1
|
Shahin-Shamsabadi A, Cappuccitti J. Muscle-specific acellular ECM fibers made with anchored cell sheet engineering support regeneration in rat models of volumetric muscle loss. Acta Biomater 2025:S1742-7061(25)00348-4. [PMID: 40399155 DOI: 10.1016/j.actbio.2025.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
Volumetric muscle loss (VML), a condition affecting millions due to trauma, represents a critical unmet need in regenerative medicine, with no established standard of care. This study introduces a de novo therapeutic strategy using tissue-specific skeletal muscle acellular extracellular matrix (aECM) fibers fabricated using scaffold-free Anchored Cell Sheet Engineering technique. These engineered fibers replicate the native ECM composition and microarchitecture of skeletal muscle, incorporating essential structural and basement membrane proteins. In a rat VML model, aECM demonstrated promising regenerative capacity compared to commercial porcine-derived small intestine submucosa (SIS) ECM. Over an 8-week period, compared to contralateral muscle, aECM preserved muscle volume and weight, regulated inflammatory and fibrotic responses, and promoted vascularization. In contrast, SIS was rapidly degraded by week 4 and associated with fibrosis. Force recovery in aECM was lower at the 8-week time point (77 % compared to 91 % in control), but histological and immunohistochemical analyses revealed newly formed, dispersed muscle fibers exclusively in aECM treatment. Importantly, muscle weight was preserved only when aECM was used, resulting in similar normalized force-to-weight across all groups (87 % in aECM vs. 88 % in SIS). The histological analyses further demonstrated ongoing tissue remodeling, indicative of sustained regeneration, in contrast to the premature fibrotic healing observed in the other groups. An innovative quantitative image analysis workflow enabled assessment of spatial tissue heterogeneity through histology and immunohistochemistry images, setting a new standard for regeneration analysis. These findings establish engineered tissue-specific aECM as a transformative approach for VML treatment, laying the groundwork for translation to clinical applications. STATEMENT OF SIGNIFICANCE: The current study introduces a transformative approach to treating volumetric muscle loss (VML) through the development of tissue-specific acellular extracellular matrix (aECM) fibers engineered using a scaffold-free biofabrication platform uniquely suited for recreation of such aECM components. The engineered fibers represent a significant advancement over current commercial options by recreating native ECM composition and microarchitecture while eliminating complications associated with xenogenic materials. Through comprehensive in vivo evaluation in a rat model, it is demonstrated that these engineered fibers maintain muscle mass and promote controlled tissue regeneration, addressing key limitations of existing treatments. The scaffold-free biofabrication of tissue-specific aECM provides a new paradigm for biomaterial design in regenerative medicine.
Collapse
Affiliation(s)
| | - John Cappuccitti
- Evolved.Bio, 280 Joseph Street, Kitchener, Ontario, N2G4Z5, Canada
| |
Collapse
|
2
|
Heisser RH, Bawa M, Shah J, Bu A, Raman R. Soft Biological Actuators for Meter-Scale Homeostatic Biohybrid Robots. Chem Rev 2025; 125:3976-4007. [PMID: 40138615 DOI: 10.1021/acs.chemrev.4c00785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Skeletal muscle's elegant protein-based architecture powers motion throughout the animal kingdom, with its constituent actomyosin complexes driving intra- and extra-cellular motion. Classical motors and recently developed soft actuators cannot match the packing density and contractility of individual muscle fibers that scale to power the motion of ants and elephants alike. Accordingly, the interdisciplinary fields of robotics and tissue engineering have combined efforts to build living muscle actuators that can power a new class of robots to be more energy-efficient, dexterous, and safe than existing motor-powered and hydraulic paradigms. Doing so ethically and at scale─creating meter-scale tissue constructs from sustainable muscle progenitor cell lines─has inspired innovations in biomaterials and tissue culture methodology. We weave discussions of muscle cell biology, materials chemistry, tissue engineering, and biohybrid design to review the state of the art in soft actuator biofabrication. Looking forward, we outline a vision for meter-scale biohybrid robotic systems and tie discussions of recent progress to long-term research goals.
Collapse
Affiliation(s)
- Ronald H Heisser
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Maheera Bawa
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Jessica Shah
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 45 Carleton St., Cambridge, Massachusetts 02142, United States of America
| | - Angel Bu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Ritu Raman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| |
Collapse
|
3
|
Tiskratok W, Chuinsiri N, Limraksasin P, Kyawsoewin M, Jitprasertwong P. Extracellular Matrix Stiffness: Mechanotransduction and Mechanobiological Response-Driven Strategies for Biomedical Applications Targeting Fibroblast Inflammation. Polymers (Basel) 2025; 17:822. [PMID: 40292716 PMCID: PMC11946729 DOI: 10.3390/polym17060822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
The extracellular matrix (ECM) is a dynamic network providing mechanical and biochemical cues that regulate cellular behavior. ECM stiffness critically influences fibroblasts, the primary ECM producers, particularly in inflammation and fibrosis. This review explores the role of ECM stiffness in fibroblast-driven inflammation and tissue remodeling, focusing on the physicochemical and biological mechanisms involved. Engineered materials, hydrogels, and polydimethylsiloxane (PDMS) are highlighted for replicating tissue-specific stiffness, enabling precise control over cell-matrix interactions. The surface functionalization of substrate materials, including collagen, polydopamine, and fibronectin, enhances bioactivity and fibroblast adhesion. Key mechanotransduction pathways, such as integrin signaling and YAP/TAZ activation, are related to regulating fibroblast behaviors and inflammatory responses. The role of fibroblasts in driving chronic inflammatory diseases emphasizes their therapeutic potentials. Advances in ECM-modifying strategies, including tunable biomaterials and hydrogel-based therapies, are explored for applications in tissue engineering, drug delivery, anti-inflammatory treatments, and diagnostic tools for the accurate diagnosis and prognosis of ECM stiffness-related inflammatory diseases. This review integrates mechanobiology with biomedical innovations, providing a comprehensive prognosis of fibroblast responses to ECM stiffness and outlining future directions for targeted therapies.
Collapse
Affiliation(s)
- Watcharaphol Tiskratok
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| | - Nontawat Chuinsiri
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| | - Phoonsuk Limraksasin
- Center of Excellence for Dental Stem Cell Biology, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (P.L.); (M.K.)
| | - Maythwe Kyawsoewin
- Center of Excellence for Dental Stem Cell Biology, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (P.L.); (M.K.)
| | - Paiboon Jitprasertwong
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| |
Collapse
|
4
|
Shi L, Ding Z, Chen J. Deciphering the role of IGFBP5 in delaying fibrosis and sarcopenia in aging skeletal muscle: therapeutic implications and molecular mechanisms. Front Pharmacol 2025; 16:1557703. [PMID: 40144669 PMCID: PMC11937025 DOI: 10.3389/fphar.2025.1557703] [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: 01/09/2025] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Sarcopenia is a condition characterized by the loss of muscle fibers and excessive deposition of extracellular matrix proteins. The interplay between muscle atrophy and fibrosis is a central feature of sarcopenia. While the mechanisms underlying skeletal muscle aging and fibrosis remain incompletely understood, cellular senescence has emerged as a key contributor. This study investigates the role of D-galactose (D-gal) in inducing fibroblasts senescence and skeletal muscle fibrosis, and aims to find the key regulator of the process to serve as a therapeutical target. Methods To discover the role of D-gal in inducing cellular senescence and fibrosis, the senescence markers and the expression of fibrosis-related proteins were assessed after introducing D-gal among fibroblasts, and muscle strength and mass. The severity of muscle atrophy and fibrosis were also verified by using H&E staining and Masson trichrome staining after D-gal treatment via subcutaneous injection among mice. Subsequently, mRNA sequencing (RNA-seq) was performed and the differential expressed genes were identified between under D-gal or control treatment, to discover the key regulator of D-GAL-driven fibroblasts senescence and fibrosis. The role of the key regulator IGFBP5 were then validated in D-GAL treated IGFBP5-knockdown fibroblasts in vitro by analyzing the level of senescence and fibrosis-related markers. And the results were further confirmed in vivo in IGFBP5-knockdown SAMP8 mice with histological examinations. Results D-gal treatment effectively induced cellular senescence and fibrosis in fibroblasts, as well as skeletal muscle atrophy, fibrosis and loss in muscle mass and function in mice. IGFBP5 was identified as a key regulator of D-GAL induced senescence and fibrosis among fibroblasts using RNA-seq. And further validation tests showed that IGFBP5-knockdown could alleviate D-GAL-induced fibroblast cellular senescence and fibrosis, as well as the severity of muscle atrophy and fibrosis in SAMP8 mice. Discussion IGFBP5 emerging as a key regulator of D-GAL-induced fibroblast cellular senescence and fibrosis. The findings provide new insights into the molecular mechanisms underlying age-related skeletal muscle fibrosis and highlight IGFBP5 as a potential therapeutic target. Further research is needed to validate these findings and explore related clinical applications.
Collapse
Affiliation(s)
| | - Zheci Ding
- *Correspondence: Zheci Ding, ; Jiwu Chen,
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
5
|
Patel PR, Tamas IP, Van Der Bas M, Kegg A, Hilliard BA, Lambi AG, Popoff SN, Barbe MF. Repetitive Overuse Injury Causes Entheseal Damage and Palmar Muscle Fibrosis in Older Rats. Int J Mol Sci 2024; 25:13546. [PMID: 39769311 PMCID: PMC11679654 DOI: 10.3390/ijms252413546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Overuse injury is a frequent diagnosis in occupational medicine and athletics. Using an established model of upper extremity overuse, we sought to characterize changes occurring in the forepaws and forelimbs of mature female rats (14-18 months of age). Thirty-three rats underwent a 4-week shaping period, before performing a high-repetition low-force (HRLF) task for 12 weeks, with the results being compared to 32 mature controls. HRLF animals showed a reduced grip strength versus controls. ELISAs carried out in the HRLF rats, versus controls, showed elevated levels of IL1-α in tendons, IL1-α and TNF-α in distal bones/entheses, and TNF-α, MIP1-α/CCL3, and CINC-2/CXCL-3 in serum, as well as IL-6 in forelimb muscles and tendons, and IL-10 in serum. HRLF rats had elevated collagen deposition in the forepaw intrinsic muscles (i.e., fibrosis), entheseal microdamage, and articular cartilage degradation versus the control rats. CD68/ED1+ osteoclasts and single-nucleated cells were elevated in distal forelimb metaphyses of the HRLF animals, versus controls. Declines in grip strength correlated with muscle fibrosis, entheseal microdamage, articular cartilage damage, distal bone/enthesis IL1-α, and serum IL-6. These data demonstrate inflammatory and persistent degradative changes in the forearm/forepaw tissues of mature female animals exposed to prolonged repetitive tasks, changes with clinical relevance to work-related overuse injuries in mature human females.
Collapse
Affiliation(s)
- Parth R. Patel
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Istvan P. Tamas
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Megan Van Der Bas
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Abby Kegg
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Brendan A. Hilliard
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
| | - Alex G. Lambi
- Plastic Surgery Section, New Mexico Veterans Administration Health Care System, Albuquerque, NM 87108, USA;
- Department of Surgery, The University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Steven N. Popoff
- Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA;
| | - Mary F. Barbe
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (P.R.P.); (I.P.T.); (M.V.D.B.); (A.K.); (B.A.H.)
- Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA;
| |
Collapse
|
6
|
Chinvattanachot G, Rivas D, Duque G. Mechanisms of muscle cells alterations and regeneration decline during aging. Ageing Res Rev 2024; 102:102589. [PMID: 39566742 DOI: 10.1016/j.arr.2024.102589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Skeletal muscles are essential for locomotion and body metabolism regulation. As muscles age, they lose strength, elasticity, and metabolic capability, leading to ineffective motion and metabolic derangement. Both cellular and extracellular alterations significantly influence muscle aging. Satellite cells (SCs), the primary muscle stem cells responsible for muscle regeneration, become exhausted, resulting in diminished population and functionality during aging. This decline in SC function impairs intercellular interactions as well as extracellular matrix production, further hindering muscle regeneration. Other muscle-resident cells, such as fibro-adipogenic progenitors (FAPs), pericytes, and immune cells, also deteriorate with age, reducing local growth factor activities and responsiveness to stress or injury. Systemic signaling, including hormonal changes, contributes to muscle cellular catabolism and disrupts muscle homeostasis. Collectively, these cellular and environmental components interact, disrupting muscle homeostasis and regeneration in advancing age. Understanding these complex interactions offers insights into potential regenerative strategies to mitigate age-related muscle degeneration.
