1
|
Qianman B, Jiasharete T, Badalihan A, Mamately A, Yeerbo N, Bahesutihan Y, Wupuer A, Aisaiding A, Wuerliebieke J, Jialihasi A, Li P, Jielile J. iTRAQ-Based Proteomic Analysis of Spontaneous Achilles Tendon Rupture. J Proteome Res 2024. [PMID: 39601082 DOI: 10.1021/acs.jproteome.4c00357] [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: 11/29/2024]
Abstract
Spontaneous Achilles tendon rupture (SATR) predominantly affects middle-aged and elderly individuals with chronic injuries. However, the exact cause and mechanism of SATR remain elusive, and potential therapeutic intervention or prevention is still insufficient. The present study aimed to uncover the key pathological molecules by using iTRAQ proteomics. The results identified 2432 candidate proteins in SATR patients using iTRAQ proteomic analysis. A total of 307 differentially expressed proteins (DEPs) were identified and linked to 211 KEGG signaling pathways including Coronavirus disease (COVID-19), focal adhesion, and ribosomes. GO enrichment analysis highlighted significant enrichment in processes such as biological adhesion, ossification, lipid (APOA4) processes, and extracellular matrix (ECM) organization (collagen). PPI network analysis identified hub genes such as serum albumin (ALB), fibronectin (FN1), and actin cytoplasmic 1. The WB analysis confirmed that FN1 and the receptor for activated C kinase (RACK1) were downregulated in the SATR tendon. Immunohistochemical staining revealed that collagen I and III were suppressed, while collagen II and APOA4 expression were higher in the SATR pathological tissue (P < 0.05). However, the primary cultured tenocytes (PCTs) from SATR patients showed enhanced proliferation and, consistent with tissue staining, reduced collagen I and III and increased collagen II. Our findings reveal vital targets and pathways in SATR's etiological progression, offering a new perspective on the diagnosis, treatment, and prognosis of this complex disorder.
Collapse
Affiliation(s)
- Bayixiati Qianman
- Department of Osteopathy and Orthopedics (Ankle) Surgery, The Sixth Teaching Hospital of Xinjiang Medical University, No. 39 Wuxing South Road, Urumqi 830001, Xinjiang Uygur Autonomous Region, China
| | - Tuomilisi Jiasharete
- Department of Clinical Medicine, Capital Medical University Class of 2024, Five-Year Clinical Medicine Class 3, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Ayinazi Badalihan
- Department of Orthopedics, Xinjiang Urumqi International and Otorhinolaryngological Hospital, No. 50 Lianhu Road, Toutunhe District Urumqi, Urumqi 830000, Xinjiang, China
| | - Abuduhilil Mamately
- Department of Orthopedic Centre, The Fifth Affiliated Hospital of Xinjiang Medical University, No. 118 West Henan Road, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Naertai Yeerbo
- Department of Geriatric Joint Surgery of Orthopedics, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi 830001, China
| | - Yemenlehan Bahesutihan
- Department of Orthopedics Surgery, Xinjiang 474 Hospital, No. 754, Beijing Middle Road, Beijing North Road, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Aikeremu Wupuer
- Department of Osteopathy and Orthopedics (Ankle) Surgery, The Sixth Teaching Hospital of Xinjiang Medical University, No. 39 Wuxing South Road, Urumqi 830001, Xinjiang Uygur Autonomous Region, China
| | - Amuding Aisaiding
- Department of Hand and Foot Microsurgery, Xinjiang Uygur Autonomous Region Children's Hospital, No. 393 Altay Road, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Jianati Wuerliebieke
- Department of Orthopedics, People's Hospital of Altay Region, No. 31, Park Road, Altay, Xinjiang 836500, China
| | - Ayidaer Jialihasi
- Department of Orthopedics, Almaty Medical Center HAK, Kazakhstan, No. 11a Otegenbater Road, Almaty 050063, Kazakhstan
| | - Ping Li
- Department of Microrepair and Reconstruction of Orthopedics Centre, The First Teaching Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
| | - Jiasharete Jielile
- Department of Osteopathy and Orthopedics (Ankle) Surgery, The Sixth Teaching Hospital of Xinjiang Medical University, No. 39 Wuxing South Road, Urumqi 830001, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
2
|
Asomugha E, Cho Y, Paudel S, Guo Y, Schon L, Zhang Z. Localization of advanced glycation end-products and their receptor in tendinopathic lesions. Histol Histopathol 2024; 39:1209-1215. [PMID: 38323686 DOI: 10.14670/hh-18-712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
This study was designed to investigate the accumulation of advanced glycation end-products (AGEs) and the expression of the receptor of AGEs (RAGE) in tendinopathic tissues. In this study, tendinopathic posterior tibial tendons (PTT) were collected from patients (n=6). Redundant autografts of flexor digitorum longus tendon (FDL; n=3) were used for controls. The control and tendinopathic tendon tissues were used for extraction of proteins for western blot and sectioned for histology and immunohistochemistry. Tendinopathy of the PTT was confirmed histologically by the presentation of disorderly organized collagen fibers, high cellularity and increased vascularity. By immunohistochemistry, heterogeneous accumulation of AGEs was detected on the PTT sections and concentrated in areas, where collagen fibers were disorderly and tangled. In the PTT, roundish tenocytes were also AGEs-positive. In contrast, AGEs were diffuse, lightly stained in the FDL. A greater number of tenocytes within the tendinopathic lesions in the PTT were RAGE positive, compared to the tenocytes in the FDL. Western blot confirmed the expression of AGEs and RAGE in both tendinopathic PTT and control FDL but their band densities were not significantly different. The spatial relation of the accumulated AGEs and RAGE- positive tenocytes within the tendinopathic lesions indicates their involvement in the molecular pathology of tendinopathy.
Collapse
Affiliation(s)
| | - Young Cho
- Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Sharada Paudel
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Yi Guo
- Department of Orthopaedic Surgery, Montefiore Medical Center, Bronx, NY, USA
| | - Lew Schon
- Institute for Foot and Ankle Reconstruction, Mercy Medical Center, Baltimore, MD, USA
- Center for Orthopaedic Innovation, Mercy Medical Center, Baltimore, MD, USA
| | - Zijun Zhang
- Center for Orthopaedic Innovation, Mercy Medical Center, Baltimore, MD, USA.
| |
Collapse
|
3
|
Schmidt L, Saynisch M, Hoegsbjerg C, Schmidt A, Mackey A, Lackmann JW, Müller S, Koch M, Brachvogel B, Kjaer M, Antczak P, Krüger M. Spatial proteomics of skeletal muscle using thin cryosections reveals metabolic adaptation at the muscle-tendon transition zone. Cell Rep 2024; 43:114374. [PMID: 38900641 DOI: 10.1016/j.celrep.2024.114374] [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: 12/07/2023] [Revised: 04/05/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024] Open
Abstract
Morphological studies of skeletal muscle tissue provide insights into the architecture of muscle fibers, the surrounding cells, and the extracellular matrix (ECM). However, a spatial proteomics analysis of the skeletal muscle including the muscle-tendon transition zone is lacking. Here, we prepare cryotome muscle sections of the mouse soleus muscle and measure each slice using short liquid chromatography-mass spectrometry (LC-MS) gradients. We generate 3,000 high-resolution protein profiles that serve as the basis for a network analysis to reveal the complex architecture of the muscle-tendon junction. Among the protein profiles that increase from muscle to tendon, we find proteins related to neuronal activity, fatty acid biosynthesis, and the renin-angiotensin system (RAS). Blocking the RAS in cultured mouse tenocytes using losartan reduces the ECM synthesis. Overall, our analysis of thin cryotome sections provides a spatial proteome of skeletal muscle and reveals that the RAS acts as an additional regulator of the matrix within muscle-tendon junctions.
Collapse
Affiliation(s)
- Luisa Schmidt
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Michael Saynisch
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Christian Hoegsbjerg
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark; Part of IOC Research Center Copenhagen and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Abigail Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark; Part of IOC Research Center Copenhagen and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan-Wilm Lackmann
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Stefan Müller
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Bent Brachvogel
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty, University of Cologne, Cologne, Germany; Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark; Part of IOC Research Center Copenhagen and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Philipp Antczak
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany.
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
4
|
Johnson BB, Cosson MV, Tsansizi LI, Holmes TL, Gilmore T, Hampton K, Song OR, Vo NTN, Nasir A, Chabronova A, Denning C, Peffers MJ, Merry CLR, Whitelock J, Troeberg L, Rushworth SA, Bernardo AS, Smith JGW. Perlecan (HSPG2) promotes structural, contractile, and metabolic development of human cardiomyocytes. Cell Rep 2024; 43:113668. [PMID: 38198277 DOI: 10.1016/j.celrep.2023.113668] [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: 05/22/2023] [Revised: 11/01/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Perlecan (HSPG2), a heparan sulfate proteoglycan similar to agrin, is key for extracellular matrix (ECM) maturation and stabilization. Although crucial for cardiac development, its role remains elusive. We show that perlecan expression increases as cardiomyocytes mature in vivo and during human pluripotent stem cell differentiation to cardiomyocytes (hPSC-CMs). Perlecan-haploinsuffient hPSCs (HSPG2+/-) differentiate efficiently, but late-stage CMs have structural, contractile, metabolic, and ECM gene dysregulation. In keeping with this, late-stage HSPG2+/- hPSC-CMs have immature features, including reduced ⍺-actinin expression and increased glycolytic metabolism and proliferation. Moreover, perlecan-haploinsuffient engineered heart tissues have reduced tissue thickness and force generation. Conversely, hPSC-CMs grown on a perlecan-peptide substrate are enlarged and display increased nucleation, typical of hypertrophic growth. Together, perlecan appears to play the opposite role of agrin, promoting cellular maturation rather than hyperplasia and proliferation. Perlecan signaling is likely mediated via its binding to the dystroglycan complex. Targeting perlecan-dependent signaling may help reverse the phenotypic switch common to heart failure.
Collapse
Affiliation(s)
- Benjamin B Johnson
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Marie-Victoire Cosson
- The Francis Crick Institute, London NW1 1AT, UK; NHLI, Imperial College London, London, UK
| | - Lorenza I Tsansizi
- The Francis Crick Institute, London NW1 1AT, UK; NHLI, Imperial College London, London, UK
| | - Terri L Holmes
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | | | - Katherine Hampton
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Ok-Ryul Song
- The Francis Crick Institute, London NW1 1AT, UK; High-Throughput Screening Science Technology Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - Nguyen T N Vo
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Aishah Nasir
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Alzbeta Chabronova
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Chris Denning
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Mandy J Peffers
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Catherine L R Merry
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK; Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - John Whitelock
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK; Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Linda Troeberg
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Stuart A Rushworth
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Andreia S Bernardo
- The Francis Crick Institute, London NW1 1AT, UK; NHLI, Imperial College London, London, UK.
| | - James G W Smith
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK.
| |
Collapse
|
5
|
Kwan KYC, Ng KWK, Rao Y, Zhu C, Qi S, Tuan RS, Ker DFE, Wang DM. Effect of Aging on Tendon Biology, Biomechanics and Implications for Treatment Approaches. Int J Mol Sci 2023; 24:15183. [PMID: 37894875 PMCID: PMC10607611 DOI: 10.3390/ijms242015183] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/07/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Tendon aging is associated with an increasing prevalence of tendon injuries and/or chronic tendon diseases, such as tendinopathy, which affects approximately 25% of the adult population. Aged tendons are often characterized by a reduction in the number and functionality of tendon stem/progenitor cells (TSPCs), fragmented or disorganized collagen bundles, and an increased deposition of glycosaminoglycans (GAGs), leading to pain, inflammation, and impaired mobility. Although the exact pathology is unknown, overuse and microtrauma from aging are thought to be major causative factors. Due to the hypovascular and hypocellular nature of the tendon microenvironment, healing of aged tendons and related injuries is difficult using current pain/inflammation and surgical management techniques. Therefore, there is a need for novel therapies, specifically cellular therapy such as cell rejuvenation, due to the decreased regenerative capacity during aging. To augment the therapeutic strategies for treating tendon-aging-associated diseases and injuries, a comprehensive understanding of tendon aging pathology is needed. This review summarizes age-related tendon changes, including cell behaviors, extracellular matrix (ECM) composition, biomechanical properties and healing capacity. Additionally, the impact of conventional treatments (diet, exercise, and surgery) is discussed, and recent advanced strategies (cell rejuvenation) are highlighted to address aged tendon healing. This review underscores the molecular and cellular linkages between aged tendon biomechanical properties and the healing response, and provides an overview of current and novel strategies for treating aged tendons. Understanding the underlying rationale for future basic and translational studies of tendon aging is crucial to the development of advanced therapeutics for tendon regeneration.
