1
|
Zhang W, Rao Y, Wong SH, Wu Y, Zhang Y, Yang R, Tsui SKW, Ker DFE, Mao C, Frith JE, Cao Q, Tuan RS, Wang DM. Transcriptome-Optimized Hydrogel Design of a Stem Cell Niche for Enhanced Tendon Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2313722. [PMID: 39417770 DOI: 10.1002/adma.202313722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Bioactive hydrogels have emerged as promising artificial niches for enhancing stem cell-mediated tendon repair. However, a substantial knowledge gap remains regarding the optimal combination of niche features for targeted cellular responses, which often leads to lengthy development cycles and uncontrolled healing outcomes. To address this critical gap, an innovative, data-driven materiomics strategy is developed. This approach is based on in-house RNA-seq data that integrates bioinformatics and mathematical modeling, which is a significant departure from traditional trial-and-error methods. It aims to provide both mechanistic insights and quantitative assessments and predictions of the tenogenic effects of adipose-derived stem cells induced by systematically modulated features of a tendon-mimetic hydrogel (TenoGel). The knowledge generated has enabled a rational approach for TenoGel design, addressing key considerations, such as tendon extracellular matrix concentration, uniaxial tensile loading, and in vitro pre-conditioning duration. Remarkably, our optimized TenoGel demonstrated robust tenogenesis in vitro and facilitated tendon regeneration while preventing undesired ectopic ossification in a rat tendon injury model. These findings shed light on the importance of tailoring hydrogel features for efficient tendon repair. They also highlight the tremendous potential of the innovative materiomics strategy as a powerful predictive and assessment tool in biomaterial development for regenerative medicine.
Collapse
Affiliation(s)
- Wanqi Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shing Hei Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yeung Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuanhao Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rui Yang
- Department of Sports Medicine, Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Stephen Kwok-Wing Tsui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dai Fei Elmer Ker
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jessica E Frith
- Materials Science and Engineering, Monash University, Clayton, 3800, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, 3800, VIC, Australia
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, 3800, VIC, Australia
| | - Qin Cao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rocky S Tuan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| |
Collapse
|
2
|
Muscat S, Nichols AEC. Leveraging in vivo animal models of tendon loading to inform tissue engineering approaches. Front Bioeng Biotechnol 2024; 12:1449372. [PMID: 39434716 PMCID: PMC11491380 DOI: 10.3389/fbioe.2024.1449372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Tendon injuries disrupt successful transmission of force between muscle and bone, resulting in reduced mobility, increased pain, and significantly reduced quality of life for affected patients. There are currently no targeted treatments to improve tendon healing beyond conservative methods such as rest and physical therapy. Tissue engineering approaches hold great promise for designing instructive biomaterials that could improve tendon healing or for generating replacement graft tissue. More recently, engineered microphysiological systems to model tendon injuries have been used to identify therapeutic targets. Despite these advances, current tissue engineering efforts that aim to regenerate, replace, or model injured tendons have largely failed due in large part to a lack of understanding of how the mechanical environment of the tendon influences tissue homeostasis and how altered mechanical loading can promote or prevent disease progression. This review article draws inspiration from what is known about tendon loading from in vivo animal models and identifies key metrics that can be used to benchmark success in tissue engineering applications. Finally, we highlight important challenges and opportunities for the field of tendon tissue engineering that should be taken into consideration in designing engineered platforms to understand or improve tendon healing.
Collapse
Affiliation(s)
- Samantha Muscat
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
- Department of Orthopedics and Physical Performance, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Anne E. C. Nichols
- Department of Orthopedics and Physical Performance, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
3
|
Zhao X, Cai Z, Luo Y, Lin Z, Wang J. Overexpression of CGRP receptor attenuates tendon graft degeneration in anterior cruciate ligament reconstruction. J Orthop Res 2024. [PMID: 39318262 DOI: 10.1002/jor.25978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Cell apoptosis or necrosis, extracellular matrix loss, and excessive inflammation may induce tendon graft degeneration. The impairment in the regeneration capability of nerve fibers and blood vessels may be the critical cause. Calcitonin gene-related peptide (CGRP), exhibiting a short half-life, favors cell proliferation, nerve fiber regeneration and angiogenesis. We aimed to investigate the effects of CGRP receptor-mediated signaling on tendon graft integrity and study if the modulation pathways are ascribed to cell proliferation, nerve fiber and blood vessel regeneration. A total of three groups in mice with ACL reconstruction were established: the control group (PBS treatment), the adenovirus vectors expressing CGRP receptor (CALCRL) treated group (Adv-Calcrl treatment), and the adenovirus vectors carrying shRNA targeting Calcrl treated group (Adv-shCalcrl treatment). The histological assessment indicated the Adv-Calcrl treatment was favored while the Adv-shCalcrl significantly impaired tendon graft integrity. TUNEL staining revealed a significant decreased number of apoptotic cells in the Adv-Calcrl group relative to the control group and the adv-shCalcrl group. Compared to the control group and the Adv-shCalcrl group, the Adv-Calcrl group showed significantly enhanced proliferation of nestin positive cells. Of note, the Adv-Calcrl treatment significantly increased EMCN expression at the tendon graft relative to the control and the Adv-shCalcrl groups, which may be ascribed to attenuation of the Hippo signaling pathway. Importantly, the Adv-Calcrl treatment significantly increased sensory nerve fibers and also PIEZO2 levels. Our results demonstrate the activation of CGRP receptor-mediated signaling attenuated tendon graft degeneration, which was ascribed to enhanced proliferation of Nestin positive cells, angiogenesis, and nerve fiber outgrowth.
Collapse
Affiliation(s)
- Xibang Zhao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Zhaoji Cai
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Ying Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Zhousheng Lin
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
4
|
Niu X, Melendez DL, Raj S, Cai J, Senadeera D, Mandelbaum J, Shestopalov IA, Martin SD, Zon LI, Schlaeger TM, Lai LP, McMahon AP, Craft AM, Galloway JL. A conserved transcription factor regulatory program promotes tendon fate. Dev Cell 2024:S1534-5807(24)00489-1. [PMID: 39216481 DOI: 10.1016/j.devcel.2024.08.006] [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: 04/03/2023] [Revised: 01/24/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Tendons, which transmit force from muscles to bones, are highly prone to injury. Understanding the mechanisms driving tendon fate would impact efforts to improve tendon healing, yet this knowledge is limited. To find direct regulators of tendon progenitor emergence, we performed a zebrafish high-throughput chemical screen. We established forskolin as a tenogenic inducer across vertebrates, functioning through Creb1a, which is required and sufficient for tendon fate. Putative enhancers containing cyclic AMP (cAMP) response elements (CREs) in humans, mice, and fish drove specific expression in zebrafish cranial and fin tendons. Analysis of these genomic regions identified motifs for early B cell factor (Ebf/EBF) transcription factors. Mutation of CRE or Ebf/EBF motifs significantly disrupted enhancer activity and specificity in tendons. Zebrafish ebf1a/ebf3a mutants displayed defects in tendon formation. Notably, Creb1a/CREB1 and Ebf1a/Ebf3a/EBF1 overexpression facilitated tenogenic induction in zebrafish and human pluripotent stem cells. Together, our work identifies the functional conservation of two transcription factors in promoting tendon fate.
Collapse
Affiliation(s)
- Xubo Niu
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Delmy L Melendez
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Suyash Raj
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Junming Cai
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dulanjalee Senadeera
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph Mandelbaum
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ilya A Shestopalov
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Scott D Martin
- Department of Sports Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Leonard I Zon
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Thorsten M Schlaeger
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lick Pui Lai
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - April M Craft
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
5
|
Pimentel Neto J, Batista RD, Rocha-Braga LC, Chacur M, Camargo PO, Ciena AP. The telocytes relationship with satellite cells: Extracellular vesicles mediate the myotendinous junction remodeling. Microsc Res Tech 2024; 87:1733-1741. [PMID: 38501548 DOI: 10.1002/jemt.24549] [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: 08/24/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
The peripheral nerve injury (PNI) affects the morphology of the whole locomotor apparatus, which can reach the myotendinous junction (MTJ) interface. In the injury condition, the skeletal muscle satellite cells (SC) are triggered, activated, and proliferated to repair their structure, and in the MTJ, the telocytes (TC) are associated to support the interface with the need for remodeling; in that way, these cells can be associated with SC. The study aimed to describe the SC and TC relationship after PNI at the MTJ. Sixteen adult Wistar rats were divided into Control Group (C, n = 8) and PNI Group (PNI, n = 8), PNI was performed by the constriction of the sciatic nerve. The samples were processed for transmission electron microscopy and immunostaining analysis. In the C group was evidenced the arrangement of sarcoplasmic evaginations and invaginations, the support collagen layer with a TC inside it, and an SC through vesicles internally and externally to then. In the PNI group were observed the disarrangement of invaginations and evaginations and sarcomeres degradation at MTJ, as the disposition of telopodes adjacent and in contact to the SC with extracellular vesicles and exosomes in a characterized paracrine activity. These findings can determine a link between the TCs and the SCs at the MTJ remodeling. RESEARCH HIGHLIGHTS: Peripheral nerve injury promotes the myotendinous junction (MTJ) remodeling. The telocytes (TC) and the satellite cells (SC) are present at the myotendinous interface. TC mediated the SC activity at MTJ.
Collapse
Affiliation(s)
- Jurandyr Pimentel Neto
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Rodrigo Daniel Batista
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Lara Caetano Rocha-Braga
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Marucia Chacur
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Paula Oliveira Camargo
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Adriano Polican Ciena
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| |
Collapse
|
6
|
Usami Y, Iijima H, Kokubun T. Exploring the role of mechanical forces on tendon development using in vivo model: A scoping review. Dev Dyn 2024; 253:550-565. [PMID: 37947268 DOI: 10.1002/dvdy.673] [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: 03/14/2023] [Revised: 08/25/2023] [Accepted: 09/27/2023] [Indexed: 11/12/2023] Open
Abstract
Tendons transmit the muscle contraction forces to bones and drive joint movement throughout life. While extensive research have indicated the essentiality of mechanical forces on tendon development, a comprehensive understanding of the fundamental role of mechanical forces still needs to be impaerted. This scoping review aimed to summarize the current knowledge about the role of mechanical forces during the tendon developmental phase. The electronic database search using PubMed, performed in May 2023, yielded 651 articles, of which 16 met the prespecified inclusion criteria. We summarized and divided the methods to reduce the mechanical force into three groups: loss of muscle, muscle dysfunction, and weight-bearing regulation. In contrast, there were few studies to analyze the increased mechanical force model. Most studies suggested that mechanical force has some roles in tendon development in the embryo to postnatal phase. However, we identified species variability and methodological heterogeneity to modulate mechanical force. To establish a comprehensive understanding, methodological commonality to modulate the mechanical force is needed in this field. Additionally, summarizing chronological changes in developmental processes across animal species helps to understand the essence of developmental tendon mechanobiology. We expect that the findings summarized in the current review serve as a groundwork for future study in the fields of tendon developmantal biology and mechanobiology.
