weeks of culture, cellular responses to osteogenic differentiation media were examined. To evaluate the osteogenic potentials of undifferentiated adipose-derived stem cells and ceiling culture-derived preadipocytes, two types of epigenetic assessment were performed using next-generation sequencing: DNA methylation assays with the Human Methylation 450K BeadChip, and chromatin immunoprecipitation assays for trimethylation of histone H3 at lysine 4. RESULTS Human ceiling culture-derived preadipocytes showed greater osteogenic differentiation ability than did adipose-derived stem cells. In an epigenetic survey of the promoters of four osteogenic regulator genes (RUNX2, SP7, ATF4, and BGLAP), the authors found a general trend toward decreased CpG methylation and increased trimethylation of histone H3 at lysine 4 levels in ceiling culture-derived preadipocytes as compared to adipose-derived stem cells, indicating that these genes were more likely to be highly expressed in ceiling culture-derived preadipocytes. CONCLUSIONS The surveyed epigenetic differences between adipose-derived stem cells and ceiling culture-derived preadipocytes were consistent with the observed differences in osteogenic potential. These results enhance the authors' understanding of these cells and will facilitate their further application in regenerative medicine.
Collapse
|
18
|
Bone Remodeling: Histone Modifications as Fate Determinants of Bone Cell Differentiation. Int J Mol Sci 2019; 20:ijms20133147. [PMID: 31252653 PMCID: PMC6651527 DOI: 10.3390/ijms20133147] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023] Open
Abstract
The bone tissue is a dynamic complex that constitutes of several interdependent systems and is continuously remodeled through the concerted actions of bone cells. Osteoblasts are mononucleated cells, derived from mesenchymal stem cells, responsible for bone formation. Osteoclasts are large multinucleated cells that differentiate from hematopoietic progenitors of the myeloid lineage and are responsible for bone resorption. The lineage-specific differentiation of bone cells requires an epigenetic regulation of gene expressions involving chromatin dynamics. The key step for understanding gene regulatory networks during bone cell development lies in characterizing the chromatin modifying enzymes responsible for reorganizing and potentiating particular chromatin structure. This review covers the histone-modifying enzymes involved in bone development, discusses the impact of enzymes on gene expression, and provides future directions and clinical significance in this area.
Collapse
|
19
|
Oncogenic and osteolytic functions of histone demethylase NO66 in castration-resistant prostate cancer. Oncogene 2019; 38:5038-5049. [DOI: 10.1038/s41388-019-0774-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/07/2018] [Accepted: 02/16/2019] [Indexed: 02/07/2023]
|
20
|
Bundred JR, Hendrix E, Coleman ML. The emerging roles of ribosomal histidyl hydroxylases in cell biology, physiology and disease. Cell Mol Life Sci 2018; 75:4093-4105. [PMID: 30151692 PMCID: PMC6182338 DOI: 10.1007/s00018-018-2903-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 12/27/2022]
Abstract
Hydroxylation is a novel protein modification catalyzed by a family of oxygenases that depend on fundamental nutrients and metabolites for activity. Protein hydroxylases have been implicated in a variety of key cellular processes that play important roles in both normal homeostasis and pathogenesis. Here, in this review, we summarize the current literature on a highly conserved sub-family of oxygenases that catalyze protein histidyl hydroxylation. We discuss the evidence supporting the biochemical assignment of these emerging enzymes as ribosomal protein hydroxylases, and provide an overview of their role in immunology, bone development, and cancer.