Collapse
Affiliation(s)
- Ka Yu Carissa Kwan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ka Wai Kerry Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chenxian Zhu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shengcai Qi
- Department of Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200040, China;
| | - Rocky S. Tuan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dai Fei Elmer Ker
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
6
|
Chabronova A, van den Akker G, Housmans BAC, Caron MMJ, Cremers A, Surtel DAM, Peffers MJ, van Rhijn LW, Marchand V, Motorin Y, Welting TJM. Depletion of SNORA33 Abolishes ψ of 28S-U4966 and Affects the Ribosome Translational Apparatus. Int J Mol Sci 2023; 24:12578. [PMID: 37628759 PMCID: PMC10454564 DOI: 10.3390/ijms241612578] [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: 07/05/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Eukaryotic ribosomes are complex molecular nanomachines translating genetic information from mRNAs into proteins. There is natural heterogeneity in ribosome composition. The pseudouridylation (ψ) of ribosomal RNAs (rRNAs) is one of the key sources of ribosome heterogeneity. Nevertheless, the functional consequences of ψ-based ribosome heterogeneity and its relevance for human disease are yet to be understood. Using HydraPsiSeq and a chronic disease model of non-osteoarthritic primary human articular chondrocytes exposed to osteoarthritic synovial fluid, we demonstrated that the disease microenvironment is capable of instigating site-specific changes in rRNA ψ profiles. To investigate one of the identified differential rRNA ψ sites (28S-ψ4966), we generated SNORA22 and SNORA33 KO SW1353 cell pools using LentiCRISPRv2/Cas9 and evaluated the ribosome translational capacity by 35S-Met/Cys incorporation, assessed the mode of translation initiation and ribosomal fidelity using dual luciferase reporters, and assessed cellular and ribosomal proteomes by LC-MS/MS. We uncovered that the depletion of SNORA33, but not SNORA22, reduced 28S-ψ4966 levels. The resulting loss of 28S-ψ4966 affected ribosomal protein composition and function and led to specific changes in the cellular proteome. Overall, our pioneering findings demonstrate that cells dynamically respond to disease-relevant changes in their environment by altering their rRNA pseudouridylation profiles, with consequences for ribosome function and the cellular proteome relevant to human disease.
Collapse
Affiliation(s)
- Alzbeta Chabronova
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Guus van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Bas A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Andy Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Don A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Mandy J Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7TX, UK
| | - Lodewijk W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Virginie Marchand
- UAR2008 IBSLor CNRS-INSERM-Université de Lorraine, F54000 Nancy, France
| | - Yuri Motorin
- UAR2008 IBSLor CNRS-INSERM-Université de Lorraine, F54000 Nancy, France
- UMR7365 IMOPA, CNRS-Université de Lorraine, F54000 Nancy, France
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, 6229 HX Maastricht, The Netherlands
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+ (MUMC+), 6229 HX Maastricht, The Netherlands
| |
Collapse
|
7
|
Stassen RHMJ, van den Akker GGH, Surtel DAM, Housmans BAC, Cremers A, Caron MMJ, Smagul A, Peffers MJ, van Rhijn LW, Welting TJM. Unravelling the Basic Calcium Phosphate crystal-dependent chondrocyte protein secretome; a role for TGF-β signaling. Osteoarthritis Cartilage 2023; 31:1035-1046. [PMID: 37075856 DOI: 10.1016/j.joca.2023.02.079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 04/21/2023]
Abstract
OBJECTIVE Basic Calcium Phosphate (BCP) crystals play an active role in the progression of osteoarthritis (OA). However, the cellular consequences remain largely unknown. Therefore, we characterized for the first time the changes in the protein secretome of human OA articular chondrocytes as a result of BCP stimulation using two unbiased proteomic analysis methods. METHOD Isolated human OA articular chondrocytes were stimulated with BCP crystals and examined by Quantitative Reverse Transcription PCR (RT-qPCR) and enzyme-linked immune sorbent assay (ELISA) after twenty-four and forty-eight hours. Forty-eight hours conditioned media were analyzed by label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) and an antibody array. The activity of BCP dependent Transforming Growth Factor Beta (TGF-β) signaling was analyzed by RT-qPCR and luciferase reporter assays. The molecular consequences regarding BCP-dependent TGF-β signaling on BCP-dependent Interleukin 6 (IL-6) were investigated using specific pathway inhibitors. RESULTS Synthesized BCP crystals induced IL-6 expression and secretion upon stimulation of human articular chondrocytes. Concomitant induction of catabolic gene expression was observed. Analysis of conditioned media revealed a complex and diverse response with a large number of proteins involved in TGF-β signaling, both in activation of latent TGF-β and TGF-β superfamily members, which were increased compared to non-stimulated OA chondrocytes. Activity of this BCP driven TGF-β signaling was confirmed by increased activity of expression of TGF-β target genes and luciferase reporters. Inhibition of BCP driven TGF-β signaling resulted in decreased IL-6 expression and secretion with a moderate effect on catabolic gene expression. CONCLUSION BCP crystal stimulation resulted in a complex and diverse chondrocyte protein secretome response. An important role for BCP-dependent TGF-β signaling was identified in development of a pro-inflammatory environment.
Collapse
Affiliation(s)
- R H M J Stassen
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - G G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - D A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - B A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - A Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - M M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - A Smagul
- Department of Musculoskeletal Biology, Life Course and Medical Sciences, University of Liverpool, UK
| | - M J Peffers
- Department of Musculoskeletal Biology, Life Course and Medical Sciences, University of Liverpool, UK
| | - L W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - T J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands; Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center +, Maastricht, The Netherlands.
| |
Collapse
|
8
|
Muller D, Thandar Y, Haffejee F. Factors Associated With Compliance With Self-Management Home Therapies for Spine Pain: A Survey of Participants Attending a Chiropractic Teaching Clinic in South Africa. J Chiropr Med 2023; 22:96-102. [PMID: 37346237 PMCID: PMC10280084 DOI: 10.1016/j.jcm.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022] Open
Abstract
Objective The purpose of this study was to determine factors that affect compliance with various prescribed home therapies based on reported feedback from participants with spine pain. Methods This was a descriptive, quantitative, cross-sectional survey. A purposive sampling method was used to recruit 121 participants with neck and back pain attending the Durban University of Technology Chiropractic Day Clinic located in KwaZulu-Natal, South Africa. Data were collected using a self-administered questionnaire. Descriptive statistics, including frequencies and percentages, were used to summarize the data, and odds ratios (ORs) were calculated. Results Most participants presented with chronic pain, reporting an average severity of 6 out of 10 and little disability from the pain. Home therapy included stretches (92.2%), heat therapy (49.1%), and ice therapy (38.8%). Almost two-thirds (62.1%) of participants reported being fully compliant with the prescribed home therapy, while 32.8% reported partial compliance. The main factors that potentially affected compliance were laziness and forgetfulness. Participants who reported having depression were less compliant (OR, 0.181), while those with chronic pain were more compliant (OR, 3.74). Those who believed that home therapy would alleviate their pain were also more compliant (OR, 3.83). Conclusion The study found that a majority of participants with spine pain were compliant with prescribed chiropractic home treatment. Key factors that potentially influenced compliance were identified.
Collapse
Affiliation(s)
- Devereaux Muller
- Department of Chiropractic, Faculty of Health Sciences, Durban University of Technology, Durban, KwaZulu-Natal, South Africa
| | - Yasmeen Thandar
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, KwaZulu-Natal, South Africa
| | - Firoza Haffejee
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
9
|
Feng HY, Zhang PP, Wang XW. Presbyphagia: Dysphagia in the elderly. World J Clin Cases 2023; 11:2363-2373. [PMID: 37123321 PMCID: PMC10131003 DOI: 10.12998/wjcc.v11.i11.2363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/08/2023] [Accepted: 03/22/2023] [Indexed: 04/06/2023] Open
Abstract
Dysphagia has been classified as a “geriatric syndrome” and can lead to serious complications that result in a tremendous burden on population health and healthcare resources worldwide. A characteristic age-related change in swallowing is defined as “presbyphagia.” Medical imaging has shown some changes that seriously affect the safety and efficacy of swallowing. However, there is a general lack of awareness of the effects of aging on swallowing function and a belief that these changes are part of normal aging. Our review provides an overview of presbyphagia, which has been a neglected health problem for a long time. Attention and awareness of dysphagia in the elderly population should be strengthened, and targeted intervention measures should be actively implemented.
Collapse
Affiliation(s)
- Hai-Yang Feng
- School of Rehabilitation Medicine, Weifang Medical University, Weifang 261021, Shandong Province, China
| | - Ping-Ping Zhang
- School of Rehabilitation Medicine, Weifang Medical University, Weifang 261021, Shandong Province, China
| | - Xiao-Wen Wang
- School of Rehabilitation Medicine, Weifang Medical University, Weifang 261021, Shandong Province, China
| |
Collapse
|
10
|
Boote C, Ma Q, Goh KL. Age-dependent mechanical properties of tail tendons in wild-type and mimecan gene-knockout mice - A preliminary study. J Mech Behav Biomed Mater 2023; 139:105672. [PMID: 36657194 DOI: 10.1016/j.jmbbm.2023.105672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/03/2023] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
Mimecan, or osteoglycin, belongs to the family of small leucine-rich proteoglycans. In connective tissues mimecan is implicated in the development and maintenance of normal collagen fibrillar organization. Since collagen fibrils are responsible for tissue reinforcement, the absence of mimecan could lead to abnormal tissue mechanical properties. Here, we carried out a preliminary investigation of possible changes in the mechanical properties of tendons in mice lacking a functional mimecan gene, as a function of age. Tail tendons were dissected from mimecan gene knockout (KO) and wild type (WT) mice at ages 1, 4 and 8 months and mechanical properties evaluated using a microtensile testing equipment. Mimecan gene knockout resulted in changes in tendon elasticity- and fracture-related properties. While tendons of WT mice exhibited enhanced mechanical properties with increasing age, this trend was notably attenuated in mimecan KO tendons, with the exception of fracture strain. When genotype and age were considered as cross factors, the diminution in the mechanical properties of mimecan KO tendons was significant for yield strength, modulus and fracture strength. This effect appeared to affect the mice at 4 month old. These preliminary results suggest that mimecan may have a role in regulating age-dependent mechanical function in mouse tail tendon.