Collapse
Affiliation(s)
- Guntarat Chinvattanachot
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - Daniel Rivas
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Gustavo Duque
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
7
|
Gustafsson T, Ulfhake B. Aging Skeletal Muscles: What Are the Mechanisms of Age-Related Loss of Strength and Muscle Mass, and Can We Impede Its Development and Progression? Int J Mol Sci 2024; 25:10932. [PMID: 39456714 PMCID: PMC11507513 DOI: 10.3390/ijms252010932] [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: 09/03/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
As we age, we lose muscle strength and power, a condition commonly referred to as sarcopenia (ICD-10-CM code (M62.84)). The prevalence of sarcopenia is about 5-10% of the elderly population, resulting in varying degrees of disability. In this review we emphasise that sarcopenia does not occur suddenly. It is an aging-induced deterioration that occurs over time and is only recognised as a disease when it manifests clinically in the 6th-7th decade of life. Evidence from animal studies, elite athletes and longitudinal population studies all confirms that the underlying process has been ongoing for decades once sarcopenia has manifested. We present hypotheses about the mechanism(s) underlying this process and their supporting evidence. We briefly review various proposals to impede sarcopenia, including cell therapy, reducing senescent cells and their secretome, utilising targets revealed by the skeletal muscle secretome, and muscle innervation. We conclude that although there are potential candidates and ongoing preclinical and clinical trials with drug treatments, the only evidence-based intervention today for humans is exercise. We present different exercise programmes and discuss to what extent the interindividual susceptibility to developing sarcopenia is due to our genetic predisposition or lifestyle factors.
Collapse
Affiliation(s)
| | - Brun Ulfhake
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden;
| |
Collapse
|
8
|
Pallotta I, Stec MJ, Schriver B, Golann DR, Considine K, Su Q, Barahona V, Napolitano JE, Stanley S, Garcia M, Feric NT, Durney KM, Aschar‐Sobbi R, Bays N, Shavlakadze T, Graziano MP. Electrical stimulation of biofidelic engineered muscle enhances myotube size, force, fatigue resistance, and induces a fast-to-slow-phenotype shift. Physiol Rep 2024; 12:e70051. [PMID: 39384537 PMCID: PMC11464147 DOI: 10.14814/phy2.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 10/11/2024] Open
Abstract
Therapeutic development for skeletal muscle diseases is challenged by a lack of ex vivo models that recapitulate human muscle physiology. Here, we engineered 3D human skeletal muscle tissue in the Biowire II platform that could be maintained and electrically stimulated long-term. Increasing differentiation time enhanced myotube formation, modulated myogenic gene expression, and increased twitch and tetanic forces. When we mimicked exercise training by applying chronic electrical stimulation, the "exercised" skeletal muscle tissues showed increased myotube size and a contractility profile, fatigue resistance, and gene expression changes comparable to in vivo models of exercise training. Additionally, tissues also responded with expected physiological changes to known pharmacological treatment. To our knowledge, this is the first evidence of a human engineered 3D skeletal muscle tissue that recapitulates in vivo models of exercise. By recapitulating key features of human skeletal muscle, we demonstrated that the Biowire II platform may be used by the pharmaceutical industry as a model for identifying and optimizing therapeutic drug candidates that modulate skeletal muscle function.
Collapse
Affiliation(s)
| | | | | | | | | | - Qi Su
- Regeneron PharmaceuticalsTarrytownNew YorkUSA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Beas-Jiménez JD, León Garrigosa A, Doñoro Cuevas P, Álvarez Recio MI, Cofré Bolados C, Alonso JM. Fibrosis and Sports Injuries: Concept and Implications: The GESMUTE-SETRADE Consensus Statement. Orthop J Sports Med 2024; 12:23259671241266604. [PMID: 39291125 PMCID: PMC11406605 DOI: 10.1177/23259671241266604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 02/16/2024] [Indexed: 09/19/2024] Open
Abstract
Background Musculoskeletal fibrosis can be frequently observed in the scope of sports medicine; however, there is no consensus regarding its definition, nor do we have substantial knowledge regarding its epidemiology or best therapeutic alternatives. Purpose The GESMUTE (Group for the Study of the Muscle Tendon System) Epidemiology Group, integrated into SETRADE (The Spanish Society for Sports Traumatology) propose a definition for musculoskeletal fibrosis within the field of physical exercise. Study Design Consensus statement. Methods A bibliographic review of the existing scientific evidence and consensus was developed by the authors on the definition of fibrosis in the field of sport. Results Our working group proposed the definition of fibrosis as an abnormal accumulation of extracellular matrix, usually with regards to an injury complication, showing various clinical findings affecting muscles, tendons, ligaments, articular capsules and nerves which, in turn, mars the recovery process, causing symptoms and finally leading to relapses. Conclusion Fibrosis in sport may be considered as an abnormal accumulation of extracellular matrix, usually related to complications of an injury, showing a wide range of clinical symptomatology, affecting muscles, tendons, ligaments, articular capsules, and nerves.
Collapse
Affiliation(s)
| | | | | | | | | | - J M Alonso
- Department of Sports Medicine, Aspetar Hospital, Doha, Qatar
| |
Collapse
|
10
|
Cumming KT, Reitzner SM, Hanslien M, Skilnand K, Seynnes OR, Horwath O, Psilander N, Sundberg CJ, Raastad T. Muscle memory in humans: evidence for myonuclear permanence and long-term transcriptional regulation after strength training. J Physiol 2024; 602:4171-4193. [PMID: 39159314 DOI: 10.1113/jp285675] [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: 09/11/2023] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
The objective of this work was to investigate myonuclear permanence and transcriptional regulation as mechanisms for cellular muscle memory after strength training in humans. Twelve untrained men and women performed 10 weeks of unilateral elbow-flexor strength training followed by 16 weeks of de-training. Thereafter, 10 weeks' re-training was conducted with both arms: the previously trained arm and the contralateral untrained control arm. Muscle biopsies were taken from the trained arm before and after both training periods and from the control arm before and after re-training. Muscle biopsies were analysed for fibre cross-sectional area (fCSA), myonuclei and global transcriptomics (RNA sequencing). During the first training period, myonuclei increased in type 1 (13 ± 17%) and type 2 (33 ± 23%) fibres together with a 30 ± 43% non-significant increase in mixed fibre fCSA (P = 0.069). Following de-training, fCSA decreased in both fibre types, whereas myonuclei were maintained, resulting in 33% higher myonuclear number in previously trained vs. control muscle in type 2 fibres. Furthermore, in the previously trained muscle, three differentially expressed genes (DEGs; EGR1, MYL5 and COL1A1) were observed. Following re-training, the previously trained muscle showed larger type 2 fCSA compared to the control (P = 0.035). However, delta change in type 2 fCSA was not different between muscles. Gene expression was more dramatically changed in the control arm (1338 DEGs) than in the previously trained arm (822 DEGs). The sustained higher number of myonuclei in the previously trained muscle confirms myonuclear accretion and permanence in humans. Nevertheless, because of the unclear effect on the subsequent hypertrophy with re-training, the physiological benefit remains to be determined. KEY POINTS: Muscle memory is a cellular mechanism that describes the capacity of skeletal muscle fibres to respond differently to training stimuli if the stimuli have been previously encountered. This study overcomes past methodological limitations related to the choice of muscles and analytical procedures. We show that myonuclear number is increased after strength training and maintained during de-training. Increased myonuclear number and differentially expressed genes related to muscle performance and development in the previously trained muscle did not translate into a clearly superior responses during re-training. Because of the unclear effect on the subsequent hypertrophy and muscle strength gain with re-training, the physiological benefit remains to be determined.
Collapse
Affiliation(s)
- Kristoffer Toldnes Cumming
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
- Faculty of Health, Welfare and Organisation, Østfold University College, Fredrikstad, Norway
| | - Stefan Markus Reitzner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Marit Hanslien
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Kenneth Skilnand
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Olivier R Seynnes
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Oscar Horwath
- Department of Physiology, Nutrition and Biomechanics, Åstrand Laboratory, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Niklas Psilander
- Department of Physiology, Nutrition and Biomechanics, Åstrand Laboratory, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Carl Johan Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Learning, Informatics, Management and Ethics, Karolinska Institutet, Stockholm, Sweden
| | - Truls Raastad
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| |
Collapse
|
11
|
Nguyen ML, Demri N, Lapin B, Di Federico F, Gropplero G, Cayrac F, Hennig K, Gomes ER, Wilhelm C, Roman W, Descroix S. Studying the impact of geometrical and cellular cues on myogenesis with a skeletal muscle-on-chip. LAB ON A CHIP 2024; 24:4147-4160. [PMID: 39072529 DOI: 10.1039/d4lc00417e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In the skeletal muscle tissue, cells are organized following an anisotropic architecture, which is both required during myogenesis when muscle precursor cells fuse to generate myotubes and for its contractile function. To build an in vitro skeletal muscle tissue, it is therefore essential to develop methods to organize cells in an anisotropic fashion, which can be particularly challenging, especially in 3D. In this study, we present a versatile muscle-on-chip system with adjustable collagen hollow tubes that can be seeded with muscle precursor cells. The collagen acts both as a tube-shaped hollow mold and as an extracellular matrix scaffold that can house other cell types for co-culture. We found that the diameter of the channel affects the organization of the muscle cells and that proper myogenesis was obtained at a diameter of 75 μm. In these conditions, muscle precursor cells fused into long myotubes aligned along these collagen channels, resulting in a fascicle-like structure. These myotubes exhibited actin striations and upregulation of multiple myogenic genes, reflecting their maturation. Moreover, we showed that our chip allowed muscle tissue culture and maturation over a month, with the possibility of fibroblast co-culture embedding in the collagen matrix.
Collapse
Affiliation(s)
- M-L Nguyen
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - N Demri
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - B Lapin
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - F Di Federico
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - G Gropplero
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - F Cayrac
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - K Hennig
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - C Wilhelm
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - W Roman
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Australian Regenerative Medicine Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - S Descroix
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| |
Collapse
|
12
|
Bagri KM, de Andrade Abraham C, Santos AT, da Silva WS, Costa ML, Mermelstein C. Rotenone inhibits embryonic chick myogenesis in a ROS-dependent mechanism. Tissue Cell 2024; 89:102423. [PMID: 38875923 DOI: 10.1016/j.tice.2024.102423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/16/2024]
Abstract
Skeletal muscle function is highly dependent on the energy supply provided by mitochondria. Besides ATP production, mitochondria have several other roles, such as calcium storage, heat production, cell death signaling, autophagy regulation and redox state modulation. Mitochondrial function is crucial for skeletal muscle fiber formation. Disorders that affect mitochondria have a major impact in muscle development and function. Here we studied the role of mitochondria during chick skeletal myogenesis. We analyzed the intracellular distribution of mitochondria in myoblasts, fibroblasts and myotubes using Mitotracker labeling. Mitochondrial respiration was investigated in chick muscle cells. Our results show that (i) myoblasts and myotubes have more mitochondria than muscle fibroblasts; (ii) mitochondria are organized in long lines within the whole cytoplasm and around the nuclei of myotubes, while in myoblasts they are dispersed in the cytoplasm; (iii) the area of mitochondria in myotubes increases during myogenesis, while in myoblasts and fibroblasts there is a slight decrease; (iv) mitochondrial length increases in the three cell types (myoblasts, fibroblasts and myotubes) during myogenesis; (v) the distance of mitochondria to the nucleus increases in myoblasts and myotubes during myogenesis; (vi) Rotenone inhibits muscle fiber formation, while FCCP increases the size of myotubes; (vii) N-acetyl cysteine (NAC), an inhibitor of ROS formation, rescues the effects of Rotenone on muscle fiber size; and (viii) Rotenone induces the production of ROS in chick myogenic cells. The collection of our results suggests a role of ROS signaling in mitochondrial function during chick myogenesis.
