1
|
Luo G, Li J, Chen S, Yuan Z, Sun Z, Lou T, Chen Z, Liu H, Zhou C, Fan C, Ruan H. Polylactic acid electrospun membranes coated with chiral hierarchical-structured hydroxyapatite nanoplates promote tendon healing based on a macrophage-homeostatic modulation strategy. Bioact Mater 2025; 47:460-480. [PMID: 40034408 PMCID: PMC11872693 DOI: 10.1016/j.bioactmat.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/30/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Tendon injury is a common and challenging problem in the motor system that lacks an effective treatment, affecting daily activities and lowering the quality of life. Limited tendon regenerative capability and immune microenvironment dyshomeostasis are considered the leading causes hindering tendon repair. The chirality of biomaterials was proved to dictate immune microenvironment and dramatically affect tissue repair. Herein, chiral hierarchical structure hydroxylapatite (CHAP) nanoplates are innovatively synthesized for immunomodulatory purposes and further coated onto polylactic acid electrospinning membranes to achieve long-term release for tendon regeneration adaption. Notably, levorotatory-chiral HAP (L-CHAP) nanoplates rather than dextral-chiral or racemic-chiral exhibit good biocompatibility and bioactivity. In vitro experiments demonstrate that L-CHAP induces macrophage M2 polarization by enhancing macrophage efferocytosis, which alleviates inflammatory damage to tendon stem cells (TDSCs) through downregulated IL-17-NF-κB signaling. Meanwhile, L-CHAP-mediated macrophage efferocytosis also promotes TDSCs proliferation and tenogenic differentiation. By establishing a rat model of Achilles tendon injury, L-CHAP was demonstrated to comprehensively promoting tendon repair by enhancing macrophage efferocytosis and M2 polarization in vivo, finally leading to improvement of tendon ultrastructural and mechanical properties and motor function. This novel strategy highlights the role of L-CHAP in tendon repair and thus provides a promising therapeutic strategy for tendon injury.
Collapse
Affiliation(s)
- Gang Luo
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Shuai Chen
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Zhengqiang Yuan
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Ziyang Sun
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Tengfei Lou
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Zhenyu Chen
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Hang Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Chao Zhou
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| | - Hongjiang Ruan
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, PR China
| |
Collapse
|
2
|
Xiong J, Ma R, Xie K, Shan C, Chen H, Wang Y, Liao Y, Deng Y, Ye G, Wang Y, Zhu Q, Zhang Y, Cai H, Guo W, Yin Y, Li Z. Recapitulation of endochondral ossification by hPSC-derived SOX9 + sclerotomal progenitors. Nat Commun 2025; 16:2781. [PMID: 40118845 PMCID: PMC11928506 DOI: 10.1038/s41467-025-58122-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
Endochondral ossification generates most of the load-bearing bones, recapitulating it in human cells remains a challenge. Here, we report generation of SOX9+ sclerotomal progenitors (scl-progenitors), a mesenchymal precursor at the pre-condensation stage, from human pluripotent stem cells and development of osteochondral induction methods for these cells. Upon lineage-specific induction, SOX9+ scl-progenitors have not only generated articular cartilage but have also undergone spontaneous condensation, cartilaginous anlagen formation, chondrocyte hypertrophy, vascular invasion, and finally bone formation with stroma, thereby recapitulating key stages during endochondral ossification. Moreover, self-organized growth plate-like structures have also been induced using SOX9+ scl-progenitor-derived fusion constructs with chondro- and osteo-spheroids, exhibiting molecular and cellular similarities to the primary growth plates. Furthermore, we have identified ITGA9 as a specific surface marker for reporter-independent isolation of SOX9+ scl-progenitors and established a culture system to support their expansion. Our work highlights SOX9+ scl-progenitors as a promising tool for modeling human skeletal development and bone/cartilage bioengineering.
Collapse
Affiliation(s)
- Jingfei Xiong
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Runxin Ma
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Kun Xie
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Ce Shan
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Hanyi Chen
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuqing Wang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuansong Liao
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yanhui Deng
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Guogen Ye
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yifu Wang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Qing Zhu
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu, China
| | - Yunqiu Zhang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Haoyang Cai
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Weihua Guo
- Yunnan Key Laboratory of Stomatology, Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Yike Yin
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China.
| | - Zhonghan Li
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China.
- Yunnan Key Laboratory of Stomatology, Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming Medical University, Kunming, China.
| |
Collapse
|
3
|
Yuan Z, Yao Z, Mao X, Gao X, Wu S, Mao H. Epigenetic mechanisms in stem cell therapies for achilles tendinopathy. Front Cell Dev Biol 2025; 13:1516250. [PMID: 40181824 PMCID: PMC11965899 DOI: 10.3389/fcell.2025.1516250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Achilles tendinopathy (AT) is a chronic degenerative tendinopathy that affects people's daily lives. Multiple clinical studies have found that current conservative treatments fail to promote quality tendon healing. Recent studies have found that stem cell therapy can target pathophysiological changes in the tendon by replenishing tendon-derived cells, promoting extracellular matrix (ECM) remodeling, and modulating the inflammatory response to improve the microenvironment of Achilles tendon regeneration. And epigenetic modifications play an important role in stem cell fate determination and function. In this review, we provided a brief overview of the biological properties of relevant stem cells. The influence of epigenetic modifications on stem cell proliferation, differentiation, and immune regulatory function in the treatment of AT was also explored. We focused on gene regulatory mechanisms controlled by DNA methylation, histones and non-coding RNAs including microRNAs, circRNAs and long non-coding RNAs. We also discuss the current challenges faced by stem cell therapies in treating AT and their potential solutions. Further research in this area will provide a more comprehensive epigenetic explanation for stem cell therapy for AT, leading to the development of stable, safe and effective stem cell therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Haijiao Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
4
|
Wang Y, Yung P, Lu G, Liu Y, Ding C, Mao C, Li ZA, Tuan RS. Musculoskeletal Organs-on-Chips: An Emerging Platform for Studying the Nanotechnology-Biology Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2401334. [PMID: 38491868 PMCID: PMC11733728 DOI: 10.1002/adma.202401334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Nanotechnology-based approaches are promising for the treatment of musculoskeletal (MSK) disorders, which present significant clinical burdens and challenges, but their clinical translation requires a deep understanding of the complex interplay between nanotechnology and MSK biology. Organ-on-a-chip (OoC) systems have emerged as an innovative and versatile microphysiological platform to replicate the dynamics of tissue microenvironment for studying nanotechnology-biology interactions. This review first covers recent advances and applications of MSK OoCs and their ability to mimic the biophysical and biochemical stimuli encountered by MSK tissues. Next, by integrating nanotechnology into MSK OoCs, cellular responses and tissue behaviors may be investigated by precisely controlling and manipulating the nanoscale environment. Analysis of MSK disease mechanisms, particularly bone, joint, and muscle tissue degeneration, and drug screening and development of personalized medicine may be greatly facilitated using MSK OoCs. Finally, future challenges and directions are outlined for the field, including advanced sensing technologies, integration of immune-active components, and enhancement of biomimetic functionality. By highlighting the emerging applications of MSK OoCs, this review aims to advance the understanding of the intricate nanotechnology-MSK biology interface and its significance in MSK disease management, and the development of innovative and personalized therapeutic and interventional strategies.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Patrick Yung
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Gang Lu
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Yuwei Liu
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- The First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhenGuangdong518037P. R. China
| | - Changhai Ding
- Clinical Research CentreZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510260China
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Zhong Alan Li
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Key Laboratory of Regenerative MedicineMinistry of EducationSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077P. R. China
- Shenzhen Research InstituteThe Chinese University of Hong KongShenzhen518172P. R. China
| | - Rocky S. Tuan
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| |
Collapse
|
5
|
Zhang W, Rao Y, Wong SH, Wu Y, Zhang Y, Yang R, Tsui SK, Ker DFE, Mao C, Frith JE, Cao Q, Tuan RS, Wang DM. Transcriptome-Optimized Hydrogel Design of a Stem Cell Niche for Enhanced Tendon Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2313722. [PMID: 39417770 PMCID: PMC11733723 DOI: 10.1002/adma.202313722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Bioactive hydrogels have emerged as promising artificial niches for enhancing stem cell-mediated tendon repair. However, a substantial knowledge gap remains regarding the optimal combination of niche features for targeted cellular responses, which often leads to lengthy development cycles and uncontrolled healing outcomes. To address this critical gap, an innovative, data-driven materiomics strategy is developed. This approach is based on in-house RNA-seq data that integrates bioinformatics and mathematical modeling, which is a significant departure from traditional trial-and-error methods. It aims to provide both mechanistic insights and quantitative assessments and predictions of the tenogenic effects of adipose-derived stem cells induced by systematically modulated features of a tendon-mimetic hydrogel (TenoGel). The knowledge generated has enabled a rational approach for TenoGel design, addressing key considerations, such as tendon extracellular matrix concentration, uniaxial tensile loading, and in vitro pre-conditioning duration. Remarkably, our optimized TenoGel demonstrated robust tenogenesis in vitro and facilitated tendon regeneration while preventing undesired ectopic ossification in a rat tendon injury model. These findings shed light on the importance of tailoring hydrogel features for efficient tendon repair. They also highlight the tremendous potential of the innovative materiomics strategy as a powerful predictive and assessment tool in biomaterial development for regenerative medicine.
