1
|
Ma N, Mourkioti F. Ex vivo two-photon imaging of whole-mount skeletal muscles to visualize stem cell behavior. STAR Protoc 2024; 5:102772. [PMID: 38085638 PMCID: PMC10733746 DOI: 10.1016/j.xpro.2023.102772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/02/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Quiescent skeletal muscle stem cells (MuSCs) are morphologically and functionally heterogeneous and exhibit different lengths of cellular extensions, which we call protrusions. Here, we present a protocol for ex vivo two-photon imaging of MuSCs in their native environment. We describe steps for muscle dissection, fixation, embedding, imaging, and analysis of datasets. This protocol allows the examination of MuSC morphology and protrusions at the single-cell level as well as stem cell numbers. For complete details on the use and execution of this protocol, please refer to Ma et al. (2022).1.
Collapse
Affiliation(s)
- Nuoying Ma
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Program, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Regenerative Medicine, Musculoskeletal Regeneration Program, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Antony JS, Birrer P, Bohnert C, Zimmerli S, Hillmann P, Schaffhauser H, Hoeflich C, Hoeflich A, Khairallah R, Satoh AT, Kappeler I, Ferreira I, Zuideveld KP, Metzger F. Local application of engineered insulin-like growth factor I mRNA demonstrates regenerative therapeutic potential in vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102055. [PMID: 37928443 PMCID: PMC10622308 DOI: 10.1016/j.omtn.2023.102055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023]
Abstract
Insulin-like growth factor I (IGF-I) is a growth-promoting anabolic hormone that fosters cell growth and tissue homeostasis. IGF-I deficiency is associated with several diseases, including growth disorders and neurological and musculoskeletal diseases due to impaired regeneration. Despite the vast regenerative potential of IGF-I, its unfavorable pharmacokinetic profile has prevented it from being used therapeutically. In this study, we resolved these challenges by the local administration of IGF-I mRNA, which ensures desirable homeostatic kinetics and non-systemic, local dose-dependent expression of IGF-I protein. Furthermore, IGF-I mRNA constructs were sequence engineered with heterologous signal peptides, which improved in vitro protein secretion (2- to 6-fold) and accelerated in vivo functional regeneration (16-fold) over endogenous IGF-I mRNA. The regenerative potential of engineered IGF-I mRNA was validated in a mouse myotoxic muscle injury and rabbit spinal disc herniation models. Engineered IGF-I mRNA had a half-life of 17-25 h in muscle tissue and showed dose-dependent expression of IGF-I over 2-3 days. Animal models confirm that locally administered IGF-I mRNA remained at the site of injection, contributing to the safety profile of mRNA-based treatment in regenerative medicine. In summary, we demonstrate that engineered IGF-I mRNA holds therapeutic potential with high clinical translatability in different diseases.
Collapse
Affiliation(s)
| | | | | | - Sina Zimmerli
- Versameb AG, Technology Park, 4057 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Pizza FX, Buckley KH. Regenerating Myofibers after an Acute Muscle Injury: What Do We Really Know about Them? Int J Mol Sci 2023; 24:12545. [PMID: 37628725 PMCID: PMC10454182 DOI: 10.3390/ijms241612545] [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: 07/12/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Injury to skeletal muscle through trauma, physical activity, or disease initiates a process called muscle regeneration. When injured myofibers undergo necrosis, muscle regeneration gives rise to myofibers that have myonuclei in a central position, which contrasts the normal, peripheral position of myonuclei. Myofibers with central myonuclei are called regenerating myofibers and are the hallmark feature of muscle regeneration. An important and underappreciated aspect of muscle regeneration is the maturation of regenerating myofibers into a normal sized myofiber with peripheral myonuclei. Strikingly, very little is known about processes that govern regenerating myofiber maturation after muscle injury. As knowledge of myofiber formation and maturation during embryonic, fetal, and postnatal development has served as a foundation for understanding muscle regeneration, this narrative review discusses similarities and differences in myofiber maturation during muscle development and regeneration. Specifically, we compare and contrast myonuclear positioning, myonuclear accretion, myofiber hypertrophy, and myofiber morphology during muscle development and regeneration. We also discuss regenerating myofibers in the context of different types of myofiber necrosis (complete and segmental) after muscle trauma and injurious contractions. The overall goal of the review is to provide a framework for identifying cellular and molecular processes of myofiber maturation that are unique to muscle regeneration.
Collapse
Affiliation(s)
- Francis X. Pizza
- Department of Exercise and Rehabilitation Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Kole H. Buckley
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA;
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 0] [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
|
5
|
Groppa E, Martini P, Derakhshan N, Theret M, Ritso M, Tung LW, Wang YX, Soliman H, Hamer MS, Stankiewicz L, Eisner C, Erwan LN, Chang C, Yi L, Yuan JH, Kong S, Weng C, Adams J, Chang L, Peng A, Blau HM, Romualdi C, Rossi FMV. Spatial compartmentalization of signaling imparts source-specific functions on secreted factors. Cell Rep 2023; 42:112051. [PMID: 36729831 DOI: 10.1016/j.celrep.2023.112051] [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: 10/04/2020] [Revised: 09/08/2022] [Accepted: 01/16/2023] [Indexed: 02/03/2023] Open
Abstract
Efficient regeneration requires multiple cell types acting in coordination. To better understand the intercellular networks involved and how they change when regeneration fails, we profile the transcriptome of hematopoietic, stromal, myogenic, and endothelial cells over 14 days following acute muscle damage. We generate a time-resolved computational model of interactions and identify VEGFA-driven endothelial engagement as a key differentiating feature in models of successful and failed regeneration. In addition, the analysis highlights that the majority of secreted signals, including VEGFA, are simultaneously produced by multiple cell types. To test whether the cellular source of a factor determines its function, we delete VEGFA from two cell types residing in close proximity: stromal and myogenic progenitors. By comparing responses to different types of damage, we find that myogenic and stromal VEGFA have distinct functions in regeneration. This suggests that spatial compartmentalization of signaling plays a key role in intercellular communication networks.
Collapse
Affiliation(s)
- Elena Groppa
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada; Borea Therapeutics, Scuola Internazionale Superiore di Studi Avanzati, Via Bonomea 265, Trieste, Italy
| | - Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Biology, University of Padova, via U. Bassi 58B, Padova, Italy
| | - Nima Derakhshan
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Marine Theret
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Morten Ritso
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Yu Xin Wang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hesham Soliman
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada; Faculty of Pharmaceutical Sciences, Minia University, Minia, Egypt; Aspect Biosystems, 1781 W 75th Avenue, Vancouver, BC, Canada
| | - Mark Stephen Hamer
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Laura Stankiewicz
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Christine Eisner
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Le Nevé Erwan
- Department of Pediatrics, Université Laval, Laval, QC, Canada
| | - Chihkai Chang
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Lin Yi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Jack H Yuan
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Sunny Kong
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Curtis Weng
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Josephine Adams
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Lucas Chang
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Anne Peng
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chiara Romualdi
- Department of Biology, University of Padova, via U. Bassi 58B, Padova, Italy
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada.
| |
Collapse
|
6
|
Bencze M. Mechanisms of Myofibre Death in Muscular Dystrophies: The Emergence of the Regulated Forms of Necrosis in Myology. Int J Mol Sci 2022; 24:ijms24010362. [PMID: 36613804 PMCID: PMC9820579 DOI: 10.3390/ijms24010362] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
Myofibre necrosis is a central pathogenic process in muscular dystrophies (MD). As post-lesional regeneration cannot fully compensate for chronic myofibre loss, interstitial tissue accumulates and impairs muscle function. Muscle regeneration has been extensively studied over the last decades, however, the pathway(s) controlling muscle necrosis remains largely unknown. The recent discovery of several regulated cell death (RCD) pathways with necrotic morphology challenged the dogma of necrosis as an uncontrolled process, opening interesting perspectives for many degenerative disorders. In this review, we focus on how cell death affects myofibres in MDs, integrating the latest research in the cell death field, with specific emphasis on Duchenne muscular dystrophy, the best-known and most common hereditary MD. The role of regulated forms of necrosis in myology is still in its infancy but there is increasing evidence that necroptosis, a genetically programmed form of necrosis, is involved in muscle degenerating disorders. The existence of apoptosis in myofibre demise will be questioned, while other forms of non-apoptotic RCDs may also have a role in myonecrosis, illustrating the complexity and possibly the heterogeneity of the cell death pathways in muscle degenerating conditions.
