1
|
Hawthorne BC, Engel S, McCarthy MBR, Cote MC, Mazzocca AD, Coyner KJ. Biologic Adjuvants to Rotator Cuff Repairs Induce Anti-inflammatory Macrophage 2 Polarization and Reduce Inflammatory Macrophage 1 Polarization In Vitro. Arthroscopy 2025; 41:32-41. [PMID: 38735413 DOI: 10.1016/j.arthro.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE To examine the effect of various biologic adjuvants on the polarization of macrophages in an in vitro model for rotator cuff tears. METHODS Tissue was harvested from 6 patients undergoing arthroscopic rotator cuff repair. An in vitro model of the supraspinatus and subacromial bursa was created and treated with control, platelet-rich plasma (PRP), autologous activated serum (AAS), or a combination of PRP+AAS. The effect of treatment on macrophage polarization between M1 proinflammatory macrophages or M2 anti-inflammatory macrophages was measured using gene expression, protein expression, flow cytometry, and nitric oxide production. RESULTS Tendon and bursa treated with PRP, AAS, and PRP+AAS significantly decreased the gene expression of M1 markers interleukin (IL)-12 and tumor necrosis factor-alpha while significantly increasing the expression of M2 markers arginase, IL-10, and transforming growth factor-β (P < .05) compared with treatment with control. Enzyme-linked immunosorbent assay analysis of protein production demonstrated that, compared with control, coculture treated with PRP, AAS, and PRP+AAS significantly decreased markers of M1-macrophages (IL-6, IL-12, and tumor necrosis factor-alpha) while significantly increasing the expression of markers of M2-macrophages (arginase, IL-10, and transforming growth factor-beta) (P < .05). Flow cytometry analysis of surface markers demonstrated that compared with control, tendon and bursa treated with PRP, AAS, and PRP+AAS significantly decreased markers of M1-macrophages (CD80, CD86, CD64, CD16) while significantly increasing the expression of markers of M2-macrophages (CD163 and CD206) (P < .05). Treatment of the coculture with PRP, AAS, and PRP+AAS consistently demonstrated a decrease in nitric oxide production (P < .05) compared with control. AAS and PRP+AAS demonstrated an increased macrophage shift to M2 compared with PRP alone, whereas there was not as uniform of a shift when comparing PRP+AAS with AAS alone. CONCLUSIONS In an in vitro model of rotator cuff tears, the treatment of supraspinatus tendon and subacromial bursa with PRP, AAS, and PRP+AAS demonstrated an increase in markers of anti-inflammatory M2-macrophages and a concomitant decrease in markers of proinflammatory M1-macrophages. AAS and PRP+AAS contributed to a large shift to macrophage polarization to the anti-inflammatory M2 compared with PRP. CLINICAL RELEVANCE The mechanism of biologic adjuvant effects on the rotator cuff remains poorly understood. This study suggests that they may contribute to polarization of macrophages for their proinflammatory (M1) state to the anti-inflammatory (M2) state.
Collapse
Affiliation(s)
| | - Sam Engel
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, U.S.A
| | - Mary Beth R McCarthy
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, U.S.A
| | - Mark C Cote
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, U.S.A
| | - Augustus D Mazzocca
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, U.S.A
| | - Katherine J Coyner
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, U.S.A..
| |
Collapse
|
2
|
Dyment NA, Kamalitdinov TB, Kuntz AF. The 2024 Kappa Delta Young Investigator Award: Leveraging Insights From Development to Improve Adult Repair: Hedgehog Signaling as a Master Regulator of Enthesis Fibrocartilage Formation. J Am Acad Orthop Surg 2024; 32:1074-1086. [PMID: 39589737 DOI: 10.5435/jaaos-d-24-00996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 11/27/2024] Open
Abstract
The work in this article summarizes findings from our group on key biochemical cues that govern the formation and repair of tendons and ligaments. Specifically, we summarize the journey that started with a serendipitous discovery that is now being translated into novel therapies to improve tendon-to-bone repair outcomes. This journey began with the discovery that the Hedgehog (Hh) signaling pathway was expressed within the enthesis during development and that its primary role was to promote fibrocartilage production and maturation. Next, we developed an anterior cruciate ligament reconstruction model in novel transgenic mice that allowed us to discover that the Hh pathway promotes fibrocartilaginous tendon-to-bone attachments during the integration process. In addition, we established that the coordinated stages of zonal tendon-to-bone integration after anterior cruciate ligament reconstruction were comparable with the stages required for enthesis formation during development. Now that we have demonstrated that the Hh pathway is a potent therapeutic target, we are currently advancing these findings to develop drug delivery systems to improve tendon-to-bone repair. Ultimately, our group aims to establish key mechanisms that govern tendon and ligament formation that can be leveraged for novel regenerative therapies to improve clinical care.
Collapse
Affiliation(s)
- Nathaniel A Dyment
- From the Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA (Dr. Dyment, Dr. Kamalitdinov, and Dr. Kuntz), and the Department of Bioengineering, University of Pennsylvania, Philadelphia, PA (Dr. Dyment and Dr. Kamalitdinov)
| | | | | |
Collapse
|
3
|
Kent RN, Huang AH, Baker BM. Augmentation of Tendon and Ligament Repair with Fiber-Reinforced Hydrogel Composites. Adv Healthc Mater 2024; 13:e2400668. [PMID: 39135411 PMCID: PMC11582515 DOI: 10.1002/adhm.202400668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/15/2024] [Indexed: 11/24/2024]
Abstract
This review highlights the promise of fiber-reinforced hydrogel composites (FRHCs) for augmenting tendon and ligament repair and regeneration. Composed of reinforcing fibers embedded in a hydrogel, these scaffolds provide both mechanical strength and a conducive microenvironment for biological processes required for connective tissue regeneration. Typical properties of FRHCs are discussed, highlighting their ability to simultaneously fulfill essential mechanical and biological design criteria for a regenerative scaffold. Furthermore, features of FRHCs are described that improve specific biological aspects of tendon healing including mesenchymal progenitor cell recruitment, early polarization to a pro-regenerative immune response, tenogenic differentiation of recruited progenitor cells, and subsequent production of a mature, aligned collagenous matrix. Finally, the review offers a perspective on clinical translation of tendon FRHCs and outlines key directions for future work.
Collapse
Affiliation(s)
- Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alice H Huang
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
4
|
Matsushima T, Hiroshi A. Molecular mechanisms of mechanosensing and plasticity of tendons and ligaments. J Biochem 2024; 176:263-269. [PMID: 38729213 PMCID: PMC11444931 DOI: 10.1093/jb/mvae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Tendons and ligaments, crucial components of the musculoskeletal system, connect muscles to bones. In the realm of sports, tendons and ligaments are vulnerable tissues, with injuries such as Achilles tendon rupture and anterior cruciate ligament tears directly impacting an athlete's career. Furthermore, repetitive trauma and tissue degeneration can lead to conditions like secondary osteoarthritis, ultimately affecting the overall quality of life. Recent research highlights the pivotal role of mechanical stress in maintaining homeostasis within tendons and ligaments. This review delves into the latest insights on the structure of tendons and ligaments and the plasticity of tendon tissue in response to mechanical loads.
Collapse
Affiliation(s)
- Takahide Matsushima
- Department of Systems BioMedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Asahara Hiroshi
- Department of Systems BioMedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
- Department of Molecular Medicine, Scripps Research, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, USA
| |
Collapse
|
5
|
Hirsinger E, Blavet C, Bonnin MA, Bellenger L, Gharsalli T, Duprez D. Limb connective tissue is organized in a continuum of promiscuous fibroblast identities during development. iScience 2024; 27:110305. [PMID: 39050702 PMCID: PMC11267076 DOI: 10.1016/j.isci.2024.110305] [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: 04/19/2024] [Revised: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Connective tissue (CT), which includes tendon and muscle CT, plays critical roles in development, in particular as positional cue provider. Nonetheless, our understanding of fibroblast developmental programs is hampered because fibroblasts are highly heterogeneous and poorly characterized. Combining single-cell RNA-sequencing-based strategies including trajectory inference and in situ hybridization analyses, we address the diversity of fibroblasts and their developmental trajectories during chicken limb fetal development. We show that fibroblasts switch from a positional information to a lineage diversification program at the fetal period onset. Muscle CT and tendon are composed of several fibroblast populations that emerge asynchronously. Once the final muscle pattern is set, transcriptionally close populations are found in neighboring locations in limbs, prefiguring the adult fibroblast layers. We propose that the limb CT is organized in a continuum of promiscuous fibroblast identities, allowing for the robust and efficient connection of muscle to bone and skin.
Collapse
Affiliation(s)
- Estelle Hirsinger
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Cédrine Blavet
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Marie-Ange Bonnin
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| | - Léa Bellenger
- Sorbonne Université, CNRS FR3631, Inserm U1156, Institut de Biologie Paris Seine (IBPS), ARTbio Bioinformatics Analysis Facility, Paris, Institut Français de Bioinformatique (IFB), 75005 Paris, France
| | - Tarek Gharsalli
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
- Inovarion, 75005 Paris, France
| | - Delphine Duprez
- Sorbonne Université, Institut Biologie Paris Seine, CNRS UMR7622, Developmental Biology Laboratory, Inserm U1156, 75005 Paris, France
| |
Collapse
|
6
|
Karlidag T, Bingol O, Kulakoglu B, Keskin OH, Durgal A, Ozdemir G. Effects of tendon elongation on plantar pressure and clinical outcomes: A comparative analysis between open repair and minimally invasive surgery. Knee Surg Sports Traumatol Arthrosc 2024; 32:1880-1890. [PMID: 38630070 DOI: 10.1002/ksa.12163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/10/2024] [Accepted: 03/12/2024] [Indexed: 07/21/2024]
Abstract
PURPOSE The aim of this study was to assess whether variances in Achilles tendon elongation are linked to dissimilarities in the plantar pressure distribution following two different surgical approaches for an Achilles tendon rupture (ATR). METHODS All patients who were treated with open or minimally invasive surgical repair (MIS) and were over 2 years post their ATR were eligible for inclusion. A total of 65 patients with an average age of 43 ± 11 years were included in the study. Thirty-five patients were treated with open repair, and 30 patients were treated with MIS. Clinical outcomes were evaluated using the American Orthopedic Foot and Ankle Society (AOFAS) and ATR Score (ATRS). Achilles tendon elongation was measured using axial and sagittal magnetic resonance imaging scans. Plantar pressure measurements for the forefoot, midfoot and hindfoot during gait were divided into percentages based on total pressure, measured in g/cm2 for each area. RESULTS The average AOFAS score was found 'excellent' (93 ± 2.8) in the MIS group, while it was found 'good' (87.4 ± 5.6) in the open repair group. In addition, the MIS group showed significantly superior ATRS scores (78.8 ± 7.4) compared to the open repair group (56.4 ± 15.4) (p < 0.001). The average tendon elongation in the MIS group was 11.3 ± 2 mm, while it was 17.3 ± 4.3 mm (p < 0.001) in the open repair group. While the open repair group showed significantly higher plantar pressure distribution in the initial contact and preswing phases compared to uninjured extremities, there was no significant difference between the uninjured extremities and the MIS group. CONCLUSION In conclusion, the findings of this study demonstrated that minimally invasive surgery was associated with less tendon elongation, more proximity to the plantar pressure distributions of the uninjured extremity and superior clinical outcomes compared to open surgical repair. Therefore, minimally invasive surgery may be considered a more suitable option for acute Achilles tendon repair to achieve overall better outcomes. LEVEL OF EVIDENCE Level III.