Collapse
Affiliation(s)
- Wanqi Zhang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Ying Rao
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Shing Hei Wong
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Yeung Wu
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Yuanhao Zhang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Rui Yang
- Department of Sports MedicineOrthopedicsSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Stephen Kwok‐Wing Tsui
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Dai Fei Elmer Ker
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongHong Kong SARChina
| | - Jessica E. Frith
- Materials Science and EngineeringMonash UniversityClayton3800VICAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClayton3800VICAustralia
- Australian Research Council Training Centre for Cell and Tissue Engineering TechnologiesMonash UniversityClayton3800VICAustralia
| | - Qin Cao
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Rocky S. Tuan
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| | - Dan Michelle Wang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| |
Collapse
|
6
|
Morya VK, Shahid H, Lang J, Kwak MK, Park SH, Noh KC. Advancements in Therapeutic Approaches for Degenerative Tendinopathy: Evaluating Efficacy and Challenges. Int J Mol Sci 2024; 25:11846. [PMID: 39519397 PMCID: PMC11545934 DOI: 10.3390/ijms252111846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
Degenerative tendinopathy results from the accumulation of minor injuries following unsuccessful tendon repair during acute tendon injuries. The process of tendon repair is prolonged and varies between individuals, making it susceptible to reinjury. Moreover, treating chronic tendinopathy often requires expensive and extensive rehabilitation, along with a variety of combined therapies to facilitate recovery. This condition significantly affects the quality of life of affected individuals, underscoring the urgent need for more efficient and cost-effective treatment options. Although traditional treatments have improved significantly and are being used as substitutes for surgical interventions, the findings have been inconsistent and conflicting. This review aims to clarify these issues by exploring the strengths and limitations of current treatments as well as recent innovations in managing various forms of degenerative tendinopathy.
Collapse
Affiliation(s)
- Vivek Kumar Morya
- Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si 18450, Republic of Korea; (V.K.M.); (J.L.)
- School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hamzah Shahid
- Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si 18450, Republic of Korea; (V.K.M.); (J.L.)
- School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jun Lang
- Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si 18450, Republic of Korea; (V.K.M.); (J.L.)
- School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Mi Kyung Kwak
- Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si 18450, Republic of Korea; (V.K.M.); (J.L.)
- School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sin-Hye Park
- Department of Food Science & Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyu-Cheol Noh
- School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Hallym University Sacred Heart Hospital, Anyang-si 14068, Republic of Korea
| |
Collapse
|
7
|
Zhang X, Li M, Mao X, Yao Z, Zhu W, Yuan Z, Gao X, Pan S, Zhang Y, Zhao J, Mao H. Small Intestinal Submucosa Hydrogel Loaded With Gastrodin for the Repair of Achilles Tendinopathy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401886. [PMID: 39185812 DOI: 10.1002/smll.202401886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Achilles tendinopathy (AT) is an injury caused by overuse of the Achilles tendon or sudden force on the Achilles tendon, with a considerable inflammatory infiltrate. As Achilles tendinopathy progresses, inflammation and inflammatory factors affect the remodeling of the extracellular matrix (ECM) of the tendon. Gastrodin(Gas), the main active ingredient of Astrodia has anti-inflammatory, antioxidant, and anti-apoptotic properties. The small intestinal submucosa (SIS) is a naturally decellularized extracellular matrix(dECM)material and has a high content of growth factors as well as good biocompatibility. However, the reparative effects of SIS and Gas on Achilles tendinopathy and their underlying mechanisms remain unknown. Here, it is found that SIS hydrogel loaded with gastrodin restored the mechanical strength of the Achilles tendon, facilitated ECM remodeling, and restored ordered collagen arrangement by promoting the translocation of protein synthesis. It also decreases the expression of inflammatory factors and reduces the infiltration of inflammatory cells by inhibiting the NF-κB signaling pathway. It is believed that through further research, Gas + SIS may be used in the future for the treatment of Achilles tendinopathy and other Achilles tendon injury disorders.
Collapse
Affiliation(s)
- Xiqian Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Mei Li
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Xufeng Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Zheyu Yao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Weilai Zhu
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Zheyang Yuan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Xiang Gao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| | - Senghao Pan
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Yijun Zhang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Jiyuan Zhao
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, P. R. China
| |
Collapse
|
8
|
Lei L, Wen Z, Cao M, Zhang H, Ling SKK, Fu BSC, Qin L, Xu J, Yung PSH. The emerging role of Piezo1 in the musculoskeletal system and disease. Theranostics 2024; 14:3963-3983. [PMID: 38994033 PMCID: PMC11234281 DOI: 10.7150/thno.96959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/15/2024] [Indexed: 07/13/2024] Open
Abstract
Piezo1, a mechanosensitive ion channel, has emerged as a key player in translating mechanical stimuli into biological signaling. Its involvement extends beyond physiological and pathological processes such as lymphatic vessel development, axon growth, vascular development, immunoregulation, and blood pressure regulation. The musculoskeletal system, responsible for structural support, movement, and homeostasis, has recently attracted attention regarding the significance of Piezo1. This review aims to provide a comprehensive summary of the current research on Piezo1 in the musculoskeletal system, highlighting its impact on bone formation, myogenesis, chondrogenesis, intervertebral disc homeostasis, tendon matrix cross-linking, and physical activity. Additionally, we explore the potential of targeting Piezo1 as a therapeutic approach for musculoskeletal disorders, including osteoporosis, muscle atrophy, intervertebral disc degeneration, and osteoarthritis.