Collapse
Affiliation(s)
- Yuna Usami
- Graduate School of Health, Medicine, and Welfare, Saitama Prefectural University, Koshigaya, Japan
| | - Hirotaka Iijima
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, Massachusetts, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
| | - Takanori Kokubun
- Graduate School of Health, Medicine, and Welfare, Saitama Prefectural University, Koshigaya, Japan
- Department of Physical Therapy, School of Health and Social Services, Saitama Prefectural University, Koshigaya, Japan
| |
Collapse
|
7
|
Callaway DA, Penkala IJ, Zhou S, Knowlton JJ, Cardenas-Diaz F, Babu A, Morley MP, Lopes M, Garcia BA, Morrisey EE. TGF-β controls alveolar type 1 epithelial cell plasticity and alveolar matrisome gene transcription in mice. J Clin Invest 2024; 134:e172095. [PMID: 38488000 PMCID: PMC10947970 DOI: 10.1172/jci172095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 01/05/2024] [Indexed: 03/19/2024] Open
Abstract
Premature birth disrupts normal lung development and places infants at risk for bronchopulmonary dysplasia (BPD), a disease disrupting lung health throughout the life of an individual and that is increasing in incidence. The TGF-β superfamily has been implicated in BPD pathogenesis, however, what cell lineage it impacts remains unclear. We show that TGFbr2 is critical for alveolar epithelial (AT1) cell fate maintenance and function. Loss of TGFbr2 in AT1 cells during late lung development leads to AT1-AT2 cell reprogramming and altered pulmonary architecture, which persists into adulthood. Restriction of fetal lung stretch and associated AT1 cell spreading through a model of oligohydramnios enhances AT1-AT2 reprogramming. Transcriptomic and proteomic analyses reveal the necessity of TGFbr2 expression in AT1 cells for extracellular matrix production. Moreover, TGF-β signaling regulates integrin transcription to alter AT1 cell morphology, which further impacts ECM expression through changes in mechanotransduction. These data reveal the cell intrinsic necessity of TGF-β signaling in maintaining AT1 cell fate and reveal this cell lineage as a major orchestrator of the alveolar matrisome.
Collapse
Affiliation(s)
- Danielle A. Callaway
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute
| | - Ian J. Penkala
- Penn-CHOP Lung Biology Institute
- Department of Cell and Developmental Biology, and
| | - Su Zhou
- Penn-CHOP Lung Biology Institute
- Department of Cell and Developmental Biology, and
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan J. Knowlton
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute
| | - Fabian Cardenas-Diaz
- Penn-CHOP Lung Biology Institute
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Apoorva Babu
- Penn-CHOP Lung Biology Institute
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael P. Morley
- Penn-CHOP Lung Biology Institute
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mariana Lopes
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benjamin A. Garcia
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward E. Morrisey
- Penn-CHOP Lung Biology Institute
- Department of Cell and Developmental Biology, and
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Kviatkovsky SA, Hickner RC, Cabre HE, Small SD, Ormsbee MJ. Collagen peptides supplementation improves function, pain, and physical and mental outcomes in active adults. J Int Soc Sports Nutr 2023; 20:2243252. [PMID: 37551682 PMCID: PMC10411303 DOI: 10.1080/15502783.2023.2243252] [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: 02/22/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023] Open
Abstract
INTRODUCTION Chronic pain affects 19% of adults in the United States, with increasing prevalence in active and aging populations. Pain can limit physical activity and activities of daily living (ADLs), resulting in declined mental and social health. Nutritional interventions for pain currently target inflammation or joint health, but few influence both. Collagen, the most abundant protein in the human body and constituent of the extra cellular matrix, is such a nutraceutical. While there have been reports of reductions in pain with short-term collagen peptide (CP) supplementation, there are no long-term studies specifically in healthy middle-aged active adults. PURPOSE To determine the effects of daily CP consumption over 3, 6, and 9 months on survey measures of pain, function, and physical and mental health using The Knee Injury & Osteoarthritis Outcomes Score (KOOS) and Veterans Rand 12 (VR-12) in middle-aged active adults. METHODS This study was a double-blind randomized control trial with three treatment groups (Placebo, 10 g/d CP, and 20 g/d CP). RESULTS Improvements in ADLs (p = .031, ηp2 = .096) and pain (p = .037, ηp2 = .164) were observed with 10 g/d CP over 6 months, although pain only improved in high frequency exercisers (>180 min/week). Additionally, VR-12 mental component scores (MCS) improved with 10 g/d of CP over 3-9 months (p = .017, ηp2 = .309), while physical component scores (PCS) improved with 20 g/d of CP over 3-9 months, but only in females (p = .013, ηp2= .582). CONCLUSION These findings suggest 10 to 20 g/d of CP supplementation over 6 to 9 months may improve ADLs, pain, MCS, and PCS in middle-aged active adults.
Collapse
Affiliation(s)
- Shiloah A. Kviatkovsky
- Florida State University, Department of Nutrition and Integrative Physiology, Tallahassee, FL, USA
- Florida State University, Institute of Sports Sciences and Medicine, Tallahassee, FL, USA
- University of Arkansas for Medical Sciences, Center for Aging and Longevity, Geriatrics, Little Rock, AR, USA
| | - Robert C. Hickner
- Florida State University, Department of Nutrition and Integrative Physiology, Tallahassee, FL, USA
- Florida State University, Institute of Sports Sciences and Medicine, Tallahassee, FL, USA
- University of KwaZulu-Natal, School of Health Sciences, Discipline of Biokinetics, Exercise and Leisure Sciences, Durban, South Africa
| | - Hannah E. Cabre
- The University of North Carolina at Chapel Hill, Applied Physiology Lab, Department of Sport Science, Chapel Hill, NC, USA
| | - Stephanie D. Small
- University of Toronto, Faculty of Kinesiology & Physical Education, Toronto, ON, Canada
| | - Michael J. Ormsbee
- Florida State University, Department of Nutrition and Integrative Physiology, Tallahassee, FL, USA
- Florida State University, Institute of Sports Sciences and Medicine, Tallahassee, FL, USA
- University of KwaZulu-Natal, School of Health Sciences, Discipline of Biokinetics, Exercise and Leisure Sciences, Durban, South Africa
| |
Collapse
|
9
|
Rajan AM, Rosin NL, Labit E, Biernaskie J, Liao S, Huang P. Single-cell analysis reveals distinct fibroblast plasticity during tenocyte regeneration in zebrafish. SCIENCE ADVANCES 2023; 9:eadi5771. [PMID: 37967180 PMCID: PMC10651129 DOI: 10.1126/sciadv.adi5771] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
Despite their importance in tissue maintenance and repair, fibroblast diversity and plasticity remain poorly understood. Using single-cell RNA sequencing, we uncover distinct sclerotome-derived fibroblast populations in zebrafish, including progenitor-like perivascular/interstitial fibroblasts, and specialized fibroblasts such as tenocytes. To determine fibroblast plasticity in vivo, we develop a laser-induced tendon ablation and regeneration model. Lineage tracing reveals that laser-ablated tenocytes are quickly regenerated by preexisting fibroblasts. By combining single-cell clonal analysis and live imaging, we demonstrate that perivascular/interstitial fibroblasts actively migrate to the injury site, where they proliferate and give rise to new tenocytes. By contrast, perivascular fibroblast-derived pericytes or specialized fibroblasts, including tenocytes, exhibit no regenerative plasticity. Active Hedgehog (Hh) signaling is required for the proliferation of activated fibroblasts to ensure efficient tenocyte regeneration. Together, our work highlights the functional diversity of fibroblasts and establishes perivascular/interstitial fibroblasts as tenocyte progenitors that promote tendon regeneration in a Hh signaling-dependent manner.
Collapse
Affiliation(s)
- Arsheen M. Rajan
- Department of Biochemistry and Molecular Biology, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Nicole L. Rosin
- Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Elodie Labit
- Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shan Liao
- Inflammation Research Network, Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
10
|
Kwan KYC, Ng KWK, Rao Y, Zhu C, Qi S, Tuan RS, Ker DFE, Wang DM. Effect of Aging on Tendon Biology, Biomechanics and Implications for Treatment Approaches. Int J Mol Sci 2023; 24:15183. [PMID: 37894875 PMCID: PMC10607611 DOI: 10.3390/ijms242015183] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/07/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Tendon aging is associated with an increasing prevalence of tendon injuries and/or chronic tendon diseases, such as tendinopathy, which affects approximately 25% of the adult population. Aged tendons are often characterized by a reduction in the number and functionality of tendon stem/progenitor cells (TSPCs), fragmented or disorganized collagen bundles, and an increased deposition of glycosaminoglycans (GAGs), leading to pain, inflammation, and impaired mobility. Although the exact pathology is unknown, overuse and microtrauma from aging are thought to be major causative factors. Due to the hypovascular and hypocellular nature of the tendon microenvironment, healing of aged tendons and related injuries is difficult using current pain/inflammation and surgical management techniques. Therefore, there is a need for novel therapies, specifically cellular therapy such as cell rejuvenation, due to the decreased regenerative capacity during aging. To augment the therapeutic strategies for treating tendon-aging-associated diseases and injuries, a comprehensive understanding of tendon aging pathology is needed. This review summarizes age-related tendon changes, including cell behaviors, extracellular matrix (ECM) composition, biomechanical properties and healing capacity. Additionally, the impact of conventional treatments (diet, exercise, and surgery) is discussed, and recent advanced strategies (cell rejuvenation) are highlighted to address aged tendon healing. This review underscores the molecular and cellular linkages between aged tendon biomechanical properties and the healing response, and provides an overview of current and novel strategies for treating aged tendons. Understanding the underlying rationale for future basic and translational studies of tendon aging is crucial to the development of advanced therapeutics for tendon regeneration.
Collapse
Affiliation(s)
- Ka Yu Carissa Kwan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ka Wai Kerry Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chenxian Zhu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shengcai Qi
- Department of Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200040, China;
| | - Rocky S. Tuan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dai Fei Elmer Ker
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.Y.C.K.); (K.W.K.N.); (Y.R.); (C.Z.); (R.S.T.); (D.F.E.K.)
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
11
|
Marvin JC, Brakewood ME, Poon MLS, Andarawis-Puri N. Regenerative MRL/MpJ tendon cells exhibit sex differences in morphology, proliferation, mechanosensitivity, and cell-ECM organization. J Orthop Res 2023; 41:2273-2286. [PMID: 37004178 DOI: 10.1002/jor.25562] [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: 09/16/2022] [Revised: 02/10/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023]
Abstract
Clinical and animal studies have reported the influence of sex on the incidence and progression of tendinopathy, which results in disparate structural and biomechanical outcomes. However, there remains a paucity in our understanding of the sex-specific biological mechanisms underlying effective tendon healing. To overcome this hurdle, our group has investigated the impact of sex on tendon regeneration using the super-healer Murphy Roths Large (MRL/MpJ) mouse strain. We have previously shown that the scarless healing capacity of MRL/MpJ patellar tendons is associated with sexually dimorphic regulation of gene expression for pathways involved in fibrosis, cell migration, adhesion, and extracellular matrix (ECM) remodeling following an acute mid-substance injury. Thus, we hypothesized that MRL/MpJ scarless tendon healing is mediated by sex-specific and temporally distinct orchestration of cell-ECM interactions. Accordingly, the present study comparatively evaluated MRL/MpJ tendon cells on two-dimensional (2D; glass) and scaffold platforms to examine cell behavior under biochemical and topographical cues associated with tendon homeostasis and healing. Female MRL/MpJ cells showed reduced 2D migration and spreading area accompanied by enhanced mechanosensing, ECM alignment, and fibronectin-mediated cell proliferation compared to male MRL/MpJ cells. Interestingly, female MRL/MpJ cells cultured on isotropic scaffolds showed diminished cell-ECM organization compared to male MRL/MpJ cells. Lastly, MRL/MpJ cells elicited enhanced cytoskeletal elongation and alignment, ECM deposition and organization, and connexin 43-mediated intercellular communication compared to male B6 cells, regardless of culture condition or sex. These results provide insight into the cellular features conserved within the MRL/MpJ phenotype and potential sex-specific targets for the development of more equitable therapeutics.
