1
|
Abdallah S, Tabebi M, Qanadilo S, Ali N, Wang J, D'Arcy P, Zhong W, Sjoberg F, Elmasry M, El-Serafi A. Modulation of biological activities in adipose derived stem cells by histone deacetylation. Sci Rep 2025; 15:3629. [PMID: 39880862 PMCID: PMC11779964 DOI: 10.1038/s41598-024-84652-1] [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: 09/23/2024] [Accepted: 12/25/2024] [Indexed: 01/31/2025] Open
Abstract
Difficult-to-heal wounds management accounts for about 4% of healthcare costs, highlighting the need for innovative solutions. Extracellular signals drive cell proliferation during tissue regeneration, while epigenetic mechanisms regulate stem cell homeostasis, differentiation, and skin repair. Exploring epigenetic regulation in adipose-derived stem cells (ADSCs) holds promise for improving skin injury treatments. We investigated the effects of histone deacetylase inhibitor (SAHA) on ADSCs to better understand its cellular and molecular impacts. ADSCs were treated with SAHA for 72 h, showing no change in cell viability at the studied concentrations. However, the expression of histone deacetylase decreased at 1000 nM, while the cell proliferation marker Ki-67 increased after SAHA treatment, as confirmed by immunofluorescence. CCND1 gene expression increased, whereas protein expression of the proliferating cell nuclear antigen (PCNA) decreased. Cell cycle analysis showed an increase in G2 phase in SAHA-treated cells. Microarray analysis revealed 74 upregulated and 40 downregulated differentially expressed genes, including upregulation of P53 targets, CDKN1A and MDM2. Proteomic analysis identified 631 upregulated and 823 downregulated proteins compared to the vehicle. Pathway enrichment analysis showed cell cycle, ATP-dependent chromatin remodeling and DNA processes were among the affected pathways. This study suggests SAHA modulates ADSCs' biological processes, highlighting its potential for skin regeneration.
Collapse
Affiliation(s)
- Sallam Abdallah
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Mouna Tabebi
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
- Clinical Genomics Linköping, Linköping University, Linköping, Sweden
| | - Sawsan Qanadilo
- Department of Biological Sciences, The University of Jordan, Amman, Jordan
| | - Neserin Ali
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jing Wang
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Pádraig D'Arcy
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Wen Zhong
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Folke Sjoberg
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Moustafa Elmasry
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
- Department of Hand Surgery and Plastic Surgery and Burns, University Hospital, Linköping, Sweden
| | - Ahmed El-Serafi
- The Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden.
- Department of Hand Surgery and Plastic Surgery and Burns, University Hospital, Linköping, Sweden.
| |
Collapse
|
2
|
Li S, Siengdee P, Hadlich F, Trakooljul N, Oster M, Reyer H, Wimmers K, Ponsuksili S. Dynamics of DNA methylation during osteogenic differentiation of porcine synovial membrane mesenchymal stem cells from two metabolically distinct breeds. Epigenetics 2024; 19:2375011. [PMID: 38956836 PMCID: PMC11225923 DOI: 10.1080/15592294.2024.2375011] [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: 01/12/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Mesenchymal stem cells (MSCs), with the ability to differentiate into osteoblasts, adipocytes, or chondrocytes, show evidence that the donor cell's metabolic type influences the osteogenic process. Limited knowledge exists on DNA methylation changes during osteogenic differentiation and the impact of diverse donor genetic backgrounds on MSC differentiation. In this study, synovial membrane mesenchymal stem cells (SMSCs) from two pig breeds (Angeln Saddleback, AS; German Landrace, DL) with distinct metabolic phenotypes were isolated, and the methylation pattern of SMSCs during osteogenic induction was investigated. Results showed that most differentially methylated regions (DMRs) were hypomethylated in osteogenic-induced SMSC group. These DMRs were enriched with genes of different osteogenic signalling pathways at different time points including Wnt, ECM, TGFB and BMP signalling pathways. AS pigs consistently exhibited a higher number of hypermethylated DMRs than DL pigs, particularly during the peak of osteogenesis (day 21). Predicting transcription factor motifs in regions of DMRs linked to osteogenic processes and donor breeds revealed influential motifs, including KLF1, NFATC3, ZNF148, ASCL1, FOXI1, and KLF5. These findings contribute to understanding the pattern of methylation changes promoting osteogenic differentiation, emphasizing the substantial role of donor the metabolic type and epigenetic memory of different donors on SMSC differentiation.
Collapse
Affiliation(s)
- Shuaichen Li
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Puntita Siengdee
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- Program in Applied Biological Sciences: Environmental Health, Chulabhorn Graduate Institute, 906 Kamphaeng Phet 6 Road, Lak-Si, Bangkok, Thailand
| | - Frieder Hadlich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Nares Trakooljul
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Michael Oster
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Henry Reyer
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Klaus Wimmers
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- Faculty of Agricultural and Environmental Sciences, University of Rostock, Rostock, Germany
| | - Siriluck Ponsuksili
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
3
|
Madkour MI, Hassan RE, Sherif NM, Awadallah S, Farahat NM, Abdelrahim DN, AlHasan FA, Taneera J, Faris ME. Changes in haptoglobin genotype-based gene expressions upon the observance of dawn-to-dusk intermittent fasting: a prospective cohort study on overweight and obese individuals. Front Nutr 2024; 11:1409344. [PMID: 39410930 PMCID: PMC11475474 DOI: 10.3389/fnut.2024.1409344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Intermittent fasting (IF) has been reported to be involved in ameliorating oxidative stress and lessening the systemic-low grade inflammation that predisposes to chronic diseases. Gene polymorphism is currently a main determining factor for the metabolic responses to different dietary and lifestyle modifications. Methods The current study was designed to explore the effect of observing four-week, dawn to dusk IF by participants with obesity on gene expression of the anti-inflammatory CD163, oxidative stress, and bioenergetics enzymes (SOD2, Nrf2, and TFAM), as well as metabolic and cellular regulatory genes (SIRT1 and SIRT3). Further, the study aimed to find out how haptoglobin (Hp) polymorphism modulates gene expression of the aforementioned genes and to determine changes in relative gene expressions of the aforementioned six genes based on Hp polymorphism in response to IF. Haptoglobin genotype was determined for the study subjects, and gene expressions were determined using qPCR. Gene expressions were assessed before and at the end of four consecutive weeks, dawn to sunset IF. Results The expressions of CD163, SOD, NfF2, and TFAM genes have significantly increased at the end of IF. At the same time, SIRT3 significantly decreased, implying that observing four consecutive weeks of dawn-to-dusk IF may enhance antioxidative stress response and reduce systemic inflammation. Conclusion Participants with genotypes Hp2-1 and Hp2-2 revealed upregulation of the antioxidant genes in response to the metabolic stress induced by IF compared with Hp1-1, implying that Hp polymorphism plays a key role in shaping the body's response to dietary modifications such as fasting.
Collapse
Affiliation(s)
- Mohamed I. Madkour
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences (RIMHS), Center of Excellence for Public Health, University of Sharjah, Sharjah, United Arab Emirates
| | - Rasha E. Hassan
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Naglaa M. Sherif
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Samir Awadallah
- Department of Medical Laboratories, College of Allied Medical Sciences, Zarqa University, Zarqa, Jordan
| | - Nada M. Farahat
- Research Institute of Medical and Health Sciences (RIMHS), Center of Excellence for Public Health, University of Sharjah, Sharjah, United Arab Emirates
| | - Dana N. Abdelrahim
- Research Institute of Medical and Health Sciences (RIMHS), Center of Excellence for Public Health, University of Sharjah, Sharjah, United Arab Emirates
| | - Fatima A. AlHasan
- Department of Medical Laboratory Medicine, King Fahd Hospital, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Jalal Taneera
- College of Medicine, Center of Excellence for Precision Medicine, Research Institute of Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, United Arab Emirates
| | - MoezAlIslam E. Faris
- Department of Clinical Nutrition and Dietetics, Faculty of Allied Medical Sciences, Applied Science Private University, Amman, Jordan
| |
Collapse
|
4
|
Alghazali R, Nugud A, El-Serafi A. Glycan Modifications as Regulators of Stem Cell Fate. BIOLOGY 2024; 13:76. [PMID: 38392295 PMCID: PMC10886185 DOI: 10.3390/biology13020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024]
Abstract
Glycosylation is a process where proteins or lipids are modified with glycans. The presence of glycans determines the structure, stability, and localization of glycoproteins, thereby impacting various biological processes, including embryogenesis, intercellular communication, and disease progression. Glycans can influence stem cell behavior by modulating signaling molecules that govern the critical aspects of self-renewal and differentiation. Furthermore, being located at the cell surface, glycans are utilized as markers for stem cell pluripotency and differentiation state determination. This review aims to provide a comprehensive overview of the current literature, focusing on the effect of glycans on stem cells with a reflection on the application of synthetic glycans in directing stem cell differentiation. Additionally, this review will serve as a primer for researchers seeking a deeper understanding of how synthetic glycans can be used to control stem cell differentiation, which may help establish new approaches to guide stem cell differentiation into specific lineages. Ultimately, this knowledge can facilitate the identification of efficient strategies for advancing stem cell-based therapeutic interventions.