Collapse
Affiliation(s)
- Lei Lei
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhenkang Wen
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mingde Cao
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haozhi Zhang
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Samuel Ka-Kin Ling
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bruma Sai-Chuen Fu
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ling Qin
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Sir Yue-Kong Pao Cancer Centre, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Sir Yue-Kong Pao Cancer Centre, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Patrick Shu-Hang Yung
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
9
|
Kimura K, Tsukamoto M, Sugisaki H, Tanaka M, Kuwamura M, Matsumoto Y, Ishihara G, Watanabe K, Okada M, Nakanishi M, Sugiura K, Hatoya S. Generation of footprint-free, high-quality feline induced pluripotent stem cells using Sendai virus vector. Regen Ther 2024; 26:708-716. [PMID: 39286639 PMCID: PMC11403254 DOI: 10.1016/j.reth.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/04/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
Companion animals, such as felines and canines, could provide an excellent platform for translational research from veterinary to human medicine. However, the use of feline induced pluripotent stems (fiPSCs) of quality in basic or clinical research has not been reported. Here, we generated footprint-free fiPSCs derived from embryonic cells, as well as juvenile feline uterus-derived cells using Sendai virus vector harboring six feline-specific pluripotency-associated genes. The fiPSCs were confirmed to be of high quality with the potential to form teratomas including all three germ layers. Furthermore, our fiPSCs were maintained under feeder-free and chemically-defined conditions using StemFit® AK02N and recombinant laminin 511, iMatrix-511. Further research on fiPSCs could result in their widespread application in veterinary regenerative medicine, which could pave the way for their use in advanced regenerative medicine research for humans.
Collapse
Affiliation(s)
- Kazuto Kimura
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Masaya Tsukamoto
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Hiroko Sugisaki
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Miyuu Tanaka
- Department of Integrated Structural Biosciences, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Mitsuru Kuwamura
- Department of Integrated Structural Biosciences, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Yuki Matsumoto
- Anicom Specialty Medical Institute, Inc., Shinjuku-ku, Tokyo 231-0033, Japan
| | - Genki Ishihara
- Anicom Specialty Medical Institute, Inc., Shinjuku-ku, Tokyo 231-0033, Japan
| | - Kei Watanabe
- Anicom Specialty Medical Institute, Inc., Shinjuku-ku, Tokyo 231-0033, Japan
| | - Mika Okada
- TOKIWA-Bio, Inc., Tsukuba, Ibaraki 305-0047, Japan
| | | | - Kikuya Sugiura
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Shingo Hatoya
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
10
|
Augustin G, Jeong JH, Kim M, Hur SS, Lee JH, Hwang Y. Stem Cell‐Based Therapies and Tissue Engineering Innovations for Tendinopathy: A Comprehensive Review of Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202300425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Indexed: 01/06/2025]
Abstract
AbstractTendon diseases commonly lead to physical disability, exerting a profound impact on the routine of affected patients. These conditions respond poorly to existing treatments, presenting a substantial challenge for orthopedic scientists. Research into clinical translational therapy has yet to yield highly versatile interventions capable of effectively addressing tendon diseases, including tendinopathy. Stem cell‐based therapies have emerged as a promising avenue for modifying the biological milieu through the secretion of regenerative and immunomodulatory factors. The current review provides an overview of the intricate tendon microenvironment, encompassing various tendon stem progenitor cells within distinct tendon sublocations, gene regulation, and pathways pertinent to tendon development, and the pathology of tendon diseases. Subsequently, the advantages of stem cell‐based therapies are discussed that utilize distinct types of autologous and allogeneic stem cells for tendon regeneration at the translational level. Moreover, this review outlines the challenges, gaps, and future innovations to propose a consolidated stem cell‐based therapy to treat tendinopathy. Finally, regenerative soluble therapies, insoluble bio‐active therapies, along with insoluble bio‐active therapies, and implantable 3D scaffolds for tendon tissue engineering are discussed, thereby presenting a pathway toward enhanced tissue regeneration and engineering.
Collapse
Affiliation(s)
- George Augustin
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Biochemistry and Biophysics Oregon State University Corvallis OR 92331 USA
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Integrated Biomedical Science Soonchunhyang University Asan‐si, Chungnam‐Do 31538 Republic of Korea
| | - Min‐Kyu Kim
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
| | - Sung Sik Hur
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
| | - Joon Ho Lee
- Department of Anesthesiology and Pain Medicine Soonchunhyang University Bucheon Hospital Soonchunhyang University College of Medicine Bucheon‐Si 14584 Republic of Korea
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi‐bio Science (SIMS) Soonchunhyang University Cheonan‐Si, Chungnam‐Do 31151 Republic of Korea
- Department of Integrated Biomedical Science Soonchunhyang University Asan‐si, Chungnam‐Do 31538 Republic of Korea
| |
Collapse
|
11
|
Tsuchiya Y, Takakura H, Osawa S, Izawa T. High-intensity interval training enhances mRNA expression of IGF1Ea in rat Achilles tendon. Mol Biol Rep 2024; 51:374. [PMID: 38421500 DOI: 10.1007/s11033-024-09306-x] [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: 10/19/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
High-intensity interval training (HIIT) reportedly enhances the functional properties of the musculoskeletal system. However, the effects of HIIT on tendons remain unclear. Sixteen male rats were randomly assigned to the control (Con) or HIIT group (n = 8 in each group). Rats in the HIIT group executed the HIIT program consisting of 2.5 min treadmill running and 4.5 min rests between the bouts, 5 days per week for 9 weeks. Running speed, number of sets, and inclination were incrementally increased during the training period. Histological analysis revealed no apparent morphological changes in the extracellular matrix structure or nuclei of tenocytes between the groups. Real-time reverse transcription polymerase chain reaction analysis revealed that Igf1Ea mRNA expression was enhanced in the HIIT group. Furthermore, Igfbp5 mRNA expression tended to be higher in the HIIT group. The 9-week HIIT program enhanced tenogenic Igf1Ea mRNA expression.
Collapse
Affiliation(s)
- Yoshifumi Tsuchiya
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto, 610-0394, Japan.
| | - Hisashi Takakura
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto, 610-0394, Japan
| | - Seita Osawa
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto, 610-0394, Japan
| | - Tetsuya Izawa
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto, 610-0394, Japan
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyoto, 610-0394, Japan
| |
Collapse
|
12
|
Hu Y, Zhang H, Wang S, Cao L, Zhou F, Jing Y, Su J. Bone/cartilage organoid on-chip: Construction strategy and application. Bioact Mater 2023; 25:29-41. [PMID: 37056252 PMCID: PMC10087111 DOI: 10.1016/j.bioactmat.2023.01.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
The necessity of disease models for bone/cartilage related disorders is well-recognized, but the barrier between ex-vivo cell culture, animal models and the real human body has been pending for decades. The organoid-on-a-chip technique showed opportunity to revolutionize basic research and drug screening for diseases like osteoporosis and arthritis. The bone/cartilage organoid on-chip (BCoC) system is a novel platform of multi-tissue which faithfully emulate the essential elements, biologic functions and pathophysiological response under real circumstances. In this review, we propose the concept of BCoC platform, summarize the basic modules and current efforts to orchestrate them on a single microfluidic system. Current disease models, unsolved problems and future challenging are also discussed, the aim should be a deeper understanding of diseases, and ultimate realization of generic ex-vivo tools for further therapeutic strategies of pathological conditions.