Collapse
Affiliation(s)
- Kayo Moreira Bagri
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Anderson Teixeira Santos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Wagner Seixas da Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Manoel Luis Costa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Claudia Mermelstein
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
13
|
Luo W, Zhang H, Wan R, Cai Y, Liu Y, Wu Y, Yang Y, Chen J, Zhang D, Luo Z, Shang X. Biomaterials-Based Technologies in Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2024; 13:e2304196. [PMID: 38712598 DOI: 10.1002/adhm.202304196] [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: 11/28/2023] [Revised: 04/26/2024] [Indexed: 05/08/2024]
Abstract
For many clinically prevalent severe injuries, the inherent regenerative capacity of skeletal muscle remains inadequate. Skeletal muscle tissue engineering (SMTE) seeks to meet this clinical demand. With continuous progress in biomedicine and related technologies including micro/nanotechnology and 3D printing, numerous studies have uncovered various intrinsic mechanisms regulating skeletal muscle regeneration and developed tailored biomaterial systems based on these understandings. Here, the skeletal muscle structure and regeneration process are discussed and the diverse biomaterial systems derived from various technologies are explored in detail. Biomaterials serve not merely as local niches for cell growth, but also as scaffolds endowed with structural or physicochemical properties that provide tissue regenerative cues such as topographical, electrical, and mechanical signals. They can also act as delivery systems for stem cells and bioactive molecules that have been shown as key participants in endogenous repair cascades. To achieve bench-to-bedside translation, the typical effect enabled by biomaterial systems and the potential underlying molecular mechanisms are also summarized. Insights into the roles of biomaterials in SMTE from cellular and molecular perspectives are provided. Finally, perspectives on the advancement of SMTE are provided, for which gene therapy, exosomes, and hybrid biomaterials may hold promise to make important contributions.
Collapse
Affiliation(s)
- Wei Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Hanli Zhang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Renwen Wan
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yuxi Cai
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yinuo Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yang Wu
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yimeng Yang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jiani Chen
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, Hong Kong
| | - Zhiwen Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xiliang Shang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| |
Collapse
|
14
|
Nelke C, Schroeter CB, Theissen L, Preusse C, Pawlitzki M, Räuber S, Dobelmann V, Cengiz D, Kleefeld F, Roos A, Schoser B, Brunn A, Neuen-Jacob E, Zschüntzsch J, Meuth SG, Stenzel W, Ruck T. Senescent fibro-adipogenic progenitors are potential drivers of pathology in inclusion body myositis. Acta Neuropathol 2023; 146:725-745. [PMID: 37773216 PMCID: PMC10564677 DOI: 10.1007/s00401-023-02637-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Inclusion body myositis (IBM) is unique across the spectrum of idiopathic inflammatory myopathies (IIM) due to its distinct clinical presentation and refractoriness to current treatment approaches. One explanation for this resistance may be the engagement of cell-autonomous mechanisms that sustain or promote disease progression of IBM independent of inflammatory activity. In this study, we focused on senescence of tissue-resident cells as potential driver of disease. For this purpose, we compared IBM patients to non-diseased controls and immune-mediated necrotizing myopathy patients. Histopathological analysis suggested that cellular senescence is a prominent feature of IBM, primarily affecting non-myogenic cells. In-depth analysis by single nuclei RNA sequencing allowed for the deconvolution and study of muscle-resident cell populations. Among these, we identified a specific cluster of fibro-adipogenic progenitors (FAPs) that demonstrated key hallmarks of senescence, including a pro-inflammatory secretome, expression of p21, increased β-galactosidase activity, and engagement of senescence pathways. FAP function is required for muscle cell health with changes to their phenotype potentially proving detrimental. In this respect, the transcriptomic landscape of IBM was also characterized by changes to the myogenic compartment demonstrating a pronounced loss of type 2A myofibers and a rarefication of acetylcholine receptor expressing myofibers. IBM muscle cells also engaged a specific pro-inflammatory phenotype defined by intracellular complement activity and the expression of immunogenic surface molecules. Skeletal muscle cell dysfunction may be linked to FAP senescence by a change in the collagen composition of the latter. Senescent FAPs lose collagen type XV expression, which is required to support myofibers' structural integrity and neuromuscular junction formation in vitro. Taken together, this study demonstrates an altered phenotypical landscape of muscle-resident cells and that FAPs, and not myofibers, are the primary senescent cell type in IBM.
Collapse
Affiliation(s)
- Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Lukas Theissen
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Corinna Preusse
- Department of Neuropathology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Saskia Räuber
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Vera Dobelmann
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Derya Cengiz
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Felix Kleefeld
- Department of Neurology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Andreas Roos
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, Centre for Neuromuscular Disorders in Children, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Benedikt Schoser
- Friedrich Baur Institute at the Department of Neurology, LMU University Hospital, LMU Munich, 80336, Munich, Germany
| | - Anna Brunn
- Institute of Neuropathology, Heinrich Heine University, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Eva Neuen-Jacob
- Institute of Neuropathology, Heinrich Heine University, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.
| |
Collapse
|
15
|
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
|
16
|
GrönholdtKlein M, Gorzi A, Wang L, Edström E, Rullman E, Altun M, Ulfhake B. Emergence and Progression of Behavioral Motor Deficits and Skeletal Muscle Atrophy across the Adult Lifespan of the Rat. BIOLOGY 2023; 12:1177. [PMID: 37759577 PMCID: PMC10526071 DOI: 10.3390/biology12091177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
The facultative loss of muscle mass and function during aging (sarcopenia) poses a serious threat to our independence and health. When activities of daily living are impaired (clinical phase), it appears that the processes leading to sarcopenia have been ongoing in humans for decades (preclinical phase). Here, we examined the natural history of sarcopenia in male outbred rats to compare the occurrence of motor behavioral deficits with the degree of muscle wasting and to explore the muscle-associated processes of the preclinical and clinical phases, respectively. Selected metrics were validated in female rats. We used the soleus muscle because of its long duty cycles and its importance in postural control. Results show that gait and coordination remain intact through middle age (40-60% of median lifespan) when muscle mass is largely preserved relative to body weight. However, the muscle shows numerous signs of remodeling with a shift in myofiber-type composition toward type I. As fiber-type prevalence shifted, fiber-type clustering also increased. The number of hybrid fibers, myofibers with central nuclei, and fibers expressing embryonic myosin increased from being barely detectable to a significant number (5-10%) at late middle age. In parallel, TGFβ1, Smad3, FBXO32, and MuRF1 mRNAs increased. In early (25-month-old) and advanced (30-month-old) aging, gait and coordination deteriorate with the progressive loss of muscle mass. In late middle age and early aging due to type II atrophy (>50%) followed by type I atrophy (>50%), the number of myofibers did not correlate with this process. In advanced age, atrophy is accompanied by a decrease in SCs and βCatenin mRNA, whereas several previously upregulated transcripts were downregulated. The re-expression of embryonic myosin in myofibers and the upregulation of mRNAs encoding the γ-subunit of the nicotinic acetylcholine receptor, the neuronal cell adhesion molecule, and myogenin that begins in late middle age suggest that one mechanism driving sarcopenia is the disruption of neuromuscular connectivity. We conclude that sarcopenia in rats, as in humans, has a long preclinical phase in which muscle undergoes extensive remodeling to maintain muscle mass and function. At later time points, these adaptive mechanisms fail, and sarcopenia becomes clinically manifest.
Collapse
Affiliation(s)
- Max GrönholdtKlein
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Ali Gorzi
- Department of Sport Sciences, University of Zanjan, Zanjan 45371-38791, Iran;
| | - Lingzhan Wang
- Department of Human Anatomy, Histology and Embryology, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Erik Edström
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Eric Rullman
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.R.); (M.A.)
| | - Mikael Altun
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.R.); (M.A.)
| | - Brun Ulfhake
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.R.); (M.A.)
| |
Collapse
|
17
|
Kanazawa Y, Miyachi R, Higuchi T, Sato H. Effects of Aging on Collagen in the Skeletal Muscle of Mice. Int J Mol Sci 2023; 24:13121. [PMID: 37685934 PMCID: PMC10487623 DOI: 10.3390/ijms241713121] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Aging affects several tissues in the body, including skeletal muscle. Multiple types of collagens are localized in the skeletal muscle and contribute to the maintenance of normal muscle structure and function. Since the effects of aging on muscle fibers vary by muscle fiber type, it is expected that the effects of aging on intramuscular collagen might be influenced by muscle fiber type. In this study, we examined the effect of aging on collagen levels in the soleus (slow-twitch muscle) and gastrocnemius (fast-twitch muscle) muscles of 3-, 10-, 24-, and 28-month-old male C57BL/6J mice using molecular and morphological analysis. It was found that aging increased collagen I, III, and VI gene expression and immunoreactivity in both slow- and fast-twitch muscles and collagen IV expression in slow-twitch muscles. However, collagen IV gene expression and immunoreactivity in fast-twitch muscle were unaffected by aging. In contrast, the expression of the collagen synthesis marker heat shock protein 47 in both slow- and fast-twitch muscles decreased with aging, while the expression of collagen degradation markers increased with aging. Overall, these results suggest that collagen gene expression and immunoreactivity are influenced by muscle fiber type and collagen type and that the balance between collagen synthesis and degradation tends to tilt toward degradation with aging.
Collapse
Affiliation(s)
- Yuji Kanazawa
- Department of Physical Therapy, Hokuriku University, Kanazawa 920-1180, Ishikawa, Japan;
| | - Ryo Miyachi
- Department of Physical Therapy, Hokuriku University, Kanazawa 920-1180, Ishikawa, Japan;
| | - Takashi Higuchi
- Department of Physical Therapy, Osaka University of Human Sciences, Settsu 566-8501, Osaka, Japan;
| | - Hiaki Sato
- Department of Medical Technology and Clinical Engineering, Hokuriku University, Kanazawa 920-1180, Ishikawa, Japan;
| |
Collapse
|
18
|
Costamagna D, Bastianini V, Corvelyn M, Duelen R, Deschrevel J, De Beukelaer N, De Houwer H, Sampaolesi M, Gayan-Ramirez G, Campenhout AV, Desloovere K. Botulinum Toxin Treatment of Adult Muscle Stem Cells from Children with Cerebral Palsy and hiPSC-Derived Neuromuscular Junctions. Cells 2023; 12:2072. [PMID: 37626881 PMCID: PMC10453788 DOI: 10.3390/cells12162072] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Botulinum neurotoxin type-A (BoNT) injections are commonly used as spasticity treatment in cerebral palsy (CP). Despite improved clinical outcomes, concerns regarding harmful effects on muscle morphology have been raised, and the BoNT effect on muscle stem cells remains not well defined. This study aims at clarifying the impact of BoNT on growing muscles (1) by analyzing the in vitro effect of BoNT on satellite cell (SC)-derived myoblasts and fibroblasts obtained from medial gastrocnemius microbiopsies collected in young BoNT-naïve children (t0) compared to age ranged typically developing children; (2) by following the effect of in vivo BoNT administration on these cells obtained from the same children with CP at 3 (t1) and 6 (t2) months post BoNT; (3) by determining the direct effect of a single and repeated in vitro BoNT treatment on neuromuscular junctions (NMJs) differentiated from hiPSCs. In vitro BoNT did not affect myogenic differentiation or collagen production. The fusion index significantly decreased in CP at t2 compared to t0. In NMJ cocultures, BoNT treatment caused axonal swelling and fragmentation. Repeated treatments impaired the autophagic-lysosomal system. Further studies are warranted to understand the long-term and collateral effects of BoNT in the muscles of children with CP.
Collapse
Affiliation(s)
- Domiziana Costamagna
- Neurorehabilitation Group, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (D.C.); (V.B.); (N.D.B.)
- Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (M.C.); (R.D.); (M.S.)
| | - Valeria Bastianini
- Neurorehabilitation Group, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (D.C.); (V.B.); (N.D.B.)
| | - Marlies Corvelyn
- Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (M.C.); (R.D.); (M.S.)
| | - Robin Duelen
- Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (M.C.); (R.D.); (M.S.)
- Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Jorieke Deschrevel
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.D.); (G.G.-R.)
| | - Nathalie De Beukelaer
- Neurorehabilitation Group, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (D.C.); (V.B.); (N.D.B.)