Collapse
Affiliation(s)
- Taner Karlidag
- Department of Orthopedics and Traumatology Surgery, Helios ENDO-Klinik, Hamburg, Germany
- Department of Orthopedics and Traumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Olgun Bingol
- Department of Orthopedics and Traumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Burak Kulakoglu
- Department of Orthopedics and Traumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Omer Halit Keskin
- Department of Orthopedics and Traumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Atahan Durgal
- Department of Orthopedics and Traumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Guzelali Ozdemir
- Department of Orthopedics and Traumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
7
|
Rajalekshmi R, Agrawal DK. Understanding Fibrous Tissue in the Effective Healing of Rotator Cuff Injury. JOURNAL OF SURGERY AND RESEARCH 2024; 7:215-228. [PMID: 38872898 PMCID: PMC11174978 DOI: 10.26502/jsr.10020363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The rotator cuff is a crucial group of muscles and tendons in the shoulder complex that plays a significant role in the stabilization of the glenohumeral joint and enabling a wide range of motion. Rotator cuff tendon tears can occur due to sudden injuries or degenerative processes that develop gradually over time, whether they are partial or full thickness. These injuries are common causes of shoulder pain and functional impairment, and their complex nature highlights the essential role of the rotator cuff in shoulder function. Scar formation is a crucial aspect of the healing process initiated following a rotator cuff tendon tear, but excessive fibrous tissue development can potentially lead to stiffness, discomfort, and movement limitations. Age is a critical risk factor, with the prevalence of these tears increasing among older individuals. This comprehensive review aims to delve deeper into the anatomy and injury mechanisms of the rotator cuff. Furthermore, it will inspect the signaling pathways involved in fibrous tissue development, evaluate the various factors affecting the healing environment, and discuss proactive measures aimed at reducing excessive fibrous tissue formation. Lastly, this review identifed gaps within existing knowledge to advance methods for better management of rotator cuff tendon injuries.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| | - Devendra K Agrawal
- Department of Translational Research, College of the Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| |
Collapse
|
8
|
Sato F, Masuda Y, Suzuki D, Hayashi T, Iwasaki T, Kim J, Matsumoto T, Maeda E. Biomechanical analysis of tendon regeneration capacity of Iberian ribbed newts following transection injury: Comparison to a mouse model. J Orthop Res 2024; 42:607-617. [PMID: 37819002 DOI: 10.1002/jor.25705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
Adult mammals are known for their poor ability to regenerate tissues, including tendons. On the other hand, urodeles have become an important model in regenerative studies for their remarkable ability to regenerate various body parts and organs throughout life, such as limbs, retinas, or even the brain. However, little is known about their capacity to regenerate injured tendons. If newts can also repair tendons without scar formation, they may be a suitable animal model for tendon regeneration studies in other adult vertebrates. Therefore, the present study used Iberian ribbed newts to characterize mechanical and structural regeneration of tendons following transection, using tensile tests and multiphoton microscopy. A digital flexor tendon in a hindlimb was transected either partially or completely, and regenerated tendon was examined 6 and 12 weeks after the operation. Tensile strength of regenerated tendons was significantly less than normal at 6 weeks, but was remarkably recovered at 12 weeks, reaching levels comparable to those of uninjured tendons. On the other hand, mouse tendons demonstrated poor recovery of strength even after 12 weeks. Multiphoton microscopy revealed that tendon-like collagenous tissue bridges residual tendon stubs in newts, but disorganized scar-like tissue filled the injured location in mice. These findings highlight the remarkable capacity of newts to recover from tendon injury and confirm the utility of newts as a model to study tendon regeneration.
Collapse
Affiliation(s)
- Fumiya Sato
- Biomechanics Laboratory, Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
| | - Yu Masuda
- Biomechanics Laboratory, Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
| | - Daisuke Suzuki
- Department of Health Science, Hokkaido Chitose College of Rehabilitation, Chitose, Hokkaido, Japan
| | - Toshinori Hayashi
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Tomohito Iwasaki
- Department of Food Science and Human Wellness, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Jeonghyun Kim
- Biomechanics Laboratory, Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
| | - Takeo Matsumoto
- Biomechanics Laboratory, Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
| | - Eijiro Maeda
- Biomechanics Laboratory, Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
9
|
Taguchi T, Lopez M, Takawira C. Viable tendon neotissue from adult adipose-derived multipotent stromal cells. Front Bioeng Biotechnol 2024; 11:1290693. [PMID: 38260742 PMCID: PMC10800559 DOI: 10.3389/fbioe.2023.1290693] [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: 09/07/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Background: Tendon healing is frequently prolonged, unpredictable, and results in poor tissue quality. Neotissue formed by adult multipotent stromal cells has the potential to guide healthy tendon tissue formation. Objectives: The objective of this study was to characterize tendon neotissue generated by equine adult adipose-derived multipotent stromal cells (ASCs) on collagen type I (COLI) templates under 10% strain in a novel bioreactor. The tested hypothesis was that ASCs assume a tendon progenitor cell-like morphology, express tendon-related genes, and produce more organized extracellular matrix (ECM) in tenogenic versus stromal medium with perfusion and centrifugal fluid motion. Methods: Equine ASCs on COLI sponge cylinders were cultured in stromal or tenogenic medium within bioreactors during combined perfusion and centrifugal fluid motion for 7, 14, or 21 days under 10% strain. Viable cell distribution and number, tendon-related gene expression, and micro- and ultra-structure were evaluated with calcein-AM/EthD-1 staining, resazurin reduction, RT-PCR, and light, transmission, and scanning electron microscopy. Fibromodulin was localized with immunohistochemistry. Cell number and gene expression were compared between culture media and among culture periods (p < 0.05). Results: Viable cells were distributed throughout constructs for up to 21 days of culture, and cell numbers were higher in tenogenic medium. Individual cells had a round or rhomboid shape with scant ECM in stromal medium in contrast to clusters of parallel, elongated cells surrounded by highly organized ECM in tenogenic medium after 21 days of culture. Transcription factor, extracellular matrix, and mature tendon gene expression profiles confirmed ASC differentiation to a tendon progenitor-like cell in tenogenic medium. Construct micro- and ultra-structure were consistent with tendon neotissue and fibromodulin was present in the ECM after culture in tenogenic medium. Conclusion: Long-term culture in custom bioreactors with combined perfusion and centrifugal tenogenic medium circulation supports differentiation of equine adult ASCs into tendon progenitor-like cells capable of neotissue formation.
Collapse
|
10
|
Mienaltowski MJ, Callahan M, Gonzales NL, Wong A. Examining the Potential of Vitamin C Supplementation in Tissue-Engineered Equine Superficial Digital Flexor Tendon Constructs. Int J Mol Sci 2023; 24:17098. [PMID: 38069418 PMCID: PMC10707379 DOI: 10.3390/ijms242317098] [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: 09/15/2023] [Revised: 11/26/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Because equine tendinopathies are slow to heal and often recur, therapeutic strategies are being considered that aid tendon repair. Given the success of utilizing vitamin C to promote tenogenesis in other species, we hypothesized that vitamin C supplementation would produce dose-dependent improvements in the tenogenic properties of tendon proper (TP) and peritenon (PERI) cells of the equine superficial digital flexor tendon (SDFT). Equine TP- and PERI-progenitor-cell-seeded fibrin three-dimensional constructs were supplemented with four concentrations of vitamin C. The gene expression profiles of the constructs were assessed with 3'-Tag-Seq and real-time quantitative polymerase chain reaction (RT-qPCR); collagen content and fibril ultrastructure were also analyzed. Moreover, cells were challenged with dexamethasone to determine the levels of cytoprotection afforded by vitamin C. Expression profiling demonstrated that vitamin C had an anti-inflammatory effect on TP and PERI cell constructs. Moreover, vitamin C supplementation mitigated the degenerative pathways seen in tendinopathy and increased collagen content in tendon constructs. When challenged with dexamethasone in two-dimensional culture, vitamin C had a cytoprotective effect for TP cells but not necessarily for PERI cells. Future studies will explore the effects of vitamin C on these cells during inflammation and within the tendon niche in vivo.
Collapse
Affiliation(s)
- Michael J. Mienaltowski
- Department of Animal Science, College of Agricultural & Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| | - Mitchell Callahan
- Department of Animal Science, College of Agricultural & Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| | - Nicole L. Gonzales
- School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Angelique Wong
- Department of Animal Science, College of Agricultural & Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
11
|
Steffen D, Mienaltowski M, Baar K. Spatial gene expression in the adult rat patellar tendon. Matrix Biol Plus 2023; 19-20:100138. [PMID: 38124714 PMCID: PMC10731370 DOI: 10.1016/j.mbplus.2023.100138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Tendons are dense connective tissues with relatively few cells which makes studying the molecular profile of the tissue challenging. There is not a consensus on the spatial location of various cell types within a tendon, nor the accompanying transcriptional profile. In the present study, we used two male rat patellar tendon samples for sequencing-based spatial transcriptomics to determine the gene expression profile. We integrated our data with a mouse Achilles single cell dataset to predict the cell type composition of the patellar tendon as a function of location within the tissue. The spatial location of the predicated cell types suggested that there were two populations of tendon fibroblasts, one located in the tendon midsubstance, while the other localized with red blood cells, pericytes, and immune cells to the tendon peripheral connective tissue. Of the highest expressed spatially variable genes, there were multiple genes with known function in tendon: Col1a1, Col1a2, Dcn, Fmod, Sparc, and Comp. Further, a novel spatially regulated gene (AABR07000398.1) with no known function was identified. The spatial gene expression of tendon associated genes (Scx, Thbs4, Tnmd, Can, Bgn, Lum, Adamts2, Lox, Ppib, Col2a1, Col3a1, Col6a2) was also visualized. Both patellar tendon samples had similar expression patterns for all these genes. This dataset provides new spatial insights into gene expression in a healthy tendon.
Collapse
Affiliation(s)
- Danielle Steffen
- Department of Neurobiology, Physiology & Behavior, University of California Davis, Davis, CA, United States
| | - Michael Mienaltowski
- Department of Animal Science, University of California Davis, Davis, CA, United States
| | - Keith Baar
- Department of Neurobiology, Physiology & Behavior, University of California Davis, Davis, CA, United States
- Physiology and Membrane Biology, University of California Davis, Davis, CA, United States
- VA Northern California Health Care System, Mather, CA 95655, United States
| |
Collapse
|
12
|
Li H, Luo S, Wang H, Chen Y, Ding M, Lu J, Jiang L, Lyu K, Huang S, Shi H, Chen H, Li S. The mechanisms and functions of TGF-β1 in tendon healing. Injury 2023; 54:111052. [PMID: 37738787 DOI: 10.1016/j.injury.2023.111052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/11/2023] [Indexed: 09/24/2023]
Abstract
Tendon injury accounts for 30% of musculoskeletal diseases and often leads to disability, pain, healthcare cost, and lost productivity. Following injury to tendon, tendon healing proceeds via three overlapping healing processes. However, due to the structural defects of the tendon itself, the tendon healing process is characterized by the formation of excessive fibrotic scar tissue, and injured tendons rarely return to native tendons, which can easily contribute to tendon reinjury. Moreover, the resulting fibrous scar is considered to be a precipitating factor for subsequent degenerative tendinopathy. Despite this, therapies are almost limited because underlying molecular mechanisms during tendon healing are still unknown. Transforming Growth Factor-β1 (TGF-β1) is known as one of most potent profibrogenic factors during tendon healing process. However, blockage TGF-β1 fails to effectively enhance tendon healing. A detailed understanding of real abilities of TGF-β1 involved in tendon healing can bring promising perspectives for therapeutic value that improve the tendon healing process. Thus, in this review, we describe recent efforts to identify and characterize the roles and mechanisms of TGF-β1 involved at each stage of the tendon healing and highlight potential roles of TGF-β1 leading to the fibrotic response to tendon injury.