Collapse
|
13
|
Tian R, Xue Z, Ruan D, Chen P, Xu Y, Dai C, Shen W, Ouyang H, Liu W, Lin J. MSdb: An integrated expression atlas of human musculoskeletal system. iScience 2023; 26:106933. [PMID: 37378342 PMCID: PMC10291471 DOI: 10.1016/j.isci.2023.106933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/26/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
The global prevalence and burden of musculoskeletal (MSK) disorders are immense. Advancements in next-generation sequencing (NGS) have generated vast amounts of data, accelerating the research of pathological mechanisms and the development of therapeutic approaches for MSK disorders. However, scattered datasets across various repositories complicate uniform analysis and comparison. Here, we introduce MSdb, a database for visualization and integrated analysis of next-generation sequencing data from human musculoskeletal system, along with manually curated patient phenotype data. MSdb provides various types of analysis, including sample-level browsing of metadata information, gene/miRNA expression, and single-cell RNA-seq dataset. In addition, MSdb also allows integrated analysis for cross-samples and cross-omics analysis, including customized differentially expressed gene/microRNA analysis, microRNA-gene network, scRNA-seq cross-sample/disease integration, and gene regulatory network analysis. Overall, systematic categorizing, standardized processing, and freely accessible knowledge features MSdb a valuable resource for MSK research community.
Collapse
Affiliation(s)
- Ruonan Tian
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China
| | - Ziwei Xue
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China
| | - Dengfeng Ruan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Pengwei Chen
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yiwen Xu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Chao Dai
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Weiliang Shen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Hongwei Ouyang
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang 310058, China
| | - Wanlu Liu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Junxin Lin
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
14
|
Huang L, Chen L, Chen H, Wang M, Jin L, Zhou S, Gao L, Li R, Li Q, Wang H, Zhang C, Wang J. Biomimetic Scaffolds for Tendon Tissue Regeneration. Biomimetics (Basel) 2023; 8:246. [PMID: 37366841 DOI: 10.3390/biomimetics8020246] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Tendon tissue connects muscle to bone and plays crucial roles in stress transfer. Tendon injury remains a significant clinical challenge due to its complicated biological structure and poor self-healing capacity. The treatments for tendon injury have advanced significantly with the development of technology, including the use of sophisticated biomaterials, bioactive growth factors, and numerous stem cells. Among these, biomaterials that the mimic extracellular matrix (ECM) of tendon tissue would provide a resembling microenvironment to improve efficacy in tendon repair and regeneration. In this review, we will begin with a description of the constituents and structural features of tendon tissue, followed by a focus on the available biomimetic scaffolds of natural or synthetic origin for tendon tissue engineering. Finally, we will discuss novel strategies and present challenges in tendon regeneration and repair.
Collapse
Affiliation(s)
- Lvxing Huang
- School of Savaid Stomatology, Hangzhou Medical College, Hangzhou 310000, China
| | - Le Chen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310000, China
| | - Hengyi Chen
- School of Savaid Stomatology, Hangzhou Medical College, Hangzhou 310000, China
| | - Manju Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310000, China
| | - Letian Jin
- School of Medical Imaging, Hangzhou Medical College, Hangzhou 310000, China
| | - Shenghai Zhou
- School of Medical Imaging, Hangzhou Medical College, Hangzhou 310000, China
| | - Lexin Gao
- School of Savaid Stomatology, Hangzhou Medical College, Hangzhou 310000, China
| | - Ruwei Li
- School of Savaid Stomatology, Hangzhou Medical College, Hangzhou 310000, China
| | - Quan Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310000, China
| | - Hanchang Wang
- School of Medical Imaging, Hangzhou Medical College, Hangzhou 310000, China
| | - Can Zhang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Junjuan Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310000, China
| |
Collapse
|
15
|
Wang H, Yu R, Wang M, Wang S, Ouyang X, Yan Z, Chen S, Wang W, Wu F, Fan C. Insulin-like growth factor binding protein 4 loaded electrospun membrane ameliorating tendon injury by promoting retention of IGF-1. J Control Release 2023; 356:162-174. [PMID: 36868516 DOI: 10.1016/j.jconrel.2023.02.039] [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: 11/24/2022] [Revised: 02/18/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023]
Abstract
Tendon injury is one of the most common musculoskeletal disorders that impair joint mobility and lower quality of life. The limited regenerative capacity of tendon remains a clinical challenge. Local delivery of bioactive protein is a viable therapeutic approach for tendon healing. Insulin-like growth factor binding protein 4 (IGFBP-4) is a secreted protein capable of binding and stabilizing insulin-like growth factor 1 (IGF-1). Here, we applied an aqueous-aqueous freezing-induced phase separation technology to obtain the IGFBP4-encapsulated dextran particles. Then, we added the particles into poly (L-lactic acid) (PLLA) solution to fabricate IGFBP4-PLLA electrospun membrane for efficient IGFBP-4 delivery. The scaffold showed excellent cytocompatibility and a sustained release of IGFBP-4 for nearly 30 days. In cellular experiments, IGFBP-4 promoted tendon-related and proliferative markers expression. In a rat Achilles tendon injury model, immunohistochemistry and quantitative real-time polymerase chain reaction confirmed better outcomes by using the IGFBP4-PLLA electrospun membrane at the molecular level. Furthermore, the scaffold effectively promoted tendon healing in functional performance, ultrastructure and biomechanical properties. We found addition of IGFBP-4 promoted IGF-1 retention in tendon postoperatively and then facilitated protein synthesis via IGF-1/AKT signaling pathway. Overall, our IGFBP4-PLLA electrospun membrane provides a promising therapeutic strategy for tendon injury.
Collapse
Affiliation(s)
- Hui Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Ruyue Yu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Meng Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shikun Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Xingyu Ouyang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Zhiwen Yan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Shuai Chen
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Wei Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China.
| | - Fei Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China.
| |
Collapse
|
16
|
Mechanisms of skeletal muscle-tendon development and regeneration/healing as potential therapeutic targets. Pharmacol Ther 2023; 243:108357. [PMID: 36764462 DOI: 10.1016/j.pharmthera.2023.108357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
Skeletal muscle contraction is essential for the movement of our musculoskeletal system. Tendons and ligaments that connect the skeletal muscles to bones in the correct position at the appropriate time during development are also required for movement to occur. Since the musculoskeletal system is essential for maintaining basic bodily functions as well as enabling interactions with the environment, dysfunctions of these tissues due to disease can significantly reduce quality of life. Unfortunately, as people live longer, skeletal muscle and tendon/ligament diseases are becoming more common. Sarcopenia, a disease in which skeletal muscle function declines, and tendinopathy, which involves chronic tendon dysfunction, are particularly troublesome because there have been no significant advances in their treatment. In this review, we will summarize previous reports on the development and regeneration/healing of skeletal muscle and tendon tissues, including a discussion of the molecular and cellular mechanisms involved that may be used as potential therapeutic targets.