- Willy Taillard Laboratory of Kinesiology, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland
| | - Hannah De Houwer
- Department of Orthopedic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium; (H.D.H.); (A.V.C.)
| | - Maurilio Sampaolesi
- Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (M.C.); (R.D.); (M.S.)
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.D.); (G.G.-R.)
| | - Anja Van Campenhout
- Department of Orthopedic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium; (H.D.H.); (A.V.C.)
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Kaat Desloovere
- Neurorehabilitation Group, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (D.C.); (V.B.); (N.D.B.)
| |
Collapse
|
19
|
Chatterjee N, Misra SK. Nanocarbon-Enforced Anisotropic MusCAMLR for Rapid Rescue of Mechanically Damaged Skeletal Muscles. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37257065 DOI: 10.1021/acsami.3c01889] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Mechanical damages to skeletal muscles could be detrimental to the active work hours and lifestyle of athletes, mountaineers, and security personnel. In this regard, the slowness of conventional treatment strategies and drug-associated side effects greatly demand the design and development of novel biomaterials, which can rescue such mechanically damaged skeletal muscles. To accomplish this demand, we have developed a musculoresponsive polymer-carbon composite for assisting myotubular regeneration (MusCAMLR). The MusCAMLR is enforced to attain anisotropic muscle-like characteristics while incorporating a smartly passivated nanoscale carbon material in the PNIPAM gel under physiological conditions as a stimulus, which is not achieved by the pristine nanocarbon system. The MusCAMLR establishes a specific mechanical interaction with muscle cells, supports myotube regeneration, maintains excellent mechanical similarity with the myotube, and restores the structural integrity and biochemical parameters of mechanically damaged muscles in a delayed onset muscle soreness (DOMS) rat model within a short period of 72 h. Concisely, this study discloses the potential of smartly passivated nanocarbon in generating an advanced biomaterial system, MusCAMLR, from a regularly used polymeric hydrogel system. This engineered polymer-carbon composite reveals its possible potential to be used as a nondrug therapeutic alternative for rescuing mechanically damaged muscles and probably can be extended for therapy of various other diseases including muscular dystrophy.
Collapse
Affiliation(s)
- Niranjan Chatterjee
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Santosh Kumar Misra
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- The Mehta family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
20
|
Song Y, Yang J, Li T, Sun X, Lin R, He Y, Sun K, Han J, Yang G, Li X, Liu B, Yang D, Dang G, Ma X, Du X, Zhang B, Hu Y, Kong W, Wang X, Zhang H, Xu Q, Feng J. CD34 + cell-derived fibroblast-macrophage cross-talk drives limb ischemia recovery through the OSM-ANGPTL signaling axis. SCIENCE ADVANCES 2023; 9:eadd2632. [PMID: 37043578 DOI: 10.1126/sciadv.add2632] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 03/10/2023] [Indexed: 06/19/2023]
Abstract
CD34+ cells improve the perfusion and function of ischemic limbs in humans and mice. However, there is no direct evidence of the differentiation potential and functional role of these cells in the ischemic muscle microenvironment. Here, we combined the single-cell RNA sequencing and genetic lineage tracing technology, then provided exact single-cell atlases of normal and ischemic limb tissues in human and mouse, and consequently found that bone marrow (BM)-derived macrophages with antigen-presenting function migrated to the ischemic site, while resident macrophages underwent apoptosis. The macrophage oncostatin M (OSM) regulatory pathway was specifically turned on by ischemia. Simultaneously, BM CD34+-derived proregenerative fibroblasts were recruited to the ischemia niche, where they received macrophage-released OSM and promoted angiopoietin-like protein-associated angiogenesis. These findings provided mechanisms on the cellular events and cell-cell communications during tissue ischemia and regeneration and provided evidence that CD34+ cells serve as fibroblast progenitors promoting tissue regeneration.
Collapse
Affiliation(s)
- Yuwei Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Junyao Yang
- Department of Clinical Laboratory, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tianrun Li
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Xiaotong Sun
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruoran Lin
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yangyan He
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Sun
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jingyan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Guangxin Yang
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Xuan Li
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Bo Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dongmin Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Guohui Dang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaolong Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xing Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Bohuan Zhang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hongkun Zhang
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
21
|
Rasaei N, Ghaffarian-Ensaf R, Gholami F, Shiraseb F, Khadem A, Fatemi SF, Mirzaei K. The association between healthy beverage index and sarcopenic obesity among women with overweight and obesity: a cross-sectional study. BMC Endocr Disord 2023; 23:25. [PMID: 36717830 PMCID: PMC9887919 DOI: 10.1186/s12902-023-01274-w] [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: 06/07/2022] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
INTRODUCTION Sarcopenic obesity is related to changes in body composition, loss of muscle mass, and raised adipose tissue. Beverage patterns are effective with changes in health status. Therefore, the aim of this study was to investigate the association between sarcopenic obesity (SO) and the healthy beverage index (HBI) in women with overweight and obesity. METHODS This cross-sectional study conducted on 210 overweight and obese (BMI ≥25 kg/m2) women aged 18-56 years. The measurement of skeletal muscle mass (SMM) and fat mass (FM) done by bioelectric impedance analyzer (BIA) (Inbody Co., Seoul, Korea) based on guidelines. The two lowest quintiles SMM and the two highest quintiles FM and body mass index (BMI) ≥30 are considered sarcopenic obesity in women. A validated and reliable semi-quantitative food-frequency questionnaire (FFQ) was used to evaluate the beverage dietary data. and RFS and NRFS was calculated. Biochemical assessments were quantified by standard approaches, and physical activity were evaluated by international physical activity questionnaire (IPAQ). RESULT In this cross-sectional study, 210 overweight and obese females took part (18-56) years old). The studies were carried out using binary logistic regression. After controlling for a wide variety of confounding variables such as age, energy intake, physical activity, education, and economic status, we found a negative association between HBI and risk of SO (OR = 0.29, 95% CI = 0.35 to 1.01, P = 0.05). CONCLUSION We observed that the odds of SO was reduced by 69% in participants with higher HBI score. More well-designed studies need to confirm our findings.
Collapse
Affiliation(s)
- Niloufar Rasaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box:14155-6117, Tehran, Iran
| | | | - Fatemeh Gholami
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box:14155-6117, Tehran, Iran
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box:14155-6117, Tehran, Iran
| | - Alireza Khadem
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyedeh Fatemeh Fatemi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box:14155-6117, Tehran, Iran
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box:14155-6117, Tehran, Iran.
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Maldonado L, Orozco-Aguilar J, Valero-Breton M, Tacchi F, Cifuentes-Silva E, Cabello-Verrugio C. Differential Fibrotic Response of Muscle Fibroblasts, Myoblasts, and Myotubes to Cholic and Deoxycholic Acids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:219-234. [PMID: 37093430 DOI: 10.1007/978-3-031-26163-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Fibrosis is a condition characterized by an increase in the components of the extracellular matrix (ECM). In skeletal muscle, the cells that participate in the synthesis of ECM are fibroblasts, myoblasts, and myotubes. These cells respond to soluble factors that increase ECM. Fibrosis is a phenomenon that develops in conditions of chronic inflammation, extensive lesions, or chronic diseases. A pathological condition with muscle weakness and increased bile acids (BA) in the blood is cholestatic chronic liver diseases (CCLD). Skeletal muscle expresses the membrane receptor for BA called TGR5. To date, muscle fibrosis in CCLD has not been evaluated. This study aims to assess whether BA can induce a fibrotic condition in muscle fibroblasts, myoblasts, and myotubes. The cells were incubated with deoxycholic (DCA) and cholic (CA) acids, and fibronectin protein levels were evaluated by Western blot. In muscle fibroblasts, both DCA and CA induced an increase in fibronectin protein levels. The same response was found in fibroblasts when activating TGR5 with the specific receptor agonist (INT-777). Interestingly, DCA reduced fibronectin protein levels in both myoblasts and myotubes, while CA did not show changes in fibronectin protein levels in myoblasts and myotubes. These results suggest that DCA and CA can induce a fibrotic phenotype in muscle-derived fibroblasts. On the other hand, DCA decreased the fibronectin in myoblasts and myotubes, whereas CA did not show any effect in these cell populations. Our results show that BA has different effects depending on the cell population to be analyzed.
Collapse
Affiliation(s)
- Luis Maldonado
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, 8370146, Santiago, Chile
- Faculty of Life Sciences, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Josué Orozco-Aguilar
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, 8370146, Santiago, Chile
- Faculty of Life Sciences, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
- Laboratorio de Ensayos Biológicos (LEBi), Universidad de Costa Rica, San José, Costa Rica
- Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| | - Mayalen Valero-Breton
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, 8370146, Santiago, Chile
- Faculty of Life Sciences, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Franco Tacchi
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, 8370146, Santiago, Chile
- Faculty of Life Sciences, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Eduardo Cifuentes-Silva
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, 8370146, Santiago, Chile
- Faculty of Life Sciences, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, 8370146, Santiago, Chile.
- Faculty of Life Sciences, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile.
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
23
|
Demri N, Dumas S, Nguyen M, Gropplero G, Abou‐Hassan A, Descroix S, Wilhelm C. Remote Magnetic Microengineering and Alignment of Spheroids into 3D Cellular Fibers. ADVANCED FUNCTIONAL MATERIALS 2022; 32. [DOI: 10.1002/adfm.202204850] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Indexed: 01/05/2025]
Abstract
AbstractDeveloping in vitro models that recapitulate the in vivo organization of living cells in a 3D microenvironment is one of the current challenges in the field of tissue engineering. In particular for anisotropic tissues where alignment of precursor cells is required for them to create functional structures. Herein, a new method is proposed that allows aligning in the direction of a uniform magnetic field both individual cells (muscle, stromal, and stem cells) or spheroids in a thermoresponsive collagen hydrogel. In an all‐in‐one approach, spheroids are generated at high throughput by magnetic engineering using microfabricated micromagnets and are used as building blocks to create 3D anisotropic tissue structures of different scales. The magnetic cells and spheroids alignment process is optimized in terms of magnetic cell labeling, concentration, and size. Anisotropic structures are induced to form fibers in the direction of the magnetic alignment, with the respective roles of the magnetic field, the mechanical stretching of hydrogel or co‐culture of the aligned cells with non‐magnetic stromal cells, being investigated. Over days, spheroids fuse into 3D tubular structures, oriented in the direction of the magnetic alignment. Moreover, in the case of the muscle cells model, multinucleated cells can be observed within the fibers.
Collapse
Affiliation(s)
- Noam Demri
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Simon Dumas
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Manh‐Louis Nguyen
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Giacomo Gropplero
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Ali Abou‐Hassan
- Institut Universitaire de France (IUF) 75231 Paris Cedex 05 France
- PHysico‐chimie des Electrolytes et Nanosystèmes InterfaciauX PHENIX CNRS UMR234 Sorbonne University 75005 Paris France
| | - Stéphanie Descroix
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| | - Claire Wilhelm
- Laboratoire Physico Chimie Curie PCC CNRS UMR168 Institut Curie Sorbonne University PSL University 75005 Paris France
| |
Collapse
|
24
|
Camman M, Joanne P, Brun J, Marcellan A, Dumont J, Agbulut O, Hélary C. Anisotropic dense collagen hydrogels with two ranges of porosity to mimic the skeletal muscle extracellular matrix. BIOMATERIALS ADVANCES 2022; 144:213219. [PMID: 36481519 DOI: 10.1016/j.bioadv.2022.213219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Despite the crucial role of the extracellular matrix (ECM) in the organotypic organization and function of skeletal muscles, most 3D models do not mimic its specific characteristics, namely its biochemical composition, stiffness, anisotropy, and porosity. Here, a novel 3D in vitro model of muscle ECM was developed reproducing these four crucial characteristics of the native ECM. An anisotropic hydrogel mimicking the muscle fascia was obtained thanks to unidirectional 3D printing of dense collagen with aligned collagen fibrils. The space between the different layers was tuned to generate an intrinsic network of pores (100 μm) suitable for nutrient and oxygen diffusion. By modulating the gelling conditions, the mechanical properties of the construct reached those measured in the physiological muscle ECM. This artificial matrix was thus evaluated for myoblast differentiation. The addition of large channels (600 μm) by molding permitted to create a second range of porosity suitable for cell colonization without altering the physical properties of the hydrogel. Skeletal myoblasts embedded in Matrigel®, seeded within the channels, organized in 3D, and differentiated into multinucleated myotubes. These results show that porous and anisotropic dense collagen hydrogels are promising biomaterials to model skeletal muscle ECM.