Collapse
Affiliation(s)
- Jason C Marvin
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Molly E Brakewood
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Mong L S Poon
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| | - Nelly Andarawis-Puri
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
- Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
12
|
Huang AH, Galloway JL. Current and emerging technologies for defining and validating tendon cell fate. J Orthop Res 2023; 41:2082-2092. [PMID: 37211925 DOI: 10.1002/jor.25632] [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: 01/13/2023] [Revised: 05/09/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023]
Abstract
The tendon field has been flourishing in recent years with the advent of new tools and model systems. The recent ORS 2022 Tendon Section Conference brought together researchers from diverse disciplines and backgrounds, showcasing studies in biomechanics and tissue engineering to cell and developmental biology and using models from zebrafish and mouse to humans. This perspective aims to summarize progress in tendon research as it pertains to understanding and studying tendon cell fate. The successful integration of new technologies and approaches have the potential to further propel tendon research into a new renaissance of discovery. However, there are also limitations with the current methodologies that are important to consider when tackling research questions. Altogether, we will highlight recent advances and technologies and propose new avenues to explore tendon biology.
Collapse
Affiliation(s)
- Alice H Huang
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
| | - Jenna L Galloway
- Department of Orthopaedic Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
13
|
Tsai SL, Villaseñor S, Shah RR, Galloway JL. Endogenous tenocyte activation underlies the regenerative capacity of the adult zebrafish tendon. NPJ Regen Med 2023; 8:52. [PMID: 37726307 PMCID: PMC10509205 DOI: 10.1038/s41536-023-00328-w] [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: 01/09/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023] Open
Abstract
Tendons are essential, frequently injured connective tissues that transmit forces from muscle to bone. Their unique highly ordered, matrix-rich structure is critical for proper function. While adult mammalian tendons heal after acute injuries, endogenous tendon cells, or tenocytes, fail to respond appropriately, resulting in the formation of disorganized fibrovascular scar tissue with impaired function and increased propensity for re-injury. Here, we show that, unlike mammals, adult zebrafish tenocytes activate upon injury and fully regenerate the tendon. Using a full tear injury model in the adult zebrafish craniofacial tendon, we defined the hallmark stages and cellular basis of tendon regeneration through multiphoton imaging, lineage tracing, and transmission electron microscopy approaches. Remarkably, we observe that zebrafish tendons regenerate and restore normal collagen matrix ultrastructure by 6 months post-injury (mpi). Tendon regeneration progresses in three main phases: inflammation within 24 h post-injury (hpi), cellular proliferation and formation of a cellular bridge between the severed tendon ends at 3-5 days post-injury (dpi), and re-differentiation and matrix remodeling beginning from 5 dpi to 6 mpi. Importantly, we demonstrate that pre-existing tenocytes are the main cellular source of regeneration, proliferating and migrating upon injury to ultimately bridge the tendon ends. Finally, we show that TGF-β signaling is required for tenocyte recruitment and bridge formation. Collectively, our work debuts and aptly positions the adult zebrafish tendon as an invaluable comparative system to elucidate regenerative mechanisms that may inspire new therapeutic strategies.
Collapse
Affiliation(s)
- Stephanie L Tsai
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steffany Villaseñor
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rishita R Shah
- Department of Biology, Barnard College, New York, NY, USA
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
14
|
Zhang T, Wan L, Xiao H, Wang L, Hu J, Lu H. Single-cell RNA sequencing reveals cellular and molecular heterogeneity in fibrocartilaginous enthesis formation. eLife 2023; 12:e85873. [PMID: 37698466 PMCID: PMC10513478 DOI: 10.7554/elife.85873] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 09/10/2023] [Indexed: 09/13/2023] Open
Abstract
The attachment site of the rotator cuff (RC) is a classic fibrocartilaginous enthesis, which is the junction between bone and tendon with typical characteristics of a fibrocartilage transition zone. Enthesis development has historically been studied with lineage tracing of individual genes selected a priori, which does not allow for the determination of single-cell landscapes yielding mature cell types and tissues. Here, in together with open-source GSE182997 datasets (three samples) provided by Fang et al., we applied Single-cell RNA sequencing (scRNA-seq) to delineate the comprehensive postnatal RC enthesis growth and the temporal atlas from as early as postnatal day 1 up to postnatal week 8. And, we furtherly performed single-cell spatial transcriptomic sequencing on postnatal day 1 mouse enthesis, in order to deconvolute bone-tendon junction (BTJ) chondrocytes onto spatial spots. In summary, we deciphered the cellular heterogeneity and the molecular dynamics during fibrocartilage differentiation. Combined with current spatial transcriptomic data, our results provide a transcriptional resource that will support future investigations of enthesis development at the mechanistic level and may shed light on the strategies for enhanced RC healing outcomes.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Liyang Wan
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Han Xiao
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Linfeng Wang
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Jianzhong Hu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South UniversityChangshaChina
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital Central South UniversityChangshaChina
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan ProvinceChangshaChina
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
15
|
Zhao B, Lv Y. A biomechanical view of epigenetic tumor regulation. J Biol Phys 2023; 49:283-307. [PMID: 37004697 PMCID: PMC10397176 DOI: 10.1007/s10867-023-09633-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/12/2023] [Indexed: 04/04/2023] Open
Abstract
The occurrence and development of tumors depend on a complex regulation by not only biochemical cues, but also biomechanical factors in tumor microenvironment. With the development of epigenetic theory, the regulation of biomechanical stimulation on tumor progress genetically is not enough to fully illustrate the mechanism of tumorigenesis. However, biomechanical regulation on tumor progress epigenetically is still in its infancy. Therefore, it is particularly important to integrate the existing relevant researches and develop the potential exploration. This work sorted out the existing researches on the regulation of tumor by biomechanical factors through epigenetic means, which contains summarizing the tumor epigenetic regulatory mode by biomechanical factors, exhibiting the influence of epigenetic regulation under mechanical stimulation, illustrating its existing applications, and prospecting the potential. This review aims to display the relevant knowledge through integrating the existing studies on epigenetic regulation in tumorigenesis under mechanical stimulation so as to provide theoretical basis and new ideas for potential follow-up research and clinical applications. Mechanical factors under physiological conditions stimulate the tumor progress through epigenetic ways, and new strategies are expected to be found with the development of epidrugs and related delivery systems.
Collapse
Affiliation(s)
- Boyuan Zhao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, No. 1 Sunshine Avenue, Jiangxia District, Wuhan, Hubei Province, 430200, People's Republic of China.
| |
Collapse
|
16
|
Subramanian A, Kanzaki LF, Schilling TF. Mechanical force regulates Sox9 expression at the developing enthesis. Development 2023; 150:dev201141. [PMID: 37497608 PMCID: PMC10445799 DOI: 10.1242/dev.201141] [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: 07/15/2022] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
Entheses transmit force from tendons and ligaments to the skeleton. Regional organization of enthesis extracellular matrix (ECM) generates differences in stiffness required for force transmission. Two key transcription factors co-expressed in entheseal tenocytes, scleraxis (Scx) and Sox9, directly control production of enthesis ECM components. Formation of embryonic craniofacial entheses in zebrafish coincides with onset of jaw movements, possibly in response to the force of muscle contraction. We show dynamic changes in scxa and sox9a mRNA levels in subsets of entheseal tenocytes that correlate with their roles in force transmission. We also show that transcription of a direct target of Scxa, Col1a, in enthesis ECM is regulated by the ratio of scxa to sox9a expression. Eliminating muscle contraction by paralyzing embryos during early stages of musculoskeletal differentiation alters relative levels of scxa and sox9a in entheses, primarily owing to increased sox9a expression. Force-dependent TGF-β (TGFβ) signaling is required to maintain this balance of scxa and sox9a expression. Thus, force from muscle contraction helps establish a balance of transcription factor expression that controls specialized ECM organization at the tendon enthesis and its ability to transmit force.
Collapse
Affiliation(s)
- Arul Subramanian
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Lauren F. Kanzaki
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
17
|
Li H, Korcari A, Ciufo D, Mendias CL, Rodeo SA, Buckley MR, Loiselle AE, Pitt GS, Cao C. Increased Ca 2+ signaling through Ca V 1.2 induces tendon hypertrophy with increased collagen fibrillogenesis and biomechanical properties. FASEB J 2023; 37:e23007. [PMID: 37261735 PMCID: PMC10254118 DOI: 10.1096/fj.202300607r] [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: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 06/02/2023]
Abstract
Tendons are tension-bearing tissues transmitting force from muscle to bone for body movement. This mechanical loading is essential for tendon development, homeostasis, and healing after injury. While Ca2+ signaling has been studied extensively for its roles in mechanotransduction, regulating muscle, bone, and cartilage development and homeostasis, knowledge about Ca2+ signaling and the source of Ca2+ signals in tendon fibroblast biology are largely unknown. Here, we investigated the function of Ca2+ signaling through CaV 1.2 voltage-gated Ca2+ channel in tendon formation. Using a reporter mouse, we found that CaV 1.2 is highly expressed in tendon during development and downregulated in adult homeostasis. To assess its function, we generated ScxCre;CaV 1.2TS mice that express a gain-of-function mutant CaV 1.2 in tendon. We found that mutant tendons were hypertrophic, with more tendon fibroblasts but decreased cell density. TEM analyses demonstrated increased collagen fibrillogenesis in the hypertrophic tendons. Biomechanical testing revealed that the hypertrophic tendons display higher peak load and stiffness, with no changes in peak stress and elastic modulus. Proteomic analysis showed no significant difference in the abundance of type I and III collagens, but mutant tendons had about two-fold increase in other ECM proteins such as tenascin C, tenomodulin, periostin, type XIV and type VIII collagens, around 11-fold increase in the growth factor myostatin, and significant elevation of matrix remodeling proteins including Mmp14, Mmp2, and cathepsin K. Taken together, these data highlight roles for increased Ca2+ signaling through CaV 1.2 on regulating expression of myostatin growth factor and ECM proteins for tendon collagen fibrillogenesis during tendon formation.
Collapse
Affiliation(s)
- Haiyin Li
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | - Antonion Korcari
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - David Ciufo
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Scott A. Rodeo
- Sports Medicine and Shoulder Service, Hospital for Special Surgery, New York, NY, USA
| | - Mark R. Buckley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Alayna E. Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Chike Cao
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopeadics, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
18
|
Dickinson AJG. Jak2 and Jaw Muscles Are Required for Buccopharyngeal Membrane Perforation during Mouth Development. J Dev Biol 2023; 11:24. [PMID: 37367478 DOI: 10.3390/jdb11020024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/23/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
The mouth is a central feature of our face, without which we could not eat, breathe, or communicate. A critical and early event in mouth formation is the creation of a "hole" which connects the digestive system and the external environment. This hole, which has also been called the primary or embryonic mouth in vertebrates, is initially covered by a 1-2 cell layer thick structure called the buccopharyngeal membrane. When the buccopharyngeal membrane does not rupture, it impairs early mouth functions and may also lead to further craniofacial malformations. Using a chemical screen in an animal model (Xenopus laevis) and genetic data from humans, we determined that Janus kinase 2 (Jak2) has a role in buccopharyngeal membrane rupture. We have determined that decreased Jak2 function, using antisense morpholinos or a pharmacological antagonist, caused a persistent buccopharyngeal membrane as well as the loss of jaw muscles. Surprisingly, we observed that the jaw muscle compartments were connected to the oral epithelium that is continuous with the buccopharyngeal membrane. Severing such connections resulted in buccopharyngeal membrane buckling and persistence. We also noted puncta accumulation of F-actin, an indicator of tension, in the buccopharyngeal membrane during perforation. Taken together, the data has led us to a hypothesis that muscles are required to exert tension across the buccopharyngeal membrane, and such tension is necessary for its perforation.