Collapse
|
17
|
Shao X, Lin X, Zhu S, Zhou H, Lu Z, Zhang Y, Wang J. Human Muscle-Derived Cells Are Capable of Tenogenic Differentiation and Contribution to Tendon Repair. Am J Sports Med 2023; 51:786-797. [PMID: 36734484 DOI: 10.1177/03635465221147486] [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] [Indexed: 02/04/2023]
Abstract
BACKGROUND It has been reported that the harvested hamstring tendon for autograft could be regenerated with well-oriented fibers and uniformly distributed spindle-shaped cells after removal. However, which cell type might participate in the repair process remains unknown. PURPOSE To investigate the tenogenic differentiation potential of human muscle-derived cells (MDCs) both in vitro and in vivo. STUDY DESIGN Controlled laboratory study. METHODS Primary human MDCs and tenocytes were isolated from discarded materials during a peroneus longus tendon-harvesting procedure. Expression of tenogenic genes were evaluated and compared among MDCs, MDCs with tenogenic induction, and tenocytes. RNA sequencing was performed to evaluate the expression profile of differentiated MDCs. Human MDCs were implanted in a tendon injury model to investigate the in vivo tenogenic differentiation potential. Histologic and functional analyses were performed to evaluate the function of MDCs for tendon repair. RESULTS The relative expression levels (in fold change) of tenogenic genes Col I, MKX, SCX, THBS4, and TNC in MDCs were significantly upregulated 11.5 ± 1.3, 957.1 ± 63.7, 19.1 ± 2.8, 61.9 ± 4.8, and 10.2 ± 2.8 after tenogenic induction, respectively. The expression profile of tenogenically differentiated MDCs was much closer to primary tenocytes. Activation of TGF-β/Smad3 signaling significantly promoted the tenogenic differentiation ability of MDCs. Transplanted human MDCs were identified in regenerated tendon and expressed tenogenic genes. As for biomechanical properties, the failure loads in the Matrigel, transplantation, and uninjured groups were 7.2 ± 0.5, 11.6 ± 0.3, and 13.9 ± 0.7 N, while the stiffness values were 4.4 ± 1.3 × 103, 7.6 ± 0.8 × 103, and 10.9 ± 1.1 × 103 N/m. Plantarflexion force, histologic morphology, and motor function were also significantly improved after MDC transplantation in a tendon injury model. CONCLUSION There exist cells with tenogenic differentiation potential in human skeletal muscles. Activation of TGF-β/Smad3 signaling plays an important role in tenogenic differentiation for human MDCs. Human MDCs contribute to structural and functional repair for the injured tendon. MDCs are a potential cell source to participate in the repair process after tendon injury. CLINICAL RELEVANCE The MDCs could be a promising cell source to repair tendon injury.
Collapse
Affiliation(s)
- Xiexiang Shao
- Department of Orthopaedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingzuan Lin
- Department of Orthopaedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyuan Zhu
- Department of Orthopaedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Zhou
- Department of Orthopaedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenfei Lu
- Department of Sports Medicine, Wuxi Hospital of Traditional Chinese Medicine, Wuxi, China
| | - Yuanyuan Zhang
- Centre Testing International Medical Laboratory, Shanghai, China
| | - Jianhua Wang
- Department of Orthopaedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Single-cell RNA sequencing in orthopedic research. Bone Res 2023; 11:10. [PMID: 36828839 PMCID: PMC9958119 DOI: 10.1038/s41413-023-00245-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 02/26/2023] Open
Abstract
Although previous RNA sequencing methods have been widely used in orthopedic research and have provided ideas for therapeutic strategies, the specific mechanisms of some orthopedic disorders, including osteoarthritis, lumbar disc herniation, rheumatoid arthritis, fractures, tendon injuries, spinal cord injury, heterotopic ossification, and osteosarcoma, require further elucidation. The emergence of the single-cell RNA sequencing (scRNA-seq) technique has introduced a new era of research on these topics, as this method provides information regarding cellular heterogeneity, new cell subtypes, functions of novel subclusters, potential molecular mechanisms, cell-fate transitions, and cell‒cell interactions that are involved in the development of orthopedic diseases. Here, we summarize the cell subpopulations, genes, and underlying mechanisms involved in the development of orthopedic diseases identified by scRNA-seq, improving our understanding of the pathology of these diseases and providing new insights into therapeutic approaches.
Collapse
|
19
|
Ning C, Li P, Gao C, Fu L, Liao Z, Tian G, Yin H, Li M, Sui X, Yuan Z, Liu S, Guo Q. Recent advances in tendon tissue engineering strategy. Front Bioeng Biotechnol 2023; 11:1115312. [PMID: 36890920 PMCID: PMC9986339 DOI: 10.3389/fbioe.2023.1115312] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Tendon injuries often result in significant pain and disability and impose severe clinical and financial burdens on our society. Despite considerable achievements in the field of regenerative medicine in the past several decades, effective treatments remain a challenge due to the limited natural healing capacity of tendons caused by poor cell density and vascularization. The development of tissue engineering has provided more promising results in regenerating tendon-like tissues with compositional, structural and functional characteristics comparable to those of native tendon tissues. Tissue engineering is the discipline of regenerative medicine that aims to restore the physiological functions of tissues by using a combination of cells and materials, as well as suitable biochemical and physicochemical factors. In this review, following a discussion of tendon structure, injury and healing, we aim to elucidate the current strategies (biomaterials, scaffold fabrication techniques, cells, biological adjuncts, mechanical loading and bioreactors, and the role of macrophage polarization in tendon regeneration), challenges and future directions in the field of tendon tissue engineering.
Collapse
Affiliation(s)
- Chao Ning
- Chinese PLA Medical School, Beijing, China
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Pinxue Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Cangjian Gao
- Chinese PLA Medical School, Beijing, China
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Liwei Fu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Zhiyao Liao
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Guangzhao Tian
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Han Yin
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Muzhe Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xiang Sui
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Zhiguo Yuan
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuyun Liu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Quanyi Guo
- Chinese PLA Medical School, Beijing, China
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
20
|
Feng S, Li J, Tian J, Lu S, Zhao Y. Application of Single-Cell and Spatial Omics in Musculoskeletal Disorder Research. Int J Mol Sci 2023; 24:2271. [PMID: 36768592 PMCID: PMC9917071 DOI: 10.3390/ijms24032271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Musculoskeletal disorders, including fractures, scoliosis, heterotopic ossification, osteoporosis, osteoarthritis, disc degeneration, and muscular injury, etc., can occur at any stage of human life. Understanding the occurrence and development mechanism of musculoskeletal disorders, as well as the changes in tissues and cells during therapy, might help us find targeted treatment methods. Single-cell techniques provide excellent tools for studying alterations at the cellular level of disorders. However, the application of these techniques in research on musculoskeletal disorders is still limited. This review summarizes the current single-cell and spatial omics used in musculoskeletal disorders. Cell isolation, experimental methods, and feasible experimental designs for single-cell studies of musculoskeletal system diseases have been reviewed based on tissue characteristics. Then, the paper summarizes the latest findings of single-cell studies in musculoskeletal disorders from three aspects: bone and ossification, joint, and muscle and tendon disorders. Recent discoveries about the cell populations involved in these diseases are highlighted. Furthermore, the therapeutic responses of musculoskeletal disorders, especially single-cell changes after the treatments of implants, stem cell therapies, and drugs are described. Finally, the application potential and future development directions of single-cell and spatial omics in research on musculoskeletal diseases are discussed.
