1
|
Hajmousa G, de Almeida RC, Bloks N, Ruiz AR, Bouma M, Slieker R, Kuipers TB, Nelissen RGHH, Ito K, Freund C, Ramos YFM, Meulenbelt I. The role of DNA methylation in chondrogenesis of human iPSCs as a stable marker of cartilage quality. Clin Epigenetics 2024; 16:141. [PMID: 39407288 PMCID: PMC11481477 DOI: 10.1186/s13148-024-01759-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Lack of insight into factors that determine purity and quality of human iPSC (hiPSC)-derived neo-cartilage precludes applications of this powerful technology toward regenerative solutions in the clinical setting. Here, we set out to generate methylome-wide landscapes of hiPSC-derived neo-cartilages from different tissues-of-origin and integrated transcriptome-wide data to identify dissimilarities in set points of methylation with associated transcription and the respective pathways in which these genes act. METHODS We applied in vitro chondrogenesis using hiPSCs generated from two different tissue sources: skin fibroblasts and articular cartilage. Upon differentiation toward chondrocytes, these are referred to as hFiCs and hCiC, respectively. Genome-wide DNA methylation and RNA sequencing datasets were generated of the hiPSC-derived neo-cartilages, and the epigenetically regulated transcriptome was compared to that of neo-cartilage deposited by human primary articular cartilage (hPAC). RESULTS Methylome-wide landscapes of neo-cartilages of hiPSCs reprogrammed from two different somatic tissues were 85% similar to that of hPACs. By integration of transcriptome-wide data, differences in transcriptionally active CpGs between hCiC relative to hPAC were prioritized. Among the CpG-gene pairs lower expressed in hCiCs relative to hPACs, we identified genes such as MGP, GDF5, and CHAD enriched in closely related pathways and involved in cartilage development that likely mark phenotypic differences in chondrocyte states. Vice versa, among the CpG-gene pairs higher expressed, we identified genes such as KIF1A or NKX2-2 enriched in neurogenic pathways and likely reflecting off target differentiation. CONCLUSIONS We did not find significant variation between the neo-cartilages derived from hiPSCs of different tissue sources, suggesting that application of a robust differentiation protocol such as we applied here is more important as compared to the epigenetic memory of the cells of origin. Results of our study could be further exploited to improve quality, purity, and maturity of hiPSC-derived neo-cartilage matrix, ultimately to realize introduction of sustainable, hiPSC-derived neo-cartilage implantation into clinical practice.
Collapse
Affiliation(s)
- Ghazaleh Hajmousa
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Niek Bloks
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Alejandro Rodríguez Ruiz
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Marga Bouma
- Department of Anatomy and Embryology and Human iPSC Hotel, 2333 ZA, Leiden, The Netherlands
| | - Roderick Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas B Kuipers
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob G H H Nelissen
- Department of Orthopedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Christian Freund
- Department of Anatomy and Embryology and Human iPSC Hotel, 2333 ZA, Leiden, The Netherlands
| | - Yolande F M Ramos
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
2
|
Castillo H, Hanna P, Sachs LM, Buisine N, Godoy F, Gilbert C, Aguilera F, Muñoz D, Boisvert C, Debiais-Thibaud M, Wan J, Spicuglia S, Marcellini S. Xenopus tropicalis osteoblast-specific open chromatin regions reveal promoters and enhancers involved in human skeletal phenotypes and shed light on early vertebrate evolution. Cells Dev 2024; 179:203924. [PMID: 38692409 DOI: 10.1016/j.cdev.2024.203924] [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: 03/09/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
While understanding the genetic underpinnings of osteogenesis has far-reaching implications for skeletal diseases and evolution, a comprehensive characterization of the osteoblastic regulatory landscape in non-mammalian vertebrates is still lacking. Here, we compared the ATAC-Seq profile of Xenopus tropicalis (Xt) osteoblasts to a variety of non mineralizing control tissues, and identified osteoblast-specific nucleosome free regions (NFRs) at 527 promoters and 6747 distal regions. Sequence analyses, Gene Ontology, RNA-Seq and ChIP-Seq against four key histone marks confirmed that the distal regions correspond to bona fide osteogenic transcriptional enhancers exhibiting a shared regulatory logic with mammals. We report 425 regulatory regions conserved with human and globally associated to skeletogenic genes. Of these, 35 regions have been shown to impact human skeletal phenotypes by GWAS, including one trps1 enhancer and the runx2 promoter, two genes which are respectively involved in trichorhinophalangeal syndrome type I and cleidocranial dysplasia. Intriguingly, 60 osteoblastic NFRs also align to the genome of the elephant shark, a species lacking osteoblasts and bone tissue. To tackle this paradox, we chose to focus on dlx5 because its conserved promoter, known to integrate regulatory inputs during mammalian osteogenesis, harbours an osteoblast-specific NFR in both frog and human. Hence, we show that dlx5 is expressed in Xt and elephant shark odontoblasts, supporting a common cellular and genetic origin of bone and dentine. Taken together, our work (i) unravels the Xt osteogenic regulatory landscape, (ii) illustrates how cross-species comparisons harvest data relevant to human biology and (iii) reveals that a set of genes including bnc2, dlx5, ebf3, mir199a, nfia, runx2 and zfhx4 drove the development of a primitive form of mineralized skeletal tissue deep in the vertebrate lineage.