Collapse
Affiliation(s)
- Marie Camman
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, CNRS, UMR 7574, F-75005, Paris, France; Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, CNRS, UMR 8256, Inserm U1164, Biological Adaptation and Ageing, F-75005, Paris, France
| | - Pierre Joanne
- Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, CNRS, UMR 8256, Inserm U1164, Biological Adaptation and Ageing, F-75005, Paris, France
| | - Julie Brun
- Sciences et Ingénierie de la Matière Molle, ESPCI Paris, Université PSL, CNRS, Sorbonne Université, F-75005, Paris, France
| | - Alba Marcellan
- Sciences et Ingénierie de la Matière Molle, ESPCI Paris, Université PSL, CNRS, Sorbonne Université, F-75005, Paris, France
| | - Julien Dumont
- CIRB Microscopy facility, Collège de France, CNRS, UMR 7241, Inserm U1050, F-75005, Paris, France
| | - Onnik Agbulut
- Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, CNRS, UMR 8256, Inserm U1164, Biological Adaptation and Ageing, F-75005, Paris, France.
| | - Christophe Hélary
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, CNRS, UMR 7574, F-75005, Paris, France.
| |
Collapse
|
25
|
Mikšiūnas R, Labeit S, Bironaitė D. The Effect of Heat Shock on Myogenic Differentiation of Human Skeletal-Muscle-Derived Mesenchymal Stem/Stromal Cells. Cells 2022; 11:3209. [PMID: 36291076 PMCID: PMC9600296 DOI: 10.3390/cells11203209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 12/18/2023] Open
Abstract
Muscle injuries, degenerative diseases and other lesions negatively affect functioning of human skeletomuscular system and thus quality of life. Therefore, the investigation of molecular mechanisms, stimulating myogenic differentiation of primary skeletal-muscle-derived mesenchymal stem/stromal cells (SM-MSCs), is actual and needed. The aim of the present study was to investigate the myogenic differentiation of CD56 (neural cell adhesion molecule, NCAM)-positive and -negative SM-MSCs and their response to the non-cytotoxic heat stimulus. The SM-MSCs were isolated from the post operation muscle tissue, sorted by flow cytometer according to the CD56 biomarker and morphology, surface profile, proliferation and myogenic differentiation has been investigated. Data show that CD56(+) cells were smaller in size, better proliferated and had significantly higher levels of CD146 (MCAM) and CD318 (CDCP1) compared with the CD56(-) cells. At control level, CD56(+) cells significantly more expressed myogenic differentiation markers MYOD1 and myogenin (MYOG) and better differentiated to the myogenic direction. The non-cytotoxic heat stimulus significantly stronger stimulated expression of myogenic markers in CD56(+) than in CD56(-) cells that correlated with the multinucleated cell formation. Data show that regenerative properties of CD56(+) SM-MSCs can be stimulated by an extracellular stimulus and be used as a promising skeletal muscle regenerating tool in vivo.
Collapse
Affiliation(s)
- Rokas Mikšiūnas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08460 Vilnius, Lithuania
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, 68169 Mannheim, Germany
- Myomedix GmbH, 69151 Neckargemünd, Germany
| | - Daiva Bironaitė
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08460 Vilnius, Lithuania
| |
Collapse
|
26
|
Kim H, Osaki T, Kamm RD, Asada HH. Tri-culture of spatially organizing human skeletal muscle cells, endothelial cells, and fibroblasts enhances contractile force and vascular perfusion of skeletal muscle tissues. FASEB J 2022; 36:e22453. [PMID: 35838893 DOI: 10.1096/fj.202200500r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 11/11/2022]
Abstract
Constructing engineered human skeletal muscle tissues that resemble the function and microstructure of human skeletal muscles is key to utilizing them in a variety of applications such as drug development, disease modeling, regenerative medicine, and engineering biological machines. However, current in vitro skeletal muscle tissues are far inferior to native muscles in terms of contractile function and lack essential cues for muscle functions, particularly heterotypic cell-cell interactions between myoblasts, endothelial cells, and fibroblasts. Here, we develop an engineered muscle tissue with a coaxial three-layered tubular structure composed of an inner endothelial cell layer, an endomysium-like layer with fibroblasts in the middle, and an outer skeletal muscle cell layer, similar to the architecture of native skeletal muscles. Engineered skeletal muscle tissues with three spatially organized cell types produced thicker myotubes and lowered Young's modulus through extracellular matrix remodeling, resulting in 43% stronger contractile force. Furthermore, we demonstrated that fibroblasts localized in the endomysium layer induced angiogenic sprouting of endothelial cells into the muscle layer more effectively than fibroblasts homogeneously distributed in the muscle layer. This layered tri-culture system enables a structured spatial configuration of the three main cell types of skeletal muscle and promotes desired paracrine signaling, resulting in improved angiogenesis and increased contractile force. This research offers new insights to efficiently obtain new human skeletal muscle models, transplantable tissues, and actuators for biological machines.
Collapse
Affiliation(s)
- Hyeonyu Kim
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Stanford Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, California, USA
| | - Tatsuya Osaki
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Roger D Kamm
- Departments of Biological and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - H Harry Asada
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
27
|
Nazri NSM, Vanoh D, Soo KL. Natural Food for Sarcopenia: A Narrative Review. Malays J Med Sci 2022; 29:28-42. [PMID: 36101538 PMCID: PMC9438863 DOI: 10.21315/mjms2022.29.4.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 01/13/2022] [Indexed: 10/26/2022] Open
Abstract
Sarcopenia is a syndrome characterised by progressive loss of skeletal muscle mass and strength. Proper nutrition is essential for management of sarcopenia. Thus, this article aims to review the association between dietary pattern or food groups consisting of natural food and sarcopenia. A literature search was performed using four databases namely PubMed, Scopus, Sage and ScienceDirect. The search terms used were 'fruits', 'vegetables', 'egg', 'fish', 'chicken', 'protein food', 'ulam', 'fresh herbs', 'sarcopenia', 'elderly and 'older adults'. A total of 18 studies were included in the final review. Adherence to Mediterranean and Japanese dietary pattern were associated with lower prevalence of sarcopenia whereas Western dietary pattern was significantly associated with higher risk of sarcopenia. For food groups, there is a significant association between dietary protein intake and sarcopenia. There are also significant associations between the intake of vegetables, fruits or both vegetables and fruits, and lower risk of sarcopenia. Consumption of natural food comprising of high-quality protein, fruits and vegetables have been associated with protection against muscle wasting and sarcopenia. Therefore, it is possible that a well-planned diet may works just as effectively as or possibly better than individual nutrient supplements for the prevention and treatment for sarcopenia among older adults.
Collapse
Affiliation(s)
- Nurul Syahidah Mohd Nazri
- Nutrition and Dietetics Programme, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Divya Vanoh
- Nutrition and Dietetics Programme, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Kah Leng Soo
- Nutrition and Dietetics Programme, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
28
|
Carraro E, Rossi L, Maghin E, Canton M, Piccoli M. 3D in vitro Models of Pathological Skeletal Muscle: Which Cells and Scaffolds to Elect? Front Bioeng Biotechnol 2022; 10:941623. [PMID: 35898644 PMCID: PMC9313593 DOI: 10.3389/fbioe.2022.941623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/21/2022] [Indexed: 12/29/2022] Open
Abstract
Skeletal muscle is a fundamental tissue of the human body with great plasticity and adaptation to diseases and injuries. Recreating this tissue in vitro helps not only to deepen its functionality, but also to simulate pathophysiological processes. In this review we discuss the generation of human skeletal muscle three-dimensional (3D) models obtained through tissue engineering approaches. First, we present an overview of the most severe myopathies and the two key players involved: the variety of cells composing skeletal muscle tissue and the different components of its extracellular matrix. Then, we discuss the peculiar characteristics among diverse in vitro models with a specific focus on cell sources, scaffold composition and formulations, and fabrication techniques. To conclude, we highlight the efficacy of 3D models in mimicking patient-specific myopathies, deepening muscle disease mechanisms or investigating possible therapeutic effects.
Collapse
Affiliation(s)
- Eugenia Carraro
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lucia Rossi
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Edoardo Maghin
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Marcella Canton
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Martina Piccoli
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- *Correspondence: Martina Piccoli,
| |
Collapse
|
29
|
Customized bioreactor enables the production of 3D diaphragmatic constructs influencing matrix remodeling and fibroblast overgrowth. NPJ Regen Med 2022; 7:25. [PMID: 35468920 PMCID: PMC9038738 DOI: 10.1038/s41536-022-00222-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
The production of skeletal muscle constructs useful for replacing large defects in vivo, such as in congenital diaphragmatic hernia (CDH), is still considered a challenge. The standard application of prosthetic material presents major limitations, such as hernia recurrences in a remarkable number of CDH patients. With this work, we developed a tissue engineering approach based on decellularized diaphragmatic muscle and human cells for the in vitro generation of diaphragmatic-like tissues as a proof-of-concept of a new option for the surgical treatment of large diaphragm defects. A customized bioreactor for diaphragmatic muscle was designed to control mechanical stimulation and promote radial stretching during the construct engineering. In vitro tests demonstrated that both ECM remodeling and fibroblast overgrowth were positively influenced by the bioreactor culture. Mechanically stimulated constructs also increased tissue maturation, with the formation of new oriented and aligned muscle fibers. Moreover, after in vivo orthotopic implantation in a surgical CDH mouse model, mechanically stimulated muscles maintained the presence of human cells within myofibers and hernia recurrence did not occur, suggesting the value of this approach for treating diaphragm defects.
Collapse
|
30
|
Iberite F, Gruppioni E, Ricotti L. Skeletal muscle differentiation of human iPSCs meets bioengineering strategies: perspectives and challenges. NPJ Regen Med 2022; 7:23. [PMID: 35393412 PMCID: PMC8991236 DOI: 10.1038/s41536-022-00216-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 03/01/2022] [Indexed: 12/31/2022] Open
Abstract
Although skeletal muscle repairs itself following small injuries, genetic diseases or severe damages may hamper its ability to do so. Induced pluripotent stem cells (iPSCs) can generate myogenic progenitors, but their use in combination with bioengineering strategies to modulate their phenotype has not been sufficiently investigated. This review highlights the potential of this combination aimed at pushing the boundaries of skeletal muscle tissue engineering. First, the overall organization and the key steps in the myogenic process occurring in vivo are described. Second, transgenic and non-transgenic approaches for the myogenic induction of human iPSCs are compared. Third, technologies to provide cells with biophysical stimuli, biomaterial cues, and biofabrication strategies are discussed in terms of recreating a biomimetic environment and thus helping to engineer a myogenic phenotype. The embryonic development process and the pro-myogenic role of the muscle-resident cell populations in co-cultures are also described, highlighting the possible clinical applications of iPSCs in the skeletal muscle tissue engineering field.
Collapse
Affiliation(s)
- Federica Iberite
- The BioRobotics Institute, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy. .,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy.
| | - Emanuele Gruppioni
- Centro Protesi INAIL, Istituto Nazionale per l'Assicurazione contro gli Infortuni sul Lavoro, 40054, Vigorso di Budrio (BO), Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy.,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy
| |
Collapse
|
31
|
Poteracki JM, Moschouris K, Yoseph BP, Zhou Y, Soker S, Criswell TL. Development of a Rat Model of Fasciotomy Treatment for Compartment Syndrome. Tissue Eng Part C Methods 2022; 28:51-60. [PMID: 35107365 PMCID: PMC9022182 DOI: 10.1089/ten.tec.2021.0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Skeletal muscle injuries are a major cause of disability for military and civilian populations. Compartment syndrome (CS) in skeletal muscle results from an edema-induced increase in intracompartmental pressure (ICP) after primary injury. Untreated ICP will occlude the tissue vasculature, tissue necrosis, and potential loss of limb. The current standard of care for CS is surgical fasciotomy, an incision through the muscle fascia to relieve ICP. Early fasciotomy will preserve the limb, but often leaves patients with long-term scarring and reduced muscle function. Our group previously developed and characterized a rat model of CS to explore the pathophysiology of CS and test new therapies. We present an expansion of this CS model, including the fasciotomy, to better simulate clinical treatment. CS was induced on the hind limb of adult male Lewis rats and fasciotomy was performed 24 h later. Less than 20% of the rats that underwent fasciotomy showed detectable force 4 days after injury, compared with the 75% of rats that underwent CS induction without fasciotomy. Muscles undergoing fasciotomy showed a significant increase in fibrosis and an increased number of macrophages, Pax7+ satellite cells, and α-smooth muscle actin+ myofibroblasts at 7 days postinjury. These data indicate that the use of fasciotomy in a rat model of CS resulted in injury sequelae that reflect the severity of human clinical disease presentation along with current standard of care. Impact Statement Current animal models of skeletal muscle injury struggle to accurately reflect the injury sequelae seen in humans, particularly in rats and mice. These animals also recover faster than humans do. More accurate recapitulation of the injury is needed to better study the injury progression, as well as screen for novel therapies. This research combines an existing model of compartment syndrome with its clinical standard of care (fasciotomy), creating a more accurate rat model of injury, and providing for a better treatment screening tool. These results show how our model leads to a sustained skeletal muscle deficit with increased inflammation.
