1
|
González-González A, Álvarez-Iglesias I, García-Sánchez D, Dotta M, Reyes R, Alfonso-Fernández A, Bolado-Carrancio A, Díaz-Rodríguez P, Pérez-Núñez MI, Rodríguez-Rey JC, Delgado-Calle J, Pérez-Campo FM. Paracrine activity of Smurf1-silenced mesenchymal stem cells enhances bone regeneration and reduces bone loss in postmenopausal osteoporosis. Stem Cell Res Ther 2025; 16:50. [PMID: 39920824 PMCID: PMC11806587 DOI: 10.1186/s13287-025-04165-0] [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: 10/25/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Osteoporosis (OP), characterized by reduced bone mass and mineral density, is a global metabolic disorder that severely impacts the quality of life in affected individuals. Although current pharmacological treatments are effective, their long-term use is often associated with adverse effects, highlighting the need for safer, more sustainable therapeutic strategies. This study investigates the pro-osteogenic and anti-resorptive potential of the secretome from Smurf1-silenced mesenchymal stem cells (MSCs) as a promising cell-free therapy for bone regeneration. METHODS Conditioned media (CM) from Smurf1-silenced rat (rCM-Smur1) and human MSCs (hCM-Smurf1) was collected and analyzed. Pro-osteogenic potential was assessed by measuring in vitro mineralization in human and rat MSCs cultures. In vivo, studies were conducted using a rat ectopic bone formation model and a post-menopausal osteoporotic mouse model. Additionally, primary human osteoporotic MSCs were preconditioned with hCM-Smurf1, and their osteogenic capacity was compared to that induced by BMP2 treatment. Ex vivo, human bone explants were treated with hCM-Smurf1 to assess anti-resorptive effects. Proteomic analysis of the soluble and vesicular CM fractions identified key proteins involved in bone regeneration. RESULTS CM from Smurf1-silenced MSCs significantly enhanced mineralization in vitro and bone formation in vivo. Preconditioning human osteoporotic MSCs with hCM-Smurf1 significantly increases in vitro mineralization, with levels comparable to those achieved with BMP2 treatment. Additionally, in ex vivo human bone cultures, treatment with hCM-Smurf1 significantly reduced RANKL expression without affecting OPG levels, indicating an anti-resorptive effect. In vivo, CM from Smurf1-silenced MSCs significantly increased bone formation in a rat ectopic model, and its local administration reduced trabecular bone loss by 50% in a post-menopausal osteoporotic mouse model after a single administration within just four weeks. Proteomic analysis revealed both soluble and vesicular fractions of hCM-Smurf1 were enriched with proteins essential for ossification and extracellular matrix organization, enhancing osteogenic differentiation. CONCLUSIONS The Smurf1-silenced MSCs' secretome shows potent osteogenic and anti-resorptive effects, significantly enhancing bone formation and reducing bone loss. This study provides compelling evidence for the therapeutic potential of Smurf1-silenced MSC-derived secretome as a non-toxic and targeted treatment for osteoporosis. These findings warrant further in vivo studies and clinical trials to validate its therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Alberto González-González
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012, Santander, Spain
| | - Itziar Álvarez-Iglesias
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012, Santander, Spain
| | - Daniel García-Sánchez
- Department of Physiology and Cell Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Monica Dotta
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012, Santander, Spain
| | - Ricardo Reyes
- Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, 38206, La Laguna, Spain
| | - Ana Alfonso-Fernández
- Department of Traumatology, Hospital Universitario Marqués de Valdecilla, University of Cantabria, 39008, Santander, Spain
| | - Alfonso Bolado-Carrancio
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Patricia Díaz-Rodríguez
- I+D Farma Group (GI-1645), Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - María Isabel Pérez-Núñez
- Department of Traumatology, Hospital Universitario Marqués de Valdecilla, University of Cantabria, 39008, Santander, Spain
| | - José Carlos Rodríguez-Rey
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012, Santander, Spain
| | - Jesús Delgado-Calle
- Department of Physiology and Cell Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Flor M Pérez-Campo
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012, Santander, Spain.
| |
Collapse
|
2
|
Premadasa LS, McDew-White M, Romero L, Gondo B, Drawec JA, Ling B, Okeoma CM, Mohan M. Epigenetic modulation of NLRP6 inflammasome sensor as a therapeutic modality to reduce necroptosis-driven gastrointestinal mucosal dysfunction in HIV/SIV infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623322. [PMID: 39605466 PMCID: PMC11601347 DOI: 10.1101/2024.11.13.623322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The epigenetic mechanisms driving persistent gastrointestinal mucosal dysfunction in HIV/SIV infection is an understudied topic. Using reduced-representation bisulfite sequencing, we identified HIV/SIV infection in combination anti-retroviral therapy (cART)-naive rhesus macaques (RMs) to induce marked hypomethylation throughout promoter-associated CpG islands (paCGIs) in genes related to inflammatory response ( NLRP6, cGAS ), cellular adhesion and proliferation in colonic epithelial cells (CEs). Moreover, low-dose delta-9-tetrahydrocannabinol (THC) administration reduced NLRP6 protein expression in CE by hypermethylating the NLRP6 paCGI and blocked polyI:C induced NLRP6 upregulation in vitro. In cART suppressed SIV-infected RMs, NLRP6 protein upregulation associated with significantly increased expression of necroptosis-driving proteins; phosphorylated-RIPK3(Ser199), phosphorylated-MLKL(Thr357/Ser358), and HMGB1. Most strikingly, supplementing cART with THC effectively reduced NLRP6 and necroptosis-driving protein expression to pre-infection levels. These findings for the first time demonstrate that NLRP6 upregulation and ensuing activation of necroptosis promote HIV/SIV-induced gastrointestinal mucosal dysfunction and that epigenetic modulation using phytocannabinoids represents a feasible therapeutic modality for alleviating HIV/SIV-induced gastrointestinal inflammation and associated comorbidities.
Collapse
|
3
|
King JS, Wan M, Wagley Y, Stestiv M, Kalajzic I, Hankenson KD, Sanjay A. Signaling pathways associated with Lgr6 to regulate osteogenesis. Bone 2024; 187:117207. [PMID: 39033993 DOI: 10.1016/j.bone.2024.117207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Fracture management largely relies on the bone's inherent healing capabilities and, when necessary, surgical intervention. Currently, there are limited osteoinductive therapies to promote healing, making targeting skeletal stem/progenitor cells (SSPCs) a promising avenue for therapeutic development. A limiting factor for this approach is our incomplete understanding of the molecular mechanisms governing SSPCs' behavior. We have recently identified that the Leucine-rich repeat-containing G-protein coupled receptor 6 (Lgr6) is expressed in sub-populations of SSPCs, and is required for maintaining bone volume during adulthood and for proper fracture healing. Lgr family members (Lgr4-6) are markers of stem cell niches and play a role in tissue regeneration primarily by binding R-Spondin (Rspo1-4). This interaction promotes canonical Wnt (cWnt) signaling by stabilizing Frizzled receptors. Interestingly, our findings here indicate that Lgr6 may also influence cWnt-independent pathways. Remarkably, Lgr6 expression was enhanced during Bmp-mediated osteogenesis of both human and murine cells. Using biochemical approaches, RNA sequencing, and bioinformatic analysis of published single-cell data, we found that elements of BMP signaling, including its target gene, pSMAD, and gene ontology pathways, are downregulated in the absence of Lgr6. Our findings uncover a molecular interdependency between the Bmp pathway and Lgr6, offering new insights into osteogenesis and potential targets for enhancing fracture healing.
Collapse
Affiliation(s)
- Justin S King
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Yadav Wagley
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Marta Stestiv
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Ivo Kalajzic
- Center for Regenerative Medicine and Skeletal Development, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, The Musculoskeletal Research Institute, UCONN Health, Farmington, CT 06032, USA.
| |
Collapse
|
4
|
Lai Z, Shu Q, Song Y, Tang A, Tian J. Effect of DNA methylation on the osteogenic differentiation of mesenchymal stem cells: concise review. Front Genet 2024; 15:1429844. [PMID: 39015772 PMCID: PMC11250479 DOI: 10.3389/fgene.2024.1429844] [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: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have promising potential for bone tissue engineering in bone healing and regeneration. They are regarded as such due to their capacity for self-renewal, multiple differentiation, and their ability to modulate the immune response. However, changes in the molecular pathways and transcription factors of MSCs in osteogenesis can lead to bone defects and metabolic bone diseases. DNA methylation is an epigenetic process that plays an important role in the osteogenic differentiation of MSCs by regulating gene expression. An increasing number of studies have demonstrated the significance of DNA methyltransferases (DNMTs), Ten-eleven translocation family proteins (TETs), and MSCs signaling pathways about osteogenic differentiation in MSCs. This review focuses on the progress of research in these areas.
Collapse
Affiliation(s)
- Zhihao Lai
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qing Shu
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yue Song
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Ao Tang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Jun Tian
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Choi J, Xu Z, Sun R. Variance-components tests for genetic association with multiple interval-censored outcomes. Stat Med 2024; 43:2560-2574. [PMID: 38636557 PMCID: PMC11116038 DOI: 10.1002/sim.10081] [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: 10/19/2022] [Revised: 02/18/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Massive genetic compendiums such as the UK Biobank have become an invaluable resource for identifying genetic variants that are associated with complex diseases. Due to the difficulties of massive data collection, a common practice of these compendiums is to collect interval-censored data. One challenge in analyzing such data is the lack of methodology available for genetic association studies with interval-censored data. Genetic effects are difficult to detect because of their rare and weak nature, and often the time-to-event outcomes are transformed to binary phenotypes for access to more powerful signal detection approaches. However transforming the data to binary outcomes can result in loss of valuable information. To alleviate such challenges, this work develops methodology to associate genetic variant sets with multiple interval-censored outcomes. Testing sets of variants such as genes or pathways is a common approach in genetic association settings to lower the multiple testing burden, aggregate small effects, and improve interpretations of results. Instead of performing inference with only a single outcome, utilizing multiple outcomes can increase statistical power by aggregating information across multiple correlated phenotypes. Simulations show that the proposed strategy can offer significant power gains over a single outcome approach. We apply the proposed test to the investigation that motivated this study, a search for the genes that perturb risks of bone fractures and falls in the UK Biobank.
Collapse
Affiliation(s)
- Jaihee Choi
- Department of Statistics, Rice University, Texas, USA
| | - Zhichao Xu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Texas, USA
| | - Ryan Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Texas, USA
| |
Collapse
|
6
|
Wang X, Ge Q, Zeng Q, Zou K, Bao Z, Ying J, Wu Z, Jin H, Chen J, Xu T. Dnmt3b ablation affects fracture repair process by regulating apoptosis. BMC Musculoskelet Disord 2024; 25:180. [PMID: 38413962 PMCID: PMC10900613 DOI: 10.1186/s12891-024-07283-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/14/2024] [Indexed: 02/29/2024] Open
Abstract
PURPOSE Previous studies have shown that DNA methyltransferase 3b (Dnmt3b) is the only Dnmt responsive to fracture repair and Dnmt3b ablation in Prx1-positive stem cells and chondrocyte cells both delayed fracture repair. Our study aims to explore the influence of Dnmt3b ablation in Gli1-positive stem cells in fracture healing mice and the underlying mechanism. METHODS We generated Gli1-CreERT2; Dnmt3bflox/flox (Dnmt3bGli1ER) mice to operated tibia fracture. Fracture callus tissues of Dnmt3bGli1ER mice and control mice were collected and analyzed by X-ray, micro-CT, biomechanical testing, histopathology and TUNEL assay. RESULTS The cartilaginous callus significantly decrease in ablation of Dnmt3b in Gli1-positive stem cells during fracture repair. The chondrogenic and osteogenic indicators (Sox9 and Runx2) in the fracture healing tissues in Dnmt3bGli1ER mice much less than control mice. Dnmt3bGli1ER mice led to delayed bone callus remodeling and decreased biomechanical properties of the newly formed bone during fracture repair. Both the expressions of Caspase-3 and Caspase-8 were upregulated in Dnmt3bGli1ER mice as well as the expressions of BCL-2. CONCLUSIONS Our study provides an evidence that Dnmt3b ablation Gli1-positive stem cells can affect fracture healing and lead to poor fracture healing by regulating apoptosis to decrease chondrocyte hypertrophic maturation.