Collapse
Affiliation(s)
- Héctor Castillo
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile.
| | - Patricia Hanna
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile
| | - Laurent M Sachs
- UMR7221, Physiologie Moléculaire et Adaptation, CNRS, MNHN, Paris Cedex 05, France
| | - Nicolas Buisine
- UMR7221, Physiologie Moléculaire et Adaptation, CNRS, MNHN, Paris Cedex 05, France
| | - Francisco Godoy
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile
| | - Clément Gilbert
- Université Paris-Saclay, CNRS, IRD, UMR Évolution, Génomes, Comportement et Écologie, 12 route 128, 91190 Gif-sur-Yvette, France
| | - Felipe Aguilera
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile
| | - David Muñoz
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile
| | - Catherine Boisvert
- School of Molecular and Life Sciences, Curtin University, Perth, WA, Australia
| | - Mélanie Debiais-Thibaud
- Institut des Sciences de l'Evolution de Montpellier, ISEM, Univ Montpellier, CNRS, IRD, Montpellier, France
| | - Jing Wan
- Aix-Marseille University, INSERM, TAGC, UMR 1090, Marseille, France; Equipe Labelisée LIGUE contre le Cancer, Marseille, France
| | - Salvatore Spicuglia
- Aix-Marseille University, INSERM, TAGC, UMR 1090, Marseille, France; Equipe Labelisée LIGUE contre le Cancer, Marseille, France
| | - Sylvain Marcellini
- Group for the Study of Developmental Processes (GDeP), School of Biological Sciences, University of Concepción, Chile.
| |
Collapse
|
3
|
Huang Y, Zhao H, Wang Y, Bi S, Zhou K, Li H, Zhou C, Wang Y, Wu W, Peng B, Tang J, Pan B, Wang B, Chen Z, Li Z, Zhang Z. The application and progress of tissue engineering and biomaterial scaffolds for total auricular reconstruction in microtia. Front Bioeng Biotechnol 2023; 11:1089031. [PMID: 37811379 PMCID: PMC10556751 DOI: 10.3389/fbioe.2023.1089031] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/21/2023] [Indexed: 10/10/2023] Open
Abstract
Microtia is a congenital deformity of the ear with an incidence of about 0.8-4.2 per 10,000 births. Total auricular reconstruction is the preferred treatment of microtia at present, and one of the core technologies is the preparation of cartilage scaffolds. Autologous costal cartilage is recognized as the best material source for constructing scaffold platforms. However, costal cartilage harvest can lead to donor-site injuries such as pneumothorax, postoperative pain, chest wall scar and deformity. Therefore, with the need of alternative to autologous cartilage, in vitro and in vivo studies of biomaterial scaffolds and cartilage tissue engineering have gradually become novel research hot points in auricular reconstruction research. Tissue-engineered cartilage possesses obvious advantages including non-rejection, minimally invasive or non-invasive, the potential of large-scale production to ensure sufficient donors and controllable morphology. Exploration and advancements of tissue-engineered cartilaginous framework are also emerging in aspects including three-dimensional biomaterial scaffolds, acquisition of seed cells and chondrocytes, 3D printing techniques, inducing factors for chondrogenesis and so on, which has greatly promoted the research process of biomaterial substitute. This review discussed the development, current application and research progress of cartilage tissue engineering in auricular reconstruction, particularly the usage and creation of biomaterial scaffolds. The development and selection of various types of seed cells and inducing factors to stimulate chondrogenic differentiation in auricular cartilage were also highlighted. There are still confronted challenges before the clinical application becomes widely available for patients, and its long-term effect remains to be evaluated. We hope to provide guidance for future research directions of biomaterials as an alternative to autologous cartilage in ear reconstruction, and finally benefit the transformation and clinical application of cartilage tissue engineering and biomaterials in microtia treatment.