Collapse
Affiliation(s)
- C Boote
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK; Department of Biomedical Engineering, National University of Singapore, Singapore; Newcastle Research and Innovation Institute (NewRIIS), Singapore
| | - Q Ma
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| | - K L Goh
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK; Newcastle Research and Innovation Institute (NewRIIS), Singapore; Faculty of Science, Agriculture and Engineering, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
11
|
Chabronova A, van den Akker GGH, Housmans BAC, Caron MMJ, Cremers A, Surtel DAM, Wichapong K, Peffers MMJ, van Rhijn LW, Marchand V, Motorin Y, Welting TJM. Ribosomal RNA-based epitranscriptomic regulation of chondrocyte translation and proteome in osteoarthritis. Osteoarthritis Cartilage 2023; 31:374-385. [PMID: 36621590 DOI: 10.1016/j.joca.2022.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/08/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Osteoarthritis-related cartilage extracellular matrix remodeling is dependent on changes in chondrocyte protein expression. Yet, the role of ribosomes in chondrocyte translation regulation is unknown. In this exploratory study, we investigated ribosomal RNA (rRNA) epitranscriptomic-based ribosome heterogeneity in human articular chondrocytes and its relevance for osteoarthritis. METHODS Sequencing-based rRNA 2'-O-methylation profiling analysis (RiboMethSeq) was performed on non-OA primary human articular chondrocytes (n = 5) exposed for 14 days to osteoarthritic synovial fluid (14 donors, pooled, 20% v/v). The SW1353 SNORD71 KO cell pool was generated using LentiCRISPRv2/Cas9. The mode of translation initiation and fidelity were determined by dual-luciferase reporters. The cellular proteome was analyzed by LC-MS/MS and collagen type I protein expression was evaluated by immunoblotting. Loading of COL1A1 mRNA into polysomes was determined by sucrose gradient ultracentrifugation and fractionation. RESULTS We discovered that osteoarthritic synovial fluid instigates site-specific changes in the rRNA 2'-O-me profile of primary human articular chondrocytes. We identified five sites with differential 2'-O-me levels. The 2'-O-me status of 5.8S-U14 (one of identified differential 2'-O-me sites; decreased by 7.7%, 95% CI [0.9-14.5%]) was targeted by depleting the level of its guide snoRNA SNORD71 (50% decrease, 95% CI [33-64%]). This resulted in an altered ribosome translation modus (e.g., CrPV IRES, FC 3, 95% CI [2.2-4.1]) and promoted translation of COL1A1 mRNA which led to increased levels of COL1A1 protein (FC 1.7, 95% CI [1.3-2.0]). CONCLUSIONS Our data identify a novel concept suggesting that articular chondrocytes employ rRNA epitranscriptomic mechanisms in osteoarthritis development.
Collapse
Affiliation(s)
- A Chabronova
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - G G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - B A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - A Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - D A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - K Wichapong
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| | - M M J Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - L W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - V Marchand
- Université de Lorraine, UAR2008 IBSLor CNRS-INSERM, BioPole, Nancy, France
| | - Y Motorin
- Université de Lorraine, UAR2008 IBSLor CNRS-INSERM, BioPole, Nancy, France; Université de Lorraine, UMR7365 IMoPA, CNRS, BioPole, Nancy, France
| | - T J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
12
|
Korcari A, Nichols AEC, Buckley MR, Loiselle AE. Scleraxis-lineage cells are required for tendon homeostasis and their depletion induces an accelerated extracellular matrix aging phenotype. eLife 2023; 12:e84194. [PMID: 36656751 PMCID: PMC9908079 DOI: 10.7554/elife.84194] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Aged tendons have disrupted homeostasis, increased injury risk, and impaired healing capacity. Understanding mechanisms of homeostatic disruption is crucial for developing therapeutics to retain tendon health through the lifespan. Here, we developed a novel model of accelerated tendon extracellular matrix (ECM) aging via depletion of Scleraxis-lineage cells in young mice (Scx-DTR). Scx-DTR recapitulates many aspects of tendon aging including comparable declines in cellularity, alterations in ECM structure, organization, and composition. Single-cell RNA sequencing demonstrated a conserved decline in tenocytes associated with ECM biosynthesis in aged and Scx-DTR tendons, identifying the requirement for Scleraxis-lineage cells during homeostasis. However, the remaining cells in aged and Scx-DTR tendons demonstrate functional divergence. Aged tenocytes become pro-inflammatory and lose proteostasis. In contrast, tenocytes from Scx-DTR tendons demonstrate enhanced remodeling capacity. Collectively, this study defines Scx-DTR as a novel model of accelerated tendon ECM aging and identifies novel biological intervention points to maintain tendon function through the lifespan.
Collapse
Affiliation(s)
- Antonion Korcari
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Anne EC Nichols
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
| | - Mark R Buckley
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| |
Collapse
|
13
|
Korcari A, Przybelski SJ, Gingery A, Loiselle AE. Impact of aging on tendon homeostasis, tendinopathy development, and impaired healing. Connect Tissue Res 2023; 64:1-13. [PMID: 35903886 PMCID: PMC9851966 DOI: 10.1080/03008207.2022.2102004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/11/2022] [Indexed: 02/03/2023]
Abstract
Aging is a complex and progressive process where the tissues of the body demonstrate a decreased ability to maintain homeostasis. During aging, there are substantial cellular and molecular changes, with a subsequent increase in susceptibility to pathological degeneration of normal tissue function. In tendon, aging results in well characterized alterations in extracellular matrix (ECM) structure and composition. In addition, the cellular environment of aged tendons is altered, including a marked decrease in cell density and metabolic activity, as well as an increase in cellular senescence. Collectively, these degenerative changes make aging a key risk factor for the development of tendinopathies and can increase the frequency of tendon injuries. However, inconsistencies in the extent of age-related degenerative impairments in tendons have been reported, likely due to differences in how "old" and "young" age-groups have been defined, differences between anatomically distinct tendons, and differences between animal models that have been utilized to study the impact of aging on tendon homeostasis. In this review, we address these issues by summarizing data by well-defined age categories (young adults, middle-aged, and aged) and from anatomically distinct tendon types. We then summarize in detail how aging affects tendon mechanics, structure, composition, and the cellular environment based on current data and underscore what is currently not known. Finally, we discuss gaps in the current understanding of tendon aging and propose key avenues for future research that can shed light on the specific mechanisms of tendon pathogenesis due to aging.
Collapse
Affiliation(s)
- Antonion Korcari
- Department of Orthopaedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | | | - Anne Gingery
- Division of Orthopedic Surgery Research, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Alayna E Loiselle
- Department of Orthopaedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| |
Collapse
|
14
|
Graça AL, Gomez-Florit M, Gomes ME, Docheva D. Tendon Aging. Subcell Biochem 2023; 103:121-147. [PMID: 37120467 DOI: 10.1007/978-3-031-26576-1_7] [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: 05/01/2023]
Abstract
Tendons are mechanosensitive connective tissues responsible for the connection between muscles and bones by transmitting forces that allow the movement of the body, yet, with advancing age, tendons become more prone to degeneration followed by injuries. Tendon diseases are one of the main causes of incapacity worldwide, leading to changes in tendon composition, structure, and biomechanical properties, as well as a decline in regenerative potential. There is still a great lack of knowledge regarding tendon cellular and molecular biology, interplay between biochemistry and biomechanics, and the complex pathomechanisms involved in tendon diseases. Consequently, this reflects a huge need for basic and clinical research to better elucidate the nature of healthy tendon tissue and also tendon aging process and associated diseases. This chapter concisely describes the effects that the aging process has on tendons at the tissue, cellular, and molecular levels and briefly reviews potential biological predictors of tendon aging. Recent research findings that are herein reviewed and discussed might contribute to the development of precision tendon therapies targeting the elderly population.
Collapse
Affiliation(s)
- Ana Luísa Graça
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Manuel Gomez-Florit
- Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
| | - Manuela Estima Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Denitsa Docheva
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
15
|
Rao Y, Zhu C, Suen HC, Huang S, Liao J, Ker DFE, Tuan RS, Wang D. Tenogenic induction of human adipose-derived stem cells by soluble tendon extracellular matrix: composition and transcriptomic analyses. Stem Cell Res Ther 2022; 13:380. [PMID: 35906661 PMCID: PMC9338462 DOI: 10.1186/s13287-022-03038-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 07/06/2022] [Indexed: 11/27/2022] Open
Abstract
Background Tendon healing is clinically challenging largely due to its inferior regenerative capacity. We have previously prepared a soluble, DNA-free, urea-extracted bovine tendon-derived extracellular matrix (tECM) that exhibits strong pro-tenogenic bioactivity on human adipose-derived stem cells (hASCs). In this study, we aimed to elucidate the mechanism of tECM bioactivity via characterization of tECM protein composition and comparison of transcriptomic profiles of hASC cultures treated with tECM versus collagen type I (Col1) as a control ECM component.
Methods The protein composition of tECM was characterized by SDS-PAGE, hydroxyproline assay, and proteomics analysis. To investigate tECM pro-tenogenic bioactivity and mechanism of action, differentiation of tECM-treated hASC cultures was compared to serum control medium or Col1-treated groups, as assessed via immunofluorescence for tenogenic markers and RNA Sequencing (RNA-Seq).
Results Urea-extracted tECM yielded consistent protein composition, including collagens (20% w/w) and at least 17 non-collagenous proteins (< 100 kDa) based on MS analysis. Compared to current literature, tECM included key tendon ECM components that are functionally involved in tendon regeneration, as well as those that are involved in similar principal Gene Ontology (GO) functions (ECM-receptor interaction and collagen formation) and signaling pathways (ECM-receptor interaction and focal adhesion). When used as a cell culture supplement, tECM enhanced hASC proliferation and tenogenic differentiation compared to the Col1 and FBS treatment groups based on immunostaining of tenogenesis-associated markers. Furthermore, RNA-Seq analysis revealed a total of 584 genes differentially expressed among the three culture groups. Specifically, Col1-treated hASCs predominantly exhibited expression of genes and pathways related to ECM-associated processes, while tECM-treated hASCs expressed a mixture of ECM- and cell activity-associated processes, which may explain in part the enhanced proliferation and tenogenic differentiation of tECM-treated hASCs. Conclusions Our findings showed that urea-extracted tECM contained 20% w/w collagens and is significantly enriched with other non-collagenous tendon ECM components. Compared to Col1 treatment, tECM supplementation enhanced hASC proliferation and tenogenic differentiation as well as induced distinct gene expression profiles. These findings provide insights into the potential mechanism of the pro-tenogenic bioactivity of tECM and support the development of future tECM-based approaches for tendon repair. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03038-0.
Collapse
Affiliation(s)
- Ying Rao
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Chenxian Zhu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Hoi Ching Suen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jinyue Liao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,Ministry of Education Key Laboratory for Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.,Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China.
| | - Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,Ministry of Education Key Laboratory for Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China. .,Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China.
| |
Collapse
|
16
|
Enthesis Healing Is Dependent on Scaffold Interphase Morphology—Results from a Rodent Patellar Model. Cells 2022; 11:cells11111752. [PMID: 35681447 PMCID: PMC9179925 DOI: 10.3390/cells11111752] [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: 04/22/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
The use of multiphasic scaffolds to treat injured tendon-to-bone entheses has shown promising results in vitro. Here, we used two versions of a biphasic silk fibroin scaffold to treat an enthesis defect created in a rat patellar model in vivo. One version presented a mixed transition between the bony and the tendon end of the construct (S-MT) while this transition was abrupt in the second version (S-AT). At 12 weeks after surgery, the S-MT scaffold promoted better healing of the injured enthesis, with minimal undesired ossification of the insertion area. The expression of tenogenic and chondrogenic markers was sustained for longer in the S-MT-treated group and the tangent modulus of the S-MT-treated samples was similar to the native tissue at 12 weeks while that of the S-AT-treated enthesis was lower. Our study highlights the important role of the transition zone of multiphasic scaffolds in the treatment of complex interphase tissues such as the tendon-to-bone enthesis.
Collapse
|
17
|
Ali OJ, Ehrle A, Comerford EJ, Canty-Laird EG, Mead A, Clegg PD, Maddox TW. Intrafascicular chondroid-like bodies in the ageing equine superficial digital flexor tendon comprise glycosaminoglycans and type II collagen. J Orthop Res 2021; 39:2755-2766. [PMID: 33580534 DOI: 10.1002/jor.25002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/24/2020] [Accepted: 01/29/2021] [Indexed: 02/04/2023]
Abstract
The superficial digital flexor tendon (SDFT) is considered functionally equivalent to the human Achilles tendon. Circular chondroid depositions scattered amongst the fascicles of the equine SDFT are rarely reported. The purpose of this study was the detailed characterization of intrafascicular chondroid-like bodies (ICBs) in the equine SDFT, and the assessment of the effect of ageing on the presence and distribution of these structures. Ultrahigh field magnetic resonance imaging (9.4T) series of SDFT samples of young (1-9 years) and aged (17-25 years) horses were obtained, and three-dimensional reconstruction of ICBs was performed. Morphological evaluation of the ICBs included histology, immunohistochemistry and transmission electron microscopy. The number, size, and position of ICBs was determined and compared between age groups. There was a significant difference (p = .008) in the ICB count between young and old horses with ICBs present in varying number (13-467; median = 47, mean = 132.6), size and distribution in the SDFT of aged horses only. There were significantly more ICBs in the tendon periphery when compared with the tendon core region (p = .010). Histological characterization identified distinctive cells associated with increased glycosaminoglycan and type II collagen extracellular matrix content. Ageing and repetitive strain frequently cause tendon micro-damage before the development of clinical tendinopathy. Documentation of the presence and distribution of ICBs is a first step towards improving our understanding of the impact of these structures on the viscoelastic properties, and ultimately their effect on the risk of age-related tendinopathy in energy-storing tendons.