Collapse
Affiliation(s)
- Hanyue Li
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Shengyu Luo
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Hao Wang
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yixuan Chen
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - MingZhe Ding
- School of Public Health, Southwest Medical University, Luzhou, China
| | - Jingwei Lu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Li Jiang
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Kexin Lyu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Shilin Huang
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Houyin Shi
- School of Public Health, Southwest Medical University, Luzhou, China
| | - Hui Chen
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Sen Li
- School of Physical Education, Southwest Medical University, Luzhou, China.
| |
Collapse
|
13
|
Kusaba Y, Kumagai K, Ishikawa K, Choe H, Ike H, Kobayashi N, Inaba Y. Bevacizumab promotes tenogenic differentiation and maturation of rat tendon-derived cells in vitro. PLoS One 2023; 18:e0293463. [PMID: 37906574 PMCID: PMC10617717 DOI: 10.1371/journal.pone.0293463] [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: 04/30/2023] [Accepted: 10/12/2023] [Indexed: 11/02/2023] Open
Abstract
Previous work suggested that tenogenic differentiation of tendon stem/progenitor cells (TSPCs) was suppressed by upregulated expression of the angiogenic marker vascular endothelial growth factor (VEGF). The purpose of this study was to test the hypothesis that anti-VEGF antibody, bevacizumab, promotes in vitro tenogenic differentiation and maturation of two distinct types of TSPCs, tendon proper-derived cells (TDCs), and paratenon-derived cells (PDCs) originating from rat Achilles tendon. TDCs and PDCs were isolated from the tendon proper and the paratenon of rat Achilles tendons. TDCs and PDCs were cultured for 3 days on plates with or without VEGF. TDCs and PDCs were also cultured in collagen gel matrix, and the blocking effect of VEGF was examined by the addition of 100 ng/mL of bevacizumab. Effects of bevacizumab on tenogenic differentiation were assessed using real-time PCR, immunofluorescent staining, and western blotting. VEGF significantly attenuated expression of the Tnmd gene in both PDCs and TDCs (P<0.05). Expressions of the Scx, Tnmd, and Col1a1 genes were significantly upregulated by the addition of bevacizumab (P<0.05). Immunofluorescent staining showed that the percentage of tenomodulin-positive PDCs and TDCs was significantly higher with bevacizumab treatment than in control cultures (P<0.05). Western blotting showed that bevacizumab suppressed pVEGFR-2 protein expression in both PDCs and TDCs. Bevacizumab promoted the in vitro tenogenic differentiation and maturation of two distinct TSPCs derived from rat Achilles tendon. Since the previous studies demonstrated that TSPCs have a potential to contribute to tendon repair, attenuating VEGF levels in TSPCs by administration of bevacizumab is a novel candidate therapeutic option for promoting tendon repair.
Collapse
Affiliation(s)
- Yohei Kusaba
- Department of Orthopaedic Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Ken Kumagai
- Department of Orthopaedic Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Kimi Ishikawa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Hyonmin Choe
- Department of Orthopaedic Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Hiroyuki Ike
- Department of Orthopaedic Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Naomi Kobayashi
- Department of Orthopaedic Surgery, Yokohama City University Medical Center, Yokohama, Japan
| | - Yutaka Inaba
- Department of Orthopaedic Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
14
|
Pechanec MY, Mienaltowski MJ. Decoding the transcriptomic expression and genomic methylation patterns in the tendon proper and its peritenon region in the aging horse. BMC Res Notes 2023; 16:267. [PMID: 37821884 PMCID: PMC10566085 DOI: 10.1186/s13104-023-06562-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/10/2023] [Indexed: 10/13/2023] Open
Abstract
OBJECTIVES Equine tendinopathies are challenging because of the poor healing capacity of tendons commonly resulting in high re-injury rates. Within the tendon, different regions - tendon proper (TP) and peritenon (PERI) - contribute to the tendon matrix in differing capacities during injury and aging. Aged tendons have decreased repair potential; the underlying transcriptional and epigenetic changes that occur in the TP and PERI regions are not well understood. The objective of this study was to assess TP and PERI regional differences in adolescent, midlife, and geriatric horses using RNA sequencing and DNA methylation techniques. RESULTS Differences existed between TP and PERI regions of equine superficial digital flexor tendons by age as evidenced by RNASeq and DNA methylation. Cluster analysis indicated that regional distinctions existed regardless of age. Genes such as DCN, COMP, FN1, and LOX maintained elevated TP expression while genes such as GSN and AHNAK were abundant in PERI. Increased gene activity was present in adolescent and geriatric populations but decreased during midlife. Regional differences in DNA methylation were also noted. Notably, when evaluating all ages of TP against PERI, five genes (HAND2, CHD9, RASL11B, ADGRD1, and COL14A1) had regions of differential methylation as well as differential gene expression.
Collapse
Affiliation(s)
- Monica Y Pechanec
- Department of Animal Science, University of California Davis, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Michael J Mienaltowski
- Department of Animal Science, University of California Davis, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA.
| |
Collapse
|
15
|
Frankewycz B, Bell R, Chatterjee M, Andarawis-Puri N. The superior healing capacity of MRL tendons is minimally influenced by the systemic environment of the MRL mouse. Sci Rep 2023; 13:17242. [PMID: 37821476 PMCID: PMC10567747 DOI: 10.1038/s41598-023-42449-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/10/2023] [Indexed: 10/13/2023] Open
Abstract
Murphy Roths Large mice (MRL) exhibit improved tendon healing and are often described as a "super-healer" strain. The underlying mechanisms that drive the superior healing response of MRL remain a controversial subject. We utilized a tendon transplantation model between MRL and "normal-healer" B6-mice to differentiate between the contribution of MRL's innate tendon and systemic environment to its improved healing capacity. Patellar tendons with a midsubstance punch injury were transplanted back into the same animal (autograft) or into an animal of the other strain (allograft). Findings at 4 weeks showed that the innate MRL tendon environment drives its improved healing capacity as demonstrated by improved stiffness and maximum load in MRL-grafts-in-B6-host-allografts compared to B6-autografts, and higher modulus in MRL-autografts compared to B6-graft-in-MRL-host-allografts. Groups with an MRL component showed an increase in pro-inflammatory cytokines in the 3 days after injury, suggesting an early enhanced inflammatory profile in MRL that ultimately resolves. A preserved range of motion of the knee joint in all MRL animals suggests a systemic "shielding effect" of MRL in regard to joint adhesiveness. Our findings 4-weeks post injury are consistent with previous studies showing tissue-driven improved healing and suggest that the systemic environment contributes to the overall healing process.
Collapse
Affiliation(s)
- Borys Frankewycz
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- University Hospital Regensburg, Regensburg, Germany
| | - Rebecca Bell
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | | - Nelly Andarawis-Puri
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.
- Hospital for Special Surgery, New York, NY, USA.
| |
Collapse
|
16
|
Chatterjee M, Evans MK, Bell R, Nguyen PK, Kamalitdinov TB, Korntner S, Kuo CK, Dyment NA, Andarawis-Puri N. Histological and immunohistochemical guide to tendon tissue. J Orthop Res 2023; 41:2114-2132. [PMID: 37321983 DOI: 10.1002/jor.25645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 06/02/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
Tendons are unique dense connective tissues with discrete zones having specific structure and function. They are juxtaposed with other tissues (e.g., bone, muscle, and fat) with different compositional, structural, and mechanical properties. Additionally, tendon properties change drastically with growth and development, disease, aging, and injury. Consequently, there are unique challenges to performing high quality histological assessment of this tissue. To address this need, histological assessment was one of the breakout session topics at the 2022 Orthopaedic Research Society (ORS) Tendon Conference hosted at the University of Pennsylvania. The purpose of the breakout session was to discuss needs from members of the ORS Tendon Section related to histological procedures, data presentation, knowledge dissemination, and guidelines for future work. Therefore, this review provides a brief overview of the outcomes of this discussion and provides a set of guidelines, based on the perspectives from our laboratories, for histological assessment to assist researchers in their quest to utilize these techniques to enhance the outcomes and interpretations of their studies.
Collapse
Affiliation(s)
- Monideepa Chatterjee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Mary K Evans
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rebecca Bell
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| | - Phong K Nguyen
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| | - Timur B Kamalitdinov
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stefanie Korntner
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Catherine K Kuo
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Orthopaedics, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Nathaniel A Dyment
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nelly Andarawis-Puri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
- Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
17
|
Leahy TP, Fung AK, Weiss SN, Dyment NA, Soslowsky LJ. Investigating the temporal roles of decorin and biglycan in tendon healing. J Orthop Res 2023; 41:2238-2249. [PMID: 37132501 PMCID: PMC10525000 DOI: 10.1002/jor.25590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/14/2023] [Accepted: 05/01/2023] [Indexed: 05/04/2023]
Abstract
The small leucine-rich proteoglycans, decorin and biglycan, are minor components of the tendon extracellular matrix that regulate fibrillogenesis and matrix assembly. Our study objective was to define the temporal roles of decorin and biglycan during tendon healing using inducible knockout mice to include genetic knockdown at specific phases of healing: time of injury, the proliferative phase, and the remodeling phase. We hypothesized that knockdown of decorin or biglycan would adversely affect tendon healing, and that by prescribing the timing of knockdown, we could elucidate the temporal roles of these proteins during healing. Contrary to our hypothesis, decorin knockdown did not affect tendon healing. However, when biglycan was knocked down, either alone or coupled with decorin, tendon modulus was increased relative to wild-type mice, and this finding was consistent among all induction timepoints. At 6 weeks postinjury, we observed increased expression of genes associated with the extracellular matrix and growth factor signaling in the biglycan knockdown and compound decorin-biglycan knockdown tendons. Interestingly, these groups demonstrated opposing trends in gene expression as a function of knockdown-induction timepoint, highlighting distinct temporal roles for decorin and biglycan. In summary, this study finds that biglycan plays multiple functions throughout tendon healing, with the most impactful, detrimental role likely occurring during late-stage healing. Statement of clinical importance: This study helps to define the molecular factors that regulate tendon healing, which may aid in the development of new clinical therapies.
Collapse
Affiliation(s)
- Thomas P. Leahy
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley K. Fung
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephanie N. Weiss
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel A. Dyment
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Louis J. Soslowsky
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
18
|
Leong NL, Wu J, Greskovich KE, Li Y, Jiang J. Pdgfrβ + lineage cells transiently increase at the site of Achilles tendon healing. J Orthop Res 2023; 41:1882-1889. [PMID: 36922361 DOI: 10.1002/jor.25552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/01/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
The purpose of this study was to track platelet-derived growth factor receptor-β (Pdgfr-β) lineage cells at the site of Achilles tendon injury over time. Pdgfr-β-CreERT2 :Ai9 mice were generated to track Pdgfr-β lineage cells in adult mice. A surgical Achilles transection injury model was employed to examine the presence of Pdgfr-β lineage cells in the healing tendon over time, with five mice per time point at 3, 7, 14, 28, and 56 days postoperatively. Histology and immunohistochemistry for tdTomato (Pdgfr-β lineage cells), PCNA (proliferating cell nuclear antigen, cell proliferation), and α-SMA (α-smooth muscle actin, myofibroblasts) were performed. The percentage of cells at the healing tendon site staining positive for tdTomato and PCNA were quantified. Over 75% of cells at the injury site were Pdgfr-β lineage cells at Days 3, 7, and 14, and this percentage decreased significantly by Days 28 and 56 postinjury. Cell proliferation at the injury site peaked on Day 7 and decreased thereafter. Immunohistochemistry for α-SMA demonstrated minimal colocalization of myofibroblasts with Pdgfr-β lineage cells. This study demonstrates that in a mouse model of Achilles tendon injury, Pdgfr-β lineage cells' presence at the injury site is transient. Thus, we conclude that they are unlikely to be the cells that differentiate into myofibroblasts and directly contribute to tendon fibrous scar formation. Clinical Significance: This study provides some insight into the presence of Pdgfr-β lineage cells (including pericytes) following Achilles injury, furthering our understanding of tendon healing.