Collapse
Affiliation(s)
- James R Bundred
- Tumour Oxygenase Group, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Eline Hendrix
- Tumour Oxygenase Group, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Mathew L Coleman
- Tumour Oxygenase Group, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
21
|
Farr JN, Almeida M. The Spectrum of Fundamental Basic Science Discoveries Contributing to Organismal Aging. J Bone Miner Res 2018; 33:1568-1584. [PMID: 30075061 PMCID: PMC6327947 DOI: 10.1002/jbmr.3564] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/22/2022]
Abstract
Aging research has undergone unprecedented advances at an accelerating rate in recent years, leading to excitement in the field as well as opportunities for imagination and innovation. Novel insights indicate that, rather than resulting from a preprogrammed series of events, the aging process is predominantly driven by fundamental non-adaptive mechanisms that are interconnected, linked, and overlap. To varying degrees, these mechanisms also manifest with aging in bone where they cause skeletal fragility. Because these mechanisms of aging can be manipulated, it might be possible to slow, delay, or alleviate multiple age-related diseases and their complications by targeting conserved genetic signaling pathways, controlled functional networks, and basic biochemical processes. Indeed, findings in various mammalian species suggest that targeting fundamental aging mechanisms (eg, via either loss-of-function or gain-of-function mutations or administration of pharmacological therapies) can extend healthspan; ie, the healthy period of life free of chronic diseases. In this review, we summarize the evidence supporting the role of the spectrum of fundamental basic science discoveries contributing to organismal aging, with emphasis on mammalian studies and in particular aging mechanisms in bone that drive skeletal fragility. These mechanisms or aging hallmarks include: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. Because these mechanisms are linked, interventions that ameliorate one hallmark can in theory ameliorate others. In the field of bone and mineral research, current challenges include defining the relative contributions of each aging hallmark to the natural skeletal aging process, better understanding the complex interconnections among the hallmarks, and identifying the most effective therapeutic strategies to safely target multiple hallmarks. Based on their interconnections, it may be feasible to simultaneously interfere with several fundamental aging mechanisms to alleviate a wide spectrum of age-related chronic diseases, including osteoporosis. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Joshua N Farr
- Division of Endocrinology and Metabolism and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
22
|
Wang FS, Lian WS, Lee MS, Weng WT, Huang YH, Chen YS, Sun YC, Wu SL, Chuang PC, Ko JY. Histone demethylase UTX counteracts glucocorticoid deregulation of osteogenesis by modulating histone-dependent and -independent pathways. J Mol Med (Berl) 2017; 95:499-512. [PMID: 28130569 DOI: 10.1007/s00109-017-1512-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 12/27/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022]
Abstract
Excess glucocorticoid administration impairs osteogenic activities, which raises the risk of osteoporotic disorders. Epigenetic methylation of DNA and histone regulates the lineage commitment of progenitor cells. This study was undertaken to delineate the actions of histone lysine demethylase 6a (UTX) with regard to the glucocorticoid impediment of osteogenic differentiation. Osteogenic progenitor cells responded to supraphysiological glucocorticoid by elevating CpG dinucleotide methylation proximal to transcription start sites within Runx2 and osterix promoters and Wnt inhibitor Dickkopf-1 (Dkk1) expression concomitant with low UTX expression. 5'-Aza-deoxycystidine demethylation of Runx2 and osterix promoters abolished the glucocorticoid inhibition of mineralized matrix accumulation. Gain of UTX function attenuated the glucocorticoid-induced loss of osteogenic differentiation, whereas UTX silencing escalated adipogenic gene expression and adipocyte formation. UTX sustained osteogenic gene transcription through maintaining its occupancy to Runx2 and osterix promoters. It also mitigated the trimethylation of histone 3 at lysine 27 (H3K27me3), which reduced H3K27me3 enrichment to Dkk1 promoter and thereby lowered Dkk1 transcription. Modulation of β-catenin and Dkk1 actions restored UTX signaling in glucocorticoid-stressed cells. In vivo, UTX inhibition by exogenous methylprednisolone and GSK-J4 administration, an effect that disturbed H3K27me3, β-catenin, Dkk1, Runx2, and osterix levels, exacerbated trabecular microarchitecture loss and marrow adiposity. Taken together, glucocorticoid reduction of UTX function hindered osteogenic differentiation. Epigenetic hypomethylation of osteogenic transcription factor promoters and H3K27 contributed to the UXT alleviation of Dkk1 transcription and osteogenesis in glucocorticoid-stressed osteogenic progenitor cells. Control of UTX action has an epigenetic perspective of curtailing glucocorticoid impairment of osteogenic differentiation and bone mass. KEY MESSAGES UTX attenuates glucocorticoid deregulation of osteogenesis and adipogenesis. UTX reduces Runx2 promoter methylation and H3K27me3 enrichment in the Dkk1 promoter. β-catenin and Dkk1 modulate the glucocorticoid inhibition of UTX signaling. UTX inhibition exacerbates bone mass, trabecular microstructure and fatty marrow. UTX signaling is indispensable in fending off glucocorticoid-impaired osteogenesis.
Collapse
Affiliation(s)
- Feng-Sheng Wang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Wei-Shiung Lian
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Mel S Lee
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Wen-Tsan Weng
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Ying-Hsien Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Yu-Shan Chen
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Yi-Chih Sun
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Shing-Long Wu
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Pei-Chin Chuang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.
| | - Jih-Yang Ko
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan. .,Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.