Collapse
Affiliation(s)
- Raghad Alghazali
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58183 Linköping, Sweden
| | - Ahmed Nugud
- Clinical Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
- Gastroenterology, Hepatology & Nutrition, Sheikh Khalifa Medical City, Abu Dhabi 51900, United Arab Emirates
| | - Ahmed El-Serafi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58183 Linköping, Sweden
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58185 Linköping, Sweden
| |
Collapse
|
5
|
Sami MM, Sherief MH, El-Abaseri TB, El-Sakka AI, El-Serafi AT. Expression of epidermal growth factor receptor and human epidermal growth factor receptor 2 in urothelial bladder carcinoma in an Egyptian cohort: Clinical implication and prognostic significance. Urologia 2023:3915603221150965. [PMID: 36670543 DOI: 10.1177/03915603221150965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Bladder cancer (BC) has a particular importance in Egyptian patients due to aggressive behavior and absence of prognostic markers. OBJECTIVE To evaluate the expression of gene and protein expression of HER2 and epidermal growth factor (EGFR) in Egyptian patients with BC and ultimately to investigate their clinical implication and prognostic significance. MATERIAL AND METHODS The study was carried out on 46 patients with urothelial bladder BC. Tissue were obtained from transurethral resection (N = 22) and radical cystectomy (N = 24) specimens. The original hematoxylin and eosin slides were re-evaluated and the formalin fixed, paraffin-embedded (FFPE) tissues which had sufficient tumor tissue (>75%) and minimal or absent tumor necrosis were selected for immunohistochemistry (IHC) and RNA extraction. Furthermore, five control biopsies were obtained from patients with cystitis. Follow-up data were retrieved from the medical records which included the treatment regimen, disease recurrence and/or progression, and survival. RESULTS EGFR and HER2 protein were overexpressed in 35% and 46% of patients respectively. EGFR was correlated with the tumor size, grade and pathological stage, with a similar trend for HER2. The recurrence rate was higher in patients with expression of any of the markers. Gene expression was significantly higher (10.6-folds) for EGFR and (21-folds) for HER2 in patients with BC in comparison to control patients. Survival analysis showed lower median disease-free survival in association with HER2 protein overexpression. CONCLUSIONS Our data highlighted the prognostic significance of EGFR and HER in BC and proposed their possible use as predictive markers and potential therapeutic targets.
Collapse
Affiliation(s)
- Manal M Sami
- Department of Pathology, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.,Department of Pathology, Faculty of Medicine - Suez Canal University, Ismailia, Egypt
| | - Mahmoud H Sherief
- Department of Urology, Faculty of Medicine - Suez Canal University, Ismailia, Egypt
| | - Taghrid B El-Abaseri
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine - Suez Canal University, Ismailia, Egypt
| | - Ahmed I El-Sakka
- Department of Urology, Faculty of Medicine - Suez Canal University, Ismailia, Egypt
| | - Ahmed T El-Serafi
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine - Suez Canal University, Ismailia, Egypt.,Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
6
|
Alghfeli L, Parambath D, Tag Eldeen LA, El-Serafi I, El-Serafi AT. Non-additive effect of the DNA methylation inhibitor, 5-Aza-dC, and glass as a culture surface on osteogenic differentiation. Heliyon 2022; 8:e12433. [PMID: 36590514 PMCID: PMC9794900 DOI: 10.1016/j.heliyon.2022.e12433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/31/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
The clinical need for bone regenerative solutions is expanding with increasing life expectancy and escalating incidence of accidents. Several strategies are being investigated to enhance the osteogenic differentiation of stem cells. We previously reported two different approaches for this purpose, in monolayer and three-dimensional cell culture. The first approach was based on pretreating cells with 5-Aza-dC, a DNA methylation inhibitor, before the applying the differentiation media. The second approach was based on culturing cells on a glass surface during differentiation. In this study, we investigated the potential effect of combining both methods. Our results suggested that both approaches were associated with decreasing global DNA methylation levels. Cells cultured as a monolayer on glass surface showed enhancement in alkaline phosphatase activity at day 10, while 5-Aza-dC pretreatment enhanced the activity at day 5, irrespective of the culture surface. In three-dimensional pellet culture, 5-Aza-dC pretreatment enhanced osteogenesis through Runx-2 and TGF-β1 upregulation while the glass surface induced Osterix. Furthermore, pellets cultured on glass showed upregulation of a group of miRNAs, including pro-osteogenesis miR- 20a and miR -148b and anti-osteogenesis miR -125b, miR -31, miR -138, and miR -133a. Interestingly, 5-Aza-dC was not associated with a change of miRNAs in cells cultured on tissue culture plastic but reverted the upregulated miRNAs on the glass to the basal level. This study confirms the two approaches for enhancing osteogenic differentiation and contradicts their combination.
Collapse
Affiliation(s)
- Latifa Alghfeli
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Divyasree Parambath
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Loaa A. Tag Eldeen
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Egypt
| | - Ibrahim El-Serafi
- Basic Medical Sciences Department, College of Medicine, Ajman University, United Arab Emirates
- Department of Biochemistry, Faculty of Medicine, Port-Said University, Egypt
| | - Ahmed T. El-Serafi
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Egypt
- Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| |
Collapse
|
7
|
Ceccarelli S, Gerini G, Megiorni F, Pontecorvi P, Messina E, Camero S, Anastasiadou E, Romano E, Onesti MG, Napoli C, Marchese C. Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis. Front Cell Dev Biol 2022; 10:926180. [PMID: 36120582 PMCID: PMC9478209 DOI: 10.3389/fcell.2022.926180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/28/2022] [Indexed: 01/10/2023] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) represent a valid therapeutic option for clinical application in several diseases, due to their ability to repair damaged tissues and to mitigate the inflammatory/immune response. A better understanding of the underlying mechanisms regulating ASC biology might represent the chance to modulate their in vitro characteristics and differentiation potential for regenerative medicine purposes. Herein, we investigated the effects of the demethylating agent 5-azacytidine (5-aza) on proliferation, clonogenicity, migration, adipogenic differentiation and senescence of ASCs, to identify the molecular pathways involved. Through functional assays, we observed a detrimental effect of 5-aza on ASC self-renewal capacity and migration, accompanied by actin cytoskeleton reorganization, with decreased stress fibers. Conversely, 5-aza treatment enhanced ASC adipogenic differentiation, as assessed by lipid accumulation and expression of lineage-specific markers. We analyzed the involvement of the Akt/mTOR, MAPK and Wnt/β-catenin pathways in these processes. Our results indicated impairment of Akt and ERK phosphorylation, potentially explaining the reduced cell proliferation and migration. We observed a 5-aza-mediated inhibition of the Wnt signaling pathway, this potentially explaining the pro-adipogenic effect of the drug. Finally, 5-aza treatment significantly induced ASC senescence, through upregulation of the p53/p21 axis. Our data may have important translational implications, by helping in clarifying the potential risks and advantages of using epigenetic treatment to improve ASC characteristics for cell-based clinical approaches.
Collapse
Affiliation(s)
- S. Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- *Correspondence: S. Ceccarelli ,
| | - G. Gerini
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - F. Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - P. Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - E. Messina
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - S. Camero
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - E. Anastasiadou
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - E. Romano
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - M. G. Onesti
- Department of Surgery “P. Valdoni”, Unit of Plastic Surgery “P. Valdoni”, Sapienza University of Rome, Rome, Italy
| | - C. Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - C. Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
8
|
Larsson L, Kavanagh NM, Nguyen TVN, Castilho RM, Berglundh T, Giannobile WV. Influence of epigenetics on periodontitis and peri-implantitis pathogenesis. Periodontol 2000 2022; 90:125-137. [PMID: 35913702 DOI: 10.1111/prd.12453] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Periodontitis is a disease characterized by tooth-associated microbial biofilms that drive chronic inflammation and destruction of periodontal-supporting tissues. In some individuals, disease progression can lead to tooth loss. A similar condition can occur around dental implants in the form of peri-implantitis. The immune response to bacterial challenges is not only influenced by genetic factors, but also by environmental factors. Epigenetics involves the study of gene function independent of changes to the DNA sequence and its associated proteins, and represents a critical link between genetic and environmental factors. Epigenetic modifications have been shown to contribute to the progression of several diseases, including chronic inflammatory diseases like periodontitis and peri-implantitis. This review aims to present the latest findings on epigenetic influences on periodontitis and to discuss potential mechanisms that may influence peri-implantitis, given the paucity of information currently available.