Collapse
Affiliation(s)
- Yeqian Huang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hanxing Zhao
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Yixi Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Siwei Bi
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Zhou
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Hairui Li
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Yudong Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqing Wu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Peng
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Jun Tang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Bo Pan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baoyun Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhixing Chen
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhenyu Zhang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Rai MF, Cai L, Zhang Q, Townsend RR, Brophy RH. Synovial Fluid Proteomics From Serial Aspirations of ACL-Injured Knees Identifies Candidate Biomarkers. Am J Sports Med 2023:3635465231169526. [PMID: 37191559 DOI: 10.1177/03635465231169526] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
BACKGROUND Anterior cruciate ligament (ACL) tears often result in knee effusion and an increased risk for developing knee osteoarthritis (OA) in the long run. The molecular profile of these effusions could be informative regarding initial steps in the development of posttraumatic OA after an ACL tear. HYPOTHESIS The proteomics of knee synovial fluid changes over time after ACL injury. STUDY DESIGN Descriptive laboratory study. METHODS Synovial fluid was collected from patients with an acute traumatic ACL tear presenting to the office for evaluation (18.31 ± 19.07 days from injury) (aspiration 1) and again at the time of surgery (35.41 ± 58.15 days after aspiration 1 (aspiration 2). High-resolution liquid chromatography mass spectrometry was used to assess the quantitative protein profile of synovial fluid, and differences in protein profile between the 2 aspirations were determined computationally. RESULTS A total of 58 synovial fluid samples collected from 29 patients (12 male, 17 female; 12 isolated ACL tear, 17 combined ACL and meniscal tear) with a mean age and body mass index of 27.01 ± 12.78 years and 26.30 ± 4.93, respectively, underwent unbiased proteomics analysis. The levels of 130 proteins in the synovial fluid changed over time (87 high, 43 low). Proteins of interest that were significantly higher in aspiration 2 included CRIP1, S100A11, PLS3, POSTN, and VIM, which represent catabolic/inflammatory activities in the joint. Proteins with a known role in chondroprotection and joint homeostasis such as CHI3L2 (YKL-39), TNFAIP6/TSG6, DEFA1, SPP1, and CILP were lower in aspiration 2. CONCLUSION Synovial fluid from knees with ACL tears exhibits an increased burden of inflammatory (catabolic) proteins relevant to OA with reduced levels of chondroprotective (anabolic) proteins. CLINICAL RELEVANCE This study identified a set of novel proteins that provide new biological insights into the aftermath of ACL tears. Elevated inflammation and decreased chondroprotection could represent initial disruption of homeostasis, potentially initiating the development of OA. Longitudinal follow-up and mechanistic studies are necessary to assess the functional role of these proteins in the joint. Ultimately, these investigations could lead to better approaches to predict and possibly improve patient outcomes.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lei Cai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qiang Zhang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - R Reid Townsend
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Kang P, Wu Z, Huang Y, Luo Z, Huo S, Chen Q. Histone H3K9 demethylase JMJD2B/KDM4B promotes osteogenic differentiation of bone marrow-derived mesenchymal stem cells by regulating H3K9me2 on RUNX2. PeerJ 2022; 10:e13862. [PMID: 36217382 PMCID: PMC9547583 DOI: 10.7717/peerj.13862] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 07/18/2022] [Indexed: 01/18/2023] Open
Abstract
Background A variety of proteins including epigenetic factors are involved in the differentiation of human bone marrow mesenchymal stem cells. These cells also exhibited an epigenetic plasticity that enabled them to trans-differentiate from adipocytes to osteoblasts (and vice versa) after commitment. Further in-depth study of their epigenetic alterations may make sense. Methods Chromatin Immunoprecipitation-PCR (ChIP-PCR) was used to detect the methylation enrichment status of H3K9me2 in the Runx2 promoter, alizarin red and alkaline phosphatase (ALP) staining were used to detect osteogenic differentiation and mineralization ability, western blot and quantitative RT-PCR were used to measure the differential expression of osteogenesis-related proteins and genes. Recombinant Lentivirus mediated gain-of-function and loss-of-function study. The scale of epigenetic modification was detected by laser confocal. Results Our results showed that compared with human bone marrow mesenchymal stem cells (hBMSCs) without osteogenic differentiation treatment, hBMSCs after osteogenic differentiation significantly promoted osteogenic differentiation and mRNA expression such as JMJD2B/KDM4B, osteogenesis-related genes like Runx2 and FAM210A in hBMSCs cells, suggesting that upregulation of JMJD2B/KDM4B is involved in the promoting effect of osteogenesis. After overexpression and silencing expression of JMJD2B, we found a completely opposite and significant difference in mRNA expression of osteogenesis-related genes and staining in hBMSCs. Overexpression of JMJD2B/KDM4B significantly promoted osteogenic differentiation, suggesting that JMJD2B/KDM4B could promote osteogenesis. In addition, ChIP-PCR showed that overexpression of JMJD2B/KDM4B significantly reversed the methylation enrichment status of H3K9me2 in Runx2 promoter. Furthermore, overexpression of JMJD2B/KDM4B significantly reverses the inhibitory effect of BIX01294 on H3K9me2, suggesting that JMJD2B/KDM4B regulates the osteogenic differentiation of hBMSCs by changing the methylation status of H3K9me2 at the Runx2 promoter. Conclusions Taken together, these results suggest that JMJD2B/ KDM4B may induce the osteogenic differentiation of hBMSCs by regulating the methylation level of H3K9me2 at the Runx2 promoter.