Collapse
Affiliation(s)
- James M. Poteracki
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA.,Address correspondence to: James M. Poteracki, MS, Wake Forest Institute of Regenerative Medicine, Wake Forest University, 391 Technology Way NE, Winston-Salem, NC 27101, USA
| | - Kathryn Moschouris
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Benyam P. Yoseph
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Yu Zhou
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Shay Soker
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Tracy L. Criswell
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| |
Collapse
|
32
|
Park J, Choi JK, Choi DH, Lee KE, Park YS. Optimization of skeletal muscle-derived fibroblast isolation and purification without the preplating method. Cell Tissue Bank 2022; 23:557-568. [DOI: 10.1007/s10561-021-09989-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 12/22/2021] [Indexed: 11/02/2022]
|
33
|
Role of TRPV4 in skeletal function and its mutant-mediated skeletal disorders. CURRENT TOPICS IN MEMBRANES 2022; 89:221-246. [DOI: 10.1016/bs.ctm.2022.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
34
|
Diagnosis and Simultaneous Treatment of Musculoskeletal Injury Using H 2O 2-Triggered Echogenic Antioxidant Polymer Nanoparticles in a Rat Model of Contusion Injury. NANOMATERIALS 2021; 11:nano11102571. [PMID: 34685012 PMCID: PMC8537538 DOI: 10.3390/nano11102571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 01/06/2023]
Abstract
Ultrasound is clinically used for diagnosis and interventions for musculoskeletal injuries like muscle contusion, but contrast of ultrasonography still remains a challenge in the field of the musculoskeletal system. A level of hydrogen peroxide (H2O2) is known to be elevated during mechanical tissue damage and therefore H2O2 can be exploited as a diagnostic and therapeutic marker for mechanical injuries in the musculoskeletal system. We previously developed poly(vanillin-oxalate) (PVO) as an inflammation-responsive polymeric prodrug of vanillin, which is designed to rapidly respond to H2O2 and exert antioxidant and anti-inflammatory activities. The primary aim of this study is to verify whether PVO nanoparticles could serve as contrast agents as well as therapeutic agents for musculoskeletal injuries simultaneously. In a rat model of contusion-induced muscle injury, PVO nanoparticles generated CO2 bubbles to enhance the ultrasound contrast in the injury site. A single intramuscular injection of PVO nanoparticles also suppressed contusion-induced muscle damages by inhibiting the expression of pro-inflammatory cytokines and inflammatory cell infiltration. We, therefore, anticipate that PVO nanoparticles have great translational potential as not only ultrasound imaging agents but also therapeutic agents for the musculoskeletal disorders such as contusion.
Collapse
|
35
|
Tonti OR, Larson H, Lipp SN, Luetkemeyer CM, Makam M, Vargas D, Wilcox SM, Calve S. Tissue-specific parameters for the design of ECM-mimetic biomaterials. Acta Biomater 2021; 132:83-102. [PMID: 33878474 PMCID: PMC8434955 DOI: 10.1016/j.actbio.2021.04.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is a complex network of biomolecules that mechanically and biochemically directs cell behavior and is crucial for maintaining tissue function and health. The heterogeneous organization and composition of the ECM varies within and between tissue types, directing mechanics, aiding in cell-cell communication, and facilitating tissue assembly and reassembly during development, injury and disease. As technologies like 3D printing rapidly advance, researchers are better able to recapitulate in vivo tissue properties in vitro; however, tissue-specific variations in ECM composition and organization are not given enough consideration. This is in part due to a lack of information regarding how the ECM of many tissues varies in both homeostatic and diseased states. To address this gap, we describe the components and organization of the ECM, and provide examples for different tissues at various states of disease. While many aspects of ECM biology remain unknown, our goal is to highlight the complexity of various tissues and inspire engineers to incorporate unique components of the native ECM into in vitro platform design and fabrication. Ultimately, we anticipate that the use of biomaterials that incorporate key tissue-specific ECM will lead to in vitro models that better emulate human pathologies. STATEMENT OF SIGNIFICANCE: Biomaterial development primarily emphasizes the engineering of new materials and therapies at the expense of identifying key parameters of the tissue that is being emulated. This can be partially attributed to the difficulty in defining the 3D composition, organization, and mechanics of the ECM within different tissues and how these material properties vary as a function of homeostasis and disease. In this review, we highlight a range of tissues throughout the body and describe how ECM content, cell diversity, and mechanical properties change in diseased tissues and influence cellular behavior. Accurately mimicking the tissue of interest in vitro by using ECM specific to the appropriate state of homeostasis or pathology in vivo will yield results more translatable to humans.
Collapse
Affiliation(s)
- Olivia R Tonti
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Hannah Larson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah N Lipp
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Callan M Luetkemeyer
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Megan Makam
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Diego Vargas
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sean M Wilcox
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States.
| |
Collapse
|
36
|
P2Y2 promotes fibroblasts activation and skeletal muscle fibrosis through AKT, ERK, and PKC. BMC Musculoskelet Disord 2021; 22:680. [PMID: 34380439 PMCID: PMC8359595 DOI: 10.1186/s12891-021-04569-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Skeletal muscle atrophy and fibrosis are pathological conditions that contribute to morbidity in numerous conditions including aging, cachexia, and denervation. Muscle atrophy is characterized as reduction of muscle fiber size and loss of muscle mass while muscle fibrosis is due to fibroblasts activation and excessive production of extracellular matrix. Purinergic receptor P2Y2 has been implicated in fibrosis. This study aims to elucidate the roles of P2Y2 in sleketal muscle atrophy and fibrosis. METHODS Primary muscle fibroblasts were isolated from wild type and P2Y2 knockout (KO) mice and their proliferating and migrating abilities were assessed by CCK-8 and Transwell migration assays respectively. Fibroblasts were activated with TGF-β1 and assessed by western blot of myofibroblast markers including α-SMA, CTGF, and collagen I. Muscle atrophy and fibrosis were induced by transection of distal sciatic nerve and assessed using Masson staining. RESULTS P2Y2 KO fibroblasts proliferated and migrated significantly slower than WT fibroblasts with or without TGF-β1.The proliferation and ECM production were enhanced by P2Y2 agonist PSB-1114 and inhibited by antagonist AR-C118925. TGF-β1 induced fibrotic activation was abolished by P2Y2 ablation and inhibited by AKT, ERK, and PKC inhibitors. Ablation of P2Y2 reduced denervation induced muscle atrophy and fibrosis. CONCLUSIONS P2Y2 is a promoter of skeletal muscle atrophy and activation of fibroblasts after muscle injury, which signaling through AKT, ERK and PKC. P2Y2 could be a potential intervention target after muscle injury.
Collapse
|
37
|
Plikus MV, Wang X, Sinha S, Forte E, Thompson SM, Herzog EL, Driskell RR, Rosenthal N, Biernaskie J, Horsley V. Fibroblasts: Origins, definitions, and functions in health and disease. Cell 2021; 184:3852-3872. [PMID: 34297930 PMCID: PMC8566693 DOI: 10.1016/j.cell.2021.06.024] [Citation(s) in RCA: 533] [Impact Index Per Article: 133.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/28/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts are diverse mesenchymal cells that participate in tissue homeostasis and disease by producing complex extracellular matrix and creating signaling niches through biophysical and biochemical cues. Transcriptionally and functionally heterogeneous across and within organs, fibroblasts encode regional positional information and maintain distinct cellular progeny. We summarize their development, lineages, functions, and contributions to fibrosis in four fibroblast-rich organs: skin, lung, skeletal muscle, and heart. We propose that fibroblasts are uniquely poised for tissue repair by easily reentering the cell cycle and exhibiting a reversible plasticity in phenotype and cell fate. These properties, when activated aberrantly, drive fibrotic disorders in humans.
Collapse
Affiliation(s)
- Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Elvira Forte
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Sean M Thompson
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | - Erica L Herzog
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Ryan R Driskell
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA; Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA.
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK.
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Valerie Horsley
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
38
|
Derk J, Jones HE, Como C, Pawlikowski B, Siegenthaler JA. Living on the Edge of the CNS: Meninges Cell Diversity in Health and Disease. Front Cell Neurosci 2021; 15:703944. [PMID: 34276313 PMCID: PMC8281977 DOI: 10.3389/fncel.2021.703944] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/08/2021] [Indexed: 12/30/2022] Open
Abstract
The meninges are the fibrous covering of the central nervous system (CNS) which contain vastly heterogeneous cell types within its three layers (dura, arachnoid, and pia). The dural compartment of the meninges, closest to the skull, is predominantly composed of fibroblasts, but also includes fenestrated blood vasculature, an elaborate lymphatic system, as well as immune cells which are distinct from the CNS. Segregating the outer and inner meningeal compartments is the epithelial-like arachnoid barrier cells, connected by tight and adherens junctions, which regulate the movement of pathogens, molecules, and cells into and out of the cerebral spinal fluid (CSF) and brain parenchyma. Most proximate to the brain is the collagen and basement membrane-rich pia matter that abuts the glial limitans and has recently be shown to have regional heterogeneity within the developing mouse brain. While the meninges were historically seen as a purely structural support for the CNS and protection from trauma, the emerging view of the meninges is as an essential interface between the CNS and the periphery, critical to brain development, required for brain homeostasis, and involved in a variety of diseases. In this review, we will summarize what is known regarding the development, specification, and maturation of the meninges during homeostatic conditions and discuss the rapidly emerging evidence that specific meningeal cell compartments play differential and important roles in the pathophysiology of a myriad of diseases including: multiple sclerosis, dementia, stroke, viral/bacterial meningitis, traumatic brain injury, and cancer. We will conclude with a list of major questions and mechanisms that remain unknown, the study of which represent new, future directions for the field of meninges biology.
Collapse
Affiliation(s)
- Julia Derk
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Aurora, CO, United States
| | - Hannah E. Jones
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Aurora, CO, United States
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Christina Como
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Aurora, CO, United States
- Neuroscience Graduate Program, University of Colorado, Aurora, CO, United States
| | - Bradley Pawlikowski
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Aurora, CO, United States
| | - Julie A. Siegenthaler
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Aurora, CO, United States
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
- Neuroscience Graduate Program, University of Colorado, Aurora, CO, United States
| |
Collapse
|
39
|
Moffatt P, Boraschi-Diaz I, Bardai G, Rauch F. Muscle transcriptome in mouse models of osteogenesis imperfecta. Bone 2021; 148:115940. [PMID: 33812081 DOI: 10.1016/j.bone.2021.115940] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Osteogenesis imperfecta (OI) is a heritable connective tissue disorder that is most often caused by mutations in collagen type I encoding genes. Even though bone fragility is the most conspicuous finding in OI, the muscle system is also affected. In the present study we explored the muscle phenotype related to collagen type I mutations on the transcriptome level. RNA sequencing was performed in gastrocnemius muscles of homozygous oim mice and of heterozygous Jrt mice, two models of severe OI. We found that oim and Jrt mice shared 27 differentially expressed genes, of which 11 were concordantly upregulated and 15 concordantly downregulated. Gene Set Enrichment Analysis revealed that in both oim and Jrt mice, genes involved in 'metabolism of lipids' were significantly enriched among upregulated genes. In addition, several genes coding for extracellular matrix components were upregulated in both oim and Jrt mice. Among downregulated genes, genes involved in 'muscle contraction' were enriched in both OI mouse models. These 'muscle contraction' genes coded for slow-twitch type I muscle fiber components. Another shared downregulated gene was Mss51, a metabolic stress-inducible factor that is found in mitochondria. These data show that two mouse models of severe OI share abnormalities in the expression of genes that code for extracellular matrix proteins, lipid and energy metabolism and structural proteins of type I muscle fibers. The muscle disturbances resulting from the collagen type I mutations in these mouse models could be viewed as a mild form of muscle dystrophy.