Collapse
Affiliation(s)
- Maximilien Bencze
- “Biology of the Neuromuscular System” Team, Institut Mondor de Recherche Biomédicale (IMRB), University Paris-Est Créteil, INSERM, U955 IMRB, 94010 Créteil, France;
- École Nationale Vétérinaire d’Alfort, IMRB, 94700 Maisons-Alfort, France
| |
Collapse
|
7
|
Jorda A, Campos-Campos J, Aldasoro C, Colmena C, Aldasoro M, Alvarez K, Valles SL. Protective action of ultrasound-guided electrolysis technique on the muscle damage induced by notexin in rats. PLoS One 2022; 17:e0276634. [PMID: 36441673 PMCID: PMC9704622 DOI: 10.1371/journal.pone.0276634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 10/10/2022] [Indexed: 11/29/2022] Open
Abstract
It is known that exercise can be one of the causes of muscular damage. In recent times, physiotherapists and medical professionals have been employing USGET techniques to stimulate muscle recovery to improve its performance after the injury. We pretend to analyse if the Ultrasound-guided electrolysis (USGET) technique could reduce muscle damage, inflammation, and pain in the present study. Female Wistar rats were assigned to one of three different groups: control (C), notexin (NOT) and notexin with USGET (electrolysis at 6mA) (NOT+USGET). We used the USGT technique, based on electrical stimulation with a continuous current of 4 pulses at an intensity of 6 mA for 5 seconds, conveyed to the muscle. The response was tested with motor function tests. In these tests, we could observe an increase in time and foot faults when crossing a beam in the NOT group compared to C group rats. On the other hand, a significant decrease in both variables was detected in the NOT+USGET compared to the NOT group. Muscle power was measured with a grip strength test, obtaining far better performances in NOT+USGET rats when compared to NOT rats. Moreover, the USGET technique prevented the increase of pro-inflammatory proteins IL-6 and chemokines CCL3 (Chemokine (C-C motif) ligand 3), CCL4 (Chemokine (C-C motif) ligand 4), and CCL5 (Chemokine (C-C motif) ligand 5) with their receptor CCR5 (C-C chemokine receptor type 5), induced by notexin in the quadriceps. At the same time, the study evidenced a decrease in both CCR8 (C-C chemokine receptor type 5,) and NF-ᴋB (nuclear factor- ᴋB) expressions after USGET treatment. On the other hand, we obtained evidence that demonstrated anti-inflammatory properties of the USGET technique, thus being the increase in IL-10 (Interleukin 10) and IL-13 (Interleukin 13) in the NOT+USGET group compared to the NOT group. Furthermore, when applying NSGET after damage, an increase in anti-inflammatory mediators and reduction of pro-inflammatory mediators, which, overall, promoted muscle regeneration, was observed. These results support the idea that the NSGET technique improves muscle recovery after toxic damages, which would justify its employment.
Collapse
Affiliation(s)
- Adrian Jorda
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
- Faculty of Nursing and Podiatry, Department of Nursing, University of Valencia, Valencia, Spain
| | - Juan Campos-Campos
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
- Faculty of Nursing and Podiatry, Department of Nursing, University of Valencia, Valencia, Spain
| | - Constanza Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Carlos Colmena
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Martin Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Kenia Alvarez
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Soraya L. Valles
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
8
|
Washington TA, Haynie WS, Schrems ER, Perry RA, Brown LA, Williams BM, Rosa-Caldwell ME, Lee DE, Brown JL. Effects of PGC-1α overexpression on the myogenic response during skeletal muscle regeneration. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 4:198-208. [PMID: 36090923 PMCID: PMC9453693 DOI: 10.1016/j.smhs.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022] Open
Abstract
The ability of skeletal muscle to regenerate from injury is crucial for locomotion, metabolic health, and quality of life. Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1A) is a transcriptional coactivator required for mitochondrial biogenesis. Increased mitochondrial biogenesis is associated with improved muscle cell differentiation, however PGC1A's role in skeletal muscle regeneration following damage requires further investigation. The purpose of this study was to investigate the role of skeletal muscle-specific PGC1A overexpression during regeneration following damage. 22 C57BL/6J (WT) and 26 PGC1A muscle transgenic (A1) mice were injected with either phosphate-buffered saline (PBS, uninjured control) or Bupivacaine (MAR, injured) into their tibialis anterior (TA) muscle to induce skeletal muscle damage. TA muscles were extracted 3- or 28-days post-injury and analyzed for markers of regenerative myogenesis and protein turnover. Pgc1a mRNA was ∼10–20 fold greater in A1 mice. Markers of protein synthesis, AKT and 4EBP1, displayed decreases in A1 mice compared to WT at both timepoints indicating a decreased protein synthetic response. Myod mRNA was ∼75% lower compared to WT 3 days post-injection. WT mice exhibited decreased cross-sectional area of the TA muscle at 28 days post-injection with bupivacaine compared to all other groups. PGC1A overexpression modifies the myogenic response during regeneration.
Collapse
Affiliation(s)
- Tyrone A. Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
- Corresponding author. University of Arkansas Department of Health, Human Performance, and Recreation, 155 Stadium Dr. HPER 309, Fayetteville, AR, 72701, USA.
| | - Wesley S. Haynie
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Eleanor R. Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Richard A. Perry
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Lemuel A. Brown
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Breanna M. Williams
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Megan E. Rosa-Caldwell
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - David E. Lee
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Jacob L. Brown
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| |
Collapse
|
9
|
Lee EJ, Lee MM, Park S, Jeong KS. Sirt2 positively regulates muscle regeneration after Notexin-induced muscle injury. Exp Mol Pathol 2022; 127:104798. [DOI: 10.1016/j.yexmp.2022.104798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/01/2022] [Accepted: 05/24/2022] [Indexed: 01/04/2023]
|
10
|
Steverink JG, Piluso S, Malda J, Verlaan JJ. Comparison of in vitro and in vivo Toxicity of Bupivacaine in Musculoskeletal Applications. FRONTIERS IN PAIN RESEARCH 2022; 2:723883. [PMID: 35295435 PMCID: PMC8915669 DOI: 10.3389/fpain.2021.723883] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
The recent societal debate on opioid use in treating postoperative pain has sparked the development of long-acting, opioid-free analgesic alternatives, often using the amino-amide local anesthetic bupivacaine as active pharmaceutical ingredient. A potential application is musculoskeletal surgeries, as these interventions rank amongst the most painful overall. Current literature showed that bupivacaine induced dose-dependent myo-, chondro-, and neurotoxicity, as well as delayed osteogenesis and disturbed wound healing in vitro. These observations did not translate to animal and clinical research, where toxic phenomena were seldom reported. An exception was bupivacaine-induced chondrotoxicity, which can mainly occur during continuous joint infusion. To decrease opioid consumption and provide sustained pain relief following musculoskeletal surgery, new strategies incorporating high concentrations of bupivacaine in drug delivery carriers are currently being developed. Local toxicity of these high concentrations is an area of further research. This review appraises relevant in vitro, animal and clinical studies on musculoskeletal local toxicity of bupivacaine.
Collapse
Affiliation(s)
- Jasper G Steverink
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Utrecht, Utrecht University, Utrecht, Netherlands
| | - Susanna Piluso
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, Enschede, Netherlands
| | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jorrit-Jan Verlaan
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
11
|
Goldman SM, Feng JP, Corona BT. Volumetric muscle loss disrupts length-dependent architectural and functional characteristics of skeletal muscle. Connect Tissue Res 2021; 62:72-82. [PMID: 32660287 DOI: 10.1080/03008207.2020.1789608] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose/Aim: Skeletal muscle architecture is a primary determinant of function. Volumetric muscle loss (VML) injury is destructive; however, the impact on muscle architecture is uncharacterized. Methods: Architectural and functional effects of VML were assessed in rat tibialis anterior (TA) muscle model 4 weeks post-injury. Results: VML caused a 31% and 33% reduction in muscle weight (p < 0.001) and fiber length (p = 0.002), respectively, culminating a 34% reduction of fiber to muscle length ratio (FL:ML; p < 0.001). Fiber pennation angle (+14%; p = 0.150) and physiological cross-sectional area (PCSA; -12%; p = 0.220) were unchanged. VML injury reduced peak isometric force (Po) by 36% (p < 0.001), specific force (sPo = Po/PCSA) by 41% (vs. Po, p > 0.999), and force per gram muscle weight (Po/mw) by 18% (vs. Po, p < 0.001). VML injury increased the length at which Po was produced (Lo) by 8% (p = 0.009), and reduced functional excursion by 35% (p = 0.035). Conclusion: The architectural changes after VML injury preserved PCSA, and therefore preserved "potential" maximal force-producing capacity. At most, only half the Po deficit was due directly to the cumulative effect of horizontal and longitudinal tissue loss. Highlighting the impact of longitudinal muscle loss, VML injury reduced fiber length, and FL:ML and grossly disrupted length-dependent functional properties. These findings raise the importance of augmenting length-dependent muscle properties to optimize functional recovery after VML injury.
Collapse
Affiliation(s)
- Stephen M Goldman
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research , Fort Sam Houston, TX, USA.,DoD-VA Extremity Trauma and Amputation Center of Excellence , Fort Sam Houston, TX, USA.,Department of Surgery, Uniformed Services University of the Health Sciences , Bethesda, MD, USA.,Department of Surgery, Walter Reed National Military Medical Center , Bethesda, MD, USA
| | - Jonathan P Feng
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research , Fort Sam Houston, TX, USA
| | - Benjamin T Corona
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research , Fort Sam Houston, TX, USA
| |
Collapse
|
12
|
Forcina L, Cosentino M, Musarò A. Mechanisms Regulating Muscle Regeneration: Insights into the Interrelated and Time-Dependent Phases of Tissue Healing. Cells 2020; 9:E1297. [PMID: 32456017 PMCID: PMC7290814 DOI: 10.3390/cells9051297] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Despite a massive body of knowledge which has been produced related to the mechanisms guiding muscle regeneration, great interest still moves the scientific community toward the study of different aspects of skeletal muscle homeostasis, plasticity, and regeneration. Indeed, the lack of effective therapies for several physiopathologic conditions suggests that a comprehensive knowledge of the different aspects of cellular behavior and molecular pathways, regulating each regenerative stage, has to be still devised. Hence, it is important to perform even more focused studies, taking the advantage of robust markers, reliable techniques, and reproducible protocols. Here, we provide an overview about the general aspects of muscle regeneration and discuss the different approaches to study the interrelated and time-dependent phases of muscle healing.