Collapse
Affiliation(s)
- Pan Kang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiming Wu
- University Medical Center Utrecht, Utrecht, Netherlands
| | - Yuxi Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhen Luo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaochuan Huo
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Qunqun Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China,The Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong Research Institute for Orthopedics & Traumatology of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Timmermans RG, Bloks NG, Tuerlings M, van Hoolwerff M, Nelissen RG, van der Wal RJ, van der Kraan PM, Blom AB, van den Bosch MH, Ramos YF, Meulenbelt I. A human in vitro 3D neo-cartilage model to explore the response of OA risk genes to hyper-physiological mechanical stress. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100231. [DOI: 10.1016/j.ocarto.2021.100231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 10/19/2022] Open
|
7
|
Hulme CH, Perry J, McCarthy HS, Wright KT, Snow M, Mennan C, Roberts S. Cell therapy for cartilage repair. Emerg Top Life Sci 2021; 5:575-589. [PMID: 34423830 PMCID: PMC8589441 DOI: 10.1042/etls20210015] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Regenerative medicine, using cells as therapeutic agents for the repair or regeneration of tissues and organs, offers great hope for the future of medicine. Cell therapy for treating defects in articular cartilage has been an exemplar of translating this technology to the clinic, but it is not without its challenges. These include applying regulations, which were designed for pharmaceutical agents, to living cells. In addition, using autologous cells as the therapeutic agent brings additional costs and logistical challenges compared with using allogeneic cells. The main cell types used in treating chondral or osteochondral defects in joints to date are chondrocytes and mesenchymal stromal cells derived from various sources such as bone marrow, adipose tissue or umbilical cord. This review discusses some of their biology and pre-clinical studies before describing the most pertinent clinical trials in this area.
Collapse
Affiliation(s)
- Charlotte H. Hulme
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, U.K
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, U.K
| | - Jade Perry
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, U.K
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, U.K
| | - Helen S. McCarthy
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, U.K
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, U.K
| | - Karina T. Wright
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, U.K
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, U.K
| | - Martyn Snow
- The Royal Orthopaedic Hospital, Birmingham, U.K
| | - Claire Mennan
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, U.K
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, U.K
| | - Sally Roberts
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, U.K
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, U.K
| |
Collapse
|
8
|
Cartilage from human-induced pluripotent stem cells: comparison with neo-cartilage from chondrocytes and bone marrow mesenchymal stromal cells. Cell Tissue Res 2021; 386:309-320. [PMID: 34241697 PMCID: PMC8557148 DOI: 10.1007/s00441-021-03498-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/23/2021] [Indexed: 11/01/2022]
Abstract
Cartilage has little intrinsic capacity for repair, so transplantation of exogenous cartilage cells is considered a realistic option for cartilage regeneration. We explored whether human-induced pluripotent stem cells (hiPSCs) could represent such unlimited cell sources for neo-cartilage comparable to human primary articular chondrocytes (hPACs) or human bone marrow-derived mesenchymal stromal cells (hBMSCs). For this, chondroprogenitor cells (hiCPCs) and hiPSC-derived mesenchymal stromal cells (hiMSCs) were generated from two independent hiPSC lines and characterized by morphology, flow cytometry, and differentiation potential. Chondrogenesis was compared to hBMSCs and hPACs by histology, immunohistochemistry, and RT-qPCR, while similarities were estimated based on Pearson correlations using a panel of 20 relevant genes. Our data show successful differentiations of hiPSC into hiMSCs and hiCPCs. Characteristic hBMSC markers were shared between hBMSCs and hiMSCs, with the exception of CD146 and CD45. However, neo-cartilage generated from hiMSCs showed low resemblances when compared to hBMSCs (53%) and hPACs (39%) characterized by lower collagen type 2 and higher collagen type 1 expression. Contrarily, hiCPC neo-cartilage generated neo-cartilage more similar to hPACs (65%), with stronger expression of matrix deposition markers. Our study shows that taking a stepwise approach to generate neo-cartilage from hiPSCs via chondroprogenitor cells results in strong similarities to neo-cartilage of hPACs within 3 weeks following chondrogenesis, making them a potential candidate for regenerative therapies. Contrarily, neo-cartilage deposited by hiMSCs seems more prone to hypertrophic characteristics compared to hPACs. We therefore compared chondrocytes derived from hiMSCs and hiCPCs with hPACs and hBMSCs to outline similarities and differences between their neo-cartilage and establish their potential suitability for regenerative medicine and disease modelling.
