1
|
Eliasberg CD, Carballo CB, J H Yao V, Piacentini A, Sanchez LA, Li TM, Havasy J, Bowen EG, Khan M, Rodeo SA. Evaluation of Novel Therapeutic Agents for Modulation of the Inflammatory Response in Rotator Cuff Tendinopathy Utilizing a Mouse Model of Subacromial Impingement. Am J Sports Med 2025; 53:1317-1327. [PMID: 40159684 DOI: 10.1177/03635465251330008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
BACKGROUND Rotator cuff disease is prevalent and can cause significant disability. Local subacromial corticosteroid injections (CSIs) have been a mainstay of nonoperative management despite concerns regarding their potential for adverse effects on tendon and muscle tissue. We identified 3 potential molecular targets that could be inhibited by alternative, currently available treatments. PURPOSE To compare the effects of losartan (LOS), anakinra (AK), and alpha-2-macroglobulin (A2M), not previously utilized in the treatment of rotator cuff disease, with CSI-treated and nontreated controls in a murine model of rotator cuff tendinopathy. STUDY DESIGN Controlled laboratory study. METHODS A total of 90 twelve-week-old male C57BL/6J mice were placed into 6 different groups (n = 15 mice per group). Group 1 underwent a sham procedure with no treatment (sham controls). Group 2 underwent placement of a metal clip in bilateral shoulders to induce impingement on the rotator cuff but received no further treatment. All mice receiving treatments (groups 3-6) underwent bilateral clip impingement surgery on day 0. On day 21, subacromial injections of CSI, AK, or A2M were performed, and LOS was administered via drinking water. All mice were sacrificed at 6 weeks after the initial impingement surgery. The supraspinatus tendons and muscles were harvested. Histology, biomechanical testing, flow cytometry, gene expression, and gait analysis were performed. The significance level was set at P = .05 for all statistical analyses. RESULTS Flow cytometry demonstrated that treatment groups exhibited individual stromal cell marker profiles more similar to sham controls than to the impingement control groups, with significantly higher percentages of CD51+, CD73+, CD90.2+, CD105+, and CD146+ cells compared with the impingement control group. Gene expression analysis demonstrated significantly decreased pathway scores for cytokine signaling, inflammasome, phagocytic cell function, oxidative stress, and proteotoxic stress in the treatment groups compared with the impingement control group. CONCLUSION These novel therapeutic agents may have utility in promoting a favorable environment for stromal progenitor cells and decreasing cytokine signaling, inflammatory responses, and stress pathways associated with subacromial impingement. CLINICAL RELEVANCE Further investigation into these agents and the underlying cellular and molecular mechanisms of inflammation may allow for the utilization of alternatives to CSIs.
Collapse
Affiliation(s)
- Claire D Eliasberg
- Sports Medicine Institute, Department of Orthopaedic Surgery, Hospital for Special Surgery, New York, New York, USA
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery Research Institute, New York, New York, USA
| | - Camila B Carballo
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery Research Institute, New York, New York, USA
| | - Vincent J H Yao
- City University of New York, School of Medicine, New York, New York, USA
| | - Alexander Piacentini
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery Research Institute, New York, New York, USA
| | | | - Thomas M Li
- University of California, San Francisco, San Francisco, California, USA
| | - Janice Havasy
- Long Island Jewish Medical Center/North Shore University Hospital, New York, New York, USA
| | | | - Marjan Khan
- University of Florida, Gainesville, Florida, USA
| | - Scott A Rodeo
- Sports Medicine Institute, Department of Orthopaedic Surgery, Hospital for Special Surgery, New York, New York, USA
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery Research Institute, New York, New York, USA
| |
Collapse
|
2
|
Yin K, Zhang C, Deng Z, Wei X, Xiang T, Yang C, Chen C, Chen Y, Luo F. FAPs orchestrate homeostasis of muscle physiology and pathophysiology. FASEB J 2024; 38:e70234. [PMID: 39676717 PMCID: PMC11647758 DOI: 10.1096/fj.202400381r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 10/26/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024]
Abstract
As a common clinical manifestation, muscle weakness is prevalent in people with mobility disorders. Further studies of muscle weakness have found that patients with muscle weakness present with persistent muscle inflammation, loss of muscle fibers, fat infiltration, and interstitial fibrosis. Therefore, we propose the concept of muscle microenvironment homeostasis, which explains the abnormal pathological changes in muscles through the imbalance of muscle microenvironment homeostasis. And we identified an interstitial progenitor cell FAP during the transition from normal muscle microenvironment homeostasis to muscle microenvironment imbalance caused by muscle damage diseases. As a kind of pluripotent stem cell, FAPs do not participate in myogenic differentiation, but can differentiate into fibroblasts, adipocytes, osteoblasts, and chondrocytes. As a kind of mesenchymal progenitor cell, it is involved in the generation of extracellular matrix, regulate muscle regeneration, and maintain neuromuscular junction. However, the muscle microenvironment is disrupted by the causative factors, and the abnormal activities of FAPs eventually contribute to the complex pathological changes in muscles. Targeting the mechanisms of these muscle pathological changes, we have identified appropriate signaling targets for FAPs to improve and even treat muscle damage diseases. In this review, we propose the construction of muscle microenvironmental homeostasis and find the key cells that cause pathological changes in muscle after homeostasis is broken. By studying the mechanism of abnormal differentiation and apoptosis of FAPs, we found a strategy to inhibit the abnormal pathological changes in muscle damage diseases and improve muscle regeneration.
Collapse
Affiliation(s)
- Kai Yin
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Chengmin Zhang
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Zihan Deng
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Xiaoyu Wei
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Tingwen Xiang
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Chuan Yang
- Department of Biomedical Materials ScienceThird Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Can Chen
- Department for Combat Casualty Care TrainingTraining Base for Army Health Care, Army Medical University (Third Military Medical University)ChongqingPeople's Republic of China
| | - Yueqi Chen
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Fei Luo
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| |
Collapse
|
3
|
Zhang H, Wague A, Diaz A, Liu M, Sang L, Youn A, Sharma S, Milan N, Kim H, Feeley B, Liu X. Overexpression of PRDM16 improves muscle function after rotator cuff tears. J Shoulder Elbow Surg 2024; 33:2725-2733. [PMID: 39032686 DOI: 10.1016/j.jse.2024.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/01/2024] [Accepted: 05/19/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Muscle atrophy, fibrosis, and fatty infiltration are commonly seen in rotator cuff tears (RCTs), which are critical factors that directly determine the clinical outcomes for patients with this injury. Therefore, improving muscle quality after RCT is crucial in improving the clinical outcome of tendon repair. In recent years, it has been discovered that adults have functional beige/brown adipose tissue (BAT) that can secrete batokines to promote muscle growth. PRDM16, a PR-domain-containing protein, was discovered with the ability to determine the brown fat cell fate and stimulate its development. Thus, the goal of this study was to discover the role of PRDM16 in improving muscle function after massive tendon tears using a transgenic mouse model with an elevated level of PRDM16 expression. METHODS Transgenic aP2-driven PRDM16-overexpressing mice and C57BL/6J mice underwent unilateral supraspinatus (SS) tendon transection and suprascapular nerve transection (TTDN) as described previously (n = 8 in each group). DigiGait was performed to evaluate forelimb function at 6 weeks post the TTDN injury. Bilateral SS muscles, interscapular brown fat, epididymal white fat, and inguinal beige fat were harvested for analysis. The expression of PRDM16 in adipose tissue was detected by Western blot. Masson Trichrome staining was conducted to evaluate the muscle fibrosis, and Oil Red O staining was used to determine the fat infiltration. Muscle fiber type was determined by major histocompatibility complex (MHC) expression via immunostaining. All data were presented in the form of mean ± standard deviation. t test and 2-way analysis of variance was performed to determine a statistically significant difference between groups. Significance was considered when P < .05. RESULTS Western blot data showed an increased expression of PRDM16 protein in both white and brown fat in PRDM16-overexpressing mice compared with wild-type (WT) mice. Even though PRDM16 overexpression had no effect on increasing muscle weight, it significantly improved the forelimbs function with longer brake, stance, and stride time and larger stride length and paw area in mice after RCT. Additionally, PRDM16-overexpressing mice showed no difference in the amount of fibrosis when compared to WT mice; however, they had a significantly reduced area of fatty infiltration. These mice also exhibited abundant MHC-IIx fiber percentage in the supraspinatus muscle after TTDN. CONCLUSION Overexpression of PRDM16 significantly improved muscle function and reduced fatty infiltration after rotator cuff tears. Promoting BAT activity is beneficial in improving rotator cuff muscle quality and shoulder function after RCT.