Collapse
Affiliation(s)
- Othman J Ali
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,Department of Surgery and Theriogenology, College of Veterinary Medicine, University of Sulaimani, Sulaymaniyah, Sulaymaniyah, Iraq.,Department of Medical Laboratory Science, Komar University of Science and Technology, Sulaymaniyah, Kurdistan Region, Iraq
| | - Anna Ehrle
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Eithne J Comerford
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK.,The Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Elizabeth G Canty-Laird
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,The Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Ashleigh Mead
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK
| | - Peter D Clegg
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK.,The Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Institute of Ageing and Chronic Disease, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Thomas W Maddox
- Department of Musculoskeletal Biology and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK
| |
Collapse
|
18
|
Tendon and multiomics: advantages, advances, and opportunities. NPJ Regen Med 2021; 6:61. [PMID: 34599188 PMCID: PMC8486786 DOI: 10.1038/s41536-021-00168-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 09/01/2021] [Indexed: 02/08/2023] Open
Abstract
Tendons heal by fibrosis, which hinders function and increases re-injury risk. Yet the biology that leads to degeneration and regeneration of tendons is not completely understood. Improved understanding of the metabolic nuances that cause diverse outcomes in tendinopathies is required to solve these problems. 'Omics methods are increasingly used to characterize phenotypes in tissues. Multiomics integrates 'omic datasets to identify coherent relationships and provide insight into differences in molecular and metabolic pathways between anatomic locations, and disease stages. This work reviews the current literature pertaining to multiomics in tendon and the potential of these platforms to improve tendon regeneration. We assessed the literature and identified areas where 'omics platforms contribute to the field: (1) Tendon biology where their hierarchical complexity and demographic factors are studied. (2) Tendon degeneration and healing, where comparisons across tendon pathologies are analyzed. (3) The in vitro engineered tendon phenotype, where we compare the engineered phenotype to relevant native tissues. (4) Finally, we review regenerative and therapeutic approaches. We identified gaps in current knowledge and opportunities for future study: (1) The need to increase the diversity of human subjects and cell sources. (2) Opportunities to improve understanding of tendon heterogeneity. (3) The need to use these improvements to inform new engineered and regenerative therapeutic approaches. (4) The need to increase understanding of the development of tendon pathology. Together, the expanding use of various 'omics platforms and data analysis resulting from these platforms could substantially contribute to major advances in the tendon tissue engineering and regenerative medicine field.
Collapse
|
19
|
Smith RKW, McIlwraith CW. "One Health" in tendinopathy research: Current concepts. J Orthop Res 2021; 39:1596-1602. [PMID: 33713481 DOI: 10.1002/jor.25035] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/02/2021] [Accepted: 03/10/2021] [Indexed: 02/04/2023]
Abstract
Tendinopathy remains one of the most common musculoskeletal disorders affecting both human and equine athletes and presents a considerable therapeutic challenge. The following workshop report comes from the third Dorothy Havemeyer Symposium of Tendinopathy which provided a unique overview of our current understanding of both the basic science and the clinical challenges for diagnosing and treating tendinopathy in both species. Pathologically, tendon demonstrates alterations in both cellular, molecular, structural, and biomechanical features, leading to a spectrum of pathological endotypes. To develop novel interventions to manage, treat or prevent tendinopathies it is vital to understand the underlying mechanisms that lead to both tendon failure, and also regeneration and resolution of inflammation. The horse shows analogous pathology with both human Achilles tendinopathy (superficial digital flexor tendon) and intrathecal rotator cuff tears (deep digital flexor tendon tears) enabling scientists and clinicians from both medical and veterinary fields to work jointly on matching naturally occurring disease models. The experience in human medicine on the design, conduct, and impact of clinical trials has much to inform clinical trials in horses. There is a need to design appropriate studies to address clear questions, socialize the study to achieve good enrollment, and consider the significance and impact of the clinical question as well as the cost of addressing it. Because economics is often a limitation in equine medicine the use of observational studies, and specifically registries, should be given careful consideration.
Collapse
Affiliation(s)
- Roger K W Smith
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hatfield, Herts, UK
| | - C Wayne McIlwraith
- Department of Clinical Sciences, Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
20
|
Kharaz YA, Birch H, Chester A, Alchorne E, Simpson D, Clegg P, Comerford E. The effect of exercise on the protein profile of rat knee joint intra- and extra-articular ligaments. Scand J Med Sci Sports 2021; 31:2033-2043. [PMID: 34271594 DOI: 10.1111/sms.14023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/15/2021] [Indexed: 01/13/2023]
Abstract
Injuries to the intra-articular anterior cruciate ligament (ACL) and the extra-articular medial collateral ligament (MCL) result in significant knee joint instability, pain, and immobility. Moderate endurance-type exercise can increase ligament strength but little is known on the effect of short-term regular bouts of high-intensity exercise on the extracellular matrix (ECM) structure of knee ligaments. Therefore, this study aimed to identify the effect of short-term regular bouts high exercise on the proteome of the rat ACL and MCL using mass spectrometry. Sprague-Dawley male rats (n = 6) were split into control and exercise groups, and subjected to high-intensity training for four 4 weeks followed by proteomic analyses of the ACL and MCL. Knee joint health status was assessed using OARSI and a validated histological scoring system. Histopathological analyses demonstrated no significant changes in either in cruciate, collateral ligaments, or cartilage between the control and exercised knee joints. However, significant proteins were found to be more abundant in the exercised ACL compared to ACL control group but not between the exercised MCL and control MCL groups. The significant abundant proteins in ACL exercise groups were mostly cytoskeletal, ribosomal and enzymes with several abundant matrisomal proteins such as collagen proteins and proteoglycans being found in this group. In conclusion, our results indicate that short-term regular bouts of high-intensity exercise have an impact on the intra-articular ACL but not extra-articular MCL ECM protein expression.
Collapse
Affiliation(s)
- Yalda A Kharaz
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Helen Birch
- Department of Orthopaedics and Musculoskeletal Science, University College London, London, UK
| | | | | | - Deborah Simpson
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Peter Clegg
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,School of Veterinary Science, Leahurst Campus, Neston, UK
| | - Eithne Comerford
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,School of Veterinary Science, Leahurst Campus, Neston, UK
| |
Collapse
|
21
|
O'Brien C, Marr N, Thorpe C. Microdamage in the equine superficial digital flexor tendon. Equine Vet J 2021; 53:417-430. [PMID: 32772396 DOI: 10.1111/evj.13331] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 06/02/2020] [Accepted: 07/30/2020] [Indexed: 12/24/2022]
Abstract
The forelimb superficial digital flexor tendon (SDFT) is an energy-storing tendon that is highly susceptible to injury during activities such as galloping and jumping, such that it is one of the most commonly reported causes of lameness in the performance horse. This review outlines the biomechanical and biothermal effects of strain on the SDFT and how these contribute to the accumulation of microdamage. The effect of age-related alterations on strain response and subsequent injury risk is also considered. Given that tendon is a slowly healing and poorly regenerative tissue, prompt detection of early stages of pathology in vivo and timely adaptations to training protocols are likely to have a greater outcome than advances in treatment. Early screening tools and detection protocols could subsequently be of benefit in identifying subclinical signs of degeneration during the training programme. This provides an opportunity for preventative strategies to be implemented to minimise incidences of SDFT injury and reduce recovery periods in elite performance horses. Therefore, this review will focus on the modalities available to implement early screening and prevention protocols as opposed to methods to diagnose and treat injuries.
Collapse
Affiliation(s)
| | - Neil Marr
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Chavaunne Thorpe
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
22
|
Joyce K, Fabra GT, Bozkurt Y, Pandit A. Bioactive potential of natural biomaterials: identification, retention and assessment of biological properties. Signal Transduct Target Ther 2021; 6:122. [PMID: 33737507 PMCID: PMC7973744 DOI: 10.1038/s41392-021-00512-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/29/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Biomaterials have had an increasingly important role in recent decades, in biomedical device design and the development of tissue engineering solutions for cell delivery, drug delivery, device integration, tissue replacement, and more. There is an increasing trend in tissue engineering to use natural substrates, such as macromolecules native to plants and animals to improve the biocompatibility and biodegradability of delivered materials. At the same time, these materials have favourable mechanical properties and often considered to be biologically inert. More importantly, these macromolecules possess innate functions and properties due to their unique chemical composition and structure, which increase their bioactivity and therapeutic potential in a wide range of applications. While much focus has been on integrating these materials into these devices via a spectrum of cross-linking mechanisms, little attention is drawn to residual bioactivity that is often hampered during isolation, purification, and production processes. Herein, we discuss methods of initial material characterisation to determine innate bioactivity, means of material processing including cross-linking, decellularisation, and purification techniques and finally, a biological assessment of retained bioactivity of a final product. This review aims to address considerations for biomaterials design from natural polymers, through the optimisation and preservation of bioactive components that maximise the inherent bioactive potency of the substrate to promote tissue regeneration.
Collapse
Affiliation(s)
- Kieran Joyce
- School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Georgina Targa Fabra
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Yagmur Bozkurt
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Galway, Ireland.
| |
Collapse
|
23
|
Narayanan N, Calve S. Extracellular matrix at the muscle - tendon interface: functional roles, techniques to explore and implications for regenerative medicine. Connect Tissue Res 2021; 62:53-71. [PMID: 32856502 PMCID: PMC7718290 DOI: 10.1080/03008207.2020.1814263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The muscle-tendon interface is an anatomically specialized region that is involved in the efficient transmission of force from muscle to tendon. Due to constant exposure to loading, the interface is susceptible to injury. Current treatment methods do not meet the socioeconomic demands of reduced recovery time without compromising the risk of reinjury, requiring the need for developing alternative strategies. The extracellular matrix (ECM) present in muscle, tendon, and at the interface of these tissues consists of unique molecules that play significant roles in homeostasis and repair. Better, understanding the function of the ECM during development, injury, and aging has the potential to unearth critical missing information that is essential for accelerating the repair at the muscle-tendon interface. Recently, advanced techniques have emerged to explore the ECM for identifying specific roles in musculoskeletal biology. Simultaneously, there is a tremendous increase in the scope for regenerative medicine strategies to address the current clinical deficiencies. Advancements in ECM research can be coupled with the latest regenerative medicine techniques to develop next generation therapies that harness ECM for treating defects at the muscle-tendon interface. The current work provides a comprehensive review on the role of muscle and tendon ECM to provide insights about the role of ECM in the muscle-tendon interface and discusses the latest research techniques to explore the ECM to gathered information for developing regenerative medicine strategies.