Collapse
Affiliation(s)
| | | | - Antonio Musarò
- Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Antonio Scarpa, 14, 00161 Rome, Italy; (L.F.); (M.C.)
| |
Collapse
|
13
|
Pablos A, Ceca D, Jorda A, Rivera P, Colmena C, Elvira L, Martínez-Arnau FM, Valles SL. Protective Effects of Foam Rolling against Inflammation and Notexin Induced Muscle Damage in Rats. Int J Med Sci 2020; 17:71-81. [PMID: 31929740 PMCID: PMC6945557 DOI: 10.7150/ijms.37981] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/14/2019] [Indexed: 01/05/2023] Open
Abstract
It is known that high-intensity exercise can cause inflammation and damage in muscle tissue, and in recent years, physical therapists and fitness professionals have begun to use foam rolling as a recovery method to improve performance. Despite the lack of basic science studies to support or refute the efficacy of foam rolling, the technique is very widely used in the sports world. In this respect, we investigated whether foam rolling could attenuate muscle damage and inflammation. Female Wistar rats were assigned to control (C), foam rolling (FR), notexin without foam rolling (N) and notexin with foam rolling (NFR) groups. A 4.5 x 2 cm foam roller was used to massage their hind legs (two 60-second repetitions twice a day for 3 days). Motor function tests (Balance Beam Test and Grip strength) were used. We detected an increase in time and foot faults when crossing a beam in the N group compared to C and FR rats. In contrast, a significant decrease was detected in both tests in NFR compared to N rats. Muscle power was measured with a grip strength test and better performance was detected in NFR rats compared to N rats. Furthermore, an increase of pro-inflammatory proteins was noted in the N group, while there was a decrease in the NFR group. On the contrary, an increase in PPAR-γ (anti-inflammatory protein) in the NFR group compared to the N group demonstrates the anti-inflammatory properties of the foam rolling technique. In summary, applying foam rolling after damage has benefits such as an increase in anti-inflammatory proteins and a reduction of pro-inflammatory proteins, resulting in muscle recovery and better performance.
Collapse
Affiliation(s)
- Ana Pablos
- Faculty of Physical Activity and Sport Sciences, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Diego Ceca
- Department of Education, Universidad Internacional de Valencia, Valencia, Spain.,Faculty of Physical Activity and Sport Sciences, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Adrián Jorda
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Pilar Rivera
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Carlos Colmena
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Laura Elvira
- Faculty of Physical Activity and Sport Sciences, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Francisco M Martínez-Arnau
- Faculty of Nursing, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain.,Departament of Physiotherapy, University of Valencia, Valencia, Spain
| | - Soraya L Valles
- Department of Physiology, School of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
14
|
Greising SM, Corona BT, McGann C, Frankum JK, Warren GL. Therapeutic Approaches for Volumetric Muscle Loss Injury: A Systematic Review and Meta-Analysis. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:510-525. [PMID: 31578930 DOI: 10.1089/ten.teb.2019.0207] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Our goal was to understand the impact of regenerative therapies on the functional capacity of skeletal muscle following volumetric muscle loss (VML) injury. An extensive database search (e.g., PubMed, Cochrane Library, and ClinicalTrials.gov) was conducted up through January 2019 to evaluate the following: "In humans or animals with VML injury, is treatment better than no treatment at recovering functional capacity?" Study eligibility criteria required studies to have both an untreated and at least one treated VML injury group. From 2312 study reports, 44 studies met the inclusion criteria. Quantitative functional capacity data (absolute and/or normalized strength) or proportional measures (histological analysis quantifying viable muscle tissue, mitochondrial function, and/or exhaustive treadmill running) were extracted for use. While both human and animal studies were included in the searches, only animal studies met the eligibility criteria. Using a random-effects model, Hedges' g was used as the effect size (ES) and calculated such that a positive ES indicated treatment efficacy. The overall ES was 0.75 (95% confidence interval: 0.53-0.96; p < 0.0000001), indicating that the treatments, on average, resulted in a significant improvement in functional capacity. From network meta-analyses, it was determined that an acellular biomaterial combined with stem and/or progenitor cells had the greatest treatment effectiveness. The findings indicate that various treatments in animal models of VML improve the functional capacity of muscle compared to leaving the injury untreated; however, the ∼16% beneficial effect is small. Our results suggest that current regenerative therapy paradigms require further maturation to achieve clinically meaningful improvements in the functional capacity of the muscle. Impact Statement Our most salient findings are that (1) various treatment approaches used in animal models of volumetric muscle loss (VML) injury improve functional capacity compared to leaving the injury untreated and (2) an acellular biomaterial in combination with cellular components was the most effective treatment to improve functional capacity following VML injury to date. The nature of our findings has substantial implications for regenerative medicine, biomedical engineering, and rehabilitative techniques currently being evaluated and developed for VML injury repair, and are pivotal to the progression of the regenerative medicine effort aimed at restoring maximal function to traumatized and disabled limbs.
Collapse
Affiliation(s)
- Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota
| | - Benjamin T Corona
- School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Christopher McGann
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| | - Jeremy K Frankum
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| | - Gordon L Warren
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| |
Collapse
|
15
|
Biotoxins in muscle regeneration research. J Muscle Res Cell Motil 2019; 40:291-297. [PMID: 31359301 DOI: 10.1007/s10974-019-09548-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/25/2019] [Indexed: 10/26/2022]
Abstract
Skeletal muscles are characterized by their unique regenerative capacity following injury due to the presence of muscle precursor cells, satellite cells. This characteristic allows researchers to study muscle regeneration using experimental injury models. These injury models should be stable and reproducible. Variety of injury models have been used, among which the intramuscular injection of myotoxic biotoxins is considered the most common and widespread method in muscle regeneration research. By using isolated biotoxins, researchers could induce acute muscle damage and regeneration in a controlled and reproducible manner. Therefore, it is considered an easy method for inducing muscle injury in order to understand the different mechanisms involved in muscle injuries and tissue response following injury. However, different toxins and venoms have different compositions and subsequently the possible effects of these toxins on skeletal muscle vary according to their composition. Moreover, regeneration of injured muscle by venoms and toxins varies according to the target of toxin or venom. Therefore, it is essential for researcher to be aware of the mechanism and possible target of toxin-induced injury. The current paper provides an overview of the biotoxins used in skeletal muscle research.
Collapse
|
16
|
Mahdy MAA, Warita K, Hosaka YZ. Glycerol induces early fibrosis in regenerating rat skeletal muscle. J Vet Med Sci 2018; 80:1646-1649. [PMID: 30282842 PMCID: PMC6261811 DOI: 10.1292/jvms.18-0328] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Glycerol has been recently used to induce muscle adiposity in mice. However, its effects on the rat muscles have not been investigated previously. Therefore, we investigated the regeneration outcomes of rat muscles following glycerol-induced injury at different time points. Glycerol injection induced myofiber degeneration with extensive inflammatory infiltration on day 4 followed by appearance of regenerating myotubes on day 7 after injury without adipocyte infiltration. Meanwhile, a significant collagen deposition at early stage of regeneration that increased together with persistent inflammatory infiltration up to day 14 after injury indicates impaired regeneration. In conclusion, glycerol injury in rats is more suitable as a fibrosis-inducing model than in mice due to earlier and higher accumulation of fibrous tissue with lacking adipogenesis.
Collapse
Affiliation(s)
- Mohamed A A Mahdy
- Laboratory of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi 753-8515, Japan.,Department of Anatomy and Embryology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt.,Department of Anatomy and Physiology, University of Pretoria, Onderstepoort 0110, South Africa
| | - Katsuhiko Warita
- Laboratory of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi 753-8515, Japan.,Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori 680-8553, Japan
| | - Yoshinao Z Hosaka
- Laboratory of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi 753-8515, Japan.,Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori 680-8553, Japan
| |
Collapse
|
17
|
Hussain N, McCartney C, Neal J, Chippor J, Banfield L, Abdallah F. Local anaesthetic-induced myotoxicity in regional anaesthesia: a systematic review and empirical analysis. Br J Anaesth 2018; 121:822-841. [DOI: 10.1016/j.bja.2018.05.076] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/16/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
|
18
|
Mahdy MAA. Glycerol-induced injury as a new model of muscle regeneration. Cell Tissue Res 2018; 374:233-241. [DOI: 10.1007/s00441-018-2846-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/24/2018] [Indexed: 01/15/2023]
|
19
|
Sung SE, Hwang M, Kim AY, Lee EM, Lee EJ, Hwang SK, Kim SY, Kim HK, Jeong KS. MyoD Overexpressed Equine Adipose-Derived Stem Cells Enhanced Myogenic Differentiation Potential. Cell Transplant 2018; 25:2017-2026. [PMID: 26892394 DOI: 10.3727/096368916x691015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells could potentially be used in the clinical treatment of muscle disorders and muscle regeneration. Adipose-derived stem cells (ADSCs) can be easily isolated from adipose tissue, as opposed to stem cells of other tissues. We believe that cell therapy using ADSCs could be applied to muscle disorders in horses and other species. We sought to improve the myogenic differentiation potential of equine ADSCs (eqADSCs) using a MyoD lentiviral vector. MyoD lentiviruses were transduced into eqADSCs and selected using puromycin. Cells were cultured in differentiation media containing 5% horse serum, and after 5 days the MyoD-transduced cells differentiated into myogenic cells (MyoD-eqADSCs). Using green fluorescent protein (GFP), MyoD-eqADSCs were purified and transplanted into the tibialis anterior muscles of mice after they were injured with the myotoxin notexin. The mice were sacrificed to examine any regeneration in the tibialis anterior muscle 4 weeks after the MyoD-eqADSCs were injected. The MyoD-eqADSCs cultured in growth media expressed murine and equine MyoD; however, they did not express late differentiation markers such as myogenin (MYOG). When cells were grown in differentiation media, the expression of MYOG was clearly observed. According to our reverse transcription polymerase chain reaction and immunocytochemistry results, MyoD-eqADSCs expressed terminal myogenic phase genes, such as those encoding dystrophin, myosin heavy chain, and troponin I. The MyoD-eqADSCs fused to each other, and the formation of myotube-like cells from myoblasts in differentiation media occurred between days 5 and 14 postplating. In mice, we observed GFP-positive myofibers, which had differentiated from the injected MyoD-eqADSCs. Our approaches improved the myogenic differentiation of eqADSCs through the forced expression of murine MyoD. Our findings suggest that limitations in the treatment of equine muscle disorders could be overcome using ADSCs.