Collapse
Affiliation(s)
- He Zhang
- Department of Physical Education, Central South University, Changsha, Hunan, China; Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Aboubacar Wague
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Agustin Diaz
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Mengyao Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Luke Sang
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Alex Youn
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sankalp Sharma
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Nesa Milan
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Hubert Kim
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Brian Feeley
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Xuhui Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Zhang H, Gu W, Wu G, Yu Y. Aging and Autophagy: Roles in Musculoskeletal System Injury. Aging Dis 2024:AD.2024.0362. [PMID: 38913046 DOI: 10.14336/ad.2024.0362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Aging is a multifactorial process that ultimately leads to a decline in physiological function and a consequent reduction in the health span, and quality of life in elderly population. In musculoskeletal diseases, aging is often associated with a gradual loss of skeletal muscle mass and strength, resulting in reduced functional capacity and an increased risk of chronic metabolic diseases, leading to impaired function and increased mortality. Autophagy is a highly conserved physiological process by which cells, under the regulation of autophagy-related genes, degrade their own organelles and large molecules by lysosomal degradation. This process is unique to eukaryotic cells and is a strict regulator of homeostasis, the maintenance of energy and substance balance. Autophagy plays an important role in a wide range of physiological and pathological processes such as cell homeostasis, aging, immunity, tumorigenesis and neurodegenerative diseases. On the one hand, under mild stress conditions, autophagy mediates the restoration of homeostasis and proliferation, reduction of the rate of aging and delay of the aging process. On the other hand, under more intense stress conditions, an inadequate suppression of autophagy can lead to cellular aging. Conversely, autophagy activity decreases during aging. Due to the interrelationship between aging and autophagy, limited literature exists on this topic. Therefore, the objective of this review is to summarize the current concepts on aging and autophagy in the musculoskeletal system. The aim is to better understand the mechanisms of age-related changes in bone, joint and muscle, as well as the interaction relationship between autophagy and aging. Its goal is to provide a comprehensive perspective for the improvement of diseases of the musculoskeletal system.
Collapse
Affiliation(s)
- Haifeng Zhang
- Department of Orthopedics Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhui Gu
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Genbin Wu
- Department of Orthopedics Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinxian Yu
- Department of Orthopedics Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Dar A, Li A, Petrigliano FA. Lineage tracing reveals a novel PDGFRβ + satellite cell subset that contributes to myo-regeneration of chronically injured rotator cuff muscle. Sci Rep 2024; 14:9668. [PMID: 38671006 PMCID: PMC11053018 DOI: 10.1038/s41598-024-58926-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Massive rotator cuff (RC) tendon tears are associated with progressive fibro-adipogenesis and muscle atrophy that altogether cause shoulder muscle wasting. Platelet derived growth factor β (PDGFRβ) lineage cells, that co-express PDGFRα have previously been shown to directly contribute to scar formation and fat accumulation in a mouse model of irreversible tendon and nerve transection (TTDN). Conversely, PDGFRβ+ lineage cells have also been shown to be myogenic in cultures and in other models of skeletal muscle injury. We therefore hypothesized that PDGFRβ demarcates two distinct RC residing subpopulations, fibro-adipogenic and myogenic, and aimed to elucidate the identity of the PDGFRβ myogenic precursors and evaluate their contribution, if any, to RC myo-regeneration. Lineage tracing revealed increasing contribution of PDGFRβ+ myo-progenitors to the formation of GFP+ myofibers, which were the most abundant myofiber type in regenerated muscle at 2 weeks post-TTDN. Muscle regeneration preceded muscle atrophy and both advanced from the lateral site of tendon transection to the farthest medial region. GFP+/PDGFRβ+Sca-1-lin-CXCR4+Integrin-β1+ marked a novel subset of satellite cells with confirmed myogenic properties. Further studies are warranted to identify the existence of PDGFRβ+ satellite cells in human and other mouse muscles and to define their myo-regenerative potential following acute and chronic muscle injury.
Collapse
Affiliation(s)
- Ayelet Dar
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Angela Li
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Gil-Melgosa L, Llombart-Blanco R, Extramiana L, Lacave I, Abizanda G, Miranda E, Agirre X, Prósper F, Pineda-Lucena A, Pons-Villanueva J, Pérez-Ruiz A. HDACi vorinostat protects muscle from degeneration after acute rotator cuff injury in mice. Bone Joint Res 2024; 13:169-183. [PMID: 38618868 PMCID: PMC11017234 DOI: 10.1302/2046-3758.134.bjr-2023-0292.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Aims Rotator cuff (RC) injuries are characterized by tendon rupture, muscle atrophy, retraction, and fatty infiltration, which increase injury severity and jeopardize adequate tendon repair. Epigenetic drugs, such as histone deacetylase inhibitors (HDACis), possess the capacity to redefine the molecular signature of cells, and they may have the potential to inhibit the transformation of the fibro-adipogenic progenitors (FAPs) within the skeletal muscle into adipocyte-like cells, concurrently enhancing the myogenic potential of the satellite cells. Methods HDACis were added to FAPs and satellite cell cultures isolated from mice. The HDACi vorinostat was additionally administered into a RC injury animal model. Histological analysis was carried out on the isolated supra- and infraspinatus muscles to assess vorinostat anti-muscle degeneration potential. Results Vorinostat, a HDACi compound, blocked the adipogenic transformation of muscle-associated FAPs in culture, promoting myogenic progression of the satellite cells. Furthermore, it protected muscle from degeneration after acute RC in mice in the earlier muscle degenerative stage after tenotomy. Conclusion The HDACi vorinostat may be a candidate to prevent early muscular degeneration after RC injury.
Collapse
Affiliation(s)
- Lara Gil-Melgosa
- Orthopedic Surgery Department of Clínica Universidad de Navarra (CUN) and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Rafael Llombart-Blanco
- Orthopedic Surgery Department of Clínica Universidad de Navarra (CUN) and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Leire Extramiana
- Technological Innovation Division, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV) and IdiSNA, Pamplona, Spain
| | | | - Gloria Abizanda
- Technological Innovation Division, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV) and IdiSNA, Pamplona, Spain
| | | | - Xabier Agirre
- Hemato-Oncology Program, FIMA-UNAV and IdiSNA, Pamplona, Spain
| | - Felipe Prósper
- Hemato-Oncology Program, FIMA-UNAV and IdiSNA, Pamplona, Spain
- Haematology Department, Clinica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | | | - Juan Pons-Villanueva
- Orthopedic Surgery Department of Clínica Universidad de Navarra (CUN) and Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Ana Pérez-Ruiz
- Technological Innovation Division, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV) and IdiSNA, Pamplona, Spain
| |
Collapse
|
7
|
Wang N, Wang H, Shen L, Liu X, Ma Y, Wang C. Aging-Related Rotator Cuff Tears: Molecular Mechanisms and Implications for Clinical Management. Adv Biol (Weinh) 2024; 8:e2300331. [PMID: 38295015 DOI: 10.1002/adbi.202300331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/15/2023] [Indexed: 02/02/2024]
Abstract
Shoulder pain and disabilities are prevalent issues among the elderly population, with rotator cuff tear (RCT) being one of the leading causes. Although surgical treatment has shown some success, high postoperative retear rates remain a great challenge, particularly in elderly patients. Aging-related degeneration of muscle, tendon, tendon-to-bone enthesis, and bone plays a critical role in the development and prognosis of RCT. Studies have demonstrated that aging worsens muscle atrophy and fatty infiltration, alters tendon structure and biomechanical properties, exacerbates enthesis degeneration, and reduces bone density. Although recent researches have contributed to understanding the pathophysiological mechanisms of aging-related RCT, a comprehensive systematic review of this topic is still lacking. Therefore, this article aims to present a review of the pathophysiological changes and their clinical significance, as well as the molecular mechanisms underlying aging-related RCT, with the goal of shedding light on new therapeutic approaches to reduce the occurrence of aging-related RCT and improve postoperative prognosis in elderly patients.
Collapse
Affiliation(s)
- Ni Wang
- Department of Rehabilitation Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haoyuan Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Longxiang Shen
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xudong Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yanhong Ma
- Department of Rehabilitation Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chongyang Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
8
|
Saveh-Shemshaki N, Barajaa MA, Otsuka T, Mirdamadi ES, Nair LS, Laurencin CT. Electroconductivity, a regenerative engineering approach to reverse rotator cuff muscle degeneration. Regen Biomater 2023; 10:rbad099. [PMID: 38020235 PMCID: PMC10676522 DOI: 10.1093/rb/rbad099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023] Open
Abstract
Muscle degeneration is one the main factors that lead to the high rate of retear after a successful repair of rotator cuff (RC) tears. The current surgical practices have failed to treat patients with chronic massive rotator cuff tears (RCTs). Therefore, regenerative engineering approaches are being studied to address the challenges. Recent studies showed the promising outcomes of electroactive materials (EAMs) on the regeneration of electrically excitable tissues such as skeletal muscle. Here, we review the most important biological mechanism of RC muscle degeneration. Further, the review covers the recent studies on EAMs for muscle regeneration including RC muscle. Finally, we will discuss the future direction toward the application of EAMs for the augmentation of RCTs.
