1
|
Gerini G, Traversa A, Cece F, Cassandri M, Pontecorvi P, Camero S, Nannini G, Romano E, Marampon F, Venneri MA, Ceccarelli S, Angeloni A, Amedei A, Marchese C, Megiorni F. Deciphering the Transcriptional Metabolic Profile of Adipose-Derived Stem Cells During Osteogenic Differentiation and Epigenetic Drug Treatment. Cells 2025; 14:135. [PMID: 39851564 PMCID: PMC11763738 DOI: 10.3390/cells14020135] [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] [Academic Contribution Register] [Received: 12/26/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/26/2025] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) are commonly employed in clinical treatment for various diseases due to their ability to differentiate into multi-lineage and anti-inflammatory/immunomodulatory properties. Preclinical studies support their use for bone regeneration, healing, and the improvement of functional outcomes. However, a deeper understanding of the molecular mechanisms underlying ASC biology is crucial to identifying key regulatory pathways that influence differentiation and enhance regenerative potential. In this study, we employed the NanoString nCounter technology, an advanced multiplexed digital counting method of RNA molecules, to comprehensively characterize differentially expressed transcripts involved in metabolic pathways at distinct time points in osteogenically differentiating ASCs treated with or without the pan-DNMT inhibitor RG108. In silico annotation and gene ontology analysis highlighted the activation of ethanol oxidation, ROS regulation, retinoic acid metabolism, and steroid hormone metabolism, as well as in the metabolism of lipids, amino acids, and nucleotides, and pinpointed potential new osteogenic drivers like AOX1 and ADH1A. RG108-treated cells, in addition to the upregulation of the osteogenesis-related markers RUNX2 and ALPL, showed statistically significant alterations in genes implicated in transcriptional control (MYCN, MYB, TP63, and IRF1), ethanol oxidation (ADH1C, ADH4, ADH6, and ADH7), and glucose metabolism (SLC2A3). These findings highlight the complex interplay of the metabolic, structural, and signaling pathways that orchestrate osteogenic differentiation. Furthermore, this study underscores the potential of epigenetic drugs like RG108 to enhance ASC properties, paving the way for more effective and personalized cell-based therapies for bone regeneration.
Collapse
Affiliation(s)
- Giulia Gerini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Alice Traversa
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy; (A.T.); (S.C.)
| | - Fabrizio Cece
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Matteo Cassandri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Simona Camero
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy; (A.T.); (S.C.)
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (G.N.); (A.A.)
| | - Enrico Romano
- Department of Sense Organs, Sapienza University of Rome, 00161 Rome, Italy;
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (G.N.); (A.A.)
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| |
Collapse
|
2
|
Roato I, Visca M, Mussano F. Suppressing the Aging Phenotype of Mesenchymal Stromal Cells: Are We Ready for Clinical Translation? Biomedicines 2024; 12:2811. [PMID: 39767719 PMCID: PMC11673080 DOI: 10.3390/biomedicines12122811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/02/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are involved in the maintenance and regeneration of a large variety of tissues due to their stemness and multi-lineage differentiation capability. Harnessing these advantageous features, a flurry of clinical trials have focused on MSCs to treat different pathologies, but only few protocols have received regulatory approval so far. Among the various causes hindering MSCs' efficacy is the emergence of cellular senescence, which has been correlated with specific characteristics, such as morphological and epigenetic alterations, DNA damage, ROS production, mitochondrial dysfunction, telomere shortening, non-coding RNAs, loss of proteostasis, and a peculiar senescence-associated secretory phenotype. Several strategies have been investigated for delaying or even hopefully reverting the onset of senescence, as assessed by the senescent phenotype of MSCs. Here, the authors reviewed the most updated literature on the potential causes of senescence, with a particular emphasis on the current and future therapeutic approaches aimed at reverting senescence and/or extending the functional lifespan of stem cells.