Collapse
Affiliation(s)
- Pierre Moffatt
- Shriners Hospital for Children-Canada, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Iris Boraschi-Diaz
- Shriners Hospital for Children-Canada, Montreal, Quebec, Canada; Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Ghalib Bardai
- Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| | - Frank Rauch
- Shriners Hospital for Children-Canada, Montreal, Quebec, Canada; Department of Pediatrics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
40
|
Contreras O, Rossi FMV, Theret M. Origins, potency, and heterogeneity of skeletal muscle fibro-adipogenic progenitors-time for new definitions. Skelet Muscle 2021; 11:16. [PMID: 34210364 PMCID: PMC8247239 DOI: 10.1186/s13395-021-00265-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Striated muscle is a highly plastic and regenerative organ that regulates body movement, temperature, and metabolism-all the functions needed for an individual's health and well-being. The muscle connective tissue's main components are the extracellular matrix and its resident stromal cells, which continuously reshape it in embryonic development, homeostasis, and regeneration. Fibro-adipogenic progenitors are enigmatic and transformative muscle-resident interstitial cells with mesenchymal stem/stromal cell properties. They act as cellular sentinels and physiological hubs for adult muscle homeostasis and regeneration by shaping the microenvironment by secreting a complex cocktail of extracellular matrix components, diffusible cytokines, ligands, and immune-modulatory factors. Fibro-adipogenic progenitors are the lineage precursors of specialized cells, including activated fibroblasts, adipocytes, and osteogenic cells after injury. Here, we discuss current research gaps, potential druggable developments, and outstanding questions about fibro-adipogenic progenitor origins, potency, and heterogeneity. Finally, we took advantage of recent advances in single-cell technologies combined with lineage tracing to unify the diversity of stromal fibro-adipogenic progenitors. Thus, this compelling review provides new cellular and molecular insights in comprehending the origins, definitions, markers, fate, and plasticity of murine and human fibro-adipogenic progenitors in muscle development, homeostasis, regeneration, and repair.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, 2052, Australia.
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Fabio M V Rossi
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Marine Theret
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
41
|
Seo H, Lee SH, Park Y, Lee HS, Hong JS, Lim CY, Kim DH, Park SS, Suh HJ, Hong KB. (-)-Epicatechin-Enriched Extract from Camellia sinensis Improves Regulation of Muscle Mass and Function: Results from a Randomized Controlled Trial. Antioxidants (Basel) 2021; 10:1026. [PMID: 34202133 PMCID: PMC8300738 DOI: 10.3390/antiox10071026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Loss of skeletal muscle mass and function with age represents an important source of frailty and functional decline in the elderly. Antioxidants from botanical extracts have been shown to enhance the development, mass, and strength of skeletal muscle by influencing age-related cellular and molecular processes. Tannase-treated green tea extract contains high levels of the antioxidants (-)-epicatechin (EC) and gallic acid that may have therapeutic benefits for age-related muscle decline. The aim of this study was to investigate the effect of tannase-treated green tea extract on various muscle-related parameters, without concomitant exercise, in a single-center, randomized, double-blind, placebo-controlled study. Administration of tannase-treated green tea extract (600 mg/day) for 12 weeks significantly increased isokinetic flexor muscle and handgrip strength in the treatment group compared with those in the placebo (control) group. In addition, the control group showed a significant decrease in arm muscle mass after 12 weeks, whereas no significant change was observed in the treatment group. Blood serum levels of follistatin, myostatin, high-sensitivity C-reactive protein (hs-CRP), interleukin (IL)-6, IL-8, insulin-like growth factor-1 (IGF-1), and cortisol were analyzed, and the decrease in myostatin resulting from the administration of tannase-treated green tea extract was found to be related to the change in muscle mass and strength. In summary, oral administration of tannase-treated green tea extract containing antioxidants without concomitant exercise can improve muscle mass and strength and may have therapeutic benefits in age-related muscle function decline.
Collapse
Affiliation(s)
- Hyeyeong Seo
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Korea;
| | - Seok-Hee Lee
- Department of Food Science and Biotechnology, Dongguk University, Goyang 10326, Korea; (S.-H.L.); (Y.P.)
| | - Yooheon Park
- Department of Food Science and Biotechnology, Dongguk University, Goyang 10326, Korea; (S.-H.L.); (Y.P.)
| | - Hee-Seok Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong 17546, Korea;
| | - Jeong Sup Hong
- Animal Center and Preclinical Evaluation Research Institute, Yonam College, Cheonan 31005, Korea;
| | - Cho Young Lim
- R&D Center, BTC Corporation, Ansan 15588, Korea; (C.Y.L.); (D.H.K.)
| | - Dong Hyeon Kim
- R&D Center, BTC Corporation, Ansan 15588, Korea; (C.Y.L.); (D.H.K.)
| | - Sung-Soo Park
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea;
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Korea;
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul 02841, Korea
| | - Ki-Bae Hong
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea;
| |
Collapse
|
42
|
Prevalence of Hypovitaminosis C and its Relationship with Frailty in Older Hospitalised Patients: A Cross-Sectional Study. Nutrients 2021; 13:nu13062117. [PMID: 34203044 PMCID: PMC8235098 DOI: 10.3390/nu13062117] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/05/2022] Open
Abstract
Frailty is common in older hospitalised patients and may be associated with micronutrient malnutrition. Only limited studies have explored the relationship between frailty and vitamin C deficiency. This study investigated the prevalence of vitamin C deficiency and its association with frailty severity in patients ≥75 years admitted under a geriatric unit. Patients (n = 160) with a mean age of 84.4 ± 6.4 years were recruited and underwent frailty assessment by use of the Edmonton Frail Scale (EFS). Patients with an EFS score <10 were classified as non-frail/vulnerable/mildly frail and those with ≥10 as moderate–severely frail. Patients with vitamin C levels between 11–28 μmol/L were classified as vitamin C depleted while those with levels <11 μmol/L were classified as vitamin C deficient. A multivariate logistic regression model determined the relationship between vitamin C deficiency and frailty severity after adjustment for various co-variates. Fifty-seven (35.6%) patients were vitamin C depleted, while 42 (26.3%) had vitamin C deficiency. Vitamin C levels were significantly lower among patients who were moderate–severely frail when compared to those who were non-frail/vulnerable/mildly frail (p < 0.05). After adjusted analysis, vitamin C deficiency was 4.3-fold more likely to be associated with moderate–severe frailty (aOR 4.30, 95% CI 1.33-13.86, p = 0.015). Vitamin C deficiency is common and is associated with a greater severity of frailty in older hospitalised patients.
Collapse
|
43
|
Costa ML, Jurberg AD, Mermelstein C. The Role of Embryonic Chick Muscle Cell Culture in the Study of Skeletal Myogenesis. Front Physiol 2021; 12:668600. [PMID: 34093232 PMCID: PMC8173222 DOI: 10.3389/fphys.2021.668600] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/21/2021] [Indexed: 11/13/2022] Open
Abstract
The mechanisms involved in the development of skeletal muscle fibers have been studied in the last 70 years and yet many aspects of this process are still not completely understood. A myriad of in vivo and in vitro invertebrate and vertebrate animal models has been used for dissecting the molecular and cellular events involved in muscle formation. Among the most used animal models for the study of myogenesis are the rodents rat and mouse, the fruit fly Drosophila, and the birds chicken and quail. Here, we describe the robustness and advantages of the chick primary muscle culture model for the study of skeletal myogenesis. In the myoblast culture obtained from embryonic chick pectoralis muscle it is possible to analyze all the steps involved in skeletal myogenesis, such as myoblast proliferation, withdrawal from cell cycle, cell elongation and migration, myoblast alignment and fusion, the assembly of striated myofibrils, and the formation of multinucleated myotubes. The fact that in vitro chick myotubes can harbor hundreds of nuclei, whereas myotubes from cell lines have only a dozen nuclei demonstrates the high level of differentiation of the autonomous chick myogenic program. This striking differentiation is independent of serum withdrawal, which points to the power of the model. We also review the major pro-myogenic and anti-myogenic molecules and signaling pathways involved in chick myogenesis, in addition to providing a detailed protocol for the preparation of embryonic chick myogenic cultures. Moreover, we performed a bibliometric analysis of the articles that used this model to evaluate which were the main explored topics of interest and their contributors. We expect that by describing the major findings, and their advantages, of the studies using the embryonic chick myogenic model we will foster new studies on the molecular and cellular process involved in muscle proliferation and differentiation that are more similar to the actual in vivo condition than the muscle cell lines.
Collapse
Affiliation(s)
- Manoel L Costa
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arnon D Jurberg
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Faculdade de Medicina-Presidente Vargas, Universidade Estácio de Sá, Rio de Janeiro, Brazil
| | - Claudia Mermelstein
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Different Lipid Signature in Fibroblasts of Long-Chain Fatty Acid Oxidation Disorders. Cells 2021; 10:cells10051239. [PMID: 34069977 PMCID: PMC8157847 DOI: 10.3390/cells10051239] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Long-chain fatty acid oxidation disorders (lc-FAOD) are a group of diseases affecting the degradation of long-chain fatty acids. In order to investigate the disease specific alterations of the cellular lipidome, we performed undirected lipidomics in fibroblasts from patients with carnitine palmitoyltransferase II, very long-chain acyl-CoA dehydrogenase, and long-chain 3-hydroxyacyl-CoA dehydrogenase. We demonstrate a deep remodeling of mitochondrial cardiolipins. The aberrant phosphatidylcholine/phosphatidylethanolamine ratio and the increased content of plasmalogens and of lysophospholipids support the theory of an inflammatory phenotype in lc-FAOD. Moreover, we describe increased ratios of sphingomyelin/ceramide and sphingomyelin/hexosylceramide in LCHAD deficiency which may contribute to the neuropathic phenotype of LCHADD/mitochondrial trifunctional protein deficiency.
Collapse
|
45
|
Westman AM, Peirce SM, Christ GJ, Blemker SS. Agent-based model provides insight into the mechanisms behind failed regeneration following volumetric muscle loss injury. PLoS Comput Biol 2021; 17:e1008937. [PMID: 33970905 PMCID: PMC8110270 DOI: 10.1371/journal.pcbi.1008937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/01/2021] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle possesses a remarkable capacity for repair and regeneration following a variety of injuries. When successful, this highly orchestrated regenerative process requires the contribution of several muscle resident cell populations including satellite stem cells (SSCs), fibroblasts, macrophages and vascular cells. However, volumetric muscle loss injuries (VML) involve simultaneous destruction of multiple tissue components (e.g., as a result of battlefield injuries or vehicular accidents) and are so extensive that they exceed the intrinsic capability for scarless wound healing and result in permanent cosmetic and functional deficits. In this scenario, the regenerative process fails and is dominated by an unproductive inflammatory response and accompanying fibrosis. The failure of current regenerative therapeutics to completely restore functional muscle tissue is not surprising considering the incomplete understanding of the cellular mechanisms that drive the regeneration response in the setting of VML injury. To begin to address this profound knowledge gap, we developed an agent-based model to predict the tissue remodeling response following surgical creation of a VML injury. Once the model was able to recapitulate key aspects of the tissue remodeling response in the absence of repair, we validated the model by simulating the tissue remodeling response to VML injury following implantation of either a decellularized extracellular matrix scaffold or a minced muscle graft. The model suggested that the SSC microenvironment and absence of pro-differentiation SSC signals were the most important aspects of failed muscle regeneration in VML injuries. The major implication of this work is that agent-based models may provide a much-needed predictive tool to optimize the design of new therapies, and thereby, accelerate the clinical translation of regenerative therapeutics for VML injuries.