Collapse
Affiliation(s)
- Xu Wang
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qinwen Ge
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qinghe Zeng
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Kaiao Zou
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhengsheng Bao
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jun Ying
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhen Wu
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Hongting Jin
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| | - Jiali Chen
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| | - Taotao Xu
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
7
|
Zhang P, Feng B, Dai G, Niu K, Zhang L. FOXC1 Promotes Osteoblastic Differentiation of Bone Marrow Mesenchymal Stem Cells via the Dnmt3b/CXCL12 Axis. Biochem Genet 2024; 62:176-192. [PMID: 37306827 DOI: 10.1007/s10528-023-10403-y] [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: 11/16/2022] [Accepted: 05/12/2023] [Indexed: 06/13/2023]
Abstract
Bone defects have remained a clinical problem in current orthopedics. Bone marrow mesenchymal stem cells (BM-MSCs) with multi-directional differentiation ability have become a research hotspot for repairing bone defects. In vitro and in vivo models were constructed, respectively. Alkaline phosphatase (ALP) staining and alizarin red staining were performed to detect osteogenic differentiation ability. Western blotting (WB) was used to detect the expression of osteogenic differentiation-related proteins. Serum inflammatory cytokine levels were detected by ELISA. Fracture recovery was evaluated by HE staining. The binding relationship between FOXC1 and Dnmt3b was verified by dual-luciferase reporter assay. The relationship between Dnmt3b and CXCL12 was explored by MSP and ChIP assays. FOXC1 overexpression promoted calcium nodule formation, upregulated osteogenic differentiation-related protein expression, promoted osteogenic differentiation, and decreased inflammatory factor levels in BM-MSCs, and promoted callus formation, upregulated osteogenic differentiation-related protein expression, and downregulated CXCL12 expression in the mouse model. Furthermore, FOXC1 targeted Dnmt3b, with Dnmt3b knockdown decreasing calcium nodule formation and downregulating osteogenic differentiation-related protein expression. Additionally, inhibiting Dnmt3b expression upregulated CXCL12 protein expression and inhibited CXCL12 methylation. Dnmt3b could be binded to CXCL12. CXCL12 overexpression attenuated the effects of FOXC1 overexpression and inhibited BM-MSCs osteogenic differentiation. This study confirmed that the FOXC1-mediated regulation of the Dnmt3b/CXCL12 axis had positive effects on the osteogenic differentiation of BM-MSCs.
Collapse
Affiliation(s)
- Peiguang Zhang
- Department of Orthopedics, The Third Affiliated Hospital, Inner Mongolia Medical University, No. 20 Shaoxian Road, Kundulun District, Baotou, 014010, Inner Mongolia, People's Republic of China
| | - Bo Feng
- Department of Orthopedics, The Third Affiliated Hospital, Inner Mongolia Medical University, No. 20 Shaoxian Road, Kundulun District, Baotou, 014010, Inner Mongolia, People's Republic of China
| | - Guangming Dai
- Department of Orthopedics, The Third Affiliated Hospital, Inner Mongolia Medical University, No. 20 Shaoxian Road, Kundulun District, Baotou, 014010, Inner Mongolia, People's Republic of China
| | - Kecheng Niu
- Department of Orthopedics, The Third Affiliated Hospital, Inner Mongolia Medical University, No. 20 Shaoxian Road, Kundulun District, Baotou, 014010, Inner Mongolia, People's Republic of China
| | - Lan Zhang
- Department of Orthopedics, The Third Affiliated Hospital, Inner Mongolia Medical University, No. 20 Shaoxian Road, Kundulun District, Baotou, 014010, Inner Mongolia, People's Republic of China.
| |
Collapse
|
8
|
Gu Y, Song Y, Pan Y, Liu J. The essential roles of m 6A modification in osteogenesis and common bone diseases. Genes Dis 2024; 11:335-345. [PMID: 37588215 PMCID: PMC10425797 DOI: 10.1016/j.gendis.2023.01.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/30/2023] [Indexed: 03/30/2023] Open
Abstract
N6-methyladenosine (m6A) is the most prevalent modification in the eukaryotic transcriptome and has a wide range of functions in coding and noncoding RNAs. It affects the fate of the modified RNA, including its stability, splicing, and translation, and plays an important role in post-transcriptional regulation. Bones play a key role in supporting and protecting muscles and other organs, facilitating the movement of the organism, ensuring blood production, etc. Bone diseases such as osteoarthritis, osteoporosis, and bone tumors are serious public health problems. The processes of bone development and osteogenic differentiation require the precise regulation of gene expression through epigenetic mechanisms including histone, DNA, and RNA modifications. As a reversible dynamic epigenetic mark, m6A modifications affect nearly every important biological process, cellular component, and molecular function, including skeletal development and homeostasis. In recent years, studies have shown that m6A modification is involved in osteogenesis and bone-related diseases. In this review, we summarized the proteins involved in RNA m6A modification and the latest progress in elucidating the regulatory role of m6A modification in bone formation and stem cell directional differentiation. We also discussed the pathological roles and potential molecular mechanisms of m6A modification in bone-related diseases like osteoporosis and osteosarcoma and suggested potential areas for new strategies that could be used to prevent or treat bone defects and bone diseases.
Collapse
Affiliation(s)
- Yuxi Gu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yidan Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yihua Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
9
|
Xiao D, Fang L, Liu Z, He Y, Ying J, Qin H, Lu A, Shi M, Li T, Zhang B, Guan J, Wang C, Abu-Amer Y, Shen J. DNA methylation-mediated Rbpjk suppression protects against fracture nonunion caused by systemic inflammation. J Clin Invest 2023; 134:e168558. [PMID: 38051594 PMCID: PMC10849763 DOI: 10.1172/jci168558] [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: 01/04/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023] Open
Abstract
Challenging skeletal repairs are frequently seen in patients experiencing systemic inflammation. To tackle the complexity and heterogeneity of the skeletal repair process, we performed single-cell RNA sequencing and revealed that progenitor cells were one of the major lineages responsive to elevated inflammation and this response adversely affected progenitor differentiation by upregulation of Rbpjk in fracture nonunion. We then validated the interplay between inflammation (via constitutive activation of Ikk2, Ikk2ca) and Rbpjk specifically in progenitors by using genetic animal models. Focusing on epigenetic regulation, we identified Rbpjk as a direct target of Dnmt3b. Mechanistically, inflammation decreased Dnmt3b expression in progenitor cells, consequently leading to Rbpjk upregulation via hypomethylation within its promoter region. We also showed that Dnmt3b loss-of-function mice phenotypically recapitulated the fracture repair defects observed in Ikk2ca-transgenic mice, whereas Dnmt3b-transgenic mice alleviated fracture repair defects induced by Ikk2ca. Moreover, Rbpjk ablation restored fracture repair in both Ikk2ca mice and Dnmt3b loss-of-function mice. Altogether, this work elucidates a common mechanism involving a NF-κB/Dnmt3b/Rbpjk axis within the context of inflamed bone regeneration. Building on this mechanistic insight, we applied local treatment with epigenetically modified progenitor cells in a previously established mouse model of inflammation-mediated fracture nonunion and showed a functional restoration of bone regeneration under inflammatory conditions through an increase in progenitor differentiation potential.
Collapse
Affiliation(s)
- Ding Xiao
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Liang Fang
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Zhongting Liu
- Department of Mechanical Engineering & Materials Sciences, School of Engineering and
| | - Yonghua He
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Jun Ying
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Haocheng Qin
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Aiwu Lu
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Meng Shi
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Tiandao Li
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA
| | - Bo Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA
| | - Jianjun Guan
- Department of Mechanical Engineering & Materials Sciences, School of Engineering and
| | - Cuicui Wang
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
- Shriners Hospital for Children, St. Louis, Missouri, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Liang X, Shi W, Zhang X, Pang R, Zhang K, Xu Q, Xu C, Wan X, Cui W, Li D, Jiang Z, Liu Z, Li H, Zhang H, Li Z. Causal association of epigenetic aging and osteoporosis: a bidirectional Mendelian randomization study. BMC Med Genomics 2023; 16:275. [PMID: 37919683 PMCID: PMC10623745 DOI: 10.1186/s12920-023-01708-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND The relationship between aging and osteoporosis is well established. However, the relationship between the body's physiological age, i.e. epigenetic age, and osteoporosis is not known. Our goal is to analyze the bidirectional causal relationship between epigenetic clocks and osteoporosis using a bidirectional Mendelian randomization study. METHODS We used SNPs closely associated with GrimAge, Hannum, PhenoAge, and HorvathAge in epigenetic age and SNPs closely associated with femoral neck bone mineral density, lumbar spine bone mineral density, and forearm bone mineral density as instrumental variables, respectively, using the inverse variance weighting method and several other MR methods to assess the bidirectional causal relationship between epigenetic age and osteoporosis. RESULT There was no evidence of a clear causal relationship of epigenetic age (GrimAge, Hannum, PhenoAge, and HorvathAge) on femoral neck bone mineral density, lumbar spine bone mineral density, and forearm bone mineral density. In reverse Mendelian randomization analysis showed a significant causal effect of lumbar spine bone mineral density on GrimAge: odds ratio (OR) = 0.692, 95% confidence interval (CI) = (0.538-0.890), p = 0.004. The results suggest that a decrease in lumbar spine bone mineral density promotes an acceleration of GrimAge. CONCLUSION There was no significant bidirectional causal relationship between epigenetic age and osteoporosis A decrease in lumbar spine bone density may lead to an acceleration of the epigenetic clock "GrimAge". Our study provides partial evidence for a bidirectional causal effect between epigenetic age and Osteoporosis.
Collapse
Affiliation(s)
- Xinyu Liang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Wei Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Xinglong Zhang
- Department of Orthopedics, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, People's Republic of China
| | - Ran Pang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Department of Orthopedics, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, People's Republic of China
| | - Kai Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Qian Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Chunlei Xu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Xin Wan
- Department of Orthopedics, Tian-Jin Union Medical Centre, Nankai University People's Hospital, Tianjin, China
| | - Wenhao Cui
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- R&D Center, Youjia (Hangzhou) Biomedical Technology Co., Ltd, Hangzhou, China
| | - Dong Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Zhaohui Jiang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Department of Orthopaedic, Wenzhou Central Hospital, Wenzhou, China
| | - Zhengxuan Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Hui Li
- Department of Orthopedics, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, People's Republic of China
| | - Huafeng Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| | - Zhijun Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
11
|
García-Sánchez D, González-González A, Álvarez-Iglesias I, del Dujo-Gutiérrez M, Bolado-Carrancio A, Certo M, Pérez-Núñez MI, Riancho JA, Rodríguez-Rey JC, Delgado-Calle J, Pérez-Campo FM. Engineering a Pro-Osteogenic Secretome through the Transient Silencing of the Gene Encoding Secreted Frizzled Related Protein 1. Int J Mol Sci 2023; 24:12399. [PMID: 37569774 PMCID: PMC10419110 DOI: 10.3390/ijms241512399] [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: 06/22/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
The evidence sustaining the regenerative properties of mesenchymal stem cells' (MSCs) secretome has prompted a paradigm change, where MSCs have shifted from being considered direct contributors to tissue regeneration toward being seen as cell factories for producing biotech medicines. We have previously designed a method to prime MSCs towards osteogenic differentiation by silencing the Wnt/β-Catenin inhibitor Sfpr1. This approach produces a significant increase in bone formation in osteoporotic mice. In this current work, we set to investigate the contribution of the secretome from the MSCs where Sfrp1 has been silenced, to the positive effect seen on bone regeneration in vivo. The conditioned media (CM) of the murine MSCs line C3H10T1/2, where Sfrp1 has been transiently silenced (CM-Sfrp1), was found to induce, in vitro, an increase in the osteogenic differentiation of this same cell line, as well as a decrease of the expression of the Wnt inhibitor Dkk1 in murine osteocytes ex vivo. A reduction in the RANKL/OPG ratio was also detected ex vivo, suggesting a negative effect of CM-Sfrp1 on osteoclastogenesis. Moreover, this CM significantly increases the mineralization of human primary MSCs isolated from osteoportotic patients in vitro. Proteomic analysis identified enrichment of proteins involved in osteogenesis within the soluble and vesicular fractions of this secretome. Altogether, we demonstrate the pro-osteogenic potential of the secretome of MSCs primmed in this fashion, suggesting that this is a valid approach to enhance the osteo-regenerative properties of MSCs' secretome.