Collapse
Affiliation(s)
- Ilaria Roato
- Department of Surgical Sciences, CIR-Dental School, University of Turin, 10126 Turin, Italy; (M.V.); (F.M.)
| | | | | |
Collapse
|
3
|
Asgharian R, Hashemi A, Javeri A, Fakhr Taha M. DNA Methyltransferase Inhibition by RG108 Improves Stemness and Multipotential Differentiation of Human Adipose Tissue-derived Stem Cells. IRANIAN JOURNAL OF BIOTECHNOLOGY 2024; 22:e3863. [PMID: 39220336 PMCID: PMC11364923 DOI: 10.30498/ijb.2024.435096.3863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 01/13/2024] [Accepted: 03/09/2024] [Indexed: 09/04/2024]
Abstract
Background DNA methylation plays important roles in regulating various biological processes, including self-renewal, differentiation and regenerative capacity of stem cells. Previous studies have demonstrated that lineage-specific differentiation of mesenchymal stem cells can be promoted using nontoxic chromatin-modifying drugs. Objectives Here we evaluated the impact of RG108, a known DNA methyltransferase inhibitor, on the expression of pluripotency genes in human adipose tissue-derived stem cells (hADSCs) and their proliferation and differentiation. Materials and Methods Human ADSCs were isolated by collagenase treatment and characterized. Then, ADSCs were treated with 5 µM RG108 for four days. The control and RG108-treated cells were analyzed for the cell cycle progression, apoptosis and the expression of pluripotency genes. Also, ADSCs were cultured in adipogenic and osteogenic differentiation media for three weeks and were assessed by Oil Red O and Alizarin Red S staining and qPCR analysis. Results We showed that RG108 treatment increased proliferation of hADSCs and upregulated the expression of pluripotency-related genes. Additionally, RG108 had a positive impact on the differentiation capability of ADSCs. This was evident through elevated levels of Oil Red O staining in the RG108 treatment group. Also, qPCR analysis showed the upregulation of some adipogenic and osteogenic markers by RG108. Conclusion These findings indicate that pretreatment with RG108 improves the differentiation potential of ADSCs, probably making these cells more beneficial for cell therapy applications.
Collapse
Affiliation(s)
- Razieh Asgharian
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Biology, Islamic Azad University, Gorgan Branch, Gorgan, Iran
| | - Afrooz Hashemi
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Biology, Islamic Azad University, Damghan Branch, Damghan, Iran
| | - Arash Javeri
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Masoumeh Fakhr Taha
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
4
|
Pereira B, Correia FP, Alves IA, Costa M, Gameiro M, Martins AP, Saraiva JA. Epigenetic reprogramming as a key to reverse ageing and increase longevity. Ageing Res Rev 2024; 95:102204. [PMID: 38272265 DOI: 10.1016/j.arr.2024.102204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/09/2023] [Revised: 12/18/2023] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
The pursuit for the fountain of youth has long been a fascination amongst scientists and humanity. Ageing is broadly characterized by a cellular decline with increased susceptibility to age-related diseases, being intimately associated with epigenetic modifications. Recently, reprogramming-induced rejuvenation strategies have begun to greatly alter longevity research not only to tackle age-related defects but also to possibly reverse the cellular ageing process. Hence, in this review, we highlight the major epigenetic changes during ageing and the state-of-art of the current emerging epigenetic reprogramming strategies leveraging on transcription factors. Notably, partial reprogramming enables the resetting of the ageing clock without erasing cellular identity. Promising chemical-based rejuvenation strategies harnessing small molecules, including DNA methyltransferase and histone deacetylase inhibitors are also discussed. Moreover, in parallel to longevity interventions, the foundations of epigenetic clocks for accurate ageing assessment and evaluation of reprogramming approaches are briefly presented. Going further, with such scientific breakthroughs, we are witnessing a rise in the longevity biotech industry aiming to extend the health span and ideally achieve human rejuvenation one day. In this context, we overview the main scenarios proposed for the future of the socio-economic and ethical challenges associated with such an emerging field. Ultimately, this review aims to inspire future research on interventions that promote healthy ageing for all.
