1
|
Lyu K, Liu X, Liu T, Lu J, Jiang L, Chen Y, Long L, Wang X, Shi H, Wang F, Li S. miRNAs contributing to the repair of tendon injury. Cell Tissue Res 2023; 393:201-215. [PMID: 37249708 PMCID: PMC10406718 DOI: 10.1007/s00441-023-03780-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 05/02/2023] [Indexed: 05/31/2023]
Abstract
Tendon injury is one of the most common disorders of the musculoskeletal system, with a higher likelihood of occurrence in elderly individuals and athletes. In posthealing tendons, two undesirable consequences, tissue fibrosis and a reduction in mechanical properties, usually occur, resulting in an increased probability of rerupture or reinjury; thus, it is necessary to propose an appropriate treatment. Currently, most methods do not sufficiently modulate the tendon healing process and restore the function and structure of the injured tendon to those of a normal tendon, since there is still inadequate information about the effects of multiple cellular and other relevant signaling pathways on tendon healing and how the expression of their components is regulated. microRNAs are vital targets for promoting tendon repair and can modulate the expression of biological components in signaling pathways involved in various physiological and pathological responses. miRNAs are a type of noncoding ribonucleic acid essential for regulating processes such as cell proliferation, differentiation, migration and apoptosis; inflammatory responses; vascularization; fibrosis; and tissue repair. This article focuses on the biogenesis response of miRNAs while presenting their mechanisms in tendon healing with perspectives and suggestions.
Collapse
Affiliation(s)
- Kexin Lyu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Xinyue Liu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Tianzhu Liu
- Neurology Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jingwei Lu
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Li Jiang
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Yixuan Chen
- School of Physical Education, Southwest Medical University, Luzhou, China
| | - Longhai Long
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoqiang Wang
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Houyin Shi
- Traumatology and Orthopedics Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Fan Wang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Sen Li
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Momen M, Brounts SH, Binversie EE, Sample SJ, Rosa GJM, Davis BW, Muir P. Selection signature analyses and genome-wide association reveal genomic hotspot regions that reflect differences between breeds of horse with contrasting risk of degenerative suspensory ligament desmitis. G3 (BETHESDA, MD.) 2022; 12:6648349. [PMID: 35866615 PMCID: PMC9526059 DOI: 10.1093/g3journal/jkac179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 06/08/2022] [Indexed: 01/07/2023]
Abstract
Degenerative suspensory ligament desmitis is a progressive idiopathic condition that leads to scarring and rupture of suspensory ligament fibers in multiple limbs in horses. The prevalence of degenerative suspensory ligament desmitis is breed related. Risk is high in the Peruvian Horse, whereas pony and draft breeds have low breed risk. Degenerative suspensory ligament desmitis occurs in families of Peruvian Horses, but its genetic architecture has not been definitively determined. We investigated contrasts between breeds with differing risk of degenerative suspensory ligament desmitis and identified associated risk variants and candidate genes. We analyzed 670k single nucleotide polymorphisms from 10 breeds, each of which was assigned one of the four breed degenerative suspensory ligament desmitis risk categories: control (Belgian, Icelandic Horse, Shetland Pony, and Welsh Pony), low risk (Lusitano, Arabian), medium risk (Standardbred, Thoroughbred, Quarter Horse), and high risk (Peruvian Horse). Single nucleotide polymorphisms were used for genome-wide association and selection signature analysis using breed-assigned risk levels. We found that the Peruvian Horse is a population with low effective population size and our breed contrasts suggest that degenerative suspensory ligament desmitis is a polygenic disease. Variant frequency exhibited signatures of positive selection across degenerative suspensory ligament desmitis breed risk groups on chromosomes 7, 18, and 23. Our results suggest degenerative suspensory ligament desmitis breed risk is associated with disturbances to suspensory ligament homeostasis where matrix responses to mechanical loading are perturbed through disturbances to aging in tendon (PIN1), mechanotransduction (KANK1, KANK2, JUNB, SEMA7A), collagen synthesis (COL4A1, COL5A2, COL5A3, COL6A5), matrix responses to hypoxia (PRDX2), lipid metabolism (LDLR, VLDLR), and BMP signaling (GREM2). Our results do not suggest that suspensory ligament proteoglycan turnover is a primary factor in disease pathogenesis.
Collapse
Affiliation(s)
- Mehdi Momen
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sabrina H Brounts
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Emily E Binversie
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Susannah J Sample
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Guilherme J M Rosa
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Brian W Davis
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Peter Muir
- Corresponding author: Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
3
|
Benage LG, Sweeney JD, Giers MB, Balasubramanian R. Dynamic Load Model Systems of Tendon Inflammation and Mechanobiology. Front Bioeng Biotechnol 2022; 10:896336. [PMID: 35910030 PMCID: PMC9335371 DOI: 10.3389/fbioe.2022.896336] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022] Open
Abstract
Dynamic loading is a shared feature of tendon tissue homeostasis and pathology. Tendon cells have the inherent ability to sense mechanical loads that initiate molecular-level mechanotransduction pathways. While mature tendons require physiological mechanical loading in order to maintain and fine tune their extracellular matrix architecture, pathological loading initiates an inflammatory-mediated tissue repair pathway that may ultimately result in extracellular matrix dysregulation and tendon degeneration. The exact loading and inflammatory mechanisms involved in tendon healing and pathology is unclear although a precise understanding is imperative to improving therapeutic outcomes of tendon pathologies. Thus, various model systems have been designed to help elucidate the underlying mechanisms of tendon mechanobiology via mimicry of the in vivo tendon architecture and biomechanics. Recent development of model systems has focused on identifying mechanoresponses to various mechanical loading platforms. Less effort has been placed on identifying inflammatory pathways involved in tendon pathology etiology, though inflammation has been implicated in the onset of such chronic injuries. The focus of this work is to highlight the latest discoveries in tendon mechanobiology platforms and specifically identify the gaps for future work. An interdisciplinary approach is necessary to reveal the complex molecular interplay that leads to tendon pathologies and will ultimately identify potential regenerative therapeutic targets.
Collapse
Affiliation(s)
- Lindsay G. Benage
- School of Chemical, Biological and Environmental Engineering, Oregon State University, Corvallis, OR, United States
| | - James D. Sweeney
- School of Chemical, Biological and Environmental Engineering, Oregon State University, Corvallis, OR, United States
| | - Morgan B. Giers
- School of Chemical, Biological and Environmental Engineering, Oregon State University, Corvallis, OR, United States
- *Correspondence: Morgan B. Giers,
| | - Ravi Balasubramanian
- School of Chemical, Biological and Environmental Engineering, Oregon State University, Corvallis, OR, United States
- School of Mechanical, Industrial and Manufacturing Engineering, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
4
|
Dede Eren A, Vermeulen S, Schmitz TC, Foolen J, de Boer J. The loop of phenotype: Dynamic reciprocity links tenocyte morphology to tendon tissue homeostasis. Acta Biomater 2022; 163:275-286. [PMID: 35584748 DOI: 10.1016/j.actbio.2022.05.019] [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: 12/02/2021] [Revised: 04/24/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022]
Abstract
Cells and their surrounding extracellular matrix (ECM) are engaged in dynamic reciprocity to maintain tissue homeostasis: cells deposit ECM, which in turn presents the signals that define cell identity. This loop of phenotype is obvious for biochemical signals, such as collagens, which are produced by and presented to cells, but the role of biomechanical signals is also increasingly recognised. In addition, cell shape goes hand in hand with cell function and tissue homeostasis. Aberrant cell shape and ECM is seen in pathological conditions, and control of cell shape in micro-fabricated platforms disclose the causal relationship between cell shape and cell function, often mediated by mechanotransduction. In this manuscript, we discuss the loop of phenotype for tendon tissue homeostasis. We describe cell shape and ECM organization in normal and diseased tissue, how ECM composition influences tenocyte shape, and how that leads to the activation of signal transduction pathways and ECM deposition. We further describe the use of technologies to control cell shape to elucidate the link between cell shape and its phenotypical markers and focus on the causal role of cell shape in the loop of phenotype. STATEMENT OF SIGNIFICANCE: The dynamic reciprocity between cells and their surrounding extracellular matrix (ECM) influences biomechanical and biochemical properties of ECM as well as cell function through activation of signal transduction pathways that regulate gene and protein expression. We refer to this reciprocity as Loop of Phenotype and it has been studied and demonstrated extensively by using micro-fabricated platforms to manipulate cell shape and cell fate. In this manuscript, we discuss this concept in tendon tissue homeostasis by giving examples in healthy and pathological tenson tissue. Furthermore, we elaborate this by showing how biomaterials are used to feed this reciprocity to manipulate cell shape and function. Finally, we elucidate the link between cell shape and its phenotypical markers and focus on the activation of signal transduction pathways and ECM deposition.
Collapse
Affiliation(s)
- Aysegul Dede Eren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Steven Vermeulen
- Maastricht University, MERLN Institute for Technology Inspired Regenerative Medicine, Instructive Biomaterial Engineering, Maastricht, the Netherlands
| | - Tara C Schmitz
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jasper Foolen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
5
|
Liu Q, Zhu Y, Zhu W, Zhang G, Yang YP, Zhao C. The role of MicroRNAs in tendon injury, repair, and related tissue engineering. Biomaterials 2021; 277:121083. [PMID: 34488121 PMCID: PMC9235073 DOI: 10.1016/j.biomaterials.2021.121083] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/27/2021] [Accepted: 08/22/2021] [Indexed: 12/15/2022]
Abstract
Tendon injuries are one of the most common musculoskeletal disorders that cause considerable morbidity and significantly compromise the patients' quality of life. The innate limited regenerative capacity of tendon poses a substantial treating challenge for clinicians. MicroRNAs (miRNAs) are a family of small non-coding RNAs that play a vital role in orchestrating many biological processes through post-transcriptional regulation. Increasing evidence reveals that miRNA-based therapeutics may serve as an innovative strategy for the treatment of tendon pathologies. In this review, we briefly present miRNA biogenesis, the role of miRNAs in tendon cell biology and their involvement in tendon injuries, followed by a summary of current miRNA-based approaches in tendon tissue engineering with a special focus on attenuating post-injury fibrosis. Next, we discuss the advantages of miRNA-functionalized scaffolds in achieving sustained and localized miRNA administration to minimize off-target effects, and thus hoping to inspire the development of effective miRNA delivery platforms specifically for tendon tissue engineering. We envision that advancement in miRNA-based therapeutics will herald a new era of tendon tissue engineering and pave a way for clinical translation for the treatments of tendon disorders.
Collapse
Affiliation(s)
- Qian Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Yaxi Zhu
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong SAR, PR China
| | - Yunzhi Peter Yang
- Department of Orthopedic Surgery, (by courtesy) Materials Science and Engineering, and Bioengineering, Stanford University, Stanford, CA, USA
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
6
|
Kharaz YA, Birch H, Chester A, Alchorne E, Simpson D, Clegg P, Comerford E. The effect of exercise on the protein profile of rat knee joint intra- and extra-articular ligaments. Scand J Med Sci Sports 2021; 31:2033-2043. [PMID: 34271594 DOI: 10.1111/sms.14023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/15/2021] [Indexed: 01/13/2023]
Abstract
Injuries to the intra-articular anterior cruciate ligament (ACL) and the extra-articular medial collateral ligament (MCL) result in significant knee joint instability, pain, and immobility. Moderate endurance-type exercise can increase ligament strength but little is known on the effect of short-term regular bouts of high-intensity exercise on the extracellular matrix (ECM) structure of knee ligaments. Therefore, this study aimed to identify the effect of short-term regular bouts high exercise on the proteome of the rat ACL and MCL using mass spectrometry. Sprague-Dawley male rats (n = 6) were split into control and exercise groups, and subjected to high-intensity training for four 4 weeks followed by proteomic analyses of the ACL and MCL. Knee joint health status was assessed using OARSI and a validated histological scoring system. Histopathological analyses demonstrated no significant changes in either in cruciate, collateral ligaments, or cartilage between the control and exercised knee joints. However, significant proteins were found to be more abundant in the exercised ACL compared to ACL control group but not between the exercised MCL and control MCL groups. The significant abundant proteins in ACL exercise groups were mostly cytoskeletal, ribosomal and enzymes with several abundant matrisomal proteins such as collagen proteins and proteoglycans being found in this group. In conclusion, our results indicate that short-term regular bouts of high-intensity exercise have an impact on the intra-articular ACL but not extra-articular MCL ECM protein expression.