Collapse
Affiliation(s)
- Soo-Eun Sung
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Meeyul Hwang
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Ah-Young Kim
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Eun-Mi Lee
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Eun-Joo Lee
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Su-Kyeong Hwang
- Department of Pediatrics, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Shin-Yoon Kim
- Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hong-Kyun Kim
- Department of Ophthalmology, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Kyu-Shik Jeong
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
20
|
Goldman SM, Henderson BEP, Walters TJ, Corona BT. Co-delivery of a laminin-111 supplemented hyaluronic acid based hydrogel with minced muscle graft in the treatment of volumetric muscle loss injury. PLoS One 2018; 13:e0191245. [PMID: 29329332 PMCID: PMC5766229 DOI: 10.1371/journal.pone.0191245] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/02/2018] [Indexed: 12/21/2022] Open
Abstract
Minced muscle autografting mediates de novo myofiber regeneration and promotes partial recovery of neuromuscular strength after volumetric muscle loss injury (VML). A major limitation of this approach is the availability of sufficient donor tissue for the treatment of relatively large VMLs without inducing donor site morbidity. This study evaluated a laminin-111 supplemented hyaluronic acid based hydrogel (HA+LMN) as a putative myoconductive scaffolding to be co-delivered with minced muscle grafts. In a rat tibialis anterior muscle VML model, delivery of a reduced dose of minced muscle graft (50% of VML defect) within HA+LMN resulted in a 42% improvement of peak tetanic torque production over unrepaired VML affected limbs. However, the improvement in strength was not improved compared to a 50% minced graft-only control group. Moreover, histological analysis revealed that the improvement in in vivo functional capacity mediated by minced grafts in HA+LMN was not accompanied by a particularly robust graft mediated regenerative response as determined through donor cell tracking of the GFP+ grafting material. Characterization of the spatial distribution and density of macrophage and satellite cell populations indicated that the combination therapy damps the heightened macrophage response while re-establishing satellite content 14 days after VML to a level consistent with an endogenously healing ischemia-reperfusion induced muscle injury. Moreover, regional analysis revealed that the combination therapy increased satellite cell density mostly in the remaining musculature, as opposed to the defect area. Based on the results, the following salient conclusions were drawn: 1) functional recovery mediated by the combination therapy is likely due to a superposition of de novo muscle fiber regeneration and augmented repair of muscle fibers within the remaining musculature, and 2) The capacity for VML therapies to augment regeneration and repair within the remaining musculature may have significant clinical impact and warrants further exploration.
Collapse
Affiliation(s)
- Stephen M. Goldman
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Beth E. P. Henderson
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Thomas J. Walters
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| | - Benjamin T. Corona
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, Texas, United States of America
| |
Collapse
|
21
|
Cezar CA, Arany P, Vermillion SA, Seo BR, Vandenburgh HH, Mooney DJ. Timed Delivery of Therapy Enhances Functional Muscle Regeneration. Adv Healthc Mater 2017; 6:10.1002/adhm.201700202. [PMID: 28703489 PMCID: PMC5641972 DOI: 10.1002/adhm.201700202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/09/2017] [Indexed: 01/14/2023]
Abstract
Cell transplantation is a promising therapeutic strategy for the treatment of traumatic muscle injury in humans. Previous investigations have typically focused on the identification of potent cell and growth factor treatments and optimization of spatial control over delivery. However, the optimal time point for cell transplantation remains unclear. Here, this study reports how myoblast and morphogen delivery timed to coincide with specific phases of the inflammatory response affects donor cell engraftment and the functional repair of severely injured muscle. Delivery of a biomaterial-based therapy timed with the peak of injury-induced inflammation leads to potent early and long-term regenerative benefits. Diminished inflammation and fibrosis, enhanced angiogenesis, and increased cell engraftment are seen during the acute stage following optimally timed treatment. Over the long term, treatment during peak inflammation leads to enhanced functional regeneration, as indicated by reduced chronic inflammation and fibrosis along with increased tissue perfusion and muscle contractile force. Treatments initiated immediately after injury or after inflammation had largely resolved provided more limited benefits. These results demonstrate the importance of appropriately timing the delivery of biologic therapy in the context of muscle regeneration. Biomaterial-based timed delivery can likely be applied to other tissues and is of potential wide utility in regenerative medicine.
Collapse
Affiliation(s)
- Christine A Cezar
- Harvard School of Engineering and Applied Sciences, 29 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford Street, Suite 403, Cambridge, MA, 02138, USA
| | - Praveen Arany
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14260, USA
| | - Sarah A Vermillion
- Harvard School of Engineering and Applied Sciences, 29 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford Street, Suite 403, Cambridge, MA, 02138, USA
| | - Bo Ri Seo
- Harvard School of Engineering and Applied Sciences, 29 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford Street, Suite 403, Cambridge, MA, 02138, USA
| | - Herman H Vandenburgh
- Department of Pathology and Lab Medicine, Brown University, Providence, RI, 02912, USA
| | - David J Mooney
- Harvard School of Engineering and Applied Sciences, 29 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford Street, Suite 403, Cambridge, MA, 02138, USA
| |
Collapse
|
22
|
Corona BT, Wenke JC, Ward CL. Pathophysiology of Volumetric Muscle Loss Injury. Cells Tissues Organs 2016; 202:180-188. [PMID: 27825160 DOI: 10.1159/000443925] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2016] [Indexed: 11/19/2022] Open
Abstract
Volumetric muscle loss (VML) injuries are prevalent in civilian and military trauma patients and are known to impart chronic functional deficits. The frank loss of muscle tissue that defines VML injuries is beyond the robust reparative and regenerative capacities of mammalian skeletal muscle. Given the nature of VML injuries, there is a clear need to develop therapies that promote de novo regeneration of skeletal muscle fibers, which can integrate with the remaining musculature and restore muscle strength. However, the pathophysiology of VML injuries is not completely defined, and, therefore, there may be other opportunities to improve functional outcomes other than de novo regeneration. Herein, clinical and preclinical studies of VML were reviewed to ascertain salient manifestations of VML injury that can impair limb function and muscle strength. The limited clinical data available highlighted proliferative fibrosis secondary to VML injury as a viable target to improve limb range of motion. Selected preclinical studies that used standardized neuromuscular functional assessments broadly identified that the muscle mass remaining after VML injury is performing suboptimally, and, therefore, percent VML strength deficits are significantly worse than can be explained by the initial frank loss of contractile machinery. Potential mechanisms of suboptimal strength of the remaining muscle mass suggested within the literature include intramuscular nerve damage, muscle architectural perturbations, and diminished transmission of force. Collectively, both clinical and preclinical data indicate a complex pathophysiology after VML that presents multiple therapeutic targets. This is a work of the US Government and is not subject to copyright protection in the USA. Foreign copyrights may apply. Published by S. Karger AG, Basel.
Collapse
|
23
|
Itoh Y, Murakami T, Mori T, Agata N, Kimura N, Inoue-Miyazu M, Hayakawa K, Hirano T, Sokabe M, Kawakami K. Training at non-damaging intensities facilitates recovery from muscle atrophy. Muscle Nerve 2016; 55:243-253. [PMID: 27301985 DOI: 10.1002/mus.25218] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2016] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Resistance training promotes recovery from muscle atrophy, but optimum training programs have not been established. We aimed to determine the optimum training intensity for muscle atrophy. METHODS Mice recovering from atrophied muscles after 2 weeks of tail suspension underwent repeated isometric training with varying joint torques 50 times per day. RESULTS Muscle recovery assessed by maximal isometric contraction and myofiber cross-sectional areas (CSAs) were facilitated at 40% and 60% maximum contraction strength (MC), but at not at 10% and 90% MC. At 60% and 90% MC, damaged and contained smaller diameter fibers were observed. Activation of myogenic satellite cells and a marked increase in myonuclei were observed at 40%, 60%, and 90% MC. CONCLUSIONS The increases in myofiber CSAs were likely caused by increased myonuclei formed through fusion of resistance-induced myofibers with myogenic satellite cells. These data indicate that resistance training without muscle damage facilitates efficient recovery from atrophy. Muscle Nerve 55: 243-253, 2017.