Collapse
Affiliation(s)
- Amanda J G Dickinson
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
19
|
Callaway DA, Penkala IJ, Zhou S, Cardenas-Diaz F, Babu A, Morley MP, Lopes M, Garcia BA, Morrisey EE. TGFβ controls alveolar type 1 epithelial cell plasticity and alveolar matrisome gene transcription. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.540035. [PMID: 37214932 PMCID: PMC10197675 DOI: 10.1101/2023.05.09.540035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Premature birth disrupts normal lung development and places infants at risk for bronchopulmonary dysplasia (BPD), a disease increasing in incidence which disrupts lung health throughout the lifespan. The TGFβ superfamily has been implicated in BPD pathogenesis, however, what cell lineage it impacts remains unclear. We show that Tgfbr2 is critical for AT1 cell fate maintenance and function. Loss of Tgfbr2 in AT1 cells during late lung development leads to AT1-AT2 cell reprogramming and altered pulmonary architecture, which persists into adulthood. Restriction of fetal lung stretch and associated AT1 cell spreading through a model of oligohydramnios enhances AT1-AT2 reprogramming. Transcriptomic and proteomic analysis reveal the necessity of Tgfbr2 expression in AT1 cells for extracellular matrix production. Moreover, TGFβ signaling regulates integrin transcription to alter AT1 cell morphology, which further impacts ECM expression through changes in mechanotransduction. These data reveal the cell intrinsic necessity of TGFβ signaling in maintaining AT1 cell fate and reveal this cell lineage as a major orchestrator of the alveolar matrisome.
Collapse
|
20
|
Jones CL, Penney BT, Theodossiou SK. Engineering Cell-ECM-Material Interactions for Musculoskeletal Regeneration. Bioengineering (Basel) 2023; 10:bioengineering10040453. [PMID: 37106640 PMCID: PMC10135874 DOI: 10.3390/bioengineering10040453] [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: 01/28/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular microenvironment regulates many of the mechanical and biochemical cues that direct musculoskeletal development and are involved in musculoskeletal disease. The extracellular matrix (ECM) is a main component of this microenvironment. Tissue engineered approaches towards regenerating muscle, cartilage, tendon, and bone target the ECM because it supplies critical signals for regenerating musculoskeletal tissues. Engineered ECM-material scaffolds that mimic key mechanical and biochemical components of the ECM are of particular interest in musculoskeletal tissue engineering. Such materials are biocompatible, can be fabricated to have desirable mechanical and biochemical properties, and can be further chemically or genetically modified to support cell differentiation or halt degenerative disease progression. In this review, we survey how engineered approaches using natural and ECM-derived materials and scaffold systems can harness the unique characteristics of the ECM to support musculoskeletal tissue regeneration, with a focus on skeletal muscle, cartilage, tendon, and bone. We summarize the strengths of current approaches and look towards a future of materials and culture systems with engineered and highly tailored cell-ECM-material interactions to drive musculoskeletal tissue restoration. The works highlighted in this review strongly support the continued exploration of ECM and other engineered materials as tools to control cell fate and make large-scale musculoskeletal regeneration a reality.
Collapse
Affiliation(s)
- Calvin L Jones
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| | - Brian T Penney
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| | - Sophia K Theodossiou
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| |
Collapse
|
21
|
Ma RC, Kocha KM, Méndez-Olivos EE, Ruel TD, Huang P. Origin and diversification of fibroblasts from the sclerotome in zebrafish. Dev Biol 2023; 498:35-48. [PMID: 36933633 DOI: 10.1016/j.ydbio.2023.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Fibroblasts play an important role in maintaining tissue integrity by secreting components of the extracellular matrix and initiating response to injury. Although the function of fibroblasts has been extensively studied in adults, the embryonic origin and diversification of different fibroblast subtypes during development remain largely unexplored. Using zebrafish as a model, we show that the sclerotome, a sub-compartment of the somite, is the embryonic source of multiple fibroblast subtypes including tenocytes (tendon fibroblasts), blood vessel associated fibroblasts, fin mesenchymal cells, and interstitial fibroblasts. High-resolution imaging shows that different fibroblast subtypes occupy unique anatomical locations with distinct morphologies. Long-term Cre-mediated lineage tracing reveals that the sclerotome also contributes to cells closely associated with the axial skeleton. Ablation of sclerotome progenitors results in extensive skeletal defects. Using photoconversion-based cell lineage analysis, we find that sclerotome progenitors at different dorsal-ventral and anterior-posterior positions display distinct differentiation potentials. Single-cell clonal analysis combined with in vivo imaging suggests that the sclerotome mostly contains unipotent and bipotent progenitors prior to cell migration, and the fate of their daughter cells is biased by their migration paths and relative positions. Together, our work demonstrates that the sclerotome is the embryonic source of trunk fibroblasts as well as the axial skeleton, and local signals likely contribute to the diversification of distinct fibroblast subtypes.
Collapse
Affiliation(s)
- Roger C Ma
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - Katrinka M Kocha
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - Emilio E Méndez-Olivos
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - Tyler D Ruel
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
22
|
Leyhr J, Sanchez S, Dollman KN, Tafforeau P, Haitina T. Enhanced contrast synchrotron X-ray microtomography for describing skeleton-associated soft tissue defects in zebrafish mutants. Front Endocrinol (Lausanne) 2023; 14:1108916. [PMID: 36950679 PMCID: PMC10025580 DOI: 10.3389/fendo.2023.1108916] [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: 11/26/2022] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
Detailed histological analyses are desirable for zebrafish mutants that are models for human skeletal diseases, but traditional histological techniques are limited to two-dimensional thin sections with orientations highly dependent on careful sample preparation. On the other hand, techniques that provide three-dimensional (3D) datasets including µCT scanning are typically limited to visualizing the bony skeleton and lack histological resolution. We combined diffusible iodine-based contrast enhancement (DICE) and propagation phase-contrast synchrotron radiation micro-computed tomography (PPC-SRµCT) to image late larval and juvenile zebrafish, obtaining high-quality 3D virtual histology datasets of the mineralized skeleton and surrounding soft tissues. To demonstrate this technique, we used virtual histological thin sections and 3D segmentation to qualitatively and quantitatively compare wild-type zebrafish and nkx3.2 -/- mutants to characterize novel soft-tissue phenotypes in the muscles and tendons of the jaw and ligaments of the Weberian apparatus, as well as the sinus perilymphaticus associated with the inner ear. We could observe disrupted fiber organization and tendons of the adductor mandibulae and protractor hyoideus muscles associated with the jaws, and show that despite this, the overall muscle volumes appeared unaffected. Ligaments associated with the malformed Weberian ossicles were mostly absent in nkx3.2 -/- mutants, and the sinus perilymphaticus was severely constricted or absent as a result of the fused exoccipital and basioccipital elements. These soft-tissue phenotypes have implications for the physiology of nkx3.2 -/- zebrafish, and demonstrate the promise of DICE-PPC-SRµCT for histopathological investigations of bone-associated soft tissues in small-fish skeletal disease models and developmental studies more broadly.
Collapse
Affiliation(s)
- Jake Leyhr
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Sophie Sanchez
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
- European Synchrotron Radiation Facility, Grenoble, France
| | | | - Paul Tafforeau
- European Synchrotron Radiation Facility, Grenoble, France
| | - Tatjana Haitina
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Lipp SN, Jacobson KR, Colling HA, Tuttle TG, Miles DT, McCreery KP, Calve S. Mechanical loading is required for initiation of extracellular matrix deposition at the developing murine myotendinous junction. Matrix Biol 2023; 116:28-48. [PMID: 36709857 PMCID: PMC10218368 DOI: 10.1016/j.matbio.2023.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
The myotendinous junction (MTJ) contributes to the generation of motion by connecting muscle to tendon. At the adult MTJ, a specialized extracellular matrix (ECM) is thought to contribute to the mechanical integrity of the muscle-tendon interface, but the factors that influence MTJ formation during mammalian development are unclear. Here, we combined 3D imaging and proteomics with murine models in which muscle contractility and patterning are disrupted to resolve morphological and compositional changes in the ECM during MTJ development. We found that MTJ-specific ECM deposition can be initiated via static loading due to growth; however, it required cyclic loading to develop a mature morphology. Furthermore, the MTJ can mature without the tendon terminating into cartilage. Based on these results, we describe a model wherein MTJ development depends on mechanical loading but not insertion into an enthesis.
Collapse
Affiliation(s)
- Sarah N Lipp
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907, United States; The Indiana University Medical Scientist/Engineer Training Program, Indianapolis, IN 46202, United States
| | - Kathryn R Jacobson
- Purdue University Interdisciplinary Life Science Program, 155 S. Grant Street, West Lafayette, IN 47907, United States
| | - Haley A Colling
- Department of Integrative Physiology, University of Colorado Boulder, 354 UCB, Boulder CO, 80309, United States
| | - Tyler G Tuttle
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO 80309, United States
| | - Dalton T Miles
- Chemical and Biological Engineering, University of Colorado Boulder, 596 UCB, CO 80309, United States
| | - Kaitlin P McCreery
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO 80309, United States
| | - Sarah Calve
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907, United States; Purdue University Interdisciplinary Life Science Program, 155 S. Grant Street, West Lafayette, IN 47907, United States; Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO 80309, United States.
| |
Collapse
|
24
|
Li H, Korcari A, Ciufo D, Mendias CL, Rodeo SA, Buckley MR, Loiselle AE, Pitt GS, Cao C. Increased Ca 2+ signaling through Ca V 1.2 induces tendon hypertrophy with increased collagen fibrillogenesis and biomechanical properties. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525119. [PMID: 36747837 PMCID: PMC9900778 DOI: 10.1101/2023.01.24.525119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Tendons are tension-bearing tissues transmitting force from muscle to bone for body movement. This mechanical loading is essential for tendon development, homeostasis, and healing after injury. While Ca 2+ signaling has been studied extensively for its roles in mechanotransduction, regulating muscle, bone and cartilage development and homeostasis, knowledge about Ca 2+ signaling and the source of Ca 2+ signals in tendon fibroblast biology are largely unknown. Here, we investigated the function of Ca 2+ signaling through Ca V 1.2 voltage-gated Ca 2+ channel in tendon formation. Using a reporter mouse, we found that Ca V 1.2 is highly expressed in tendon during development and downregulated in adult homeostasis. To assess its function, we generated ScxCre;Ca V 1.2 TS mice that express a gain-of-function mutant Ca V 1.2 channel (Ca V 1.2 TS ) in tendon. We found that tendons in the mutant mice were approximately 2/3 larger and had more tendon fibroblasts, but the cell density of the mutant mice decreased by around 22%. TEM analyses demonstrated increased collagen fibrillogenesis in the hypertrophic tendon. Biomechanical testing revealed that the hypertrophic Achilles tendons display higher peak load and stiffness, with no changes in peak stress and elastic modulus. Proteomics analysis reveals no significant difference in the abundance of major extracellular matrix (ECM) type I and III collagens, but mutant mice had about 2-fold increase in other ECM proteins such as tenascin C, tenomodulin, periostin, type XIV and type VIII collagens, around 11-fold increase in the growth factor of TGF-β family myostatin, and significant elevation of matrix remodeling proteins including Mmp14, Mmp2 and cathepsin K. Taken together, these data highlight roles for increased Ca 2+ signaling through Ca V 1.2 on regulating expression of myostatin growth factor and ECM proteins for tendon collagen fibrillogenesis during tendon formation.