Collapse
Affiliation(s)
- Yalda A Kharaz
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Helen Birch
- Department of Orthopaedics and Musculoskeletal Science, University College London, London, UK
| | | | | | - Deborah Simpson
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Peter Clegg
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,School of Veterinary Science, Leahurst Campus, Neston, UK
| | - Eithne Comerford
- Department of Musculoskeletal and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.,School of Veterinary Science, Leahurst Campus, Neston, UK
| |
Collapse
|
7
|
Ding L, Wang M, Qin S, Xu L. The Roles of MicroRNAs in Tendon Healing and Regeneration. Front Cell Dev Biol 2021; 9:687117. [PMID: 34277629 PMCID: PMC8283311 DOI: 10.3389/fcell.2021.687117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/11/2021] [Indexed: 01/20/2023] Open
Abstract
Tendons connect the muscle abdomen of skeletal muscles to the bone, which transmits the force generated by the muscle abdomen contraction and pulls the bone into motion. Tendon injury is a common clinical condition occurring in certain populations, such as repeated tendon strains in athletes. And it can lead to substantial pain and loss of motor function, in severe cases, significant disability. Tendon healing and regeneration have attracted growing interests. Some treatments including growth factors, stem cell therapies and rehabilitation programs have been tried to improve tendon healing. However, the basic cellular biology and pathology of tendons are still not fully understood, and the management of tendon injury remains a considerable challenge. Regulating gene expression at post-transcriptional level, microRNA (miRNA) has been increasingly recognized as essential regulators in the biological processes of tendon healing and regeneration. A wide range of miRNAs in tendon injury have been shown to play vital roles in maintaining and regulating its physiological function, as well as regulating the tenogenic differentiation potential of stem cells. In this review, we show the summary of the latest information on the role of miRNAs in tendon healing and regeneration, and also discuss potentials for miRNA-directed diagnosis and therapy in tendon injuries and tendinopathy, which may provide new theoretical foundation for tenogenesis and tendon healing.
Collapse
Affiliation(s)
- Lingli Ding
- Lingnan Medical Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Wang
- Lingnan Medical Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shengnan Qin
- Department of Orthopaedics, Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Liangliang Xu
- Lingnan Medical Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Stadnik PS, Gilbert SJ, Tarn J, Charlton S, Skelton AJ, Barter MJ, Duance VC, Young DA, Blain EJ. Regulation of microRNA-221, -222, -21 and -27 in articular cartilage subjected to abnormal compressive forces. J Physiol 2020; 599:143-155. [PMID: 33052608 PMCID: PMC8132181 DOI: 10.1113/jp279810] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/09/2020] [Indexed: 01/01/2023] Open
Abstract
Key points microRNAs (miRs) are small non‐coding molecules that regulate post‐transcriptional target gene expression. miRs are involved in regulating cellular activities in response to mechanical loading in all physiological systems, although it is largely unknown whether this response differs with increasing magnitudes of load. miR‐221, miR‐222, miR‐21‐5p and miR‐27a‐5p were significantly increased in ex vivo cartilage explants subjected to increasing load magnitude and in in vivo joint cartilage exposed to abnormal loading. TIMP3 and CPEB3 are putative miR targets in chondrocytes Identification of mechanically regulated miRs that have potential to impact on tissue homeostasis provides a mechanism by which load‐induced tissue behaviour is regulated, in both health and pathology, in all physiological systems.
Abstract MicroRNAs (miRs) are small non‐coding molecules that regulate post‐transcriptional target gene expression and are involved in mechano‐regulation of cellular activities in all physiological systems. It is unknown whether such epigenetic mechanisms are regulated in response to increasing magnitudes of load. The present study investigated mechano‐regulation of miRs in articular cartilage subjected to ‘physiological’ and ‘non‐physiological’ compressive loads in vitro as a model system and validated findings in an in vivo model of abnormal joint loading. Bovine full‐depth articular cartilage explants were loaded to 2.5 MPa (physiological) or 7 MPa (non‐physiological) (1 Hz, 15 min) and mechanically‐regulated miRs identified using next generation sequencing and verified using a quantitative PCR. Downstream targets were verified using miR‐specific mimics or inhibitors in conjunction with 3′‐UTR luciferase activity assays. A subset of miRs were mechanically‐regulated in ex vivo cartilage explants and in vivo joint cartilage. miR‐221, miR‐222, miR‐21‐5p and miR‐27a‐5p were increased and miR‐483 levels decreased with increasing load magnitude. Tissue inhibitor of metalloproteinase 3 (TIMP3) and cytoplasmic polyadenylation element binding protein 3 (CPEB3) were identified as putative downstream targets. Our data confirm miR‐221 and ‐222 mechano‐regulation and demonstrates novel mechano‐regulation of miR‐21‐5p and miR‐27a‐5p in ex vivo and in vivo cartilage loading models. TIMP3 and CPEB3 are putative miR targets in chondrocytes. Identification of specific miRs that are regulated by increasing load magnitude, as well as their potential to impact on tissue homeostasis, has direct relevance to other mechano‐sensitive physiological systems and provides a mechanism by which load‐induced tissue behaviour is regulated, in both health and pathology. microRNAs (miRs) are small non‐coding molecules that regulate post‐transcriptional target gene expression. miRs are involved in regulating cellular activities in response to mechanical loading in all physiological systems, although it is largely unknown whether this response differs with increasing magnitudes of load. miR‐221, miR‐222, miR‐21‐5p and miR‐27a‐5p were significantly increased in ex vivo cartilage explants subjected to increasing load magnitude and in in vivo joint cartilage exposed to abnormal loading. TIMP3 and CPEB3 are putative miR targets in chondrocytes Identification of mechanically regulated miRs that have potential to impact on tissue homeostasis provides a mechanism by which load‐induced tissue behaviour is regulated, in both health and pathology, in all physiological systems.
Collapse
Affiliation(s)
- Paulina S Stadnik
- Biomechanics and Bioengineering Research Centre Versus Arthritis, Biomedicine Division, School of Biosciences, The Sir Martin Evans Building, Cardiff University, Cardiff, Wales, UK
| | - Sophie J Gilbert
- Biomechanics and Bioengineering Research Centre Versus Arthritis, Biomedicine Division, School of Biosciences, The Sir Martin Evans Building, Cardiff University, Cardiff, Wales, UK
| | - Jessica Tarn
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Sarah Charlton
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew J Skelton
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew J Barter
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Victor C Duance
- Biomechanics and Bioengineering Research Centre Versus Arthritis, Biomedicine Division, School of Biosciences, The Sir Martin Evans Building, Cardiff University, Cardiff, Wales, UK
| | - David A Young
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Emma J Blain
- Biomechanics and Bioengineering Research Centre Versus Arthritis, Biomedicine Division, School of Biosciences, The Sir Martin Evans Building, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
9
|
Zamboulis DE, Thorpe CT, Ashraf Kharaz Y, Birch HL, Screen HR, Clegg PD. Postnatal mechanical loading drives adaptation of tissues primarily through modulation of the non-collagenous matrix. eLife 2020; 9:58075. [PMID: 33063662 PMCID: PMC7593091 DOI: 10.7554/elife.58075] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Mature connective tissues demonstrate highly specialised properties, remarkably adapted to meet their functional requirements. Tissue adaptation to environmental cues can occur throughout life and poor adaptation commonly results in injury. However, the temporal nature and drivers of functional adaptation remain undefined. Here, we explore functional adaptation and specialisation of mechanically loaded tissues using tendon; a simple aligned biological composite, in which the collagen (fascicle) and surrounding predominantly non-collagenous matrix (interfascicular matrix) can be interrogated independently. Using an equine model of late development, we report the first phase-specific analysis of biomechanical, structural, and compositional changes seen in functional adaptation, demonstrating adaptation occurs postnatally, following mechanical loading, and is almost exclusively localised to the non-collagenous interfascicular matrix. These novel data redefine adaptation in connective tissue, highlighting the fundamental importance of non-collagenous matrix and suggesting that regenerative medicine strategies should change focus from the fibrous to the non-collagenous matrix of tissue.
Collapse
Affiliation(s)
- Danae E Zamboulis
- Institute of Ageing and Chronic Disease, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Chavaunne T Thorpe
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, United Kingdom
| | - Yalda Ashraf Kharaz
- Institute of Ageing and Chronic Disease, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Helen L Birch
- University College London, Department of Orthopaedics and Musculoskeletal Science, Stanmore Campus, Royal National Orthopaedic Hospital, Stanmore, United Kingdom
| | - Hazel Rc Screen
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
10
|
Laguette MJN, Barrow K, Firfirey F, Dlamini S, Saunders CJ, Dandara C, Gamieldien J, Collins M, September AV. Exploring new genetic variants within COL5A1 intron 4-exon 5 region and TGF-β family with risk of anterior cruciate ligament ruptures. J Orthop Res 2020; 38:1856-1865. [PMID: 31922278 DOI: 10.1002/jor.24585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/03/2020] [Indexed: 02/04/2023]
Abstract
Variants within genes encoding structural and regulatory elements of ligaments have been associated with musculoskeletal soft tissue injury risk. The role of intron 4-exon 5 variants within the α1 chain of type V collagen (COL5A1) gene and genes of the transforming growth factor-β (TGF-β) family, TGFBR3 and TGFBI, was investigated on the risk of anterior cruciate ligament (ACL) ruptures. A case-control genetic association study was performed on 210 control (CON) and 249 participants with surgically diagnosed ruptures (ACL), of which 147 reported a noncontact mechanism of injury (NON). Whole-exome sequencing data were used to prioritize variants of potential functional relevance. Genotyping for COL5A1 (rs3922912 G>A, rs4841926 C>T, and rs3124299 C>T), TGFBR3 (rs1805113 G>A and rs1805117 T>C), and TGFBI (rs1442 G>C) was performed using Taqman SNP genotyping assays. Significant overrepresentation of the G allele of TGFBR3 rs1805113 was observed in CON vs ACL (P = .014) and NON groups (P = .021). Similar results were obtained in a female with the G allele (CON vs ACL: P = .029; CON vs NON: P = .016). The TGFBI rs1442 CC genotype was overrepresented in the female ACL vs CON (P = .013). Associations of inferred allele combinations were observed in line with the above results. COL5A1 intron 4-exon 5 genomic interval was not associated with the risk of ACL ruptures. Instead, this novel study is the first to use this approach to identify variants within the TGF-β signaling pathway to be implicated in the risk of ACL ruptures. A genetic susceptibility interval was identified to be explored in the context of extracellular matrix remodeling.
Collapse
Affiliation(s)
- Mary-Jessica N Laguette
- Division of Exercise Science and Sports Medicine (ESSM), University of Cape Town, Cape Town, South Africa.,International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, ESSM, University of Cape Town, Cape Town, South Africa.,Research Centre for Health Through Physical Activity and Sport, University of Cape Town, Cape Town, South Africa
| | - Kelly Barrow
- Department of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - Firzana Firfirey
- Division of Exercise Science and Sports Medicine (ESSM), University of Cape Town, Cape Town, South Africa.,International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, ESSM, University of Cape Town, Cape Town, South Africa.,Research Centre for Health Through Physical Activity and Sport, University of Cape Town, Cape Town, South Africa
| | - Senanile Dlamini
- Division of Exercise Science and Sports Medicine (ESSM), University of Cape Town, Cape Town, South Africa.,International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, ESSM, University of Cape Town, Cape Town, South Africa.,Research Centre for Health Through Physical Activity and Sport, University of Cape Town, Cape Town, South Africa
| | - Colleen J Saunders
- South African National Bioinformatics Institute/MRC Unit for Bioinformatics Capacity, University of the Western Cape, Cape Town, Bellville, South Africa.,Division of Emergency Medicine, Department of Surgery, University of Cape Town, Cape Town, South Africa
| | - Collet Dandara
- Department of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - Junaid Gamieldien
- South African National Bioinformatics Institute/MRC Unit for Bioinformatics Capacity, University of the Western Cape, Cape Town, Bellville, South Africa
| | - Malcolm Collins
- Division of Exercise Science and Sports Medicine (ESSM), University of Cape Town, Cape Town, South Africa.,International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, ESSM, University of Cape Town, Cape Town, South Africa.,Research Centre for Health Through Physical Activity and Sport, University of Cape Town, Cape Town, South Africa
| | - Alison V September
- Division of Exercise Science and Sports Medicine (ESSM), University of Cape Town, Cape Town, South Africa.,International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, ESSM, University of Cape Town, Cape Town, South Africa.,Research Centre for Health Through Physical Activity and Sport, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
11
|
Gumucio JP, Schonk MM, Kharaz YA, Comerford E, Mendias CL. Scleraxis is required for the growth of adult tendons in response to mechanical loading. JCI Insight 2020; 5:138295. [PMID: 32463804 DOI: 10.1172/jci.insight.138295] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Scleraxis is a basic helix-loop-helix transcription factor that plays a central role in promoting tenocyte proliferation and matrix synthesis during embryonic tendon development. However, the role of scleraxis in the growth and adaptation of adult tendons is not known. We hypothesized that scleraxis is required for tendon growth in response to mechanical loading and that scleraxis promotes the specification of progenitor cells into tenocytes. We conditionally deleted scleraxis in adult mice using a tamoxifen-inducible Cre-recombinase expressed from the Rosa26 locus (ScxΔ) and then induced tendon growth in Scx+ and ScxΔ adult mice via plantaris tendon mechanical overload. Compared with the WT Scx+ group, ScxΔ mice demonstrated blunted tendon growth. Transcriptional and proteomic analyses revealed significant reductions in cell proliferation, protein synthesis, and extracellular matrix genes and proteins. Our results indicate that scleraxis is required for mechanically stimulated adult tendon growth by causing the commitment of CD146+ pericytes into the tenogenic lineage and by promoting the initial expansion of newly committed tenocytes and the production of extracellular matrix proteins.