Collapse
Affiliation(s)
- Naagarajan Narayanan
- Paul M. Rady Department of Mechanical Engineering, University of Colorado – Boulder, 1111 Engineering Drive, Boulder, Colorado 80309 – 0427
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado – Boulder, 1111 Engineering Drive, Boulder, Colorado 80309 – 0427
| |
Collapse
|
24
|
Siadat SM, Zamboulis DE, Thorpe CT, Ruberti JW, Connizzo BK. Tendon Extracellular Matrix Assembly, Maintenance and Dysregulation Throughout Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:45-103. [PMID: 34807415 DOI: 10.1007/978-3-030-80614-9_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In his Lissner Award medal lecture in 2000, Stephen Cowin asked the question: "How is a tissue built?" It is not a new question, but it remains as relevant today as it did when it was asked 20 years ago. In fact, research on the organization and development of tissue structure has been a primary focus of tendon and ligament research for over two centuries. The tendon extracellular matrix (ECM) is critical to overall tissue function; it gives the tissue its unique mechanical properties, exhibiting complex non-linear responses, viscoelasticity and flow mechanisms, excellent energy storage and fatigue resistance. This matrix also creates a unique microenvironment for resident cells, allowing cells to maintain their phenotype and translate mechanical and chemical signals into biological responses. Importantly, this architecture is constantly remodeled by local cell populations in response to changing biochemical (systemic and local disease or injury) and mechanical (exercise, disuse, and overuse) stimuli. Here, we review the current understanding of matrix remodeling throughout life, focusing on formation and assembly during the postnatal period, maintenance and homeostasis during adulthood, and changes to homeostasis in natural aging. We also discuss advances in model systems and novel tools for studying collagen and non-collagenous matrix remodeling throughout life, and finally conclude by identifying key questions that have yet to be answered.
Collapse
Affiliation(s)
| | - Danae E Zamboulis
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Chavaunne T Thorpe
- Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Brianne K Connizzo
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|
25
|
Riasat K, Bardell D, Goljanek-Whysall K, Clegg PD, Peffers MJ. Epigenetic mechanisms in Tendon Ageing. Br Med Bull 2020; 135:90-107. [PMID: 32827252 PMCID: PMC7585832 DOI: 10.1093/bmb/ldaa023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Tendon is a composite material with a well-ordered hierarchical structure exhibiting viscoelastic properties designed to transfer force. It is recognized that the incidence of tendon injury increases with age, suggesting a deterioration in homeostatic mechanisms or reparative processes. This review summarizes epigenetic mechanisms identified in ageing healthy tendon. SOURCES OF DATA We searched multiple databases to produce a systematic review on the role of epigenetic mechanisms in tendon ageing. AREAS OF AGREEMENT Epigenetic mechanisms are important in predisposing ageing tendon to injury. AREAS OF CONTROVERSY The relative importance of epigenetic mechanisms are unknown in terms of promoting healthy ageing. It is also unknown whether these changes represent protective mechanisms to function or predispose to pathology. GROWING POINT Epigenetic markers in ageing tendon, which are under-researched including genome-wide chromatin accessibility, should be investigated. AREAS TIMELY FOR DEVELOPING RESEARCH Metanalysis through integration of multiple datasets and platforms will enable a holistic understanding of the epigenome in ageing and its relevance to disease.
Collapse
Affiliation(s)
- Kiran Riasat
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - David Bardell
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK.,Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, Wirral CH64 7TE, UK
| | - Katarzyna Goljanek-Whysall
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Peter D Clegg
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Mandy J Peffers
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| |
Collapse
|
26
|
Farnebo S, Wiig M, Holm B, Ghafouri B. Differentially Expressed Proteins in Intra Synovial Compared to Extra Synovial Flexor Tendon Grafts in a Rabbit Tendon Transplantation Model. Biomedicines 2020; 8:biomedicines8100408. [PMID: 33053838 PMCID: PMC7650534 DOI: 10.3390/biomedicines8100408] [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: 09/02/2020] [Revised: 10/07/2020] [Accepted: 10/10/2020] [Indexed: 11/16/2022] Open
Abstract
Uncomplicated healing of grafts for tendon reconstruction remains an unsolved problem in hand surgery. Results are limited by adhesion formation and decreased strength properties, especially within the tight fibro-osseous sheath of the digits. This is especially problematic when an extra synovial tendon graft is used to replace an intra synovial flexor tendon. Compositional differences are likely to play an important role in these processes. The aim of this study was, therefore, to compare protein expression in pair-matched intra synovial tendon grafts with extra synovial tendon grafts, using a rabbit tendon injury model. We hypothesized that there would be significant differences in proteins critical for response to tensile loading and adhesion formation between the two groups. Using mass spectrometry and multivariate statistical data analysis, we found tissue-specific differences in 22 proteins, where 7 explained 93% (R2) of the variation, with a prediction of 81% (Q2). Among the highest discriminating proteins were Galectin, Histone H2A, and Periostin, which were found in a substantially larger amount in the extra synovial tendons compared to the intra synovial tendons. These findings may contribute to improved understanding of the differences in outcome seen after tendon reconstruction using tendon grafts with intra synovial and extra synovial grafts.
Collapse
Affiliation(s)
- Simon Farnebo
- Department of Hand Surgery, Plastic Surgery, and Burns, Linköping University, SE 581 83 Linköping, Sweden
- Correspondence: (S.F.); (B.G.)
| | - Monica Wiig
- Department of Surgical Science, Hand Surgery, Uppsala University, and Uppsala University Hospital, 751 85 Uppsala, Sweden; (M.W.); (B.H.)
| | - Björn Holm
- Department of Surgical Science, Hand Surgery, Uppsala University, and Uppsala University Hospital, 751 85 Uppsala, Sweden; (M.W.); (B.H.)
| | - Bijar Ghafouri
- Pain and Rehabilitation Centre, and Department of Health, Medicine and Caring Sciences, Linköping University, SE 581 83 Linköping, Sweden
- Correspondence: (S.F.); (B.G.)
| |
Collapse
|
27
|
Zhang C, Svensson RB, Montagna C, Carstensen H, Buhl R, Schoof EM, Kjaer M, Magnusson SP, Yeung CYC. Comparison of Tenocyte Populations from the Core and Periphery of Equine Tendons. J Proteome Res 2020; 19:4137-4144. [PMID: 32822197 DOI: 10.1021/acs.jproteome.0c00591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tendon is a highly organized, dense connective tissue that has been demonstrated to have very little turnover. In spite of the low turnover, tendon can grow in response to loading, which may take place primarily at the periphery. Tendon injuries and recurrence of injuries are common in both humans and animals in sports. It is unclear why some areas of the tendon are more susceptible to such injuries and whether this is due to intrinsic regional differences in extracellular matrix (ECM) production or tissue turnover. This study aimed to compare populations of tenocytes derived from the tendon core and periphery. Tenocytes were isolated from equine superficial digital flexor tendons (SDFTs), and the proliferation capacity was determined. ECM production was characterized by immuno- and histological staining and by liquid chromatography-mass spectrometry-based proteomics. Core and periphery SDFT cultures exhibited comparable proliferation rates and had very similar proteome profiles, but showed biological variation in collagen type I deposition. In conclusion, the intrinsic properties of tenocytes from different regions of the tendon are very similar, and other factors in the tissue may contribute to how specific areas respond to loading or injury.
Collapse
Affiliation(s)
- Cheng Zhang
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital and Center for Healthy Aging, University of Copenhagen, 2400 Copenhagen, Denmark
| | - Rene B Svensson
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital and Center for Healthy Aging, University of Copenhagen, 2400 Copenhagen, Denmark
| | - Costanza Montagna
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital and Center for Healthy Aging, University of Copenhagen, 2400 Copenhagen, Denmark
| | - Helena Carstensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2630 Taastrup, Denmark
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2630 Taastrup, Denmark
| | - Erwin M Schoof
- Proteomics Core, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital and Center for Healthy Aging, University of Copenhagen, 2400 Copenhagen, Denmark
| | - S Peter Magnusson
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital and Center for Healthy Aging, University of Copenhagen, 2400 Copenhagen, Denmark.,Department of Physical and Occupational Therapy, Bispebjerg Hospital, 2400 Copenhagen, Denmark
| | - Ching-Yan Chloé Yeung
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital and Center for Healthy Aging, University of Copenhagen, 2400 Copenhagen, Denmark
| |
Collapse
|
28
|
Impaired chondrocyte U3 snoRNA expression in osteoarthritis impacts the chondrocyte protein translation apparatus. Sci Rep 2020; 10:13426. [PMID: 32778764 PMCID: PMC7417995 DOI: 10.1038/s41598-020-70453-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Although pathways controlling ribosome activity have been described to regulate chondrocyte homeostasis in osteoarthritis, ribosome biogenesis in osteoarthritis is unexplored. We hypothesized that U3 snoRNA, a non-coding RNA involved in ribosomal RNA maturation, is critical for chondrocyte protein translation capacity in osteoarthritis. U3 snoRNA was one of a number of snoRNAs with decreased expression in osteoarthritic cartilage and osteoarthritic chondrocytes. OA synovial fluid impacted U3 snoRNA expression by affecting U3 snoRNA gene promoter activity, while BMP7 was able to increase its expression. Altering U3 snoRNA expression resulted in changes in chondrocyte phenotype. Interference with U3 snoRNA expression led to reduction of rRNA levels and translational capacity, whilst induced expression of U3 snoRNA was accompanied by increased 18S and 28S rRNA levels and elevated protein translation. Whole proteome analysis revealed a global impact of reduced U3 snoRNA expression on protein translational processes and inflammatory pathways. For the first time we demonstrate implications of a snoRNA in osteoarthritis chondrocyte biology and investigated its role in the chondrocyte differentiation status, rRNA levels and protein translational capacity.
Collapse
|
29
|
Kelly E, Smith R, Dudhia J, Faragher RGA. Science-in-brief: The importance of senescence in tendinopathy: New opportunities. Equine Vet J 2020; 52:349-351. [PMID: 32259376 DOI: 10.1111/evj.13228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 01/12/2020] [Indexed: 11/27/2022]
Affiliation(s)
- E Kelly
- Department of Clinical Sciences and Services, The Royal Veterinary College, North Mymms, Hertfordshire, UK
| | - R Smith
- Department of Clinical Sciences and Services, The Royal Veterinary College, North Mymms, Hertfordshire, UK
| | - J Dudhia
- Department of Clinical Sciences and Services, The Royal Veterinary College, North Mymms, Hertfordshire, UK
| | - R G A Faragher
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, East Sussex, UK
| |
Collapse
|
30
|
Durgam S, Singh B, Cole SL, Brokken MT, Stewart M. Quantitative Assessment of Tendon Hierarchical Structure by Combined Second Harmonic Generation and Immunofluorescence Microscopy. Tissue Eng Part C Methods 2020; 26:253-262. [PMID: 32228165 DOI: 10.1089/ten.tec.2020.0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Histological evaluation of healing tendons is primarily focused on monitoring restoration of longitudinal collagen alignment, although the elastic property of energy-storing flexor tendons is largely attributed to interfascicular sliding facilitated by the interfascicular matrix (IFM). The objectives of this study were to explore the utility of second harmonic generation (SHG) imaging to objectively assess cross-sectional tendon fascicle architecture, to combine SHG microscopy with elastin immunofluorescence to assess the ultrastructure of collagen and elastin in longitudinal and transverse sections, and lastly, to quantify changes in IFM elastin and fascicle collagen alignment of normal and collagenase-injured flexor tendons. Paraffin-embedded transverse and longitudinal histological sections (10-μm thickness) derived from normal and collagenase-injured (6- and 16-week time-points) equine superficial digital flexor tendons were de-paraffinized, treated with Tris EDTA at 80°C for epitope retrieval, and incubated with mouse monoclonal anti-elastin antibody (1:100 dilution) overnight. Anti-mouse IgG Alexa Flour 546 secondary antibody was applied, and sections were mounted with ProLong Gold reagent with 4',6-diamidino-2-phenylindole (DAPI). Nuclei (DAPI) and elastin (Alexa Fluor 546) signals were captured by using standard confocal imaging with 405 and 543 nm excitation wavelengths, respectively. The SHG signal was captured by using a tunable Ti:Sapphire laser tuned to 950 nm to visualize type I collagen. Quantitative measurements of fascicle cross-sectional area (CSA), IFM thickness in transverse SHG-DAPI merged z-stacks, fascicle/IFM elastin area fraction (%), and elastin-collagen alignment in longitudinal SHG-elastin merged z-stacks were conducted by using ImageJ software. Using this methodology, fascicle CSA, IFM thickness, and IFM elastin area fraction (%) at 6 weeks (∼2.25-fold; ∼2.8-fold; 60% decrease; p < 0.001) and 16 weeks (∼2-fold; ∼1.5-fold; 70% decrease; p < 0.001) after collagenase injection, respectively, were found to be significantly different from normal tendon. IFM elastin and fascicle collagen alignment characterized via fast Fourier transform (FFT) frequency plots at 16 weeks demonstrated that collagen re-alignment was more advanced than that of elastin. The integration of SHG-derived quantitative measurements in transverse and longitudinal tendon sections supports comprehensive assessment of tendon structure. Our findings demonstrate the importance of including IFM and non-collagenous proteins in tendon histological evaluations, tasks that can be effectively carried out by using SHG and immunofluorescence microscopy. Impact statement This work demonstrated that second harmonic generation microscopy in conjunction with elastin immunofluorescence provided a comprehensive assessment of multiscale structural re-organization in healing tendon than when restricted to longitudinal collagen fiber alignment alone. Utilizing this approach for tendon histomorphometry is ideal not only to improve our understanding of hierarchical structural changes that occur after tendon injury and during remodeling but also to monitor the efficacy of therapeutic approaches.