Collapse
Affiliation(s)
- Natalie L Leong
- Baltimore VA Medical Center, VA Maryland Healthcare System, Baltimore, Maryland, USA
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jocelyn Wu
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kathryn E Greskovich
- Baltimore VA Medical Center, VA Maryland Healthcare System, Baltimore, Maryland, USA
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yang Li
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jie Jiang
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Koch DW, Schnabel LV. Mesenchymal stem cell licensing: enhancing MSC function as a translational approach for the treatment of tendon injury. Am J Vet Res 2023; 84:1-8. [PMID: 37669745 PMCID: PMC11027115 DOI: 10.2460/ajvr.23.07.0154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/14/2023] [Indexed: 09/07/2023]
Abstract
Tendon injuries are common in both veterinary and human clinical patients and result in morbidity, pain, and lost athletic performance. Consequently, utilizing naturally occurring injuries in veterinary patients as a comparative model could inform the development of novel therapies and increase translation for the treatment of human tendon injuries. Mesenchymal stem cells (MSCs) have shown considerable efficacy for the treatment of experimental and clinical superficial digital flexor tendon injury in the horse; however, the reinjury rate following treatment can remain high and MSC efficacy in treating other tendons is less well known. Additionally, the translation of MSC therapy to human tendon injury has remained poor. Recent evidence indicates that naïve MSC function can be enhanced through exogenous stimulation or manipulation of their environment. This stimulation or activation, herein termed MSC licensing, markedly alters MSC functions associated with immunomodulation, extracellular matrix remodeling, vascular development, bioactive factor production, and endogenous stromal/progenitor cell support. Additionally, a variety of licensing strategies has proven to influence MSC-secreted factors that have positively influenced outcome parameters in both in vitro and in vivo disease models separate from musculoskeletal tissues. Therefore, identifying the optimal licensing strategy for MSCs could ultimately provide an avenue for reliable and repeatable treatment of a broad range of tendon injuries of both veterinary and human clinical patients. This article details current evidence on the effects of licensed MSCs in both in vitro and in vivo disease models of different species and provides commentary on how those effector functions identified may be translated to the treatment of tendon injuries.
Collapse
Affiliation(s)
- Drew W. Koch
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Lauren V. Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| |
Collapse
|
20
|
Watanabe G, Yamamoto M, Taniguchi S, Sugiyama Y, Hirouchi H, Ishizuka S, Kitamura K, Mizoguchi T, Takayama T, Hayashi K, Abe S. Chronological Changes in the Expression and Localization of Sox9 between Achilles Tendon Injury and Functional Recovery in Mice. Int J Mol Sci 2023; 24:11305. [PMID: 37511063 PMCID: PMC10379325 DOI: 10.3390/ijms241411305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Tendons help transmit forces from the skeletal muscles and bones. However, tendons have inferior regenerative ability compared to muscles. Despite studies on the regeneration of muscles and bone tissue, only a few have focused on tendinous tissue regeneration, especially tendon regeneration. Sex-determining region Y-box transcription factor 9 (Sox9) is an SRY-related transcription factor with a DNA-binding domain and is an important control factor for cartilage formation. Sox9 is critical to the early-to-middle stages of tendon development. However, how Sox9 participates in the healing process after tendon injury is unclear. We hypothesized that Sox9 is expressed in damaged tendons and is crucially involved in restoring tendon functions. We constructed a mouse model of an Achilles tendon injury by performing a 0.3 mm wide partial excision in the Achilles tendon of mice, and chronologically evaluated the function restoration and localization of the Sox9 expressed in the damaged sites. The results reveal that Sox9 was expressed simultaneously with the formation of the pre-structure of the epitenon, an essential part of the tendinous tissue, indicating that its expression is linked to the functional restoration of tendons. Lineage tracing for Sox9 expressed during tendon restoration revealed the tendon restoration involvement of cells that switched into Sox9-expressing cells after tendon injury. The stem cells involved in tendon regeneration may begin to express Sox9 after injury.
Collapse
Affiliation(s)
- Genji Watanabe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Masahito Yamamoto
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Shuichirou Taniguchi
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Yuki Sugiyama
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Hidetomo Hirouchi
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Satoshi Ishizuka
- Department of Pharmacology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Kei Kitamura
- Department of Histology and Developmental Biology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Toshihide Mizoguchi
- Oral Health Science Center, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Takashi Takayama
- Department of Dentistry, The Jikei University School of Medicine, 3-19-18 Nishi-shinnbashi, Minato, Tokyo 105-8471, Japan
| | - Katsuhiko Hayashi
- Department of Dentistry, The Jikei University School of Medicine, 3-19-18 Nishi-shinnbashi, Minato, Tokyo 105-8471, Japan
| | - Shinichi Abe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| |
Collapse
|
21
|
Pechanec MY, Beall JM, Katzman S, Maga EA, Mienaltowski MJ. Examining the Effects of In Vitro Co-Culture of Equine Adipose-Derived Mesenchymal Stem Cells With Tendon Proper and Peritenon Cells. J Equine Vet Sci 2023; 126:104262. [PMID: 36841345 DOI: 10.1016/j.jevs.2023.104262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 01/26/2023] [Accepted: 02/20/2023] [Indexed: 02/27/2023]
Abstract
Tendinopathies remain the leading contributor to career-ending injuries in horses because of the complexity of tendon repair. As such, cell-based therapies like injections of adipose-derived mesenchymal stem cells (ADMSCs, or MSCs) into injured tendons are becoming increasingly popular though their long-term efficacy on a molecular and wholistic level remains contentious. Thus, we co-cultured equine MSCs with intrinsic (tendon proper) and extrinsic (peritenon) tendon cell populations to examine interactions between these cells. Gene expression for common tenogenic, perivascular, and differentiation markers was quantified at 48 and 120 hours. Additionally, cellular metabolism of proliferation was examined every 24 hours for peritenon and tendon proper cells co-cultured with MSCs. MSCs co-cultured with tendon proper or peritenon cells had altered expression profiles demonstrating trend toward tenogenic phenotype with the exception of decreases in type I collagen (COL1A1). Peritenon cells co-cultured with MSCs had a trending and significant decrease in biglycan (BGN) and CSPG4 at 48 hours and 120 hours but overall significant increases in lysyl oxidase (LOX), mohawk (MKX), and scleraxis (SCX) within 48 hours. Tendon proper cells co-cultured with MSCs also exhibited increases in LOX and SCX at 48 hours. Furthermore, cell proliferation improved overall for tendon proper cells co-cultured with MSCs. The co-culture study results suggest that adipose-derived MSCs contribute beneficially to tenogenic stimulation of peritenon or tendon proper cells.
Collapse
Affiliation(s)
- Monica Y Pechanec
- Department of Animal Science, University of California Davis, Davis, CA
| | - Jessica M Beall
- Department of Animal Science, University of California Davis, Davis, CA
| | - Scott Katzman
- School of Veterinary Medicine, University of California Davis, Davis, CA
| | - Elizabeth A Maga
- Department of Animal Science, University of California Davis, Davis, CA
| | | |
Collapse
|
22
|
Hardy M, Feehan L, Savvides G, Wong J. How controlled motion alters the biophysical properties of musculoskeletal tissue architecture. J Hand Ther 2023; 36:269-279. [PMID: 37029054 DOI: 10.1016/j.jht.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/06/2022] [Indexed: 04/09/2023]
Abstract
INTRODUCTION Movement is fundamental to the normal behaviour of the hand, not only for day-to-day activity, but also for fundamental processes like development, tissue homeostasis and repair. Controlled motion is a concept that hand therapists apply to their patients daily for functional gains, yet the scientific understanding of how this works is poorly understood. PURPOSE OF THE ARTICLE To review the biology of the tissues in the hand that respond to movement and provide a basic science understanding of how it can be manipulated to facilitate better functionThe review outlines the concept of controlled motion and actions across the scales of tissue architecture, highlighting the the role of movement forces in tissue development, homeostasis and repair. The biophysical behaviour of mechanosensitve tissues of the hand such as skin, tendon, bone and cartilage are discussed. CONCLUSION Controlled motion during early healing is a form of controlled stress and can be harnessed to generate appropriate reparative tissues. Understanding the temporal and spatial biology of tissue repair allows therapists to tailor therapies that allow optimal recovery based around progressive biophysical stimuli by movement.
Collapse
Affiliation(s)
- Maureen Hardy
- Past Director Rehab Services and Hand Management Center, St. Dominic Hospital, Jackson, MS, USA
| | - Lynne Feehan
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Georgia Savvides
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, Manchester Academic Health Science Centre, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Jason Wong
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, Manchester Academic Health Science Centre, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
23
|
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
|
24
|
Nichols AE, Wagner NW, Ketonis C, Loiselle AE. Epitenon-derived cells comprise a distinct progenitor population that contributes to both tendon fibrosis and regeneration following acute injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526242. [PMID: 36778469 PMCID: PMC9915485 DOI: 10.1101/2023.01.30.526242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Flexor tendon injuries are common and heal poorly owing to both the deposition of function- limiting peritendinous scar tissue and insufficient healing of the tendon itself. Therapeutic options are limited due to a lack of understanding of the cell populations that contribute to these processes. Here, we identified a bi-fated progenitor cell population that originates from the epitenon and goes on to contribute to both peritendinous fibrosis and regenerative tendon healing following acute tendon injury. Using a combination of genetic lineage tracing and single cell RNA-sequencing (scRNA-seq), we profiled the behavior and contributions of each cell fate to the healing process in a spatio-temporal manner. Branched pseudotime trajectory analysis identified distinct transcription factors responsible for regulation of each fate. Finally, integrated scRNA-seq analysis of mouse healing with human peritendinous scar tissue revealed remarkable transcriptional similarity between mouse epitenon- derived cells and fibroblasts present in human peritendinous scar tissue, which was further validated by immunofluorescent staining for conserved markers. Combined, these results clearly identify the epitenon as the cellular origin of an important progenitor cell population that could be leveraged to improve tendon healing.
Collapse
|
25
|
Ackerman JE, Best KT, Muscat SN, Pritchett EM, Nichols AE, Wu CL, Loiselle AE. Defining the spatial-molecular map of fibrotic tendon healing and the drivers of Scleraxis-lineage cell fate and function. Cell Rep 2022; 41:111706. [PMID: 36417854 PMCID: PMC9741867 DOI: 10.1016/j.celrep.2022.111706] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
Tendon injuries heal via a scar-mediated response, and there are no biological approaches to promote more regenerative healing. Mouse flexor tendons heal through the formation of spatially distinct tissue areas: a highly aligned tissue bridge between the native tendon stubs that is enriched for adult Scleraxis-lineage cells and a disorganized outer shell associated with peri-tendinous scar formation. However, the specific molecular programs that underpin these spatially distinct tissue profiles are poorly defined. In the present study, we combine lineage tracing of adult Scleraxis-lineage cells with spatial transcriptomic profiling to define the overarching molecular programs that govern tendon healing and cell-fate decisions. Pseudotime analysis identified three fibroblast trajectories (synthetic, fibrotic, and reactive) and key transcription factors regulating these fate-switching decisions, including the progression of adult Scleraxis-lineage cells through the reactive trajectory. Collectively, this resource defines the molecular mechanisms that coordinate the temporo-spatial healing phenotype, which can be leveraged to inform therapeutic candidate selection.