Collapse
Affiliation(s)
- Jonathan P Gumucio
- Department of Molecular & Integrative Physiology and.,Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Yalda A Kharaz
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, and Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing, University of Liverpool, Liverpool, United Kingdom
| | - Eithne Comerford
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, and Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing, University of Liverpool, Liverpool, United Kingdom
| | - Christopher L Mendias
- Department of Molecular & Integrative Physiology and.,Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Hospital for Special Surgery, New York, New York, USA.,Department of Physiology & Biophysics, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
12
|
Giordano L, Porta GD, Peretti GM, Maffulli N. Therapeutic potential of microRNA in tendon injuries. Br Med Bull 2020; 133:79-94. [PMID: 32219416 DOI: 10.1093/bmb/ldaa002] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/07/2020] [Accepted: 01/24/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The regulatory role of microRNA (miRNA) in several conditions has been studied, but their function in tendon healing remains elusive. This review summarizes how miRNAs are related to the pathogenesis of tendon injuries and highlights their clinical potential, focusing on the issues related to their delivery for clinical purposes. SOURCES OF DATA We searched multiple databases to perform a systematic review on miRNA in relation to tendon injuries. We included in the present work a total of 15 articles. AREAS OF AGREEMENT The mechanism of repair of tendon injuries is probably mediated by resident tenocytes. These maintain a fine equilibrium between anabolic and catabolic events of the extracellular matrix. Specific miRNAs regulate cytokine expression and orchestrate proliferation and differentiation of stromal cell lines involved in the composition of the extracellular matrix. AREAS OF CONTROVERSY The lack of effective delivery systems poses serious obstacles to the clinical translation of these basic science findings. GROWING POINT In vivo studies should be planned to better explore the relationship between miRNA and tendon injuries and evaluate the most suitable delivery system for these molecules. AREAS TIMELY FOR DEVELOPING RESEARCH Investigations ex vivo suggest therapeutic opportunities of miRNA for the management of tendon injuries. Given the poor pharmacokinetic properties of miRNAs, these must be delivered by an adequate adjuvant transport system.
Collapse
Affiliation(s)
- Lorenzo Giordano
- Department of Musculoskeletal Disorder, Faculty of Medicine, Surgery and Dentistry, University of Salerno, Via San Leonardo 1, 84131 Salerno, Italy
| | - Giovanna Della Porta
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy
| | - Giuseppe M Peretti
- Department of Biomedical Sciences for Health, University of Milan, Via Riccardo Galeazzi 4, 20161, Milan, Italy.,IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi 4, 20161 Milan, Italy
| | - Nicola Maffulli
- Department of Musculoskeletal Disorder, Faculty of Medicine, Surgery and Dentistry, University of Salerno, Via San Leonardo 1, 84131 Salerno, Italy.,Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy.,Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Mile End Hospital, 275 Bancroft Road, London E1 4DG, England.,School of Pharmacy and Bioengineering, Keele University School of Medicine, Thornburrow Drive, Stoke on Trent ST5 5B, England
| |
Collapse
|
13
|
Cho SH, Kim YR, Kim JH, An HJ, Kim JO, Ko JJ, Lee WS, Kim NK. The association of miR-25T>C, miR-32C>A, miR-125C>T, and miR-222G>T polymorphisms with a risk of primary ovarian insufficiency in Korean women. Menopause 2020; 26:409-416. [PMID: 30422934 DOI: 10.1097/gme.0000000000001258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate the association of microRNA polymorphisms (miR-25T>C, miR-32C>A, miR-125C>T, and miR-222G>T) with primary ovarian insufficiency (POI) in Korean women. METHODS We conducted a case-control study of Korean women: 142 participants with POI and 266 controls with at least 1 live birth and no history of pregnancy loss. RESULTS The haplotype-based multifactor dimensionality reduction analysis revealed that the T-C-T-G (miR-25/-32/-125/-222), T-A-C-G (miR-25/-32/-125/-222), C-T-G (miR-32/-125/-222), A-C-G (miR-32/-125/-222), T-G (miR-122/-222), C-T (miR-32/-125), and C-C (miR-25/-32) inferred haplotypes were significantly less frequent in POI (P < 0.05), which suggested potential protective effects. Participants with POI had significantly increased luteinizing hormone levels (P < 0.05), but hormonal levels, including luteinizing hormone, were not significantly different between POI women and control women with miR-32/-125/-222. CONCLUSIONS After considering multiple comparisons, we concluded that miR-25T>C, miR-32C>A, miR-125C>T, and miR-222G>T had no relation with POI.
Collapse
Affiliation(s)
- Sung Hwan Cho
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Young Ran Kim
- Department of Obstetrics and Gynecology, School of Medicine, CHA University, Seongnam, Korea
| | - Ji Hyang Kim
- Department of Obstetrics and Gynecology, School of Medicine, CHA University, Seongnam, Korea
| | - Hui Jeong An
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Jung Oh Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Jung Jae Ko
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Woo Sik Lee
- Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| |
Collapse
|
14
|
Tendon and ligament mechanical loading in the pathogenesis of inflammatory arthritis. Nat Rev Rheumatol 2020; 16:193-207. [PMID: 32080619 DOI: 10.1038/s41584-019-0364-x] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
Mechanical loading is an important factor in musculoskeletal health and disease. Tendons and ligaments require physiological levels of mechanical loading to develop and maintain their tissue architecture, a process that is achieved at the cellular level through mechanotransduction-mediated fine tuning of the extracellular matrix by tendon and ligament stromal cells. Pathological levels of force represent a biological (mechanical) stress that elicits an immune system-mediated tissue repair pathway in tendons and ligaments. The biomechanics and mechanobiology of tendons and ligaments form the basis for understanding how such tissues sense and respond to mechanical force, and the anatomical extent of several mechanical stress-related disorders in tendons and ligaments overlaps with that of chronic inflammatory arthritis in joints. The role of mechanical stress in 'overuse' injuries, such as tendinopathy, has long been known, but mechanical stress is now also emerging as a possible trigger for some forms of chronic inflammatory arthritis, including spondyloarthritis and rheumatoid arthritis. Thus, seemingly diverse diseases of the musculoskeletal system might have similar mechanisms of immunopathogenesis owing to conserved responses to mechanical stress.
Collapse
|
15
|
Willard K, Laguette MJN, Alves de Souza Rios L, D'Alton C, Nel M, Prince S, Collins M, September AV. Altered expression of proteoglycan, collagen and growth factor genes in a TGF-β1 stimulated genetic risk model for musculoskeletal soft tissue injuries. J Sci Med Sport 2020; 23:695-700. [PMID: 32061523 DOI: 10.1016/j.jsams.2020.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVES To investigate the functional effect of implicated variants within BGN and COL5A1 on gene expression of components of the extracellular matrix (ECM) in a TGF-β-stimulated risk model for musculoskeletal soft tissue injuries. DESIGN Experimental research, laboratory study. METHODS Skin biopsies were obtained from nine healthy participants with either a combined increased or reduced risk profile for COL5A1 rs12722 C>T and BGN rs1126499 C>T - rs1042103 G>A, and primary fibroblast cell lines were established. Total RNA was extracted at baseline (10% FBS), after serum starvation (1% FBS) and TGF-β1 treatment (1% FBS, 10ng/mL TGF-1β). Relative mRNA levels of BGN, COL5A1, DCN and VEGFA was quantified using Taqman® array pre-spotted plate assays (Applied Biosystems, Foster city, CA, USA). RESULTS At baseline, the reduced risk group had 2.5, 1.9 and 2 fold increases (p<0.001) in relative BGN, COL5A1 and VEGFA mRNA levels respectively. In the serum starved experiments, except for a significant 1.5 fold (p=0.017) increase in relative DCN mRNA expression in the reduced risk group, similar observations were noted for the other three genes. After TGF-1β treatment, the reduced risk group had 1.3 (p=0.011) and 1.4 fold (p=0.001) increases in the relative COL5A1 and VEGFA mRNA levels, respectively. CONCLUSIONS Altered mRNA levels associated with genetic risk profiles for musculoskeletal soft injury risk at baseline (BGN, COL5A1 and VEGFA), with serum starvation (DCN) and after TGF-β1 treatment (COL5A1 and VEGFA) provide additional functional evidence to support the association of implicated genetic loci with several musculoskeletal soft tissue injuries. Implication of altered gene expression profiles underpinning these genetic risk associated loci potentially highlight key therapeutic targets for management of these injuries.
Collapse
Affiliation(s)
- Kyle Willard
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, South Africa; UCT Research Centre for Health through Physical Activity, Lifestyle and Sport, South Africa; International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, South Africa
| | - Mary-Jessica Nancy Laguette
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, South Africa; UCT Research Centre for Health through Physical Activity, Lifestyle and Sport, South Africa; International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, South Africa
| | | | - Caroline D'Alton
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, South Africa; UCT Research Centre for Health through Physical Activity, Lifestyle and Sport, South Africa; International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, South Africa
| | - Melissa Nel
- Division of Cell BiologyDepartment of Human Biology, University of Cape Town, South Africa
| | - Sharon Prince
- Division of Cell BiologyDepartment of Human Biology, University of Cape Town, South Africa
| | - Malcolm Collins
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, South Africa; UCT Research Centre for Health through Physical Activity, Lifestyle and Sport, South Africa; International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, South Africa
| | - Alison Victoria September
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, South Africa; UCT Research Centre for Health through Physical Activity, Lifestyle and Sport, South Africa; International Federation of Sports Medicine (FIMS) Collaborative Centre of Sports Medicine, South Africa.
| |
Collapse
|
16
|
Lin CC, Wu PT, Chang CW, Lin RW, Wang GJ, Jou IM, Lai KA. A single-pulsed electromagnetic field enhances collagen synthesis in tendon cells. Med Eng Phys 2020; 77:130-136. [PMID: 31954614 DOI: 10.1016/j.medengphy.2019.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 12/03/2019] [Accepted: 12/08/2019] [Indexed: 01/18/2023]
Abstract
Tendinopathy is a progressive pathology of tendon that is characteristic of imbalance between matrix synthesis and degeneration and is often caused by failure to adapt to mechanical loading. Non-steroidal anti-inflammatory medications (NSAIDS) are used as a conventional treatment to alleviate pain and swelling in the short term, but the ideal treatment for tendinopathy remains unclear. Here, we show a single pulsed electromagnetic field (SPEMF, 0.2 Hz) that up-regulated tenogenic gene expression (Col1a1, Col3a1, Scx, Dcn) and down-regulated inflammatory gene expression (Mmp1) in vitro. After five days of SPEMF stimulation (3 min/day), the collagen type I and total collagen synthesis protein levels were significantly increased. Under pro-inflammatory cytokine (IL-1β) irritation, the decreased expression of Col1a1/Col3a1 was up-regulated by SPEMF treatment, and the increased expression of Mmp1 was also reversed. From the above, it can be inferred that SPEMF that enhances matrix synthesis and reduces matrix degeneration may counteract the imbalance in tendinopathy. SPEMF application may be developed as a potential future strategy for therapeutic intervention in tendon disorders.