Collapse
Affiliation(s)
- Lena Larsson
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Department of Periodontology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nolan M Kavanagh
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Trang V N Nguyen
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Rogerio M Castilho
- Department of Periodontics and Oral Medicine and Laboratory of Epithelial Biology, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Tord Berglundh
- Department of Periodontology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - William V Giannobile
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Khan AA, Khattak MNK, Parambath D, El-Serafi AT. Significant transcriptomic changes are associated with the inhibitory effects of 5-aza-2-deoxycytidine during adipogenic differentiation of MG-63 cells. Saudi J Biol Sci 2021; 28:7336-7348. [PMID: 34867036 PMCID: PMC8626271 DOI: 10.1016/j.sjbs.2021.08.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/25/2021] [Accepted: 08/12/2021] [Indexed: 11/30/2022] Open
Abstract
Our previous study revealed that the treatment of 5-aza-2-deoxycytidine (5-aza) inhibited while treatment of suberoylanilide hydroxamic acid (SAHA) enhanced the adipogenic differentiation of MG-63 cells. In this study, we examined the transcriptomic profiles of the derived adipocyte-like cells from MG-63 cells in the presence of 5-aza (Treatment 1) and SAHA (Treatment 2). Genome wide expression analysis showed high within sample variability for the adipocytes derived with 5-aza versus vehicle. Additionally, the expression profile of 5-aza derived cells was separated from the other sample groups. Differential analysis on the pairwise comparison of 5-aza versus control and SAHA versus 5-aza identified 1290 and 1086 differentially expressed (DE) genes, respectively. Furthermore, some overlap was observed between the up and down-regulated DE genes of 5-aza versus control and SAHA versus control (jaccard score 0.3) as well as between the differentially regulated genes of 5-aza versus control and 5-aza versus SAHA (jaccard score 0.29). A total of 73 transcription factors (TFs) were differentially expressed across all the pair wise comparisons with some overlap between the under and over expressed TFs of 5-aza versus control and 5-aza versus SAHA (jaccard score 0.29). Unsupervised clustering of TFs showed that the samples within the group are consistent in expression and the samples cluster in accordance with the group. Several GO terms related to enhanced adipogenesis such as neutral lipid biosynthetic process, lipid metabolic processes, cellular amide metabolic processes and cellular carbohydrate metabolic processes were enriched in the down regulated genes of 5-aza derived adipocytes versus control, indicating 5-aza inhibit the adipogenic differentiation of MG-63 cells. GSEA analysis on selected gene sets of MAPK and PI3K signaling pathway in MSigDB identified the pathways were up-regulated in 5-aza versus control. This study revealed that inhibition of MG-63 adipogenesis due to 5-aza treatment is associated with large transcriptomics changes and further research is needed to unravel the roles of these genes in the adipogenesis.
Collapse
Affiliation(s)
- Amir Ali Khan
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.,Human Genetics and Stem Cells Research Group, Research Institute of Sciences & Engineering (RISE), University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Muhammad Nasir Khan Khattak
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.,Human Genetics and Stem Cells Research Group, Research Institute of Sciences & Engineering (RISE), University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Divyasree Parambath
- Sharjah Institute for Medical and Health Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahmed Taher El-Serafi
- Sharjah Institute for Medical and Health Research, University of Sharjah, Sharjah 27272, United Arab Emirates.,Department of Biomedical and Clinical Sciences (BKV), Linköping University, P.O. Box 581 83, Linköping, Sweden.,Medical Biochemistry department, Faculty of Medicine, Suez Canal University, 41522, Ismailia, Egypt
| |
Collapse
|
10
|
Alghfeli L, Parambath D, Manzoor S, Roach HI, Oreffo RO, El-Serafi AT. Synthesis of scaffold-free, three dimensional, osteogenic constructs following culture of skeletal osteoprogenitor cells on glass surfaces. Bone Rep 2021; 15:101143. [PMID: 34746337 PMCID: PMC8554168 DOI: 10.1016/j.bonr.2021.101143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Efficient differentiation of stem cells into three-dimensional (3D) osteogenic construct is still an unmet challenge. These constructs can be crucial for patients with bone defects due to congenital or traumatic reasons. The modulation of cell fate and function as a consequence of interaction with the physical and chemical properties of materials is well known. METHODS The current study has examined the osteogenic differentiation potential of human skeletal populations following culture on glass surfaces, as a monolayer, or in glass tubes as a pellet culture. The 3D prosperities were assessed morphometrically and the differentiation was evaluated through molecular characterization as well as matrix formation. RESULTS Early temporal expression of alkaline phosphatase expression of skeletal populations was observed following culture on glass surfaces. Skeletal populations seeded on glass tubes, adhered as a monolayer to the tube base and subsequently formed 3D pellets at the air -media interface. The pellets cultured on glass displayed 4.9 ± 1.3 times the weight and 2.9 ± 0.1 the diameter of their counterpart cultured in plastic tubes and displayed enhanced production of osteogenic matrix proteins, such a collagen I and osteonectin. The size and weight of the pellets correlated with surface area in contrast to cell numbers seeded. Global DNA methylation level was decreased in pellets cultured on glass. In contrast, gene expression analysis confirmed upregulation extracellular matrix proteins and osteogenesis-related growth factors. CONCLUSION This simple approach to the culture of skeletal cells on glass tubes provides a scaffold-free, 3D construct platform for generating pellets enabling analysis and evaluation of tissue development and integration of multiple constructs with implications for tissue repair and regenerative application on scale-up.
Collapse
Key Words
- 3D culture
- 3D, three-dimensional
- A/S, Alcian blue/Sirius red/Weigert's haematoxylin
- ALP, Alkaline Phosphatase
- BMP, bone morphogenic protein
- BMSC, human bone marrow stromal
- CSF, colony stimulating factor
- Ct, Cycle threshold
- Differentiation
- EGF, epidermal growth factor
- FC, Fetal bone cells
- FCS, Fetal Calf Serum
- FGF, fibroblast growth factor
- FN1, fibronectin
- GLI, GLI family zinc finger 1
- HIPPIE, Human Integrated Protein Interaction Reference
- ITGA3, integrin A3
- MMP, matrix metalloprotease
- Osteogenesis
- Osteoprogenitor cells
- P/S, penicillin and streptomycin
- Pellets
- R, receptor
- TGF, β transforming growth factor beta
- TGFBR2 transforming growth factor beta receptor 2 VDR, vitamin D receptor
- gDNA, genomic DNA
- iMSC, immortalized human bone marrow derived, mesenchymal stem cells
- vWF, von Willebrand factor
Collapse
Affiliation(s)
- Latifa Alghfeli
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Divyasree Parambath
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Shaista Manzoor
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Helmtrud I. Roach
- Bone and Joint Research Group, Institute of Developmental Sciences, University of Southampton, School of Medicine, UK
| | - Richard O.C. Oreffo
- Bone and Joint Research Group, Institute of Developmental Sciences, University of Southampton, School of Medicine, UK
| | - Ahmed T. El-Serafi
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
- Medical Biochemistry Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Sweden
| |
Collapse
|
11
|
Man K, Mekhileri NV, Lim KS, Jiang LH, Woodfield TBF, Yang XB. MI192 induced epigenetic reprogramming enhances the therapeutic efficacy of human bone marrows stromal cells for bone regeneration. Bone 2021; 153:116138. [PMID: 34339909 DOI: 10.1016/j.bone.2021.116138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022]
Abstract
Human bone marrow stromal cells (hBMSCs) have been extensively utilised for bone tissue engineering applications. However, they are associated with limitations that hinder their clinical utility for bone regeneration. Cell fate can be modulated via altering their epigenetic functionality. Inhibiting histone deacetylase (HDAC) enzymes have been reported to promote osteogenic differentiation, with HDAC3 activity shown to be causatively associated with osteogenesis. Therefore, this study aimed to investigate the potential of using an HDAC2 & 3 selective inhibitor - MI192 to induce epigenetic reprogramming of hBMSCs and enhance its therapeutic efficacy for bone formation. Treatment with MI192 caused a time-dose dependant reduction in hBMSCs viability. MI192 was also found to substantially alter hBMSCs epigenetic function through reduced HDAC activity and increased histone acetylation. hBMSCs were pre-treated with MI192 (50 μM) for 48 h prior to osteogenic induction. MI192 pre-treatment significantly upregulated osteoblast-related gene/protein expression (Runx2, ALP, Col1a and OCN) and enhanced alkaline phosphatase specific activity (ALPSA) (1.43-fold) (P ≤ 0.001). Moreover, MI192 substantially increased hBMSCs extracellular matrix calcium deposition (1.4-fold) (P ≤ 0.001) and mineralisation when compared to the untreated control. In 3D microtissue culture, MI192 significantly promoted hBMSCs osteoblast-related gene expression and ALPSA (> 2.41-fold) (P ≤ 0.001). Importantly, MI192 substantially enhanced extracellular matrix deposition (ALP, Col1a, OCN) and mineralisation (1.67-fold) (P ≤ 0.001) within the bioassembled-microtissue (BMT) construct. Following 8-week intraperitoneal implantation within nude mice, MI192 treated hBMSCs exhibited enhanced extracellular matrix deposition and mineralisation (2.39-fold) (P ≤ 0.001) within the BMT when compared to the untreated BMT construct. Taken together, these results demonstrate that MI192 effectively altered hBMSCs epigenetic functionality and is capable of promoting hBMSCs osteogenic differentiation in vitro and in vivo, indicating the potential of using epigenetic reprogramming to enhance the therapeutic efficacy of hBMSCs for bone augmentation strategies.