Collapse
|
9
|
Jauković A, Abadjieva D, Trivanović D, Stoyanova E, Kostadinova M, Pashova S, Kestendjieva S, Kukolj T, Jeseta M, Kistanova E, Mourdjeva M. Specificity of 3D MSC Spheroids Microenvironment: Impact on MSC Behavior and Properties. Stem Cell Rev Rep 2021; 16:853-875. [PMID: 32681232 DOI: 10.1007/s12015-020-10006-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSC) have been considered the promising candidates for the regenerative and personalized medicine due to their self-renewal potential, multilineage differentiation and immunomodulatory capacity. Although these properties have encouraged profound MSC studies in recent years, the majority of research has been based on standard 2D culture utilization. The opportunity to resemble in vivo characteristics of cells native niche has been provided by implementation of 3D culturing models such as MSC spheroid formation assesed through cells self-assembling. In this review, we address the current literature on physical and biochemical features of 3D MSC spheroid microenvironment and their impact on MSC properties and behaviors. Starting with the reduction in the cells' dimensions and volume due to the changes in adhesion molecules expression and cytoskeletal proteins rearrangement resembling native conditions, through the microenvironment shifts in oxygen, nutrients and metabolites gradients and demands, we focus on distinctive and beneficial features of MSC in spheroids compared to cells cultured in 2D conditions. By summarizing the data for 3D MSC spheroids regarding cell survival, pluripotency, differentiation, immunomodulatory activities and potential to affect tumor cells growth we highlighted advantages and perspectives of MSC spheroids use in regenerative medicine. Further detailed analyses are needed to deepen our understanding of mechanisms responsible for modified MSC behavior in spheroids and to set future directions for MSC clinical application.
Collapse
Affiliation(s)
- Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia
| | - Desislava Abadjieva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Drenka Trivanović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia.,IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics, Röntgenring 11, D-97070, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Germany
| | - Elena Stoyanova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Milena Kostadinova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Shina Pashova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Snejana Kestendjieva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia
| | - Michal Jeseta
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Obilní trh 11, 602 00, Brno, Czech Republic.,Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Kamýcká 129, 165 00, Suchdol, Praha 6, Czech Republic
| | - Elena Kistanova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Milena Mourdjeva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria.
| |
Collapse
|
10
|
Ramos YFM, Mobasheri A. MicroRNAs and Regulation of Autophagy in Chondrocytes. Methods Mol Biol 2021; 2245:179-194. [PMID: 33315203 DOI: 10.1007/978-1-0716-1119-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Chondrocytes are the main cells responsible for the maintenance of cartilage homeostasis and integrity. During development, extracellular matrix (ECM) macromolecules are produced and deposited by chondrocyte precursors. Autophagy, a highly dynamic process aimed at degradation of dysfunctional or pathogenic proteins, organelles, and intracellular microbes that can damage tissues, is one of the key processes required for sustained cartilage homeostasis. In different cell types it has been shown that, among others, autophagy is regulated by epigenetic mechanisms such as small noncoding RNAs (miRNAs, ~22 base pairs). Increasing evidence suggests that miRNAs are also involved in the regulation of autophagy in chondrocytes. Based on our previous research of gene and miRNA expression in articular cartilage, in this chapter we provide a summary of the tools models to direct in vitro and in vivo studies aimed at gaining a better understanding of the regulatory roles of miRNAs in chondrocyte autophagy.
Collapse
Affiliation(s)
- Yolande F M Ramos
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Queen's Medical Centre, Nottingham, UK.,Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| |
Collapse
|
11
|
van Hoolwerff M, Metselaar PI, Tuerlings M, Suchiman HED, Lakenberg N, Ramos YFM, Cats D, Nelissen RGHH, Broekhuis D, Mei H, de Almeida RC, Meulenbelt I. Elucidating Epigenetic Regulation by Identifying Functional cis-Acting Long Noncoding RNAs and Their Targets in Osteoarthritic Articular Cartilage. Arthritis Rheumatol 2020; 72:1845-1854. [PMID: 32840049 PMCID: PMC7702083 DOI: 10.1002/art.41396] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/02/2020] [Indexed: 12/23/2022]
Abstract
Objective To identify robustly differentially expressed long noncoding RNAs (lncRNAs) with osteoarthritis (OA) pathophysiology in cartilage and to explore potential target messenger RNA (mRNA) by establishing coexpression networks, followed by functional validation. Methods RNA sequencing was performed on macroscopically lesioned and preserved OA cartilage from patients who underwent joint replacement surgery due to OA (n = 98). Differential expression analysis was performed on lncRNAs that were annotated in GENCODE and Ensembl databases. To identify potential interactions, correlations were calculated between the identified differentially expressed lncRNAs and the previously reported differentially expressed protein‐coding genes in the same samples. Modulation of chondrocyte lncRNA expression was achieved using locked nucleic acid GapmeRs. Results By applying our in‐house pipeline, we identified 5,053 lncRNAs that were robustly expressed, of which 191 were significantly differentially expressed (according to false discovery rate) between lesioned and preserved OA cartilage. Upon integrating mRNA sequencing data, we showed that intergenic and antisense differentially expressed lncRNAs demonstrate high, positive correlations with their respective flanking sense genes. To functionally validate this observation, we selected P3H2‐AS1, which was down‐regulated in primary chondrocytes, resulting in the down‐regulation of P3H2 gene expression levels. As such, we can confirm that P3H2‐AS1 regulates its sense gene P3H2. Conclusion By applying an improved detection strategy, robustly differentially expressed lncRNAs in OA cartilage were detected. Integration of these lncRNAs with differential mRNA expression levels in the same samples provided insight into their regulatory networks. Our data indicate that intergenic and antisense lncRNAs play an important role in regulating the pathophysiology of OA.