Collapse
Affiliation(s)
- Sushmitha Durgam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Benjamin Singh
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sara L Cole
- Campus Microscopy Imaging Facility, The Ohio State University, Columbus, Ohio, USA
| | - Matthew T Brokken
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Matthew Stewart
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois, Urbana, Illinois, USA
| |
Collapse
|
31
|
Chen M, Li Y, Xiao L, Dai G, Lu P, Wang Y, Rui Y. AQP1 modulates tendon stem/progenitor cells senescence during tendon aging. Cell Death Dis 2020; 11:193. [PMID: 32188840 PMCID: PMC7080760 DOI: 10.1038/s41419-020-2386-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
The link between tendon stem/progenitor cells (TSPCs) senescence and tendon aging has been well recognized. However, the cellular and molecular mechanisms of TSPCs senescence are still not fully understood. In present study, we investigated the role of Aquaporin 1 (AQP1) in TSPCs senescence. We showed that AQP1 expression declines with age during tendon aging. In aged TSPCs, overexpression of AQP1 significantly attenuated TSPCs senescence. In addition, AQP1 overexpression also restored the age-related dysfunction of self-renewal, migration and tenogenic differentiation. Furthermore, we demonstrated that the JAK-STAT signaling pathway is activated in aged TSPCs, and AQP1 overexpression inhibited the JAK-STAT signaling pathway activation which indicated that AQP1 attenuates senescence and age-related dysfunction of TSPCs through the repression of JAK−STAT signaling pathway. Taken together, our findings demonstrated the critical role of AQP1 in the regulation of TSPCs senescence and provided a novel target for antagonizing tendon aging.
Collapse
Affiliation(s)
- Minhao Chen
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Yingjuan Li
- China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China.,Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Longfei Xiao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Guangchun Dai
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China
| | - Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China. .,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, Jiangsu, China. .,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China. .,China Orthopedic Regenerative Medicine Group, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
32
|
Steinmann S, Pfeifer CG, Brochhausen C, Docheva D. Spectrum of Tendon Pathologies: Triggers, Trails and End-State. Int J Mol Sci 2020; 21:ijms21030844. [PMID: 32013018 PMCID: PMC7037288 DOI: 10.3390/ijms21030844] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/18/2020] [Accepted: 01/24/2020] [Indexed: 12/31/2022] Open
Abstract
The biggest compartment of the musculoskeletal system is the tendons and ligaments. In particular, tendons are dense tissues connecting muscle to bone that are critical for the integrity, function and locomotion of this system. Due to the increasing age of our society and the overall rise in engagement in extreme and overuse sports, there is a growing prevalence of tendinopathies. Despite the recent advances in tendon research and due to difficult early diagnosis, a multitude of risk factors and vague understanding of the underlying biological mechanisms involved in the progression of tendon injuries, the toolbox of treatment strategies remains limited and non-satisfactory. This review is designed to summarize the current knowledge of triggers, trails and end state of tendinopathies.
Collapse
Affiliation(s)
- Sara Steinmann
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Am Biopark 9, 93053 Regensburg, Germany; (S.S.); (C.G.P.)
| | - Christian G. Pfeifer
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Am Biopark 9, 93053 Regensburg, Germany; (S.S.); (C.G.P.)
- Department of Trauma Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany;
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center Regensburg, Am Biopark 9, 93053 Regensburg, Germany; (S.S.); (C.G.P.)
- Department of Medical Biology, Medical University-Plovdiv, 15A Vassil Aprilov Blvd., 4002 Plovdiv, Bulgaria
- Correspondence: ; Tel.: +49 941 943-1605
| |
Collapse
|
33
|
Lui PPY, Wong CM. Biology of Tendon Stem Cells and Tendon in Aging. Front Genet 2020; 10:1338. [PMID: 32010194 PMCID: PMC6976534 DOI: 10.3389/fgene.2019.01338] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 12/09/2019] [Indexed: 12/22/2022] Open
Abstract
Both tendon injuries and tendinopathies, particularly rotator cuff tears, increase with tendon aging. Tendon stem cells play important roles in promoting tendon growth, maintenance, and repair. Aged tendons show a decline in regenerative potential coupled with a loss of stem cell function. Recent studies draw attention to aging primarily a disorder of stem cells. The micro-environment (“niche”) where stem cells resided in vivo provides signals that direct them to metabolize, self-renew, differentiate, or remain quiescent. These signals include receptors and secreted soluble factors for cell-cell communication, extracellular matrix, oxidative stress, and vascularity. Both intrinsic cellular deficits and aged niche, coupled with age-associated systemic changes of hormonal and metabolic signals can inhibit or alter the functions of tendon stem cells, resulting in reduced fitness of these primitive cells and hence more frequent injuries and poor outcomes of tendon repair. This review aims to summarize the biological changes of aged tendons. The biological changes of tendon stem cells in aging are reviewed after a systematic search of the PubMed. Relevant factors of stem cell aging including cell-intrinsic factors, changes of microenvironment, and age-associated systemic changes of hormonal and metabolic signals are examined, with findings related to tendon stem cells highlighted when literature is available. Future research directions on the aging mechanisms of tendon stem cells are discussed. Better understanding of the molecular mechanisms underlying the functional decline of aged tendon stem cells would provide insight for the rational design of rejuvenating therapies.
Collapse
Affiliation(s)
| | - Chi Ming Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| |
Collapse
|
34
|
Connizzo BK, Piet JM, Shefelbine SJ, Grodzinsky AJ. Age-associated changes in the response of tendon explants to stress deprivation is sex-dependent. Connect Tissue Res 2020; 61:48-62. [PMID: 31411079 PMCID: PMC6884684 DOI: 10.1080/03008207.2019.1648444] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose of the Study: The incidence of tendon injuries increases dramatically with age, which presents a major clinical burden. While previous studies have sought to identify age-related changes in extracellular matrix structure and function, few have been able to explain fully why aged tissues are more prone to degeneration and injury. In addition, recent studies have also demonstrated that age-related processes in humans may be sex-dependent, which could be responsible for muddled conclusions in changes with age. In this study, we investigate short-term responses through an ex vivo explant culture model of stress deprivation that specifically questions how age and sex differentially affect the ability of tendons to respond to altered mechanical stimulus.Materials and Methods: We subjected murine flexor explants from young (4 months of age) and aged (22-24 months of age) male and female mice to stress-deprived culture conditions for up to 1 week and investigated changes in viability, cell metabolism and proliferation, matrix biosynthesis and composition, gene expression, and inflammatory responses throughout the culture period.Results and Conclusions: We found that aging did have a significant influence on the response to stress deprivation, demonstrating that aged explants have a less robust response overall with reduced metabolic activity, viability, proliferation, and biosynthesis. However, age-related changes appeared to be sex-dependent. Together, this work demonstrates that the aging process and the subsequent effect of age on the ability of tendons to respond to stress-deprivation are inherently different based on sex, where male explants favor increased activity, apoptosis, and matrix remodeling while female explants favor reduced activity and tissue preservation.
Collapse
Affiliation(s)
- Brianne K. Connizzo
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Correspondence: Brianne K. Connizzo, 70 Massachusetts Avenue, NE47-377, Cambridge, MA 02139, T: 617-253-2469,
| | - Judith M. Piet
- Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Sandra J. Shefelbine
- Department of Bioengineering, Northeastern University, Boston, MA 02115, United States,Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, United States
| | - Alan J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| |
Collapse
|
35
|
McDermott BT, Peffers MJ, McDonagh B, Tew SR. Translational regulation contributes to the secretory response of chondrocytic cells following exposure to interleukin-1β. J Biol Chem 2019; 294:13027-13039. [PMID: 31300557 PMCID: PMC6721953 DOI: 10.1074/jbc.ra118.006865] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 06/12/2019] [Indexed: 01/18/2023] Open
Abstract
Osteoarthritis is a chronic disease characterized by the loss of articular cartilage in synovial joints through a process of extracellular matrix destruction that is strongly associated with inflammatory stimuli. Chondrocytes undergo changes to their protein translational capacity during osteoarthritis, but a study of how disease-relevant signals affect chondrocyte protein translation at the transcriptomic level has not previously been performed. In this study, we describe how the inflammatory cytokine interleukin 1-β (IL-1β) rapidly affects protein translation in the chondrocytic cell line SW1353. Using ribosome profiling we demonstrate that IL-1β induced altered translation of inflammatory-associated transcripts such as NFKB1, TNFAIP2, MMP13, CCL2, and CCL7, as well as a number of ribosome-associated transcripts, through differential translation and the use of multiple open reading frames. Proteomic analysis of the cellular layer and the conditioned media of these cells identified changes in a number of the proteins that were differentially translated. Translationally regulated secreted proteins included a number of chemokines and cytokines, underlining the rapid, translationally mediated inflammatory cascade that is initiated by IL-1β. Although fewer cellular proteins were found to be regulated in both ribosome profiling and proteomic data sets, we did find increased levels of SOD2, indicative of redox changes within SW1353 cells being modulated at the translational level. In conclusion, we have produced combined ribosome profiling and proteomic data sets that provide a valuable resource in understanding the processes that occur during cytokine stimulation of chondrocytic cells.
Collapse
Affiliation(s)
- Benjamin T McDermott
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom.
| | - Mandy J Peffers
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom
| | - Brian McDonagh
- Department of Physiology, School of Medicine, National University of Ireland (NUI), Galway H91 TK33, Ireland
| | - Simon R Tew
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom
| |
Collapse
|
36
|
Grote C, Reinhardt D, Zhang M, Wang J. Regulatory mechanisms and clinical manifestations of musculoskeletal aging. J Orthop Res 2019; 37:1475-1488. [PMID: 30919498 PMCID: PMC9202363 DOI: 10.1002/jor.24292] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/13/2019] [Indexed: 02/04/2023]
Abstract
Aging is the strongest risk factor for degenerative bone and joint diseases. Clinical therapies for age-related musculoskeletal disorders face significant challenges as their pathogenic mechanisms remain largely unclear. This review article focuses on the recent advances in the understanding of regulatory mechanisms of musculoskeletal aging and their clinical relevance. We begin with the prevalence and socioeconomic impacts of major age-related musculoskeletal disorders such as sarcopenia, osteoporosis, osteoarthritis, and degenerative tendinopathy. The current understanding of responsible biological mechanisms involved in general aging is then summarized. Proposed molecular, cellular, and biomechanical mechanisms relevant to the clinical manifestations of aging in the musculoskeletal system are discussed in detail, with a focus on the disorders affecting muscle, bone, articular cartilage, and tendon. Although musculoskeletal aging processes share many common pathways with the aging of other body systems, unique molecular and cellular mechanisms may be involved in the aging processes of musculoskeletal tissues. Advancements in the understanding of regulatory mechanisms of musculoskeletal aging may promote the development of novel treatments for age-related musculoskeletal disorders. Finally, future research directions for major musculoskeletal tissues including functional interaction between the tissues and their clinical relevance to age-related musculoskeletal disorders are highlighted in the Future Prospects section. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1475-1488, 2019.