Collapse
Affiliation(s)
- Jessica E. Ackerman
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, USA,Department of Pathology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - Katherine T. Best
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, USA,Department of Pathology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - Samantha N. Muscat
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, USA,Department of Pathology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - Elizabeth M. Pritchett
- Genomics Research Center, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - Anne E.C. Nichols
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Chia-Lung Wu
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, USA,Senior author
| | - Alayna E. Loiselle
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, USA,Department of Pathology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA,Senior author,Lead contact,Correspondence:
| |
Collapse
|
26
|
Amini M, Venkatesan JK, Liu W, Leroux A, Nguyen TN, Madry H, Migonney V, Cucchiarini M. Advanced Gene Therapy Strategies for the Repair of ACL Injuries. Int J Mol Sci 2022; 23:ijms232214467. [PMID: 36430947 PMCID: PMC9695211 DOI: 10.3390/ijms232214467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
The anterior cruciate ligament (ACL), the principal ligament for stabilization of the knee, is highly predisposed to injury in the human population. As a result of its poor intrinsic healing capacities, surgical intervention is generally necessary to repair ACL lesions, yet the outcomes are never fully satisfactory in terms of long-lasting, complete, and safe repair. Gene therapy, based on the transfer of therapeutic genetic sequences via a gene vector, is a potent tool to durably and adeptly enhance the processes of ACL repair and has been reported for its workability in various experimental models relevant to ACL injuries in vitro, in situ, and in vivo. As critical hurdles to the effective and safe translation of gene therapy for clinical applications still remain, including physiological barriers and host immune responses, biomaterial-guided gene therapy inspired by drug delivery systems has been further developed to protect and improve the classical procedures of gene transfer in the future treatment of ACL injuries in patients, as critically presented here.
Collapse
Affiliation(s)
- Mahnaz Amini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Jagadeesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Amélie Leroux
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Tuan Ngoc Nguyen
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Véronique Migonney
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
- Correspondence: or
| |
Collapse
|
27
|
Leiphart RJ, Weiss SN, DiStefano MS, Mavridis AA, Adams SA, Dyment NA, Soslowsky LJ. Collagen V deficiency during murine tendon healing results in distinct healing outcomes based on knockdown severity. J Biomech 2022; 144:111315. [PMID: 36201909 PMCID: PMC10108665 DOI: 10.1016/j.jbiomech.2022.111315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/11/2022] [Accepted: 09/16/2022] [Indexed: 10/31/2022]
Abstract
Tendon function is dependent on proper organization and maintenance of the collagen I tissue matrix. Collagen V is a critical regulator of collagen I fibrils, and while prior studies have shown a negative impact of collagen V deficiency on tendon healing outcomes, these studies are confounded by collagen V deficiency through tendon development. The specific role of collagen V in regulating healing tendon properties is therefore unknown. By using inducible Col5a1 knockdown models and analyzing gene expression, fibril and histological tendon morphology, and tendon mechanical properties, this study defines the isolated role of collagen V through tendon healing. Patellar tendon injury caused large changes in tendon gene expression, and Col5a1 knockdown resulted in dysregulated expression of several genes through tendon healing. Col5a1 knockdown also impacted collagen fibril size and shape without observable changes in scar tissue formation. Surprisingly, heterozygous Col5a1 knockdown resulted in improved stiffness of healing tendons that was not observed with homozygous Col5a1 knockdown. Together, these results present an unexpected and dynamic role of collagen V deficiency on tendon healing outcomes following injury. This work suggests a model of tendon healing in which quasi-static mechanics may be improved through titration of collagen fibril size and shape with modulation of collagen V expression and activity.
Collapse
Affiliation(s)
- R J Leiphart
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - S N Weiss
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M S DiStefano
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A A Mavridis
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - S A Adams
- University of South Florida, Morsani College of Medicine, Tampa, FL 33612, USA
| | - N A Dyment
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - L J Soslowsky
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
Korcari A, Muscat S, McGinn E, Buckley MR, Loiselle AE. Depletion of Scleraxis-lineage cells during tendon healing transiently impairs multi-scale restoration of tendon structure during early healing. PLoS One 2022; 17:e0274227. [PMID: 36240193 PMCID: PMC9565440 DOI: 10.1371/journal.pone.0274227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022] Open
Abstract
Tendons are composed of a heterogeneous cell environment, with Scleraxis-lineage (ScxLin) cells being the predominant population. Although ScxLin cells are required for maintenance of tendon homeostasis, their functions during tendon healing are unknown. To this end, we first characterized the spatiotemporal dynamics of ScxLin cells during tendon healing, and identified that the overall ScxLin pool continuously expands up to early remodeling healing phase. To better define the function of ScxLin cells during the late proliferative phase of healing, we inducibly depleted ScxLin cells from day 14-18 post-surgery using the Scx-Cre; Rosa-DTR mouse model, with local administration of diphtheria toxin inducing apoptosis of ScxLin cells in the healing tendon. At D28 post-surgery, ScxLin cell depleted tendons (DTRScxLin) had substantial impairments in structure and function, relative to WT, demonstrating the importance of ScxLin cells during tendon healing. Next, bulk RNAseq was utilized to identify the underlying mechanisms that were impaired with depletion and revealed that ScxLin depletion induced molecular and morphological stagnation of the healing process at D28. However, this stagnation was transient, such that by D56 tendon mechanics in DTRScxLin were not significantly different than wildtype repairs. Collectively, these data offer fundamental knowledge on the dynamics and roles of ScxLin cells during tendon healing.
Collapse
Affiliation(s)
- Antonion Korcari
- Department of Orthopaedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States of America
| | - Samantha Muscat
- Department of Orthopaedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Elizabeth McGinn
- Department of Orthopaedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States of America
| | - Mark R. Buckley
- Department of Orthopaedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States of America
| | - Alayna E. Loiselle
- Department of Orthopaedics & Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States of America
| |
Collapse
|
29
|
Dong C, Gingery A, Amadio PC, An KN, Moran SL, Zhao C. Apoptotic Body-Rich Media from Tenocytes Enhance Proliferation and Migration of Tenocytes and Bone Marrow Stromal Cells. Int J Mol Sci 2022; 23:11475. [PMID: 36232777 PMCID: PMC9569589 DOI: 10.3390/ijms231911475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
The intrinsic healing following tendon injury is ideal, in which tendon progenitor cells proliferate and migrate to the injury site to directly bridge or regenerate tendon tissue. However, the mechanism determining why and how those cells are attracted to the injury site for tendon healing is not understood. Since the tenocytes near the injury site go through apoptosis or necrosis following injury, we hypothesized that secretions from injured tenocytes might have biological effects on cell proliferation and migration to enhance tendon healing. Tenocyte apoptosis was induced by 24 h cell starvation. Apoptotic body-rich media (T-ABRM) and apoptotic body-depleted media (T-ABDM) were collected from culture media after centrifuging. Tenocytes and bone marrow-derived stem cells (BMDSCs) were isolated and cultured with the following four media: (1) T-ABRM, (2) T-ABDM, (3) GDF-5, or (4) basal medium with 2% fetal calf serum (FCS). The cell activities and functions were evaluated. Both T-ABRM and T-ABDM treatments significantly stimulated the cell proliferation, migration, and extracellular matrix synthesis for both tenocytes and BMDSCs compared to the control groups (GDF-5 and basal medium). However, cell proliferation, migration, and extracellular matrix production of T-ABRM-treated cells were significantly higher than the T-ABDM, which indicates the apoptotic bodies are critical for cell activities. Our study revealed the possible mechanism of the intrinsic healing of the tendon in which apoptotic bodies, in the process of apoptosis, following tendon injury promote tenocyte and stromal cell proliferation, migration, and production. Future studies should analyze the components of the apoptotic bodies that play this role, and, thus, the targeting of therapeutics can be developed.
Collapse
Affiliation(s)
- Chenhui Dong
- Biomechanics & Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, Rochester, MN 55905, USA
- Department of Sports medicine, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou 730050, China
| | - Anne Gingery
- Biomechanics & Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Peter C Amadio
- Biomechanics & Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Kai-Nan An
- Biomechanics & Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Steven L Moran
- Biomechanics & Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Chunfeng Zhao
- Biomechanics & Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
30
|
Walia B, Li T, Crosio G, Montero A, Huang A. Axin2-lineage cells contribute to neonatal tendon regeneration. Connect Tissue Res 2022; 63:530-543. [PMID: 35180018 PMCID: PMC9491382 DOI: 10.1080/03008207.2022.2036732] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 01/17/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Tendon injuries are a challenging clinical problem with few treatment options. Identifying the molecular regulators of tendon is required for the development of new therapies. While the Wnt pathway is critical for the maintenance and differentiation of many tissues, the role of Wnt signaling in tendon cell biology remains largely unexplored. METHODS The effects of Wnt activation were tested in vitro using neonatal tendon-derived cells cultured in 2D and 3D conditions. The inducible Axin2CreERT2 was then used to label Axin2+ cells in vivo and cells were traced during neonatal tendon regeneration. RESULTS We showed that activation of Wnt signaling results in proliferation of neonatal tendon cells. While tendon marker expression was inhibited by Wnt activation under 2D conditions, Scx expression was not affected under 3D uniaxial tension, suggesting that the microenvironment contextualizes tendon cell response to Wnt signaling. Using an in vivo model of neonatal tendon regeneration, we further showed that Wnt signaling cells comprise a subpopulation of tenocyte and epitenon cells that proliferate after injury and are recruited during regeneration. DISCUSSION Collectively, these studies suggest that Wnt signaling may play a role in tendon cell proliferation, differentiation, and regeneration.
Collapse
Affiliation(s)
- B. Walia
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - T.M. Li
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - G. Crosio
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - A.M. Montero
- Department of Orthopedic Surgery, Columbia University, New York, NY
| | - A.H. Huang
- Department of Orthopedic Surgery, Columbia University, New York, NY
| |
Collapse
|
31
|
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
|
32
|
Development and Regeneration of Muscle, Tendon, and Myotendinous Junctions in Striated Skeletal Muscle. Int J Mol Sci 2022; 23:ijms23063006. [PMID: 35328426 PMCID: PMC8950615 DOI: 10.3390/ijms23063006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Owing to a rapid increase in aging population in recent years, the deterioration of motor function in older adults has become an important social problem, and several studies have aimed to investigate the mechanisms underlying muscle function decline. Furthermore, structural maintenance of the muscle–tendon–bone complexes in the muscle attachment sites is important for motor function, particularly for joints; however, the development and regeneration of these complexes have not been studied thoroughly and require further elucidation. Recent studies have provided insights into the roles of mesenchymal progenitors in the development and regeneration of muscles and myotendinous junctions. In particular, studies on muscles and myotendinous junctions have—through the use of the recently developed scRNA-seq—reported the presence of syncytia, thereby suggesting that fibroblasts may be transformed into myoblasts in a BMP-dependent manner. In addition, the high mobility group box 1—a DNA-binding protein found in nuclei—is reportedly involved in muscle regeneration. Furthermore, studies have identified several factors required for the formation of locomotor apparatuses, e.g., tenomodulin (Tnmd) and mohawk (Mkx), which are essential for tendon maturation.
Collapse
|
33
|
Kallenbach JG, Freeberg MAT, Abplanalp D, Alenchery RG, Ajalik RE, Muscat S, Myers JA, Ashton JM, Loiselle A, Buckley MR, van Wijnen AJ, Awad HA. Altered TGFB1 regulated pathways promote accelerated tendon healing in the superhealer MRL/MpJ mouse. Sci Rep 2022; 12:3026. [PMID: 35194136 PMCID: PMC8863792 DOI: 10.1038/s41598-022-07124-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 02/11/2022] [Indexed: 12/23/2022] Open
Abstract
To better understand the molecular mechanisms of tendon healing, we investigated the Murphy Roth's Large (MRL) mouse, which is considered a model of mammalian tissue regeneration. We show that compared to C57Bl/6J (C57) mice, injured MRL tendons have reduced fibrotic adhesions and cellular proliferation, with accelerated improvements in biomechanical properties. RNA-seq analysis revealed that differentially expressed genes in the C57 healing tendon at 7 days post injury were functionally linked to fibrosis, immune system signaling and extracellular matrix (ECM) organization, while the differentially expressed genes in the MRL injured tendon were dominated by cell cycle pathways. These gene expression changes were associated with increased α-SMA+ myofibroblast and F4/80+ macrophage activation and abundant BCL-2 expression in the C57 injured tendons. Transcriptional analysis of upstream regulators using Ingenuity Pathway Analysis showed positive enrichment of TGFB1 in both C57 and MRL healing tendons, but with different downstream transcriptional effects. MRL tendons exhibited of cell cycle regulatory genes, with negative enrichment of the cell senescence-related regulators, compared to the positively-enriched inflammatory and fibrotic (ECM organization) pathways in the C57 tendons. Serum cytokine analysis revealed decreased levels of circulating senescence-associated circulatory proteins in response to injury in the MRL mice compared to the C57 mice. These data collectively demonstrate altered TGFB1 regulated inflammatory, fibrosis, and cell cycle pathways in flexor tendon repair in MRL mice, and could give cues to improved tendon healing.