| |
Collapse
|
23
|
Wu H, Gordon JAR, Whitfield TW, Tai PWL, van Wijnen AJ, Stein JL, Stein GS, Lian JB. Chromatin dynamics regulate mesenchymal stem cell lineage specification and differentiation to osteogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:438-449. [PMID: 28077316 DOI: 10.1016/j.bbagrm.2017.01.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 12/10/2016] [Accepted: 01/06/2017] [Indexed: 01/08/2023]
Abstract
Multipotent mesenchymal stromal cells (MSCs) are critical for regeneration of multiple tissues. Epigenetic mechanisms are fundamental regulators of lineage specification and cell fate, and as such, we addressed the question of which epigenetic modifications characterize the transition of nascent MSCs to a tissue specific MSC-derived phenotype. By profiling the temporal changes of seven histone marks correlated to gene expression during proliferation, early commitment, matrix deposition, and mineralization stages, we identified distinct epigenetic mechanisms that regulate transcriptional programs necessary for tissue-specific phenotype development. Patterns of stage-specific enrichment of histone modifications revealed distinct modes of repression and activation of gene expression that would not be detected using single endpoint analysis. We discovered that at commitment, H3K27me3 is removed from genes that are upregulated and is not acquired on downregulated genes. Additionally, we found that the absence of H3K4me3 modification at promoters defined a subset of osteoblast-specific upregulated genes, indicating that acquisition of acetyl modifications drive activation of these genes. Significantly, loss or gain of H3K36me3 was the primary predictor of dynamic changes in temporal gene expression. Using unsupervised pattern discovery analysis the signature of osteogenic-related histone modifications identified novel functional cis regulatory modules associated with enhancer regions that control tissue-specific genes. Our work provides a cornerstone to understand the epigenetic regulation of transcriptional programs that are important for MSC lineage commitment and lineage, as well as insights to facilitate MSC-based therapeutic interventions.
Collapse
Affiliation(s)
- Hai Wu
- Department of Biochemistry and Vermont Cancer Center, University of Vermont, Burlington, VT, United States.
| | - Jonathan A R Gordon
- Department of Biochemistry and Vermont Cancer Center, University of Vermont, Burlington, VT, United States.
| | - Troy W Whitfield
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, United States.
| | - Phillip W L Tai
- Department of Biochemistry and Vermont Cancer Center, University of Vermont, Burlington, VT, United States.
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Janet L Stein
- Department of Biochemistry and Vermont Cancer Center, University of Vermont, Burlington, VT, United States.
| | - Gary S Stein
- Department of Biochemistry and Vermont Cancer Center, University of Vermont, Burlington, VT, United States.
| | - Jane B Lian
- Department of Biochemistry and Vermont Cancer Center, University of Vermont, Burlington, VT, United States.
| |
Collapse
|
24
|
Jian CX, Fan QS, Hu YH, He Y, Li MZ, Zheng WY, Ren Y, Li CJ. IL-7 suppresses osteogenic differentiation of periodontal ligament stem cells through inactivation of mitogen-activated protein kinase pathway. Organogenesis 2016; 12:183-193. [PMID: 27579861 DOI: 10.1080/15476278.2016.1229726] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Periodontal ligament stem cells (PDLSCs) are tissue-specific mesenchymal stem cells (MSCs), having an important role in regenerative therapy for teeth loss. Interleukin-7 (IL-7) is a key cytokine produced by stromal cells including MSCs, and exhibits specific roles for B and T cell development and osteoblasts differentiation of multiple myeloma. However, the effect of IL-7 on osteogenic differentiation of PDLSCs remains unclear. Therefore, in the present study we determined whether IL-7 affects the proliferation and osteogenic differentiation of PDLSCs in vitro and explored the associated signaling pathways for IL-7-mediated cell differentiation. The results demonstrated that the isolated human PDLSCs possessed MSCs features, highly expressing CD90, CD44, CD105, CD29 and CD73, and almost did not expressed CD34, CD45, CD11b, CD14 and CD117. IL-7 could not significantly affect the proliferation of PDLSCs, but it decreased their osteogenic differentiation and inhibited alkaline phosphatase (ALP) activity. The results of quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blotting exhibited that the expression levels of Runx-2, SP7 and osteocalcin (OCN) were significantly reduced by IL-7. Further studies indicated that IL-7 did not significantly change JNK, ERK1/2 and p38 protein production, but markedly suppressed their phosphorylation levels. These data suggest that IL-7 inhibits the osteogenic differentiation of PDLSCs probably via inactivation of mitogen-activated protein kinase (MAPK) signaling pathway.