Collapse
Affiliation(s)
- Nikoo Saveh-Shemshaki
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Mohammed A Barajaa
- Department of Biomedical Engineering, Imam Abdulrahman Bin Faisal University, Dammam 31451, Saudi Arabia
| | - Takayoshi Otsuka
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
| | - Elnaz S Mirdamadi
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Lakshmi S Nair
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Cato T Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
9
|
Tanaka S, Portilla D, Okusa MD. Role of perivascular cells in kidney homeostasis, inflammation, repair and fibrosis. Nat Rev Nephrol 2023; 19:721-732. [PMID: 37608184 DOI: 10.1038/s41581-023-00752-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/24/2023]
Abstract
Perivascular niches in the kidney comprise heterogeneous cell populations, including pericytes and fibroblasts, with distinct functions. These perivascular cells have crucial roles in preserving kidney homeostasis as they maintain microvascular networks by stabilizing the vasculature and regulating capillary constriction. A subset of kidney perivascular cells can also produce and secrete erythropoietin; this ability can be enhanced with hypoxia-inducible factor-prolyl hydroxylase inhibitors, which are used to treat anaemia in chronic kidney disease. In the pathophysiological state, kidney perivascular cells contribute to the progression of kidney fibrosis, partly via transdifferentiation into myofibroblasts. Moreover, perivascular cells are now recognized as major innate immune sentinels in the kidney that produce pro-inflammatory cytokines and chemokines following injury. These mediators promote immune cell infiltration, leading to persistent inflammation and progression of kidney fibrosis. The crosstalk between perivascular cells and tubular epithelial, immune and endothelial cells is therefore a key process in physiological and pathophysiological states. Here, we examine the multiple roles of kidney perivascular cells in health and disease, focusing on the latest advances in this field of research.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Didier Portilla
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA, USA
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
10
|
Loomis T, Smith LR. Thrown for a loop: fibro-adipogenic progenitors in skeletal muscle fibrosis. Am J Physiol Cell Physiol 2023; 325:C895-C906. [PMID: 37602412 PMCID: PMC11932532 DOI: 10.1152/ajpcell.00245.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023]
Abstract
Fibro-adipogenic progenitors (FAPs) are key regulators of skeletal muscle regeneration and homeostasis. However, dysregulation of these cells leads to fibro-fatty infiltration across various muscle diseases. FAPs are the key source of extracellular matrix (ECM) deposition in muscle, and disruption to this process leads to a pathological accumulation of ECM, known as fibrosis. The replacement of contractile tissue with fibrotic ECM functionally impairs the muscle and increases muscle stiffness. FAPs and fibrotic muscle form a progressively degenerative feedback loop where, as a muscle becomes fibrotic, it induces a fibrotic FAP phenotype leading to further development of fibrosis. In this review, we summarize FAPs' role in fibrosis in terms of their activation, heterogeneity, contributions to fibrotic degeneration, and role across musculoskeletal diseases. We also discuss current research on potential therapeutic avenues to attenuate fibrosis by targeting FAPs.
Collapse
Affiliation(s)
- Taryn Loomis
- Biomedical Engineering Graduate Group, University of California, Davis, California, United States
| | - Lucas R Smith
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California, United States
- Department of Physical Medicine and Rehabilitation, University of California, Davis, California, United States
| |
Collapse
|
11
|
Gomez-Salazar MA, Wang Y, Thottappillil N, Hardy RW, Alexandre M, Höller F, Martin N, Gonzalez-Galofre ZN, Stefancova D, Medici D, James AW, Péault B. Aldehyde Dehydrogenase, a Marker of Normal and Malignant Stem Cells, Typifies Mesenchymal Progenitors in Perivascular Niches. Stem Cells Transl Med 2023; 12:474-484. [PMID: 37261440 PMCID: PMC10651226 DOI: 10.1093/stcltm/szad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 04/07/2023] [Indexed: 06/02/2023] Open
Abstract
Innate mesenchymal stem cells exhibiting multilineage differentiation and tissue (re)generative-or pathogenic-properties reside in perivascular niches. Subsets of these progenitors are committed to either osteo-, adipo-, or fibrogenesis, suggesting the existence of a developmental organization in blood vessel walls. We evaluated herein the activity of aldehyde dehydrogenase, a family of enzymes catalyzing the oxidation of aldehydes into carboxylic acids and a reported biomarker of normal and malignant stem cells, within human adipose tissue perivascular areas. A progression of ALDHLow to ALDHHigh CD34+ cells was identified in the tunica adventitia. Mesenchymal stem cell potential was confined to ALDHHigh cells, as assessed by proliferation and multilineage differentiation in vitro of cells sorted by flow cytometry with a fluorescent ALDH substrate. RNA sequencing confirmed and validated that ALDHHigh cells have a progenitor cell phenotype and provided evidence that the main isoform in this fraction is ALDH1A1, which was confirmed by immunohistochemistry. This demonstrates that ALDH activity, which marks hematopoietic progenitors and stem cells in diverse malignant tumors, also typifies native, blood vessel resident mesenchymal stem cells.
Collapse
Affiliation(s)
- Mario A Gomez-Salazar
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | | | - Reef W Hardy
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Manon Alexandre
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Polytech Marseille, Aix Marseille University, Marseille, France
| | - Fabian Höller
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Niall Martin
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Zaniah N Gonzalez-Galofre
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Dorota Stefancova
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Daniele Medici
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | - Bruno Péault
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
12
|
Li A, Anbuchelvan M, Fathi A, Abu-Zahra M, Evseenko D, Petrigliano FA, Dar A. Distinct human skeletal muscle-derived CD90 progenitor subsets for myo-fibro-adipogenic disease modeling and treatment in multiplexed conditions. Front Cell Dev Biol 2023; 11:1173794. [PMID: 37143896 PMCID: PMC10151706 DOI: 10.3389/fcell.2023.1173794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/03/2023] [Indexed: 05/06/2023] Open
Abstract
Chronic muscle injuries, such as massive rotator cuff tears, are associated with progressive muscle wasting, fibrotic scarring, and intramuscular fat accumulation. While progenitor cell subsets are usually studied in culture conditions that drive either myogenic, fibrogenic, or adipogenic differentiation, it is still unknown how combined myo-fibro-adipogenic signals, which are expected to occur in vivo, modulate progenitor differentiation. We therefore evaluated the differentiation potential of retrospectively generated subsets of primary human muscle mesenchymal progenitors in multiplexed conditions in the presence or absence of 423F drug, a modulator of gp130 signaling. We identified a novel CD90+CD56- non-adipogenic progenitor subset that maintained a lack of adipogenic potential in single and multiplexed myo-fibro-adipogenic culture conditions. CD90-CD56- demarcated fibro-adipogenic progenitors (FAP) and CD56+CD90+ progenitors were typified as myogenic. These human muscle subsets exhibited varying degrees of intrinsically regulated differentiation in single and mixed induction cultures. Modulation of gp130 signaling via 423F drug mediated muscle progenitor differentiation in a dose-, induction-, and cell subset-dependent manner and markedly decreased fibro-adipogenesis of CD90-CD56- FAP. Conversely, 423F promoted myogenesis of CD56+CD90+ myogenic subset, indicated by increased myotube diameter and number of nuclei per myotube. 423F treatment eliminated FAP-derived mature adipocytes from mixed adipocytes-FAP cultures but did not modify the growth of non-differentiated FAP in these cultures. Collectively, these data demonstrate that capability of myogenic, fibrogenic, or adipogenic differentiation is largely dependent on the intrinsic features of cultured subsets, and that the degree of lineage differentiation varies when signals are multiplexed. Moreover, our tests performed in primary human muscle cultures reveal and confirm the potential triple-therapeutic effects of 423F drug which simultaneously attenuates degenerative fibrosis, fat accumulation and promotes myo-regeneration.