Collapse
Affiliation(s)
- Site Feng
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jiahao Li
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| | - Jingjing Tian
- Medical Science Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Sheng Lu
- The Key Laboratory of Digital Orthopaedics of Yunnan Provincial, Department of Orthopedic Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Yu Zhao
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| |
Collapse
|
21
|
Satake T, Komura S, Aoki H, Hirakawa A, Imai Y, Akiyama H. Induction of iPSC-derived Prg4-positive cells with characteristics of superficial zone chondrocytes and fibroblast-like synovial cells. BMC Mol Cell Biol 2022; 23:30. [PMID: 35870887 PMCID: PMC9308249 DOI: 10.1186/s12860-022-00431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022] Open
Abstract
Background Lubricin, a proteoglycan encoded by the PRG4 gene, is synthesised by superficial zone (SFZ) chondrocytes and synovial cells. It reduces friction between joints and allows smooth sliding of tendons. Although lubricin has been shown to be effective against osteoarthritis and synovitis in animals, its clinical application remains untested. In this study, we aimed to induce lubricin-expressing cells from pluripotent stem cells (iPSCs) and applied them locally via cell transplantation. Methods To generate iPSCs, OCT3/4, SOX2, KLF4, and L-MYC were transduced into fibroblasts derived from Prg4-mRFP1 transgenic mice. We established a protocol for the differentiation of iPSC-derived Prg4-mRFP1-positive cells and characterised their mRNA expression profile. Finally, we injected Prg4-mRFP1-positive cells into the paratenon, surrounding the Achilles tendons and knee joints of severe combined immunodeficient mice and assessed lubricin expression. Result Wnt3a, activin A, TGF-β1, and bFGF were applied to induce the differentiation of iPSC-derived Prg4-mRFP1-positive cells. Markers related to SFZ chondrocytes and fibroblast-like synovial cells (FLSs) were expressed during differentiation. RNA-sequencing indicated that iPSC-derived Prg4-mRFP1-positive cells manifested expression profiles typical of SFZ chondrocytes and FLSs. Transplanted iPSC-derived Prg4-mRFP1-positive cells survived around the Achilles tendons and in knee joints. Conclusions The present study describes a protocol for the differentiation of iPSC-derived Prg4-positive cells with characteristics of SFZ chondrocytes and FLSs. Transplantation of lubricin-expressing cells offers promise as a therapy against arthritis and synovitis.
Collapse
|
22
|
Schulze-Tanzil GG, Delgado-Calcares M, Stange R, Wildemann B, Docheva D. Tendon healing: a concise review on cellular and molecular mechanisms with a particular focus on the Achilles tendon. Bone Joint Res 2022; 11:561-574. [PMID: 35920195 PMCID: PMC9396922 DOI: 10.1302/2046-3758.118.bjr-2021-0576.r1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tendon is a bradytrophic and hypovascular tissue, hence, healing remains a major challenge. The molecular key events involved in successful repair have to be unravelled to develop novel strategies that reduce the risk of unfavourable outcomes such as non-healing, adhesion formation, and scarring. This review will consider the diverse pathophysiological features of tendon-derived cells that lead to failed healing, including misrouted differentiation (e.g. de- or transdifferentiation) and premature cell senescence, as well as the loss of functional progenitors. Many of these features can be attributed to disturbed cell-extracellular matrix (ECM) or unbalanced soluble mediators involving not only resident tendon cells, but also the cross-talk with immigrating immune cell populations. Unrestrained post-traumatic inflammation could hinder successful healing. Pro-angiogenic mediators trigger hypervascularization and lead to persistence of an immature repair tissue, which does not provide sufficient mechano-competence. Tendon repair tissue needs to achieve an ECM composition, structure, strength, and stiffness that resembles the undamaged highly hierarchically ordered tendon ECM. Adequate mechano-sensation and -transduction by tendon cells orchestrate ECM synthesis, stabilization by cross-linking, and remodelling as a prerequisite for the adaptation to the increased mechanical challenges during healing. Lastly, this review will discuss, from the cell biological point of view, possible optimization strategies for augmenting Achilles tendon (AT) healing outcomes, including adapted mechanostimulation and novel approaches by restraining neoangiogenesis, modifying stem cell niche parameters, tissue engineering, the modulation of the inflammatory cells, and the application of stimulatory factors.Cite this article: Bone Joint Res 2022;11(8):561-574.
Collapse
Affiliation(s)
| | - Manuel Delgado-Calcares
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute for Musculoskeletal Medicine (IMM), University Hospital Münster, Münster, Germany
| | - Britt Wildemann
- Department of Experimental Trauma Surgery, University Hospital Jena, Jena, Germany
| | - Denitsa Docheva
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Zhu S, He Z, Ji L, Zhang W, Tong Y, Luo J, Zhang Y, Li Y, Meng X, Bi Q. Advanced Nanofiber-Based Scaffolds for Achilles Tendon Regenerative Engineering. Front Bioeng Biotechnol 2022; 10:897010. [PMID: 35845401 PMCID: PMC9280267 DOI: 10.3389/fbioe.2022.897010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/20/2022] [Indexed: 11/22/2022] Open
Abstract
The Achilles tendon (AT) is responsible for running, jumping, and standing. The AT injuries are very common in the population. In the adult population (21–60 years), the incidence of AT injuries is approximately 2.35 per 1,000 people. It negatively impacts people’s quality of life and increases the medical burden. Due to its low cellularity and vascular deficiency, AT has a poor healing ability. Therefore, AT injury healing has attracted a lot of attention from researchers. Current AT injury treatment options cannot effectively restore the mechanical structure and function of AT, which promotes the development of AT regenerative tissue engineering. Various nanofiber-based scaffolds are currently being explored due to their structural similarity to natural tendon and their ability to promote tissue regeneration. This review discusses current methods of AT regeneration, recent advances in the fabrication and enhancement of nanofiber-based scaffolds, and the development and use of multiscale nanofiber-based scaffolds for AT regeneration.