Collapse
Affiliation(s)
- Jacob G Kallenbach
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Margaret A T Freeberg
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - David Abplanalp
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Rahul G Alenchery
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Raquel E Ajalik
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Samantha Muscat
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacquelyn A Myers
- UR Genomics Research Center (GRC), University of Rochester Medical Center, Rochester, NY, USA
| | - John M Ashton
- UR Genomics Research Center (GRC), University of Rochester Medical Center, Rochester, NY, USA
| | - Alayna Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
| | - Mark R Buckley
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Hani A Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.
| |
Collapse
|
34
|
Leiphart RJ, Pham H, Harvey T, Komori T, Kilts TM, Shetye SS, Weiss SN, Adams SM, Birk DE, Soslowsky LJ, Young MF. Coordinate roles for collagen VI and biglycan in regulating tendon collagen fibril structure and function. Matrix Biol Plus 2022; 13:100099. [PMID: 35036900 PMCID: PMC8749075 DOI: 10.1016/j.mbplus.2021.100099] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 11/28/2022] Open
Abstract
Tendon is a vital musculoskeletal tissue that is prone to degeneration. Proper tendon maintenance requires complex interactions between extracellular matrix components that remain poorly understood. Collagen VI and biglycan are two matrix molecules that localize pericellularly within tendon and are critical regulators of tissue properties. While evidence suggests that collagen VI and biglycan interact within the tendon matrix, the relationship between the two molecules and its impact on tendon function remains unknown. We sought to elucidate potential coordinate roles of collagen VI and biglycan within tendon by defining tendon properties in knockout models of collagen VI, biglycan, or both molecules. We first demonstrated co-expression and co-localization of collagen VI and biglycan within the healing tendon, providing further evidence of cooperation between the two molecules during nascent tendon matrix formation. Deficiency in collagen VI and/or biglycan led to significant reductions in collagen fibril size and tendon mechanical properties. However, collagen VI-null tendons displayed larger reductions in fibril size and mechanics than seen in biglycan-null tendons. Interestingly, knockout of both molecules resulted in similar properties to collagen VI knockout alone. These results indicate distinct and non-additive roles for collagen VI and biglycan within tendon. This work provides better understanding of regulatory interactions between two critical tendon matrix molecules.
Collapse
Affiliation(s)
- Ryan J. Leiphart
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Hai Pham
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tyler Harvey
- Carnegie Institution for Science, Department of Embryology, The Johns Hopkins University, USA
| | - Taishi Komori
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tina M. Kilts
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Snehal S. Shetye
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephanie N. Weiss
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Sheila M. Adams
- University of South Florida, Morsani College of Medicine, Tampa, FL 33612, USA
| | - David E. Birk
- University of South Florida, Morsani College of Medicine, Tampa, FL 33612, USA
| | - Louis J. Soslowsky
- McKay Orthopedic Research Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Marian F. Young
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Ding L, Zhou B, Hou Y, Xu L. Stem cells in tendon regeneration and factors governing tenogenesis. Curr Stem Cell Res Ther 2022; 17:503-512. [PMID: 35086458 DOI: 10.2174/1574888x17666220127111135] [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: 08/23/2021] [Revised: 11/16/2021] [Accepted: 12/08/2021] [Indexed: 11/22/2022]
Abstract
Tendons are connective tissue structures of paramount importance to the human ability of locomotion. Tendinopathy and tendon rupture can be resistant to treatment and often recurs, thus resulting in a significant health problem with a relevant social impact worldwide. Unfortunately, existing treatment approaches are suboptimal. A better understanding of the basic biology of tendons may provide a better way to solve these problems and promote tendon regeneration. Stem cells, either obtained from tendons or non-tendon sources, such as bone marrow (BMSCs), adipose tissue (AMSCs), as well as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have received increasing attention toward enhancing tendon healing. There are many studies showing that stem cells can contribute to improving tendon healing. Hence, in this review, the current knowledge of BMSCs, AMSCs, TSPCs, ESCs and iPSCs for tendon regeneration, as well as the advantages and limitations among them, has been highlighted. Moreover, the transcriptional and bioactive factors governing tendon healing processes have been discussed.
Collapse
Affiliation(s)
- Lingli Ding
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - BingYu Zhou
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yonghui Hou
- Key Laboratory of Orthopaedics & Traumatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Liangliang Xu
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
36
|
He P, Ruan D, Huang Z, Wang C, Xu Y, Cai H, Liu H, Fei Y, Heng BC, Chen W, Shen W. Comparison of Tendon Development Versus Tendon Healing and Regeneration. Front Cell Dev Biol 2022; 10:821667. [PMID: 35141224 PMCID: PMC8819183 DOI: 10.3389/fcell.2022.821667] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/07/2022] [Indexed: 12/27/2022] Open
Abstract
Tendon is a vital connective tissue in human skeletal muscle system, and tendon injury is very common and intractable in clinic. Tendon development and repair are two closely related but still not fully understood processes. Tendon development involves multiple germ layer, as well as the regulation of diversity transcription factors (Scx et al.), proteins (Tnmd et al.) and signaling pathways (TGFβ et al.). The nature process of tendon repair is roughly divided in three stages, which are dominated by various cells and cell factors. This review will describe the whole process of tendon development and compare it with the process of tendon repair, focusing on the understanding and recent advances in the regulation of tendon development and repair. The study and comparison of tendon development and repair process can thus provide references and guidelines for treatment of tendon injuries.
Collapse
Affiliation(s)
- Peiwen He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Zizhan Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yiwen Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Honglu Cai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Hengzhi Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School of Stomatology, Bejing, China
| | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| |
Collapse
|
37
|
Tan GK, Pryce BA, Stabio A, Keene DR, Tufa SF, Schweitzer R. Cell autonomous TGFβ signaling is essential for stem/progenitor cell recruitment into degenerative tendons. Stem Cell Reports 2021; 16:2942-2957. [PMID: 34822771 PMCID: PMC8693658 DOI: 10.1016/j.stemcr.2021.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/03/2022] Open
Abstract
Understanding cell recruitment in damaged tendons is critical for improvements in regenerative therapy. We recently reported that targeted disruption of transforming growth factor beta (TGFβ) type II receptor in the tendon cell lineage (Tgfbr2ScxCre) resulted in resident tenocyte dedifferentiation and tendon deterioration in postnatal stages. Here we extend the analysis and identify direct recruitment of stem/progenitor cells into the degenerative mutant tendons. Cre-mediated lineage tracing indicates that these cells are not derived from tendon-ensheathing tissues or from a Scleraxis-expressing lineage, and they turned on tendon markers only upon entering the mutant tendons. Through immunohistochemistry and inducible gene deletion, we further find that the recruited cells originated from a Sox9-expressing lineage and their recruitment was dependent on cell autonomous TGFβ signaling. The cells identified in this study thus differ from previous reports of cell recruitment into injured tendons and suggest a critical role for TGFβ signaling in cell recruitment, providing insights that may support improvements in tendon repair. Targeted deletion of TGFβ signaling led to degenerative changes in mouse tendons Stem/progenitor cells were recruited into the degenerative mutant tendons The recruited cells are different from the ones so far reported in tendon injury Recruitment was dependent on cell autonomous TGFβ signaling in the recruited cells
Collapse
Affiliation(s)
- Guak-Kim Tan
- Research Division, Shriners Hospital for Children, Portland, OR 97239, USA; Department of Orthopaedics and Rehabilitation, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Brian A Pryce
- Research Division, Shriners Hospital for Children, Portland, OR 97239, USA
| | - Anna Stabio
- Research Division, Shriners Hospital for Children, Portland, OR 97239, USA
| | - Douglas R Keene
- Research Division, Shriners Hospital for Children, Portland, OR 97239, USA
| | - Sara F Tufa
- Research Division, Shriners Hospital for Children, Portland, OR 97239, USA
| | - Ronen Schweitzer
- Research Division, Shriners Hospital for Children, Portland, OR 97239, USA; Department of Orthopaedics and Rehabilitation, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
38
|
Secretome from In Vitro Mechanically Loaded Myoblasts Induces Tenocyte Migration, Transition to a Fibroblastic Phenotype and Suppression of Collagen Production. Int J Mol Sci 2021; 22:ijms222313089. [PMID: 34884895 PMCID: PMC8657858 DOI: 10.3390/ijms222313089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
It is known that mechanical loading of muscles increases the strength of healing tendon tissue, but the mechanism involved remains elusive. We hypothesized that the secretome from myoblasts in co-culture with tenocytes affects tenocyte migration, cell phenotype, and collagen (Col) production and that the effect is dependent on different types of mechanical loading of myoblasts. To test this, we used an in vitro indirect transwell co-culture system. Myoblasts were mechanically loaded using the FlexCell® Tension system. Tenocyte cell migration, proliferation, apoptosis, collagen production, and several tenocyte markers were measured. The secretome from myoblasts decreased the Col I/III ratio and increased the expression of tenocyte specific markers as compared with tenocytes cultured alone. The secretome from statically loaded myoblasts significantly enhanced tenocyte migration and Col I/III ratio as compared with dynamic loading and controls. In addition, the secretome from statically loaded myoblasts induced tenocytes towards a myofibroblast-like phenotype. Taken together, these results demonstrate that the secretome from statically loaded myoblasts has a profound influence on tenocytes, affecting parameters that are related to the tendon healing process.
Collapse
|
39
|
LeVasseur MR, Hawthorne BC, Mancini MR, McCarthy MBR, Wellington IJ, Cote MP, Solovyova O, Williams VJ, Mazzocca AD. Trochanteric Bursa Is a Source of Connective Tissue Progenitor Cells. Arthrosc Sports Med Rehabil 2021; 3:e1661-e1670. [PMID: 34977618 PMCID: PMC8689227 DOI: 10.1016/j.asmr.2021.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/25/2021] [Indexed: 12/27/2022] Open
Abstract
Purpose To investigate the presence of connective tissue progenitor cells (CTPs) in the trochanteric bursa harvested over the gluteus medius muscle belly and tendon during open hip procedures. Methods Trochanteric bursa samples from nine patients (63.1 ± 8.6 years) undergoing total hip arthroplasty for primary osteoarthritis were obtained from 2 sites: over the gluteus medius tendon at the greater trochanter and over the muscle belly. Bursal tissue was digested with collagenase and grown in culture. The nucleated cell count, cellular concentration, cellular proliferation, fluorescence-activated cell sorting (FACS) analysis, and differentiation using immunostaining and quantitative polymerase chain reaction (PCR) were used to determine and quantify the presence of CTPs. Results Bursa-derived CTPs were identified in all harvested samples. At t = 0, there was no difference in nucleated cell count over muscle and tendon (1.69 ± 1.26 × 108 and 1.41 ± 1.12 × 108 cells/g, respectively; P = .162). Similarly, the cellular concentration at 3 weeks was not significantly different between bursa harvested over muscle and tendon (6.61 ± 5.14 × 106 and 5.58 ± 4.70 × 106 cells/g, respectively; P = .532). High cellular proliferation was identified for both bursal tissue overlying muscle and tendon (2.28 ± .95 and 1.66 ± 1.05, respectively; P = .194). FACS analysis revealed high positivity rates (>95%) of CTP-specific surface epitopes (CD105, CD90, and CD73) and low positivity rates (<1.3%) of negative markers (CD45, CD31). Osteogenic, adipogenic, and chondrogenic differentiation potential was demonstrated with immunostaining and quantitative PCR for gene expression. Conclusions Connective tissue progenitor cells are found in the trochanteric bursa overlying the muscle and tendon of the hip abductors. Clinical Relevance During open hip procedures, the trochanteric bursa is often partially excised to identify muscular boundaries and tissue planes for surgical exposure. The function of the trochanteric bursa remains largely unknown. However, this tissue is a source of connective tissue progenitor cells, which may be important in the healing response of surgically repaired abductor tendons.