Collapse
Affiliation(s)
- Daniel García-Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Alberto González-González
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Itzíar Álvarez-Iglesias
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Mónica del Dujo-Gutiérrez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Alfonso Bolado-Carrancio
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK;
| | - Matilde Certo
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - María Isabel Pérez-Núñez
- Department of Traumatology, Hospital Universitario Marqués de Valdecilla, University of Cantabria, 39008 Santander, Spain;
| | - José A. Riancho
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, CEBERER, 39012 Santander, Spain;
| | - José Carlos Rodríguez-Rey
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Jesús Delgado-Calle
- Department of Physiology and Cell Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Flor María Pérez-Campo
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| |
Collapse
|
12
|
la Torre A, Lo Vecchio F, Greco A. Epigenetic Mechanisms of Aging and Aging-Associated Diseases. Cells 2023; 12:cells12081163. [PMID: 37190071 DOI: 10.3390/cells12081163] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Aging is an inevitable outcome of life, characterized by a progressive decline in tissue and organ function. At a molecular level, it is marked by the gradual alterations of biomolecules. Indeed, important changes are observed on the DNA, as well as at a protein level, that are influenced by both genetic and environmental parameters. These molecular changes directly contribute to the development or progression of several human pathologies, including cancer, diabetes, osteoporosis, neurodegenerative disorders and others aging-related diseases. Additionally, they increase the risk of mortality. Therefore, deciphering the hallmarks of aging represents a possibility for identifying potential druggable targets to attenuate the aging process, and then the age-related comorbidities. Given the link between aging, genetic, and epigenetic alterations, and given the reversible nature of epigenetic mechanisms, the precisely understanding of these factors may provide a potential therapeutic approach for age-related decline and disease. In this review, we center on epigenetic regulatory mechanisms and their aging-associated changes, highlighting their inferences in age-associated diseases.
Collapse
Affiliation(s)
- Annamaria la Torre
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Filomena Lo Vecchio
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| | - Antonio Greco
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
| |
Collapse
|
13
|
Doherty L, Wan M, Peterson A, Youngstrom DW, King JS, Kalajzic I, Hankenson KD, Sanjay A. Wnt-associated adult stem cell marker Lgr6 is required for osteogenesis and fracture healing. Bone 2023; 169:116681. [PMID: 36708855 PMCID: PMC10015414 DOI: 10.1016/j.bone.2023.116681] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
Despite the remarkable regenerative capacity of skeletal tissues, nonunion of bone and failure of fractures to heal properly presents a significant clinical concern. Stem and progenitor cells are present in bone and become activated following injury; thus, elucidating mechanisms that promote adult stem cell-mediated healing is important. Wnt-associated adult stem marker Lgr6 is implicated in the regeneration of tissues with well-defined stem cell niches in stem cell-reliant organs. Here, we demonstrate that Lgr6 is dynamically expressed in osteoprogenitors in response to fracture injury. We used an Lgr6-null mouse model and found that Lgr6 expression is necessary for maintaining bone volume and efficient postnatal bone regeneration in adult mice. Skeletal progenitors isolated from Lgr6-null mice have reduced colony-forming potential and reduced osteogenic differentiation capacity due to attenuated cWnt signaling. Lgr6-null mice consist of a lower proportion of self-renewing stem cells. In response to fracture injury, Lgr6-null mice have a deficiency in the proliferation of periosteal progenitors and reduced ALP activity. Further, analysis of the bone regeneration phase and remodeling phase of fracture healing in Lgr6-null mice showed impaired endochondral ossification and decreased mineralization. We propose that in contrast to not being required for successful skeletal development, Lgr6-positive cells have a direct role in endochondral bone repair.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA; School of Dental Medicine, UConn Health, Farmington, CT 06030, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Anna Peterson
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Justin S King
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA
| | - Ivo Kalajzic
- School of Dental Medicine, UConn Health, Farmington, CT 06030, USA; Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmington, CT 06030, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, School of Medicine, USA.
| |
Collapse
|
14
|
Wang Z, Wen S, Zhong M, Yang Z, Xiong W, Zhang K, Yang S, Li H, Guo S. Epigenetics: Novel crucial approach for osteogenesis of mesenchymal stem cells. J Tissue Eng 2023; 14:20417314231175364. [PMID: 37342486 PMCID: PMC10278427 DOI: 10.1177/20417314231175364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/26/2023] [Indexed: 06/23/2023] Open
Abstract
Bone has a robust regenerative potential, but its capacity to repair critical-sized bone defects is limited. In recent years, stem cells have attracted significant interest for their potential in tissue engineering. Applying mesenchymal stem cells (MSCs) for enhancing bone regeneration is a promising therapeutic strategy. However, maintaining optimal cell efficacy or viability of MSCs is limited by several factors. Epigenetic modification can cause changes in gene expression levels without changing its sequence, mainly including nucleic acids methylation, histone modification, and non-coding RNAs. This modification is believed to be one of the determinants of MSCs fate and differentiation. Understanding the epigenetic modification of MSCs can improve the activity and function of stem cells. This review summarizes recent advances in the epigenetic mechanisms of MSCs differentiation into osteoblast lineages. We expound that epigenetic modification of MSCs can be harnessed to treat bone defects and promote bone regeneration, providing potential therapeutic targets for bone-related diseases.
Collapse
Affiliation(s)
- Zhaohua Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Si Wen
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Meiqi Zhong
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ziming Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Kuo Zhang
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, Liaoning Province, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Huizheng Li
- Department of Otorhinolaryngology & Head and Neck Surgery, Dalian Friendship Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
15
|
Epigenetic Alterations in Sports-Related Injuries. Genes (Basel) 2022; 13:genes13081471. [PMID: 36011382 PMCID: PMC9408207 DOI: 10.3390/genes13081471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
It is a well-known fact that physical activity benefits people of all age groups. However, highly intensive training, maladaptation, improper equipment, and lack of sufficient rest lead to contusions and sports-related injuries. From the perspectives of sports professionals and those performing regular–amateur sports activities, it is important to maintain proper levels of training, without encountering frequent injuries. The bodily responses to physical stress and intensive physical activity are detected on many levels. Epigenetic modifications, including DNA methylation, histone protein methylation, acetylation, and miRNA expression occur in response to environmental changes and play fundamental roles in the regulation of cellular activities. In the current review, we summarise the available knowledge on epigenetic alterations present in tissues and organs (e.g., muscles, the brain, tendons, and bones) as a consequence of sports-related injuries. Epigenetic mechanism observations have the potential to become useful tools in sports medicine, as predictors of approaching pathophysiological alterations and injury biomarkers that have already taken place.
Collapse
|
16
|
Effects of Systemic or Local Administration of Mesenchymal Stem Cells from Patients with Osteoporosis or Osteoarthritis on Femoral Fracture Healing in a Mouse Model. Biomolecules 2022; 12:biom12050722. [PMID: 35625649 PMCID: PMC9138345 DOI: 10.3390/biom12050722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
The purpose of this study was to analyze the regenerative capacity of mesenchymal stem cells (MSCs) in the treatment of fractures. MSCs extracted from patients with osteoporotic hip fractures or hip osteoarthritis undergoing hip replacement surgeries were cultured and injected into mice with femoral fracture. Two experimental models were established, one for the systemic administration of MSCs (n = 29) and another one for local administration (n = 30). Fracture consolidation was assessed by micro-CT and histology. The degree of radiological consolidation and corticalization was better with MSCs from osteoporosis than from osteoarthritis, being significant after systemic administration (p = 0.0302 consolidation; p = 0.0243 corticalization). The histological degree of consolidation was also better with MSCs from osteoporosis than from osteoarthritis. Differences in histological scores after systemic infusion were as follows: Allen, p = 0.0278; Huo, p = 0.3471; and Bone Bridge, p = 0.0935. After local administration at the fracture site, differences in histological scores were as follows: Allen, p = 0.0764; Huo, p = 0.0256; and Bone Bridge, p = 0.0012. As osteoporosis and control groups were similar, those differences depended on an inhibitory influence by MSCs from patients with osteoarthritis. In conclusion, we found an unexpected impairment of consolidation induced by MSCs from patients with osteoarthritis. However, MSCs from patients with osteoporosis compared favorably with cells from patients with osteoarthritis. In other words, based on this study and previous studies, MSCs from patients with osteoporosis do not appear to have worse bone-regenerating capabilities than MSCs from non-osteoporotic individuals of similar age.
Collapse
|
17
|
López-Delgado L, Del Real A, Sañudo C, Garcia-Ibarbia C, Laguna E, Menendez G, Garcia-Montesinos B, Santurtun A, Merino J, Pérez-Núñez MI, Riancho JA. Osteogenic capacity of mesenchymal stem cells from patients with osteoporotic hip fractures in vivo. Connect Tissue Res 2022; 63:243-255. [PMID: 33618587 DOI: 10.1080/03008207.2021.1894140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Human mesenchymal stem cells (MSCs) have the ability to differentiate into bone-forming osteoblasts. The aim of this study was to elucidate if MSCs from patients with OP show a senescent phenotype and explore their bone-forming ability in vivo. MATERIALS AND METHODS MSCs from patients with OP and controls with osteoarthritis (OA) were implanted into the subcutaneous tissue of immunodeficient mice for histological analysis and expression of human genes by RT-PCR. The expression of senescence-associated phenotype (SASP) genes, as well as p16, p21, and galactosidase, was studied in cultures of MSCs. RESULTS In vivo bone formation was evaluated in 103 implants (47 OP, 56 OA). New bone was observed in 45% of the implants with OP cells and 46% of those with OA cells (p = 0.99). The expression of several bone-related genes (collagen, osteocalcin, alkaline phosphatase, sialoprotein) was also similar in both groups. There were no differences between groups in SASP gene expression, p16, and p21 expression, or in senescence-associated galactosidase activity. CONCLUSION Senescence markers and the osteogenic capacity in vivo of MSCs from patients with OP are not inferior to that of cells from controls of similar age with OA. This supports the interest of future studies to evaluate the potential use of autologous MSCs from OP patients in bone regeneration procedures.