Collapse
Affiliation(s)
- Caleb Grote
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Daniel Reinhardt
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mingcai Zhang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
37
|
Nichols AEC, Best KT, Loiselle AE. The cellular basis of fibrotic tendon healing: challenges and opportunities. Transl Res 2019; 209:156-168. [PMID: 30776336 PMCID: PMC6545261 DOI: 10.1016/j.trsl.2019.02.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 12/11/2022]
Abstract
Tendon injuries are common and can dramatically impair patient mobility and productivity, resulting in a significant socioeconomic burden and reduced quality of life. Because the tendon healing process results in the formation of a fibrotic scar, injured tendons never regain the mechanical strength of the uninjured tendon, leading to frequent reinjury. Many tendons are also prone to the development of peritendinous adhesions and excess scar formation, which further reduce tendon function and lead to chronic complications. Despite this, there are currently no treatments that adequately improve the tendon healing process due in part to a lack of information regarding the contributions of various cell types to tendon healing and how their activity may be modulated for therapeutic value. In this review, we summarize recent efforts to identify and characterize the distinct cell populations involved at each stage of tendon healing. In addition, we examine the mechanisms through which different cell populations contribute to the fibrotic response to tendon injury, and how these responses can be affected by systemic factors and comorbidities. We then discuss gaps in our current understanding of tendon fibrosis and highlight how new technologies and research areas are shedding light on this clinically important and intractable challenge. A better understanding of the complex cellular environment during tendon healing is crucial to the development of new therapies to prevent fibrosis and promote tissue regeneration.
Collapse
Affiliation(s)
- Anne E C Nichols
- Department of Orthopedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Katherine T Best
- Department of Orthopedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Alayna E Loiselle
- Department of Orthopedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|
38
|
Steinbusch MMF, Caron MMJ, Surtel DAM, van den Akker GGH, van Dijk PJ, Friedrich F, Zabel B, van Rhijn LW, Peffers MJ, Welting TJM. The antiviral protein viperin regulates chondrogenic differentiation via CXCL10 protein secretion. J Biol Chem 2019; 294:5121-5136. [PMID: 30718282 PMCID: PMC6442052 DOI: 10.1074/jbc.ra119.007356] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 01/29/2019] [Indexed: 01/10/2023] Open
Abstract
Viperin (also known as radical SAM domain–containing 2 (RSAD2)) is an interferon-inducible and evolutionary conserved protein that participates in the cell's innate immune response against a number of viruses. Viperin mRNA is a substrate for endoribonucleolytic cleavage by RNase mitochondrial RNA processing (MRP) and mutations in the RNase MRP small nucleolar RNA (snoRNA) subunit of the RNase MRP complex cause cartilage-hair hypoplasia (CHH), a human developmental condition characterized by metaphyseal chondrodysplasia and severe dwarfism. It is unknown how CHH-pathogenic mutations in RNase MRP snoRNA interfere with skeletal development, and aberrant processing of RNase MRP substrate RNAs is thought to be involved. We hypothesized that viperin plays a role in chondrogenic differentiation. Using immunohistochemistry, real-time quantitative PCR, immunoblotting, ELISA, siRNA-mediated gene silencing, plasmid-mediated gene overexpression, label-free MS proteomics, and promoter reporter bioluminescence assays, we discovered here that viperin is expressed in differentiating chondrocytic cells and regulates their protein secretion and the outcome of chondrogenic differentiation by influencing transforming growth factor β (TGF-β)/SMAD family 2/3 (SMAD2/3) activity via C-X-C motif chemokine ligand 10 (CXCL10). Of note, we observed disturbances in this viperin–CXCL10–TGF-β/SMAD2/3 axis in CHH chondrocytic cells. Our results indicate that the antiviral protein viperin controls chondrogenic differentiation by influencing secretion of soluble proteins and identify a molecular route that may explain impaired chondrogenic differentiation of cells from individuals with CHH.
Collapse
Affiliation(s)
- Mandy M F Steinbusch
- From the Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery and
| | - Marjolein M J Caron
- From the Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery and
| | - Don A M Surtel
- From the Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery and
| | | | - Paul J van Dijk
- the Department of Anatomy and Embryology, Maastricht University, NL-6202 AZ Maastricht, The Netherlands
| | - Franziska Friedrich
- the University Heart Centre Freiburg, Faculty of Medicine, University of Freiburg, Institute for Experimental Cardiovascular Medicine, 79110 Freiburg, Germany
| | - Bernhard Zabel
- the Medical Faculty, Otto van Guericke University of Magdeburg, 39106 Magdeburg, Germany, and
| | - Lodewijk W van Rhijn
- From the Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery and
| | - Mandy J Peffers
- the Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Tim J M Welting
- From the Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery and
| |
Collapse
|
39
|
Bonilla-Gutiérrez AF, López C, Carmona JU. Regenerative Therapies for the Treatment of Tenodesmic Injuries in Horses. J Equine Vet Sci 2019. [DOI: 10.1016/j.jevs.2018.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
40
|
Lee KJ, Comerford EJ, Simpson DM, Clegg PD, Canty-Laird EG. Identification and Characterization of Canine Ligament Progenitor Cells and Their Extracellular Matrix Niche. J Proteome Res 2019; 18:1328-1339. [DOI: 10.1021/acs.jproteome.8b00933] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Katie J Lee
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, United Kingdom
| | - Eithne J Comerford
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, United Kingdom
- School of Veterinary Science, Leahurst Campus, University of Liverpool, Chester High Road, Neston, CH64 7TE, United Kingdom
| | - Deborah M Simpson
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | - Peter D Clegg
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, United Kingdom
- School of Veterinary Science, Leahurst Campus, University of Liverpool, Chester High Road, Neston, CH64 7TE, United Kingdom
- The MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool L7 8TX, United Kingdom
| | - Elizabeth G Canty-Laird
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, United Kingdom
- The MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool L7 8TX, United Kingdom
| |
Collapse
|
41
|
Guzzoni V, Selistre-de-Araújo HS, Marqueti RDC. Tendon Remodeling in Response to Resistance Training, Anabolic Androgenic Steroids and Aging. Cells 2018; 7:E251. [PMID: 30544536 PMCID: PMC6316563 DOI: 10.3390/cells7120251] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022] Open
Abstract
Exercise training (ET), anabolic androgenic steroids (AAS), and aging are potential factors that affect tendon homeostasis, particularly extracellular matrix (ECM) remodeling. The goal of this review is to aggregate findings regarding the effects of resistance training (RT), AAS, and aging on tendon homeostasis. Data were gathered from our studies regarding the impact of RT, AAS, and aging on the calcaneal tendon (CT) of rats. We demonstrated a series of detrimental effects of AAS and aging on functional and biomechanical parameters, including the volume density of blood vessel cells, adipose tissue cells, tendon calcification, collagen content, the regulation of the major proteins related to the metabolic/development processes of tendons, and ECM remodeling. Conversely, RT seems to mitigate age-related tendon dysfunction. Our results suggest that AAS combined with high-intensity RT exert harmful effects on ECM remodeling, and also instigate molecular and biomechanical adaptations in the CT. Moreover, we provide further information regarding the harmful effects of AAS on tendons at a transcriptional level, and demonstrate the beneficial effects of RT against the age-induced tendon adaptations of rats. Our studies might contribute in terms of clinical approaches in favor of the benefits of ET against tendinopathy conditions, and provide a warning on the harmful effects of the misuse of AAS on tendon development.
Collapse
Affiliation(s)
- Vinicius Guzzoni
- Departamento de Biologia Molecular e Celular, Universidade Federal da Paraíba, João Pessoa 58051-970, Paraíba, Brazil.
| | | | - Rita de Cássia Marqueti
- Graduate Program of Rehabilitation Science, University of Brasilia, Distrito Federal, Brasília 70840-901, Distrito Federal, Brazil.
| |
Collapse
|
42
|
Fêo HB, Biancalana A, Romero Nakagaki W, Aparecida de Aro A, Gomes L. Morphological Alterations and Increased Gelatinase Activity in the Superficial Digital Flexor Tendon of Chickens During Growth and Maturation. Anat Rec (Hoboken) 2018; 302:964-972. [DOI: 10.1002/ar.24027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 07/30/2018] [Accepted: 08/28/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Haline Ballestero Fêo
- Department of Structural and Functional BiologyInstitute of Biology, State University of Campinas – UNICAMP Campinas Brazil
| | - Adriano Biancalana
- Department of Structural and Functional BiologyInstitute of Biology, State University of Campinas – UNICAMP Campinas Brazil
- Laboratory of Cellular and Molecular BiologyFederal University of Pará – UFPA Soure Brazil
| | - Wilson Romero Nakagaki
- Department of Structural and Functional BiologyInstitute of Biology, State University of Campinas – UNICAMP Campinas Brazil
- Master's Program in Health SciencesUniversity of Western São Paulo – UNOESTE Presidente Prudente Brazil
| | - Andrea Aparecida de Aro
- Department of Structural and Functional BiologyInstitute of Biology, State University of Campinas – UNICAMP Campinas Brazil
- Biomedical Sciences Graduate ProgramHerminio Ometto University Center –UNIARARAS Araras Brazil
| | - Laurecir Gomes
- Department of Structural and Functional BiologyInstitute of Biology, State University of Campinas – UNICAMP Campinas Brazil
| |
Collapse
|
43
|
Thankam FG, Dilisio MF, Gross RM, Agrawal DK. Collagen I: a kingpin for rotator cuff tendon pathology. Am J Transl Res 2018; 10:3291-3309. [PMID: 30662587 PMCID: PMC6291732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/21/2018] [Indexed: 06/09/2023]
Abstract
Derangements in tendon matrisome are pathognomonic for musculoskeletal disorders including rotator cuff tendinopathies (RCT). Collagen type-1 accounts for more than 85% of the dry weight of tendon extracellular matrix (ECM). The understanding of basic tendon physiology, organization of ECM, structure and function of component biomolecules of matrisome and the underlying regulatory mechanisms reveal the pathological events associated with RCT. Histomorphological evidence from RCT patients and animal models illustrate that ECM disorganization is the major hallmark in tendinopathy where a significant decrease in type-1 collagen is prevalent. However, the molecular events and regulatory signals associated with the regulation of collagen organization and its composition switch in response to pathological stimuli are largely unknown. The elucidation of various regulatory signalling pathways associated with collagen type-1 gene expression could benefit to develop novel promising therapeutic approaches to restore the tendon ECM. The major focus of the article is to critically evaluate tendon architecture regarding type-1 collagen, the molecular events associated with gene expression, secretion and maturation, the possible mechanisms of type-1 collagen regulation and its translational significance in RCT management.
Collapse
Affiliation(s)
- Finosh G Thankam
- Department of Clinical and Translational Science and Orthopedic Surgery, School of Medicine, Creighton University Omaha, NE 68178, USA
| | - Matthew F Dilisio
- Department of Clinical and Translational Science and Orthopedic Surgery, School of Medicine, Creighton University Omaha, NE 68178, USA
| | - Richard M Gross
- Department of Clinical and Translational Science and Orthopedic Surgery, School of Medicine, Creighton University Omaha, NE 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science and Orthopedic Surgery, School of Medicine, Creighton University Omaha, NE 68178, USA
| |
Collapse
|
44
|
Devaud YR, Avilla-Royo E, Trachsel C, Grossmann J, Martin I, Lutolf MP, Ehrbar M. Label-Free Quantification Proteomics for the Identification of Mesenchymal Stromal Cell Matrisome Inside 3D Poly(Ethylene Glycol) Hydrogels. Adv Healthc Mater 2018; 7:e1800534. [PMID: 30260582 DOI: 10.1002/adhm.201800534] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/20/2018] [Indexed: 01/31/2023]
Abstract
Cells modulate the functional properties of their environment by depositing extracellular matrix (ECM) proteins during biological processes in vivo and in vitro. Despite the ECMs central role in tissue formation, its quantification in hydrogels like Matrigel, which have a complex materials-inherent biopolymer composition is exceptionally challenging. Here, the use of protein-free, synthetic poly(ethylene glycol) hydrogels enables the analysis of deposited human bone marrow mesenchymal stromal cells ECM directly harvested from fresh 3D cell cultures by a tandem mass spectrometry (LC-MS/MS) method. In this study, it is proved that a label-free LC-MS/MS quantification method can selectively identify proteins deposited in 3D synthetic hydrogels following different growth factor (GF) treatments. Furthermore, it is shown that the sequence in which GFs are administered and the choice of stimuli significantly influences the number and abundance of ECM proteins. Therefore, this provides a versatile method to optimize GF treatments in synthetic hydrogel-based regenerative medicine and tissue engineering approaches.