Collapse
Affiliation(s)
- Chih-Chun Lin
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan; Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Ting Wu
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Wei Chang
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ru-Wei Lin
- Institute of Food Safety Management, College of Agriculture, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Gwo-Jaw Wang
- Departments of Orthopedics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Department of Orthopedic Surgery, University of Virginia, Charlottesville, VA, USA
| | - I-Ming Jou
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuo-An Lai
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
17
|
Tan KT, Ang STJ, Tsai SY. Sarcopenia: Tilting the Balance of Protein Homeostasis. Proteomics 2019; 20:e1800411. [PMID: 31722440 DOI: 10.1002/pmic.201800411] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/04/2019] [Indexed: 12/14/2022]
Abstract
Sarcopenia, defined as age-associated decline of muscle mass and function, is a risk factor for mortality and disability, and comorbid with several chronic diseases such as type II diabetes and cardiovascular diseases. Clinical trials showed that nutritional supplements had positive effects on muscle mass, but not on muscle function and strength, demonstrating our limited understanding of the molecular events involved in the ageing muscle. Protein homeostasis, the equilibrium between protein synthesis and degradation, is proposed as the major mechanism underlying the development of sarcopenia. As the key central regulator of protein homeostasis, the mammalian target of rapamycin (mTOR) is proposed to be essential for muscle hypertrophy. Paradoxically, sustained activation of mTOR complex 1 (mTORC1) is associated with a loss of sensitivity to extracellular signaling in the elderly. It is not understood why sustained mTORC1 activity, which should induce muscle hypertrophy, instead results in muscle atrophy. Here, recent findings on the implications of disrupting protein homeostasis on muscle physiology and sarcopenia development in the context of mTOR/protein kinase B (AKT) signaling are reviewed. Understanding the role of these molecular mechanisms during the ageing process will contribute towards the development of targeted therapies that will improve protein metabolism and reduce sarcopenia.
Collapse
Affiliation(s)
- Kuan Ting Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9 Admin Office, Singapore, 117597, Singapore
| | - Seok-Ting Jamie Ang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9 Admin Office, Singapore, 117597, Singapore
| | - Shih-Yin Tsai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9 Admin Office, Singapore, 117597, Singapore
| |
Collapse
|
18
|
Guo S, Chen Q, Sun Y, Chen J. Nicotinamide protects against skeletal muscle atrophy in streptozotocin-induced diabetic mice. Arch Physiol Biochem 2019; 125:470-477. [PMID: 31291133 DOI: 10.1080/13813455.2019.1638414] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/14/2019] [Indexed: 02/08/2023]
Abstract
Context: Skeletal muscle atrophy is a complication of diabetes, partially induced by nicotinamide adenine dinucleotide (NAD+) deficiency. Objective: This study investigates the potential of nicotinamide (NAM) supplementation, a precursor of NAD+, against muscle atrophy. Methods: Mice were separated into normal control group, normal control with NAM administration group, diabetic group, and diabetic mice with NAM administration group. Basic characteristics, muscle weight, maximal grip strength, and myofibers cross-sectional area were analysed. Markers reflecting muscle atrophy and hypertrophy, and transforming growth factor β1/Smad2 (TGF-β1/Smad2) pathway were examined. Results: NAM did not influence body weight and blood glucose. In diabetic mice, NAM increased NAD+ level, rescued muscle weight and strength loss, and increased myofibers cross-sectional area. NAM inhibited MuRF1 and Atrogin1, while elevated phosphorylation of Akt. Overactivation of TGF-β1/Smad2 pathway was repressed by NAM. Conclusion: NAM ameliorated diabetic muscle atrophy by rebalancing protein anabolism and catabolism, probably through de-activation of TGF-β1/Smad2 signaling.
Collapse
Affiliation(s)
- Shizhe Guo
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University , Shanghai , China
| | - Qingyan Chen
- Department of Biology, College of Arts and Sciences, Boston University , Boston , MA , USA
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University , Shanghai , China
| | - Jiwu Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University , Shanghai , China
| |
Collapse
|
19
|
Disser NP, Sugg KB, Talarek JR, Sarver DC, Rourke BJ, Mendias CL. Insulin-like growth factor 1 signaling in tenocytes is required for adult tendon growth. FASEB J 2019; 33:12680-12695. [PMID: 31536390 DOI: 10.1096/fj.201901503r] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tenocytes serve to synthesize and maintain collagen fibrils and other extracellular matrix proteins in tendon. Despite the high prevalence of tendon injury, the underlying biologic mechanisms of postnatal tendon growth and repair are not well understood. IGF1 plays an important role in the growth and remodeling of numerous tissues but less is known about IGF1 in tendon. We hypothesized that IGF1 signaling is required for proper tendon growth in response to mechanical loading through regulation of collagen synthesis and cell proliferation. To test this hypothesis, we conditionally deleted the IGF1 receptor (IGF1R) in scleraxis (Scx)-expressing tenocytes using a tamoxifen-inducible Cre-recombinase system and caused tendon growth in adult mice via mechanical overload of the plantaris tendon. Compared with control Scx-expressing IGF1R-positive (Scx:IGF1R+) mice, in which IGF1R is present in tenocytes, mice that lacked IGF1R in their tenocytes [Scx-expressing IGF1R-negative (Scx:IGF1RΔ) mice] demonstrated reduced cell proliferation and smaller tendons in response to mechanical loading. Additionally, we identified that both the PI3K/protein kinase B and ERK pathways are activated downstream of IGF1 and interact in a coordinated manner to regulate cell proliferation and protein synthesis. These studies indicate that IGF1 signaling is required for proper postnatal tendon growth and support the potential use of IGF1 in the treatment of tendon disorders.-Disser, N. P., Sugg, K. B., Talarek, J. R., Sarver, D. C., Rourke, B. J., Mendias, C. L. Insulin-like growth factor 1 signaling in tenocytes is required for adult tendon growth.
Collapse
Affiliation(s)
| | - Kristoffer B Sugg
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Section of Plastic and Reconstructive Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jeffrey R Talarek
- Hospital for Special Surgery, New York, New York, USA.,Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Dylan C Sarver
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Brennan J Rourke
- Hospital for Special Surgery, New York, New York, USA.,Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Christopher L Mendias
- Hospital for Special Surgery, New York, New York, USA.,Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
20
|
Freedman BR, Mooney DJ. Biomaterials to Mimic and Heal Connective Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806695. [PMID: 30908806 PMCID: PMC6504615 DOI: 10.1002/adma.201806695] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/27/2019] [Indexed: 05/11/2023]
Abstract
Connective tissue is one of the four major types of animal tissue and plays essential roles throughout the human body. Genetic factors, aging, and trauma all contribute to connective tissue dysfunction and motivate the need for strategies to promote healing and regeneration. The goal here is to link a fundamental understanding of connective tissues and their multiscale properties to better inform the design and translation of novel biomaterials to promote their regeneration. Major clinical problems in adipose tissue, cartilage, dermis, and tendon are discussed that inspire the need to replace native connective tissue with biomaterials. Then, multiscale structure-function relationships in native soft connective tissues that may be used to guide material design are detailed. Several biomaterials strategies to improve healing of these tissues that incorporate biologics and are biologic-free are reviewed. Finally, important guidance documents and standards (ASTM, FDA, and EMA) that are important to consider for translating new biomaterials into clinical practice are highligted.
Collapse
Affiliation(s)
- Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
21
|
Cui H, He Y, Chen S, Zhang D, Yu Y, Fan C. Macrophage-Derived miRNA-Containing Exosomes Induce Peritendinous Fibrosis after Tendon Injury through the miR-21-5p/Smad7 Pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 14:114-130. [PMID: 30594070 PMCID: PMC6307349 DOI: 10.1016/j.omtn.2018.11.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/13/2018] [Accepted: 11/13/2018] [Indexed: 01/02/2023]
Abstract
Following tendon injury, the development of fibrotic healing response impairs tendon function and restricts tendon motion. Peritendinous tissue fibrosis poses a major clinical problem in hand surgery. Communication between macrophages and tendon cells has a critical role in regulating the tendon-healing process. Yet, the mechanisms employed by macrophages to control peritendinous fibrosis are not fully understood. Here we analyze the role of macrophages in tendon adhesion in mice by pharmacologically depleting them. Such macrophage-depleted mice have less peritendinous fibrosis formation around the injured tendon compared with wild-type littermates. Macrophage-depleted mice restart fibrotic tendon healing by treatment with bone marrow macrophage-derived exosomes. We show that bone marrow macrophages secrete exosomal miR-21-5p that directly targets Smad7, leading to the activation of fibrogenesis in tendon cells. These results demonstrate that intercellular crosstalk between bone marrow macrophages and tendon cells is mediated by macrophage-derived miR-21-5p-containing exosomes that control the fibrotic healing response, providing potential targets for the prevention and treatment of tendon adhesion.
Collapse
Affiliation(s)
- Haomin Cui
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Department of Orthopaedics, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, China
| | - Yu He
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100010, China
| | - Shuai Chen
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Deming Zhang
- Zhejiang Province's Key Laboratory of 3D Printing and Equipment, College of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yaling Yu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Department of Orthopaedics, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, China.
| |
Collapse
|
22
|
Dubin JA, Greenberg DR, Iglinski-Benjamin KC, Abrams GD. Effect of micro-RNA on tenocytes and tendon-related gene expression: A systematic review. J Orthop Res 2018; 36:2823-2829. [PMID: 29873411 DOI: 10.1002/jor.24064] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/21/2018] [Indexed: 02/04/2023]
Abstract
The purpose of the review was to synthesize the current literature regarding the effect of miRNA on biological processes known to be involved in tendon and tenocyte development and homeostasis. Using multiple databases, a systematic review was performed with a customized search term crafted to identify any study examining micro-RNA in relation to tendon and/or tenocytes. Results were classified based on the following categories: Gene expression, tenocyte development and differentiation, tendon tissue repair, and tenocyte senescence. A total of 3,112 potentially relevant studies were reviewed, and after exclusion criteria was applied, 15 investigations were included in the final analysis. There were 14 specific miRNA included in this review, with 11 studies reporting on tendon-related gene expression, five reporting on tendon development and/or tenocyte differentiation, six reporting on tendon tissue repair, and five reporting on tenocyte senescence. The miR-29 family was the most commonly reported micro-RNA in the investigation. We also report on a number of micro-RNA which are associated with both positive and negative effects on tendon homeostasis. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:2823-2829, 2018.
Collapse
Affiliation(s)
- Jeremy A Dubin
- Veterans Administration-Palo Alto, Palo Alto, California
| | - Daniel R Greenberg
- Department of Orthopedic Surgery, Stanford University School of Medicine, 341 Galvez Street Mail Code 6175, Stanford, California
| | - Kag C Iglinski-Benjamin
- Veterans Administration-Palo Alto, Palo Alto, California.,Department of Orthopedic Surgery, Stanford University School of Medicine, 341 Galvez Street Mail Code 6175, Stanford, California
| | - Geoffrey D Abrams
- Veterans Administration-Palo Alto, Palo Alto, California.,Department of Orthopedic Surgery, Stanford University School of Medicine, 341 Galvez Street Mail Code 6175, Stanford, California
| |
Collapse
|
23
|
Wang S, Wen X, Han X, Wang Y, Shen M, Fan S, Zhuang J, Zhang Z, Shan Q, Li M, Hu B, Sun C, Wu D, Lu J, Zheng Y. Repression of microRNA-382 inhibits glomerular mesangial cell proliferation and extracellular matrix accumulation via FoxO1 in mice with diabetic nephropathy. Cell Prolif 2018; 51:e12462. [PMID: 29701296 PMCID: PMC6528942 DOI: 10.1111/cpr.12462] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/08/2018] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Diabetic nephropathy (DN) is a nerve damaging disorder, characterized by glomerular mesangial cell expansion and accumulation of extracellular matrix (ECM) proteins. In this study, we aimed to investigate mesangial cell proliferation and ECM accumulation when promoting or suppressing endogenous miR-382 in glomerular mesangial cells of DN. MATERIALS AND METHODS Model establishment consisted of DN induction by streptozotocin (STZ) in mice. The underlying regulatory mechanisms of miR-382 were analysed in concert with the treatment of miR-382 mimics, miR-382 inhibitors or siRNA against FoxO1 in cultured glomerular mesangial cells isolated from DN mice. RESULTS FoxO1 was identified as the downregulated gene in DN based on the microarray data of GSE1009. We found that miR-382 was significantly upregulated in renal tissues of DN mice and its downregulation dephosphorylated FoxO1, reduced glomerular mesangial cell proliferation and ECM accumulation in vitro. The determination of luciferase activity suggested that miR-382 negatively targeted FoxO1. Expectedly, distinct levels of phosphorylated FoxO1 were observed in the renal cortices of DN mice, while the silencing of FoxO1 was found to increase glomerular mesangial cell proliferation and ECM accumulation in vitro. Reduced glomerular mesangial cell proliferation and ECM accumulation elicited by miR-382 inhibitors were reversed by silencing FoxO1. CONCLUSIONS This study demonstrates miR-382 suppression exerts a potent anti-proliferative effect that may be applied to inhibit glomerular mesangial cell proliferation and ECM accumulation in DN.