Collapse
Affiliation(s)
- Matthew R LeVasseur
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Benjamin C Hawthorne
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Michael R Mancini
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Mary Beth R McCarthy
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Ian J Wellington
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Mark P Cote
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Olga Solovyova
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Vincent J Williams
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Augustus D Mazzocca
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| |
Collapse
|
40
|
Mienaltowski MJ, Gonzales NL, Beall JM, Pechanec MY. Basic Structure, Physiology, and Biochemistry of Connective Tissues and Extracellular Matrix Collagens. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:5-43. [PMID: 34807414 DOI: 10.1007/978-3-030-80614-9_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The physiology of connective tissues like tendons and ligaments is highly dependent upon the collagens and other such extracellular matrix molecules hierarchically organized within the tissues. By dry weight, connective tissues are mostly composed of fibrillar collagens. However, several other forms of collagens play essential roles in the regulation of fibrillar collagen organization and assembly, in the establishment of basement membrane networks that provide support for vasculature for connective tissues, and in the formation of extensive filamentous networks that allow for cell-extracellular matrix interactions as well as maintain connective tissue integrity. The structures and functions of these collagens are discussed in this chapter. Furthermore, collagen synthesis is a multi-step process that includes gene transcription, translation, post-translational modifications within the cell, triple helix formation, extracellular secretion, extracellular modifications, and then fibril assembly, fibril modifications, and fiber formation. Each step of collagen synthesis and fibril assembly is highly dependent upon the biochemical structure of the collagen molecules created and how they are modified in the cases of development and maturation. Likewise, when the biochemical structures of collagens or are compromised or these molecules are deficient in the tissues - in developmental diseases, degenerative conditions, or injuries - then the ultimate form and function of the connective tissues are impaired. In this chapter, we also review how biochemistry plays a role in each of the processes involved in collagen synthesis and assembly, and we describe differences seen by anatomical location and region within tendons. Moreover, we discuss how the structures of the molecules, fibrils, and fibers contribute to connective tissue physiology in health, and in pathology with injury and repair.
Collapse
Affiliation(s)
| | - Nicole L Gonzales
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - Jessica M Beall
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - Monica Y Pechanec
- Department of Animal Science, University of California Davis, Davis, CA, USA
| |
Collapse
|
41
|
Notermans T, Khayyeri H, Isaksson H. Predicting the effect of reduced load level and cell infiltration on spatio-temporal Achilles tendon healing. J Biomech 2021; 139:110853. [PMID: 34838291 DOI: 10.1016/j.jbiomech.2021.110853] [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: 03/29/2021] [Revised: 10/13/2021] [Accepted: 10/27/2021] [Indexed: 10/19/2022]
Abstract
Mechanobiology plays an important role in tendon healing. However, the relationship between mechanical loading and spatial and temporal evolution of tendon properties during healing is not well understood. This study builds on a recently presented mechanoregulatory computational framework that couples mechanobiological tendon healing to tissue production and collagen orientation. In this study, we investigated how different magnitudes of mechanical stimulation (principal strain) affect the spatio-temporal evolution of tissue production and the temporal evolution of elastic and viscoelastic mechanical parameters. Specifically, we examined the effect of cell infiltration (mimicking migration and proliferation) in the callus on the resulting tissue production by modeling production to depend on local cell density. The model predictions were carefully compared with experimental data from Achilles tendons in rats, at 1, 2 and 4 weeks of healing. In the experiments, the rat tendons had been subjected to free cage activity or reduced load levels through intramuscular botox injections. The simulations that included cell infiltration and strain-regulated collagen production predicted spatio-temporal tissue distributions and mechanical properties similarly to that observed experimentally. In addition, lack of matrix-producing cells in the tendon core during early healing may result in reduced collagen content, regardless of the daily load level. This framework is the first to computationally investigate mechanobiological mechanisms underlying spatial and temporal variations during tendon healing for various magnitudes of loading. This framework will allow further characterization of biomechanical, biological, or mechanobiological processes underlying tendon healing.
Collapse
Affiliation(s)
- Thomas Notermans
- Department of Biomedical Engineering, Lund University, BMC D13, 22184 Lund, Sweden.
| | - Hanifeh Khayyeri
- Department of Biomedical Engineering, Lund University, BMC D13, 22184 Lund, Sweden
| | - Hanna Isaksson
- Department of Biomedical Engineering, Lund University, BMC D13, 22184 Lund, Sweden
| |
Collapse
|
42
|
Delgado Caceres M, Angerpointner K, Galler M, Lin D, Michel PA, Brochhausen C, Lu X, Varadarajan AR, Warfsmann J, Stange R, Alt V, Pfeifer CG, Docheva D. Tenomodulin knockout mice exhibit worse late healing outcomes with augmented trauma-induced heterotopic ossification of Achilles tendon. Cell Death Dis 2021; 12:1049. [PMID: 34741033 PMCID: PMC8571417 DOI: 10.1038/s41419-021-04298-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 12/23/2022]
Abstract
Heterotopic ossification (HO) represents a common problem after tendon injury with no effective treatment yet being developed. Tenomodulin (Tnmd), the best-known mature marker for tendon lineage cells, has important effects in tendon tissue aging and function. We have reported that loss of Tnmd leads to inferior early tendon repair characterized by fibrovascular scaring and therefore hypothesized that its lack will persistently cause deficient repair during later stages. Tnmd knockout (Tnmd-/-) and wild-type (WT) animals were subjected to complete Achilles tendon surgical transection followed by end-to-end suture. Lineage tracing revealed a reduction in tendon-lineage cells marked by ScleraxisGFP, but an increase in alpha smooth muscle actin myofibroblasts in Tnmd-/- tendon scars. At the proliferative stage, more pro-inflammatory M1 macrophages and larger collagen II cartilaginous template were detected in this group. At the remodeling stage, histological scoring revealed lower repair quality in the injured Tnmd-/- tendons, which was coupled with higher HO quantified by micro-CT. Tendon biomechanical properties were compromised in both groups upon injury, however we identified an abnormal stiffening of non-injured Tnmd-/- tendons, which possessed higher static and dynamic E-moduli. Pathologically thicker and abnormally shaped collagen fibrils were observed by TEM in Tnmd-/- tendons and this, together with augmented HO, resulted in diminished running capacity of Tnmd-/- mice. These novel findings demonstrate that Tnmd plays a protecting role against trauma-induced endochondral HO and can inspire the generation of novel therapeutics to accelerate repair.
Collapse
Affiliation(s)
- Manuel Delgado Caceres
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Katharina Angerpointner
- Hand, Elbow and Plastic Surgery Department, Schön Klinik München Harlaching, Munich, Germany
| | - Michael Galler
- Department of Trauma Surgery, Caritas Hospital St. Josef, Regensburg, Germany
| | - Dasheng Lin
- Orthopaedic Center of People's Liberation Army, The Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Philipp A Michel
- Department of Trauma-, Hand-, and Reconstructive Surgery, University Hospital Münster, Münster, Germany
| | | | - Xin Lu
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Adithi R Varadarajan
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Jens Warfsmann
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute for Musculoskeletal Medicine, University Hospital Münster, Westfälische Wilhelms-University, Münster, Germany
| | - Volker Alt
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Clinic and Policlinic for Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Christian G Pfeifer
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Clinic and Policlinic for Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
43
|
Stauber T, Wolleb M, Duss A, Jaeger PK, Heggli I, Hussien AA, Blache U, Snedeker JG. Extrinsic Macrophages Protect While Tendon Progenitors Degrade: Insights from a Tissue Engineered Model of Tendon Compartmental Crosstalk. Adv Healthc Mater 2021; 10:e2100741. [PMID: 34494401 PMCID: PMC11468160 DOI: 10.1002/adhm.202100741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/30/2021] [Indexed: 12/15/2022]
Abstract
Tendons are among the most mechanically stressed tissues of the body, with a functional core of type-I collagen fibers maintained by embedded stromal fibroblasts known as tenocytes. The intrinsic load-bearing core compartment of tendon is surrounded, nourished, and repaired by the extrinsic peritendon, a synovial-like tissue compartment with access to tendon stem/progenitor cells as well as blood monocytes. In vitro tendon model systems generally lack this important feature of tissue compartmentalization, while in vivo models are cumbersome when isolating multicellular mechanisms. To bridge this gap, an improved in vitro model of explanted tendon core stromal tissue (mouse tail tendon fascicles) surrounded by cell-laden collagen hydrogels that mimic extrinsic tissue compartments is suggested. Using this model, CD146+ tendon stem/progenitor cell and CD45+ F4/80+ bone-marrow derived macrophage activity within a tendon injury-like niche are recapitulated. It is found that extrinsic stromal progenitors recruit to the damaged core, contribute to an overall increase in catabolic ECM gene expression, and accelerate the decrease in mechanical properties. Conversely, it is found that extrinsic bone-marrow derived macrophages in these conditions adopt a proresolution phenotype that mitigates rapid tissue breakdown by outwardly migrated tenocytes and F4/80+ "tenophages" from the intrinsic tissue core.
Collapse
Affiliation(s)
- Tino Stauber
- Department of OrthopedicsBalgrist University HospitalUniversity of ZurichLengghalde 5Zurich8008Switzerland
- Institute for BiomechanicsETH ZurichZurich8093Switzerland
| | - Maja Wolleb
- Department of OrthopedicsBalgrist University HospitalUniversity of ZurichLengghalde 5Zurich8008Switzerland
- Institute for BiomechanicsETH ZurichZurich8093Switzerland
| | - Anja Duss
- Department of OrthopedicsBalgrist University HospitalUniversity of ZurichLengghalde 5Zurich8008Switzerland
- Institute for BiomechanicsETH ZurichZurich8093Switzerland
| | - Patrick K. Jaeger
- Department of OrthopedicsBalgrist University HospitalUniversity of ZurichLengghalde 5Zurich8008Switzerland
- Institute for BiomechanicsETH ZurichZurich8093Switzerland
| | - Irina Heggli
- Center of Experimental RheumatologyDepartment of RheumatologyUniversity Hospital, University of ZurichLengghalde 5Zurich8008Switzerland
| | - Amro A. Hussien
- Department of OrthopedicsBalgrist University HospitalUniversity of ZurichLengghalde 5Zurich8008Switzerland
- Institute for BiomechanicsETH ZurichZurich8093Switzerland
| | - Ulrich Blache
- Department of OrthopedicsBalgrist University HospitalUniversity of ZurichLengghalde 5Zurich8008Switzerland
- Institute for BiomechanicsETH ZurichZurich8093Switzerland
- Fraunhofer Institute for Cell Therapy and Immunology04103LeipzigGermany
| | - Jess G. Snedeker
- Department of OrthopedicsBalgrist University HospitalUniversity of ZurichLengghalde 5Zurich8008Switzerland
- Institute for BiomechanicsETH ZurichZurich8093Switzerland
| |
Collapse
|
44
|
Hou J, Yang R, Vuong I, Li F, Kong J, Mao HQ. Biomaterials strategies to balance inflammation and tenogenesis for tendon repair. Acta Biomater 2021; 130:1-16. [PMID: 34082095 DOI: 10.1016/j.actbio.2021.05.043] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/15/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022]
Abstract
Adult tendon tissue demonstrates a limited regenerative capacity, and the natural repair process leaves fibrotic scar tissue with inferior mechanical properties. Surgical treatment is insufficient to provide the mechanical, structural, and biochemical environment necessary to restore functional tissue. While numerous strategies including biodegradable scaffolds, bioactive factor delivery, and cell-based therapies have been investigated, most studies have focused exclusively on either suppressing inflammation or promoting tenogenesis, which includes tenocyte proliferation, ECM production, and tissue formation. New biomaterials-based approaches represent an opportunity to more effectively balance the two processes and improve regenerative outcomes from tendon injuries. Biomaterials applications that have been explored for tendon regeneration include formation of biodegradable scaffolds presenting topographical, mechanical, and/or immunomodulatory cues conducive to tendon repair; delivery of immunomodulatory or tenogenic biomolecules; and delivery of therapeutic cells such as tenocytes and stem cells. In this review, we provide the biological context for the challenges in tendon repair, discuss biomaterials approaches to modulate the immune and regenerative environment during the healing process, and consider the future development of comprehensive biomaterials-based strategies that can better restore the function of injured tendon. STATEMENT OF SIGNIFICANCE: Current strategies for tendon repair focus on suppressing inflammation or enhancing tenogenesis. Evidence indicates that regulated inflammation is beneficial to tendon healing and that excessive tissue remodeling can cause fibrosis. Thus, it is necessary to adopt an approach that balances the benefits of regulated inflammation and tenogenesis. By reviewing potential treatments involving biodegradable scaffolds, biological cues, and therapeutic cells, we contrast how each strategy promotes or suppresses specific repair steps to improve the healing outcome, and highlight the advantages of a comprehensive approach that facilitates the clearance of necrotic tissue and recruitment of cells during the inflammatory stage, followed by ECM synthesis and organization in the proliferative and remodeling stages with the goal of restoring function to the tendon.