Collapse
Affiliation(s)
- Angela Li
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Madhavan Anbuchelvan
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Amir Fathi
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Maya Abu-Zahra
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Frank A. Petrigliano
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ayelet Dar
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
13
|
Sahai A, Jones DL, Hughes M, Pu A, Williams K, Iyer SR, Rathinam C, Davis DL, Lovering RM, Gilotra MN. Fibroadipogenic progenitor cell response peaks prior to progressive fatty infiltration after rotator cuff tendon tear. J Orthop Res 2022; 40:2743-2753. [PMID: 35239216 PMCID: PMC9440165 DOI: 10.1002/jor.25321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023]
Abstract
Fibroadipogenic progenitor (FAP) cells are implicated as a major source of fatty infiltration (FI) in murine rotator cuff (RC) injury, but FAP cell response after RC tear in a rabbit model is unknown. This study determined whether changes in FAP cell count after an RC tear predate muscle degeneration in a clinically relevant rabbit model. We hypothesized increases in FAP cell count correlate temporally with RC degeneration. New Zealand white rabbits (n = 26) were evaluated at 1, 2, 4, and 6 weeks after unilateral full-thickness tenotomy of supraspinatus and infraspinatus tendons. FI area and adipocyte size were histologically analyzed, muscle density was measured by computerized tomography, and quantification of FAP cells was measured by flow cytometry and immunohistochemistry. The percentage of intrafascicular adipocyte area increased over time in supraspinatus muscle samples (p = 0.03), significantly between 1- and 6-week samples (p = 0.04). There were no differences in perifascicular adipocyte area percentages between time points. Peak increase in FAP cell count occurred at 1-week (p = 0.03), with a decrease in the following weeks. There was a negative correlation between supraspinatus adipocyte area and FAP cell count (p < 0.05). On computed tomography (CT) scan, maximal decrease in muscle density was observed in the 4th to 6th weeks. In summary, FAP cell response occurred early after tenotomy and did not correlate temporally with increases in FI. This suggests that FAP cell response may predate degenerative changes, and early targeting of FAPs before adipocyte maturation could blunt FI after RC tear.
Collapse
Affiliation(s)
- Amil Sahai
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Derek L. Jones
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Marcus Hughes
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Alex Pu
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Katrina Williams
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Shama R. Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Chozha Rathinam
- Department of Immunology, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Derik L. Davis
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Richard M. Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| | - Mohit N. Gilotra
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Baltimore, USA
| |
Collapse
|
14
|
Zhang G, Zhou X, Hu S, Jin Y, Qiu Z. Large animal models for the study of tendinopathy. Front Cell Dev Biol 2022; 10:1031638. [PMID: 36393858 PMCID: PMC9640604 DOI: 10.3389/fcell.2022.1031638] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Tendinopathy has a high incidence in athletes and the aging population. It can cause pain and movement disorders, and is one of the most difficult problems in orthopedics. Animal models of tendinopathy provide potentially efficient and effective means to develop understanding of human tendinopathy and its underlying pathological mechanisms and treatments. The selection of preclinical models is essential to ensure the successful translation of effective and innovative treatments into clinical practice. Large animals can be used in both micro- and macro-level research owing to their similarity to humans in size, structure, and function. This article reviews the application of large animal models in tendinopathy regarding injuries to four tendons: rotator cuff, patellar ligament, Achilles tendon, and flexor tendon. The advantages and disadvantages of studying tendinopathy with large animal models are summarized. It is hoped that, with further development of animal models of tendinopathy, new strategies for the prevention and treatment of tendinopathy in humans will be developed.
Collapse
Affiliation(s)
- Guorong Zhang
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xuyan Zhou
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Shuang Hu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Ye Jin, ; Zhidong Qiu,
| | - Zhidong Qiu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Ye Jin, ; Zhidong Qiu,
| |
Collapse
|
15
|
Wang D. Fatty Infiltration and Atrophy in Rotator Cuff Muscle Degeneration: What More Should We Look For?: Commentary on an article by Lindsey Ruderman, BA, et al.: "Histologic Differences in Human Rotator Cuff Muscle Based on Tear Characteristics". J Bone Joint Surg Am 2022; 104:e58. [PMID: 35793801 DOI: 10.2106/jbjs.22.00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Dean Wang
- Department of Orthopaedic Surgery, University of California Irvine, Orange, California
| |
Collapse
|
16
|
Takada Y, Matsumura N, Shirasawa H, Yoda M, Matsumoto M, Nakamura M, Horiuchi K. Aging Aggravates the Progression of Muscle Degeneration After Rotator Cuff Tears in Mice. Arthroscopy 2022; 38:752-760. [PMID: 34571183 DOI: 10.1016/j.arthro.2021.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE The purpose of this study was to evaluate the impact of aging on muscle degeneration after rotator cuff tear (RCT) in mice. METHODS Young (12-week-old) and aged (50-to-60-week-old) female C57BL/6 mice were used (n = 29 for each group). The rotator cuff was transected, and the proximal humerus was removed to induce degeneration of the rotator cuff muscles. The mice were euthanized 4 and 12 weeks after the procedure (referred to as RCT-4wk mice and RCT-12wk mice, respectively) and compared with the sham-treated mice. The supraspinatus muscles were collected for histology, Western blot analysis, and gene expression analyses. RESULTS There was a significant increase in fat tissue in aged RCT-4wk mice (P = .001) and aged RCT-12wk mice (P < .001) compared with sham-treated aged mice, and aged RCT-12wk mice had a significantly increased fat area ratio compared with aged RCT-4wk mice (P < .001). The fat area was significantly larger in both the aged RCT-4wk (P = .002) and RCT-12wk mice (P < .001) than in the corresponding young mice. Muscular fibrosis was significantly increased in aged RCT-12wk mice compared with aged sham-treated mice (P = .005) and young RCT-12wk mice (P = .016). There were also significant increases in the expression of perilipin and transcripts of adipogenic and fibrogenic differentiation markers in aged RCT mice compared with young RCT mice. CONCLUSION The present results show that aging is critically involved in the pathology of muscular fatty infiltration and fibrosis after RCT, and muscular degeneration progresses over time in aged mice. CLINICAL RELEVANCE Aging promotes the progression of muscle degeneration in a mouse RCT model. Furthermore, this study shows that muscle degeneration occurs in aged mice even without denervation and that the model described in the present study is a useful tool for studying the pathology of muscle degeneration.
Collapse
Affiliation(s)
- Yuhei Takada
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Noboru Matsumura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan.
| | - Hideyuki Shirasawa
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Yoda
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| |
Collapse
|
17
|
Craig DJ, James AW, Wang Y, Tavian M, Crisan M, Péault BM. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:35-43. [PMID: 35641167 PMCID: PMC8895497 DOI: 10.1093/stcltm/szab001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022] Open
Abstract
The vascular wall is comprised of distinct layers controlling angiogenesis, blood flow, vessel anchorage within organs, and cell and molecule transit between blood and tissues. Moreover, some blood vessels are home to essential stem-like cells, a classic example being the existence in the embryo of hemogenic endothelial cells at the origin of definitive hematopoiesis. In recent years, microvascular pericytes and adventitial perivascular cells were observed to include multi-lineage progenitor cells involved not only in organ turnover and regeneration but also in pathologic remodeling, including fibrosis and atherosclerosis. These perivascular mesodermal elements were identified as native forerunners of mesenchymal stem cells. We have presented in this brief review our current knowledge on vessel wall-associated tissue remodeling cells with respect to discriminating phenotypes, functional diversity in health and disease, and potential therapeutic interest.
Collapse
Affiliation(s)
- David J Craig
- Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Mihaela Crisan
- Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Bruno M Péault
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, USA
- Corresponding author: Bruno Péault, PhD, Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, 615 Charles E. Young Drive South, Los Angeles, CA 90095-7358, USA.
| |
Collapse
|
18
|
Bogdanov J, Lan R, Chu TN, Bolia IK, Weber AE, Petrigliano FA. Fatty degeneration of the rotator cuff: pathogenesis, clinical implications, and future treatment. JSES REVIEWS, REPORTS, AND TECHNIQUES 2021; 1:301-308. [PMID: 37588720 PMCID: PMC10426606 DOI: 10.1016/j.xrrt.2021.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Chronic rotator cuff pathology is often complicated by fatty degeneration of the rotator cuff (FDRC) muscles, an insidious process associated with poor prognosis with or without surgical intervention. Currently there is no treatment for FDRC, and many studies have described a natural course for this disease almost always resulting in further degeneration and morbidity. Recapitulating FDRC using animal injury models, and using imaging-based studies of human FDRC, the pathophysiology of this disease continues to be further characterized. Researchers studying mesenchymal stem cell-derived progenitor cells and known fibrogenic and adipogenic signaling pathways implicated in FDRC seek to clarify the underlying processes driving these changes. While new cell- and molecular-based therapies are being developed, currently the strongest available avenue for improved management of FDRC is the use of novel imaging techniques which allow for more accurate and personalized staging of fatty degeneration. This narrative review summarizes the evidence on the molecular and pathophysiologic mechanisms of FDRC and provides a clinical update on the diagnosis and management of this condition based on the existing knowledge. We also sought to examine the role of newer biologic therapies in the management of RC fatty degeneration and to identify areas of future research.