Collapse
Affiliation(s)
- Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Xin‐Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Yong‐Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Shao‐Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Juan Zhuang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,School of Environment Science and Spatial InformaticsChina University of Mining and TechnologyXuzhouChina,Jiangsu Key Laboratory for Eco‐Agricultural Biotechnology around Hongze LakeSchool of Life SciencesHuaiyin Normal UniversityHuaianChina
| | - Zi‐Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Meng‐Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Chun‐Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Dong‐Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| | - Yuan‐Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu ProvinceSchool of Life ScienceJiangsu Normal UniversityXuzhouChina,College of Health SciencesJiangsu Normal UniversityXuzhouChina
| |
Collapse
|
24
|
Sarver DC, Kharaz YA, Sugg KB, Gumucio JP, Comerford E, Mendias CL. Sex differences in tendon structure and function. J Orthop Res 2017; 35:2117-2126. [PMID: 28071813 PMCID: PMC5503813 DOI: 10.1002/jor.23516] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/06/2017] [Indexed: 02/04/2023]
Abstract
Tendons play a critical role in the transmission of forces between muscles and bones, and chronic tendon injuries and diseases are among the leading causes of musculoskeletal disability. Little is known about sex-based differences in tendon structure and function. Our objective was to evaluate the mechanical properties, biochemical composition, transcriptome, and cellular activity of plantarflexor tendons from 4 month old male and female C57BL/6 mice using in vitro biomechanics, mass spectrometry-based proteomics, genome-wide expression profiling, and cell culture techniques. While the Achilles tendons of male mice were approximately 6% larger than female mice (p < 0.05), the cell density of female mice was around 19% greater than males (p < 0.05). No significant differences in mechanical properties (p > 0.05) of plantaris tendons were observed. Mass spectrometry proteomics analysis revealed no significant difference between sexes in the abundance of major extracellular matrix (ECM) proteins such as collagen types I (p = 0.30) and III (p = 0.68), but female mice had approximately twofold elevations (p < 0.05) in less abundant ECM proteins such as fibronectin, periostin, and tenascin C. The transcriptome of male and female tendons differed by only 1%. In vitro, neither the sex of the serum that fibroblasts were cultured in, nor the sex of the ECM in which they were embedded, had profound effects on the expression of collagen and cell proliferation genes. Our results indicate that while male mice expectedly had larger tendons, male and female tendons have very similar mechanical properties and biochemical composition, with small increases in some ECM proteins and proteoglycans evident in female tendons. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2117-2126, 2017.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Orthopaedic Surgery, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yalda Ashraf Kharaz
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Kristoffer B Sugg
- Department of Orthopaedic Surgery, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA,Department of Molecular & Integrative Physiology, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA,Department of Surgery, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jonathan P Gumucio
- Department of Orthopaedic Surgery, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA,Department of Molecular & Integrative Physiology, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eithne Comerford
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Christopher L Mendias
- Department of Orthopaedic Surgery, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA,Department of Molecular & Integrative Physiology, Section of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA,Corresponding Author: Christopher L Mendias, PhD, Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 2017, Ann Arbor, MI 48109-2200, 734-764-3250, 734-647-0003 fax,
| |
Collapse
|
25
|
Foolen J, Wunderli SL, Loerakker S, Snedeker JG. Tissue alignment enhances remodeling potential of tendon-derived cells - Lessons from a novel microtissue model of tendon scarring. Matrix Biol 2017. [PMID: 28636876 DOI: 10.1016/j.matbio.2017.06.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tendinopathy is a widespread and unresolved clinical challenge, in which associated pain and hampered mobility present a major cause for work-related disability. Tendinopathy associates with a change from a healthy tissue with aligned extracellular matrix (ECM) and highly polarized cells that are connected head-to-tail, towards a diseased tissue with a disorganized ECM and randomly distributed cells, scar-like features that are commonly attributed to poor innate regenerative capacity of the tissue. A fundamental clinical dilemma with this scarring process is whether treatment strategies should focus on healing the affected (disorganized) tissue or strengthen the remaining healthy (anisotropic) tissue. The question was thus asked whether the intrinsic remodeling capacity of tendon-derived cells depends on the organization of the 3D extracellular matrix (isotropic vs anisotropic). Progress in this field is hampered by the lack of suitable in vitro tissue platforms. We aimed at filling this critical gap by creating and exploiting a next generation tissue platform that mimics aspects of the tendon scarring process; cellular response to a gradient in tissue organization from isotropic (scarred/non-aligned) to highly anisotropic (unscarred/aligned) was studied, as was a transient change from isotropic towards highly anisotropic. Strikingly, cells residing in an 'unscarred' anisotropic tissue indicated superior remodeling capacity (increased gene expression levels of collagen, matrix metalloproteinases MMPs, tissue inhibitors of MMPs), when compared to their 'scarred' isotropic counterparts. A numerical model then supported the hypothesis that cellular remodeling capacity may correlate to cellular alignment strength. This in turn may have improved cellular communication, and could thus relate to the more pronounced connexin43 gap junctions observed in anisotropic tissues. In conclusion, increased tissue anisotropy was observed to enhance the cellular potential for functional remodeling of the matrix. This may explain the poor regenerative capacity of tenocytes in chronic tendinopathy, where the pathological process has resulted in ECM disorganization. Additionally, it lends support to treatment strategies that focus on strengthening the remaining healthy tissue, rather than regenerating scarred tissue.
Collapse
Affiliation(s)
- Jasper Foolen
- Department of Orthopaedics, University Hospital Balgrist, Lengghalde 5, CH-8008 Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Lengghalde 5, CH-8008 Zurich, Switzerland; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Stefania L Wunderli
- Department of Orthopaedics, University Hospital Balgrist, Lengghalde 5, CH-8008 Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Lengghalde 5, CH-8008 Zurich, Switzerland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jess G Snedeker
- Department of Orthopaedics, University Hospital Balgrist, Lengghalde 5, CH-8008 Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Lengghalde 5, CH-8008 Zurich, Switzerland.
| |
Collapse
|
26
|
Reddy S, Hu DQ, Zhao M, Blay E, Sandeep N, Ong SG, Jung G, Kooiker KB, Coronado M, Fajardo G, Bernstein D. miR-21 is associated with fibrosis and right ventricular failure. JCI Insight 2017; 2:91625. [PMID: 28469078 DOI: 10.1172/jci.insight.91625] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Combined pulmonary insufficiency (PI) and stenosis (PS) is a common long-term sequela after repair of many forms of congenital heart disease, causing progressive right ventricular (RV) dilation and failure. Little is known of the mechanisms underlying this combination of preload and afterload stressors. We developed a murine model of PI and PS (PI+PS) to identify clinically relevant pathways and biomarkers of disease progression. Diastolic dysfunction was induced (restrictive RV filling, elevated RV end-diastolic pressures) at 1 month after generation of PI+PS and progressed to systolic dysfunction (decreased RV shortening) by 3 months. RV fibrosis progressed from 1 month (4.4% ± 0.4%) to 3 months (9.2% ± 1%), along with TGF-β signaling and tissue expression of profibrotic miR-21. Although plasma miR-21 was upregulated with diastolic dysfunction, it was downregulated with the onset of systolic dysfunction), correlating with RV fibrosis. Plasma miR-21 in children with PI+PS followed a similar pattern. A model of combined RV volume and pressure overload recapitulates the evolution of RV failure unique to patients with prior RV outflow tract surgery. This progression was characterized by enhanced TGF-β and miR-21 signaling. miR-21 may serve as a plasma biomarker of RV failure, with decreased expression heralding the need for valve replacement.
Collapse
Affiliation(s)
- Sushma Reddy
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Dong-Qing Hu
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Mingming Zhao
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Eddie Blay
- Department of Surgery, Temple University, Philadelphia, Pennsylvania, USA
| | - Nefthi Sandeep
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Sang-Ging Ong
- Cardiovascular Institute, Stanford University, Stanford, California, USA
| | - Gwanghyun Jung
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Kristina B Kooiker
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Michael Coronado
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Giovanni Fajardo
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Daniel Bernstein
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| |
Collapse
|
27
|
Qin LL, An MX, Liu YL, Xu HC, Lu ZQ. MicroRNA-126: a promising novel biomarker in peripheral blood for diabetic retinopathy. Int J Ophthalmol 2017; 10:530-534. [PMID: 28503423 DOI: 10.18240/ijo.2017.04.05] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 12/02/2016] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the content of serum microRNA-126 (miR-126) and its role in screening retinal endothelial injury and early diagnosis of proliferative diabetic retinopathy. METHODS The study included 184 serum samples, 59 samples from healthy individuals, 44 samples from diabetes mellitus (DM) patients without diabetic retinopathy (NDR), 42 from non-proliferative diabetic retinopathy (NPDR) patients and 39 samples from proliferative diabetic retinopathy (PDR) patients. The expression of miR-126 was evaluated using a real-time quantitative polymerase chain reaction. RESULTS The serum content of miR-126 declined as the damage degree in the retina. There was significant difference between the two retinopathy groups (P<0.001). No difference was observed in miR-126 content between healthy individuals and NDR patients (P>0.05). Receiver operating characteristic curve (ROC) analyses indicated that serum miR-126 had significant diagnostic value for PDR. It yielded an area under the curve (AUC) of ROC of 0.976 with 81.21% sensitivity and 90.34% specificity in discriminating PDR from healthy controls, and an AUC of ROC of 0.919 with 84.75% sensitivity and 94.41% specificity in discriminating NDR and NPDR from healthy controls. When the diagnostic threshold was greater than or equal to 8.43, there was an increase in the possibility of NPDR. When the content of miR-126 was less than or equal to 5.02, the possibility of the occurrence of PDR increased. CONCLUSION Serum miR-126 can serve as a non-invasive biomarker for screening retinal endothelial injury and early diagnosis PDR.
Collapse
Affiliation(s)
- Li-Li Qin
- Department of Ophthalmology, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong Province, China
| | - Mei-Xia An
- Department of Ophthalmology, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong Province, China
| | - Yan-Li Liu
- Department of Ophthalmology, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong Province, China
| | - Han-Chun Xu
- Department of Ophthalmology, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong Province, China
| | - Zhi-Qing Lu
- Department of Ophthalmology, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong Province, China
| |
Collapse
|
28
|
Fong G, Backman LJ, Alfredson H, Scott A, Danielson P. The effects of substance P and acetylcholine on human tenocyte proliferation converge mechanistically via TGF-β1. PLoS One 2017; 12:e0174101. [PMID: 28301610 PMCID: PMC5354451 DOI: 10.1371/journal.pone.0174101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/04/2017] [Indexed: 01/10/2023] Open
Abstract
Previous in vitro studies on human tendon cells (tenocytes) have demonstrated that the exogenous administration of substance P (SP) and acetylcholine (ACh) independently result in tenocyte proliferation, which is a prominent feature of tendinosis. Interestingly, the possible link between SP and ACh has not yet been explored in human tenocytes. Recent studies in other cell types demonstrate that both SP and ACh independently upregulate TGF-β1 expression via their respective receptors, the neurokinin 1 receptor (NK-1R) and muscarinic ACh receptors (mAChRs). Furthermore, TGF-β1 has been shown to downregulate NK-1R expression in human keratocytes. The aim of this study was to examine if TGF-β1 is the intermediary player involved in mediating the proliferative pathway shared by SP and ACh in human tenocytes. The results showed that exogenous administration of SP and ACh both caused significant upregulation of TGF-β1 at the mRNA and protein levels. Exposing cells to TGF-β1 resulted in increased cell viability of tenocytes, which was blocked in the presence of the TGFβRI/II kinase inhibitor. In addition, the proliferative effects of SP and ACh on tenocytes were reduced by the TGFβRI/II kinase inhibitor; this supports the hypothesis that the proliferative effects of these signal substances are mediated via the TGF-β axis. Furthermore, exogenous TGF-β1 downregulated NK-1R and mAChRs expression at both the mRNA and protein levels, and these effects were negated by simultaneous exposure to the TGFβRI/II kinase inhibitor, suggesting a negative feedback loop. In conclusion, the results indicate that TGF-β1 is the intermediary player through which the proliferative actions of both SP and ACh converge mechanistically.