Collapse
Affiliation(s)
- Yannick R. Devaud
- Department of Obstetrics; University Hospital Zurich; University of Zurich; Schmelzbergstr. 12 8091 Zurich Switzerland
| | - Eva Avilla-Royo
- Department of Obstetrics; University Hospital Zurich; University of Zurich; Schmelzbergstr. 12 8091 Zurich Switzerland
- Institute for Biomechanics; Swiss Federal Institute of Technology; CH-8008 Zurich Switzerland
| | - Christian Trachsel
- Functional Genomics Center University of Zurich and ETH Zurich; Winterthurerstr. 190 8057 Zürich Switzerland
| | - Jonas Grossmann
- Functional Genomics Center University of Zurich and ETH Zurich; Winterthurerstr. 190 8057 Zürich Switzerland
| | - Ivan Martin
- Department of Biomedicine and Department of Surgery; University Hospital Basel; University of Basel; Hebelstrasse 20 4031 Basel Switzerland
| | - Matthias P. Lutolf
- Institute of Bioengineering; Ecole Polytechnique Fédérale de Lausanne (EPFL); Station 15, Bld AI 1109 1015 Lausanne Switzerland
| | - Martin Ehrbar
- Department of Obstetrics; University Hospital Zurich; University of Zurich; Schmelzbergstr. 12 8091 Zurich Switzerland
| |
Collapse
|
45
|
Connizzo BK, Grodzinsky AJ. Multiscale Poroviscoelastic Compressive Properties of Mouse Supraspinatus Tendons Are Altered in Young and Aged Mice. J Biomech Eng 2018; 140:2666618. [PMID: 29238818 PMCID: PMC5816244 DOI: 10.1115/1.4038745] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/04/2017] [Indexed: 02/02/2023]
Abstract
Rotator cuff disorders are one of the most common causes of shoulder pain and disability in the aging population but, unfortunately, the etiology is still unknown. One factor thought to contribute to the progression of disease is the external compression of the rotator cuff tendons, which can be significantly increased by age-related changes such as muscle weakness and poor posture. The objective of this study was to investigate the baseline compressive response of tendon and determine how this response is altered during maturation and aging. We did this by characterizing the compressive mechanical, viscoelastic, and poroelastic properties of young, mature, and aged mouse supraspinatus tendons using macroscale indentation testing and nanoscale high-frequency AFM-based rheology testing. Using these multiscale techniques, we found that aged tendons were stiffer than their mature counterparts and that both young and aged tendons exhibited increased hydraulic permeability and energy dissipation. We hypothesize that regional and age-related variations in collagen morphology and organization are likely responsible for changes in the multiscale compressive response as these structural parameters may affect fluid flow. Importantly, these results suggest a role for age-related changes in the progression of tendon degeneration, and we hypothesize that decreased ability to resist compressive loading via fluid pressurization may result in damage to the extracellular matrix (ECM) and ultimately tendon degeneration. These studies provide insight into the regional multiscale compressive response of tendons and indicate that altered compressive properties in aging tendons may be a major contributor to overall tendon degeneration.
Collapse
Affiliation(s)
- Brianne K. Connizzo
- Department of Biological Engineering,
Massachusetts Institute of Technology,
Cambridge, MA 02139
| | - Alan J. Grodzinsky
- Department of Biological Engineering,Massachusetts Institute of Technology,
Cambridge, MA 02139;
Center for Biomedical Engineering,Massachusetts Institute of Technology,
Cambridge, MA 02139;
Department of Electrical Engineeringand Computer Science,
Massachusetts Institute of Technology,
Cambridge, MA 02139;
Department of Mechanical Engineering,Massachusetts Institute of Technology,
Cambridge, MA 02139
e-mail:
| |
Collapse
|
46
|
Turlo AJ, Ashraf Kharaz Y, Clegg PD, Anderson J, Peffers MJ. Donor age affects proteome composition of tenocyte-derived engineered tendon. BMC Biotechnol 2018; 18:2. [PMID: 29338716 PMCID: PMC5771075 DOI: 10.1186/s12896-018-0414-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 01/03/2018] [Indexed: 12/27/2022] Open
Abstract
Background The concept of tissue engineering is to deliver to the injury site biological scaffolds carrying functional cells that will enhance healing response. The preferred cell source is autologous in order to reduce immune response in the treated individual. However, in elderly patients age-related changes in synthetic activity of the implanted cells and subsequent alterations in tissue protein content may affect therapeutic outcomes. In this study we investigated the effect of donor age on proteome composition of tenocyte-derived tendon tissue-engineered constructs. Results Liquid chromatography tandem mass spectrometry was used to assess the proteome of tissue-engineered constructs derived from young and old equine tenocytes. Ageing was associated with altered extracellular matrix composition, especially accumulation of collagens (type I, III and XIV), and lower cytoskeletal turnover. Proteins involved in cell responsiveness to mechanical stimuli and cell-extracellular matrix interaction (calponin 1, palladin, caldesmon 1, cortactin) were affected. Conclusions This study demonstrated significant changes in proteome of engineered tendon derived from young and old tenocytes, indicating the impact of donor age on composition of autologous constructs. Electronic supplementary material The online version of this article (10.1186/s12896-018-0414-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Agnieszka J Turlo
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Science, ul. Nowoursynowska 159c, 02-776, Warsaw, Poland.
| | - Yalda Ashraf Kharaz
- Institute of Ageing and Chronic Disease, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - James Anderson
- Institute of Ageing and Chronic Disease, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Mandy J Peffers
- Institute of Ageing and Chronic Disease, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| |
Collapse
|
47
|
Abstract
The extracellular matrix (ECM) provides the environment for many cells types within the body and, in addition to the well recognised role as a structural support, influences many important cell process within the body. As a result, age-related changes to the proteins of the ECM have far reaching consequences with the potential to disrupt many different aspects of homeostasis and healthy function. The proteins collagen and elastin are the most abundant in the ECM and their ability to function as a structural support and provide mechanical stability results from the formation of supra-molecular structures. Collagen and elastin have a long half-life, as required by their structural role, which leaves them vulnerable to a range of post-translational modifications. In this chapter the role of the ECM is discussed and the component proteins introduced. Major age-related modifications including glycation, carbamylation and fragmentation and the impact these have on ECM function are reviewed.
Collapse
|
48
|
Gagliano N, Menon A, Cabitza F, Compagnoni R, Randelli P. Morphological and molecular characterization of human hamstrings shows that tendon features are not influenced by donor age. Knee Surg Sports Traumatol Arthrosc 2018; 26:343-352. [PMID: 28770299 DOI: 10.1007/s00167-017-4661-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/26/2017] [Indexed: 01/12/2023]
Abstract
PURPOSE Age-related modifications of tendons, such as reduced tenocyte proliferation and modified extracellular matrix (ECM) turnover, have been previously described, but results are often incomplete and discordant. The aim of this study was to investigate, using morphological and molecular methods, the effect of ageing on human tendons and tenocytes, especially focusing on the collagen turnover pathways, in order to understand how the ageing process could influence tendon biology and structure. METHODS Morphological analysis was performed on fragments from human semitendinosus and gracilis tendons harvested from 10 adult (mean age 41.8 ± 13.3 years) and 6 aged healthy patients (mean age 72.7 ± 7.0 years) by haematoxylin and eosin, Sirius red and Alcian blue staining. The expression of genes and proteins involved in collagen turnover and focal adhesions was assessed by real-time PCR, slot blot and zymography in cultured tenocytes. Cytoskeleton arrangement was studied by immunofluorescence and cell migration by wound healing assay. RESULTS The structure and composition of ECM in ageing tendons are preserved as well as the expression of genes and proteins involved in collagen turnover pathways. Although morphological analysis revealed that ageing tenocytes tended to an impaired migration potential and that actin filaments are occasionally shorter and randomly distributed, the expression of proteins involved in focal adhesion formation is preserved. CONCLUSION Results of this study suggest that the structure of ageing tendons is preserved and that ageing tenocytes maintain their ability for ECM remodelling, supporting the hypothesis that ageing tendons maintain their biomechanical properties. The biological reliability of aged tendons has a clinical relevance, supporting the use of tendon autografts also in the elderly patients. Since the common and successful orthopaedic procedure of anterior cruciate ligament reconstruction using either autografts or allografts is becoming more common in older age groups, these findings suggest that the donor age would not significantly influence the clinical outcome.
Collapse
Affiliation(s)
- Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy.
| | - Alessandra Menon
- 1st Department, Azienda Socio Sanitaria Territoriale Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan, Italy
| | - Federico Cabitza
- Istituto Ortopedico Galeazzi, Milan, Italy
- Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Riccardo Compagnoni
- 1st Department, Azienda Socio Sanitaria Territoriale Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan, Italy
| | - Pietro Randelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
- 1st Department, Azienda Socio Sanitaria Territoriale Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan, Italy
| |
Collapse
|
49
|
Ackerman JE, Bah I, Jonason JH, Buckley MR, Loiselle AE. Aging does not alter tendon mechanical properties during homeostasis, but does impair flexor tendon healing. J Orthop Res 2017; 35:2716-2724. [PMID: 28419543 PMCID: PMC5645212 DOI: 10.1002/jor.23580] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/12/2017] [Indexed: 02/04/2023]
Abstract
Aging is an important factor in disrupted homeostasis of many tissues. While an increased incidence of tendinopathy and tendon rupture are observed with aging, it is unclear whether this is due to progressive changes in tendon cell function and mechanics over time, or an impaired repair reaction from aged tendons in response to insult or injury. In the present study, we examined changes in the mechanical properties of Flexor Digitorum Longus (FDL), Flexor Carpi Ulnaris (FCU), and tail fascicles in both male and female C57Bl/6 mice between 3 and 27 months of age to better understand the effects of sex and age on tendon homeostasis. No change in max load at failure was observed in any group over the course of aging, although there were significant decreases in toe and linear stiffness in female mice from 3 to 15 months, and 3 to 27 months. No changes in cell proliferation were observed with aging, although an observable decrease in cellularity occurred in 31-month old tendons. Given that aging did not dramatically alter tendon mechanical homeostasis we hypothesized that a disruption in tendon homeostasis, via acute injury would result in an impaired healing response. Significant decreases in max load, stiffness, and yield load were observed in repairs of 22-month old mice, relative to 4-month old mice. No changes in cell proliferation were observed between young and aged, however, a dramatic loss of bridging collagen extracellular matrix was observed in aged repairs suggest that matrix production, but not cell proliferation leads to impaired tendon healing with aging. Results © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2716-2724, 2017.
Collapse
Affiliation(s)
- Jessica E. Ackerman
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642
| | - Ibrahima Bah
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642,Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642
| | - Jennifer H. Jonason
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642
| | - Mark R. Buckley
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642,Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642
| | - Alayna E. Loiselle
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642,Corresponding Author: Alayna E. Loiselle, PhD, Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, Phone: 585-275-7239, Fax: 585-276-2177,
| |
Collapse
|
50
|
Barin FR, Durigan JLQ, Oliveira KDS, Migliolo L, Almeida JA, Carvalho M, Petriz B, Selistre-de-Araujo HS, Fontes W, Franco OL, Marqueti RDC. Beneficial effects of resistance training on the protein profile of the calcaneal tendon during aging. Exp Gerontol 2017; 100:54-62. [DOI: 10.1016/j.exger.2017.10.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 09/23/2017] [Accepted: 10/20/2017] [Indexed: 01/08/2023]
|