Collapse
Affiliation(s)
- Laura López-Delgado
- Department of Internal Medicine, Hospital Universitario Marqués De Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Alvaro Del Real
- Department of Internal Medicine, Hospital Universitario Marqués De Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Carolina Sañudo
- Department of Internal Medicine, Hospital Universitario Marqués De Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Carmen Garcia-Ibarbia
- Department of Internal Medicine, Hospital Universitario Marqués De Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Esther Laguna
- Department of Traumatology and Orthopedic Surgery, Hospital Universitario Marqués De Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Guillermo Menendez
- Department of Traumatology and Orthopedic Surgery, Hospital Universitario Marqués De Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | | | - Ana Santurtun
- Unit of Legal Medicine, University of Cantabria, IDIVAL, Santander, Spain
| | - Jesus Merino
- Department of Molecular Biology, University of Cantabria, IDIVAL, Santander, Spain
| | - María I Pérez-Núñez
- Department of Traumatology and Orthopedic Surgery, Hospital Universitario Marqués De Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Jose A Riancho
- Department of Internal Medicine, Hospital Universitario Marqués De Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| |
Collapse
|
18
|
Tsagiopoulou M, Pechlivanis N, Maniou M, Psomopoulos F. InterTADs: integration of multi-omics data on topologically associated domains, application to chronic lymphocytic leukemia. NAR Genom Bioinform 2022; 4:lqab121. [PMID: 35047813 PMCID: PMC8759567 DOI: 10.1093/nargab/lqab121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/21/2021] [Accepted: 12/13/2021] [Indexed: 11/25/2022] Open
Abstract
The integration of multi-omics data can greatly facilitate the advancement of research in Life Sciences by highlighting new interactions. However, there is currently no widespread procedure for meaningful multi-omics data integration. Here, we present a robust framework, called InterTADs, for integrating multi-omics data derived from the same sample, and considering the chromatin configuration of the genome, i.e. the topologically associating domains (TADs). Following the integration process, statistical analysis highlights the differences between the groups of interest (normal versus cancer cells) relating to (i) independent and (ii) integrated events through TADs. Finally, enrichment analysis using KEGG database, Gene Ontology and transcription factor binding sites and visualization approaches are available. We applied InterTADs to multi-omics datasets from 135 patients with chronic lymphocytic leukemia (CLL) and found that the integration through TADs resulted in a dramatic reduction of heterogeneity compared to individual events. Significant differences for individual events and on TADs level were identified between patients differing in the somatic hypermutation status of the clonotypic immunoglobulin genes, the core biological stratifier in CLL, attesting to the biomedical relevance of InterTADs. In conclusion, our approach suggests a new perspective towards analyzing multi-omics data, by offering reasonable execution time, biological benchmarking and potentially contributing to pattern discovery through TADs.
Collapse
|
19
|
Abstract
Osteoporosis, characterised by low bone mass, poor bone structure, and an increased risk of fracture, is a major public health problem. There is increasing evidence that the influence of the environment on gene expression, through epigenetic processes, contributes to variation in BMD and fracture risk across the lifecourse. Such epigenetic processes include DNA methylation, histone and chromatin modifications and non-coding RNAs. Examples of associations with phenotype include DNA methylation in utero linked to maternal vitamin D status, and to methylation of target genes such as OPG and RANKL being associated with osteoporosis in later life. Epigenome-wide association studies and multi-omics technologies have further revealed susceptibility loci, and histone acetyltransferases, deacetylases and methylases are being considered as therapeutic targets. This review encompasses recent advances in our understanding of epigenetic mechanisms in the regulation of bone mass and osteoporosis development, and outlines possible diagnostic and prognostic biomarker applications.
Collapse
Affiliation(s)
| | | | - Cyrus Cooper
- MRC Lifecourse Epidemiology Centre, University of Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| |
Collapse
|
20
|
Xu J, Liu G, Wang X, Hu Y, Luo H, Ye L, Feng Z, Li C, Kuang M, Zhang L, Zhou Y, Qi X. hUC-MSCs: evaluation of acute and long-term routine toxicity testing in mice and rats. Cytotechnology 2022; 74:17-29. [PMID: 35185283 PMCID: PMC8817012 DOI: 10.1007/s10616-021-00502-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/17/2021] [Indexed: 02/03/2023] Open
Abstract
Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) are present in human umbilical connective tissue and can differentiate into various cell types. Our previous studies have proved that hUC-MSCs do not lead to allergies and tumorigenesis. In the present study, the acute and long-term toxicity of hUC-MSCs in mice and rats was evaluated. The acute toxicity of hUC-MSCs was assessed in 8-week-old mice receiving two caudal intravenous (i.v.) injections of hUC-MSCs at the maximum tolerated dose of 1.5 × 107 cells/kg with an interval of 8 h and the observation period sustained for 14 days. For the long-term toxicity evaluation, rats were randomly divided into control, low-dose (3.0 × 105 cells/kg), mid-dose (1.5 × 106 cells/kg), and high-dose (7.5 × 106 cells/kg) groups, which were treated with hUC-MSCs via a caudal i.v. injection every 3 days for 90 days. Weight and food intake evaluation was performed for all rats for 2 weeks after the hUC-MSC administration. The animals were then sacrificed for hematological, blood biochemical, and pathological analyses, as well as organ index determination. We observed no obvious acute toxicity of hUC-MSCs in mice at the maximum tolerated dose. Long-term toxicity tests in rats showed no significant differences between HUC-MSC-treated and control groups in the following parameters: body weight, hematological and blood biochemical parameters, and histopathologic changes in the heart, liver, kidneys, and lungs. This study provides evidence of the safety of i.v. hUC-MSCs infusion for future clinical therapies.
Collapse
Affiliation(s)
- Jianwei Xu
- grid.413458.f0000 0000 9330 9891National Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guizhou Medical University, Guiyang, China ,Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China ,grid.413458.f0000 0000 9330 9891Department of Pharmacology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Gang Liu
- grid.413458.f0000 0000 9330 9891Department of Pharmacology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Xianyao Wang
- grid.413458.f0000 0000 9330 9891National Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guizhou Medical University, Guiyang, China
| | - Ya’nan Hu
- grid.263761.70000 0001 0198 0694Department of Cell Biology, Medical College of Soochow University, Suzhou, China
| | - Hongyang Luo
- Department of Otorhinolaryngology, People’s Hospital of Wudang District, Guiyang, China
| | - Lan Ye
- grid.413458.f0000 0000 9330 9891Department of Pharmacology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Zhanhui Feng
- grid.452244.1Neurological Department, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chen Li
- Department of Oncology, General Hospital of the Yangtze River Shipping, Wuhan, China
| | - Menglan Kuang
- grid.413458.f0000 0000 9330 9891School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Lijuan Zhang
- grid.413458.f0000 0000 9330 9891School of Nursing, Guizhou Medical University, Guiyang, China
| | - Yixia Zhou
- grid.443382.a0000 0004 1804 268XSchool of Nursing, Guizhou University of Traditional Chinese Medicine, 9# Beijing Road, Guiyang, China ,grid.452244.1Department of Nursing, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiaolan Qi
- grid.413458.f0000 0000 9330 9891Key Laboratory of Endemic and Ethnic Diseases (Guizhou Medical University), Ministry of Education, 9# Beijing Road, Guiyang, People’s Republic of China ,grid.413458.f0000 0000 9330 9891Key Laboratory of Medical Molecular Biology (Guizhou Medical University), Guiyang, 550004 People’s Republic of China
| |
Collapse
|
21
|
González-González A, García-Sánchez D, Alfonso-Fernández A, Haider KH, Rodríguez-Rey JC, Pérez-Campo FM. Regenerative Medicine Applied to the Treatment of Musculoskeletal Pathologies. HANDBOOK OF STEM CELL THERAPY 2022:1123-1158. [DOI: 10.1007/978-981-19-2655-6_50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Rauner M, Foessl I, Formosa MM, Kague E, Prijatelj V, Lopez NA, Banerjee B, Bergen D, Busse B, Calado Â, Douni E, Gabet Y, Giralt NG, Grinberg D, Lovsin NM, Solan XN, Ostanek B, Pavlos NJ, Rivadeneira F, Soldatovic I, van de Peppel J, van der Eerden B, van Hul W, Balcells S, Marc J, Reppe S, Søe K, Karasik D. Perspective of the GEMSTONE Consortium on Current and Future Approaches to Functional Validation for Skeletal Genetic Disease Using Cellular, Molecular and Animal-Modeling Techniques. Front Endocrinol (Lausanne) 2021; 12:731217. [PMID: 34938269 PMCID: PMC8686830 DOI: 10.3389/fendo.2021.731217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/30/2021] [Indexed: 12/26/2022] Open
Abstract
The availability of large human datasets for genome-wide association studies (GWAS) and the advancement of sequencing technologies have boosted the identification of genetic variants in complex and rare diseases in the skeletal field. Yet, interpreting results from human association studies remains a challenge. To bridge the gap between genetic association and causality, a systematic functional investigation is necessary. Multiple unknowns exist for putative causal genes, including cellular localization of the molecular function. Intermediate traits ("endophenotypes"), e.g. molecular quantitative trait loci (molQTLs), are needed to identify mechanisms of underlying associations. Furthermore, index variants often reside in non-coding regions of the genome, therefore challenging for interpretation. Knowledge of non-coding variance (e.g. ncRNAs), repetitive sequences, and regulatory interactions between enhancers and their target genes is central for understanding causal genes in skeletal conditions. Animal models with deep skeletal phenotyping and cell culture models have already facilitated fine mapping of some association signals, elucidated gene mechanisms, and revealed disease-relevant biology. However, to accelerate research towards bridging the current gap between association and causality in skeletal diseases, alternative in vivo platforms need to be used and developed in parallel with the current -omics and traditional in vivo resources. Therefore, we argue that as a field we need to establish resource-sharing standards to collectively address complex research questions. These standards will promote data integration from various -omics technologies and functional dissection of human complex traits. In this mission statement, we review the current available resources and as a group propose a consensus to facilitate resource sharing using existing and future resources. Such coordination efforts will maximize the acquisition of knowledge from different approaches and thus reduce redundancy and duplication of resources. These measures will help to understand the pathogenesis of osteoporosis and other skeletal diseases towards defining new and more efficient therapeutic targets.
Collapse
Affiliation(s)
- Martina Rauner
- Department of Medicine III, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- University Hospital Carl Gustav Carus, Dresden, Germany
| | - Ines Foessl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| | - Melissa M. Formosa
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Erika Kague
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Vid Prijatelj
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- The Generation R Study, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nerea Alonso Lopez
- Rheumatology and Bone Disease Unit, CGEM, Institute of Genetics and Cancer (IGC), Edinburgh, United Kingdom
| | - Bodhisattwa Banerjee
- Musculoskeletal Genetics Laboratory, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Dylan Bergen
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ângelo Calado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Eleni Douni
- Department of Biotechnology, Agricultural University of Athens, Athens, Greece
- Institute for Bioinnovation, B.S.R.C. “Alexander Fleming”, Vari, Greece
| | - Yankel Gabet
- Department of Anatomy & Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Natalia García Giralt
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - Nika M. Lovsin
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Xavier Nogues Solan
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - Barbara Ostanek
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Nathan J. Pavlos
- Bone Biology & Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | | | - Ivan Soldatovic
- Institute of Medical Statistics and Informatic, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Bram van der Eerden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Wim van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Susanna Balcells
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - Janja Marc
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Sjur Reppe
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
- Marcus Research Institute, Hebrew SeniorLife, Boston, MA, United States
| |
Collapse
|
23
|
García-Sánchez D, González-González A, García-García P, Reyes R, Pérez-Núñez MI, Riancho JA, Évora C, Rodríguez-Rey JC, Pérez-Campo FM. Effective Osteogenic Priming of Mesenchymal Stem Cells through LNA-ASOs-Mediated Sfrp1 Gene Silencing. Pharmaceutics 2021; 13:1277. [PMID: 34452242 PMCID: PMC8398380 DOI: 10.3390/pharmaceutics13081277] [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] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 01/21/2023] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has emerged as a promising approach for bone regeneration. Importantly, the beneficial effects of MSCs can be improved by modulating the expression levels of specific genes to stimulate MSC osteogenic differentiation. We have previously shown that Smurf1 silencing by using Locked Nucleic Acid-Antisense Oligonucleotides, in combination with a scaffold that sustainably releases low doses of BMP-2, was able to increase the osteogenic potential of MSCs in the presence of BMP-2 doses significantly smaller than those currently used in the clinic. This would potentially allow an important reduction in this protein in MSs-based treatments, and thus of the side effects linked to its administration. We have further improved this system by specifically targeting the Wnt pathway modulator Sfrp1. This approach not only increases MSC bone regeneration efficiency, but is also able to induce osteogenic differentiation in osteoporotic human MSCs, bypassing the need for BMP-2 induction, underscoring the regenerative potential of this system. Achieving successful osteogenesis with the sole use of LNA-ASOs, without the need of administering pro-osteogenic factors such as BMP-2, would not only reduce the cost of treatments, but would also open the possibility of targeting these LNA-ASOs specifically to MSCs in the bone marrow, allowing us to treat systemic bone loss such as that associated with osteoporosis.