Collapse
Affiliation(s)
- Gloria Fong
- Dept. of Integrative Medical Biology, Anatomy, Umeå University, Umeå, Sweden
- Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Ludvig J. Backman
- Dept. of Integrative Medical Biology, Anatomy, Umeå University, Umeå, Sweden
| | - Håkan Alfredson
- Dept. of Community Medicine and Rehabilitation, Sports Medicine, Umeå University, Umeå, Sweden
| | - Alex Scott
- Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Patrik Danielson
- Dept. of Integrative Medical Biology, Anatomy, Umeå University, Umeå, Sweden
- Dept. of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
29
|
Maeda E, Pian H, Ohashi T. Temporal regulation of gap junctional communication between tenocytes subjected to static tensile strain with physiological and non-physiological amplitudes. Biochem Biophys Res Commun 2017; 482:1170-1175. [DOI: 10.1016/j.bbrc.2016.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/02/2016] [Indexed: 01/28/2023]
|
30
|
Peffers MJ, Goljanek-Whysall K, Collins J, Fang Y, Rushton M, Loughlin J, Proctor C, Clegg PD. Decoding the Regulatory Landscape of Ageing in Musculoskeletal Engineered Tissues Using Genome-Wide DNA Methylation and RNASeq. PLoS One 2016; 11:e0160517. [PMID: 27533049 PMCID: PMC4988628 DOI: 10.1371/journal.pone.0160517] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSC) are capable of multipotent differentiation into connective tissues and as such are an attractive source for autologous cell-based regenerative medicine and tissue engineering. Epigenetic mechanisms, like DNA methylation, contribute to the changes in gene expression in ageing. However there was a lack of sufficient knowledge of the role that differential methylation plays during chondrogenic, osteogenic and tenogenic differentiation from ageing MSCs. This study undertook genome level determination of the effects of DNA methylation on expression in engineered tissues from chronologically aged MSCs. We compiled unique DNA methylation signatures from chondrogenic, osteogenic, and tenogenic engineered tissues derived from young; n = 4 (21.8 years ± 2.4 SD) and old; n = 4 (65.5 years±8.3SD) human MSCs donors using the Illumina HumanMethylation 450 Beadchip arrays and compared these to gene expression by RNA sequencing. Unique and common signatures of global DNA methylation were identified. There were 201, 67 and 32 chondrogenic, osteogenic and tenogenic age-related DE protein-coding genes respectively. Findings inferred the nature of the transcript networks was predominantly for 'cell death and survival', 'cell morphology', and 'cell growth and proliferation'. Further studies are required to validate if this gene expression effect translates to cell events. Alternative splicing (AS) was dysregulated in ageing with 119, 21 and 9 differential splicing events identified in chondrogenic, osteogenic and tenogenic respectively, and enrichment in genes associated principally with metabolic processes. Gene ontology analysis of differentially methylated loci indicated age-related enrichment for all engineered tissue types in 'skeletal system morphogenesis', 'regulation of cell proliferation' and 'regulation of transcription' suggesting that dynamic epigenetic modifications may occur in genes associated with shared and distinct pathways dependent upon engineered tissue type. An altered phenotype in engineered tissues was observed with ageing at numerous levels. These changes represent novel insights into the ageing process, with implications for stem cell therapies in older patients. In addition we have identified a number of tissue-dependant pathways, which warrant further studies.
Collapse
Affiliation(s)
- Mandy Jayne Peffers
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst, Chester High Road, Neston, Wirral, UK, CH64 7TE
| | - Katarzyna Goljanek-Whysall
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst, Chester High Road, Neston, Wirral, UK, CH64 7TE
| | - John Collins
- Thurston Arthritis Research Centre, School Of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA, 27599
| | - Yongxiang Fang
- Centre for Genomic Research, Institute of Integrative Biology, Biosciences Building, Crown Street, University of Liverpool, Liverpool, UK, L69 7ZB
| | - Michael Rushton
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK, NE2 4HH
| | - John Loughlin
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK, NE2 4HH
| | - Carole Proctor
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK, NE2 4HH
- Newcastle University Institute for Ageing, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK, NE4 5PL
| | - Peter David Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst, Chester High Road, Neston, Wirral, UK, CH64 7TE
| |
Collapse
|
31
|
Hudgens JL, Sugg KB, Grekin JA, Gumucio JP, Bedi A, Mendias CL. Platelet-Rich Plasma Activates Proinflammatory Signaling Pathways and Induces Oxidative Stress in Tendon Fibroblasts. Am J Sports Med 2016; 44:1931-40. [PMID: 27400714 PMCID: PMC4970921 DOI: 10.1177/0363546516637176] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Tendon injuries are one of the most common musculoskeletal conditions in active patients. Platelet-rich plasma (PRP) has shown some promise in the treatment of tendon disorders, but little is known as to the mechanisms by which PRP can improve tendon regeneration. PRP contains numerous different growth factors and cytokines that activate various cellular signaling cascades, but it has been difficult to determine precisely which signaling pathways and cellular responses are activated after PRP treatment. Additionally, macrophages play an important role in modulating tendon regeneration, but the influence of PRP on determining whether macrophages assume a proinflammatory or anti-inflammatory phenotype remains unknown. PURPOSE To use genome-wide expression profiling, bioinformatics, and protein analysis to determine the cellular pathways activated in fibroblasts treated with PRP. The effect of PRP on macrophage polarization was also evaluated. STUDY DESIGN Controlled laboratory study. METHODS Tendon fibroblasts or macrophages from rats were cultured and treated with either platelet-poor plasma (PPP) or PRP. RNA or protein was isolated from cells and analyzed using microarrays, quantitative polymerase chain reaction, immunoblotting, or bioinformatics techniques. RESULTS Pathway analysis determined that the most highly induced signaling pathways in PRP-treated tendon fibroblasts were TNFα and NFκB pathways. PRP also downregulated the expression of extracellular matrix genes and induced the expression of autophagy-related genes and reactive oxygen species (ROS) genes and protein markers in tendon fibroblasts. PRP failed to have a major effect on markers of macrophage polarization. CONCLUSION PRP induces an inflammatory response in tendon fibroblasts, which leads to the formation of ROS and the activation of oxidative stress pathways. PRP does not appear to significantly modulate macrophage polarization. CLINICAL RELEVANCE PRP might act by inducing a transient inflammatory event, which could then trigger a tissue regeneration response.
Collapse
Affiliation(s)
- Joshua L Hudgens
- Departments of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Kristoffer B Sugg
- Departments of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109,Departments of Molecular & Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109,Departments of Surgery, Section of Plastic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Jeremy A Grekin
- Departments of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Jonathan P Gumucio
- Departments of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109,Departments of Molecular & Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Asheesh Bedi
- Departments of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Christopher L Mendias
- Departments of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109,Departments of Molecular & Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109,To whom correspondence should be addressed: Christopher L Mendias, PhD, ATC, Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 2017, Ann Arbor, MI 48109-2200, 734-764-3250,
| |
Collapse
|
32
|
The role of TGF-ß1 in osteoarthritis of the temporomandibular joint in two genetic mouse models. Arch Oral Biol 2016; 67:68-73. [DOI: 10.1016/j.archoralbio.2016.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/16/2016] [Accepted: 03/13/2016] [Indexed: 11/20/2022]
|
33
|
El-Ahwany E, Nagy F, Zoheiry M, Shemis M, Nosseir M, Taleb HA, El Ghannam M, Atta R, Zada S. Circulating miRNAs as Predictor Markers for Activation of Hepatic Stellate Cells and Progression of HCV-Induced Liver Fibrosis. Electron Physician 2016; 8:1804-10. [PMID: 26955452 PMCID: PMC4768932 DOI: 10.19082/1804] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 12/28/2015] [Indexed: 12/17/2022] Open
Abstract
Introduction Liver fibrosis is the excessive accumulation of extracellular matrix that occurs by activation of hepatic stellate cells (HSCs), which has been identified as the major driver of liver fibrosis. Several studies confirmed that miRNAs have regulatory effects on the activation of HSCs by affecting the signaling pathways. The aim of this study was to develop non-invasive diagnostic markers by measuring different circulating miRNAs in serum as predictor markers for early diagnosis of liver fibrosis and its progression. Methods In this case-control study, we enrolled 66 subjects with chronic hepatitis C (CHC) with early stage of fibrosis and 65 subjects with CHC with late-stage fibrosis. Also, 40 subjects were included as normal controls. The six main miRNAs, i.e., miR-138, miR-140, miR-143, miR-325, miR-328, and miR-349, were measured using the reverse transcription-polymerase chain reaction. Results In the cases of CHC both with early and late stage of fibrosis, the circulating levels of the six main miRNAs were significantly higher than the levels in the control group. ROC analysis indicated that the sensitivity and specificity of miR-138 were 89.3% and 71.43%, respectively, in the early stage of fibrosis. In the late stage, the sensitivity and specificity of miR-138 were 89.3 and 93.02%, respectively, whereas, for miR-143, they were 75.0 and 88.4%, respectively. Conclusions Circulating miR-138 could serve as a non-invasive biomarker for the detection of early fibrosis. Also, miR-138 and miR-143 could be specific biomarkers for indicating the late stage of liver fibrosis.
Collapse
Affiliation(s)
- Eman El-Ahwany
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Faten Nagy
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mona Zoheiry
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mohamed Shemis
- Biochemistry Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mona Nosseir
- Pathology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Hoda Abu Taleb
- Environmental Research Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Maged El Ghannam
- Gastroenterology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Rafaat Atta
- Gastroenterology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Suher Zada
- Biology Department, American University in Cairo, Cairo, Egypt
| |
Collapse
|
34
|
Tenogenic differentiation of mesenchymal stem cells and noncoding RNA: From bench to bedside. Exp Cell Res 2015; 341:237-42. [PMID: 26724570 DOI: 10.1016/j.yexcr.2015.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 11/21/2022]
Abstract
Tendon is a critical unit of musculoskeletal system that connects muscle to bone to control bone movement. More population participate in physical activities, tendon injuries, such as acute tendon rupture and tendinopathy due to overuse, are common causing unbearable pain and disability. However, the process of tendon development and the pathogenesis of tendinopathy are not well defined, limiting the development of clinical therapy for tendon injuries. Studying the tendon differentiation control pathways may help to develop novel therapeutic strategies. This review summarized the novel molecular and cellular events in tendon development and highlighted the clinical application potential of non-coding RNAs and tendon-derived stem cells in gene and cell therapy for tendon injuries, which may bring insights into research and new therapy for tendon disorders.
Collapse
|
35
|
Mohamed JS, Hajira A, Lopez MA, Boriek AM. Genome-wide Mechanosensitive MicroRNA (MechanomiR) Screen Uncovers Dysregulation of Their Regulatory Networks in the mdm Mouse Model of Muscular Dystrophy. J Biol Chem 2015; 290:24986-5011. [PMID: 26272747 DOI: 10.1074/jbc.m115.659375] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Indexed: 11/06/2022] Open
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of genetic and neuromuscular disorders, which result in severe loss of motor ability and skeletal muscle mass and function. Aberrant mechanotransduction and dysregulated-microRNA pathways are often associated with the progression of MD. Here, we hypothesized that dysregulation of mechanosensitive microRNAs (mechanomiRs) in dystrophic skeletal muscle plays a major role in the progression of MD. To test our hypothesis, we performed a genome-wide expression profile of anisotropically regulated mechanomiRs and bioinformatically analyzed their target gene networks. We assessed their functional roles in the advancement of MD using diaphragm muscles from mdm (MD with myositis) mice, an animal model of human tibial MD (titinopathy), and their wild-type littermates. We were able to show that ex vivo anisotropic mechanical stretch significantly alters the miRNA expression profile in diaphragm muscles from WT and mdm mice; as a result, some of the genes associated with MDs are dysregulated in mdm mice due to differential regulation of a distinct set of mechanomiRs. Interestingly, we found a contrasting expression pattern of the highly expressed let-7 family mechanomiRs, let-7e-5p and miR-98-5p, and their target genes associated with the extracellular matrix and TGF-β pathways, respectively, between WT and mdm mice. Gain- and loss-of-function analysis of let-7e-5p in myocytes isolated from the diaphragms of WT and mdm mice confirmed Col1a1, Col1a2, Col3a1, Col24a1, Col27a1, Itga1, Itga4, Scd1, and Thbs1 as target genes of let-7e-5p. Furthermore, we found that miR-98 negatively regulates myoblast differentiation. Our study therefore introduces additional biological players in the regulation of skeletal muscle structure and myogenesis that may contribute to unexplained disorders of MD.