Collapse
Affiliation(s)
- Amanda M. Westman
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shayn M. Peirce
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Ophthalmology, University of Virginia, Charlottesville, Virginia, United States of America
| | - George J. Christ
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (GJC); (SSB)
| | - Silvia S. Blemker
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Ophthalmology, University of Virginia, Charlottesville, Virginia, United States of America
- Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (GJC); (SSB)
| |
Collapse
|
46
|
Smith LR, Pichika R, Meza RC, Gillies AR, Baliki MN, Chambers HG, Lieber RL. Contribution of extracellular matrix components to the stiffness of skeletal muscle contractures in patients with cerebral palsy. Connect Tissue Res 2021; 62:287-298. [PMID: 31779492 PMCID: PMC7253322 DOI: 10.1080/03008207.2019.1694011] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: Joint contractures in children with cerebral palsy contain muscle tissue that is mechanically stiffer with higher collagen content than typically developing children. Interestingly, the correlation between collagen content and stiffness is weak. To date, no data are available on collagen types or other extracellular matrix proteins in these muscles, nor any information regarding their function. Thus, our purpose was to measure specific extracellular protein composition in cerebral palsy and typically developing human muscles along with structural aspects of extracellular matrix architecture to determine the extent to which these explain mechanical properties. Materials and Methods: Biopsies were collected from children with cerebral palsy undergoing muscle lengthening procedures and typically developing children undergoing anterior cruciate ligament reconstruction. Tissue was prepared for the determination of collagen types I, III, IV, and VI, proteoglycan, biglycan, decorin, hyaluronic acid/uronic acid and collagen crosslinking. Results: All collagen types increased in cerebral palsy along with pyridinoline crosslinks, total proteoglycan, and uronic acid. In all cases, type I or total collagen and total proteoglycan were positive predictors, while biglycan was a negative predictor of stiffness. Together these parameters accounted for a greater degree of variance within groups than across groups, demonstrating an altered relationship between extracellular matrix and stiffness with cerebral palsy. Further, stereological analysis revealed a significant increase in collagen fibrils organized in cables and an increased volume fraction of fibroblasts in CP muscle. Conclusions: These data demonstrate a novel adaptation of muscle extracellular matrix in children with cerebral palsy that includes alterations in extracellular matrix protein composition and structure related to mechanical function.
Collapse
Affiliation(s)
- Lucas R. Smith
- Departments of Neurobiology, Physiology, and Behavior and Physical Medicine and Rehabilitation, University of California, Davis, CA, 95616, USA
| | - Rajeswari Pichika
- Shirley Ryan AbilityLab and Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA
| | - Rachel C. Meza
- Department of Orthopaedic Surgery, University of California San Diego,La Jolla, CA, 92093-0863, USA,Department of Biology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Allison R. Gillies
- Department of Orthopaedic Surgery, University of California San Diego,La Jolla, CA, 92093-0863, USA
| | - Marwan N. Baliki
- Shirley Ryan AbilityLab and Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA
| | - Henry G. Chambers
- Department of Orthopaedics, Rady Children’s Hospital, San Diego, CA, USA
| | - Richard L. Lieber
- Shirley Ryan AbilityLab and Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA,Department of Orthopaedic Surgery, University of California San Diego,La Jolla, CA, 92093-0863, USA,Hines V.A. Medical Center, Maywood, IL, USA
| |
Collapse
|
47
|
Ollewagen T, Myburgh KH, van de Vyver M, Smith C. Rheumatoid cachexia: the underappreciated role of myoblast, macrophage and fibroblast interplay in the skeletal muscle niche. J Biomed Sci 2021; 28:15. [PMID: 33658022 PMCID: PMC7931607 DOI: 10.1186/s12929-021-00714-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/11/2021] [Indexed: 12/24/2022] Open
Abstract
Although rheumatoid arthritis affects 1% of the global population, the role of rheumatoid cachexia, which occurs in up to a third of patients, is relatively neglected as research focus, despite its significant contribution to decreased quality of life in patients. A better understanding of the cellular and molecular processes involved in rheumatoid cachexia, as well as its potential treatment, is dependent on elucidation of the intricate interactions of the cells involved, such as myoblasts, fibroblasts and macrophages. Persistent RA-associated inflammation results in a relative depletion of the capacity for regeneration and repair in the satellite cell niche. The repair that does proceed is suboptimal due to dysregulated communication from the other cellular role players in this multi-cellular environment. This includes the incomplete switch in macrophage phenotype resulting in a lingering pro-inflammatory state within the tissues, as well as fibroblast-associated dysregulation of the dynamic control of the extracellular matrix. Additional to this endogenous dysregulation, some treatment strategies for RA may exacerbate muscle wasting and no multi-cell investigation has been done in this context. This review summarizes the most recent literature characterising clinical RA cachexia and links these features to the roles of and complex communication between multiple cellular contributors in the muscle niche, highlighting the importance of a targeted approach to therapeutic intervention.
Collapse
Affiliation(s)
- T Ollewagen
- Department of Physiological Sciences, Science Faculty, Stellenbosch University, Stellenbosch, South Africa
| | - K H Myburgh
- Department of Physiological Sciences, Science Faculty, Stellenbosch University, Stellenbosch, South Africa
| | - M van de Vyver
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa
| | - C Smith
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa.
| |
Collapse
|
48
|
Crescioli C. Vitamin D Restores Skeletal Muscle Cell Remodeling and Myogenic Program: Potential Impact on Human Health. Int J Mol Sci 2021; 22:1760. [PMID: 33578813 PMCID: PMC7916580 DOI: 10.3390/ijms22041760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle cells, albeit classified as vitamin D receptor (VDR)-poor cells, are finely controlled by vitamin D through genomic and non-genomic mechanisms. Skeletal muscle constantly undergoes cell remodeling, a complex system under multilevel regulation, mainly orchestrated by the satellite niche in response to a variety of stimuli. Cell remodeling is not limited to satisfy reparative and hypertrophic needs, but, through myocyte transcriptome/proteome renewal, it warrants the adaptations necessary to maintain tissue integrity. While vitamin D insufficiency promotes cell maladaptation, restoring vitamin D levels can correct/enhance the myogenic program. Hence, vitamin D fortified foods or supplementation potentially represents the desired approach to limit or avoid muscle wasting and ameliorate health. Nevertheless, consensus on protocols for vitamin D measurement and supplementation is still lacking, due to the high variability of lab tests and of the levels required in different contexts (i.e., age, sex, heath status, lifestyle). This review aims to describe how vitamin D can orchestrate skeletal muscle cell remodeling and myogenic programming, after reviewing the main processes and cell populations involved in this important process, whose correct progress highly impacts on human health. Topics on vitamin D optimal levels, supplementation and blood determination, which are still under debate, will be addressed.
Collapse
Affiliation(s)
- Clara Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome "Foro Italico", Piazza L. de Bosis 6, 00135 Rome, Italy
| |
Collapse
|
49
|
Venter C, Myburgh KH, Niesler CU. Co-culture of pro-inflammatory macrophages and myofibroblasts: Evaluating morphological phenotypes and screening the effects of signaling pathway inhibitors. Physiol Rep 2021; 9:e14704. [PMID: 33463904 PMCID: PMC7814483 DOI: 10.14814/phy2.14704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle regeneration is a complex process influenced by non-myogenic macrophages and fibroblasts, which acquire different phenotypes in response to changes in the injury milieu or changes in experimental conditions. In vitro, serum stimulates the differentiation of fibroblasts into myofibroblasts, while lipopolysaccharide (LPS) stimulates the polarization of unstimulated (M0) macrophages to acquire an M1 pro-inflammatory phenotype. We characterized these phenotypes using morphology (with circularity as shape descriptor; perfect circularity = 1.0) and phenotype-specific markers. Myofibroblasts (high α-smooth muscle actin [SMA] expression) had high circularity (mean 0.60 ± 0.03). Their de-differentiation to fibroblasts (low α-SMA expression) significantly lessened circularity (0.47 ± 0.01 and 0.35 ± 0.02 in 2% or 0% serum culture media respectively (p < 0.05). Unstimulated (M0) macrophages (no CD86 expression) had high circularity (0.72 ± 0.02) which decreased when stimulated to M1 macrophages (CD86 expression) (LPS; 0.61 ± 0.02; p < 0.05). Utilizing these established conditions, we then co-cultured M1 macrophages with myofibroblasts or myoblasts. M1 macrophages significantly decreased relative myofibroblast numbers (from 223 ± 22% to 64 ± 7%), but not myoblast numbers. This pro-inflammatory co-culture model was used to rapidly screen the following four compounds for ability to prevent M1 macrophage-mediated decrease in myofibroblast numbers: L-NAME (inducible nitric oxide synthase inhibitor), SB203580 (p38 mitogen-activated protein kinase inhibitor), SP600125 (c-Jun N-terminal kinase inhibitor) and LY294002 (phosphoinositide 3-kinase [PI3K] inhibitor). We found that LY294002 rescued myofibroblasts and decreased macrophage numbers. Myofibroblast rescue did not occur with L-NAME, SB203580 or SP600125 incubation. In conclusion, these data suggest a PI3K-associated mechanism whereby myofibroblasts can be rescued, despite simulated pro-inflammatory conditions.
Collapse
Affiliation(s)
- Colin Venter
- Discipline of BiochemistrySchool of Life SciencesUniversity of KwaZulu‐NatalScottsvilleSouth Africa
| | - Kathryn H. Myburgh
- Department Physiological SciencesStellenbosch UniversityMatielandSouth Africa
| | - Carola U. Niesler
- Discipline of BiochemistrySchool of Life SciencesUniversity of KwaZulu‐NatalScottsvilleSouth Africa
| |
Collapse
|
50
|
Lewis LN, Hayhoe RPG, Mulligan AA, Luben RN, Khaw KT, Welch AA. Lower Dietary and Circulating Vitamin C in Middle- and Older-Aged Men and Women Are Associated with Lower Estimated Skeletal Muscle Mass. J Nutr 2020; 150:2789-2798. [PMID: 32851397 PMCID: PMC7549302 DOI: 10.1093/jn/nxaa221] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/06/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Age-related loss of skeletal muscle mass contributes to poor outcomes including sarcopenia, physical disability, frailty, type 2 diabetes, and mortality. Vitamin C has physiological relevance to skeletal muscle and may protect it during aging, but few studies have investigated its importance in older populations. OBJECTIVES We aimed to investigate cross-sectional associations of dietary and plasma vitamin C with proxy measures of skeletal muscle mass in a large cohort of middle- and older-aged individuals. METHODS We analyzed data from >13,000 men and women in the European Prospective Investigation into Cancer and Nutrition-Norfolk cohort, aged 42-82 y. Fat-free mass (FFM), as a proxy for skeletal muscle mass, was estimated using bioelectrical impedance analysis and expressed as a percentage of total mass (FFM%) or standardized by BMI (FFMBMI). Dietary vitamin C intakes were calculated from 7-d food diary data, and plasma vitamin C was measured in peripheral blood. Multivariable regression models, including relevant lifestyle, dietary, and biological covariates, were used to determine associations between FFM measures and quintiles of dietary vitamin C or insufficient compared with sufficient plasma vitamin C (<50 μmol/L and ≥50 μmol/L). RESULTS Positive trends were found across quintiles of dietary vitamin C and FFM measures for both sexes, with interquintile differences in FFM% and FFMBMI of 1.0% and 2.3% for men and 1.9% and 2.9% for women, respectively (all P < 0.001). Similarly, FFM% and FFMBMI measures were higher in participants with sufficient than with insufficient plasma vitamin C: by 1.6% and 2.0% in men, and 3.4% and 3.9% in women, respectively (all P < 0.001). Associations were also evident in analyses stratified into <65-y and ≥65-y age groups. CONCLUSIONS Our findings of positive associations, of both dietary and circulating vitamin C with measures of skeletal muscle mass in middle- and older-aged men and women, suggest that dietary vitamin C intake may be useful for reducing age-related muscle loss.
Collapse
Affiliation(s)
- Lucy N Lewis
- Department of Epidemiology and Public Health, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - Richard P G Hayhoe
- Department of Epidemiology and Public Health, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - Angela A Mulligan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Robert N Luben
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Ailsa A Welch
- Department of Epidemiology and Public Health, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|