Collapse
Affiliation(s)
- Jacob Bogdanov
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Rae Lan
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Timothy N. Chu
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Ioanna K. Bolia
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Alexander E. Weber
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Frank A. Petrigliano
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| |
Collapse
|
19
|
Hsu GCY, Cherief M, Sono T, Wang Y, Negri S, Xu J, Peault B, James AW. Divergent effects of distinct perivascular cell subsets for intra-articular cell therapy in posttraumatic osteoarthritis. J Orthop Res 2021; 39:2388-2397. [PMID: 33512030 PMCID: PMC8319216 DOI: 10.1002/jor.24997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/30/2020] [Accepted: 01/24/2021] [Indexed: 02/04/2023]
Abstract
Intra-articular injection of mesenchymal stem cells has shown benefit for the treatment of osteoarthritis (OA). However, mesenchymal stem/stromal cells at the origin of these clinical results are heterogenous cell populations with limited cellular characterization. Here, two transgenic reporter mice were used to examine the differential effects of two precisely defined perivascular cell populations (Pdgfrα+ and Pdgfrβ+ cells) from white adipose tissue for alleviation of OA. Perivascular mesenchymal cells were isolated from transgenic Pdgfrα-and Pdgfrβ-CreERT2 reporter animals and delivered as a one-time intra-articular dose to C57BL/6J mice after destabilization of the medial meniscus (DMM). Both Pdgfrα+ and Pdgfrβ+ cell preparations improved metrics of cartilage degradation and reduced markers of chondrocyte hypertrophy. While some similarities in cell distribution were identified within the synovial and perivascular spaces, injected Pdgfrα+ cells remained in the superficial layers of articular cartilage, while Pdgfrβ+ cells were more widely dispersed. Pdgfrβ+ cell therapy prevented subchondral sclerosis induced by DMM, while Pdgfrα+ cell therapy had no effect. In summary, while both cell therapies showed beneficial effects in the DMM model, important differences in cell incorporation, persistence, and subchondral sclerosis were identified.
Collapse
Affiliation(s)
- Ginny Ching-Yun Hsu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States;,Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095;,Center For Cardiovascular Science and Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| |
Collapse
|
20
|
Fu C, Huang AH, Galatz LM, Han WM. Cellular and molecular modulation of rotator cuff muscle pathophysiology. J Orthop Res 2021; 39:2310-2322. [PMID: 34553789 DOI: 10.1002/jor.25179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/04/2021] [Accepted: 09/07/2021] [Indexed: 02/04/2023]
Abstract
Rotator cuff (RC) tendon tears are common shoulder injuries that result in irreversible and persistent degeneration of the associated muscles, which is characterized by severe inflammation, atrophy, fibrosis, and fatty infiltration. Although RC muscle degeneration strongly dictates the overall clinical outcomes, strategies to stimulate RC muscle regeneration have largely been overlooked to date. In this review, we highlight the current understanding of the cellular processes that coordinate muscle regeneration, and the roles of muscle resident cells, including immune cells, fibroadipogenic progenitors, and muscle satellite cells in the pathophysiologic regulation of RC muscles following injury. This review also provides perspectives for potential therapies to alleviate the hallmarks of RC muscle degeneration to address current limitations in postsurgical recovery.
Collapse
Affiliation(s)
- Chengcheng Fu
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Department of Orthopedic Surgery, Columbia University, New York City, New York, USA
| | - Leesa M Galatz
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Woojin M Han
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
21
|
Shaw IW, O'Sullivan ED, Pisco AO, Borthwick G, Gallagher KM, Péault B, Hughes J, Ferenbach DA. Aging modulates the effects of ischemic injury upon mesenchymal cells within the renal interstitium and microvasculature. Stem Cells Transl Med 2021; 10:1232-1248. [PMID: 33951342 PMCID: PMC8284778 DOI: 10.1002/sctm.20-0392] [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: 08/19/2020] [Revised: 02/05/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022] Open
Abstract
The renal mesenchyme contains heterogeneous cells, including interstitial fibroblasts and pericytes, with key roles in wound healing. Although healing is impaired in aged kidneys, the effect of age and injury on the mesenchyme remains poorly understood. We characterized renal mesenchymal cell heterogeneity in young vs old animals and after ischemia‐reperfusion‐injury (IRI) using multiplex immunolabeling and single cell transcriptomics. Expression patterns of perivascular cell markers (α‐SMA, CD146, NG2, PDGFR‐α, and PDGFR‐β) correlated with their interstitial location. PDGFR‐α and PDGFR‐β co‐expression labeled renal myofibroblasts more efficiently than the current standard marker α‐SMA, and CD146 was a superior murine renal pericyte marker. Three renal mesenchymal subtypes; pericytes, fibroblasts, and myofibroblasts, were recapitulated with data from two independently performed single cell transcriptomic analyzes of murine kidneys, the first dataset an aging cohort and the second dataset injured kidneys following IRI. Mesenchymal cells segregated into subtypes with distinct patterns of expression with aging and following injury. Baseline uninjured old kidneys resembled post‐ischemic young kidneys, with this phenotype further exaggerated following IRI. These studies demonstrate that age modulates renal perivascular/interstitial cell marker expression and transcriptome at baseline and in response to injury and provide tools for the histological and transcriptomic analysis of renal mesenchymal cells, paving the way for more accurate classification of renal mesenchymal cell heterogeneity and identification of age‐specific pathways and targets.
Collapse
Affiliation(s)
- Isaac W Shaw
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Eoin D O'Sullivan
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | | - Gary Borthwick
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Kevin M Gallagher
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Bruno Péault
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK.,Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Jeremy Hughes
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - David A Ferenbach
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| |
Collapse
|
22
|
Theret M, Rossi FMV, Contreras O. Evolving Roles of Muscle-Resident Fibro-Adipogenic Progenitors in Health, Regeneration, Neuromuscular Disorders, and Aging. Front Physiol 2021; 12:673404. [PMID: 33959042 PMCID: PMC8093402 DOI: 10.3389/fphys.2021.673404] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Normal skeletal muscle functions are affected following trauma, chronic diseases, inherited neuromuscular disorders, aging, and cachexia, hampering the daily activities and quality of life of the affected patients. The maladaptive accumulation of fibrous intramuscular connective tissue and fat are hallmarks of multiple pathologies where chronic damage and inflammation are not resolved, leading to progressive muscle replacement and tissue degeneration. Muscle-resident fibro-adipogenic progenitors are adaptable stromal cells with multilineage potential. They are required for muscle homeostasis, neuromuscular integrity, and tissue regeneration. Fibro-adipogenic progenitors actively regulate and shape the extracellular matrix and exert immunomodulatory functions via cross-talk with multiple other residents and non-resident muscle cells. Remarkably, cumulative evidence shows that a significant proportion of activated fibroblasts, adipocytes, and bone-cartilage cells, found after muscle trauma and disease, descend from these enigmatic interstitial progenitors. Despite the profound impact of muscle disease on human health, the fibrous, fatty, and ectopic bone tissues' origins are poorly understood. Here, we review the current knowledge of fibro-adipogenic progenitor function on muscle homeostatic integrity, regeneration, repair, and aging. We also discuss how scar-forming pathologies and disorders lead to dysregulations in their behavior and plasticity and how these stromal cells can control the onset and severity of muscle loss in disease. We finally explore the rationale of improving muscle regeneration by understanding and modulating fibro-adipogenic progenitors' fate and behavior.
Collapse
Affiliation(s)
- Marine Theret
- Biomedical Research Centre, Department of Medical Genetics, School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Biomedical Research Centre, Department of Medical Genetics, School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Osvaldo Contreras
- Departamento de Biología Celular y Molecular, Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, Australia
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| |
Collapse
|
23
|
Jiang Z, Feng T, Lu Z, Wei Y, Meng J, Lin CP, Zhou B, Liu C, Zhang H. PDGFRb + mesenchymal cells, but not NG2 + mural cells, contribute to cardiac fat. Cell Rep 2021; 34:108697. [PMID: 33535029 DOI: 10.1016/j.celrep.2021.108697] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/01/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Understanding cellular origins of cardiac adipocytes (CAs) can offer important implications for the treatment of fat-associated cardiovascular diseases. Here, we perform lineage tracing studies by using various genetic models and find that cardiac mesenchymal cells (MCs) contribute to CAs in postnatal development and adult homeostasis. Although PDGFRa+ and PDGFRb+ MCs both give rise to intramyocardial adipocytes, PDGFRb+ MCs are demonstrated to be the major source of intramyocardial adipocytes. Moreover, we find that PDGFRb+ cells are heterogenous, as PDGFRb is expressed not only in pericytes and smooth muscle cells (SMCs) but also in some subendocardial, pericapillary, or adventitial PDGFRa+ fibroblasts. Dual-recombinase-mediated intersectional genetic lineage tracing reveals that PDGFRa+PDGFRb+ double-positive periendothelial fibroblasts contribute to intramyocardial adipocytes. In contrast, SMCs and NG2+ pericytes do not contribute to CAs. These in vivo findings demonstrate that PDGFRb+ MCs, but not NG2+ coronary vascular mural cells, are the major source of intramyocardial adipocytes.