Collapse
Affiliation(s)
- Junaith S Mohamed
- From the Pulmonary and Critical Care Section, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Ameena Hajira
- From the Pulmonary and Critical Care Section, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Michael A Lopez
- From the Pulmonary and Critical Care Section, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Aladin M Boriek
- From the Pulmonary and Critical Care Section, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
36
|
Freedman BR, Bade ND, Riggin CN, Zhang S, Haines PG, Ong KL, Janmey PA. The (dys)functional extracellular matrix. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3153-64. [PMID: 25930943 DOI: 10.1016/j.bbamcr.2015.04.015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/11/2015] [Accepted: 04/13/2015] [Indexed: 10/23/2022]
Abstract
The extracellular matrix (ECM) is a major component of the biomechanical environment with which cells interact, and it plays important roles in both normal development and disease progression. Mechanical and biochemical factors alter the biomechanical properties of tissues by driving cellular remodeling of the ECM. This review provides an overview of the structural, compositional, and mechanical properties of the ECM that instruct cell behaviors. Case studies are reviewed that highlight mechanotransduction in the context of two distinct tissues: tendons and the heart. Although these two tissues demonstrate differences in relative cell-ECM composition and mechanical environment, they share similar mechanisms underlying ECM dysfunction and cell mechanotransduction. Together, these topics provide a framework for a fundamental understanding of the ECM and how it may vary across normal and diseased tissues in response to mechanical and biochemical cues. This article is part of a Special Issue entitled: Mechanobiology.
Collapse
Affiliation(s)
- Benjamin R Freedman
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathan D Bade
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Corinne N Riggin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sijia Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip G Haines
- Division of Cardiovascular Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katy L Ong
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Janmey
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
37
|
MicroRNA29a regulates IL-33-mediated tissue remodelling in tendon disease. Nat Commun 2015; 6:6774. [PMID: 25857925 PMCID: PMC4403384 DOI: 10.1038/ncomms7774] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 02/25/2015] [Indexed: 01/04/2023] Open
Abstract
MicroRNA (miRNA) has the potential for cross-regulation and functional integration of discrete biological processes during complex physiological events. Utilizing the common human condition tendinopathy as a model system to explore the cross-regulation of immediate inflammation and matrix synthesis by miRNA we observed that elevated IL-33 expression is a characteristic of early tendinopathy. Using in vitro tenocyte cultures and in vivo models of tendon damage, we demonstrate that such IL-33 expression plays a pivotal role in the transition from type 1 to type 3 collagen (Col3) synthesis and thus early tendon remodelling. Both IL-33 effector function, via its decoy receptor sST2, and Col3 synthesis are regulated by miRNA29a. Downregulation of miRNA29a in human tenocytes is sufficient to induce an increase in Col3 expression. These data provide a molecular mechanism of miRNA-mediated integration of the early pathophysiologic events that facilitate tissue remodelling in human tendon after injury.
Collapse
|
38
|
Wei FL, Wang JH, Ding G, Yang SY, Li Y, Hu YJ, Wang SL. Mechanical force-induced specific MicroRNA expression in human periodontal ligament stem cells. Cells Tissues Organs 2015; 199:353-63. [PMID: 25823370 DOI: 10.1159/000369613] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2014] [Indexed: 01/14/2023] Open
Abstract
It remains unclear how the expression of microRNAs (miRNAs) in human periodontal ligament stem cells (PDLSCs) might respond to mechanical stretch. To investigate specific miRNA expression in stretched PDLSCs, we used a Flexcell® FX-5000™ tension system to achieve external mechanical stimulation. Then, a custom-designed microarray assay was performed to investigate and describe the genome-wide differential expression of miRNAs in normal and stretched PDLSCs. Finally, we implemented integrative miRNA target prediction and network analysis approaches to construct an interaction network of the key miRNAs and their putative targets. We found that stretching induced morphological changes and increased alkaline phosphatase (ALP) activity, runt-related transcription factor 2 (RUNX2), osteocalcin (OCN), and bone sialoprotein (BSP) expression in PDLSCs. The microarray data showed that 53 miRNAs were differentially expressed with stretching. With an interaction network, we examined the connections between 10 selected key miRNAs and their putative target genes, which were related to mechanical force. The results from the interaction network provided a basis for postulating the functional roles of miRNAs in PDLSCs.
Collapse
Affiliation(s)
- F L Wei
- Department of Orthodontics, Shandong Provincial Key Laboratory of Oral Biomedicine, School of Stomatology, Shandong University, Jinan, PR China
| | | | | | | | | | | | | |
Collapse
|
39
|
p38 MAPK signaling in postnatal tendon growth and remodeling. PLoS One 2015; 10:e0120044. [PMID: 25768932 PMCID: PMC4359143 DOI: 10.1371/journal.pone.0120044] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/03/2015] [Indexed: 12/16/2022] Open
Abstract
Tendon is a dynamic tissue whose structure and function is influenced by mechanical loading, but little is known about the fundamental mechanisms that regulate tendon growth and remodeling in vivo. Data from cultured tendon fibroblasts indicated that the p38 MAPK pathway plays an important role in tendon fibroblast proliferation and collagen synthesis in vitro. To gain greater insight into the mechanisms of tendon growth, and explore the role of p38 MAPK signaling in this process, we tested the hypotheses that inducing plantaris tendon growth through the ablation of the synergist Achilles tendon would result in rapid expansion of a neotendon matrix surrounding the original tendon, and that treatment with the p38 MAPK inhibitor SB203580 would prevent this growth. Rats were treated with vehicle or SB203580, and subjected to synergist ablation by bilateral tenectomy of the Achilles tendon. Changes in histological and biochemical properties of plantaris tendons were analyzed 3, 7, or 28 days after overload, and comparisons were made to non-overloaded animals. By 28 days after overload, tendon mass had increased by 30% compared to non-overloaded samples, and cross-sectional area (CSA) increased by around 50%, with most of the change occurring in the neotendon. The expansion in CSA initially occurred through the synthesis of a hyaluronic acid rich matrix that was progressively replaced with mature collagen. Pericytes were present in areas of active tendon growth, but never in the original tendon ECM. Inhibition of p38 MAPK resulted in a profound decrease in IL6 expression, and had a modest effect on the expression of other ECM and cell proliferation genes, but had a negligible impact on overall tendon growth. The combined results from this study provided novel insights into tendon mechanobiology, and suggest that p38 MAPK signaling does not appear to be necessary for tendon growth in vivo.
Collapse
|
40
|
Mendias CL, Lynch EB, Gumucio JP, Flood MD, Rittman DS, Van Pelt DW, Roche SM, Davis CS. Changes in skeletal muscle and tendon structure and function following genetic inactivation of myostatin in rats. J Physiol 2015; 593:2037-52. [PMID: 25640143 DOI: 10.1113/jphysiol.2014.287144] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/23/2015] [Indexed: 11/08/2022] Open
Abstract
Myostatin is a negative regulator of skeletal muscle and tendon mass. Myostatin deficiency has been well studied in mice, but limited data are available on how myostatin regulates the structure and function of muscles and tendons of larger animals. We hypothesized that, in comparison to wild-type (MSTN(+/+) ) rats, rats in which zinc finger nucleases were used to genetically inactivate myostatin (MSTN(Δ/Δ) ) would exhibit an increase in muscle mass and total force production, a reduction in specific force, an accumulation of type II fibres and a decrease and stiffening of connective tissue. Overall, the muscle and tendon phenotype of myostatin-deficient rats was markedly different from that of myostatin-deficient mice, which have impaired contractility and pathological changes to fibres and their extracellular matrix. Extensor digitorum longus and soleus muscles of MSTN(Δ/Δ) rats demonstrated 20-33% increases in mass, 35-45% increases in fibre number, 20-57% increases in isometric force and no differences in specific force. The insulin-like growth factor-1 pathway was activated to a greater extent in MSTN(Δ/Δ) muscles, but no substantial differences in atrophy-related genes were observed. Tendons of MSTN(Δ/Δ) rats had a 20% reduction in peak strain, with no differences in mass, peak stress or stiffness. The general morphology and gene expression patterns were similar between tendons of both genotypes. This large rodent model of myostatin deficiency did not have the negative consequences to muscle fibres and extracellular matrix observed in mouse models, and suggests that the greatest impact of myostatin in the regulation of muscle mass may not be to induce atrophy directly, but rather to block hypertrophy signalling.
Collapse
Affiliation(s)
- Christopher L Mendias
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Chen Q, Lu H, Yang H. Chitosan inhibits fibroblasts growth in Achilles tendon via TGF-β1/Smad3 pathway by miR-29b. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8462-8470. [PMID: 25674210 PMCID: PMC4313955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/01/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND AND AIM Chitosan, is a natural polymer, plays an important role in prevention of tendon adhesion in tendon healing process. However, the molecular mechanisms underlying the prevention effect is unclear. Here we investigated the effects of chitosan on Achilles tendon injury rats and fibroblasts. METHODS Eight weeks after surgery, gliding excursion and the content of collagen fibers in Achilles tendon injury rats were determined to evaluate the chitosan effect on tendon healing. Fibroblasts isolated from scar tissue of repaired tendon were treated with different concentration of chitosan, and then cell inhibition, apoptosis and cell cycle were measured using MTT and Flow Cytometry respectively. The expression of microRNAs (miRNAs) was quantified by real-time PCR and protein expression of TGF-β1, Smad3 and P21 were quantified by western blotting. MiR-29b inhibitor was transfected in cells to evaluate the mechanism underlying the effects of chitosan on tendon fibroblasts. RESULTS The gliding excursion of repaired tendon was increased and the content of collagen fibers was decreased by chitosan in rats. Chitosan inhibited the fibroblasts growth and arrested cells in G1 phase. Chitosan also elevated the expression of miR-29b and P21 while reduced the levels of TGFβ1 and Smad3 in both repaired tendon and fibroblasts. In addition, miR-29b inhibitor revered the effects of chitosan on fibroblasts. CONCLUSIONS The current study demonstrated that chitosan improving the condition of tendon healing after surgery, which is reduced by the high expression of miR-29b and its down-regulation of TGF-β1/Smad3 level and inhibition of fibroblasts growth.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Hand Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University China
| | - Hui Lu
- Department of Hand Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University China
| | - Hu Yang
- Department of Hand Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University China
| |
Collapse
|
42
|
RUNX1 is essential for mesenchymal stem cell proliferation and myofibroblast differentiation. Proc Natl Acad Sci U S A 2014; 111:16389-94. [PMID: 25313057 DOI: 10.1073/pnas.1407097111] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Myofibroblasts are a key cell type in wound repair, cardiovascular disease, and fibrosis and in the tumor-promoting microenvironment. The high accumulation of myofibroblasts in reactive stroma is predictive of the rate of cancer progression in many different tumors, yet the cell types of origin and the mechanisms that regulate proliferation and differentiation are unknown. We report here, for the first time to our knowledge, the characterization of normal human prostate-derived mesenchymal stem cells (MSCs) and the TGF-β1-regulated pathways that modulate MSC proliferation and myofibroblast differentiation. Human prostate MSCs combined with prostate cancer cells expressing TGF-β1 resulted in commitment to myofibroblasts. TGF-β1-regulated runt-related transcription factor 1 (RUNX1) was required for cell cycle progression and proliferation of progenitors. RUNX1 also inhibited, yet did not block, differentiation. Knockdown of RUNX1 in prostate or bone marrow-derived MSCs resulted in cell cycle arrest, attenuated proliferation, and constitutive differentiation to myofibroblasts. These data show that RUNX1 is a key transcription factor for MSC proliferation and cell fate commitment in myofibroblast differentiation. This work also shows that the normal human prostate gland contains tissue-derived MSCs that exhibit multilineage differentiation similar to bone marrow-derived MSCs. Targeting RUNX1 pathways may represent a therapeutic approach to affect myofibroblast proliferation and biology in multiple disease states.