Collapse
Affiliation(s)
- Yuta Itoh
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Faculty of Rehabilitation Science, Nagoya Gakuin University, Seto, Japan
| | - Taro Murakami
- Faculty of Wellness, Shigakkan University, Ohbu, Japan
| | - Tomohiro Mori
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhide Agata
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Faculty of Health and Medical Sciences, Tokoha University, Hamamatsu, Japan
| | - Nahoko Kimura
- Aiche Medical College for Physical and Occupational Therapy, Kiyosu, Japan
| | | | - Kimihide Hayakawa
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takayuki Hirano
- Faculty of Rehabilitation Science, Nagoya Gakuin University, Seto, Japan
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keisuke Kawakami
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Faculty of Welfare and Health Sciences, Oita University, Dannoharu 700, Oita City, 870-1192, Japan
| |
Collapse
|
24
|
Corona BT, Greising SM. Challenges to acellular biological scaffold mediated skeletal muscle tissue regeneration. Biomaterials 2016; 104:238-46. [DOI: 10.1016/j.biomaterials.2016.07.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/11/2016] [Accepted: 07/16/2016] [Indexed: 02/08/2023]
|
25
|
Cho TG, Park SW, Kim YB. Chronic Paraspinal Muscle Injury Model in Rat. J Korean Neurosurg Soc 2016; 59:430-6. [PMID: 27651859 PMCID: PMC5028601 DOI: 10.3340/jkns.2016.59.5.430] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/27/2016] [Accepted: 07/07/2016] [Indexed: 01/26/2023] Open
Abstract
Objective The objective of this study is to establish an animal model of chronic paraspinal muscle injury in rat. Methods Fifty four Sprague-Dawley male rats were divided into experimental group (n=30), sham (n=15), and normal group (n=9). Incision was done from T7 to L2 and paraspinal muscles were detached from spine and tied at each level. The paraspinal muscles were exposed and untied at 2 weeks after surgery. Sham operation was done by paraspinal muscles dissection at the same levels and wound closure was done without tying. Kyphotic index and thoracolumbar Cobb's angle were measured at preoperative, 2, 4, 8, and 12 weeks after the first surgery for all groups. The rats were sacrificed at 4, 8, and 12 weeks after the first surgery, and performed histological examinations. Results At 4 weeks after surgery, the kyphotic index decreased, but, Cobb's angle increased significantly in the experimental group (p<0.05), and then that were maintained until the end of the experiment. However, there were no significant differences of the kyphotic index and Cobb's angle between sham and normal groups. In histological examinations, necrosis and fibrosis were observed definitely and persisted until 12 weeks after surgery. There were also presences of regenerated muscle cells which nucleus is at the center of cytoplasm, centronucleated myofibers. Conclusion Our chronic injury model of paraspinal muscles in rats shows necrosis and fibrosis in the muscles for 12 weeks after surgery, which might be useful to study the pathophysiology of the degenerative thoracolumbar kyphosis or degeneration of paraspinal muscles.
Collapse
Affiliation(s)
- Tack Geun Cho
- Department of Neurosurgery, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Seung Won Park
- Department of Neurosurgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | - Young Baeg Kim
- Department of Neurosurgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Perry RA, Brown LA, Lee DE, Brown JL, Baum JI, Greene NP, Washington TA. Differential effects of leucine supplementation in young and aged mice at the onset of skeletal muscle regeneration. Mech Ageing Dev 2016; 157:7-16. [PMID: 27327351 DOI: 10.1016/j.mad.2016.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 11/29/2022]
Abstract
Aging decreases the ability of skeletal muscle to respond to injury. Leucine has been demonstrated to target protein synthetic pathways in skeletal muscle thereby enhancing this response. However, the effect of aging on leucine-induced alterations in protein synthesis at the onset of skeletal muscle regeneration has not been fully elucidated. The purpose of this study was to determine if aging alters skeletal muscle regeneration and leucine-induced alterations in markers of protein synthesis. The tibialis anterior of young (3 months) and aged (24 months) female C57BL/6J mice were injected with either bupivacaine or PBS, and the mice were given ad libitum access to leucine-supplemented or normal drinking water. Protein and gene expression of markers of protein synthesis and degradation, respectively, were analyzed at three days post-injection. Following injury in young mice, leucine supplementation was observed to elevate only p-p70S6K. In aged mice, leucine was shown to elicit higher p-mTOR content with and without injury, and p-4EBP-1 content post-injury. Additionally in aged mice, leucine was shown to elicit higher content of relative p70S6K post-injury. Our study shows that leucine supplementation affects markers of protein synthesis at the onset of skeletal muscle regeneration differentially in young and aged mice.
Collapse
Affiliation(s)
- Richard A Perry
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, United States
| | - Lemuel A Brown
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, United States
| | - David E Lee
- Integrative Muscle Metabolism Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, United States
| | - Jacob L Brown
- Integrative Muscle Metabolism Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, United States
| | - Jamie I Baum
- Department of Food Science, University of Arkansas, Fayetteville AR 72701, United States
| | - Nicholas P Greene
- Integrative Muscle Metabolism Laboratory, Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, United States
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, United States.
| |
Collapse
|
27
|
Joanisse S, Nederveen JP, Baker JM, Snijders T, Iacono C, Parise G. Exercise conditioning in old mice improves skeletal muscle regeneration. FASEB J 2016; 30:3256-68. [PMID: 27306336 DOI: 10.1096/fj.201600143rr] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 06/07/2016] [Indexed: 12/13/2022]
Abstract
Skeletal muscle possesses the ability to regenerate after injury, but this ability is impaired or delayed with aging. Regardless of age, muscle retains the ability to positively respond to stimuli, such as exercise. We examined whether exercise is able to improve regenerative response in skeletal muscle of aged mice. Twenty-two-month-old male C57Bl/6J mice (n = 20) underwent an 8-wk progressive exercise training protocol [old exercised (O-Ex) group]. An old sedentary (O-Sed) and a sedentary young control (Y-Ctl) group were included. Animals were subjected to injections of cardiotoxin into the tibialis anterior muscle. The tibialis anterior were harvested before [O-Ex/O-Sed/Y-Ctl control (CTL); n = 6], 10 d (O-Ex/O-Sed/Y-Ctl d 10; n = 8), and 28 d (O-Ex/O-Sed/Y-Ctl d 28; n = 6) postinjection. Average fiber cross-sectional area was reduced in all groups at d 10 (CTL: O-Ex: 2499 ± 140; O-Sed: 2320 ± 165; Y-Ctl: 2474 ± 269; d 10: O-Ex: 1191 ± 100; O-Sed: 1125 ± 99; Y-Ctl: 1481 ± 167 µm(2); P < 0.05), but was restored to control values in O-Ex and Y-Ctl groups at d 28 (O-Ex: 2257 ± 181; Y-Ctl: 2398 ± 171 µm(2); P > 0.05). Satellite cell content was greater at CTL in O-Ex (2.6 ± 0.4 satellite cells/100 fibers) compared with O-Sed (1.0 ± 0.1% satellite cells/100 fibers; P < 0.05). Exercise conditioning appears to improve ability of skeletal muscle to regenerate after injury in aged mice.-Joanisse, S., Nederveen, J. P., Baker, J. M., Snijders, T., Iacono, C., Parise, G. Exercise conditioning in old mice improves skeletal muscle regeneration.
Collapse
Affiliation(s)
- Sophie Joanisse
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and
| | - Joshua P Nederveen
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and
| | - Jeff M Baker
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and
| | - Tim Snijders
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and
| | - Carlo Iacono
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and
| | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and Department of Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
28
|
Xu X, Wilschut KJ, Kouklis G, Tian H, Hesse R, Garland C, Sbitany H, Hansen S, Seth R, Knott PD, Hoffman WY, Pomerantz JH. Human Satellite Cell Transplantation and Regeneration from Diverse Skeletal Muscles. Stem Cell Reports 2016; 5:419-34. [PMID: 26352798 PMCID: PMC4618654 DOI: 10.1016/j.stemcr.2015.07.016] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/31/2015] [Accepted: 07/31/2015] [Indexed: 12/24/2022] Open
Abstract
Identification of human satellite cells that fulfill muscle stem cell criteria is an unmet need in regenerative medicine. This hurdle limits understanding how closely muscle stem cell properties are conserved among mice and humans and hampers translational efforts in muscle regeneration. Here, we report that PAX7 satellite cells exist at a consistent frequency of 2-4 cells/mm of fiber in muscles of the human trunk, limbs, and head. Xenotransplantation into mice of 50-70 fiber-associated, or 1,000-5,000 FACS-enriched CD56(+)/CD29(+) human satellite cells led to stable engraftment and formation of human-derived myofibers. Human cells with characteristic PAX7, CD56, and CD29 expression patterns populated the satellite cell niche beneath the basal lamina on the periphery of regenerated fibers. After additional injury, transplanted satellite cells robustly regenerated to form hundreds of human-derived fibers. Together, these findings conclusively delineate a source of bona-fide endogenous human muscle stem cells that will aid development of clinical applications.
Collapse
Affiliation(s)
- Xiaoti Xu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Karlijn J Wilschut
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gayle Kouklis
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hua Tian
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Robert Hesse
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Catharine Garland
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hani Sbitany
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Scott Hansen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Rahul Seth
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - P Daniel Knott
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William Y Hoffman
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason H Pomerantz
- Division of Plastic and Reconstructive Surgery, Departments of Surgery and Orofacial Sciences, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
29
|
Neuromuscular electrical stimulation promotes development in mice of mature human muscle from immortalized human myoblasts. Skelet Muscle 2016; 6:4. [PMID: 26925213 PMCID: PMC4769538 DOI: 10.1186/s13395-016-0078-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 01/06/2016] [Indexed: 12/25/2022] Open
Abstract
Background Studies of the pathogenic mechanisms underlying human myopathies and muscular dystrophies often require animal models, but models of some human diseases are not yet available. Methods to promote the engraftment and development of myogenic cells from individuals with such diseases in mice would accelerate such studies and also provide a useful tool for testing therapeutics. Here, we investigate the ability of immortalized human myogenic precursor cells (hMPCs) to form mature human myofibers following implantation into the hindlimbs of non-obese diabetic-Rag1nullIL2rγnull (NOD-Rag)-immunodeficient mice. Results We report that hindlimbs of NOD-Rag mice that are X-irradiated, treated with cardiotoxin, and then injected with immortalized control hMPCs or hMPCs from an individual with facioscapulohumeral muscular dystrophy (FSHD) develop mature human myofibers. Furthermore, intermittent neuromuscular electrical stimulation (iNMES) of the peroneal nerve of the engrafted limb enhances the development of mature fibers in the grafts formed by both immortal cell lines. With control cells, iNMES increases the number and size of the human myofibers that form and promotes closer fiber-to-fiber packing. The human myofibers in the graft are innervated, fully differentiated, and minimally contaminated with murine myonuclei. Conclusions Our results indicate that control and FSHD human myofibers can form in mice engrafted with hMPCs and that iNMES enhances engraftment and subsequent development of mature human muscle.