Collapse
Affiliation(s)
- Cong-Xiang Jian
- a Department of Stomatolog , PLA General Hospital of Chengdu Military Region, Tianhui Town , Jinniu District, Chengdu , Sichuan Province , China
| | - Quan-Shui Fan
- b Chengdu Military Garrison Center for Disease Control and Prevention , Chengdu , Sichuan , China
| | - Yong-He Hu
- b Chengdu Military Garrison Center for Disease Control and Prevention , Chengdu , Sichuan , China
| | - Yong He
- a Department of Stomatolog , PLA General Hospital of Chengdu Military Region, Tianhui Town , Jinniu District, Chengdu , Sichuan Province , China
| | - Ming-Zhe Li
- a Department of Stomatolog , PLA General Hospital of Chengdu Military Region, Tianhui Town , Jinniu District, Chengdu , Sichuan Province , China
| | - Wei-Yin Zheng
- a Department of Stomatolog , PLA General Hospital of Chengdu Military Region, Tianhui Town , Jinniu District, Chengdu , Sichuan Province , China
| | - Yu Ren
- a Department of Stomatolog , PLA General Hospital of Chengdu Military Region, Tianhui Town , Jinniu District, Chengdu , Sichuan Province , China
| | - Chen-Jun Li
- a Department of Stomatolog , PLA General Hospital of Chengdu Military Region, Tianhui Town , Jinniu District, Chengdu , Sichuan Province , China
| |
Collapse
|
25
|
Wang C, Liao H, Cao Z. Role of Osterix and MicroRNAs in Bone Formation and Tooth Development. Med Sci Monit 2016; 22:2934-42. [PMID: 27543160 PMCID: PMC4994932 DOI: 10.12659/msm.896742] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Osterix (Osx) is an osteoblast-specific transcription factor that is essential for bone formation. MicroRNAs (miRNAs) are ~22-nucleotide-long noncoding RNAs that play important regulatory roles in animals and plants by targeting mRNAs for cleavage or translational repression. They can also control osteoblast-mediated bone formation and osteoclast-related bone remodeling. The vital roles of Osx and miRNAs during bone formation have been well studied, but very few studies have discussed their co-functions and the relationships between them. In this review, we outline the significant functions of Osx and miRNAs on certain cell types during osteogenesis and illustrate their roles during tooth development. More importantly, we discuss the relationship between Osx and miRNAs, which we believe could lead to a new treatment for skeletal and periodontal diseases.
Collapse
Affiliation(s)
- Chuan Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) and Key Laboratory for Oral Biomedical Engineering of Ministry of Education (KLOBME), School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China (mainland)
| | - Haiqing Liao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) and Key Laboratory for Oral Biomedical Engineering of Ministry of Education (KLOBME), School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China (mainland)
| | - Zhengguo Cao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) and Key Laboratory for Oral Biomedical Engineering of Ministry of Education (KLOBME), School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
26
|
Boeckel JN, Derlet A, Glaser SF, Luczak A, Lucas T, Heumüller AW, Krüger M, Zehendner CM, Kaluza D, Doddaballapur A, Ohtani K, Treguer K, Dimmeler S. JMJD8 Regulates Angiogenic Sprouting and Cellular Metabolism by Interacting With Pyruvate Kinase M2 in Endothelial Cells. Arterioscler Thromb Vasc Biol 2016; 36:1425-33. [PMID: 27199445 DOI: 10.1161/atvbaha.116.307695] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 04/25/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Jumonji C (JmjC) domain-containing proteins modify histone and nonhistone proteins thereby controlling cellular functions. However, the role of JmjC proteins in angiogenesis is largely unknown. Here, we characterize the expression of JmjC domain-containing proteins after inducing endothelial differentiation of murine embryonic stem cells and study the function of JmjC domain-only proteins in endothelial cell (EC) functions. APPROACH AND RESULTS We identified a large number of JmjC domain-containing proteins regulated by endothelial differentiation of murine embryonic stem cells. Among the family of JmjC domain-only proteins, Jmjd8 was significantly upregulated on endothelial differentiation. Knockdown of Jmjd8 in ECs significantly decreased in vitro network formation and sprouting in the spheroid assay. JMJD8 is exclusively detectable in the cytoplasm, excluding a function as a histone-modifying enzyme. Mass spectrometry analysis revealed JMJD8-interacting proteins with known functions in cellular metabolism like pyruvate kinase M2. Accordingly, knockdown of pyruvate kinase M2 in human umbilical vein ECs decreased endothelial sprouting in the spheroid assay. Knockdown of JMJD8 caused a reduction of EC metabolism as measured by Seahorse Bioscience extracellular flux analysis. Conversely, overexpression of JMJD8 enhanced cellular oxygen consumption rate of ECs, reflecting an increased mitochondrial respiration. CONCLUSIONS Jmjd8 is upregulated during endothelial differentiation and regulates endothelial sprouting and metabolism by interacting with pyruvate kinase M2.