Collapse
Affiliation(s)
| | | | | | | | - Nico Lakenberg
- Leiden University Medical Center, Leiden, The Netherlands
| | | | - Davy Cats
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Hailiang Mei
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
12
|
Zhao W, Zou T, Cui H, Lv Y, Gao D, Ruan C, Zhang X, Zhang Y. Parathyroid hormone (1-34) promotes the effects of 3D printed scaffold-seeded bone marrow mesenchymal stem cells on meniscus regeneration. Stem Cell Res Ther 2020; 11:328. [PMID: 32731897 PMCID: PMC7394673 DOI: 10.1186/s13287-020-01845-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/01/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Background Cell-based tissue engineering represents a promising management for meniscus repair and regeneration. The present study aimed to investigate whether the injection of parathyroid hormone (PTH) (1-34) could promote the regeneration and chondroprotection of 3D printed scaffold seeded with bone marrow mesenchymal stem cells (BMSCs) in a canine total meniscal meniscectomy model. Methods 3D printed poly(e-caprolactone) scaffold seeded with BMSCs was cultured in vitro, and the effects of in vitro culture time on cell growth and matrix synthesis of the BMSCs–scaffold construct were evaluated by microscopic observation and cartilage matrix content detection at 7, 14, 21, and 28 days. After that, the tissue-engineered meniscus based on BMSCs–scaffold cultured for the appropriate culture time was selected for in vivo implantation. Sixteen dogs were randomly divided into four groups: PTH + BMSCs–scaffold, BMSCs–scaffold, total meniscectomy, and sham operation. The regeneration of the implanted tissue and the degeneration of articular cartilage were assessed by gross, histological, and immunohistochemical analysis at 12 weeks postoperatively. Results In vitro study showed that the glycosaminoglycan (GAG)/DNA ratio and the expression of collagen type II (Col2) were significantly higher on day 21 as compared to the other time points. In vivo study showed that, compared with the BMSCs–scaffold group, the PTH + BMSCs–scaffold group showed better regeneration of the implanted tissue and greater similarity to native meniscus concerning gross appearance, cell composition, and cartilage extracellular matrix deposition. This group also showed less expression of terminal differentiation markers of BMSC chondrogenesis as well as lower cartilage degeneration with less damage on the knee cartilage surface, higher expression of Col2, and lower expression of degeneration markers. Conclusions Our results demonstrated that PTH (1-34) promotes the regenerative and chondroprotective effects of the BMSCs–3D printed meniscal scaffold in a canine model, and thus, their combination could be a promising strategy for meniscus tissue engineering.
Collapse
Affiliation(s)
- Wen Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Tong Zou
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hao Cui
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yangou Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Dengke Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chenmei Ruan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xia Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yihua Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
13
|
van den Bosch MHJ, Ramos YFM, den Hollander W, Bomer N, Nelissen RGHH, Bovée JVMG, van den Berg WB, van Lent PLEM, Blom AB, van der Kraan PM, Meulenbelt I. Increased WISP1 expression in human osteoarthritic articular cartilage is epigenetically regulated and decreases cartilage matrix production. Rheumatology (Oxford) 2020; 58:1065-1074. [PMID: 30649473 DOI: 10.1093/rheumatology/key426] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/21/2018] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Previously, we have shown the involvement of Wnt-activated protein Wnt-1-induced signaling protein 1 (WISP1) in the development of OA in mice. Here, we aimed to characterize the relation between WISP1 expression and human OA and its regulatory epigenetic determinants. METHODS Preserved and lesioned articular cartilage from end-stage OA patients and non-OA-diagnosed individuals was collected. WISP1 expression was determined using immunohistochemistry and damage was classified using Mankin scoring. RNA expression and DNA methylation were assessed in silico from genome-wide datasets (microarray analysis and RNA sequencing, and 450 k-methylationarrays, respectively). Effects of WISP1 were tested in pellet cultures of primary human chondrocytes. RESULTS WISP1 expression in cartilage of OA patients was increased compared with non-OA-diagnosed controls and, within OA patients, WISP1 was even higher in lesioned compared with preserved regions, with expression strongly correlating with Mankin score. In early symptomatic OA patients with disease progression, higher synovial WISP1 expression was observed as compared with non-progressors. Notably, increased WISP1 expression was inversely correlated with methylation levels of a positional CpG-dinucleotide (cg10191240), with lesioned areas showing strong hypomethylation for this CpG as compared with preserved cartilage. Additionally, we observed that methylation levels were allele-dependent for an intronic single-nucleotide polymorphism nearby cg10191240. Finally, addition of recombinant WISP1 to pellets of primary chondrocytes strongly inhibited deposition of extracellular matrix as reflected by decreased pellet circumference, proteoglycan content and decreased expression of matrix components. CONCLUSION Increased WISP1 expression is found in lesioned human articular cartilage, and appears epigenetically regulated via DNA methylation. In vitro assays suggest that increased WISP1 is detrimental for cartilage integrity.