Collapse
Affiliation(s)
- Daniel García-Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (J.C.R.-R.)
| | - Alberto González-González
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (J.C.R.-R.)
| | - Patricia García-García
- Department of Chemical Engineering and Pharmaceutical Technology, Institute of Biomedical Technologies (ITB), University of La Laguna, 38206 La Laguna, Spain; (P.G.-G.); (C.É.)
| | - Ricardo Reyes
- Department of Biochemistry, Microbiology, Cellular Biology and Genetics, Institute of Biomedical Technologies (ITB), University of La Laguna, 38200 La Laguna, Spain;
| | - María Isabel Pérez-Núñez
- Department of Traumatology, Hospital Universitario Marqués de Valdecilla, University of Cantabria, 39008 Santander, Spain;
| | - José A. Riancho
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, 39012 Santander, Spain;
| | - Carmen Évora
- Department of Chemical Engineering and Pharmaceutical Technology, Institute of Biomedical Technologies (ITB), University of La Laguna, 38206 La Laguna, Spain; (P.G.-G.); (C.É.)
| | - José Carlos Rodríguez-Rey
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (J.C.R.-R.)
| | - Flor M. Pérez-Campo
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (J.C.R.-R.)
| |
Collapse
|
24
|
Riquier S, Mathieu M, Bessiere C, Boureux A, Ruffle F, Lemaitre JM, Djouad F, Gilbert N, Commes T. Long non-coding RNA exploration for mesenchymal stem cell characterisation. BMC Genomics 2021; 22:412. [PMID: 34088266 PMCID: PMC8178833 DOI: 10.1186/s12864-020-07289-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/28/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The development of RNA sequencing (RNAseq) and the corresponding emergence of public datasets have created new avenues of transcriptional marker search. The long non-coding RNAs (lncRNAs) constitute an emerging class of transcripts with a potential for high tissue specificity and function. Therefore, we tested the biomarker potential of lncRNAs on Mesenchymal Stem Cells (MSCs), a complex type of adult multipotent stem cells of diverse tissue origins, that is frequently used in clinics but which is lacking extensive characterization. RESULTS We developed a dedicated bioinformatics pipeline for the purpose of building a cell-specific catalogue of unannotated lncRNAs. The pipeline performs ab initio transcript identification, pseudoalignment and uses new methodologies such as a specific k-mer approach for naive quantification of expression in numerous RNAseq data. We next applied it on MSCs, and our pipeline was able to highlight novel lncRNAs with high cell specificity. Furthermore, with original and efficient approaches for functional prediction, we demonstrated that each candidate represents one specific state of MSCs biology. CONCLUSIONS We showed that our approach can be employed to harness lncRNAs as cell markers. More specifically, our results suggest different candidates as potential actors in MSCs biology and propose promising directions for future experimental investigations.
Collapse
Affiliation(s)
- Sébastien Riquier
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Marc Mathieu
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Chloé Bessiere
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Anthony Boureux
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Florence Ruffle
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Jean-Marc Lemaitre
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Farida Djouad
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Nicolas Gilbert
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| | - Thérèse Commes
- IRMB, University of Montpellier, INSERM, 80 rue Augustin Fliche, Montpellier, France
| |
Collapse
|
25
|
Xu F, Li W, Yang X, Na L, Chen L, Liu G. The Roles of Epigenetics Regulation in Bone Metabolism and Osteoporosis. Front Cell Dev Biol 2021; 8:619301. [PMID: 33569383 PMCID: PMC7868402 DOI: 10.3389/fcell.2020.619301] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/31/2020] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis is a metabolic disease characterized by decreased bone mineral density and the destruction of bone microstructure, which can lead to increased bone fragility and risk of fracture. In recent years, with the deepening of the research on the pathological mechanism of osteoporosis, the research on epigenetics has made significant progress. Epigenetics refers to changes in gene expression levels that are not caused by changes in gene sequences, mainly including DNA methylation, histone modification, and non-coding RNAs (lncRNA, microRNA, and circRNA). Epigenetics play mainly a post-transcriptional regulatory role and have important functions in the biological signal regulatory network. Studies have shown that epigenetic mechanisms are closely related to osteogenic differentiation, osteogenesis, bone remodeling and other bone metabolism-related processes. Abnormal epigenetic regulation can lead to a series of bone metabolism-related diseases, such as osteoporosis. Considering the important role of epigenetic mechanisms in the regulation of bone metabolism, we mainly review the research progress on epigenetic mechanisms (DNA methylation, histone modification, and non-coding RNAs) in the osteogenic differentiation and the pathogenesis of osteoporosis to provide a new direction for the treatment of bone metabolism-related diseases.
Collapse
Affiliation(s)
- Fei Xu
- College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Collaborative Innovation Center, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Wenhui Li
- Collaborative Innovation Center, Shanghai University of Medicine and Health Sciences, Shanghai, China
- College of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xiao Yang
- Traditional Chinese Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lixin Na
- Collaborative Innovation Center, Shanghai University of Medicine and Health Sciences, Shanghai, China
- College of Public Health, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Linjun Chen
- College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Guobin Liu
- Traditional Chinese Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Chen J, Tian Y, Zhang Q, Ren D, Zhang Q, Yan X, Wang L, He Z, Zhang W, Zhang T, Yuan X. Novel Insights Into the Role of N6-Methyladenosine RNA Modification in Bone Pathophysiology. Stem Cells Dev 2020; 30:17-28. [PMID: 33231507 DOI: 10.1089/scd.2020.0157] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Thus far, there are more than known 150 modifications to RNA, in which common internal modifications of mRNA include N6-methyladenosine (m6A), N1-methyladenosine, and 5-methylcytosine. Among them, m6A RNA modification is one of the highest abundance modifications in eukaryotes, regulating mechanisms controlling gene expression at the post-transcription level. As an invertible and dynamic epigenetic marker, m6A base modification influences almost all vital biological processes, cellular components, and molecular functions. Once the m6A modification process is abnormal, a series of diseases-including cancer, neurological diseases, and growth disorders-will be caused. Besides, several base modification activities also have been created by noncoding RNAs (ncRNAs), for instance, microRNAs, and circular RNAs, long ncRNAs, which were dynamically regulated during bone and cartilage pathophysiology processes. Therefore, it has now been clear that dynamic modification on coding RNAs and ncRNAs represents a completely new way to modulate genetic information. In this review, we highlight up-to-date progress and applications of m6A RNA modification in bone and cartilage pathophysiology, and we discuss the pathological roles and underlying molecular mechanism of m6A modifications in osteoarthritis and osteoporosis and osteosarcoma pathogenesis.
Collapse
Affiliation(s)
- Junbo Chen
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Yihong Tian
- School of Stomatology, Qingdao University, Qingdao, China
| | - Qi Zhang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Dapeng Ren
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qiang Zhang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao Yan
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lingzhi Wang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Zijing He
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Wei Zhang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Tianzhen Zhang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| | - Xiao Yuan
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology, Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Li Y, Wei Z, Huang S, Yang B. mRNA expression and DNA methylation analysis of the inhibitory mechanism of H 2O 2 on the proliferation of A549 cells. Oncol Lett 2020; 20:288. [PMID: 33014166 PMCID: PMC7520746 DOI: 10.3892/ol.2020.12151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 08/18/2020] [Indexed: 01/29/2023] Open
Abstract
Reactive oxygen species, particularly hydrogen peroxide (H2O2), can induce proliferation inhibition and death of A549 cells via oxidative stress. Oxidative stress has effect on DNA methylation. Oxidative stress and DNA methylation feature a common denominator: The one carbon cycle. To explore the inhibitory mechanism of H2O2 on the proliferation of lung cancer cells, the present study analysed the mRNA expression and methylation profiles in A549 cells treated with H2O2 for 24 h, as adenocarcinoma is the most common pathological type of lung cancer. The DNA methylation profile was constructed using reduced representation bisulphite sequencing, which identified 29,755 differentially methylated sites (15,365 upregulated and 14,390 downregulated), and 1,575 differentially methylated regions located in the gene promoters were identified using the methylKit. Analysis of the assocaition between gene expression and methylation levels revealed that several genes were downregulated and hypermethylated, including cyclin-dependent kinase inhibitor 3, denticleless E3 ubiquitin protein ligase homolog, centromere protein (CENP)F, kinesin family member (KIF)20A, CENPA, KIF11, PCNA clamp-associated factor and GINS complex subunit 2, which may be involved in the inhibitory process of H2O2 on the proliferation of A549 cells.
Collapse
Affiliation(s)
- Yepeng Li
- Department of Oncology, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Zhongheng Wei
- Department of Oncology, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Shiqing Huang
- Department of Oncology, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Bo Yang
- Key Laboratory of Guangxi College and Universities, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| |
Collapse
|
28
|
Ren JC, Wang T, Wu H, Zhang GH, Sun D, Guo K, Li H, Zhang F, Wu W, Xia ZL. Promoter hypermethylation in CSF3R induces peripheral neutrophil reduction in benzene-exposure poisoning. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:786-796. [PMID: 32329128 DOI: 10.1002/em.22382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/22/2020] [Accepted: 04/19/2020] [Indexed: 06/11/2023]
Abstract
Benzene is a global pollutant and has been established to cause leukemia. To better understand the role of DNA methylation in benzene toxicity, peripheral blood mononuclear cells were collected from six benzene-poisoning patients and six matched controls for genome-wide DNA methylation screening by Illumina Infinium Methylation 450 BeadChip. The Gene Chip Human Gene 2.0 ST Array (Affymetrix) was used to analyze global mRNA expression. Compared with the corresponding sites of controls, 442 sites in patients were hypermethylated, corresponding to 253 genes, and 237 sites were hypomethylated, corresponding to 130 genes. The promoter methylation and mRNA expression of CSF3R, CREB5, and F2R were selected for verification by bisulfite sequencing and real-time PCR in a larger data set with 21 cases and 23 controls. The results indicated that promoter methylation of CSF3R (p = .005) and F2R (p = .015) was significantly higher in cases than in controls. Correlation analysis showed that the promoter methylation of CSF3R (p < .001) and F2R (p < .001) was highly correlated with its mRNA expression. In the poisoning cases, neutrophil percentage was significantly different among the high, middle, and low CSF3R-methylation groups (p = .002). In particular, the neutrophil percentage in the high CSF3R-methylation group (48.10 ± 9.63%) was significantly lower than that in the low CSF3R-methylation group (59.30 ± 6.26%) (p = .012). The correlation coefficient between promoter methylation in CSF3R and the neutrophil percentage was -0.445 (p = .020) in cases and - 0.398 (p = .060) in controls. These results imply that hypermethylation occurs in the CSF3R promoter due to benzene exposure and is significantly associated with a reduction in neutrophils.