Collapse
Affiliation(s)
- Jes-Niels Boeckel
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Anja Derlet
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Simone F Glaser
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Annika Luczak
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Tina Lucas
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Andreas W Heumüller
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Marcus Krüger
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Christoph M Zehendner
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - David Kaluza
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Anuradha Doddaballapur
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Kisho Ohtani
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Karine Treguer
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.)
| | - Stefanie Dimmeler
- From the Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (J.-N.B., A.D., S.F.G., A.L., T.L., A.W.H., C.M.Z., D.K., A.D., K.O., K.T., S.D.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.K.); Department of Cardiology, Internal Medicine III, Goethe-University Hospital, Frankfurt, Germany (C.M.Z.); and German Center of Cardiovascular Research (DZHK), RheinMain, Frankfurt, Germany (J.-N.B., A.D., S.F.G., T.L., C.M.Z., A.D., S.D.).
| |
Collapse
|
27
|
Deng P, Chen QM, Hong C, Wang CY. Histone methyltransferases and demethylases: regulators in balancing osteogenic and adipogenic differentiation of mesenchymal stem cells. Int J Oral Sci 2015; 7:197-204. [PMID: 26674421 PMCID: PMC5153596 DOI: 10.1038/ijos.2015.41] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2015] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are characterized by their self-renewing capacity and differentiation potential into multiple tissues. Thus, management of the differentiation capacities of MSCs is important for MSC-based regenerative medicine, such as craniofacial bone regeneration, and in new treatments for metabolic bone diseases, such as osteoporosis. In recent years, histone modification has been a growing topic in the field of MSC lineage specification, in which the Su(var)3–9, enhancer-of-zeste, trithorax (SET) domain-containing family and the Jumonji C (JmjC) domain-containing family represent the major histone lysine methyltransferases (KMTs) and histone lysine demethylases (KDMs), respectively. In this review, we summarize the current understanding of the epigenetic mechanisms by which SET domain-containing KMTs and JmjC domain-containing KDMs balance the osteogenic and adipogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Peng Deng
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA.,State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Qian-Ming Chen
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Christine Hong
- Section of Orthodontics, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Cun-Yu Wang
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California at Los Angeles, Los Angeles, USA
| |
Collapse
|
28
|
Gordon JAR, Stein JL, Westendorf JJ, van Wijnen AJ. Chromatin modifiers and histone modifications in bone formation, regeneration, and therapeutic intervention for bone-related disease. Bone 2015; 81:739-745. [PMID: 25836763 PMCID: PMC4591092 DOI: 10.1016/j.bone.2015.03.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/13/2015] [Indexed: 02/07/2023]
Abstract
Post-translational modifications of chromatin such as DNA methylation and different types of histone acetylation, methylation and phosphorylation are well-appreciated epigenetic mechanisms that confer information to progeny cells during lineage commitment. These distinct epigenetic modifications have defined roles in bone, development, tissue regeneration, cell commitment and differentiation, as well as disease etiologies. In this review, we discuss the role of these chromatin modifications and the enzymes regulating these marks (methyltransferases, demethylases, acetyltransferases, and deacetylases) in progenitor cells, osteoblasts and bone-related cells. In addition, the clinical relevance of deregulated histone modifications and enzymes as well as current and potential therapeutic interventions targeting chromatin modifiers are addressed.
Collapse
Affiliation(s)
| | - Janet L Stein
- Department of Biochemistry, University of Vermont, Burlington, VT, USA.
| | | | | |
Collapse
|
29
|
Wang C, Zhang Q, Hang T, Tao Y, Ma X, Wu M, Zhang X, Zang J. Structure of the JmjC domain-containing protein NO66 complexed with ribosomal protein Rpl8. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2015; 71:1955-64. [PMID: 26327385 PMCID: PMC4556315 DOI: 10.1107/s1399004715012948] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/05/2015] [Indexed: 11/10/2022]
Abstract
The JmjC domain-containing proteins belong to a large family of oxygenases possessing distinct substrate specificities which are involved in the regulation of different biological processes, such as gene transcription, RNA processing and translation. Nucleolar protein 66 (NO66) is a JmjC domain-containing protein which has been reported to be a histone demethylase and a ribosome protein 8 (Rpl8) hydroxylase. The present biochemical study confirmed the hydroxylase activity of NO66 and showed that oligomerization is required for NO66 to efficiently catalyze the hydroxylation of Rpl8. The structures of NO66(176-C) complexed with Rpl8(204-224) in a tetrameric form and of the mutant protein M2 in a dimeric form were solved. Based on the results of structural and biochemical analyses, the consensus sequence motif NHXH recognized by NO66 was confirmed. Several potential substrates of NO66 were found by a BLAST search according to the consensus sequence motif. When binding to substrate, the relative positions of each subunit in the NO66 tetramer shift. Oligomerization may facilitate the motion of each subunit in the NO66 tetramer and affect the catalytic activity.