Collapse
Affiliation(s)
- Senbo Zhu
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zeju He
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lichen Ji
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Zhang
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yu Tong
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Junchao Luo
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yin Zhang
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yong Li
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Xiang Meng
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Qing Bi
- Center for Rehabilitation Medicine, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qing Bi,
| |
Collapse
|
24
|
Watson-Levings RS, Palmer GD, Levings PP, Dacanay EA, Evans CH, Ghivizzani SC. Gene Therapy in Orthopaedics: Progress and Challenges in Pre-Clinical Development and Translation. Front Bioeng Biotechnol 2022; 10:901317. [PMID: 35837555 PMCID: PMC9274665 DOI: 10.3389/fbioe.2022.901317] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
In orthopaedics, gene-based treatment approaches are being investigated for an array of common -yet medically challenging- pathologic conditions of the skeletal connective tissues and structures (bone, cartilage, ligament, tendon, joints, intervertebral discs etc.). As the skeletal system protects the vital organs and provides weight-bearing structural support, the various tissues are principally composed of dense extracellular matrix (ECM), often with minimal cellularity and vasculature. Due to their functional roles, composition, and distribution throughout the body the skeletal tissues are prone to traumatic injury, and/or structural failure from chronic inflammation and matrix degradation. Due to a mixture of environment and endogenous factors repair processes are often slow and fail to restore the native quality of the ECM and its function. In other cases, large-scale lesions from severe trauma or tumor surgery, exceed the body’s healing and regenerative capacity. Although a wide range of exogenous gene products (proteins and RNAs) have the potential to enhance tissue repair/regeneration and inhibit degenerative disease their clinical use is hindered by the absence of practical methods for safe, effective delivery. Cumulatively, a large body of evidence demonstrates the capacity to transfer coding sequences for biologic agents to cells in the skeletal tissues to achieve prolonged delivery at functional levels to augment local repair or inhibit pathologic processes. With an eye toward clinical translation, we discuss the research progress in the primary injury and disease targets in orthopaedic gene therapy. Technical considerations important to the exploration and pre-clinical development are presented, with an emphasis on vector technologies and delivery strategies whose capacity to generate and sustain functional transgene expression in vivo is well-established.
Collapse
Affiliation(s)
- Rachael S. Watson-Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Glyn D. Palmer
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Padraic P. Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - E. Anthony Dacanay
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Christopher H. Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MI, United States
| | - Steven C. Ghivizzani
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
- *Correspondence: Steven C. Ghivizzani,
| |
Collapse
|
25
|
Gomez-Florit M, Labrador-Rached CJ, Domingues RM, Gomes ME. The tendon microenvironment: Engineered in vitro models to study cellular crosstalk. Adv Drug Deliv Rev 2022; 185:114299. [PMID: 35436570 DOI: 10.1016/j.addr.2022.114299] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022]
Abstract
Tendinopathy is a multi-faceted pathology characterized by alterations in tendon microstructure, cellularity and collagen composition. Challenged by the possibility of regenerating pathological or ruptured tendons, the healing mechanisms of this tissue have been widely researched over the past decades. However, so far, most of the cellular players and processes influencing tendon repair remain unknown, which emphasizes the need for developing relevant in vitro models enabling to study the complex multicellular crosstalk occurring in tendon microenvironments. In this review, we critically discuss the insights on the interaction between tenocytes and the other tendon resident cells that have been devised through different types of existing in vitro models. Building on the generated knowledge, we stress the need for advanced models able to mimic the hierarchical architecture, cellularity and physiological signaling of tendon niche under dynamic culture conditions, along with the recreation of the integrated gradients of its tissue interfaces. In a forward-looking vision of the field, we discuss how the convergence of multiple bioengineering technologies can be leveraged as potential platforms to develop the next generation of relevant in vitro models that can contribute for a deeper fundamental knowledge to develop more effective treatments.
Collapse
|
26
|
Xue Y, Kim HJ, Lee J, Liu Y, Hoffman T, Chen Y, Zhou X, Sun W, Zhang S, Cho HJ, Lee J, Kang H, WonHyoung R, Chang-Moon L, Ahadian S, Dokmeci MR, Lei B, Lee K, Khademhosseini A. Co-Electrospun Silk Fibroin and Gelatin Methacryloyl Sheet Seeded with Mesenchymal Stem Cells for Tendon Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107714. [PMID: 35487761 PMCID: PMC9714686 DOI: 10.1002/smll.202107714] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/07/2022] [Indexed: 05/03/2023]
Abstract
Silk fibroin (SF) is a promising biomaterial for tendon repair, but its relatively rigid mechanical properties and low cell affinity have limited its application in regenerative medicine. Meanwhile, gelatin-based polymers have advantages in cell attachment and tissue remodeling but have insufficient mechanical strength to regenerate tough tissue such as tendons. Taking these aspects into account, in this study, gelatin methacryloyl (GelMA) is combined with SF to create a mechanically strong and bioactive nanofibrous scaffold (SG). The mechanical properties of SG nanofibers can be flexibly modulated by varying the ratio of SF and GelMA. Compared to SF nanofibers, mesenchymal stem cells (MSCs) seeded on SG fibers with optimal composition (SG7) exhibit enhanced growth, proliferation, vascular endothelial growth factor production, and tenogenic gene expression behavior. Conditioned media from MSCs cultured on SG7 scaffolds can greatly promote the migration and proliferation of tenocytes. Histological analysis and tenogenesis-related immunofluorescence staining indicate SG7 scaffolds demonstrate enhanced in vivo tendon tissue regeneration compared to other groups. Therefore, rational combinations of SF and GelMA hybrid nanofibers may help to improve therapeutic outcomes and address the challenges of tissue-engineered scaffolds for tendon regeneration.
Collapse
Affiliation(s)
- Yumeng Xue
- State Key Laboratory of Solidification Processing, Center for Nano Energy Materials, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi’an, 710072, China
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, 710000, China
| | - Han-Jun Kim
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Junmin Lee
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Mechanical Engineering, YONSEI University, Seoul, 03722, South Korea
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Yaowen Liu
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- College of Food Science, Sichuan Agricultural University, Yaan, 625014, China
| | - Tyler Hoffman
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yi Chen
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xingwu Zhou
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wujin Sun
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Shiming Zhang
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Hyun-Jong Cho
- College of Pharmacy, Kangwon National University, Chuncheon, 23431, South Korea
| | - JiYong Lee
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Mechanical Engineering, YONSEI University, Seoul, 03722, South Korea
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, South Korea
| | - Ryu WonHyoung
- Department of Mechanical Engineering, YONSEI University, Seoul, 03722, South Korea
| | - Lee Chang-Moon
- Department of Healthcare and Biomedical Engineering, Chonnam National University, Yeosu, 59626, South Korea
| | - Samad Ahadian
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Mehmet R. Dokmeci
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Bo Lei
- Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, 710000, China
| | - KangJu Lee
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Healthcare and Biomedical Engineering, Chonnam National University, Yeosu, 59626, South Korea
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Ali Khademhosseini
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| |
Collapse
|
27
|
Ramires LC, Jeyaraman M, Muthu S, Shankar A N, Santos GS, da Fonseca LF, Lana JF, Rajendran RL, Gangadaran P, Jogalekar MP, Cardoso AA, Eickhoff A. Application of Orthobiologics in Achilles Tendinopathy: A Review. Life (Basel) 2022; 12:399. [PMID: 35330150 PMCID: PMC8954398 DOI: 10.3390/life12030399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Orthobiologics are biological materials that are intended for the regeneration of bone, cartilage, and soft tissues. In this review, we discuss the application of orthobiologics in Achilles tendinopathy, more specifically. We explain the concepts and definitions of each orthobiologic and the literature regarding its use in tendon disorders. The biological potential of these materials can be harnessed and administered into injured tissues, particularly in areas where standard healing is disrupted, a typical feature of Achilles tendinopathy. These products contain a wide variety of cell populations, cytokines, and growth factors, which have been shown to modulate many other cells at local and distal sites in the body. Collectively, they can shift the state of escalated inflammation and degeneration to reestablish tissue homeostasis. The typical features of Achilles tendinopathy are failed healing responses, persistent inflammation, and predominant catabolic reactions. Therefore, the application of orthobiologic tools represents a viable solution, considering their demonstrated efficacy, safety, and relatively easy manipulation. Perhaps a synergistic approach regarding the combination of these orthobiologics may promote more significant clinical outcomes rather than individual application. Although numerous optimistic results have been registered in the literature, additional studies and clinical trials are still highly desired to further illuminate the clinical utility and efficacy of these therapeutic strategies in the management of tendinopathies.