Collapse
Affiliation(s)
- Kenny Man
- Biomaterial and Tissue Engineering Group, School of Dentistry, University of Leeds, Leeds, UK; School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Naveen V Mekhileri
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Khoon S Lim
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Tim B F Woodfield
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Xuebin B Yang
- Biomaterial and Tissue Engineering Group, School of Dentistry, University of Leeds, Leeds, UK.
| |
Collapse
|
12
|
Analysis of Gene Expression Patterns of Epigenetic Enzymes Dnmt3a, Tet1 and Ogt in Murine Chondrogenic Models. Cells 2021; 10:cells10102678. [PMID: 34685658 PMCID: PMC8534543 DOI: 10.3390/cells10102678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 10/02/2021] [Indexed: 12/30/2022] Open
Abstract
We investigated the gene expression pattern of selected enzymes involved in DNA methylation and the effects of the DNA methylation inhibitor 5-azacytidine during in vitro and in vivo cartilage formation. Based on the data of a PCR array performed on chondrifying BMP2-overexpressing C3H10T1/2 cells, the relative expressions of Tet1 (tet methylcytosine dioxygenase 1), Dnmt3a (DNA methyltransferase 3), and Ogt (O-linked N-acetylglucosamine transferase) were further examined with RT-qPCR in murine cell line-based and primary chondrifying micromass cultures. We found very strong but gradually decreasing expression of Tet1 throughout the entire course of in vitro cartilage differentiation along with strong signals in the cartilaginous embryonic skeleton using specific RNA probes for in situ hybridization on frozen sections of 15-day-old mouse embryos. Dnmt3a and Ogt expressions did not show significant changes with RT-qPCR and gave weak in situ hybridization signals. The DNA methylation inhibitor 5-azacytidine reduced cartilage-specific gene expression and cartilage formation when applied during the early stages of chondrogenesis. In contrast, it had a stimulatory effect when added to differentiated chondrocytes, and quantitative methylation-specific PCR proved that the DNA methylation pattern of key chondrogenic marker genes was altered by the treatment. Our results indicate that the DNA demethylation inducing Tet1 plays a significant role during chondrogenesis, and inhibition of DNA methylation exerts distinct effects in different phases of in vitro cartilage formation.
Collapse
|
13
|
de Nigris F, Ruosi C, Colella G, Napoli C. Epigenetic therapies of osteoporosis. Bone 2021; 142:115680. [PMID: 33031975 DOI: 10.1016/j.bone.2020.115680] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 12/29/2022]
Abstract
The study of epigenetics reaches its 50th anniversary, however, its clinical application is gradually coming into the clinical setting. Osteoporosis is one of the major and widely diffused bone diseases. Pathogenic mechanisms at the epigenetic level may interfere with bone remodeling occurring during osteoporosis. Preclinical models were used to understand whether such events may interfere with the disease. Besides, observational clinical trials investigated epigenetic-related biomarkers. This effort leads to some epigenetic-related therapies in clinical trials for the treatment of osteoporosis. Bisphosphonates (BPs), target therapy blocking RANK/RANKL pathway, and anti-sclerostin antibody (SOST) are the main therapeutic approaches. However, future large trials will reveal whether epigenetic therapies of osteoporosis will remain a work in progress or data will become more robust in the real-world management of these frailty patients.
Collapse
Affiliation(s)
- Filomena de Nigris
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Carlo Ruosi
- Department of Public Health, Federico II University, 80132 Naples, Italy
| | - Gianluca Colella
- Department of Public Health, Federico II University, 80132 Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy; IRCCS SDN, 80134 Naples, Italy
| |
Collapse
|
14
|
Shahin H, Elmasry M, Steinvall I, Söberg F, El-Serafi A. Vascularization is the next challenge for skin tissue engineering as a solution for burn management. BURNS & TRAUMA 2020; 8:tkaa022. [PMID: 32766342 PMCID: PMC7396265 DOI: 10.1093/burnst/tkaa022] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/23/2020] [Indexed: 12/19/2022]
Abstract
Skin regeneration represents a promising line of management for patients with skin loss, including burn victims. The current approach of spraying single cells over the defective areas results in variable success rates in different centers. The modern approach is to synthesize a multilayer skin construct that is based on autologous stem cells. One of the main complications with different types of transplants is sloughing due to the absence of proper vascularization. Ensuring proper vascularization will be crucial for the integration of skin constructs with the surrounding tissues. Combination of the right cells with scaffolds of proper physico-chemical properties, vascularization can be markedly enhanced. The material effect, pore size and adsorption of certain proteins, as well as the application of appropriate growth factors, such as vascular endothelial growth factors, can have an additive effect. A selection of the most effective protocols is discussed in this review.
Collapse
Affiliation(s)
- Hady Shahin
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
- Faculty of Biotechnology, MSA University, 26 July Mehwar Road, 125 85, 6th October City. Egypt
| | - Moustafa Elmasry
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
| | - Ingrid Steinvall
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
| | - Folke Söberg
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
| | - Ahmed El-Serafi
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
| |
Collapse
|
15
|
5-Aza-2-deoxycytidine inhibits osteolysis induced by titanium particles by regulating RANKL/OPG ratio. Biochem Biophys Res Commun 2020; 529:629-634. [PMID: 32736684 DOI: 10.1016/j.bbrc.2020.05.192] [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: 05/06/2020] [Accepted: 05/25/2020] [Indexed: 12/20/2022]
Abstract
Periprosthetic osteolysis (PIO) caused by wear particles is the main cause of implant failure, which is regulated by nuclear factor κ B receptor activator ligand (RANKL)/osteoprotegerin (OPG) system. At present, there is a lack of effective drugs to prevent or treat PIO. Previous studies have confirmed that DNA methylation is closely related to postmenopausal osteoporosis and can affect the expression of OPG and RANKL. However, the relationship between DNA methylation and PIO is not clear. In this study, we investigated the inhibitory effect of 5-Aza-2-deoxycytidine (AzadC) on osteolysis induced by titanium particles in a mouse model. This inhibition mechanism is achieved by changing the ratio of RANKL/OPG in the osteolysis model. In conclusion, there is a relationship between DNA methylation and PIO. AzadC has a certain inhibitory effect on osteolysis induced by titanium particles. Regulating DNA methylation may be a new way to treat PIO. Our findings lay a foundation for epigenetic understanding and intervention of osteolysis.
Collapse
|
16
|
DNA hypomethylation during MSC chondrogenesis occurs predominantly at enhancer regions. Sci Rep 2020; 10:1169. [PMID: 31980739 PMCID: PMC6981252 DOI: 10.1038/s41598-020-58093-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022] Open
Abstract
Regulation of transcription occurs in a cell type specific manner orchestrated by epigenetic mechanisms including DNA methylation. Methylation changes may also play a key role in lineage specification during stem cell differentiation. To further our understanding of epigenetic regulation in chondrocytes we characterised the DNA methylation changes during chondrogenesis of mesenchymal stem cells (MSCs) by Infinium 450 K methylation array. Significant DNA hypomethylation was identified during chondrogenic differentiation including changes at many key cartilage gene loci. Integration with chondrogenesis gene expression data revealed an enrichment of significant CpGs in upregulated genes, while characterisation of significant CpG loci indicated their predominant localisation to enhancer regions. Comparison with methylation profiles of other tissues, including healthy and diseased adult cartilage, identified chondrocyte-specific regions of hypomethylation and the overlap with differentially methylated CpGs in osteoarthritis. Taken together we have associated DNA methylation levels with the chondrocyte phenotype. The consequences of which has potential to improve cartilage generation for tissue engineering purposes and also to provide context for observed methylation changes in cartilage diseases such as osteoarthritis.