Collapse
Affiliation(s)
- Chengliang Wang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, Collaborative Innovation Center of Chemistry for Life Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, People’s Republic of China
- Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, People’s Republic of China
| | - Qiongdi Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, Collaborative Innovation Center of Chemistry for Life Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, People’s Republic of China
| | - Tianrong Hang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, Collaborative Innovation Center of Chemistry for Life Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, People’s Republic of China
- Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, People’s Republic of China
| | - Yue Tao
- Institute for Pediatric Translational Medicine, Shanghai Children’s Medical Center, 1678 Dongfang Road, Pudong, Shanghai 200120, People’s Republic of China
| | - Xukai Ma
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, Collaborative Innovation Center of Chemistry for Life Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, People’s Republic of China
| | - Minhao Wu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, Collaborative Innovation Center of Chemistry for Life Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, People’s Republic of China
- Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, People’s Republic of China
| | - Xuan Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, Collaborative Innovation Center of Chemistry for Life Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, People’s Republic of China
- Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, People’s Republic of China
| | - Jianye Zang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, Collaborative Innovation Center of Chemistry for Life Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, People’s Republic of China
- Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, People’s Republic of China
| |
Collapse
|
30
|
Montesi M, Panseri S, Iafisco M, Adamiano A, Tampieri A. Coupling Hydroxyapatite Nanocrystals with Lactoferrin as a Promising Strategy to Fine Regulate Bone Homeostasis. PLoS One 2015; 10:e0132633. [PMID: 26148296 PMCID: PMC4492779 DOI: 10.1371/journal.pone.0132633] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/16/2015] [Indexed: 12/13/2022] Open
Abstract
Lactoferrin (LF) is an interesting glycoprotein in the field of bone biology for its regulatory effect on cells involved in bone remodeling, that results compromised in several pathological conditions, as osteoporosis. In a previous study we observed that the coupling of LF and biomimetic hydroxyapatite nanocrystals (HA), a material well-known for its bioactivity and osteoconductive properties, leads to a combined effect in the induction of osteogenic differentiation of mesenchymal stem cells. On the basis of this evidence, the present study is an extension of our previous work aiming to investigate the synergistic effect of the coupling of HA and LF on bone homeostasis. Biomimetic HA nanocrystals were synthesized and functionalized with LF (HA-LF) and then pre-osteoblasts (MC3T3-E1) and monocyte/macrophage cells lines (RAW 264.7), using as osteoclastogenesis in vitro model, were cultured separately or in co-culture in presence of HA-LF. The results clearly revealed that HA and LF act in synergism in the regulation of the bone homeostasis, working as anabolic factor for osteoblasts differentiation and bone matrix deposition, and as inhibitor of the osteoclast formation and activity.
Collapse
Affiliation(s)
- Monica Montesi
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
- * E-mail:
| | - Silvia Panseri
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| | - Michele Iafisco
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| | - Alessio Adamiano
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| | - Anna Tampieri
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| |
Collapse
|
31
|
Lee SH, Jeong HM, Han Y, Cheong H, Kang BY, Lee KY. Prolyl isomerase Pin1 regulates the osteogenic activity of Osterix. Mol Cell Endocrinol 2015; 400:32-40. [PMID: 25463757 DOI: 10.1016/j.mce.2014.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/27/2014] [Accepted: 11/20/2014] [Indexed: 02/07/2023]
Abstract
Osterix is an essential transcription factor for osteoblast differentiation and bone formation. The mechanism of regulation of Osterix by post-translational modification remains unknown. Peptidyl-prolyl isomerase 1 (Pin1) catalyzes the isomerization of pSer/Thr-Pro bonds and induces a conformational change in its substrates, subsequently regulating diverse cellular processes. In this study, we demonstrated that Pin1 interacts with Osterix and influences its protein stability and transcriptional activity. This regulation is likely due to the suppression of poly-ubiquitination-mediated proteasomal degradation of Osterix. Collectively, our data demonstrate that Pin1 is a novel regulator of Osterix and may play an essential role in the regulation of osteogenic differentiation.