Collapse
Affiliation(s)
- Jing-Chao Ren
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Tongshuai Wang
- Department of Occupational Health and Toxicology, School of Public Health, Fudan University, Shanghai, China
| | - Hantian Wu
- Department of Occupational Health and Toxicology, School of Public Health, Fudan University, Shanghai, China
| | - Guang-Hui Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Daoyuan Sun
- Tongji University Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Kongrong Guo
- Tongji University Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Haibin Li
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Fengquan Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Zhao-Lin Xia
- Department of Occupational Health and Toxicology, School of Public Health, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Del Real A, López-Delgado L, Sañudo C, García-Ibarbia C, Laguna E, Perez-Campo FM, Menéndez G, Alfonso A, Fakkas M, García-Montesinos B, Valero C, Pérez-Núñez MI, Riancho JA. Long Noncoding RNAs as Bone Marrow Stem Cell Regulators in Osteoporosis. DNA Cell Biol 2020; 39:1691-1699. [PMID: 32700968 DOI: 10.1089/dna.2020.5672] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) contribute toward regulating gene expression and cell differentiation and may be involved in the pathogenesis of several diseases. The objective of this study was to determine the expression patterns of lncRNAs in bone marrow mesenchymal stem cells (BMSCs) derived from patients with osteoporotic fractures and their relevance to osteogenic function. The BMSCs were isolated from the femoral head of patients with hip fractures (FRX) and controls with osteoarthritis (OA). We found 74 differentially expressed genes between FRX and OA, of which 33 were of the lncRNA type. Among them, 52 genes (20 lncRNAs) were replicated in another independent dataset. The differentially expressed lncRNAs were over-represented among those correlated with differentially expressed protein-coding genes. In addition, the comparison of pre- and post-differentiated paired samples revealed 163 differentially expressed genes, of which 99 were of the lncRNA type. Among them, the overexpression of LINC00341 induced an upregulation of typical osteoblastic genes. In conclusion, the analysis of lncRNA expression in BMSCs shows specific patterns in patients with osteoporotic fractures, as well as changes associated with osteogenic differentiation. The regulation of bone genes through lncRNAs might bring new opportunities for designing bone anabolic therapies in systemic and localized bone disorders.
Collapse
Affiliation(s)
- Alvaro Del Real
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Laura López-Delgado
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Carolina Sañudo
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Carmen García-Ibarbia
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Esther Laguna
- Department of Traumatology and Orthopedic Surgery, Hospital UM Valdecilla, University of Cantabria-IDIVAL, Santander, Spain
| | - Flor M Perez-Campo
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, Santander, Spain
| | - Guillermo Menéndez
- Department of Traumatology and Orthopedic Surgery, Hospital UM Valdecilla, University of Cantabria-IDIVAL, Santander, Spain
| | - Ana Alfonso
- Department of Traumatology and Orthopedic Surgery, Hospital UM Valdecilla, University of Cantabria-IDIVAL, Santander, Spain
| | - Michel Fakkas
- Department of Traumatology and Orthopedic Surgery, Hospital UM Valdecilla, University of Cantabria-IDIVAL, Santander, Spain
| | - Belén García-Montesinos
- Department of Maxillofacial Surgery, Faculty of Medicine, University of Cantabria-IDIVAL, Santander, Spain
| | - Carmen Valero
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| | - Maria Isabel Pérez-Núñez
- Department of Traumatology and Orthopedic Surgery, Hospital UM Valdecilla, University of Cantabria-IDIVAL, Santander, Spain
| | - Jose A Riancho
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, Santander, Spain
| |
Collapse
|
30
|
Cakouros D, Gronthos S. The changing epigenetic landscape of Mesenchymal Stem/Stromal Cells during aging. Bone 2020; 137:115440. [PMID: 32445894 DOI: 10.1016/j.bone.2020.115440] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/17/2020] [Accepted: 05/17/2020] [Indexed: 12/17/2022]
Abstract
There is mounting evidence in the literature that mesenchymal stromal/stem cell (MSC) like populations derived from different tissues, undergo epigenetic changes during aging, leading to compromised connective tissue integrity and function. This body of work has linked the biological aging of MSC to changes in their epigenetic signatures affecting growth, lifespan, self-renewal and multi-potential, due to deregulation of processes such as cellular senescence, oxidative stress, DNA damage, telomere shortening and DNA damage. This review addresses recent findings examining DNA methylation, histone modifications and miRNA changes in aging MSC populations. Moreover, we explore how epigenetic factors alter cellular pathways and associated biological networks, contributing to the MSC aging phenotype. Finally we discuss the crucial areas requiring a greater understanding of these processes, in order to piece together a global picture of the changing epigenetic landscape in MSC during aging.
Collapse
Affiliation(s)
- Dimitrios Cakouros
- Mesenchymal Stem Cell Laboratory, School of Medical Sciences, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, School of Medical Sciences, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| |
Collapse
|
31
|
Anagliptin stimulates osteoblastic cell differentiation and mineralization. Biomed Pharmacother 2020; 129:109796. [PMID: 32559615 DOI: 10.1016/j.biopha.2019.109796] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 01/08/2023] Open
Abstract
Osteoporosis is a common debilitating bone disease characterized by loss of bone mass and degradation of the bone architecture, which is primarily driven by dysregulated differentiation of mesenchymal stem cells into bone-producing osteoblasts. Osteoblasts contribute to bone formation by secreting various proteins that guide the deposition of bone extracellular matrix, such as alkaline phosphatase (ALP), osteocalcin (OCN), and osteopontin (OPN). The Wnt/β-catenin pathway is widely recognized as a regulator of bone mass and is required to maintain bone homeostasis. Hormones have long been recognized as playing a key role in bone metabolism, and in recent years, growing evidence has shown that diabetes is a risk factor for osteoporosis. In the present study, we investigated the effects of the antidiabetic drug anagliptin on the differentiation and mineralization of osteoblasts induced by osteogenic medium. Anagliptin promotes insulin production via inhibition of dipeptidyl peptidase IV (DPP-4), an enzyme that targets the incretin hormone glucagon-like peptide 1 (GLP-1) for degradation. Our findings show that anagliptin significantly increases the differentiation of MSCs into osteoblasts via activation of RUNX2. Anagliptin significantly increased matrix deposition and mineralization by osteoblasts, as evidenced by elevated levels of ALP, OCN, OPN, and BMP-2. We further demonstrate that anagliptin activates the canonical and noncannonical Wnt signaling pathways and that silencing of Wnt/β-catenin signaling completely abolished the effects of anagliptin. Thus, anagliptin might be a safe, effective therapy for type II diabetes that might show promise as a therapy against osteoporosis.
Collapse
|
32
|
Efficacy of MSC for steroid-refractory acute GVHD associates with MSC donor age and a defined molecular profile. Bone Marrow Transplant 2020; 55:2188-2192. [PMID: 32346077 DOI: 10.1038/s41409-020-0910-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 12/30/2022]
|
33
|
Seelbinder B, Scott AK, Nelson I, Schneider SE, Calahan K, Neu CP. TENSCell: Imaging of Stretch-Activated Cells Reveals Divergent Nuclear Behavior and Tension. Biophys J 2020; 118:2627-2640. [PMID: 32407683 DOI: 10.1016/j.bpj.2020.03.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/08/2020] [Accepted: 03/13/2020] [Indexed: 12/22/2022] Open
Abstract
Mechanisms of cellular and nuclear mechanosensation are unclear, partially because of a lack of methods that can reveal dynamic processes. Here, we present a new concept for a low-cost, three-dimensionally printed device that enables high-magnification imaging of cells during stretch. We observed that nuclei of mouse embryonic skin fibroblasts underwent rapid (within minutes) and divergent responses, characterized by nuclear area expansion during 5% strain but nuclear area shrinkage during 20% strain. Only responses to low strain were dependent on calcium signaling, whereas actin inhibition abrogated all nuclear responses and increased nuclear strain transfer and DNA damage. Imaging of actin dynamics during stretch revealed similar divergent trends, with F-actin shifting away from (5% strain) or toward (20% strain) the nuclear periphery. Our findings emphasize the importance of simultaneous stimulation and data acquisition to capture mechanosensitive responses and suggest that mechanical confinement of nuclei through actin may be a protective mechanism during high mechanical stretch or loading.
Collapse
Affiliation(s)
- Benjamin Seelbinder
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Adrienne K Scott
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Isabel Nelson
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Stephanie E Schneider
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Kristin Calahan
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Corey P Neu
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado.
| |
Collapse
|
34
|
Ying J, Xu T, Wang C, Jin H, Tong P, Guan J, Abu-Amer Y, O’Keefe R, Shen J. Dnmt3b ablation impairs fracture repair through upregulation of Notch pathway. JCI Insight 2020; 5:131816. [PMID: 32051335 PMCID: PMC7098799 DOI: 10.1172/jci.insight.131816] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
We previously established that DNA methyltransferase 3b (Dnmt3b) is the sole Dnmt responsive to fracture repair and that Dnmt3b expression is induced in progenitor cells during fracture repair. In the current study, we confirmed that Dnmt3b ablation in mesenchymal progenitor cells (MPCs) resulted in impaired endochondral ossification, delayed fracture repair, and reduced mechanical strength of the newly formed bone in Prx1-Cre;Dnmt3bf/f (Dnmt3bPrx1) mice. Mechanistically, deletion of Dnmt3b in MPCs led to reduced chondrogenic and osteogenic differentiation in vitro. We further identified Rbpjκ as a downstream target of Dnmt3b in MPCs. In fact, we located 2 Dnmt3b binding sites in the murine proximal Rbpjκ promoter and gene body and confirmed Dnmt3b interaction with the 2 binding sites by ChIP assays. Luciferase assays showed functional utilization of the Dnmt3b binding sites in murine C3H10T1/2 cells. Importantly, we showed that the MPC differentiation defect observed in Dnmt3b deficiency cells was due to the upregulation of Rbpjκ, evident by restored MPC differentiation upon Rbpjκ inhibition. Consistent with in vitro findings, Rbpjκ blockage via dual antiplatelet therapy reversed the differentiation defect and accelerated fracture repair in Dnmt3bPrx1 mice. Collectively, our data suggest that Dnmt3b suppresses Notch signaling during MPC differentiation and is necessary for normal fracture repair.
Collapse
Affiliation(s)
- Jun Ying
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Institute of Orthopaedics and Traumatology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Taotao Xu
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Institute of Orthopaedics and Traumatology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuicui Wang
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianjun Guan
- Department of Biomedical Engineering, School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Shriners Hospital for Children, St. Louis, Missouri, USA
| | - Regis O’Keefe
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
35
|
García‐García P, Ruiz M, Reyes R, Delgado A, Évora C, Riancho JA, Rodríguez‐Rey JC, Pérez‐Campo FM. Smurf1 Silencing Using a LNA-ASOs/Lipid Nanoparticle System to Promote Bone Regeneration. Stem Cells Transl Med 2019; 8:1306-1317. [PMID: 31631568 PMCID: PMC6877774 DOI: 10.1002/sctm.19-0145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/17/2019] [Indexed: 12/19/2022] Open
Abstract
Despite the great advance of bone tissue engineering in the last few years, repair of bone defects remains a major problem. Low cell engraftment and dose-dependent side effects linked to the concomitant administration of bone morphogenetic proteins (BMPs) are the main problems currently hindering the clinical use of mesenchymal stem cell (MSC)-based therapies in this field. We have managed to bypass these drawbacks by combining the silencing the Smurf1 ubiquitin ligase in MSCs with the use of a scaffold that sustainably releases low doses of BMP-2. In this system, Smurf1 silencing is achieved by using GapmeRs, a clinically safe method that avoids the use of viral vectors, facilitating its translation to the clinic. Here, we show that a single transient transfection with a small quantity of a Smurf1-specific GapmeR is able to induce a significant level of silencing of the target gene, enough to prime MSCs for osteogenic differentiation. Smurf1 silencing highly increases MSCs responsiveness to BMP-2, allowing a dramatic reduction of the dose needed to achieve the desired therapeutic effect. The combination of these primed cells with alginate scaffolds designed to sustainably and locally release low doses of BMP-2 to the defect microenvironment is able to induce the formation of a mature bone matrix both in an osteoporotic rat calvaria system and in a mouse ectopic model. Importantly, this approach also enhances osteogenic differentiation in MSCs from osteoporotic patients, characterized by a reduced bone-forming potential, even at low BMP doses, underscoring the regenerative potential of this system. Stem Cells Translational Medicine 2019;8:1306&1317.