Collapse
|
25
|
Mechanochemistry of collagen. Acta Biomater 2023; 163:50-62. [PMID: 36669548 DOI: 10.1016/j.actbio.2023.01.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/02/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023]
Abstract
The collagen molecular family is the result of nearly one billion years of evolution. It is a unique family of proteins, the majority of which provide general mechanical support to biological tissues. Fibril forming collagens are the most abundant collagens in vertebrate animals and are generally found in positions that resist tensile loading. In animals, cells produce fibril-forming collagen molecules that self-assemble into larger structures known as collagen fibrils. Collagen fibrils are the fundamental, continuous, load-bearing elements in connective tissues, but are often further aggregated into larger load-bearing structures, fascicles in tendon, lamellae in cornea and in intervertebral disk. We know that failure to form fibrillar collagen is embryonic lethal, and excessive collagen formation/growth (fibrosis) or uncontrolled enzymatic remodeling (type II collagen: osteoarthritis) is pathological. Collagen is thus critical to vertebrate viability and instrumental in maintaining efficient mechanical structures. However, despite decades of research, our understanding of collagen matrix formation is not complete, and we know still less about the detailed mechanisms that drive collagen remodeling, growth, and pathology. In this perspective, we examine the known role of mechanical force on the formation and development of collagenous structure. We then discuss a mechanochemical mechanism that has the potential to unify our understanding of collagenous tissue assembly dynamics, which preferentially deposits and grows collagen fibrils directly in the path of mechanical force, where the energetics should be dissuasive and where collagen fibrils are most required. We term this mechanism: Mechanochemical force-structure causality. STATEMENT OF SIGNIFICANCE: Our mechanochemical-force structure causality postulate suggests that collagen molecules are components of mechanochemically-sensitive and dynamically-responsive fibrils. Collagen molecules assemble preferentially in the path of applied strain, can be grown in place by mechanical extension, and are retained in the path of force through strain-stabilization. The mechanisms that drive this behavior operate at the level of the molecules themselves and are encoded into the structure of the biomaterial. The concept might change our understanding of structure formation, enhance our ability to treat injuries, and accelerate the development of therapeutics to prevent pathologies such as fibrosis. We suggest that collagen is a mechanochemically responsive dynamic element designed to provide a substantial "material assist" in the construction of adaptive carriers of mechanical signals.
Collapse
|
26
|
Lu K, Zhou M, Wang L, Wang Y, Tang H, He G, Wang H, Tang C, He J, Wang W, Tang K, Wang Y, Deng Z. N-Acetyl-L-cysteine facilitates tendon repair and promotes the tenogenic differentiation of tendon stem/progenitor cells by enhancing the integrin α5/β1/PI3K/AKT signaling. BMC Mol Cell Biol 2023; 24:1. [PMID: 36604630 DOI: 10.1186/s12860-022-00463-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Tendon injury is associated with oxidative stress, leading to reactive oxygen species (ROS) production and inflammation. N-acetyl-L-cysteine (NAC) is a potent antioxidant. However, how NAC affects the biological functions of tendon stem/progenitor cells (TSPCs) and tendon repair has not been clarified. METHOD: The impacts of NAC on the viability, ROS production, and differentiation of TSPCs were determined with the cell counting kit-8, fluorescence staining, Western blotting, and immunofluorescence. The effect of NAC on gene transcription in TSPCs was analyzed by transcriptomes and bioinformatics and validated by Western blotting. The potential therapeutic effect of NAC on tendon repair was tested in a rat model of Achilles tendon injury. RESULTS Compared with the untreated control, treatment with 500 µM NAC greatly promoted the proliferation of TSPCs and significantly mitigated hydrogen peroxide-induced ROS production and cytotoxicity in vitro. NAC treatment significantly increased the relative protein expression of collagen type 1 alpha 1 (COL1A1), tenascin C (TNC), scleraxis (SCX), and tenomodulin (TNMD) in TPSCs. Bioinformatics analyses revealed that NAC modulated transcriptomes, particularly in the integrin-related phosphoinositide 3-kinase (PI3K)/AKT signaling, and Western blotting revealed that NAC enhanced integrin α5β1 expression and PI3K/AKT activation in TSPCs. Finally, NAC treatment mitigated the tendon injury, but enhanced the protein expression of SCX, TNC, TNMD, and COLIA1 in the injured tissue regions of the rats. CONCLUSION NAC treatment promoted the survival and differentiation of TSPCs to facilitate tendon repair after tendon injury in rats. Thus, NAC may be valuable for the treatment of tendon injury.
Collapse
Affiliation(s)
- Kang Lu
- Department of Orthopedics-Spine Surgery Center, the Second Affiliated Hospital, Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, China
| | - Mei Zhou
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, No. 29, Yanzheng Street, Gaotan, Shapingba District, Chongqing, China
| | - Liyuan Wang
- Department of Orthopedics-Spine Surgery Center, the Second Affiliated Hospital, Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, China
| | - Yang Wang
- Department of Orthopedics-Spine Surgery Center, the Second Affiliated Hospital, Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, China
| | - Hong Tang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, No. 29, Yanzheng Street, Gaotan, Shapingba District, Chongqing, China
| | - Gang He
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, No. 29, Yanzheng Street, Gaotan, Shapingba District, Chongqing, China
| | - Huan Wang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, No. 29, Yanzheng Street, Gaotan, Shapingba District, Chongqing, China
| | - Chuyue Tang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, No. 29, Yanzheng Street, Gaotan, Shapingba District, Chongqing, China
| | - Jie He
- Department of Orthopedics-Spine Surgery Center, the Second Affiliated Hospital, Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, China
| | - Wei Wang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, No. 29, Yanzheng Street, Gaotan, Shapingba District, Chongqing, China
| | - Kanglai Tang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, No. 29, Yanzheng Street, Gaotan, Shapingba District, Chongqing, China.
| | - Yunjiao Wang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, No. 29, Yanzheng Street, Gaotan, Shapingba District, Chongqing, China.
| | - Zhongliang Deng
- Department of Orthopedics-Spine Surgery Center, the Second Affiliated Hospital, Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
27
|
Peserico A, Barboni B, Russo V, Bernabò N, El Khatib M, Prencipe G, Cerveró-Varona A, Haidar-Montes AA, Faydaver M, Citeroni MR, Berardinelli P, Mauro A. Mammal comparative tendon biology: advances in regulatory mechanisms through a computational modeling. Front Vet Sci 2023; 10:1175346. [PMID: 37180059 PMCID: PMC10174257 DOI: 10.3389/fvets.2023.1175346] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023] Open
Abstract
There is high clinical demand for the resolution of tendinopathies, which affect mainly adult individuals and animals. Tendon damage resolution during the adult lifetime is not as effective as in earlier stages where complete restoration of tendon structure and property occurs. However, the molecular mechanisms underlying tendon regeneration remain unknown, limiting the development of targeted therapies. The research aim was to draw a comparative map of molecules that control tenogenesis and to exploit systems biology to model their signaling cascades and physiological paths. Using current literature data on molecular interactions in early tendon development, species-specific data collections were created. Then, computational analysis was used to construct Tendon NETworks in which information flow and molecular links were traced, prioritized, and enriched. Species-specific Tendon NETworks generated a data-driven computational framework based on three operative levels and a stage-dependent set of molecules and interactions (embryo-fetal or prepubertal) responsible, respectively, for signaling differentiation and morphogenesis, shaping tendon transcriptional program and downstream modeling of its fibrillogenesis toward a mature tissue. The computational network enrichment unveiled a more complex hierarchical organization of molecule interactions assigning a central role to neuro and endocrine axes which are novel and only partially explored systems for tenogenesis. Overall, this study emphasizes the value of system biology in linking the currently available disjointed molecular data, by establishing the direction and priority of signaling flows. Simultaneously, computational enrichment was critical in revealing new nodes and pathways to watch out for in promoting biomedical advances in tendon healing and developing targeted therapeutic strategies to improve current clinical interventions.
Collapse
|
28
|
Luderman LN, Michaels MT, Levic DS, Knapik EW. Zebrafish Erc1b mediates motor innervation and organization of craniofacial muscles in control of jaw movement. Dev Dyn 2023; 252:104-123. [PMID: 35708710 DOI: 10.1002/dvdy.511] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Movement of the lower jaw, a common behavior observed among vertebrates, is required for eating and processing food. This movement is controlled by signals sent from the trigeminal motor nerve through neuromuscular junctions (NMJs) to the masticatory muscles. Dysfunctional jaw movements contribute to craniomandibular disorders, yet the pathophysiology of these disorders is not well understood, as limited studies have been conducted on the molecular mechanisms of jaw movement. RESULTS Using erc1b/kimm533 genetic loss of function mutant, we evaluated lower jaw muscle organization and innervation by the cranial motor nerves in developing zebrafish. Using time-lapse confocal imaging of the erc1b mutant in a transgenic fluorescent reporter line, we found delayed trigeminal nerve growth and disrupted nerve branching architecture during muscle innervation. By automated 3D image analysis of NMJ distribution, we identified an increased number of small, disorganized NMJ clusters in erc1b mutant larvae compared to WT siblings. Using genetic replacement experiments, we determined the Rab GTPase binding domain of Erc1b is required for cranial motor nerve branching, but not NMJ organization or muscle attachment. CONCLUSIONS We identified Erc1b/ERC1 as a novel component of a genetic pathway contributing to muscle organization, trigeminal nerve outgrowth, and NMJ spatial distribution during development that is required for jaw movement.
Collapse
Affiliation(s)
- Lauryn N Luderman
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mackenzie T Michaels
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Daniel S Levic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
- Neuroscience Graduate Program, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Ela W Knapik
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
29
|
Tendon-Specific Activation of Tenogenic Transcription Factors Enables Keeping Tenocytes' Identity In Vitro. Int J Mol Sci 2022; 23:ijms232214078. [PMID: 36430562 PMCID: PMC9695818 DOI: 10.3390/ijms232214078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
We generated a novel tetracycline-inducible transgenic mouse line with the tendon-specific expression of a series of tendon-critical transcription factors. Primary tenocytes derived from this mouse line consistently expressed green fluorescent protein reporter transcription factors in response to doxycycline. The tenocytes maintained their tendon cell properties for a longer time after the transient induction in the absence of growth factors and mechanical stress. Four key transcription factors for tendon development and the green fluorescent protein reporter were linked with different viral 2A self-cleaving peptides. They were expressed under the control of the tet-responsive element. In combination with the expression of BFP, which reports on the tendon-specific collagen I, and mScarlet, which reports on the tendon-specific transcription factor Scleraxis (Scx), we observed the more extended maintenance of the tendon cell identity of in vitro cultured tendon cells and Achilles tendon explants. This means that the Scleraxis bHLH transcription factor (Scx), mohawk homeobox (Mkx), early growth response 1 (Egr1) and early growth response 2 (Egr2) contributed to the maintenance of tenocytes' identity in vitro, providing a new model for studying extracellular matrix alterations and identifying alternative biomaterials in vitro.