Collapse
Affiliation(s)
- Zhen Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Teng Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhengkai Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanxin Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jufeng Meng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Bin Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chen Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Hui Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
24
|
Wu G, Hu VJ, McClintick DJ, Gatto JD, Aderibigbe T, Lu L, Jensen AR, Dar A, Petrigliano FA. Lateral to medial fibro-adipogenic degeneration are greater in infraspinatus than supraspinatus following nerve and tendon injury of murine rotator cuff. J Orthop Res 2021; 39:184-195. [PMID: 32886404 DOI: 10.1002/jor.24847] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 02/04/2023]
Abstract
Small animal models of massive tears of the rotator cuff (RC) were introduced a decade ago and have been extensively used to study the pathophysiology of chronically injured RC. Transection of rodent suprascapular nerve and RC tendon results in progressive muscle atrophy, fibrosis and fat accumulation and affect the infraspinatus and supraspinatus muscles similarly to that seen in the setting of massive RC tears in humans. The purpose of this study was to perform a comprehensive and detailed analysis of the kinetics of fibrotic scar and adipose tissue development comparing phenotypic differences between chronically injured infraspinatus and supraspinatus. Automatic mosaic imaging was used to create large image of whole infraspinatus or supraspinatus sectioned area for quantification of spatial heterogeneity of muscle damage. Pathologic changes advanced from the lateral site of transection to the medial region far from the transection site. A prominent, accelerated muscle fibrosis and fat accumulation was measured in injured infraspinatus compared to supraspinatus. Furthermore, adipose tissue occupied significantly larger area than that of fibrotic tissue in both muscles but was greater in infraspinatus within 6 weeks post induction of injury. Our findings confirm that infraspinatus is more susceptible to accelerated chronic degeneration and can be used to identify the physiological functions that distinguish between the response of infraspinatus and supraspinatus in the setting of massive tears. Whether these pathologic differences observed in mice are reflected in humans is one key aspect that awaits clarification.
Collapse
Affiliation(s)
- Genbin Wu
- Department of Joint Surgery, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, China.,Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Vivian J Hu
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Daniel J McClintick
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Jonathan D Gatto
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Temidayo Aderibigbe
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Liangyu Lu
- Department of Joint Surgery, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Andrew R Jensen
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Ayelet Dar
- Epstein Family Center for Sports Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Frank A Petrigliano
- Epstein Family Center for Sports Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
25
|
Sono T, Hsu CY, Negri S, Miller S, Wang Y, Xu J, Meyers CA, Peault B, James AW. Platelet-derived growth factor receptor-β (PDGFRβ) lineage tracing highlights perivascular cell to myofibroblast transdifferentiation during post-traumatic osteoarthritis. J Orthop Res 2020; 38:2484-2494. [PMID: 32134140 PMCID: PMC7483913 DOI: 10.1002/jor.24648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 02/04/2023]
Abstract
Pericytes ubiquitously surround capillaries and microvessels within vascularized tissues and have diverse functions after tissue injury. In addition to regulation of angiogenesis and tissue regeneration after injury, pericytes also contribute to organ fibrosis. Destabilization of the medial meniscus (DMM) phenocopies post-traumatic osteoarthritis, yet little is known regarding the impact of DMM surgery on knee joint-associated pericytes and their cellular descendants. Here, inducible platelet-derived growth factor receptor-β (PDGFRβ)-CreERT2 reporter mice were subjected to DMM surgery, and lineage tracing studies performed over an 8-week period. Results showed that at baseline PDGFRβ reporter activity highlights abluminal perivascular cells within synovial and infrapatellar fat pad (IFP) tissues. DMM induces a temporospatially patterned increase in vascular density within synovial and subsynovial tissues. Marked vasculogenesis within IFP was accompanied by expansion of PDGFRβ reporter+ perivascular cell numbers, detachment of mGFP+ descendants from vessel walls, and aberrant adoption of myofibroblastic markers among mGFP+ cells including α-SMA, ED-A, and TGF-β1. At later timepoints, fibrotic changes and vascular maturation occurred within subsynovial tissues, with the redistribution of PDGFRβ+ cellular descendants back to their perivascular niche. In sum, PDGFRβ lineage tracing allows for tracing of perivascular cell fate within the diarthrodial joint. Further, destabilization of the joint induces vascular and fibrogenic changes of the IFP accompanied by perivascular to myofibroblast transdifferentiation.
Collapse
Affiliation(s)
- Takashi Sono
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States.,Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606–8507, Japan
| | - Ching-Yun Hsu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Sarah Miller
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095, University of Edinburgh, Edinburgh, United Kingdom,Center For Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
26
|
Feeley BT, Liu M, Ma CB, Agha O, Aung M, Lee C, Liu X. Human Rotator Cuff Tears Have an Endogenous, Inducible Stem Cell Source Capable of Improving Muscle Quality and Function After Rotator Cuff Repair. Am J Sports Med 2020; 48:2660-2668. [PMID: 32730704 PMCID: PMC9262007 DOI: 10.1177/0363546520935855] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The muscle quality of the rotator cuff (RC), measured by atrophy and fatty infiltration (FI), is a key determinant of outcomes in RC injury and repair. The ability to regenerate muscle after repair has been shown to be limited. PURPOSE To determine if there is a source of resident endogenous stem cells, fibroadipogenic progenitor cells (FAPs), within RC injury patients, and if these cells are capable of adipogenic, fibrogenic, and pro-myogenic differentiation. STUDY DESIGN Controlled laboratory study. METHODS A total of 20 patients between the ages of 40 and 75 years with partial- or full-thickness RC tears of the supraspinatus and evidence of atrophy and FI Goutallier grade 1, 2, or 3 were selected from 2 surgeons at an orthopaedic center. During the surgical repair procedure, supraspinatus muscle biopsy specimens were obtained for analysis as were deltoid muscle biopsy specimens to serve as the control. FAPs and satellite cells were quantified using fluorescence-activated cell sorting. Muscle FI and fibrosis was quantified using Oil Red O and Masson trichrome staining. FAP differentiation and gene expression profiles were compared across tear sizes after culture in adipogenic, fibrogenic, and beta-3 agonist (amibegron) conditions. Analysis of variance was used for statistical comparisons between groups, with P < .05 as statistically significant. RESULTS Histologic analysis confirmed the presence of fat in biopsy specimens from patients with full-thickness tears. There were more FAPs in the full-thickness tear group compared with the partial-thickness tear group (9.43% ± 4.25% vs 3.84% ± 2.54%; P < .01). Full-thickness tears were divided by tear size, with patients with larger tears having significantly more FAPs than those with smaller tears. FAPs from muscles with full-thickness tendon tears had more adipogenic and fibrogenic potential than those with partial tears. Induction of a beige adipose tissue (BAT) phenotype in FAPs was possible, as demonstrated by increased expression of BAT markers and pro-myogenic genes including insulin-like growth factor 1 and follistatin. CONCLUSION Endogenous FAPs are present within the RC and likely are the source of FI. These FAPs were increased in muscles with in larger tears but are capable of adopting a pro-myogenic BAT phenotype that could be utilized to improve muscle quality and patient function after RC repair.