Collapse
Affiliation(s)
- Patricia García‐García
- Department of Chemical Engineering and Pharmaceutical TechnologyInstitute of Biomedical Technologies (ITB), University of La LagunaLa LagunaSpain
| | - Mario Ruiz
- Department of Molecular Biology, Faculty of MedicineUniversity of Cantabria, IDIVALSantanderSpain
| | - Ricardo Reyes
- Department of Biochemistry, Microbiology, Cellular Biology and GeneticsInstitute of Biomedical Technologies (ITB), University of La LagunaLa LagunaSpain
| | - Araceli Delgado
- Department of Chemical Engineering and Pharmaceutical TechnologyInstitute of Biomedical Technologies (ITB), University of La LagunaLa LagunaSpain
| | - Carmen Évora
- Department of Chemical Engineering and Pharmaceutical TechnologyInstitute of Biomedical Technologies (ITB), University of La LagunaLa LagunaSpain
| | - José Antonio Riancho
- Department of Internal Medicine, Hospital U M ValdecillaUniversity of Cantabria, IDIVALSantanderSpain
| | | | - Flor María Pérez‐Campo
- Department of Molecular Biology, Faculty of MedicineUniversity of Cantabria, IDIVALSantanderSpain
| |
Collapse
|
36
|
Li Y, Xie B, Jiang Z, Yuan B. Relationship between osteoporosis and osteoarthritis based on DNA methylation. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3399-3407. [PMID: 31934183 PMCID: PMC6949860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 07/23/2019] [Indexed: 06/10/2023]
Abstract
The aim of this study was to investigate the relationship between osteoporosis and osteoarthritis by analyzing the DNA methylation in osteoporosis and osteoarthritis. The cancellous bone specimens were collected from a total of 12 hospitalized patients and divided into the osteoporosis group (OA), the osteoarthritis group (OP), the osteoporosis combined with osteoarthritis group (OA & OP), and the normal control group (N). The cancellous bone specimens of each group were detected and the differences in gene expression profiles by the MeDIP-chip technique were compared. Compared with Group OA & OP, the methylation levels in Group OA and Group OP were statistically higher, P < 0.05. In the microarray analysis, a total of 1,222 sites occurred hypermethylation. The analysis targeting the differentially expressed genes between Group OA & OP and Group N revealed that group OA and group OP had 4 common genes: PPIL3, NIF3L1, SMTN, and CALHM2. The level of genomic methylation is lower in the patients with osteoporosis and/or osteoarthritis. The common difference between osteoarthritis and osteoporosis is reflected in some specific promoters, which may participate in the processes of diseases through different pathways.
Collapse
Affiliation(s)
- Ying Li
- Department of Orthopedics, Guangdong Province Integrative Medicine Hospital Foshan, China
| | - Bing Xie
- Department of Orthopedics, Guangdong Province Integrative Medicine Hospital Foshan, China
| | - Zhiqiang Jiang
- Department of Orthopedics, Guangdong Province Integrative Medicine Hospital Foshan, China
| | - Binbin Yuan
- Department of Orthopedics, Guangdong Province Integrative Medicine Hospital Foshan, China
| |
Collapse
|
37
|
Lee H, Min SK, Song Y, Park YH, Park JB. Bone morphogenetic protein-7 upregulates genes associated with osteoblast differentiation, including collagen I, Sp7 and IBSP in gingiva-derived stem cells. Exp Ther Med 2019; 18:2867-2876. [PMID: 31555377 PMCID: PMC6755424 DOI: 10.3892/etm.2019.7904] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 07/19/2019] [Indexed: 12/22/2022] Open
Abstract
The present study was performed to evaluate the effects of short-term application of bone morphogenetic protein-7 (BMP-7) on human gingiva-derived mesenchymal stem cells with next-generation sequencing. Human gingiva-derived stem cells were treated with a final concentration of 100 ng/ml BMP-7 and the same concentration of a vehicle control. mRNA sequencing and data analysis were performed along using gene ontology and pathway analysis. RT-qPCR of mRNA of collagen I, Sp7, IBSP and western blot analysis of collagen I, osterix and bone sialoprotein was also performed. A total of 25,737 mRNAs were identified to be differentially expressed. Regarding osteoblast differentiation, 14 mRNAs were upregulated and 10 were downregulated when the results of the BMP-7 at 3 h were compared with the control at 3 h. The expression of collagen I was increased following the application of BMP-7 at 3 h, and this increase was also observed following western blot analysis. The effects of BMP-7 on stem cells were evaluated with mRNA sequencing, and the expression was validated with RT-qPCR and western blot analysis. The short-term application of BMP-7 produced an increased expression of collagen I, which was associated with target genes selected for osteoblast differentiation. This study may provide novel insights into the role of BMP-7 using mRNA sequencing.
Collapse
Affiliation(s)
- Hyunjin Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sae Kyung Min
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Youngmin Song
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | | | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
38
|
Cortés-Pereira E, Fernández-Tajes J, Fernández-Moreno M, Vázquez-Mosquera ME, Relaño S, Ramos-Louro P, Durán-Sotuela A, Dalmao-Fernández A, Oreiro N, Blanco FJ, Rego-Pérez I. Differential Association of Mitochondrial DNA Haplogroups J and H With the Methylation Status of Articular Cartilage: Potential Role in Apoptosis and Metabolic and Developmental Processes. Arthritis Rheumatol 2019; 71:1191-1200. [PMID: 30747498 DOI: 10.1002/art.40857] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/07/2019] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To analyze the influence of mitochondrial genome variation on the DNA methylome of articular cartilage. METHODS DNA methylation profiling was performed using data deposited in the NCBI Gene Expression Omnibus database (accession no. GSE43269). Data were obtained for 14 cartilage samples from subjects with haplogroup J and 20 cartilage samples from subjects with haplogroup H. Subsequent validation was performed in an independent subset of 7 subjects with haplogroup J and 9 with haplogroup H by RNA-seq. Correlated genes were validated by real-time polymerase chain reaction in an independent cohort of 12 subjects with haplogroup J and 12 with haplogroup H. Appropriate analyses were performed using R Bioconductor and qBasePlus software, and gene ontology analysis was conducted using DAVID version 6.8. RESULTS DNA methylation profiling revealed 538 differentially methylated loci, while whole-transcriptome profiling identified 2,384 differentially expressed genes, between cartilage samples from subjects with haplogroup H and those with haplogroup J. Seventeen genes showed an inverse correlation between methylation and expression. In terms of gene ontology, differences in correlations between methylation and expression were also detected between cartilage from subjects with haplogroup H and those with haplogroup J, highlighting a significantly enhanced apoptotic process in cartilage from subjects with haplogroup H (P = 0.007 for methylation and P = 0.019 for expression) and repressed apoptotic process in cartilage from subjects with haplogroup J (P = 0.021 for methylation), as well as a significant enrichment of genes related to metabolic processes (P = 1.93 × 10-4 for methylation and P = 6.79 x 10-4 for expression) and regulation of gene expression (P = 0.012 for methylation) in cartilage from subjects with haplogroup H, and to developmental processes (P = 0.015 for methylation and P = 8.25 x 10-12 for expression) in cartilage from subjects with haplogroup J. CONCLUSION Mitochondrial DNA variation differentially associates with the methylation status of articular cartilage by acting on key mechanisms involved in osteoarthritis, such as apoptosis and metabolic and developmental processes.
Collapse
Affiliation(s)
- Estefanía Cortés-Pereira
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | | | | | - María E Vázquez-Mosquera
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Sara Relaño
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Paula Ramos-Louro
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Alejandro Durán-Sotuela
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Andrea Dalmao-Fernández
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Natividad Oreiro
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Francisco J Blanco
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Ignacio Rego-Pérez
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| |
Collapse
|
39
|
Yalaev BI, Tyurin AV, Mirgalieva RY, Khusainova RI. The role of DNA methylation in the disorders of bone metabolism. Vavilovskii Zhurnal Genet Selektsii 2019; 23:67-74. [DOI: 10.18699/vj19.463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Osteoporosis is one of multifactorial diseases, it develops from interactions between the genetic component and the environment. However, the universal epigenetic markers of osteoporosis are not yet defined. Finding the risk factors will predict the risk of osteoporosis at the preclinical stage, help define the course and severity of the disease, and develop preventive measures based on them to reduce the risk of fractures. Expanding knowledge in the field of bone biology, especially in the genetics of osteoporosis and osteoimmunology, showed that osteoporosis is a disease that occurs not only due to hormonal or mechanical disorders, but also as a clinically and genetically heterogeneous disease, and there are still unknown pathogenetic links in its structure. Decreases in bone mass and matrix mineralization as well as changes in bone microarchitecture can have different pathogenetic patterns of development and, moreover, there are unknown links of the pathogenesis of osteoporosis. It is possible that DNA methylation is one of these links and a mechanism for epigenetic regulation of gene expression. Evidence exists that this mechanism alongside regulatory miRNAs and post-translational modifications makes a significant contribution to the central processes of bone remodeling; however, the results of various studies vary greatly, and, therefore, there is a need to understand the significance of the accumulated data and to make them consistent. The purpose of this review is to compile and systematize data on the role of DNA methylation in bone metabolism in normal and pathological conditions, in the formation of osteoporosis, and to assess achievements and trends in this field of research and technologies for studying DNA methylation.
Collapse
Affiliation(s)
- B. I. Yalaev
- Institute of Biochemistry and Genetics of Ufa Science Centre RAS
| | | | | | - R. I. Khusainova
- Institute of Biochemistry and Genetics of Ufa Science Centre RAS; Republican Medical-Genetic Center
| |
Collapse
|
40
|
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
|
41
|
Koromani F, Trajanoska K, Rivadeneira F, Oei L. Recent Advances in the Genetics of Fractures in Osteoporosis. Front Endocrinol (Lausanne) 2019; 10:337. [PMID: 31231309 PMCID: PMC6559287 DOI: 10.3389/fendo.2019.00337] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 05/10/2019] [Indexed: 12/15/2022] Open
Abstract
Genetic susceptibility, together with old age, female sex, and low bone mineral density (BMD) are amongst the strongest determinants of fracture risk. Tmost recent large-scale genome-wide association study (GWAS) meta-analysis has yielded fifteen loci. This review focuses on the advances in the research of genetic determinants of fracture risk. We first discuss the genetic architecture of fracture risk, touching upon different methods and overall findings. We then discuss in a second paragraph the most recent advances in the field and focus on the genetics of fracture risk and also of other endophenotypes closely related to fracture risk such as bone mineral density (BMD). Application of state-of-the-art methodology such as Mendelian randzation in fracture GWAS are reviewed. The final part of this review touches upon potential future directions in genetic research of osteoporotic fractures.
Collapse
Affiliation(s)
- Fjorda Koromani
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Ling Oei
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- *Correspondence: Ling Oei
| |
Collapse
|
42
|
Affiliation(s)
- Alvaro Del Real
- Department of Internal Medicine, Hospital U.M. Valdecilla IDIVAL, University of Cantabria, Santander, Spain
| | | | - Jose A Riancho
- Department of Internal Medicine, Hospital U.M. Valdecilla IDIVAL, University of Cantabria, Santander, Spain
| |
Collapse
|
43
|
Jakob F, Ebert R, Fernandez-Rebollo E, Bischof O, Wagner W. Response to Letter to the Editor: Epigenetic Aging in Osteoporosis. J Bone Miner Res 2018; 33:1904-1905. [PMID: 30589450 DOI: 10.1002/jbmr.3569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Eduardo Fernandez-Rebollo
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany.,Institute for Biomedical Technology-Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Oliver Bischof
- Laboratory of Nuclear Organization and Oncogenesis, Department of Cell Biology and Infection, INSERM U.993, Institute Pasteur, Paris, France
| | - Wolfgang Wagner
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany.,Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW 'Omics' technologies developed for the massive analysis of the major biologically relevant molecules (genes, proteins, metabolites) have been applied to the study of osteoarthritis (OA) for more than a decade. RECENT FINDINGS 'Omics' studies have undoubtedly contributed to increase the knowledge on pathogenic processes related with OA and have provided hundreds to thousands of molecules that might have a putative biomarker utility for this disease. SUMMARY This review describes the most recent 'omics' studies in OA research, their conclusions, and discuss those remaining challenges. Still many validation studies must be performed in large and well-characterized cohorts for the translation of the findings from 'omics' strategies to clinical applications. The development of tools for the intelligent integration of 'omics' data with clinical and imaging information is also mandatory to take full profit of the work that has been already performed.