Collapse
|
30
|
The Influence of Different Modes of Exercise on Healthy and Injured Tendons. Stem Cells Int 2022; 2022:3945210. [PMID: 36117720 PMCID: PMC9481386 DOI: 10.1155/2022/3945210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/03/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Tendons are essential components of the musculoskeletal system that links the skeletal muscle to the skeleton. This dense connective tissue exhibits great plasticity. Therefore, research on the influence of types of exercise, including acute and long-term training, on the structural and mechanical properties of tendons in athletic and sedentary populations is of critical importance in the design of scientific-based exercise plans and effective tendinopathy treatment. Here, we review recent studies on the relationship between exercise and tendon health and tendinopathy repair to provide a general understanding of how exercise may reshape tendons.
Collapse
|
31
|
Korntner SH, Jana A, Kinnard E, Leo E, Beane T, Li X, Sengupta R, Becker L, Kuo CK. Craniofacial tendon development—Characterization of extracellular matrix morphology and spatiotemporal protein distribution. Front Cell Dev Biol 2022; 10:944126. [PMID: 36158210 PMCID: PMC9490420 DOI: 10.3389/fcell.2022.944126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Craniofacial (CF) tendons are often affected by traumatic injuries and painful disorders that can severely compromise critical jaw functions, such as mastication and talking. Unfortunately, tendons lack the ability to regenerate, and there are no solutions to restore their native properties or function. An understanding of jaw tendon development could inform tendon regeneration strategies to restore jaw function, however CF tendon development has been relatively unexplored. Using the chick embryo, we identified the jaw-closing Tendon of the musculus Adductor Mandibulae Externus (TmAM) and the jaw-opening Tendon of the musculus Depressor Mandibulae (TmDM) that have similar functions to the masticatory tendons in humans. Using histological and immunohistochemical (IHC) analyses, we characterized the TmAM and TmDM on the basis of cell and extracellular matrix (ECM) morphology and spatiotemporal protein distribution from early to late embryonic development. The TmAM and TmDM were detectable as early as embryonic day (d) 9 based on histological staining and tenascin-C (TNC) protein distribution. Collagen content increased and became more organized, cell density decreased, and cell nuclei elongated over time during development in both the TmAM and TmDM. The TmAM and TmDM exhibited similar spatiotemporal patterns for collagen type III (COL3), but differential spatiotemporal patterns for TNC, lysyl oxidase (LOX), and matrix metalloproteinases (MMPs). Our results demonstrate markers that play a role in limb tendon formation are also present in jaw tendons during embryonic development, implicate COL3, TNC, LOX, MMP2, and MMP9 in jaw tendon development, and suggest TmAM and TmDM possess different developmental programs. Taken together, our study suggests the chick embryo may be used as a model with which to study CF tendon extracellular matrix development, the results of which could ultimately inform therapeutic approaches for CF tendon injuries and disorders.
Collapse
Affiliation(s)
- Stefanie H. Korntner
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Aniket Jana
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Elizabeth Kinnard
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Emily Leo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Timothy Beane
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Xianmu Li
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Rohit Sengupta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Lauren Becker
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Catherine K. Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Maryland Medical Center, Baltimore, MD, United States
- *Correspondence: Catherine K. Kuo,
| |
Collapse
|
32
|
Zhang JL, Richetti S, Ramezani T, Welcker D, Lütke S, Pogoda HM, Hatzold J, Zaucke F, Keene DR, Bloch W, Sengle G, Hammerschmidt M. Vertebrate extracellular matrix protein hemicentin-1 interacts physically and genetically with basement membrane protein nidogen-2. Matrix Biol 2022; 112:132-154. [PMID: 36007682 PMCID: PMC10015821 DOI: 10.1016/j.matbio.2022.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/19/2022]
Abstract
Hemicentins are large proteins of the extracellular matrix that belong to the fibulin family and play pivotal roles during development and homeostasis of a variety of invertebrate and vertebrate tissues. However, bona fide interaction partners of hemicentins have not been described as yet. Here, applying surface plasmon resonance spectroscopy and co-immunoprecipitation, we identify the basement membrane protein nidogen-2 (NID2) as a binding partner of mouse and zebrafish hemicentin-1 (HMCN1), in line with the formerly described essential role of mouse HMCN1 in basement membrane integrity. We show that HMCN1 binds to the same protein domain of NID2 (G2) as formerly shown for laminins, but with an approximately 3.5-fold lower affinity and in a competitive manner. Furthermore, immunofluorescence and immunogold labeling revealed that HMCN1/Hmcn1 is localized close to basement membranes and in partial overlap with NID2/Nid2a in different tissues of mouse and zebrafish. Genetic knockout and antisense-mediated knockdown studies in zebrafish further show that loss of Nid2a leads to similar defects in fin fold morphogenesis as the loss of Laminin-α5 (Lama5) or Hmcn1. Finally, combined partial loss-of-function studies indicated that nid2a genetically interacts with both hmcn1 and lama5. Together, these findings suggest that despite their mutually exclusive physical binding, hemicentins, nidogens, and laminins tightly cooperate and support each other during formation, maintenance, and function of basement membranes to confer tissue linkage.
Collapse
Affiliation(s)
- Jin-Li Zhang
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Stefania Richetti
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Thomas Ramezani
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Daniela Welcker
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Steffen Lütke
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hans-Martin Pogoda
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Julia Hatzold
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Frank Zaucke
- Research Unit for Osteoarthritis, Department for Orthopedics, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Douglas R Keene
- Micro-Imaging Center, Shriners Hospital for Children, Portland, OR, United States
| | - Wilhelm Bloch
- Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cologne Center for Musculoskeletal Biomechanics (CCMB), University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Matthias Hammerschmidt
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
33
|
Differential biological responses of adherent and non-adherent (cancer and non-cancerous) cells to variable extremely low frequency magnetic fields. Sci Rep 2022; 12:14225. [PMID: 35987807 PMCID: PMC9392794 DOI: 10.1038/s41598-022-18210-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 08/08/2022] [Indexed: 11/08/2022] Open
Abstract
Extremely low-frequency electromagnetic field (ELF-EMF) induces biological effects on different cells through various signaling pathways. To study the impact of the ELF-EMF on living cells under an optimal physiological condition, we have designed and constructed a novel system that eliminates several limitations of other ELF-EMF systems. Apoptosis and cell number were assessed by flow cytometry and the Trypan Blue dye exclusion method, respectively. In vitro cell survival was evaluated by colony formation assay. The distribution of cells in the cell cycle, intracellular ROS level, and autophagy were analyzed by flow cytometer. Suspended cells differentiation was assessed by phagocytosis of latex particles and NBT reduction assay. Our results showed that response to the exposure to ELF-EMF is specific and depends on the biological state of the cell. For DU145, HUVEC, and K562 cell lines the optimum results were obtained at the frequency of 0.01 Hz, while for MDA-MB-231, the optimum response was obtained at 1 Hz. Long-term exposure to ELF-EMF in adherent cells effectively inhibited proliferation by arresting the cell population at the cell cycle G2/M phase and increased intracellular ROS level, leading to morphological changes and cell death. The K562 cells exposed to the ELF-EMF differentiate via induction of autophagy and decreasing the cell number. Our novel ELF-EMF instrument could change morphological and cell behaviors, including proliferation, differentiation, and cell death.
Collapse
|
34
|
Noriega-González DC, Drobnic F, Caballero-García A, Roche E, Perez-Valdecantos D, Córdova A. Effect of Vitamin C on Tendinopathy Recovery: A Scoping Review. Nutrients 2022; 14:2663. [PMID: 35807843 PMCID: PMC9267994 DOI: 10.3390/nu14132663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Tendinopathies represent 30-50% of all sports injuries. The tendon response is influenced by the load (volume, intensity, and frequency) that the tendon support, resulting in irritability and pain, among others. The main molecular component of tendons is collagen I (60-85%). The rest consist of glycosaminoglycans-proteoglycans, glycoproteins, and other collagen subtypes. This study's aim was to critically evaluate the efficacy of vitamin C supplementation in the treatment of tendinopathies. At the same time, the study aims to determine the optimal conditions (dose and time) for vitamin C supplementation. A structured search was carried out in the SCOPUS, Medline (PubMed), and Web of Science (WOS) databases. The inclusion criteria took into account studies describing optimal tendon recovery when using vitamin C alone or in combination with other compounds. The study design was considered, including randomized, double-blind controlled, and parallel designs in animal models or humans. The main outcome is that vitamin C supplementation is potentially useful as a therapeutic approach for tendinopathy recovery. Vitamin C supplementation, alone or in combination with other products, increases collagen synthesis with a consequent improvement in the patient's condition. On the other hand, vitamin C deficiency is mainly associated with a decrease in procollagen synthesis and reduced hydroxylation of proline and lysine residues, hindering the tendon repair process.
Collapse
Affiliation(s)
- David C. Noriega-González
- Department of Surgery, Ophthalmology, Otorhinolaryngology and Physiotherapy, Faculty of Medicine, Hospital Clínico Universitario de Valladolid, 47002 Valladolid, Spain;
| | | | - Alberto Caballero-García
- Department of Anatomy and Radiology, Faculty of Health Sciences, GIR Physical Exercise and Aging, Campus Los Pajaritos, University of Valladolid, 42004 Soria, Spain;
| | - Enrique Roche
- Department of Applied Biology-Nutrition, Institute of Bioengineering, University Miguel Hernández, 03202 Elche, Spain;
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Daniel Perez-Valdecantos
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Health Sciences, GIR Physical Exercise and Aging, Campus Duques de Soria, University of Valladolid, 42004 Soria, Spain;
| | - Alfredo Córdova
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Health Sciences, GIR Physical Exercise and Aging, Campus Duques de Soria, University of Valladolid, 42004 Soria, Spain;
| |
Collapse
|
35
|
Lu J, Jiang L, Chen Y, Lyu K, Zhu B, Li Y, Liu X, Liu X, Long L, Wang X, Xu H, Wang D, Li S. The Functions and Mechanisms of Basic Fibroblast Growth Factor in Tendon Repair. Front Physiol 2022; 13:852795. [PMID: 35770188 PMCID: PMC9234302 DOI: 10.3389/fphys.2022.852795] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Tendon injury is a disorder of the musculoskeletal system caused by overuse or trauma, which is characterized by pain and limitations in joint function. Since tendon healing is slowly and various treatments are generally ineffective, it remains a clinically challenging problem. Recent evidences suggest that basic fibroblast growth factor (bFGF) not only plays an important role in tendon healing, but also shows a positive effect in laboratory experimentations. The purpose of this review is to summarize the effects of bFGF in the tendon healing. Firstly, during the inflammatory phase, bFGF stimulates the proliferation and differentiation of vascular endothelial cells to foster neovascularization. Furthermore, bFGF enhances the production of pro-inflammatory factors during the early phase of tendon healing, thereby accelerating the inflammatory response. Secondly, the cell proliferation phase is accompanied by the synthesis of a large number of extracellular matrix components. bFGF speeds up tendon healing by stimulating fibroblasts to secrete type III collagen. Lastly, the remodeling phase is characterized by the transition from type III collagen to type I collagen, which can be promoted by bFGF. However, excessive injection of bFGF can cause tendon adhesions as well as scar tissue formation. In future studies, we need to explore further applications of bFGF in the tendon healing process.