Collapse
|
30
|
Cohen TV, Many GM, Fleming BD, Gnocchi VF, Ghimbovschi S, Mosser DM, Hoffman EP, Partridge TA. Upregulated IL-1β in dysferlin-deficient muscle attenuates regeneration by blunting the response to pro-inflammatory macrophages. Skelet Muscle 2015; 5:24. [PMID: 26251696 PMCID: PMC4527226 DOI: 10.1186/s13395-015-0048-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/16/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Loss-of-function mutations in the dysferlin gene (DYSF) result in a family of muscle disorders known collectively as the dysferlinopathies. Dysferlin-deficient muscle is characterized by inflammatory foci and macrophage infiltration with subsequent decline in muscle function. Whereas macrophages function to remove necrotic tissue in acute injury, their prevalence in chronic myopathy is thought to inhibit resolution of muscle regeneration. Two major classes of macrophages, classical (M1) and alternative (M2a), play distinct roles during the acute injury process. However, their individual roles in chronic myopathy remain unclear and were explored in this study. METHODS To test the roles of the two macrophage phenotypes on regeneration in dysferlin-deficient muscle, we developed an in vitro co-culture model of macrophages and muscle cells. We assayed the co-cultures using ELISA and cytokine arrays to identify secreted factors and performed transcriptome analysis of molecular networks induced in the myoblasts. RESULTS Dysferlin-deficient muscle contained an excess of M1 macrophage markers, compared with WT, and regenerated poorly in response to toxin injury. Co-culturing macrophages with muscle cells showed that M1 macrophages inhibit muscle regeneration whereas M2a macrophages promote it, especially in dysferlin-deficient muscle cells. Examination of soluble factors released in the co-cultures and transcriptome analysis implicated two soluble factors in mediating the effects: IL-1β and IL-4, which during acute injury are secreted from M1 and M2a macrophages, respectively. To test the roles of these two factors in dysferlin-deficient muscle, myoblasts were treated with IL-4, which improved muscle differentiation, or IL-1β, which inhibited it. Importantly, blockade of IL-1β signaling significantly improved differentiation of dysferlin-deficient cells. CONCLUSIONS We propose that the inhibitory effects of M1 macrophages on myogenesis are mediated by IL-1β signals and suppression of the M1-mediated immune response may improve muscle regeneration in dysferlin deficiency. Our studies identify a potential therapeutic approach to promote muscle regeneration in dystrophic muscle.
Collapse
Affiliation(s)
- Tatiana V. Cohen
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
- />Center for Genetic Muscle Disorders, Kennedy Krieger Institute, 707 N. Broadway, Baltimore, MD 21205 USA
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Gina M. Many
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Bryan D. Fleming
- />Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| | - Viola F. Gnocchi
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Svetlana Ghimbovschi
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - David M. Mosser
- />Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| | - Eric P. Hoffman
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Terence A. Partridge
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| |
Collapse
|
31
|
Simonato M, Morbiato L, Zorzi V, Caccin P, Fernández J, Massimino ML, Polverino de Laureto P, Tonello F. Production in Escherichia coli, folding, purification and characterization of notexin with wild type sequence and with N-terminal and catalytic site mutations. Toxicon 2014; 88:11-20. [DOI: 10.1016/j.toxicon.2014.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/31/2014] [Accepted: 06/11/2014] [Indexed: 12/11/2022]
|
32
|
Church JE, Trieu J, Sheorey R, Chee AYM, Naim T, Baum DM, Ryall JG, Gregorevic P, Lynch GS. Functional β-adrenoceptors are important for early muscle regeneration in mice through effects on myoblast proliferation and differentiation. PLoS One 2014; 9:e101379. [PMID: 25000590 PMCID: PMC4084885 DOI: 10.1371/journal.pone.0101379] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 06/05/2014] [Indexed: 12/25/2022] Open
Abstract
Muscles can be injured in different ways and the trauma and subsequent loss of function and physical capacity can impact significantly on the lives of patients through physical impairments and compromised quality of life. The relative success of muscle repair after injury will largely determine the extent of functional recovery. Unfortunately, regenerative processes are often slow and incomplete, and so developing novel strategies to enhance muscle regeneration is important. While the capacity to enhance muscle repair by stimulating β2-adrenoceptors (β-ARs) using β2-AR agonists (β2-agonists) has been demonstrated previously, the exact role β-ARs play in regulating the regenerative process remains unclear. To investigate β-AR-mediated signaling in muscle regeneration after myotoxic damage, we examined the regenerative capacity of tibialis anterior and extensor digitorum longus muscles from mice lacking either β1-AR (β1-KO) and/or β2-ARs (β2-KO), testing the hypothesis that muscles from mice lacking the β2-AR would exhibit impaired functional regeneration after damage compared with muscles from β1-KO or β1/β2-AR null (β1/β2-KO) KO mice. At 7 days post-injury, regenerating muscles from β1/β2-KO mice produced less force than those of controls but muscles from β1-KO or β2-KO mice did not exhibit any delay in functional restoration. Compared with controls, β1/β2-KO mice exhibited an enhanced inflammatory response to injury, which delayed early muscle regeneration, but an enhanced myoblast proliferation later during regeneration ensured a similar functional recovery (to controls) by 14 days post-injury. This apparent redundancy in the β-AR signaling pathway was unexpected and may have important implications for manipulating β-AR signaling to improve the rate, extent and efficacy of muscle regeneration to enhance functional recovery after injury.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Proliferation
- Gene Knockout Techniques
- Mice
- Muscle Strength
- Muscle, Skeletal/anatomy & histology
- Muscle, Skeletal/cytology
- Muscle, Skeletal/physiology
- Myoblasts/cytology
- Organ Size
- Receptors, Adrenergic, beta-1/deficiency
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/metabolism
- Receptors, Adrenergic, beta-2/deficiency
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Regeneration
Collapse
Affiliation(s)
- Jarrod E. Church
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Jennifer Trieu
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Radhika Sheorey
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Annabel Y. -M. Chee
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Timur Naim
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Dale M. Baum
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - James G. Ryall
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| | - Paul Gregorevic
- Laboratory for Muscle Research & Therapeutics Development, Baker IDI Heart and Diabetes Institute, Victoria, Australia
| | - Gordon S. Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
33
|
|
34
|
Head SI, Houweling PJ, Chan S, Chen G, Hardeman EC. Properties of regenerated mouse extensor digitorum longus muscle following notexin injury. Exp Physiol 2014; 99:664-74. [PMID: 24414176 DOI: 10.1113/expphysiol.2013.077289] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Muscles of mdx mice are known to be more susceptible to contraction-induced damage than wild-type muscle. However, it is not clear whether this is because of dystrophin deficiency or because of the abnormal branching morphology of dystrophic muscle fibres. This distinction has an important bearing on our traditional understanding of the function of dystrophin as a mechanical stabilizer of the sarcolemma. In this study, we address the question: 'Does dystrophin-positive, regenerated muscle containing branched fibres also show an increased susceptibility to contraction-induced damage?' We produced a model of fibre branching by injecting dystrophin-positive extensor digitorum longus muscles with notexin. The regenerated muscle was examined at 21 days postinjection. Notexin-injected muscle contained 29% branched fibres and was not more susceptible to damage from mild eccentric contractions than contralateral saline-injected control muscle. Regenerated muscles also had greater mass, greater cross-sectional area and lower specific force than control muscles. We conclude that the number of branched fibres in this regenerated muscle is below the threshold needed to increase susceptibility to damage. However, it would serve as an ideal control for muscles of young mdx mice, allowing for clearer differentiation of the effects of dystrophin deficiency from the effects of fibre regeneration and morphology.
Collapse
Affiliation(s)
- S I Head
- * Faculty of Medicine, The University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | | | | | |
Collapse
|
35
|
Arnett AL, Konieczny P, Ramos JN, Hall J, Odom G, Yablonka-Reuveni Z, Chamberlain JR, Chamberlain JS. Adeno-associated viral (AAV) vectors do not efficiently target muscle satellite cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:S2329-0501(16)30105-X. [PMID: 25580445 PMCID: PMC4288464 DOI: 10.1038/mtm.2014.38] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Adeno-associated viral (AAV) vectors are becoming an important tool for gene therapy of numerous genetic and other disorders. Several recombinant AAV vectors (rAAV) have the ability to transduce striated muscles in a variety of animals following intramuscular and intravascular administration, and have attracted widespread interest for therapy of muscle disorders such as the muscular dystrophies. However, most studies have focused on the ability to transduce mature muscle cells, and have not examined the ability to target myogenic stem cells such as skeletal muscle satellite cells. Here we examined the relative ability of rAAV vectors derived from AAV6 to target myoblasts, myocytes, and myotubes in culture and satellite cells and myofibers in vivo. AAV vectors are able to transduce proliferating myoblasts in culture, albeit with reduced efficiency relative to postmitotic myocytes and myotubes. In contrast, quiescent satellite cells are refractory to transduction in adult mice. These results suggest that while muscle disorders characterized by myofiber regeneration can be slowed or halted by AAV transduction, little if any vector transduction can be obtained in myogenic stems cells that might other wise support ongoing muscle regeneration.