Collapse
Affiliation(s)
| | - Yolande F M Ramos
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter den Hollander
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nils Bomer
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob G H H Nelissen
- Department of Orthopedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wim B van den Berg
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ingrid Meulenbelt
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
DNA hypomethylation during MSC chondrogenesis occurs predominantly at enhancer regions. Sci Rep 2020; 10:1169. [PMID: 31980739 PMCID: PMC6981252 DOI: 10.1038/s41598-020-58093-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022] Open
Abstract
Regulation of transcription occurs in a cell type specific manner orchestrated by epigenetic mechanisms including DNA methylation. Methylation changes may also play a key role in lineage specification during stem cell differentiation. To further our understanding of epigenetic regulation in chondrocytes we characterised the DNA methylation changes during chondrogenesis of mesenchymal stem cells (MSCs) by Infinium 450 K methylation array. Significant DNA hypomethylation was identified during chondrogenic differentiation including changes at many key cartilage gene loci. Integration with chondrogenesis gene expression data revealed an enrichment of significant CpGs in upregulated genes, while characterisation of significant CpG loci indicated their predominant localisation to enhancer regions. Comparison with methylation profiles of other tissues, including healthy and diseased adult cartilage, identified chondrocyte-specific regions of hypomethylation and the overlap with differentially methylated CpGs in osteoarthritis. Taken together we have associated DNA methylation levels with the chondrocyte phenotype. The consequences of which has potential to improve cartilage generation for tissue engineering purposes and also to provide context for observed methylation changes in cartilage diseases such as osteoarthritis.
Collapse
|
15
|
Zhao Z, Fan C, Chen F, Sun Y, Xia Y, Ji A, Wang DA. Progress in Articular Cartilage Tissue Engineering: A Review on Therapeutic Cells and Macromolecular Scaffolds. Macromol Biosci 2019; 20:e1900278. [PMID: 31800166 DOI: 10.1002/mabi.201900278] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Repair and regeneration of articular cartilage lesions have always been a major challenge in the medical field due to its peculiar structure (e.g., sparsely distributed chondrocytes, no blood supply, no nerves). Articular cartilage tissue engineering is considered as one promising strategy to achieve reconstruction of cartilage. With this perspective, the articular cartilage tissue engineering has been widely studied. Here, the recent progress of articular cartilage tissue engineering is reviewed. The ad hoc therapeutic cells and growth factors for cartilage regeneration are summarized and discussed. Various types of bio/macromolecular scaffolds together with their pros and cons are also reviewed and elaborated.
Collapse
Affiliation(s)
- Zhongyi Zhao
- Department of Traumatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changjiang Fan
- Department of Human Anatomy, Histology and Embryology, College of Medicine, Qingdao University, Qingdao, 266021, China.,Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, P. R. China
| | - Feng Chen
- Department of Traumatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yutai Sun
- School of Information Engineering, Shandong Vocational College of Science & Technology, Weifang, 261053, P. R. China
| | - Yujun Xia
- Department of Human Anatomy, Histology and Embryology, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Aiyu Ji
- Department of Traumatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong SAR
| |
Collapse
|
16
|
Kiapour AM, Sieker JT, Proffen BL, Lam TT, Fleming BC, Murray MM. Synovial fluid proteome changes in ACL injury-induced posttraumatic osteoarthritis: Proteomics analysis of porcine knee synovial fluid. PLoS One 2019; 14:e0212662. [PMID: 30822327 PMCID: PMC6396923 DOI: 10.1371/journal.pone.0212662] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 02/07/2019] [Indexed: 01/26/2023] Open
Abstract
Surgical transection of the anterior cruciate ligament (ACL) in the porcine model leads to posttraumatic osteoarthritis if left untreated. However, a recently developed surgical treatment, bridge-enhanced ACL repair, prevents further cartilage damage. Since the synovial fluid bathes all the intrinsic structures of knee, we reasoned that a comparative analysis of synovial fluid protein contents could help to better understand the observed chondroprotective effects of the bridge-enhanced ACL repair. We hypothesized that post-surgical changes in the synovial fluid proteome would be different in the untreated and repaired knees, and those changes would correlate with the degree of cartilage damage. Thirty adolescent Yucatan mini-pigs underwent unilateral ACL transection and were randomly assigned to either no further treatment (ACLT, n = 14) or bridge-enhanced ACL repair (BEAR, n = 16). We used an isotopically labeled high resolution LC MS/MS-based proteomics approach to analyze the protein profile of synovial fluid at 6 and 12 months after ACL transection in untreated and repaired porcine knees. A linear mixed effect model was used to compare the normalized protein abundance levels between the groups at each time point. Bivariate linear regression analyses were used to assess the correlations between the macroscopic cartilage damage (total lesion area) and normalized abundance levels of each of the identified secreted proteins. There were no significant differences in cartilage lesion area or quantitative abundance levels of the secreted proteins between the ACLT and BEAR groups at 6 months. However, by 12 months, greater cartilage damage was seen in the ACLT group compared to the BEAR group (p = 0.005). This damage was accompanied by differences in the abundance levels of secreted proteins, with higher levels of Vitamin K-dependent protein C (p = 0.001), and lower levels of Apolipoprotein A4 (p = 0.021) and Cartilage intermediate layer protein 1 (p = 0.049) in the ACLT group compared to the BEAR group. There were also group differences in the secreted proteins that significantly changed in abundance between 6 and 12 months in ACLT and BEAR knees. Increased concentration of Ig lambda-1 chain C regions and decreased concentration of Hemopexin, Clusterin, Coagulation factor 12 and Cartilage intermediate layer protein 1 were associated with greater cartilage lesion area. In general, ACLT knees had higher concentrations of pro-inflammatory proteins and lower concentrations of anti-inflammatory proteins than BEAR group. In addition, the ACLT group had a lower and declining synovial concentrations of CILP, in contrast to a consistently high abundance of CILP in repaired knees. These differences suggest that the knees treated with bridge-enhanced ACL repair may be maintaining an environment that is more protective of the extracellular matrix, a function which is not seen in the ACLT knees.