Collapse
Affiliation(s)
- Beatriz Pereira
- Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | | | - Inês A Alves
- Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Margarida Costa
- Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Mariana Gameiro
- Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Ana P Martins
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Jorge A Saraiva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
5
|
Roato I, Baima G, Orrico C, Mosca Balma A, Alotto D, Romano F, Ferracini R, Aimetti M, Mussano F. Senescent Markers Expressed by Periodontal Ligament-Derived Stem Cells (PDLSCs) Harvested from Patients with Periodontitis Can Be Rejuvenated by RG108. Biomedicines 2023; 11:2535. [PMID: 37760976 PMCID: PMC10526252 DOI: 10.3390/biomedicines11092535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/27/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Periodontal ligament (PDL) has become an elective source of mesenchymal stem cells (PDLSCs) in dentistry. This research aimed to compare healthy PDLSCs (hPDLSCs) and periodontitis PDLSCs (pPDLSCs) to ascertain any possible functional differences owing to their milieux of origin. Cells were tested in terms of colony-forming unit efficiency; multi differentiating capacity; immunophenotype, stemness, and senescent state were studied by flow cytometry, immunofluorescence, and β-galactosidase staining; gene expression using RT-PCR. Both hPDLSCs and pPDLSCs were comparable in terms of their immunophenotype and multilineage differentiation capabilities, but pPDLSCs showed a senescent phenotype more frequently. Thus, a selective small molecule inhibitor of DNA methyltransferase (DNMT), RG108, known for its effect on senescence, was used to possibly reverse this phenotype. RG108 did not affect the proliferation and apoptosis of PDLSCs, and it showed little effect on hPDLSCs, while a significant reduction of both p16 and p21 was detected along with an increase of SOX2 and OCT4 in pPDLSCs after treatment at 100 μM RG108. Moreover, the subset of PDLSCs co-expressing OCT4 and p21 decreased, and adipogenic potential increased in pPDLSCs after treatment. pPDLSCs displayed a senescent phenotype that could be reversed, opening new perspectives for the treatment of periodontitis.
Collapse
Affiliation(s)
- Ilaria Roato
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy; (I.R.); (A.M.B.); (F.R.); (M.A.); (F.M.)
| | - Giacomo Baima
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy; (I.R.); (A.M.B.); (F.R.); (M.A.); (F.M.)
- Politecnico di Torino, 10129 Turin, Italy
| | - Clarissa Orrico
- Fondazione Ricerca Molinette-Onlus, A.O.U. Città della Salute e della Scienza, 10129 Turin, Italy;
| | - Alessandro Mosca Balma
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy; (I.R.); (A.M.B.); (F.R.); (M.A.); (F.M.)
| | - Daniela Alotto
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Città della Salute e Della Scienza, 10126 Turin, Italy;
| | - Federica Romano
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy; (I.R.); (A.M.B.); (F.R.); (M.A.); (F.M.)
| | - Riccardo Ferracini
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, 16132 Genoa, Italy;
| | - Mario Aimetti
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy; (I.R.); (A.M.B.); (F.R.); (M.A.); (F.M.)
| | - Federico Mussano
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy; (I.R.); (A.M.B.); (F.R.); (M.A.); (F.M.)
| |
Collapse
|
6
|
Cheng M, Yuan W, Moshaverinia A, Yu B. Rejuvenation of Mesenchymal Stem Cells to Ameliorate Skeletal Aging. Cells 2023; 12:998. [PMID: 37048071 PMCID: PMC10093211 DOI: 10.3390/cells12070998] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/15/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
Advanced age is a shared risk factor for many chronic and debilitating skeletal diseases including osteoporosis and periodontitis. Mesenchymal stem cells develop various aging phenotypes including the onset of senescence, intrinsic loss of regenerative potential and exacerbation of inflammatory microenvironment via secretory factors. This review elaborates on the emerging concepts on the molecular and epigenetic mechanisms of MSC senescence, such as the accumulation of oxidative stress, DNA damage and mitochondrial dysfunction. Senescent MSCs aggravate local inflammation, disrupt bone remodeling and bone-fat balance, thereby contributing to the progression of age-related bone diseases. Various rejuvenation strategies to target senescent MSCs could present a promising paradigm to restore skeletal aging.
Collapse
Affiliation(s)
- Mingjia Cheng
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Weihao Yuan
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Alireza Moshaverinia
- Section of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Bo Yu
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Wang X, Yu F, Ye L. Epigenetic control of mesenchymal stem cells orchestrates bone regeneration. Front Endocrinol (Lausanne) 2023; 14:1126787. [PMID: 36950693 PMCID: PMC10025550 DOI: 10.3389/fendo.2023.1126787] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/18/2022] [Accepted: 02/17/2023] [Indexed: 03/08/2023] Open
Abstract
Recent studies have revealed the vital role of MSCs in bone regeneration. In both self-healing bone regeneration processes and biomaterial-induced healing of bone defects beyond the critical size, MSCs show several functions, including osteogenic differentiation and thus providing seed cells. However, adverse factors such as drug intake and body senescence can significantly affect the functions of MSCs in bone regeneration. Currently, several modalities have been developed to regulate MSCs' phenotype and promote the bone regeneration process. Epigenetic regulation has received much attention because of its heritable nature. Indeed, epigenetic regulation of MSCs is involved in the pathogenesis of a variety of disorders of bone metabolism. Moreover, studies using epigenetic regulation to treat diseases are also being reported. At the same time, the effects of epigenetic regulation on MSCs are yet to be fully understood. This review focuses on recent advances in the effects of epigenetic regulation on osteogenic differentiation, proliferation, and cellular senescence in MSCs. We intend to illustrate how epigenetic regulation of MSCs orchestrates the process of bone regeneration.