Collapse
Affiliation(s)
- Andrea Lh Arnett
- Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, USA ; Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA ; Molecular and Cellular Biology Program, University of Washington School of Medicine, Seattle, WA, USA
| | - Patryk Konieczny
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Julian N Ramos
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA ; Molecular and Cellular Biology Program, University of Washington School of Medicine, Seattle, WA, USA
| | - John Hall
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Guy Odom
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA, USA
| | - Joel R Chamberlain
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA ; Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
36
|
Corona BT, Garg K, Ward CL, McDaniel JS, Walters TJ, Rathbone CR. Autologous minced muscle grafts: a tissue engineering therapy for the volumetric loss of skeletal muscle. Am J Physiol Cell Physiol 2013; 305:C761-75. [DOI: 10.1152/ajpcell.00189.2013] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Volumetric muscle loss (VML) results in a large void deficient in the requisite materials for regeneration for which there is no definitive clinical standard of care. Autologous minced muscle grafts (MG), which contain the essential components for muscle regeneration, may embody an ideal tissue engineering therapy for VML. The purpose of this study was to determine if orthotopic transplantation of MG acutely after VML in the tibialis anterior muscle of male Lewis rats promotes functional tissue regeneration. Herein we report that over the first 16 wk postinjury, MG transplantation 1) promotes remarkable regeneration of innervated muscle fibers within the defect area (i.e., de novo muscle fiber regeneration); 2) reduced evidence of chronic injury in the remaining muscle mass compared with nonrepaired muscles following VML (i.e., transplantation attenuated chronically upregulated transforming growth factor-β1 gene expression and the presence of centrally located nuclei in 30% of fibers observed in nonrepaired muscles); and 3) significantly improves net torque production (i.e., ∼55% of the functional deficit in nonrepaired muscles was restored). Additionally, voluntary wheel running was shown to reduce the heightened accumulation of extracellular matrix deposition observed within the regenerated tissue of MG-repaired sedentary rats 8 wk postinjury (collagen 1% area: sedentary vs. runner, ∼41 vs. 30%), which may have been the result of an augmented inflammatory response [i.e., M1 (CCR7) and M2 (CD163) macrophage expression was significantly greater in runner than sedentary MG-repaired muscles 2 wk postinjury]. These findings support further exploration of autologous minced MGs for the treatment of VML.
Collapse
Affiliation(s)
- B. T. Corona
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - K. Garg
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - C. L. Ward
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - J. S. McDaniel
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - T. J. Walters
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - C. R. Rathbone
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| |
Collapse
|
37
|
Washington TA, Brown L, Smith DA, Davis G, Baum J, Bottje W. Monocarboxylate transporter expression at the onset of skeletal muscle regeneration. Physiol Rep 2013; 1:e00075. [PMID: 24303150 PMCID: PMC3831894 DOI: 10.1002/phy2.75] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/24/2013] [Accepted: 07/26/2013] [Indexed: 12/24/2022] Open
Abstract
The onset of skeletal muscle regeneration is characterized by proliferating myoblasts. Proliferating myoblasts have an increased energy demand and lactate exchange across the sarcolemma can be used to address this increased demand. Monocarboxylate transporters (MCTs) are involved in lactate transport across the sarcolemma and are known to be affected by various physiological stimuli. However, MCT expression at the onset of skeletal muscle regeneration has not been determined. The purpose of this study was to determine if skeletal muscle regeneration altered MCT expression in regenerating tibialis anterior (TA) muscle. Male C57/BL6 mice were randomly assigned to either a control (uninjured) or bupivacaine (injured) group. Three days post injection, the TA was extracted for determination of protein and gene expression. A 21% decrease in muscle mass to tibia length (2.4 ± 0.1 mg/mm vs. 1.9 ± 0.2 mg/mm, P < 0.02) was observed. IGF-1 and MyoD gene expression increased 5.0-fold (P < 0.05) and 3.5-fold (P < 0.05), respectively, 3 days post bupivacaine injection. MCT-1 protein was decreased 32% (P < 0.03); however, MCT-1 gene expression was not altered. There was no difference in MCT4 protein or gene expression. Lactate dehydrogenase (LDH)-A protein expression increased 71% (P < 0.0004). Protein levels of LDH-B and mitochondrial enzyme cytochrome C oxidase subunit decreased 3 days post bupivacaine injection. CD147 and PKC-θ protein increased 64% (P < 0.03) and 79% (P < 0.02), respectively. MCT1 but not MCT4 expression is altered at the onset of skeletal muscle regeneration possibly in an attempt to regulate lactate uptake and use by skeletal muscle cells.
Collapse
Affiliation(s)
- Tyrone A Washington
- Exercise Muscle Biology Laboratory, University of Arkansas Fayetteville, Arkansas, 72701 ; Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas Fayetteville, Arkansas, 72701
| | | | | | | | | | | |
Collapse
|
38
|
Lee ASJ, Anderson JE, Joya JE, Head SI, Pather N, Kee AJ, Gunning PW, Hardeman EC. Aged skeletal muscle retains the ability to fully regenerate functional architecture. BIOARCHITECTURE 2013; 3:25-37. [PMID: 23807088 PMCID: PMC3715540 DOI: 10.4161/bioa.24966] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
While the general understanding of muscle regenerative capacity is that it declines with increasing age due to impairments in the number of muscle progenitor cells and interaction with their niche, studies vary in their model of choice, indices of myogenic repair, muscle of interest and duration of studies. We focused on the net outcome of regeneration, functional architecture, compared across three models of acute muscle injury to test the hypothesis that satellite cells maintain their capacity for effective myogenic regeneration with age. Muscle regeneration in extensor digitorum longus muscle (EDL) of young (3 mo-old), old (22 mo-old) and senescent female mice (28 mo-old) was evaluated for architectural features, fiber number and central nucleation, weight, collagen and fat deposition. The 3 injury paradigms were: a myotoxin (notexin) which leaves the blood vessels and nerves intact, freezing (FI) that damages local muscle, nerve and blood vessels and denervation-devascularization (DD) which dissociates the nerves and blood vessels from the whole muscle. Histological analyses revealed successful architectural regeneration following notexin injury with negligible fibrosis and fully restored function, regardless of age. In comparison, the regenerative response to injuries that damaged the neurovascular supply (FI and DD) was less effective, but similar across the ages. The focus on net regenerative outcome demonstrated that old and senescent muscle has a robust capacity to regenerate functional architecture.
Collapse
Affiliation(s)
- Antonio S J Lee
- Neuromuscular and Regenerative Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Hunt LC, Gorman C, Kintakas C, McCulloch DR, Mackie EJ, White JD. Hyaluronan synthesis and myogenesis: a requirement for hyaluronan synthesis during myogenic differentiation independent of pericellular matrix formation. J Biol Chem 2013; 288:13006-21. [PMID: 23493399 PMCID: PMC3642344 DOI: 10.1074/jbc.m113.453209] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 03/12/2013] [Indexed: 11/06/2022] Open
Abstract
Exogenous hyaluronan is known to alter muscle precursor cell proliferation, migration, and differentiation, ultimately inhibiting myogenesis in vitro. The aim of the current study was to investigate the role of endogenous hyaluronan synthesis during myogenesis. In quantitative PCR studies, the genes responsible for synthesizing hyaluronan were found to be differentially regulated during muscle growth, repair, and pathology. Although all Has genes (Has1, Has2, and Has3) were differentially regulated in these models, only Has2 gene expression consistently associated with myogenic differentiation. During myogenic differentiation in vitro, Has2 was the most highly expressed of the synthases and increased after induction of differentiation. To test whether this association between Has2 expression and myogenesis relates to a role for Has2 in myoblast differentiation and fusion, C2C12 myoblasts were depleted of Has2 by siRNA and induced to differentiate. Depletion of Has2 inhibited differentiation and caused a loss of cell-associated hyaluronan and the hyaluronan-dependent pericellular matrix. The inhibition of differentiation caused by loss of hyaluronan was confirmed with the hyaluronan synthesis inhibitor 4-methylumbelliferone. In hyaluronan synthesis-blocked cultures, restoration of the pericellular matrix could be achieved through the addition of exogenous hyaluronan and the proteoglycan versican, but this was not sufficient to restore differentiation to control levels. These data indicate that intrinsic hyaluronan synthesis is necessary for myoblasts to differentiate and form syncytial muscle cells, but the hyaluronan-dependent pericellular matrix is not sufficient to support differentiation alone; additional hyaluronan-dependent cell functions that are yet unknown may be required for myogenic differentiation.