Collapse
|
17
|
Elsharkawi I, Parambath D, Saber-Ayad M, Khan AA, El-Serafi AT. Exploring the effect of epigenetic modifiers on developing insulin-secreting cells. Hum Cell 2019; 33:1-9. [PMID: 31755075 DOI: 10.1007/s13577-019-00292-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/11/2019] [Indexed: 12/24/2022]
Abstract
Diabetes is a worldwide health problem with increasing incidence. The current management modalities did not succeed to decrease comorbidities. This study aimed at enhancing the regenerative solution for diabetes by improving the differentiation of mesenchymal stromal cells (MSC) into glucose-sensitive, insulin-secreting cells through an epigenetic modification approach. A 3-day treatment protocol with the epigenetic modifiers, either decitabine (5-aza-2'-deoxycytidine; Aza); a DNA methylation inhibitor or Vorinostat (suberoylanilide hydroxamic acid; SAHA); a histone deacetylase inhibitor was added to two different human stem cell lines. The cells followed a multi-step differentiation protocol that provided the critical triggers in a temporal approach. Aza-pretreated group showed higher intracellular expression of insulin and the transcription factor 'PDX-1'. The cells responded to the high glucose challenge by secreting insulin in the media, as shown by ELISA. Gene expression showed induction of the genes for insulin, the glucose transporter 2, glucokinase, as well as the transcription factors MafA and NKX6.1. Although SAHA showed upregulation of insulin secretion, in comparison to control, the cells could not respond to the high glucose challenge. Interestingly, Aza-treated cells showed a significant decrease in the global DNA methylation level at the end of the culture. In conclusion, this additional step with Aza could enhance the response of MSC to the classical differentiation protocol for insulin-secreting cells and may help in establishing a regenerative solution for patients with diabetes.
Collapse
Affiliation(s)
| | | | - Maha Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, UAE.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE.,College of Medicine, Cairo University, Cairo, Egypt
| | - Amir Ali Khan
- Department of Applied Biology, College of Science, University of Sharjah, Sharjah, UAE
| | - Ahmed T El-Serafi
- College of Medicine, University of Sharjah, Sharjah, UAE. .,Faculty of Medicine, Suez Canal University, Ismailia, Egypt. .,Department of Hand Surgery and Plastic Surgery and Burns, 401A, Building 462, Floor 11, Linköping University Hospital, P. O. Box: 581 85, Linköping, Sweden.
| |
Collapse
|
18
|
Allas L, Boumédiene K, Baugé C. Epigenetic dynamic during endochondral ossification and articular cartilage development. Bone 2019; 120:523-532. [PMID: 30296494 DOI: 10.1016/j.bone.2018.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/23/2022]
Abstract
Within the last decade epigenetics has emerged as fundamental regulator of numerous cellular processes, including those orchestrating embryonic and fetal development. As such, epigenetic factors play especially crucial roles in endochondral ossification, the process by which bone tissue is created, as well during articular cartilage formation. In this review, we summarize the recent discoveries that characterize how DNA methylation, histone post-translational modifications and non-coding RNA (e.g., miRNA and lcnRNA) epigenetically regulate endochondral ossification and chondrogenesis.
Collapse
Affiliation(s)
- Lyess Allas
- Normandie Univ, UNICAEN, EA7451 BioConnecT, Caen, France
| | | | | |
Collapse
|
19
|
Paradoxical effects of the epigenetic modifiers 5-aza-deoxycytidine and suberoylanilide hydroxamic acid on adipogenesis. Differentiation 2019; 106:1-8. [PMID: 30818187 DOI: 10.1016/j.diff.2019.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/12/2019] [Accepted: 02/13/2019] [Indexed: 01/17/2023]
Abstract
Adipogenesis is an important biological process that is linked to obesity and metabolic disorders. On the other hand, fat regeneration is crucial as a restorative approach following mastectomy or severe burn injury. Furthermore, optimizing an in-vitro model of adipogenesis, which would help in understanding the possible effects and/or side effects of fat-soluble drugs and anti-obesity remedies, in addition to the developmental studies. Epigenetic is an important factor that is involved in cellular differentiation and commitment. This study aimed at investigating the effect of DNA methylation and histone deactylases inhibitors, 5-Aza-deoxycytidine (5-Aza-dC) and Suberoylanilide hydroxamic acid (SAHA), on the adipogenic differentiation process. The two modifiers were applied according to our previously published protocol, followed by three cycles of a classical, two-step adipogenesis protocol. The cells pretreated with SAHA showed enhanced expression of the many adipogenic genes, including peroxisome proliferator-activated receptor-γ as well as the accumulation of intracytoplasmic fat as shown by oil red and Nile red staining and the secretion of adipokines, such as MCP-1 and IP-10. On contrary, 5-Aza-dC inhibited all these markers. In conclusion, adding the reported step with SAHA to the differentiation protocols could have an impact on the progress of the in-vitro fat regenerative approach. The possible role of 5-Aza-dC in the inhibition of adipogenesis can be of clinical interest and will need further characterization in the future.
Collapse
|
20
|
van Meurs JB, Boer CG, Lopez-Delgado L, Riancho JA. Role of Epigenomics in Bone and Cartilage Disease. J Bone Miner Res 2019; 34:215-230. [PMID: 30715766 DOI: 10.1002/jbmr.3662] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/03/2018] [Accepted: 01/02/2019] [Indexed: 12/14/2022]
Abstract
Phenotypic variation in skeletal traits and diseases is the product of genetic and environmental factors. Epigenetic mechanisms include information-containing factors, other than DNA sequence, that cause stable changes in gene expression and are maintained during cell divisions. They represent a link between environmental influences, genome features, and the resulting phenotype. The main epigenetic factors are DNA methylation, posttranslational changes of histones, and higher-order chromatin structure. Sometimes non-coding RNAs, such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are also included in the broad term of epigenetic factors. There is rapidly expanding experimental evidence for a role of epigenetic factors in the differentiation of bone cells and the pathogenesis of skeletal disorders, such as osteoporosis and osteoarthritis. However, different from genetic factors, epigenetic signatures are cell- and tissue-specific and can change with time. Thus, elucidating their role has particular difficulties, especially in human studies. Nevertheless, epigenomewide association studies are beginning to disclose some disease-specific patterns that help to understand skeletal cell biology and may lead to development of new epigenetic-based biomarkers, as well as new drug targets useful for treating diffuse and localized disorders. Here we provide an overview and update of recent advances on the role of epigenomics in bone and cartilage diseases. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Cindy G Boer
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Laura Lopez-Delgado
- Department of Internal Medicine, Hospital U M Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Jose A Riancho
- Department of Internal Medicine, Hospital U M Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| |
Collapse
|
21
|
Wang Y, Shi ZY, Feng J, Cao JK. HDAC6 regulates dental mesenchymal stem cells and osteoclast differentiation. BMC Oral Health 2018; 18:190. [PMID: 30463548 PMCID: PMC6247693 DOI: 10.1186/s12903-018-0624-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 09/16/2018] [Indexed: 02/07/2023] Open
Abstract
Background Dental and periodontal tissue development is a complicated process involving a finely regulated network of communication among various cell types. Understanding the mechanisms involved in regulating dental mesenchymal stem cells (MSCs) and osteoclast cell differentiation is critical. However, it is still unclear whether histone deacetylase HDAC6 is involved in dental MSCs fate determination and osteoclast differentiation. Methods We used shRNA and siRNA knockdown to explore the role of HDAC6 in dental MSCs odontogenic differentiation and osteoclasts maturation. Results Based on HDAC6 knockdown dental MSCs, our data suggest that HDAC6 knockdown significantly increases alkaline phosphate activity and mineralized nodules formation. Additionally, mRNA expression of odontogenic marker genes (OSX, OCN, and OPN) was induced by HDAC6 knockdown. By using HDAC6 siRNA, we knocked down HDAC6 in osteoclast precursor RAW 264.7 cells. Our data suggests that HDAC6 knockdown significantly inhibited osteoclasts differentiation. Additionally, mRNA expression of osteoclast marker genes Trap, Mmp9, and Ctsk was decreased by HDAC6 knockdown. Conclusions Our study demonstrated that HDAC6 plays an important role in regulating dental MSCs and osteoclasts differentiation.