Collapse
Affiliation(s)
- Brian T. Feeley
- San Francisco Veteran Affairs Health Care System, San Francisco, California, USA.,Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, California, USA.,Address correspondence to Brian T. Feeley, MD, Department of Orthopedic Surgery, University of California, San Francisco, 1700 Owens Street, San Francisco, CA 94158, USA ()
| | - Mengyao Liu
- San Francisco Veteran Affairs Health Care System, San Francisco, California, USA.,Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - C. Benjamin Ma
- San Francisco Veteran Affairs Health Care System, San Francisco, California, USA.,Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Obiajulu Agha
- San Francisco Veteran Affairs Health Care System, San Francisco, California, USA.,Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Mya Aung
- San Francisco Veteran Affairs Health Care System, San Francisco, California, USA
| | - Carlin Lee
- San Francisco Veteran Affairs Health Care System, San Francisco, California, USA.,Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Xuhui Liu
- San Francisco Veteran Affairs Health Care System, San Francisco, California, USA.,Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
27
|
Rationale for the Use of Orthobiologics in Sports Medicine. OPER TECHN SPORT MED 2020. [DOI: 10.1016/j.otsm.2020.150753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
28
|
Agha O, Diaz A, Davies M, Kim HT, Liu X, Feeley BT. Rotator cuff tear degeneration and the role of fibro-adipogenic progenitors. Ann N Y Acad Sci 2020; 1490:13-28. [PMID: 32725671 DOI: 10.1111/nyas.14437] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/06/2020] [Accepted: 06/18/2020] [Indexed: 12/25/2022]
Abstract
The high prevalence of rotator cuff tears poses challenges to individual patients and the healthcare system at large. This orthopedic injury is complicated further by high rates of retear after surgical repair. Outcomes following repair are highly dependent upon the quality of the injured rotator cuff muscles, and it is, therefore, crucial that the pathophysiology of rotator cuff degeneration continues to be explored. Fibro-adipogenic progenitors, a major population of resident muscle stem cells, have emerged as the main source of intramuscular fibrosis and fatty infiltration, both of which are key features of rotator cuff muscle degeneration. Improvements to rotator cuff repair outcomes will likely require addressing the muscle pathology produced by these cells. The aim of this review is to summarize the current rotator cuff degeneration assessment tools, the effects of poor muscle quality on patient outcomes, the role of fibro-adipogenic progenitors in mediating muscle pathology, and how these cells could be leveraged for potential therapeutics to augment current rotator cuff surgical and rehabilitative strategies.
Collapse
Affiliation(s)
- Obiajulu Agha
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California.,San Francisco Veteran Affairs Health Care System, San Francisco, California
| | - Agustin Diaz
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California.,San Francisco Veteran Affairs Health Care System, San Francisco, California
| | - Michael Davies
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California.,San Francisco Veteran Affairs Health Care System, San Francisco, California
| | - Hubert T Kim
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California.,San Francisco Veteran Affairs Health Care System, San Francisco, California
| | - Xuhui Liu
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California.,San Francisco Veteran Affairs Health Care System, San Francisco, California
| | - Brian T Feeley
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California.,San Francisco Veteran Affairs Health Care System, San Francisco, California
| |
Collapse
|
29
|
Jensen AR, Taylor AJ, Sanchez-Sotelo J. Factors Influencing the Reparability and Healing Rates of Rotator Cuff Tears. Curr Rev Musculoskelet Med 2020; 13:572-583. [PMID: 32681307 DOI: 10.1007/s12178-020-09660-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF THE REVIEW To discuss tear- and patient-related factors that influence the healing potential of rotator cuff tears and to clarify the terminology surrounding this topic. RECENT FINDINGS Over the last few years, further insight has been gained regarding rotator cuff tear features that are associated with poor healing rates after rotator cuff repair. Some of these features have been incorporated in prediction models developed to accurately predict rotator cuff healing rates utilizing preoperative risk factors weighted by importance. Rotator cuff tears may be considered functionally irreparable based on their size, chronicity, absence of adequate tendon length, atrophy, and fatty infiltration. Furthermore, advanced age, use of tobacco products, diabetes, and other patient-related factors may impair tendon healing. Careful analysis and discussion of all these factors with patients is essential to determine if surgical repair of a rotator cuff tear should be recommended, or if it is best to proceed with one of the several salvage procedures reviewed in this topical collection, including augmentation of the repair, superior capsular reconstruction, tendon transfers, and other.
Collapse
Affiliation(s)
- Andrew R Jensen
- Department of Orthopaedic Surgery, University of California, Los Angeles, CA, 90403, USA
| | - Adam J Taylor
- Department of Orthopedic Surgery, Mayo Clinic, 200 First Street, Rochester, MN, 55905, USA
| | - Joaquin Sanchez-Sotelo
- Department of Orthopaedic Surgery, Harbor-UCLA Medical Center, Torrance, CA, 90502, USA.
| |
Collapse
|
30
|
Perivascular Fibro-Adipogenic Progenitor Tracing during Post-Traumatic Osteoarthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1909-1920. [PMID: 32533926 DOI: 10.1016/j.ajpath.2020.05.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/03/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022]
Abstract
Perivascular mural cells surround capillaries and microvessels and have diverse regenerative or fibrotic functions after tissue injury. Subsynovial fibrosis is a well-known pathologic feature of osteoarthritis, yet transgenic animals for use in visualizing perivascular cell contribution to fibrosis during arthritic changes have not been developed. Here, inducible Pdgfra-CreERT2 reporter mice were subjected to joint-destabilization surgery to induce arthritic changes, and cell lineage was traced over an 8-week period with a focus on the joint-associated fat pad. Results showed that, at baseline, inducible Pdgfra reporter activity highlighted adventitial and, to a lesser extent, pericytic cells within the infrapatellar fat pad. Joint-destabilization surgery was associated with marked fibrosis of the infrapatellar fat pad, accompanied by an expansion of perivascular Pdgfra-expressing cellular descendants, many of which adopted α-smooth muscle actin expression. Gene expression analysis of microdissected infrapatellar fat pad confirmed enrichment in membrane-bound green fluorescent protein/Pdgfra-expressing cells, along with a gene signature that corresponded with injury-associated fibro-adipogenic progenitors. Our results highlight dynamic changes in joint-associated perivascular fibro-adipogenic progenitors during osteoarthritis.
Collapse
|
31
|
Flück M, Fitze D, Ruoss S, Valdivieso P, von Rechenberg B, Bratus-Neuenschwander A, Opitz L, Hu J, Laczko E, Wieser K, Gerber C. Down-Regulation of Mitochondrial Metabolism after Tendon Release Primes Lipid Accumulation in Rotator Cuff Muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1513-1529. [PMID: 32305353 DOI: 10.1016/j.ajpath.2020.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 02/14/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Atrophy and fat accumulation are debilitating aspects of muscle diseases and are rarely prevented. Using a vertical approach combining anatomic techniques with omics methodology in a tenotomy-induced sheep model of rotator cuff disease, we tested whether mitochondrial dysfunction is implicated in muscle wasting and perturbed lipid metabolism, speculating that both can be prevented by the stimulation of β-oxidation with l-carnitine. The infraspinatus muscle lost 22% of its volume over the first 6 weeks after tenotomy before the area-percentage of lipid increased from 8% to 18% at week 16. Atrophy was associated with the down-regulation of mitochondrial transcripts and protein and a slow-to-fast shift in muscle composition. Correspondingly, amino acid levels were increased 2 weeks after tendon release, when the levels of high-energy phosphates and glycerophospholipids were lowered. l-Carnitine administration (0.9 g/kg per day) prevented atrophy over the first 2 weeks, and mitigated alterations of glutamate, glycerophospholipids, and carnitine levels in released muscle, but did not prevent the level decrease in high-energy phosphates or protein constituents of mitochondrial respiration, promoting the accumulation of longer lipids with an increasing saturation. We conclude that the early phase of infraspinatus muscle degeneration after tendon release involves the elimination of oxidative characteristics associated with an aberrant accumulation of lipid species but is largely unrelated to the prevention of atrophy with oral l-carnitine administration.
Collapse
Affiliation(s)
- Martin Flück
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland.