Collapse
Affiliation(s)
- Sung Ho Lee
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Hyung Min Jeong
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Younho Han
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Heesun Cheong
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 410-769, Republic of Korea
| | - Bok Yun Kang
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Kwang Youl Lee
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea.
| |
Collapse
|
32
|
Pires-Luís AS, Vieira-Coimbra M, Vieira FQ, Costa-Pinheiro P, Silva-Santos R, Dias PC, Antunes L, Lobo F, Oliveira J, Gonçalves CS, Costa BM, Henrique R, Jerónimo C. Expression of histone methyltransferases as novel biomarkers for renal cell tumor diagnosis and prognostication. Epigenetics 2015; 10:1033-43. [PMID: 26488939 PMCID: PMC4844211 DOI: 10.1080/15592294.2015.1103578] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/21/2015] [Accepted: 09/24/2015] [Indexed: 12/13/2022] Open
Abstract
Renal cell tumors (RCTs) are the most lethal of the common urological cancers. The widespread use of imaging entailed an increased detection of small renal masses, emphasizing the need for accurate distinction between benign and malignant RCTs, which is critical for adequate therapeutic management. Histone methylation has been implicated in renal tumorigenesis, but its potential clinical value as RCT biomarker remains mostly unexplored. Hence, the main goal of this study was to identify differentially expressed histone methyltransferases (HMTs) and histone demethylases (HDMs) that might prove useful for RCT diagnosis and prognostication, emphasizing the discrimination between oncocytoma (a benign tumor) and renal cell carcinoma (RCC), especially the chromophobe subtype (chRCC). We found that the expression levels of 3 genes--SMYD2, SETD3, and NO66--was significantly altered in a set of RCTs, which was further validated in a large independent cohort. Higher expression levels were found in RCTs compared to normal renal tissues (RNTs) and in chRCCs comparatively to oncocytomas. SMYD2 and SETD3 mRNA levels correlated with protein expression assessed by immunohistochemistry. SMYD2 transcript levels discriminated RCTs from RNT, with 82.1% sensitivity and 100% specificity [area under curve (AUC) = 0.959], and distinguished chRCCs from oncocytomas, with 71.0% sensitivity and 73.3% specificity (AUC = 0.784). Low expression levels of SMYD2, SETD3, and NO66 were significantly associated with shorter disease-specific and disease-free survival, especially in patients with non-organ confined tumors. We conclude that expression of selected HMTs and HDMs might constitute novel biomarkers to assist in RCT diagnosis and assessment of tumor aggressiveness.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/diagnosis
- Carcinoma, Renal Cell/enzymology
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- Diagnosis, Differential
- Dioxygenases
- Early Detection of Cancer
- Gene Expression Regulation, Neoplastic
- Histone Demethylases/genetics
- Histone Demethylases/metabolism
- Histone Methyltransferases
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Humans
- Kidney Neoplasms/diagnosis
- Kidney Neoplasms/enzymology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Prognosis
- Survival Analysis
- Up-Regulation
Collapse
Affiliation(s)
- Ana Sílvia Pires-Luís
- Cancer Biology and Epigenetics Group – Research Center; Portuguese Oncology Institute – Porto; Porto, Portugal
- Department of Pathology; Portuguese Oncology Institute – Porto; Porto, Portugal
| | - Márcia Vieira-Coimbra
- Cancer Biology and Epigenetics Group – Research Center; Portuguese Oncology Institute – Porto; Porto, Portugal
- Department of Pathology; Portuguese Oncology Institute – Porto; Porto, Portugal
| | - Filipa Quintela Vieira
- Cancer Biology and Epigenetics Group – Research Center; Portuguese Oncology Institute – Porto; Porto, Portugal
- School of Allied Health Sciences (ESTSP); Polytechnic of Porto; Porto, Portugal
| | - Pedro Costa-Pinheiro
- Cancer Biology and Epigenetics Group – Research Center; Portuguese Oncology Institute – Porto; Porto, Portugal
| | - Rui Silva-Santos
- Cancer Biology and Epigenetics Group – Research Center; Portuguese Oncology Institute – Porto; Porto, Portugal
| | - Paula C Dias
- Department of Pathology; Portuguese Oncology Institute – Porto; Porto, Portugal
| | - Luís Antunes
- Department of Epidemiology; Portuguese Oncology Institute – Porto; Porto, Portugal
| | - Francisco Lobo
- Department of Urology; Portuguese Oncology Institute – Porto; Porto, Portugal
| | - Jorge Oliveira
- Department of Urology; Portuguese Oncology Institute – Porto; Porto, Portugal
| | - Céline S Gonçalves
- Life and Health Sciences Research Institute (ICVS); School of Health Sciences; University of Minho; Braga, Portugal
- ICVS/3B's – PT Government Associate Laboratory; University of Minho; Braga/Guimarães; Portugal
| | - Bruno M Costa
- Life and Health Sciences Research Institute (ICVS); School of Health Sciences; University of Minho; Braga, Portugal