Collapse
Affiliation(s)
- Ata M. Kiapour
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States of America
- * E-mail:
| | - Jakob T. Sieker
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Benedikt L. Proffen
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - TuKiet T. Lam
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, United States of America
- MS & Proteomics Resource, W.M. Keck Biotechnology Resource Laboratory, Yale University, New Haven, CT, United States of America
| | - Braden C. Fleming
- Department of Orthopaedics, Warren Alpert Medical School of Brown University & Rhode Island Hospital, Providence, RI, United States of America
| | - Martha M. Murray
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
17
|
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: 9.0] [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
|
18
|
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: 5.2] [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
|
19
|
Naranda J, Gradišnik L, Gorenjak M, Vogrin M, Maver U. Isolation and characterization of human articular chondrocytes from surgical waste after total knee arthroplasty (TKA). PeerJ 2017; 5:e3079. [PMID: 28344902 PMCID: PMC5363257 DOI: 10.7717/peerj.3079] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/09/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Cartilage tissue engineering is a fast-evolving field of biomedical engineering, in which the chondrocytes represent the most commonly used cell type. Since research in tissue engineering always consumes a lot of cells, simple and cheap isolation methods could form a powerful basis to boost such studies and enable their faster progress to the clinics. Isolated chondrocytes can be used for autologous chondrocyte implantation in cartilage repair, and are the base for valuable models to investigate cartilage phenotype preservation, as well as enable studies of molecular features, nature and scales of cellular responses to alterations in the cartilage tissue. METHODS Isolation and consequent cultivation of primary human adult articular chondrocytes from the surgical waste obtained during total knee arthroplasty (TKA) was performed. To evaluate the chondrogenic potential of the isolated cells, gene expression of collagen type 2 (COL2), collagen 1 (COL1) and aggrecan (ACAN) was evaluated. Immunocytochemical staining of all mentioned proteins was performed to evaluate chondrocyte specific production. RESULTS Cartilage specific gene expression of COL2 and ACAN has been shown that the proposed protocol leads to isolation of cells with a high chondrogenic potential, possibly even specific phenotype preservation up to the second passage. COL1 expression has confirmed the tendency of the isolated cells dedifferentiation into a fibroblast-like phenotype already in the second passage, which confirms previous findings that higher passages should be used with care in cartilage tissue engineering. To evaluate the effectiveness of our approach, immunocytochemical staining of the evaluated chondrocyte specific products was performed as well. DISCUSSION In this study, we developed a protocol for isolation and consequent cultivation of primary human adult articular chondrocytes with the desired phenotype from the surgical waste obtained during TKA. TKA is a common and very frequently performed orthopaedic surgery during which both femoral condyles are removed. The latter present the ideal source for a simple and relatively cheap isolation of chondrocytes as was confirmed in our study.
Collapse
Affiliation(s)
- Jakob Naranda
- Department of Orthopaedics, University Medical Centre Maribor, Maribor, Slovenia
| | - Lidija Gradišnik
- Institute of Biomedical Sciences, University of Maribor, Faculty of Medicine, Maribor, Slovenia
| | - Mario Gorenjak
- Center for Human Molecular Genetics and Pharmacogenomics, University of Maribor, Faculty of Medicine, Maribor, Slovenia
| | - Matjaž Vogrin
- Department of Orthopaedics, University Medical Centre Maribor, Maribor, Slovenia
| | - Uroš Maver
- Institute of Biomedical Sciences, University of Maribor, Faculty of Medicine, Maribor, Slovenia
| |
Collapse
|