Collapse
|
45
|
Microscopic characterisation of local strain field in healing tissue in the central third defect of mouse patellar tendon at early-phase of healing. J Mech Behav Biomed Mater 2021; 123:104702. [PMID: 34365097 DOI: 10.1016/j.jmbbm.2021.104702] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/26/2021] [Accepted: 07/03/2021] [Indexed: 11/20/2022]
Abstract
Tendons exhibit a hierarchical collagen structure, wherein higher-level components, such as collagen fibres and fascicles, are elongated, slid, and rotated during macroscopic stretching. These mechanical behaviours of collagen fibres play important roles in stimulating tenocytes, imposing stretching, compression, and shear deformation. It was hypothesised that a lack of local fibre behaviours in healing tendon tissue may result in a limited application of mechanical stimuli to cells within the tissue, leading to incomplete recovery of tissue structure and functions in regenerated tendons. Therefore, the present study aimed to measure the microscopic strain field in the healing tendon tissue. A central third defect was created in the patellar tendon of mice, and the regenerated tissue in the defect was examined by tensile testing, collagen fibre analysis, and local strain measurement using confocal microscopy at 3 and 6 weeks after surgery. Healing tissue at 3 weeks exhibited a significantly lower strength and disorganised collagen fibre structure compared with the normal tendon. These characteristics at 6 weeks remained significantly different from those of the normal tendon. Moreover, the magnitude of local shear strain in the healing tissue under 4% tissue strain was significantly smaller than that in the normal tendon. Differences in the local strain field may be reflected in the cell nuclear shape and possibly the amount of mechanical stimuli applied to the cells during tendon deformation. Accordingly, restoration of a normal local mechanical environment in the healing tissue may be key to a better healing outcome of tendon injury.
Collapse
|
46
|
Arvind V, Huang AH. Reparative and Maladaptive Inflammation in Tendon Healing. Front Bioeng Biotechnol 2021; 9:719047. [PMID: 34350166 PMCID: PMC8327090 DOI: 10.3389/fbioe.2021.719047] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/28/2021] [Indexed: 12/26/2022] Open
Abstract
Tendon injuries are common and debilitating, with non-regenerative healing often resulting in chronic disease. While there has been considerable progress in identifying the cellular and molecular regulators of tendon healing, the role of inflammation in tendon healing is less well understood. While inflammation underlies chronic tendinopathy, it also aids debris clearance and signals tissue repair. Here, we highlight recent findings in this area, focusing on the cells and cytokines involved in reparative inflammation. We also discuss findings from other model systems when research in tendon is minimal, and explore recent studies in the treatment of human tendinopathy to glean further insights into the immunobiology of tendon healing.
Collapse
Affiliation(s)
- Varun Arvind
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alice H. Huang
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Orthopedic Surgery, Columbia University, New York, NY, United States
| |
Collapse
|
47
|
Nichols AEC, Muscat SN, Miller SE, Green LJ, Richards MS, Loiselle AE. Impact of isolation method on cellular activation and presence of specific tendon cell subpopulations during in vitro culture. FASEB J 2021; 35:e21733. [PMID: 34160846 DOI: 10.1096/fj.202100405r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 11/11/2022]
Abstract
Tendon injuries are common and heal poorly, due in part to a lack of understanding of fundamental tendon cell biology. A major impediment to the study of tendon cells is the absence of robust, well-characterized in vitro models. Unlike other tissue systems, current tendon cell models do not account for how differences in isolation methodology may affect the activation state of tendon cells or the presence of various tendon cell subpopulations. The objective of this study was to characterize how common isolation methods affect the behavior, fate, and lineage composition of tendon cell cultures. Tendon cells isolated by explant exhibited reduced proliferative capacity, decreased expression of tendon marker genes, and increased expression of genes associated with fibroblast activation compared to digested cells. Consistently, explanted cells also displayed an increased propensity to differentiate to myofibroblasts compared to digested cells. Explanted cultures from multiple different tendons were substantially enriched for the presence of scleraxis-lineage (Scx-lin+) cells compared to digested cultures, while the overall percentage of S100a4-lineage (S100a4-lin+) cells was dependent on both isolation method and tendon of origin. Neither isolation methods preserved the ratios of Scx-lin+ or S100a4-lin+ to non-lineage cells seen in tendons in vivo. Combined, these data indicate that further refinement of in vitro cultures models is required in order to more accurately understand the effects of various stimuli on tendon cell behavior. Statement of clinical significance: The development of informed in vitro tendon cell models will facilitate enhanced screening of potential therapeutic candidates to improve tendon healing.
Collapse
Affiliation(s)
- Anne E C Nichols
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Samantha N Muscat
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Sarah E Miller
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Luke J Green
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Michael S Richards
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
48
|
The interaction between human rotator cuff tendon and subacromial bursal tissue in co-culture. J Shoulder Elbow Surg 2021; 30:1494-1502. [PMID: 33197595 DOI: 10.1016/j.jse.2020.09.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/07/2020] [Accepted: 09/21/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND The role of subacromial bursa in rotator cuff pathology is unclear. Along with recognized inflammatory potential, current data demonstrate the presence of mesenchymal stem cells and potential regenerative properties of the bursa. The purpose of this study was to (1) approximate an in vitro co-culture model that represents interaction between torn rotator cuff tendon and subacromial bursa, (2) quantify the cellular activity of tendon and bursa and their interactions, (3) use this model to induce a state of inflammation present with rotator cuff pathology. METHODS In part 1, tendon and bursa samples were obtained from 6 patients undergoing rotator cuff repair. Tendon and bursa were cultured alone and together in co-culture wells for 21 days. Markers specific for tenocyte gene expression (tenascin C, decorin, etc) were measured in both tendon and bursa alone and compared to co-culture models. In part 2 of the study, an inflammatory state was induced with interleukin-1β treatment, and markers of inflammation were measured via protein assay at 0 and 21 days in samples from 7 additional patients. RESULTS There was an increase in tendon and bursa markers in nearly all groups as evidenced by increased gene expression of known tendon and bursa markers. There was a significant increase in gene expression when torn tendon was co-cultured with bursa compared with culturing alone. Additionally, a state of inflammation was induced as evidenced by increased markers of inflammation, inflammatory protein concentration, and inflammatory cells and disruption of histologic morphology. CONCLUSION There is a clear interaction between rotator cuff tendon and the milieu produced by the subacromial bursa in this in vitro co-culture system that is significantly different when compared to an isolated culture of tendon and bursa. This system was successfully used to induce a state of inflammation that may represent in vivo inflammation. This in vitro model of rotator cuff pathology can aid investigators in testing effects of agents proposed to improve rotator cuff healing. This can lead to further knowledge regarding effective treatment options.
Collapse
|
49
|
Bobzin L, Roberts RR, Chen HJ, Crump JG, Merrill AE. Development and maintenance of tendons and ligaments. Development 2021; 148:239823. [PMID: 33913478 DOI: 10.1242/dev.186916] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tendons and ligaments are fibrous connective tissues vital to the transmission of force and stabilization of the musculoskeletal system. Arising in precise regions of the embryo, tendons and ligaments share many properties and little is known about the molecular differences that differentiate them. Recent studies have revealed heterogeneity and plasticity within tendon and ligament cells, raising questions regarding the developmental mechanisms regulating tendon and ligament identity. Here, we discuss recent findings that contribute to our understanding of the mechanisms that establish and maintain tendon progenitors and their differentiated progeny in the head, trunk and limb. We also review the extent to which these findings are specific to certain anatomical regions and model organisms, and indicate which findings similarly apply to ligaments. Finally, we address current research regarding the cellular lineages that contribute to tendon and ligament repair, and to what extent their regulation is conserved within tendon and ligament development.
Collapse
Affiliation(s)
- Lauren Bobzin
- Division of Biomedical Sciences, Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA.,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ryan R Roberts
- Division of Biomedical Sciences, Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA.,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Hung-Jhen Chen
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - J Gage Crump
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amy E Merrill
- Division of Biomedical Sciences, Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA.,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
50
|
Mondrinos MJ, Alisafaei F, Yi AY, Ahmadzadeh H, Lee I, Blundell C, Seo J, Osborn M, Jeon TJ, Kim SM, Shenoy VB, Huh D. Surface-directed engineering of tissue anisotropy in microphysiological models of musculoskeletal tissue. SCIENCE ADVANCES 2021; 7:7/11/eabe9446. [PMID: 33712463 PMCID: PMC7954445 DOI: 10.1126/sciadv.abe9446] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/27/2021] [Indexed: 05/11/2023]
Abstract
Here, we present an approach to model and adapt the mechanical regulation of morphogenesis that uses contractile cells as sculptors of engineered tissue anisotropy in vitro. Our method uses heterobifunctional cross-linkers to create mechanical boundary constraints that guide surface-directed sculpting of cell-laden extracellular matrix hydrogel constructs. Using this approach, we engineered linearly aligned tissues with structural and mechanical anisotropy. A multiscale in silico model of the sculpting process was developed to reveal that cell contractility increases as a function of principal stress polarization in anisotropic tissues. We also show that the anisotropic biophysical microenvironment of linearly aligned tissues potentiates soluble factor-mediated tenogenic and myogenic differentiation of mesenchymal stem cells. The application of our method is demonstrated by (i) skeletal muscle arrays to screen therapeutic modulators of acute oxidative injury and (ii) a 3D microphysiological model of lung cancer cachexia to study inflammatory and oxidative muscle injury induced by tumor-derived signals.
Collapse
Affiliation(s)
- Mark J Mondrinos
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Farid Alisafaei
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex Y Yi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hossein Ahmadzadeh
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Insu Lee
- Department of Mechanical Engineering, Inha University, Incheon, Korea
| | - Cassidy Blundell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeongyun Seo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Osborn
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tae-Joon Jeon
- Department of Biological Engineering, Inha University, Incheon, Korea
| | - Sun Min Kim
- Department of Mechanical Engineering, Inha University, Incheon, Korea
| | - Vivek B Shenoy
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|