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Epigenetic mechanisms modify gene activity in a stable manner without altering DNA sequence. They participate in the adaptation to the environment, as well as in the pathogenesis of common complex disorders. We provide an overview of the role of epigenetic mechanisms in bone biology and pathology. RECENT FINDINGS Extensive evidence supports the involvement of epigenetic mechanisms (DNA methylation, post-translational modifications of histone tails, and non-coding RNAs) in the differentiation of bone cells and mechanotransduction. A variety of epigenetic abnormalities have been described in patients with osteoporosis, osteoarthritis, and skeletal cancers, but their actual pathogenetic roles are still unclear. A few drugs targeting epigenetic marks have been approved for neoplastic disorders, and many more are being actively investigated. Advances in the field of epigenetics underscore the complex interactions between genetic and environmental factors as determinants of osteoporosis and other common disorders. Likewise, they help to explain the mechanisms by which prenatal and post-natal external factors, from nutrition to psychological stress, impact our body and influence the risk of later disease.
Collapse
Affiliation(s)
- Alvaro Del Real
- Department of Internal Medicine, Hospital U.M. Valdecilla IDIVAL, University of Cantabria, 39008, Santander, Spain
| | | | - Laura López-Delgado
- Department of Internal Medicine, Hospital U.M. Valdecilla IDIVAL, University of Cantabria, 39008, Santander, Spain
| | - José A Riancho
- Department of Internal Medicine, Hospital U.M. Valdecilla IDIVAL, University of Cantabria, 39008, Santander, Spain.
| |
Collapse
|
46
|
Sun H, Zhao X, Zhang C, Zhang Z, Lun J, Liao W, Zhang Z. MiR-455-3p inhibits the degenerate process of chondrogenic differentiation through modification of DNA methylation. Cell Death Dis 2018; 9:537. [PMID: 29748607 PMCID: PMC5945650 DOI: 10.1038/s41419-018-0565-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/19/2018] [Accepted: 04/05/2018] [Indexed: 12/21/2022]
Abstract
The aim of this work was to determine whether miR-455-3p regulates DNA methylation during chondrogenic differentiation of hMSCs. The expression of miR-455-3p and de novo methyltransferase DNMT3A was assessed in micromass culture of hBMSCs, which induced chondrogenic differentiation in vitro, and in E16.5 mice in vivo. A luciferase reporter assay was used to confirm whether miR-455-3p directly targets DNMT3A by interaction with the 3′-UTR. Using an Illumina Infinium Methylation EPIC microarray, genome-wide DNA methylation of hBMSCs with or without overexpressed miR-455-3p was examined for 28 days during induced chondrogenic differentiation. Here, we showed that miR-455-3p was more expressed during the middle stage of hBMSC chondrogenic differentiation, and less expressed in the late stage. DNMT3A was less expressed in the middle stage and more expressed in the late stage, and was also more expressed in the palms of miR-455-3p deletion mice compared to those of wild-type mice. The luciferase reporter assay demonstrated that miR-455-3p directly targets DNMT3A 3′-UTR. miR-455-3p overexpression inhibits the degenerate process during chondrogenic differentiation, while deletion of miR-455-3p in mice accelerated cartilage degeneration. Genome-wide DNA methylation analysis showed miR-455-3p overexpression regulates DNA methylation of cartilage-specific genes. GO analysis revealed PI3K-Akt signaling pathway was most hypomethylated. Our data show that miR-455-3p can regulate hMSC chondrogenic differentiation by affecting DNA methylation. Overexpression of miR-455-3p and DNA methylation inhibitors can thus potentially be utilized to optimize chondrogenic differentiation.
Collapse
Affiliation(s)
- Hao Sun
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Xiaoyi Zhao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Chengyun Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Ziji Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Jiayong Lun
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Weiming Liao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China.
| | - Zhiqi Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China.
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Growing evidence supports the critical role of transcriptional mechanisms in promoting the spatial and temporal progression of bone healing. In this review, we evaluate and discuss new transcriptional and post-transcriptional regulatory mechanisms of secondary bone repair, along with emerging evidence for epigenetic regulation of fracture healing. RECENT FINDINGS Using the candidate gene approach has identified new roles for several transcription factors in mediating the reactive, reparative, and remodeling phases of fracture repair. Further characterization of the different epigenetic controls of fracture healing and fracture-driven transcriptome changes between young and aged fracture has identified key biological pathways that may yield therapeutic targets. Furthermore, exogenously delivered microRNA to post-transcriptionally control gene expression is quickly becoming an area with great therapeutic potential. Activation of specific transcriptional networks can promote the proper progression of secondary bone healing. Targeting these key factors using small molecules or through microRNA may yield effective therapies to enhance and possibly accelerate fracture healing.
Collapse
Affiliation(s)
- Joseph L Roberts
- Department of Orthopaedics, School of Medicine, Emory University, Atlanta, GA, USA
- Nutrition and Health Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - David N Paglia
- Department of Orthopaedics, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Hicham Drissi
- Department of Orthopaedics, School of Medicine, Emory University, Atlanta, GA, USA.
- Nutrition and Health Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA.
- Atlanta VA Medical Center, 1670 Clairmont Rd, Decatur, GA, 30033, USA.
| |
Collapse
|
48
|
Lien TG, Borgan Ø, Reppe S, Gautvik K, Glad IK. Integrated analysis of DNA-methylation and gene expression using high-dimensional penalized regression: a cohort study on bone mineral density in postmenopausal women. BMC Med Genomics 2018; 11:24. [PMID: 29514638 PMCID: PMC5842543 DOI: 10.1186/s12920-018-0341-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 02/21/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Using high-dimensional penalized regression we studied genome-wide DNA-methylation in bone biopsies of 80 postmenopausal women in relation to their bone mineral density (BMD). The women showed BMD varying from severely osteoporotic to normal. Global gene expression data from the same individuals was available, and since DNA-methylation often affects gene expression, the overall aim of this paper was to include both of these omics data sets into an integrated analysis. METHODS The classical penalized regression uses one penalty, but we incorporated individual penalties for each of the DNA-methylation sites. These individual penalties were guided by the strength of association between DNA-methylations and gene transcript levels. DNA-methylations that were highly associated to one or more transcripts got lower penalties and were therefore favored compared to DNA-methylations showing less association to expression. Because of the complex pathways and interactions among genes, we investigated both the association between DNA-methylations and their corresponding cis gene, as well as the association between DNA-methylations and trans-located genes. Two integrating penalized methods were used: first, an adaptive group-regularized ridge regression, and secondly, variable selection was performed through a modified version of the weighted lasso. RESULTS When information from gene expressions was integrated, predictive performance was considerably improved, in terms of predictive mean square error, compared to classical penalized regression without data integration. We found a 14.7% improvement in the ridge regression case and a 17% improvement for the lasso case. Our version of the weighted lasso with data integration found a list of 22 interesting methylation sites. Several corresponded to genes that are known to be important in bone formation. Using BMD as response and these 22 methylation sites as covariates, least square regression analyses resulted in R2=0.726, comparable to an average R2=0.438 for 10000 randomly selected groups of DNA-methylations with group size 22. CONCLUSIONS Two recent types of penalized regression methods were adapted to integrate DNA-methylation and their association to gene expression in the analysis of bone mineral density. In both cases predictions clearly benefit from including the additional information on gene expressions.
Collapse
Affiliation(s)
- Tonje G. Lien
- University of Oslo, Department of Mathematics, P.O Box 1053, Oslo, 0316 Norway
| | - Ørnulf Borgan
- University of Oslo, Department of Mathematics, P.O Box 1053, Oslo, 0316 Norway
| | - Sjur Reppe
- Oslo University Hospital, Department of Medical Biochemistry, Oslo, Norway
- Lovisenberg Diakonale Hospital, Unger-Vetlesen Institute, Oslo, Norway
| | - Kaare Gautvik
- Lovisenberg Diakonale Hospital, Unger-Vetlesen Institute, Oslo, Norway
- University of Oslo, Institute of Basic Medical Sciences, Oslo, Norway
| | | |
Collapse
|
49
|
Peffers MJ, Balaskas P, Smagul A. Osteoarthritis year in review 2017: genetics and epigenetics. Osteoarthritis Cartilage 2018; 26:304-311. [PMID: 28989115 PMCID: PMC6292677 DOI: 10.1016/j.joca.2017.09.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The purpose of this review is to describe highlights from original research publications related to osteoarthritis (OA), epigenetics and genomics with the intention of recognising significant advances. DESIGN To identify relevant papers a Pubmed literature search was conducted for articles published between April 2016 and April 2017 using the search terms 'osteoarthritis' together with 'genetics', 'genomics', 'epigenetics', 'microRNA', 'lncRNA', 'DNA methylation' and 'histone modification'. RESULTS The search term OA generated almost 4000 references. Publications using the combination of descriptors OA and genetics provided the most references (82 references). However this was reduced compared to the same period in the previous year; 8.1-2.1% (expressed as a percentage of the total publications combining the terms OA and genetics). Publications combining the terms OA with genomics (29 references), epigenetics (16 references), long non-coding RNA (lncRNA) (11 references; including the identification of novel lncRNAs in OA), DNA methylation (21 references), histone modification (3 references) and microRNA (miR) (79 references) were reviewed. Potential OA therapeutics such as histone deacetylase (HDAC) inhibitors have been identified. A number of non-coding RNAs may also provide targets for future treatments. CONCLUSION There continues to be a year on year increase in publications researching miRs in OA (expressed as a percentage of the total publications), with a doubling over the last 4 years. An overview on the last year's progress within the fields of epigenetics and genomics with respect to OA will be given.
Collapse
Affiliation(s)
- M J Peffers
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - P Balaskas
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - A Smagul
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| |
Collapse
|
50
|
Fernandez-Rebollo E, Eipel M, Seefried L, Hoffmann P, Strathmann K, Jakob F, Wagner W. Primary Osteoporosis Is Not Reflected by Disease-Specific DNA Methylation or Accelerated Epigenetic Age in Blood. J Bone Miner Res 2018; 33:356-361. [PMID: 28926142 DOI: 10.1002/jbmr.3298] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/14/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
Osteoporosis is an age-related metabolic bone disease. Hence, osteoporotic patients might suffer from molecular features of accelerated aging, which is generally reflected by specific age-associated DNA methylation (DNAm) changes. In this study, we analyzed genomewide DNAm profiles of peripheral blood from patients with manifest primary osteoporosis and non-osteoporotic controls. Statistical analysis did not reveal any individual CG dinucleotides (CpG sites) with significant aberrant DNAm in osteoporosis. Subsequently, we analyzed if age-associated DNAm patterns are increased in primary osteoporosis (OP). Using three independent age-predictors we did not find any evidence for accelerated epigenetic age in blood of osteoporotic patients. Taken together, osteoporosis is not reflected by characteristic DNAm patterns of peripheral blood that might be used as biomarker for the disease. The prevalence of osteoporosis is age-associated-but it is not associated with premature epigenetic aging in peripheral blood. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Eduardo Fernandez-Rebollo
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University Medical School, Aachen, Germany.,Institute for Biomedical Technology-Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Monika Eipel
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University Medical School, Aachen, Germany.,Institute for Biomedical Technology-Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Lothar Seefried
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Per Hoffmann
- Department of Genomics, Institute of Human Genetics, University of Bonn, Bonn, Germany.,Human Genomics Research Group, Department of Biomedicine, University of Basel, Switzerland
| | - Klaus Strathmann
- Institute for Transfusion Medicine, RWTH Aachen University Medical School, Aachen, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University Medical School, Aachen, Germany.,Institute for Biomedical Technology-Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| |
Collapse
|