Collapse
|
43
|
Gumucio JP, Phan AC, Ruehlmann DG, Noah AC, Mendias CL. Synergist ablation induces rapid tendon growth through the synthesis of a neotendon matrix. J Appl Physiol (1985) 2014; 117:1287-91. [PMID: 25277742 DOI: 10.1152/japplphysiol.00720.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mechanical loading can increase tendon cross-sectional area (CSA), but the mechanisms by which this occurs are largely unknown. To gain a greater understanding of the cellular mechanisms of adult tendon growth in response to mechanical loading, we used a synergist ablation model whereby a tenectomy of the Achilles tendon was performed to induce growth of the synergist plantaris tendon. We hypothesized that after synergist ablation progenitor cells in the epitenon would proliferate and increase the size of the existing tendon matrix. Adult male mice were subjected to a bilateral Achilles tenectomy, and plantaris tendons were isolated from mice at 0, 2, 7, 14, and 28 days after surgery. Tendons were sectioned stained with either fast green and hematoxylin, prepared for fluorescent microscopy, or prepared for gene expression of scleraxis and type I collagen. After overload, there was a dramatic increase in total CSA of tendons, whereas the size of the original tendon matrix was not changed. Growth primarily occurred through the formation of a neotendon matrix between the original tendon and the epitenon, and contained cells that were proliferative and scleraxis positive. Additionally, an initial expansion of fibroblast cells occurred before the synthesis of new extracellular matrix. Fibroblasts in the original tendon did not re-enter the cell cycle. The results from this study provide new insight into the mechanisms of tendon growth, indicate tendon consists mostly of postmitotic cells, and that growth of tendon primarily occurs from the most superficial layers outward.
Collapse
Affiliation(s)
- Jonathan P Gumucio
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan and Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Anthony C Phan
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan and
| | - David G Ruehlmann
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan and
| | - Andrew C Noah
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan and
| | - Christopher L Mendias
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan and Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
44
|
Liu H, Zhu S, Zhang C, Lu P, Hu J, Yin Z, Ma Y, Chen X, OuYang H. Crucial transcription factors in tendon development and differentiation: their potential for tendon regeneration. Cell Tissue Res 2014; 356:287-98. [PMID: 24705622 DOI: 10.1007/s00441-014-1834-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/30/2014] [Indexed: 12/22/2022]
Abstract
Tendons that connect muscles to bone are often the targets of sports injuries. The currently unsatisfactory state of tendon repair is largely attributable to the limited understanding of basic tendon biology. A number of tendon lineage-related transcription factors have recently been uncovered and provide clues for the better understanding of tendon development. Scleraxis and Mohawk have been identified as critical transcription factors in tendon development and differentiation. Other transcription factors, such as Sox9 and Egr1/2, have also been recently reported to be involved in tendon development. However, the molecular mechanisms and application of these transcription factors remain largely unclear and this prohibits their use in tendon therapy. Here, we systematically review and analyze recent findings and our own data concerning tendon transcription factors and tendon regeneration. Based on these findings, we provide interaction and temporal programming maps of transcription factors, as a basis for future tendon therapy. Finally, we discuss future directions for tendon regeneration with differentiation and trans-differentiation approaches based on transcription factors.
Collapse
Affiliation(s)
- Huanhuan Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou, 310058, China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Freedman BR, Gordon JA, Soslowsky LJ. The Achilles tendon: fundamental properties and mechanisms governing healing. Muscles Ligaments Tendons J 2014; 4:245-255. [PMID: 25332943 PMCID: PMC4187594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This review highlights recent research on Achilles tendon healing, and comments on the current clinical controversy surrounding the diagnosis and treatment of injury. The processes of Achilles tendon healing, as demonstrated through changes in its structure, composition, and biomechanics, are reviewed. Finally, a review of tendon developmental biology and mechano transductive pathways is completed to recognize recent efforts to augment injured Achilles tendons, and to suggest potential future strategies for therapeutic intervention and functional tissue engineering. Despite an abundance of clinical evidence suggesting that current treatments and rehabilitation strategies for Achilles tendon ruptures are equivocal, significant questions remain to fully elucidate the basic science mechanisms governing Achilles tendon injury, healing, treatment, and rehabilitation.
Collapse
Affiliation(s)
| | | | - Louis J. Soslowsky
- Corresponding Author: Louis J. Soslowsky, McKay Orthopaedic Research Laboratory, University of Pennsylvania, 424 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104-6081, USA, E-mail:
| |
Collapse
|
46
|
Li J, Ohliger J, Pei M. Significance of epigenetic landscape in cartilage regeneration from the cartilage development and pathology perspective. Stem Cells Dev 2014; 23:1178-94. [PMID: 24555773 DOI: 10.1089/scd.2014.0002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Regenerative therapies for cartilage defects have been greatly advanced by progress in both the stem cell biology and tissue engineering fields. Despite notable successes, significant barriers remain including shortage of autologous cell sources and generation of a stable chondrocyte phenotype using progenitor cells. Increasing demands for the treatment of degenerative diseases, such as osteoarthritis and rheumatoid arthritis, highlight the importance of epigenetic remodeling in cartilage regeneration. Epigenetic regulatory mechanisms, such as microRNAs, DNA methylation, and histone modifications, have been intensively studied due to their direct regulatory role on gene expression. However, a thorough understanding of the environmental factors that initiate these epigenetic events may provide greater insight into the prevention of degenerative diseases and improve the efficacy of treatments. In other words, if we could identify a specific factor from the environment and its downstream signaling events, then we could stop or retard degradation and enhance cartilage regeneration. A more operational definition of epigenetic remodeling has recently been proposed by categorizing the signals during the epigenetic process into epigenators, initiators, and maintainers. This review seeks to compile and reorganize the existing literature pertaining to epigenetic remodeling events placing emphasis on perceiving the landscape of epigenetic mechanisms during cartilage regeneration with the new operational definition, especially from the environmental factors' point of view. Progress in understanding epigenetic regulatory mechanisms could benefit cartilage regeneration and engineering on a larger scale and provide more promising therapeutic applications.
Collapse
Affiliation(s)
- Jingting Li
- 1 Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University , Morgantown, West Virginia
| | | | | |
Collapse
|
47
|
Holladay C, Abbah SA, O'Dowd C, Pandit A, Zeugolis DI. Preferential tendon stem cell response to growth factor supplementation. J Tissue Eng Regen Med 2014; 10:783-98. [PMID: 24474722 DOI: 10.1002/term.1852] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 09/30/2013] [Accepted: 11/06/2013] [Indexed: 12/14/2022]
Abstract
Tendon injuries are increasingly prevalent around the world, accounting for more than 100 000 new clinical cases/year in the USA alone. Cell-based therapies have been proposed as a therapeutic strategy, with recent data advocating the use of tendon stem cells (TSCs) as a potential cell source with clinical relevance for tendon regeneration. However, their in vitro expansion is problematic, as they lose their multipotency and change their protein expression profile in culture. Herein, we ventured to assess the influence of insulin-like growth factor 1 (IGF-1), growth and differentiation factor-5 (GDF-5) and transforming growth factor-β1 (TGFβ1) supplementation in TSC culture. IGF-1 preserved multipotency for up to 28 days. Upregulation of decorin and scleraxis expression was observed as compared to freshly isolated cells. GDF-5 treated cells exhibited reduced differentiation along adipogenic and chondrogenic pathways after 28 days, and decorin, scleraxis and collagen type I expression was increased. After 28 days, TGFβ1 supplementation led to increased scleraxis, osteonectin and collagen type II expression. The varied responses to each growth factor may reflect their role in tendon repair, suggesting that: GDF-5 promotes the transition of tendon stem cells towards tenocytes; TGFβ1 induces differentiation along several pathways, including a phenotype indicative of fibrocartilage or calcified tendon, common problems in tendon healing; and IGF-1 promotes proliferation and maintenance of TSC phenotypes, thereby creating a population sufficient to have a beneficial effect. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Carolyn Holladay
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland Galway (NUI Galway), Ireland.,Vornia Biomaterials, Galway, Ireland
| | - Sunny-Akogwu Abbah
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland Galway (NUI Galway), Ireland
| | | | - Abhay Pandit
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland Galway (NUI Galway), Ireland
| | - Dimitrios I Zeugolis
- Network of Excellence for Functional Biomaterials (NFB), National University of Ireland Galway (NUI Galway), Ireland.
| |
Collapse
|
48
|
Barnette DN, Hulin A, Ahmed ASI, Colige AC, Azhar M, Lincoln J. Tgfβ-Smad and MAPK signaling mediate scleraxis and proteoglycan expression in heart valves. J Mol Cell Cardiol 2013; 65:137-46. [PMID: 24157418 DOI: 10.1016/j.yjmcc.2013.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/10/2013] [Accepted: 10/09/2013] [Indexed: 11/17/2022]
Abstract
Mature heart valves are complex structures consisting of three highly organized extracellular matrix layers primarily composed of collagens, proteoglycans and elastin. Collectively, these diverse matrix components provide all the necessary biomechanical properties for valve function throughout life. In contrast to healthy valves, myxomatous valve disease is the most common cause of mitral valve prolapse in the human population and is characterized by an abnormal abundance of proteoglycans within the valve tri-laminar structure. Despite the clinical significance, the etiology of this phenotype is not known. Scleraxis (Scx) is a basic-helix-loop-helix transcription factor that we previously showed to be required for establishing heart valve structure during remodeling stages of valvulogenesis. In this study, we report that remodeling heart valves from Scx null mice express decreased levels of proteoglycans, particularly chondroitin sulfate proteoglycans (CSPGs), while overexpression in embryonic avian valve precursor cells and adult porcine valve interstitial cells increases CSPGs. Using these systems we further identify that Scx is positively regulated by canonical Tgfβ2 signaling during this process and this is attenuated by MAPK activity. Finally, we show that Scx is increased in myxomatous valves from human patients and mouse models, and overexpression in human mitral valve interstitial cells modestly increases proteoglycan expression consistent with myxomatous mitral valve phenotypes. Together, these studies identify an important role for Scx in regulating proteoglycans in embryonic and mature valve cells and suggest that imbalanced regulation could influence myxomatous pathogenesis.
Collapse
Affiliation(s)
- Damien N Barnette
- Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, P.O. Box 016189 (R-189), Miami, FL, USA; Center for Cardiovascular and Pulmonary Research at Nationwide Children's Hospital Research Institute, 700 Children's Drive, Columbus, OH 43205, USA; The Heart Center at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | | | | | | | | | | |
Collapse
|
49
|
Gumucio JP, Flood MD, Phan AC, Brooks SV, Mendias CL. Targeted inhibition of TGF-β results in an initial improvement but long-term deficit in force production after contraction-induced skeletal muscle injury. J Appl Physiol (1985) 2013; 115:539-45. [PMID: 23766498 DOI: 10.1152/japplphysiol.00374.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a proinflammatory cytokine that regulates the response of many tissues following injury. Previous studies in our lab have shown that treating muscles with TGF-β results in a dramatic accumulation of type I collagen, substantial fiber atrophy, and a marked decrease in force production. Because TGF-β promotes atrophy and fibrosis, our objective was to investigate whether the inhibition of TGF-β after injury would enhance the recovery of muscle following injury. We hypothesized that inhibiting TGF-β after contraction-induced injury would improve the functional recovery of muscles by preventing muscle fiber atrophy and weakness, and by limiting the accumulation of fibrotic scar tissue. To test this hypothesis, we induced an injury using a series of in situ lengthening contractions to extensor digitorum longus muscles of mice treated with either a bioneutralizing antibody against TGF-β or a sham antibody. Compared with controls, muscles from mice receiving TGF-β inhibitor showed a greater recovery in force 3 days and 7 days after injury but had a decrease in force compared with controls at the 21-day time point. The early enhancement in force in the TGF-β inhibitor group was associated with an initial improvement in tissue morphology, but, at 21 days, while the control group was fully recovered, the TGF-β inhibitor group displayed an irregular extracellular matrix and an increase in atrogin-1 gene expression. These results indicate that the inhibition of TGF-β promotes the early recovery of muscle function but is detrimental overall to full muscle recovery following moderate to severe muscle injuries.
Collapse
Affiliation(s)
- Jonathan P Gumucio
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
Sarcopenia is one of the leading causes of disability in the elderly. Despite the growing prevalence of sarcopenia, the molecular mechanisms that control aging-related changes in muscle mass are not fully understood. The ubiquitin proteasome system is one of the major pathways that regulate muscle protein degradation, and this system plays a central role in controlling muscle size. Atrogin-1 and MuRF-1 are two E3 ubiquitin ligases that are important regulators of ubiquitin-mediated protein degradation in skeletal muscle. In this review, we will discuss: (i) aging-related changes to skeletal muscle structure and function; (ii) the regulation of protein synthesis and protein degradation by IGF-1, TGF-β, and myostatin, with emphasis on the control of atrogin-1 and MuRF-1 expression; and (iii) the potential for modulating atrogin-1 and MuRF-1 expression to treat or prevent sarcopenia.
Collapse
Affiliation(s)
- Jonathan P Gumucio
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 2017, Ann Arbor, MI, 48109-2200, USA
| | | |
Collapse
|