Collapse
Affiliation(s)
- Yi Wang
- Department of Stomatology, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, People's Republic of China
| | - Zhi Yun Shi
- Department of Stomatology, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, People's Republic of China
| | - Jin Feng
- Department of Stomatology, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, People's Republic of China
| | - Jun Kai Cao
- Department of Stomatology, Chinese PLA General Hospital, 28th Fuxing Road, Beijing, 100853, People's Republic of China.
| |
Collapse
|
22
|
Meng F, Li Z, Zhang Z, Yang Z, Kang Y, Zhao X, Long D, Hu S, Gu M, He S, Wu P, Chang Z, He A, Liao W. MicroRNA-193b-3p regulates chondrogenesis and chondrocyte metabolism by targeting HDAC3. Theranostics 2018; 8:2862-2883. [PMID: 29774080 PMCID: PMC5957014 DOI: 10.7150/thno.23547] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/12/2018] [Indexed: 12/17/2022] Open
Abstract
Histone deacetylase 3 (HDAC3) plays a pivotal role in the repression of cartilage-specific gene expression in human chondrocytes. The aim of this study was to determine whether microRNA-193b-3p (miR-193b-3p) regulates the expression of HDAC3 during chondrogenesis and chondrocyte metabolism. Methods: miR-193b-3p expression was assessed in a human mesenchymal stem cell (hMSC) model of chondrogenesis, in interleukin-1β (IL-1β)-treated primary human chondrocytes (PHCs), and in non-degraded and degraded cartilage. hMSCs and PHCs were transfected with miR-193b-3p or its antisense inhibitor. A direct interaction between miR-193b-3p and its putative binding site in the 3'-untranslated region (3'-UTR) of HDAC3 mRNA was confirmed by performing luciferase reporter assays. Chondrocytes were transfected with miR-193b-3p before performing a chromatin immunoprecipitation assay with an anti-acetylated histone H3 antibody. To investigate miR-193b-3p-transfected PHCs in vivo, they were seeded in tricalcium phosphate-collagen-hyaluronate (TCP-COL-HA) scaffolds, which were then implanted in nude mice. In addition, plasma exosomal miR-193b-3p in samples from normal controls and patients with osteoarthritis (OA) were measured. Results: miR-193b-3p expression was elevated in chondrogenic and hypertrophic hMSCs, while expression was significantly reduced in degraded cartilage compared to non-degraded cartilage. In addition, miR-193b-3p suppressed the activity of reporter constructs containing the 3'-UTR of HDAC3, inhibited HDAC3 expression, and promoted histone H3 acetylation in the COL2A1, AGGRECAN, COMP, and SOX9 promoters. Treatment with the HDAC inhibitor trichostatin A (TSA) increased cartilage-specific gene expression and enhanced hMSCs chondrogenesis. TSA also increased AGGRECAN expression and decreased MMP13 expression in IL-1β-treated PHCs. Further, 8 weeks after implanting PHC-seeded TCP-COL-HA scaffolds subcutaneously in nude mice, we found that miR-193b overexpression strongly enhanced in vivo cartilage formation compared to that found under control conditions. We also found that patients with OA had lower plasma exosomal miR-193b levels than control subjects. Conclusions: These findings indicate that miR-193b-3p directly targets HDAC3, promotes H3 acetylation, and regulates hMSC chondrogenesis and metabolism in PHCs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Aishan He
- Department of Joint Surgery, First Affiliated Hospital of SunYat-sen University, Guangzhou, Guangdong 510080, China
| | - Weiming Liao
- Department of Joint Surgery, First Affiliated Hospital of SunYat-sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
23
|
Larsson L, Pilipchuk SP, Giannobile WV, Castilho RM. When epigenetics meets bioengineering-A material characteristics and surface topography perspective. J Biomed Mater Res B Appl Biomater 2017; 106:2065-2071. [PMID: 28741893 DOI: 10.1002/jbm.b.33953] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 06/05/2017] [Accepted: 06/15/2017] [Indexed: 12/15/2022]
Abstract
The field of tissue engineering and regenerative medicine (TE/RM) involves regeneration of tissues and organs using implantable biomaterials. The term epigenetics refers to changes in gene expression that are not encoded in the DNA sequence, leading to remodeling of the chromatin and activation or inactivation of gene expression. Recently, studies have demonstrated that these modifications are influenced not only by biological cues but also by mechanical and topographical signals. This review highlights the current knowledge on emerging approaches in TE/RM with a focus on the effect of materials and topography on the epigenetic expression pattern in cells with potential impacts on modulating regenerative biology. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 2065-2071, 2018.
Collapse
Affiliation(s)
- Lena Larsson
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan.,Department of Periodontology, Institute of Odontology, University of Gothenburg, Sweden
| | - Sophia P Pilipchuk
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
| | - William V Giannobile
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan.,Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
| | - Rogerio M Castilho
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan.,Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan
| |
Collapse
|
24
|
Lee J, Im GI. Effects of Trichostatin A on the Chondrogenesis from Human Mesenchymal Stem Cells. Tissue Eng Regen Med 2017; 14:403-410. [PMID: 30603496 DOI: 10.1007/s13770-017-0041-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/17/2016] [Accepted: 09/12/2016] [Indexed: 12/01/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) are a class of compounds that suppress the function of histone deacetylases (HDACs). This study was performed to examine the effects of Trichostatin A (TSA), a typical HDACi, on chondrogenesis of human bone marrow mesenchymal stem cells (hBMMSCs) and related molecular pathways. After evaluating the concentration for cytotoxicity and HDAC activity, hBMMSCs underwent chondrogenic differentiation in pellet culture with or without TSA for 21 days. The weight of TSA-treated pellets was 25% lower than that of untreated pellets. DNA level was not significantly different, but glycosaminoglycan content per DNA level was lower in TSA-treated pellets than that of untreated pellets. Gene expression of the chondrogenic markers (SOX9, Aggrecan, and Col2A1) decreased by by 12.9-fold, 8.9-fold, and 7.6-fold respectively in TSA-treated pellets compared with that in TSA-untreated pellets. TSA-treated pellets had lower cell density and lower proteoglycan staining content compared with those of TSA-untreated pellets. A microarray analysis from TSA-treated pellets showed that 1,467 chondrogenic-related genes were downregulated and 1,524 were upregulated by more than 2-fold compared with TSA-untreated pellets. Col10A1, TGF-β3, and SOX9 decreased significantly by 10-fold, 2.1-fold, and 3.2-fold respectively in TSA-treated pellets compared with those in untreated pellets, whereas expression of BMP4 and FGFR3 increased significantly by 2.1-fold and 5.4-fold respectively. It is concluded that TSA inhibits chondrogenesis and does not seem to be useful for cartilage tissue engineering of hBMMSCs.
Collapse
Affiliation(s)
- Jimin Lee
- Department of Orthopaedics, Dongguk University Ilsan Hospital, 27, Dongguk-ro, Goyang, 10326 Republic of Korea
| | - Gun-Il Im
- Department of Orthopaedics, Dongguk University Ilsan Hospital, 27, Dongguk-ro, Goyang, 10326 Republic of Korea
| |
Collapse
|
25
|
Human Pluripotent Stem Cells: Advances in Chondrogenic Differentiation and Articular Cartilage Regeneration. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s40610-016-0041-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
26
|
Pérez-Campo FM, Riancho JA. Epigenetic Mechanisms Regulating Mesenchymal Stem Cell Differentiation. Curr Genomics 2015; 16:368-383. [PMID: 27019612 PMCID: PMC4765524 DOI: 10.2174/1389202916666150817202559] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 03/27/2015] [Accepted: 04/07/2015] [Indexed: 12/28/2022] Open
Abstract
Human Mesenchymal Stem Cells (hMSCs) have emerged in the last few years as one of the most promising therapeutic cell sources and, in particular, as an important tool for regenerative medicine of skeletal tissues. Although they present a more restricted potency than Embryonic Stem (ES) cells, the use of hMCS in regenerative medicine avoids many of the drawbacks characteristic of ES cells or induced pluripotent stem cells. The challenge in using these cells lies into developing precise protocols for directing cellular differentiation to generate a specific cell lineage. In order to achieve this goal, it is of the upmost importance to be able to control de process of fate decision and lineage commitment. This process requires the coordinate regulation of different molecular layers at transcriptional, posttranscriptional and translational levels. At the transcriptional level, switching on and off different sets of genes is achieved not only through transcriptional regulators, but also through their interplay with epigenetic modifiers. It is now well known that epigenetic changes take place in an orderly way through development and are critical in the determination of lineage-specific differentiation. More importantly, alteration of these epigenetic changes would, in many cases, lead to disease generation and even tumour formation. Therefore, it is crucial to elucidate how epigenetic factors, through their interplay with transcriptional regulators, control lineage commitment in hMSCs.