- ICVS/3B's – PT Government Associate Laboratory; University of Minho; Braga/Guimarães; Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group – Research Center; Portuguese Oncology Institute – Porto; Porto, Portugal
- Department of Pathology; Portuguese Oncology Institute – Porto; Porto, Portugal
- Department of Pathology and Molecular Immunology; Institute of Biomedical Sciences Abel Salazar (ICBAS) – University of Porto; Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group – Research Center; Portuguese Oncology Institute – Porto; Porto, Portugal
- Department of Pathology and Molecular Immunology; Institute of Biomedical Sciences Abel Salazar (ICBAS) – University of Porto; Porto, Portugal
| |
Collapse
|
33
|
Artigas N, Ureña C, Rodríguez-Carballo E, Rosa JL, Ventura F. Mitogen-activated protein kinase (MAPK)-regulated interactions between Osterix and Runx2 are critical for the transcriptional osteogenic program. J Biol Chem 2014; 289:27105-27117. [PMID: 25122769 PMCID: PMC4175347 DOI: 10.1074/jbc.m114.576793] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 08/06/2014] [Indexed: 11/06/2022] Open
Abstract
The transcription factors Runx2 and Osx (Osterix) are required for osteoblast differentiation and bone formation. Runx2 expression occurs at early stages of osteochondroprogenitor determination, followed by Osx induction during osteoblast maturation. We demonstrate that coexpression of Osx and Runx2 leads to cooperative induction of expression of the osteogenic genes Col1a1, Fmod, and Ibsp. Functional interaction of Osx and Runx2 in the regulation of these promoters is mediated by enhancer regions with adjacent Sp1 and Runx2 DNA-binding sites. These enhancers allow formation of a cooperative transcriptional complex, mediated by the binding of Osx and Runx2 to their specific DNA promoter sequences and by the protein-protein interactions between them. We also identified the domains involved in the interaction between Osx and Runx2. These regions contain the amino acids in Osx and Runx2 known to be phosphorylated by p38 and ERK MAPKs. Inhibition of p38 and ERK kinase activities or mutation of their known phosphorylation sites in Osx or Runx2 strongly disrupts their physical interaction and cooperative transcriptional effects. Altogether, our results provide a molecular description of a mechanism for Osx and Runx2 transcriptional cooperation that is subject to further regulation by MAPK-activating signals during osteogenesis.
Collapse
Affiliation(s)
- Natalia Artigas
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, L'Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), E-08907 L'Hospitalet de Llobregat, Spain
| | - Carlos Ureña
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, L'Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), E-08907 L'Hospitalet de Llobregat, Spain
| | - Edgardo Rodríguez-Carballo
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, L'Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), E-08907 L'Hospitalet de Llobregat, Spain
| | - José Luis Rosa
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, L'Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), E-08907 L'Hospitalet de Llobregat, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, L'Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), E-08907 L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
34
|
Tassano E, Accogli A, Panigada S, Ronchetto P, Cuoco C, Gimelli G. Phenotypic and genetic characterization of a patient with a de novo interstitial 14q24.1q24.3 deletion. Mol Cytogenet 2014; 7:49. [PMID: 25076984 PMCID: PMC4115490 DOI: 10.1186/1755-8166-7-49] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 06/30/2014] [Indexed: 11/23/2022] Open
Abstract
Background Interstitial deletions of chromosome bands 14q24.1q24.3 are very rare with only three reported cases. Results We describe a 7-year-old boy with a 5.345 Mb de novo interstitial deletion at 14q24.1q24.3 band detected by array-CGH who had a complex phenotype characterized by seizures, congenital heart defects, dysmorphisms, psychomotor delay, and bronchopulmonary, skeletal, and brain anomalies. Conclusion The deleted region contains numerous genes, but we focused our attention on three of them (C14orf169, NUMB, and PSEN1), which could account, at least partially, for the phenotype of the boy. We therefore discuss the involvement of these genes and the observed phenotype compared to that of previously described patients.
Collapse
Affiliation(s)
- Elisa Tassano
- Laboratorio di Citogenetica, Istituto G.Gaslini, L.go G.Gaslini 5, Genoa 16147, Italy
| | - Andrea Accogli
- Pediatric Pulmonology and Allergy Unit, Istituto Giannina Gaslini, Genoa, Italy
| | - Serena Panigada
- Pediatric Pulmonology and Allergy Unit, Istituto Giannina Gaslini, Genoa, Italy
| | - Patrizia Ronchetto
- Laboratorio di Citogenetica, Istituto G.Gaslini, L.go G.Gaslini 5, Genoa 16147, Italy
| | - Cristina Cuoco
- Laboratorio di Citogenetica, Istituto G.Gaslini, L.go G.Gaslini 5, Genoa 16147, Italy
| | - Giorgio Gimelli
- Laboratorio di Citogenetica, Istituto G.Gaslini, L.go G.Gaslini 5, Genoa 16147, Italy
| |
Collapse
|