Collapse
Affiliation(s)
- Jingwei Lu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Li Jiang
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Yixuan Chen
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Kexin Lyu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Bin Zhu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Yujie Li
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Xueli Liu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Xinyue Liu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Longhai Long
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaoqiang Wang
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Houping Xu
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- *Correspondence: Houping Xu, ; Dingxuan Wang, ; Sen Li,
| | - Dingxuan Wang
- School of Physical Education, Southwest Medical University, Luzhou, China
- *Correspondence: Houping Xu, ; Dingxuan Wang, ; Sen Li,
| | - Sen Li
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- *Correspondence: Houping Xu, ; Dingxuan Wang, ; Sen Li,
| |
Collapse
|
36
|
Nakai K, Tanaka H, Fukuzawa K, Nakajima J, Ozaki M, Kato N, Kawato T. Effects of Electric-Toothbrush Vibrations on the Expression of Collagen and Non-Collagen Proteins through the Focal Adhesion Kinase Signaling Pathway in Gingival Fibroblasts. Biomolecules 2022; 12:biom12060771. [PMID: 35740896 PMCID: PMC9221308 DOI: 10.3390/biom12060771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 11/27/2022] Open
Abstract
Electric-toothbrush vibrations, which remove plaque, are transmitted to the gingival connective tissue via epithelial cells. Physical energy affects cell function; however, the effects of electric-toothbrush vibrations on gingival extracellular matrix (ECM) protein expression remain unknown. We aimed to examine the effects of these vibrations on the expression of ECM proteins—type I collagen (col I), type III collagen (col III), elastin, and fibronectin (FN)—using human gingival fibroblasts (HGnFs). HGnFs were seeded for 5 days in a six-well plate with a hydrophilic surface, exposed to electric-toothbrush vibrations, and cultured for 7 days. Subsequently, the mRNA and protein levels of col I, col III, elastin, and FN were examined. To investigate the role of focal adhesion kinase (FAK) signaling on ECM protein expression in vibration-stimulated cells, the cells were treated with siRNA against protein tyrosine kinase (PTK). Electric-toothbrush vibrations increased col I, col III, elastin, and FN expression; promoted collagen and non-collagen protein production; and enhanced FAK phosphorylation in HGnFs. Moreover, PTK2 siRNA completely blocked the effects of these vibrations on the expression of col I, col III and elastin mRNA. The results suggest that electric-toothbrush vibrations increase collagen, elastin, and FN production through the FAK-signaling pathway in fibroblasts.
Collapse
Affiliation(s)
- Kumiko Nakai
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo 101-8310, Japan; (H.T.); (K.F.); (M.O.); (N.K.); (T.K.)
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo 101-8310, Japan
- Correspondence: ; Tel.: +81-3-3219-8128
| | - Hideki Tanaka
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo 101-8310, Japan; (H.T.); (K.F.); (M.O.); (N.K.); (T.K.)
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| | - Kyoko Fukuzawa
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo 101-8310, Japan; (H.T.); (K.F.); (M.O.); (N.K.); (T.K.)
| | - Jyunya Nakajima
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry, Tokyo 101-8310, Japan;
| | - Manami Ozaki
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo 101-8310, Japan; (H.T.); (K.F.); (M.O.); (N.K.); (T.K.)
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| | - Nobue Kato
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo 101-8310, Japan; (H.T.); (K.F.); (M.O.); (N.K.); (T.K.)
| | - Takayuki Kawato
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo 101-8310, Japan; (H.T.); (K.F.); (M.O.); (N.K.); (T.K.)
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| |
Collapse
|
37
|
Tsuchiya Y, Bayer ML, Schjerling P, Soendenbroe C, Kjaer M. CRediT author statement (Author contributions)Yoshifumi Tsuchiya: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Resources, Writing – original draft, Visualization, Supervision, Project administration, Funding acquisition. Monika Lucia Bayer: Investigation, Resources. Peter Schjerling: Investigation, Writing – review & editing. . Casper Soendenbroe: Validation, Writing – review & editing. Michael Kjaer: Writing – review & editing, Supervision, Project administration, Funding acquisition acquisition.Human derived tendon cells contribute to myotube formation in vitro. Exp Cell Res 2022; 417:113164. [DOI: 10.1016/j.yexcr.2022.113164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 11/04/2022]
|
38
|
Earp JE, Gesick H, Angelino D, Adami A. Effects of isometric loading intensity on patellar tendon microvascular response. Scand J Med Sci Sports 2022; 32:1182-1191. [PMID: 35485297 PMCID: PMC9283377 DOI: 10.1111/sms.14175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/07/2022] [Accepted: 04/19/2022] [Indexed: 11/30/2022]
Abstract
Acute increases in tendon blood flow and oxygenation after stress (i.e., hyperemic response) can enhance tendon recovery. While loading intensity is a fundamental part of resistance training programs, its effects on tendon's hyperemic response are unknown. This study aimed to compare acute changes in total (total hemoglobin [THb]) and oxygenated hemoglobin (HbO2 ) concentrations in the patellar tendon after isometric exercise at different intensities. Thirteen participants performed 8 (5 s) isometric knee extensions at 25%, 50%, and 75% maximal load (maximal voluntarily isometric contraction [MVIC]), separated by 20 min recovery, prescribed in randomized and counterbalanced order. Changes in patellar tendon THb, HbO2 and deoxygenated hemoglobin (HHb) in response to exercise at each intensity were measured using near-infrared spectroscopy. Post-exercise, HbO2 increased with 50% ( η p 2 $$ {\eta}_p^2 $$ = 0.305, f = 5.26, p < 0.01) and 75% ( η p 2 $$ {\eta}_p^2 $$ = 0.245, f = 4.56, p < 0.01) but not 25% ( η p 2 $$ {\eta}_p^2 $$ = 0.088, f = 1.16, p = 0.339) MVIC, while THb increased in 50% ( η p 2 $$ {\eta}_p^2 $$ = 0.305, f = 5.26, p = 0.01) but not 25% ( η p 2 $$ {\eta}_p^2 $$ = 0.067, f = 0.865, p = 0.51) or 75% ( η p 2 $$ {\eta}_p^2 $$ = 0.126, f = 1.729, p = 0.14) MVIC. Additionally, increasing load from 25% to 50% MVIC resulted in greater THb (f = 2.459, p = 0.43), HbO2 (f = 3.389, p = 0.13) and HHb (f = 0.320, p = 0.01) post-exercise responses, but no differences were observed between 50% and 75% MVIC (THb: f = 0.748, p = 0.59; HbO2 : f = 0.825, p = 0.54; HHb: f = 0.713, p = 0.62). Our results suggest there is a loading threshold at ~50% MVIC at which the tendon hyperemic response is fully achieved. Training above this intensity is not expected to provide any additional change to the tendon microvascular response. Therefore, moderate loading seems to be sufficient to fully elicit the patellar tendon hyperemic response that's believed to stimulate tendon healing.
Collapse
Affiliation(s)
- Jacob E Earp
- Department of Kinesiology, University of Connecticut, Storrs, Connecticut, USA.,Department of Kinesiology, University of Rhode Island, Kingston, Rhode Island, USA
| | - Haley Gesick
- Department of Kinesiology, University of Connecticut, Storrs, Connecticut, USA
| | - Domenic Angelino
- Department of Kinesiology, University of Rhode Island, Kingston, Rhode Island, USA
| | - Alessandra Adami
- Department of Kinesiology, University of Rhode Island, Kingston, Rhode Island, USA
| |
Collapse
|
39
|
Fan C, Zhao Y, Chen Y, Qin T, Lin J, Han S, Yan R, Lei T, Xie Y, Wang T, Gu S, Ouyang H, Shen W, Yin Z, Chen X. A Cd9 +Cd271 + stem/progenitor population and the SHP2 pathway contribute to neonatal-to-adult switching that regulates tendon maturation. Cell Rep 2022; 39:110762. [PMID: 35476985 DOI: 10.1016/j.celrep.2022.110762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/06/2022] [Accepted: 04/08/2022] [Indexed: 11/03/2022] Open
Abstract
Tendon maturation lays the foundation for postnatal tendon development, its proper mechanical function, and regeneration, but the critical cell populations and the entangled mechanisms remain poorly understood. Here, by integrating the structural, mechanical, and molecular properties, we show that post-natal days 7-14 are the crucial transitional stage for mouse tendon maturation. We decode the cellular and molecular regulatory networks at the single-cell level. We find that a nerve growth factor (NGF)-secreting Cd9+Cd271+ tendon stem/progenitor cell population mainly prompts conversion from neonate to adult tendon. Through single-cell gene regulatory network analysis, in vitro inhibitor identification, and in vivo tendon-specific Shp2 deletion, we find that SHP2 signaling is a regulator for tendon maturation. Our research comprehensively reveals the dynamic cell population transition during tendon maturation, implementing insights into the critical roles of the maturation-related stem cell population and SHP2 signaling pathway during tendon differentiation and regeneration.
Collapse
Affiliation(s)
- Chunmei Fan
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Yanyan Zhao
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Yangwu Chen
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Tian Qin
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Junxin Lin
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Shan Han
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong SAR, China
| | - Ruojin Yan
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Tingyun Lei
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Yuanhao Xie
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Tingzhang Wang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Shen Gu
- School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Weiliang Shen
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| | - Xiao Chen
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
40
|
Park S, Kim H, Wang Y, Eom DS, Allard J. Zebrafish airinemes optimize their shape between ballistic and diffusive search. eLife 2022; 11:75690. [PMID: 35467525 PMCID: PMC9098217 DOI: 10.7554/elife.75690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
In addition to diffusive signals, cells in tissue also communicate via long, thin cellular protrusions, such as airinemes in zebrafish. Before establishing communication, cellular protrusions must find their target cell. Here we demonstrate that the shapes of airinemes in zebrafish are consistent with a finite persistent random walk model. The probability of contacting the target cell is maximized for a balance between ballistic search (straight) and diffusive search (highly curved, random). We find that the curvature of airinemes in zebrafish, extracted from live cell microscopy, is approximately the same value as the optimum in the simple persistent random walk model. We also explore the ability of the target cell to infer direction of the airineme's source, finding that there is a theoretical trade-off between search optimality and directional information. This provides a framework to characterize the shape, and performance objectives, of non-canonical cellular protrusions in general.
Collapse
Affiliation(s)
- Sohyeon Park
- Center for Complex Biological Systems, University of California, Irvine, Irvine, United States
| | - Hyunjoong Kim
- Department of Mathematics, University of Pennsylvania, Pennsylvania, United States
| | - Yi Wang
- Center for Complex Biological Systems, University of California, Irvine, Irvine, United States
| | - Dae Seok Eom
- Center for Complex Biological Systems, University of California, Irvine, Irvine, United States
| | - Jun Allard
- dDepartment of Physics and Astronomy, University of California, Irvine, Irvine, United States
| |
Collapse
|
41
|
Growth and mechanobiology of the tendon-bone enthesis. Semin Cell Dev Biol 2022; 123:64-73. [PMID: 34362655 PMCID: PMC8810906 DOI: 10.1016/j.semcdb.2021.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022]
Abstract
Tendons are cable-like connective tissues that transfer both active and passive forces generated by skeletal muscle to bone. In the mature skeleton, the tendon-bone enthesis is an interfacial zone of transitional tissue located between two mechanically dissimilar tissues: compliant, fibrous tendon to rigid, dense mineralized bone. In this review, we focus on emerging areas in enthesis development related to its structure, function, and mechanobiology, as well as highlight established and emerging signaling pathways and physiological processes that influence the formation and adaptation of this important transitional tissue.