| | - Daniel Fitze
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Severin Ruoss
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Paola Valdivieso
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Brigitte von Rechenberg
- Vetsuisse Faculty, Musculoskeletal Research Unit, Competence Center for Applied Biotechnology, University of Zurich, Zurich, Switzerland
| | | | - Lennart Opitz
- Functional Genomics Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Junmin Hu
- Functional Genomics Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Endre Laczko
- Functional Genomics Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Karl Wieser
- Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland
| | - Christian Gerber
- Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland
| |
Collapse
|
32
|
Gomez-Salazar M, Gonzalez-Galofre ZN, Casamitjana J, Crisan M, James AW, Péault B. Five Decades Later, Are Mesenchymal Stem Cells Still Relevant? Front Bioeng Biotechnol 2020; 8:148. [PMID: 32185170 PMCID: PMC7058632 DOI: 10.3389/fbioe.2020.00148] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells are culture-derived mesodermal progenitors isolatable from all vascularized tissues. In spite of multiple fundamental, pre-clinical and clinical studies, the native identity and role in tissue repair of MSCs have long remained elusive, with MSC selection in vitro from total cell suspensions essentially unchanged as a mere primary culture for half a century. Recent investigations have helped understand the tissue origin of these progenitor cells, and uncover alternative effects of MSCs on tissue healing via growth factor secretion and interaction with the immune system. In this review, we describe current trends in MSC biology and discuss how these may improve the use of these therapeutic cells in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mario Gomez-Salazar
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zaniah N Gonzalez-Galofre
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Joan Casamitjana
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mihaela Crisan
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Bruno Péault
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom.,Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
33
|
Sharma AK, Levian B, Shah P, Mosich GM, Husman R, Ariniello A, Gatto JD, Hu VJ, McClintick DJ, Jensen AR, McAllister DR, Péault B, Dar A, Petrigliano FA. Aged Mice Demonstrate Greater Muscle Degeneration of Chronically Injured Rotator Cuff. J Orthop Res 2020; 38:320-328. [PMID: 31517395 DOI: 10.1002/jor.24468] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 09/03/2019] [Indexed: 02/04/2023]
Abstract
Massive tears of the rotator cuff (RC) are often associated with progressive and irreversible muscle degeneration due to fibrosis, fatty infiltration, and muscle atrophy. RC tears are common in individuals older than 60 years and the repair of these tears is amongst the most prevalent of orthopedic procedures. However, most current models of this injury are established in young animals, which may not accurately recapitulate the clinical condition. In this study, we used a murine model of massive RC tears to evaluate age-related muscle degeneration following chronic injury. The expression of the fibro-adipogenic genes encoding collagen type III and leptin was higher in aged RC compared with matched injured young tissue at 2 weeks post-injury, and development of fibrosis was accelerated in aged mice within 5 days post-injury. Furthermore, the synthesis of collagens type I and III and fat tissue accumulation were significantly higher in injured RCs of aged mice. Similar frequency of fibro-adipogenic PDGFRβ+ PDGFRα+ progenitor cells was measured in non-injured RC of aged and young mice, but PDGFRβ+ PDGFRα+ cells contributed to significantly larger fibrotic lesions in aged RCs within 2 weeks post-injury, implying a more robust fibrotic environment in the aged injured muscle. Altogether, these findings demonstrate age-dependent differences in RC response to chronic injury with a more profound fibro-adipogenic change in aged muscles. Clinically, cell therapies for muscular pathologies should not only consider the cell type being transplanted but also the recipient milieu into which these cells are seeded. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:320-328, 2020.
Collapse
Affiliation(s)
- Abhinav K Sharma
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Brandon Levian
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Paras Shah
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Gina M Mosich
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Regina Husman
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Allison Ariniello
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Jonathan D Gatto
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Vivian J Hu
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Daniel J McClintick
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Andrew R Jensen
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - David R McAllister
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Bruno Péault
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245.,Center for Cardiovascular Science and MRC, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Ayelet Dar
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, David Geffen School of Medicine, Orthopaedic Hospital Research Center, University of California, Toyota Sports Performance Center 555 N. Nash St., Suite BEl Segundo, Los Angeles, California, 90245
| |
Collapse
|
34
|
Mosich GM, Husman R, Shah P, Sharma A, Rezzadeh K, Aderibigbe T, Hu VJ, McClintick DJ, Wu G, Gatto JD, Xi H, Pyle AD, Péault B, Petrigliano FA, Dar A. Non-fibro-adipogenic pericytes from human embryonic stem cells attenuate degeneration of the chronically injured mouse muscle. JCI Insight 2019; 4:125334. [PMID: 31852842 DOI: 10.1172/jci.insight.125334] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/31/2019] [Indexed: 12/22/2022] Open
Abstract
Massive tears of the rotator cuff (RC) are associated with chronic muscle degeneration due to fibrosis, fatty infiltration, and muscle atrophy. The microenvironment of diseased muscle often impairs efficient engraftment and regenerative activity of transplanted myogenic precursors. Accumulating myofibroblasts and fat cells disrupt the muscle stem cell niche and myogenic cell signaling and deposit excess disorganized connective tissue. Therefore, restoration of the damaged stromal niche with non-fibro-adipogenic cells is a prerequisite to successful repair of an injured RC. We generated from human embryonic stem cells (hES) a potentially novel subset of PDGFR-β+CD146+CD34-CD56- pericytes that lack expression of the fibro-adipogenic cell marker PDGFR-α. Accordingly, the PDGFR-β+PDGFR-α- phenotype typified non-fibro-adipogenic, non-myogenic, pericyte-like derivatives that maintained non-fibro-adipogenic properties when transplanted into chronically injured murine RCs. Although administered hES pericytes inhibited developing fibrosis at early and late stages of progressive muscle degeneration, transplanted PDGFR-β+PDGFR-α+ human muscle-derived fibro-adipogenic progenitors contributed to adipogenesis and greater fibrosis. Additionally, transplanted hES pericytes substantially attenuated muscle atrophy at all tested injection time points after injury. Coinciding with this observation, conditioned medium from cultured hES pericytes rescued atrophic myotubes in vitro. These findings imply that non-fibro-adipogenic hES pericytes recapitulate the myogenic stromal niche and may be used to improve cell-based treatments for chronic muscle disorders.
Collapse
Affiliation(s)
- Gina M Mosich
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| | - Regina Husman
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| | - Paras Shah
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| | - Abhinav Sharma
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| | - Kevin Rezzadeh
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| | | | - Vivian J Hu
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| | | | - Genbin Wu
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| | - Jonathan D Gatto
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| | - Haibin Xi
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, UCLA, California, USA
| | - April D Pyle
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, UCLA, California, USA
| | - Bruno Péault
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, UCLA, California, USA.,Center for Cardiovascular Science and MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Frank A Petrigliano
- Epstein Family Center for Sports Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Ayelet Dar
- Orthopaedic Hospital Research Center, David Geffen School of Medicine, and
| |
Collapse
|
35
|
Wang Y, Xu J, Meyers CA, Gao Y, Tian Y, Broderick K, Peault B, James AW. PDGFRα marks distinct perivascular populations with different osteogenic potential within adipose tissue. Stem Cells 2019; 38:276-290. [PMID: 31742801 DOI: 10.1002/stem.3108] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/11/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022]
Abstract
The perivascular niche within adipose tissue is known to house multipotent cells, including osteoblast precursors. However, the identity of perivascular subpopulations that may mineralize or ossify most readily is not known. Here, we utilize inducible PDGFRα (platelet-derived growth factor alpha) reporter animals to identify subpopulations of perivascular progenitor cells. Results showed that PDGFRα-expressing cells are present in four histologic niches within inguinal fat, including two perivascular locations. PDGFRα+ cells are most frequent within the tunica adventitia of arteries and veins, where PDGFRα+ cells populate the inner aspects of the adventitial layer. Although both PDGFRα+ and PDGFRα- fractions are multipotent progenitor cells, adipose tissue-derived PDGFRα+ stromal cells proliferate faster and mineralize to a greater degree than their PDGFRα- counterparts. Likewise, PDGFRα+ ectopic implants reconstitute the perivascular niche and ossify to a greater degree than PDGFRα- cell fractions. Adventicytes can be further grouped into three distinct groups based on expression of PDGFRα and/or CD34. When further partitioned, adventicytes co-expressing PDGFRα and CD34 represented a cell fraction with the highest mineralization potential. Long-term tracing studies showed that PDGFRα-expressing adventicytes give rise to adipocytes, but not to other cells within the vessel wall under homeostatic conditions. However, upon bone morphogenetic protein 2 (BMP2)-induced ossicle formation, descendants of PDGFRα+ cells gave rise to osteoblasts, adipocytes, and "pericyte-like" cells within the ossicle. In sum, PDGFRα marks distinct perivascular osteoprogenitor cell subpopulations within adipose tissue. The identification of perivascular osteoprogenitors may contribute to our improved understanding of pathologic mineralization/ossification.
Collapse
Affiliation(s)
- Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Yongxing Gao
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Ye Tian
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Kristen Broderick
- Department of Plastic Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California.,Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| |
Collapse
|
36
|
Fitzsimmons REB, Mazurek MS, Soos A, Simmons CA. Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering. Stem Cells Int 2018; 2018:8031718. [PMID: 30210552 PMCID: PMC6120267 DOI: 10.1155/2018/8031718] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/31/2018] [Accepted: 07/17/2018] [Indexed: 02/08/2023] Open
Abstract
As a result of over five decades of investigation, mesenchymal stromal/stem cells (MSCs) have emerged as a versatile and frequently utilized cell source in the fields of regenerative medicine and tissue engineering. In this review, we summarize the history of MSC research from the initial discovery of their multipotency to the more recent recognition of their perivascular identity in vivo and their extraordinary capacity for immunomodulation and angiogenic signaling. As well, we discuss long-standing questions regarding their developmental origins and their capacity for differentiation toward a range of cell lineages. We also highlight important considerations and potential risks involved with their isolation, ex vivo expansion, and clinical use. Overall, this review aims to serve as an overview of the breadth of research that has demonstrated the utility of MSCs in a wide range of clinical contexts and continues to unravel the mechanisms by which these cells exert their therapeutic effects.
Collapse
Affiliation(s)
- Ross E. B. Fitzsimmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Matthew S. Mazurek
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB, Canada T2N 4Z6
| | - Agnes Soos
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Craig A. Simmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada M5S 3G8
| |
Collapse
|