Collapse
Affiliation(s)
- Xiaofeng Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Fanyuan Yu, ; Ling Ye,
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Fanyuan Yu, ; Ling Ye,
| |
Collapse
|
8
|
Roszkowski L, Jaszczyk B, Plebańczyk M, Ciechomska M. S100A8 and S100A12 Proteins as Biomarkers of High Disease Activity in Patients with Rheumatoid Arthritis That Can Be Regulated by Epigenetic Drugs. Int J Mol Sci 2022; 24:ijms24010710. [PMID: 36614150 PMCID: PMC9820830 DOI: 10.3390/ijms24010710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune chronic inflammatory disease that is still not well understood in terms of its pathogenesis and presents diagnostic and therapeutic challenges. Monocytes are key players in initiating and maintaining inflammation through the production of pro-inflammatory cytokines and S100 proteins in RA. This study aimed to test a specific DNA methylation inhibitor (RG108) and activator (budesonide) in the regulation of pro-inflammatory mediators-especially the S100 proteins. We also searched for new biomarkers of high disease activity in RA patients. RNA sequencing analysis of healthy controls (HCs) and RA monocytes was performed. Genes such as the S100 family, TNF, and IL-8 were validated by qRT-PCR following DNA-methylation-targeted drug treatment in a monocytic THP-1 cell line. The concentrations of the S100A8, S100A11, and S100A12 proteins in the sera and synovial fluids of RA patients were tested and correlated with clinical parameters. We demonstrated that RA monocytes had significantly increased levels of S100A8, S100A9, S100A11, S100A12, MYD88, JAK3, and IQGAP1 and decreased levels of IL10RA and TGIF1 transcripts. In addition, stimulation of THP-1 cells with budesonide statistically reduced the expression of the S100 family, IL-8, and TNF genes. In contrast, THP-1 cells treated with RG108 had increased levels of the S100 family and TNF genes. We also revealed a significant upregulation of S100A8, S100A11, and S100A12 in RA patients, especially in early RA compared to HC sera. In addition, protein levels of S100A8, S100A11, and S100A12 in RA synovial fluids compared to HC sera were significantly increased. Overall, our data suggest that the S100A8 and S100A12 proteins are strongly elevated during ongoing inflammation, so they could be used as a better biomarker of disease activity than CRP. Interestingly, epigenetic drugs can regulate these S100 proteins, suggesting their potential use in targeting RA inflammation.
Collapse
Affiliation(s)
- Leszek Roszkowski
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation (NIGRiR), 02-637 Warsaw, Poland
| | - Bożena Jaszczyk
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology and Rehabilitation (NIGRiR), 02-637 Warsaw, Poland
| | - Magdalena Plebańczyk
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation (NIGRiR), 02-637 Warsaw, Poland
| | - Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation (NIGRiR), 02-637 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-670-95-63
| |
Collapse
|
9
|
Yu X, Li M, Guo C, Wu Y, Zhao L, Shi Q, Song J, Song B. Therapeutic Targeting of Cancer: Epigenetic Homeostasis. Front Oncol 2021; 11:747022. [PMID: 34765551 PMCID: PMC8576334 DOI: 10.3389/fonc.2021.747022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/25/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
A large number of studies have revealed that epigenetics plays an important role in cancer development. However, the currently-developed epigenetic drugs cannot achieve a stable curative effect. Thus, it may be necessary to redefine the role of epigenetics in cancer development. It has been shown that embryonic development and tumor development share significant similarities in terms of biological behavior and molecular expression patterns, and epigenetics may be the link between them. Cell differentiation is likely a manifestation of epigenetic homeostasis at the cellular level. In this article, we introduced the importance of epigenetic homeostasis in cancer development and analyzed the shortcomings of current epigenetic treatment regimens. Understanding the dynamic process of epigenetic homeostasis in organ development can help us characterize cancer according to its differentiation stages, explore new targets for cancer treatment, and improve the clinical prognosis of patients with cancer.
Collapse
Affiliation(s)
- Xiaoyuan Yu
- Department of Oncology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Menglu Li
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunyan Guo
- Department of Oncology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuesheng Wu
- Department of Oncology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Li Zhao
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Qinying Shi
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Jianbo Song
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Bin Song
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|