Collapse
Affiliation(s)
- Luciano C. Ramires
- Department of Orthopaedics and Sports Medicine, Centro Clínico Mãe de Deus, Porto Alegre 90110-270, Brazil;
| | - Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, India;
- Department of Orthopaedics, Apollo Hospitals, Greams Road, Chennai 600006, India;
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, India
| | - Navaladi Shankar A
- Department of Orthopaedics, Apollo Hospitals, Greams Road, Chennai 600006, India;
| | - Gabriel Silva Santos
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, Brazil; (L.F.d.F.); (J.F.L.)
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, Brazil; (L.F.d.F.); (J.F.L.)
- Department of Orthopaedics, The Federal University of São Paulo, São Paulo 04024-002, Brazil
| | - José Fábio Lana
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba 13334-170, Brazil; (L.F.d.F.); (J.F.L.)
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Manasi P. Jogalekar
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA;
| | - Alfredo A. Cardoso
- Department of Oncology-Integrative Medicine-Pain Care, IAC—Instituto Ana Cardoso de Práticas Integrativas e Medicina Regenerative, Gramado 95670-000, Brazil;
| | - Alex Eickhoff
- Department of Orthopaedics, Centro Ortopédico Eickhoff, Três de Maio 98910-000, Brazil;
| |
Collapse
|
28
|
Marqueti RDC, Kjaer M, Moriscot AS. Editorial: Trends in Muscle and Tendon Molecular and Cell Biology. Front Physiol 2022; 12:832613. [PMID: 35185607 PMCID: PMC8851330 DOI: 10.3389/fphys.2021.832613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/30/2021] [Indexed: 11/15/2022] Open
Affiliation(s)
- Rita de Cássia Marqueti
- Graduate Program in Rehabilitation Sciences and Graduate Program of Sciences and Technology of Health, Faculty of Ceilândia, University of Brasília, Brasília, Brazil
- *Correspondence: Rita de Cássia Marqueti ;
| | - Michael Kjaer
- Department of Orthopedic Surgery, Institute of Sports Medicine, Copenhagen University Hospital - Bispebjerg Frederiksberg, and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anselmo Sigari Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
29
|
Tsutsumi H, Kurimoto R, Nakamichi R, Chiba T, Matsushima T, Fujii Y, Sanada R, Kato T, Shishido K, Sakamaki Y, Kimura T, Kishida A, Asahara H. Generation of a tendon-like tissue from human iPS cells. J Tissue Eng 2022; 13:20417314221074018. [PMID: 35083031 PMCID: PMC8785341 DOI: 10.1177/20417314221074018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/01/2022] [Indexed: 12/29/2022] Open
Abstract
Tendons and ligaments are essential connective tissues that connect the muscle and bone. Their recovery from injuries is known to be poor, highlighting the crucial need for an effective therapy. A few reports have described the development of artificial ligaments with sufficient strength from human cells. In this study, we successfully generated a tendon-like tissue (bio-tendon) using human induced pluripotent stem cells (iPSCs). We first differentiated human iPSCs into mesenchymal stem cells (iPSC-MSCs) and transfected them with Mohawk (Mkx) to obtain Mkx-iPSC-MSCs, which were applied to a newly designed chamber with a mechanical stretch incubation system. The embedded Mkx-iPSC-MSCs created bio-tendons and exhibited an aligned extracellular matrix structure. Transplantation of the bio-tendons into a mouse Achilles tendon rupture model showed host-derived cell infiltration with improved histological score and biomechanical properties. Taken together, the bio-tendon generated in this study has potential clinical applications for tendon/ligament-related injuries and diseases.
Collapse
Affiliation(s)
- Hiroki Tsutsumi
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Ryota Kurimoto
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Ryo Nakamichi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Tomoki Chiba
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Takahide Matsushima
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Yuta Fujii
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Risa Sanada
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Tomomi Kato
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Kana Shishido
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Yuriko Sakamaki
- Research Core, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Tsuyoshi Kimura
- Department of Material-Based Medical Engineering, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Akio Kishida
- Department of Material-Based Medical Engineering, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Hiroshi Asahara
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
30
|
Joint-on-chip platforms: entering a new era of in vitro models for arthritis. Nat Rev Rheumatol 2022; 18:217-231. [PMID: 35058618 DOI: 10.1038/s41584-021-00736-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
|
31
|
Ajalik RE, Alenchery RG, Cognetti JS, Zhang VZ, McGrath JL, Miller BL, Awad HA. Human Organ-on-a-Chip Microphysiological Systems to Model Musculoskeletal Pathologies and Accelerate Therapeutic Discovery. Front Bioeng Biotechnol 2022; 10:846230. [PMID: 35360391 PMCID: PMC8964284 DOI: 10.3389/fbioe.2022.846230] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Human Microphysiological Systems (hMPS), otherwise known as organ- and tissue-on-a-chip models, are an emerging technology with the potential to replace in vivo animal studies with in vitro models that emulate human physiology at basic levels. hMPS platforms are designed to overcome limitations of two-dimensional (2D) cell culture systems by mimicking 3D tissue organization and microenvironmental cues that are physiologically and clinically relevant. Unlike animal studies, hMPS models can be configured for high content or high throughput screening in preclinical drug development. Applications in modeling acute and chronic injuries in the musculoskeletal system are slowly developing. However, the complexity and load bearing nature of musculoskeletal tissues and joints present unique challenges related to our limited understanding of disease mechanisms and the lack of consensus biomarkers to guide biological therapy development. With emphasis on examples of modeling musculoskeletal tissues, joints on chips, and organoids, this review highlights current trends of microphysiological systems technology. The review surveys state-of-the-art design and fabrication considerations inspired by lessons from bioreactors and biological variables emphasizing the role of induced pluripotent stem cells and genetic engineering in creating isogenic, patient-specific multicellular hMPS. The major challenges in modeling musculoskeletal tissues using hMPS chips are identified, including incorporating biological barriers, simulating joint compartments and heterogenous tissue interfaces, simulating immune interactions and inflammatory factors, simulating effects of in vivo loading, recording nociceptors responses as surrogates for pain outcomes, modeling the dynamic injury and healing responses by monitoring secreted proteins in real time, and creating arrayed formats for robotic high throughput screens. Overcoming these barriers will revolutionize musculoskeletal research by enabling physiologically relevant, predictive models of human tissues and joint diseases to accelerate and de-risk therapeutic discovery and translation to the clinic.
Collapse
Affiliation(s)
- Raquel E. Ajalik
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Rahul G. Alenchery
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - John S. Cognetti
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Benjamin L. Miller
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Department of Dermatology, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- *Correspondence: Hani A. Awad,
| |
Collapse
|