Collapse
|
42
|
Yin Z, Sun L, Shi L, Nie H, Dai J, Zhang C. Bioinspired bimodal micro-nanofibrous scaffolds promote the tenogenic differentiation of tendon stem/progenitor cells for achilles tendon regeneration. Biomater Sci 2022; 10:753-769. [PMID: 34985056 DOI: 10.1039/d1bm01287h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Poor tendon repair remains a clinical problem due to the difficulties in replicating the complex multiscale hierarchical structure of native tendons. In this work, a bioinspired fibrous scaffold with bimodal micro-nanofibers and a teno-inductive aligned topography was developed to replicate microscale collagen fibers and nanoscale collagen fibrils that compose native tendons. The results showed indicated that the combination of micro- and nanofibers enhanced the mechanical properties. Furthermore, their biological performance was assessed using tendon stem/progenitor cells (TSPCs). Micro-nanofibers induced a higher cell aspect ratio and enhanced the tenogenic differentiation of TSPCs compared to micro- and nanocontrols. Interestingly, it was observed that scaffold nanotopography and microstructures promoted tenogenesis via activating the TGF-β/Smad2/3-mediated signaling pathway. The in situ implantation study confirmed that micro-nanofibrous scaffolds promoted the structural and mechanical properties of the regenerated Achilles tendon. Overall, our study shows that the bimodal micro-nanofibrous scaffold developed here presents a promising potential to improve the outcomes of tendon tissue engineering.
Collapse
Affiliation(s)
- Zhiwei Yin
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China.
| | - Lu Sun
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China.
| | - Liyang Shi
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China.
| | - Hemin Nie
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China.
| | - Jianwu Dai
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China. .,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Can Zhang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China.
| |
Collapse
|
43
|
Hoyle DJ, Dranow DB, Schilling TF. Pthlha and mechanical force control early patterning of growth zones in the zebrafish craniofacial skeleton. Development 2022; 149:dev199826. [PMID: 34919126 PMCID: PMC8917414 DOI: 10.1242/dev.199826] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022]
Abstract
Secreted signals in patterning systems often induce repressive signals that shape their distributions in space and time. In developing growth plates (GPs) of endochondral long bones, Parathyroid hormone-like hormone (Pthlh) inhibits Indian hedgehog (Ihh) to form a negative-feedback loop that controls GP progression and bone size. Whether similar systems operate in other bones and how they arise during embryogenesis remain unclear. We show that Pthlha expression in the zebrafish craniofacial skeleton precedes chondrocyte differentiation and restricts where cells undergo hypertrophy, thereby initiating a future GP. Loss of Pthlha leads to an expansion of cells expressing a novel early marker of the hypertrophic zone (HZ), entpd5a, and later HZ markers, such as ihha, whereas local Pthlha misexpression induces ectopic entpd5a expression. Formation of this early pre-HZ correlates with onset of muscle contraction and requires mechanical force; paralysis leads to loss of entpd5a and ihha expression in the pre-HZ, mislocalized pthlha expression and no subsequent ossification. These results suggest that local Pthlh sources combined with force determine HZ locations, establishing the negative-feedback loop that later maintains GPs.
Collapse
Affiliation(s)
| | | | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92693, USA
| |
Collapse
|
44
|
Viganò M, Lugano G, Orfei CP, Menon A, Ragni E, Colombini A, de Luca P, Talò G, Randelli PS, de Girolamo L. Tendon Cells Derived From The Long Head Of The Biceps And The Supraspinatus Tendons Of Patients Affected By Rotator Cuff Tears Show Different Expression Of Inflammatory Markers. Connect Tissue Res 2021; 62:570-579. [PMID: 32921180 DOI: 10.1080/03008207.2020.1816993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIM OF THE STUDY Tendons are exposed to mechanical stress constantly during movements and thus they are frequently subjected to injuries. Rotator cuff tears are common musculoskeletal disorders, mainly involving the supraspinatus tendon. The characterization of the tenocytes derived from this tendon and the comparison to cells isolated from the long head of the biceps tendon obtained from donors affected by rotator cuff disease may improve the knowledge of the cellular mechanisms involved in the initiation and progression of the pathology. Thus, the aim of the present study was to characterize and compare donor-matched human tendon cells (TCs) isolated from the long head of the biceps (LHB-TCs) and the supraspinatus tendons (SSP-TCs) of patients affected by rotator cuff tears. METHODS donor-matched LHB-TCs and SSP-TCs were isolated and cultured up to passage 3. Phenotypic appearance, metabolic activity, DNA content, production of soluble mediators (IL-1Ra, IL-1β, IL-6, and VEGF) and gene expression of tendon markers (SCX, COL1A1, COL3A1), inflammatory (PTGS2), and catabolic enzymes (MMP-1, MMP-3) were evaluated. RESULTS LHB-TCs showed an elongated fibroblast-like shape, while SSP-TCs appeared irregular with jagged membrane. SSP-TCs gene expression revealed an augmented production of PTGS2, a marker of inflammation, whereas they produced a reduced amount of IL-6, in respect to LHB-TCs. CONCLUSION SSP-TCs showed higher cellular stress and expression of inflammatory markers with respect to donor-matched LHB-TCs, suggesting that addressing the physio-pathological state of supraspinatus tendon cells during treatment of rotator cuff tears could favor tissue healing and possibly prevent relapses.
Collapse
Affiliation(s)
- Marco Viganò
- Orthopedics Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Gaia Lugano
- Orthopedics Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | | | - Alessandra Menon
- Laboratory of Applied Biomechanics, Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy.,U.O.C. 1° Clinica Ortopedica, ASST Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan, Italy.,Research Center for Adult and Pediatric Rheumatic Diseases (RECAP-RD), Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - Enrico Ragni
- Orthopedics Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | | | - Paola de Luca
- Orthopedics Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Giuseppe Talò
- Orthopedics Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Pietro S Randelli
- Laboratory of Applied Biomechanics, Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy.,U.O.C. 1° Clinica Ortopedica, ASST Centro Specialistico Ortopedico Traumatologico Gaetano Pini-CTO, Milan, Italy.,Research Center for Adult and Pediatric Rheumatic Diseases (RECAP-RD), Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - Laura de Girolamo
- Orthopedics Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
45
|
Paredes J, Pekmezian A, Andarawis-Puri N. MRL/MpJ tendon matrix-derived therapeutic promotes improved healing outcomes in scar-mediated canonical tendon healing. J Orthop Res 2021; 39:1548-1560. [PMID: 32441819 PMCID: PMC7680300 DOI: 10.1002/jor.24754] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/10/2020] [Accepted: 05/08/2020] [Indexed: 02/04/2023]
Abstract
Tendons are commonly injured connective soft tissues characterized by an ineffective healing response that results in scar formation and loss of functional and structural properties. Naturally occurring extracellular matrix (ECM) constructs have become a promising therapeutic for tendon injuries due to their capacity to harness a complex biological environment. However, in tendon, the ECM properties needed for improved healing remain unknown. Interestingly, we have determined that the improved tendon healing response of the scarless-healing MRL/MpJ is driven by intrinsic properties with therapeutic potential to modulate the proliferative and morphological behavior of cells derived from a canonically healing model in vitro. We hypothesize that a distinct composition of ECM deposited during the early healing response of the MRL/MpJ will harnesses the biological cues to stimulate improved structure and function in vivo of canonically healing B6 mice. Accordingly, MRL/MpJ and B6 patellar tendons were injured via midsubstance punch defects. Healing tendons were isolated after 3 or 7 days and encapsulated in PEG-4MAL hydrogels to develop ECM-derived therapeutic constructs. Constructs were then introduced into B6 mice as a treatment following full thickness midsubstance-punch injuries. Treatment with ECM-derived constructs from MRL/MpJ tendons after 7-days post-injury (M7) resulted in improved matrix alignment, tissue stiffness, decreased collagen III content and improved cell morphology in B6 tendons after 6 weeks post-injury. Furthermore, proteomic analysis showed that M7 contained a unique compositional profile rich in glycoproteins, thereby elucidating a valuable naturally-derived platform for the treatment of tendon injuries. Overall this work highlights promising targets for future therapeutic development and tissue engineering applications.
Collapse
Affiliation(s)
- Juan Paredes
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Ashley Pekmezian
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Nelly Andarawis-Puri
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York,Hospital for Special Surgery, New York, New York
| |
Collapse
|
46
|
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: 2.3] [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
|
47
|
Abstract
Nature faces the challenge of stably attaching soft muscles to a stiff skeleton. A new study combines live imaging and fly genetics to reveal that mechanical tension and a putative intracellular chaperone assist in assembling the gigantic extracellular matrix protein Dumpy at fly tendon-skeleton interfaces.
Collapse
Affiliation(s)
- Clara Sidor
- Turing Center for Living Systems, Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Frank Schnorrer
- Turing Center for Living Systems, Aix Marseille University, CNRS, IBDM, 13288 Marseille, France.
| |
Collapse
|
48
|
Wang D, Zhang X, Huang S, Liu Y, Fu BSC, Mak KKL, Blocki AM, Yung PSH, Tuan RS, Ker DFE. Engineering multi-tissue units for regenerative Medicine: Bone-tendon-muscle units of the rotator cuff. Biomaterials 2021; 272:120789. [PMID: 33845368 DOI: 10.1016/j.biomaterials.2021.120789] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Our body systems are comprised of numerous multi-tissue units. For the musculoskeletal system, one of the predominant functional units is comprised of bone, tendon/ligament, and muscle tissues working in tandem to facilitate locomotion. To successfully treat musculoskeletal injuries and diseases, critical consideration and thoughtful integration of clinical, biological, and engineering aspects are necessary to achieve translational bench-to-bedside research. In particular, identifying ideal biomaterial design specifications, understanding prior and recent tissue engineering advances, and judicious application of biomaterial and fabrication technologies will be crucial for addressing current clinical challenges in engineering multi-tissue units. Using rotator cuff tears as an example, insights relevant for engineering a bone-tendon-muscle multi-tissue unit are presented. This review highlights the tissue engineering strategies for musculoskeletal repair and regeneration with implications for other bone-tendon-muscle units, their derivatives, and analogous non-musculoskeletal tissue structures.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Yang Liu
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Bruma Sai-Chuen Fu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | | | - Anna Maria Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Patrick Shu-Hang Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
49
|
Tsai SL, Noedl MT, Galloway JL. Bringing tendon biology to heel: Leveraging mechanisms of tendon development, healing, and regeneration to advance therapeutic strategies. Dev Dyn 2021; 250:393-413. [PMID: 33169466 PMCID: PMC8486356 DOI: 10.1002/dvdy.269] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Tendons are specialized matrix-rich connective tissues that transmit forces from muscle to bone and are essential for movement. As tissues that frequently transfer large mechanical loads, tendons are commonly injured in patients of all ages. Following injury, mammalian tendons heal poorly through a slow process that forms disorganized fibrotic scar tissue with inferior biomechanical function. Current treatments are limited and patients can be left with a weaker tendon that is likely to rerupture and an increased chance of developing degenerative conditions. More effective, alternative treatments are needed. However, our current understanding of tendon biology remains limited. Here, we emphasize why expanding our knowledge of tendon development, healing, and regeneration is imperative for advancing tendon regenerative medicine. We provide a comprehensive review of the current mechanisms governing tendon development and healing and further highlight recent work in regenerative tendon models including the neonatal mouse and zebrafish. Importantly, we discuss how present and future discoveries can be applied to both augment current treatments and design novel strategies to treat tendon injuries.
Collapse
Affiliation(s)
- Stephanie L. Tsai
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Marie-Therese Noedl
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Jenna L. Galloway
- Center for Regenerative Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02138
| |
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: 8.3] [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
|