Collapse
Affiliation(s)
- Flor M. Pérez-Campo
- Department of Internal Medicine, Hospital U. Marqués de Valdecilla-IDIVAL Universidad de Cantabria, 39008 Santander, Cantabria, Spain
| | | |
Collapse
|
27
|
Baker EK, Taylor S, Gupte A, Chalk AM, Bhattacharya S, Green AC, Martin TJ, Strbenac D, Robinson MD, Purton LE, Walkley CR. Wnt inhibitory factor 1 (WIF1) is a marker of osteoblastic differentiation stage and is not silenced by DNA methylation in osteosarcoma. Bone 2015; 73:223-32. [PMID: 25571841 DOI: 10.1016/j.bone.2014.12.063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/03/2014] [Accepted: 12/28/2014] [Indexed: 12/29/2022]
Abstract
Wnt pathway targeting is of high clinical interest for treating bone loss disorders such as osteoporosis. These therapies inhibit the action of negative regulators of osteoblastic Wnt signaling. The report that Wnt inhibitory factor 1 (WIF1) was epigenetically silenced via promoter DNA methylation in osteosarcoma (OS) raised potential concerns for such treatment approaches. Here we confirm that Wif1 expression is frequently reduced in OS. However, we demonstrate that silencing is not driven by DNA methylation. Treatment of mouse and human OS cells showed that Wif1 expression was robustly induced by HDAC inhibition but not by methylation inhibition. Consistent with HDAC dependent silencing, the Wif1 locus in OS was characterized by low acetylation levels and a bivalent H3K4/H3K27-trimethylation state. Wif1 expression marked late stages of normal osteoblast maturation and stratified OS tumors based on differentiation stage across species. Culture of OS cells under differentiation inductive conditions increased expression of Wif1. Together these results demonstrate that Wif1 is not targeted for silencing by DNA methylation in OS. Instead, the reduced expression of Wif1 in OS cells is in context with their stage in differentiation.
Collapse
Affiliation(s)
- Emma K Baker
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Australia.
| | - Scott Taylor
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia
| | - Ankita Gupte
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia
| | - Alistair M Chalk
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Australia
| | - Shreya Bhattacharya
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Australia
| | - Alanna C Green
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Australia
| | - T John Martin
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Australia; Bone Cell Biology and Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia
| | - Dario Strbenac
- Cancer Epigenetics, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Louise E Purton
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Australia
| | - Carl R Walkley
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Australia.
| |
Collapse
|
28
|
Abstract
Due to a blood supply shortage, articular cartilage has a limited capacity for self-healing once damaged. Articular chondrocytes, cartilage progenitor cells, embryonic stem cells, and mesenchymal stem cells are candidate cells for cartilage regeneration. Significant current attention is paid to improving chondrogenic differentiation capacity; unfortunately, the potential chondrogenic hypertrophy of differentiated cells is largely overlooked. Consequently, the engineered tissue is actually a transient cartilage rather than a permanent one. The development of hypertrophic cartilage ends with the onset of endochondral bone formation which has inferior mechanical properties. In this review, current strategies for inhibition of chondrogenic hypertrophy are comprehensively summarized; the impact of cell source options is discussed; and potential mechanisms underlying these strategies are also categorized. This paper aims to provide guidelines for the prevention of hypertrophy in the regeneration of cartilage tissue. This knowledge may also facilitate the retardation of osteophytes in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Song Chen
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- Department of Joint Surgery, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, China
| | - Peiliang Fu
- Department of Joint Surgery, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, China
| | - Ruijun Cong
- Department of Orthopaedics, The 10th People's Hospital of Shanghai, Affiliated with Tongji University, Shanghai 200072, China
| | - HaiShan Wu
- Department of Joint Surgery, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV 26506, USA
- Corresponding author. Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, PO Box 9196, One Medical Center Drive, Morgantown, WV 26506-9196, USA. Tel.: +1 304 293 1072; fax: +1 304 293 7070.
| |
Collapse
|
29
|
Maher S, Kolieb E, Sabik NA, Abd-Elhalim D, El-Serafi AT, El-Wazir Y. Comparison of the osteogenic differentiation potential of mesenchymal cells isolated from human bone marrow, umbilical cord blood and placenta derived stem cells. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2015. [DOI: 10.1016/j.bjbas.2015.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
30
|
Teklemariam T, Purandare B, Zhao L, Hantash BM. Inhibition of DNA methylation enhances HLA-G expression in human mesenchymal stem cells. Biochem Biophys Res Commun 2014; 452:753-9. [PMID: 25204503 DOI: 10.1016/j.bbrc.2014.08.152] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs) are immunosuppressive multipotent cells under investigation for potential therapeutic applications in regenerative medicine and prevention of graft-versus-host disease. Human leukocyte antigen (HLA)-G contributes to the immunomodulatory properties of MSCs. HLA-G expression in MSCs is very low and diminishes during in vitro expansion. Epigenetic regulation activates HLA-G expression in some cancer cell lines but not in MSCs. In the present study, adipose- and bone marrow-derived MSCs were exposed to the DNA demethylating agent 5-aza-2-deoxycytidine (5-aza-dC) and histone deacetylase inhibitor valproic acid (VPA) and HLA-G mRNA levels assessed using semi-quantitative reverse-transcription PCR. Exposure to 5-aza-dC resulted in HLA-G1 and -G3 upregulation in both early and late passage MSCs. VPA treatment did not induce HLA-G expression in both bone marrow and adipose derived MSCs. Our results provide the first evidence that HLA-G3 could be expressed in MSCs and that methylation-mediated repression is partly responsible for the observed low levels of HLA-G expression in MSCs. Our findings provide insight that treatment of MSCs with specific epigenetic regulatory modulators may improve their immunoregulatory capability for therapeutic applications.
Collapse
Affiliation(s)
| | | | - Longmei Zhao
- Escape Therapeutics, Inc., San Jose, CA, United States
| | | |
Collapse
|
31
|
Li J, Ohliger J, Pei M. Significance of epigenetic landscape in cartilage regeneration from the cartilage development and pathology perspective. Stem Cells Dev 2014; 23:1178-94. [PMID: 24555773 DOI: 10.1089/scd.2014.0002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Regenerative therapies for cartilage defects have been greatly advanced by progress in both the stem cell biology and tissue engineering fields. Despite notable successes, significant barriers remain including shortage of autologous cell sources and generation of a stable chondrocyte phenotype using progenitor cells. Increasing demands for the treatment of degenerative diseases, such as osteoarthritis and rheumatoid arthritis, highlight the importance of epigenetic remodeling in cartilage regeneration. Epigenetic regulatory mechanisms, such as microRNAs, DNA methylation, and histone modifications, have been intensively studied due to their direct regulatory role on gene expression. However, a thorough understanding of the environmental factors that initiate these epigenetic events may provide greater insight into the prevention of degenerative diseases and improve the efficacy of treatments. In other words, if we could identify a specific factor from the environment and its downstream signaling events, then we could stop or retard degradation and enhance cartilage regeneration. A more operational definition of epigenetic remodeling has recently been proposed by categorizing the signals during the epigenetic process into epigenators, initiators, and maintainers. This review seeks to compile and reorganize the existing literature pertaining to epigenetic remodeling events placing emphasis on perceiving the landscape of epigenetic mechanisms during cartilage regeneration with the new operational definition, especially from the environmental factors' point of view. Progress in understanding epigenetic regulatory mechanisms could benefit cartilage regeneration and engineering on a larger scale and provide more promising therapeutic applications.
Collapse
Affiliation(s)
- Jingting Li
- 1 Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University , Morgantown, West Virginia
| | | | | |
Collapse
|
32
|
Zych J, Stimamiglio MA, Senegaglia AC, Brofman PRS, Dallagiovanna B, Goldenberg S, Correa A. The epigenetic modifiers 5-aza-2'-deoxycytidine and trichostatin A influence adipocyte differentiation in human mesenchymal stem cells. Braz J Med Biol Res 2014; 46:405-16. [PMID: 23797495 PMCID: PMC3854397 DOI: 10.1590/1414-431x20132893] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 03/11/2013] [Indexed: 01/10/2023] Open
Abstract
Epigenetic mechanisms such as DNA methylation and histone modification are
important in stem cell differentiation. Methylation is principally associated
with transcriptional repression, and histone acetylation is correlated with an
active chromatin state. We determined the effects of these epigenetic mechanisms
on adipocyte differentiation in mesenchymal stem cells (MSCs) derived from bone
marrow (BM-MSCs) and adipose tissue (ADSCs) using the chromatin-modifying agents
trichostatin A (TSA), a histone deacetylase inhibitor, and
5-aza-2′-deoxycytidine (5azadC), a demethylating agent. Subconfluent MSC
cultures were treated with 5, 50, or 500 nM TSA or with 1, 10, or
100 µM 5azadC for 2 days before the initiation of adipogenesis. The
differentiation was quantified and expression of the adipocyte genes PPARG and
FABP4 and of the anti-adipocyte gene GATA2 was evaluated. TSA decreased
adipogenesis, except in BM-MSCs treated with 5 nM TSA. Only treatment
with 500 nM TSA decreased cell proliferation. 5azadC treatment decreased
proliferation and adipocyte differentiation in all conditions evaluated,
resulting in the downregulation of PPARG and FABP4 and the upregulation of
GATA2. The response to treatment was stronger in ADSCs than in BM-MSCs,
suggesting that epigenetic memories may differ between cells of different
origins. As epigenetic signatures affect differentiation, it should be possible
to direct the use of MSCs in cell therapies to improve process efficiency by
considering the various sources available.
Collapse
Affiliation(s)
- J Zych
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, FIOCRUZ, Curitiba, PR, Brasil.
| | | | | | | | | | | | | |
Collapse
|
33
|
Vrtačnik P, Marc J, Ostanek B. Epigenetic mechanisms in bone. Clin Chem Lab Med 2014; 52:589-608. [DOI: 10.1515/cclm-2013-0770] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 11/19/2013] [Indexed: 12/11/2022]
|
34
|
de Andrés MC, Kingham E, Imagawa K, Gonzalez A, Roach HI, Wilson DI, Oreffo ROC. Epigenetic regulation during fetal femur development: DNA methylation matters. PLoS One 2013; 8:e54957. [PMID: 23383012 PMCID: PMC3557259 DOI: 10.1371/journal.pone.0054957] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 12/18/2012] [Indexed: 01/09/2023] Open
Abstract
Epigenetic modifications are heritable changes in gene expression without changes in DNA sequence. DNA methylation has been implicated in the control of several cellular processes including differentiation, gene regulation, development, genomic imprinting and X-chromosome inactivation. Methylated cytosine residues at CpG dinucleotides are commonly associated with gene repression; conversely, strategic loss of methylation during development could lead to activation of lineage-specific genes. Evidence is emerging that bone development and growth are programmed; although, interestingly, bone is constantly remodelled throughout life. Using human embryonic stem cells, human fetal bone cells (HFBCs), adult chondrocytes and STRO-1+ marrow stromal cells from human bone marrow, we have examined a spectrum of developmental stages of femur development and the role of DNA methylation therein. Using pyrosequencing methodology we analysed the status of methylation of genes implicated in bone biology; furthermore, we correlated these methylation levels with gene expression levels using qRT-PCR and protein distribution during fetal development evaluated using immunohistochemistry. We found that during fetal femur development DNA methylation inversely correlates with expression of genes including iNOS (NOS2) and COL9A1, but not catabolic genes including MMP13 and IL1B. Furthermore, significant demethylation was evident in the osteocalcin promoter between the fetal and adult developmental stages. Increased TET1 expression and decreased expression of DNA (cytosine-5-)-methyltransferase 1 (DNMT1) in adult chondrocytes compared to HFBCs could contribute to the loss of methylation observed during fetal development. HFBC multipotency confirms these cells to be an ideal developmental system for investigation of DNA methylation regulation. In conclusion, these findings demonstrate the role of epigenetic regulation, specifically DNA methylation, in bone development, informing and opening new possibilities in development of strategies for bone repair/tissue engineering.
Collapse
Affiliation(s)
- María C. de Andrés
- Bone and Joint Research Group, University of Southampton, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, United Kingdom
- Instituto de Investigación Sanitaria-Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Emmajayne Kingham
- Bone and Joint Research Group, University of Southampton, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, United Kingdom
| | - Kei Imagawa
- Bone and Joint Research Group, University of Southampton, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, United Kingdom
- Tohoku University School of Medicine, Sendai, Japan
| | - Antonio Gonzalez
- Instituto de Investigación Sanitaria-Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Helmtrud I. Roach
- Bone and Joint Research Group, University of Southampton, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, United Kingdom
| | - David I. Wilson
- Centre for Human Development, Stem Cells and Regeneration Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, United Kingdom
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, University of Southampton, Southampton, United Kingdom
- Centre for Human Development, Stem Cells and Regeneration Human Development and Health, Institute of Developmental Sciences, University of Southampton, Southampton, United Kingdom
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
- * E-mail:
| |
Collapse
|
35
|
Delgado-Calle J, Fernández AF, Sainz J, Zarrabeitia MT, Sañudo C, García-Renedo R, Pérez-Núñez MI, García-Ibarbia C, Fraga MF, Riancho JA. Genome-wide profiling of bone reveals differentially methylated regions in osteoporosis and osteoarthritis. ACTA ACUST UNITED AC 2012; 65:197-205. [DOI: 10.1002/art.37753] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 10/09/2012] [Indexed: 12/20/2022]
|
36
|
Delgado-Calle J, Sañudo C, Bolado A, Fernández AF, Arozamena J, Pascual-Carra MA, Rodriguez-Rey JC, Fraga MF, Bonewald L, Riancho JA. DNA methylation contributes to the regulation of sclerostin expression in human osteocytes. J Bone Miner Res 2012; 27:926-37. [PMID: 22162201 DOI: 10.1002/jbmr.1491] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Sclerostin, encoded by the SOST gene, is a potent inhibitor of bone formation, produced by osteocytes, not by osteoblasts, but little is known about the molecular mechanisms controlling its expression. We aimed to test the hypothesis that epigenetic mechanisms, specifically DNA methylation, modulate SOST expression. We found two CpG-rich regions in SOST: region 1, located in the proximal promoter, and region 2, around exon 1. qMSP and pyrosequencing analysis of DNA methylation showed that region 2 was largely methylated in all samples analyzed. In contrast, marked differences were observed in region 1. Whereas the CpG-rich region 1 was hypermethylated in osteoblasts, this region was largely hypomethylated in microdissected human osteocytes. Bone lining cells showed a methylation profile between primary osteoblasts and osteocytes. Whereas SOST expression was detected at very low level or not at all by RT-qPCR in several human osteoblastic and nonosteoblastic cell lines, and human primary osteoblasts under basal conditions, it was dramatically upregulated (up to 1300-fold) by the demethylating agent AzadC. Experiments using reporter vectors demonstrated the functional importance of the region -581/+30 of the SOST gene, which contains the CpG-rich region 1. In vitro methylation of this CpG-island impaired nuclear protein binding and led to a 75 ± 12% inhibition of promoter activity. In addition, BMP-2-induced expression of SOST was markedly enhanced in cells demethylated by AzadC. Overall, these results strongly suggest that DNA methylation is involved in the regulation of SOST expression during osteoblast-osteocyte transition, presumably by preventing the binding of transcription factors to the proximal promoter. To our knowledge, our data provide first ever evidence of the involvement of DNA methylation in the regulation of SOST expression and may help to establish convenient experimental models for further studies of human sclerostin.
Collapse
Affiliation(s)
- Jesús Delgado-Calle
- Department of Internal Medicine, Hospital U.M. Valdecilla-IFIMAV, University of Cantabria, RETICEF, Santander, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Owing to their potential for differentiation into multiple cell types, multipotent stem cells extracted from many adult tissues are an attractive stem cell resource for the replacement of damaged tissues in regenerative medicine. The requirements for cellular differentiation of an adult stem cell are a loss of proliferation potential and a gain of cell-type identity. These processes could be restricted by epigenetic modifications that prevent the risks of lineage-unrelated gene expression or the undifferentiated features of stem cells in adult somatic cells. In this review, we focus on the role of DNA methylation in controlling the transcriptional activity of genes important for self-renewal, the dynamism of CpG methylation of tissue-specific genes during several differentiation programs, and whether the multilineage potential of adult stem cells could be imposed early in the original precursor stem cells through CpG methylation. Additionally, we draw attention to the role of DNA methylation in adult stem cell differentiation by reviewing the reports on spontaneous differentiation after treatment with demethylating agents and by considering the evidence provided by reprogramming of somatic cells into undifferentiated cells (that is, somatic nuclear transfer or generation of induced pluripotent cells). It is clear from the evidence that DNA methylation is necessary for controlling stem cell proliferation and differentiation, but their exact contribution in each lineage program is still unclear. As a consequence, in a clinical setting, caution should be exerted before employing adult stem cells or their derivatives in regenerative medicine and appropriate tests should be applied to ensure the integrity of the genome and epigenome.
Collapse
Affiliation(s)
- María Berdasco
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, 08908, L'Hospitalet de Llobregat, Av, Gran Via 199-203, Barcelona, Catalonia, Spain
| | | |
Collapse
|