Collapse
Affiliation(s)
- Liam C. Hunt
- From the Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Chris Gorman
- From the Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria 3052, Australia
- the School of Veterinary Science, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Christopher Kintakas
- From the Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria 3052, Australia
- the School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia, and
| | - Daniel R. McCulloch
- the School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia, and
| | - Eleanor J. Mackie
- the School of Veterinary Science, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jason D. White
- From the Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria 3052, Australia
- the School of Veterinary Science, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
40
|
Seppanen EJ, Hodgson SS, Khosrotehrani K, Bou-Gharios G, Fisk NM. Fetal microchimeric cells in a fetus-treats-its-mother paradigm do not contribute to dystrophin production in serially parous mdx females. Stem Cells Dev 2012; 21:2809-16. [PMID: 22731493 DOI: 10.1089/scd.2012.0047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Throughout every pregnancy, genetically distinct fetal microchimeric stem/progenitor cells (FMCs) engraft in the mother, persist long after delivery, and may home to damaged maternal tissues. Phenotypically normal fetal lymphoid progenitors have been described to develop in immunodeficient mothers in a fetus-treats-its-mother paradigm. Since stem cells contribute to muscle repair, we assessed this paradigm in the mdx mouse model of Duchenne muscular dystrophy. mdx females were bred serially to either ROSAeGFP males or mdx males to obtain postpartum microchimeras that received either wild-type FMCs or dystrophin-deficient FMCs through serial gestations. To enhance regeneration, notexin was injected into the tibialis anterior of postpartum mice. FMCs were detected by qPCR at a higher frequency in injected compared to noninjected side muscle (P=0.02). However, the number of dystrophin-positive fibers was similar in mothers delivering wild-type compared to mdx pups. In addition, there was no correlation between FMC detection and percentage dystrophin, and no GFP+ve FMCs were identified that expressed dystrophin. In 10/11 animals, GFP+ve FMCs were detected by immunohistochemistry, of which 60% expressed CD45 with 96% outside the basal lamina defining myofiber contours. Finally we confirmed lack of FMC contribution to statellite cells in postpartum mdx females mated with Myf5-LacZ males. We conclude that the FMC contribution to regenerating muscles is insufficient to have a functional impact.
Collapse
Affiliation(s)
- Elke Jane Seppanen
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia.
| | | | | | | | | |
Collapse
|
41
|
Röder IV, Strack S, Reischl M, Dahley O, Khan MM, Kassel O, Zaccolo M, Rudolf R. Participation of myosin Va and Pka type I in the regeneration of neuromuscular junctions. PLoS One 2012; 7:e40860. [PMID: 22815846 PMCID: PMC3397957 DOI: 10.1371/journal.pone.0040860] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/14/2012] [Indexed: 11/21/2022] Open
Abstract
Background The unconventional motor protein, myosin Va, is crucial for the development of the mouse neuromuscular junction (NMJ) in the early postnatal phase. Furthermore, the cooperative action of protein kinase A (PKA) and myosin Va is essential to maintain the adult NMJ. We here assessed the involvement of myosin Va and PKA in NMJ recovery during muscle regeneration. Methodology/Principal Findings To address a putative role of myosin Va and PKA in the process of muscle regeneration, we used two experimental models the dystrophic mdx mouse and Notexin-induced muscle degeneration/regeneration. We found that in both systems myosin Va and PKA type I accumulate beneath the NMJs in a fiber maturation-dependent manner. Morphologically intact NMJs were found to express stable nicotinic acetylcholine receptors and to accumulate myosin Va and PKA type I in the subsynaptic region. Subsynaptic cAMP signaling was strongly altered in dystrophic muscle, particularly in fibers with severely subverted NMJ morphology. Conclusions/Significance Our data show a correlation between the subsynaptic accumulation of myosin Va and PKA type I on the one hand and NMJ regeneration status and morphology, AChR stability and specificity of subsynaptic cAMP handling on the other hand. This suggests an important role of myosin Va and PKA type I for the maturation of NMJs in regenerating muscle.
Collapse
Affiliation(s)
- Ira Verena Röder
- Institut für Toxikologie und Genetik, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Siegfried Strack
- Institut für Toxikologie und Genetik, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Markus Reischl
- Institut für Angewandte Informatik, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Oliver Dahley
- Institut für Toxikologie und Genetik, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Muzamil Majid Khan
- Institut für Toxikologie und Genetik, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Olivier Kassel
- Institut für Toxikologie und Genetik, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Manuela Zaccolo
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom
| | - Rüdiger Rudolf
- Institut für Toxikologie und Genetik, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
- Institut für Medizintechnologie, Universität Heidelberg und Hochschule Mannheim, Mannheim, Germany
- Institut für Molekular- und Zellbiologie, Hochschule Mannheim, Mannheim, Germany
- * E-mail:
| |
Collapse
|
42
|
S1P lyase in skeletal muscle regeneration and satellite cell activation: exposing the hidden lyase. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:167-75. [PMID: 22750505 DOI: 10.1016/j.bbalip.2012.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 06/18/2012] [Accepted: 06/20/2012] [Indexed: 01/12/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid whose actions are essential for many physiological processes including angiogenesis, lymphocyte trafficking and development. In addition, S1P serves as a muscle trophic factor that enables efficient muscle regeneration. This is due in part to S1P's ability to activate quiescent muscle stem cells called satellite cells (SCs) that are needed for muscle repair. However, the molecular mechanism by which S1P activates SCs has not been well understood. Further, strategies for harnessing S1P signaling to recruit SCs for therapeutic benefit have been lacking. S1P is irreversibly catabolized by S1P lyase (SPL), a highly conserved enzyme that catalyzes the cleavage of S1P at carbon bond C(2-3), resulting in formation of hexadecenal and ethanolamine-phosphate. SPL enhances apoptosis through substrate- and product-dependent events, thereby regulating cellular responses to chemotherapy, radiation and ischemia. SPL is undetectable in resting murine skeletal muscle. However, we recently found that SPL is dynamically upregulated in skeletal muscle after injury. SPL upregulation occurred in the context of a tightly orchestrated genetic program that resulted in a transient S1P signal in response to muscle injury. S1P activated quiescent SCs via a sphingosine-1-phosphate receptor 2 (S1P2)/signal transducer and activator of transcription 3 (STAT3)-dependent pathway, thereby facilitating skeletal muscle regeneration. Mdx mice, which serve as a model for muscular dystrophy (MD), exhibited skeletal muscle SPL upregulation and S1P deficiency. Pharmacological SPL inhibition raised skeletal muscle S1P levels, enhanced SC recruitment and improved mdx skeletal muscle regeneration. These findings reveal how S1P can activate SCs and indicate that SPL suppression may provide a therapeutic strategy for myopathies. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
|
43
|
Murphy KT, Allen AM, Chee A, Naim T, Lynch GS. Disruption of muscle renin-angiotensin system in AT1a-/- mice enhances muscle function despite reducing muscle mass but compromises repair after injury. Am J Physiol Regul Integr Comp Physiol 2012; 303:R321-31. [PMID: 22673782 DOI: 10.1152/ajpregu.00007.2012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of the renin-angiotensin system (RAS) in vasoregulation is well established, but a localized RAS exists in multiple tissues and exerts diverse functions including autonomic control and thermogenesis. The role of the RAS in the maintenance and function of skeletal muscle is not well understood, especially the role of angiotensin peptides, which appear to contribute to muscle atrophy. We tested the hypothesis that mice lacking the angiotensin type 1A receptor (AT(1A)(-/-)) would exhibit enhanced whole body and skeletal muscle function and improved regeneration after severe injury. Despite 18- to 20-wk-old AT(1A)(-/-) mice exhibiting reduced muscle mass compared with controls (P < 0.05), the tibialis anterior (TA) muscles produced a 25% higher maximum specific (normalized) force (P < 0.05). Average fiber cross-sectional area (CSA) and fiber oxidative capacity was not different between groups, but TA muscles from AT(1A)(-/-) mice had a reduced number of muscle fibers as well as a higher proportion of type IIx/b fibers and a lower proportion of type IIa fibers (P < 0.05). Measures of whole body function (grip strength, rotarod performance, locomotor activity) were all improved in AT(1A)(-/-) mice (P < 0.05). Surprisingly, the recovery of muscle mass and fiber CSA following myotoxic injury was impaired in AT(1A)(-/-) mice, in part by impaired myoblast fusion, prolonged collagen infiltration and inflammation, and delayed expression of myogenic regulatory factors. The findings support the therapeutic potential of RAS inhibition for enhancing whole body and skeletal muscle function, but they also reveal the importance of RAS signaling in the maintenance of muscle mass and for normal fiber repair after injury.
Collapse
Affiliation(s)
- Kate T Murphy
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
44
|
Kao PH, Chiou YL, Chen YJ, Lin SR, Chang LS. Guanidination of notexin promotes its phospholipase A(2) activity-independent fusogenicity on vesicles with lipid-supplied negative curvature. Toxicon 2011; 59:47-58. [PMID: 22030836 DOI: 10.1016/j.toxicon.2011.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 09/27/2011] [Accepted: 10/11/2011] [Indexed: 11/30/2022]
Abstract
To address the requirement of phospholipase A(2) (PLA(2)) activity in membrane fusion events and membrane perturbation activity of notexin and guanidinated notexin (Gu-notexin), the present study was conducted. Notexin and Gu-notexin did not show PLA(2) activity after the removal of Ca(2+) with EDTA. Metal-free notexin and Gu-notexin were found to induce membrane leakage and fusion of phospholipid vesicles. Fusogenic activity of native and modified notexin correlated positively with their membrane-damaging activity underlying the deprivation of PLA(2) activity. Compared with Ca(2+)-bound Gu-notexin, fusogenicity of metal-free Gu-notexin was notably increased by incorporation of cholesterol, cholesterol sulfate, phosphatidylethanolamine, α-tocopherol and phosphatidic acid that supplied negative curvature into phospholipid bilayer. The ability of Gu-notexin to induce membrane fusion of vesicles with lipid-supplied negative curvature was higher than that of notexin regardless of the absence or presence of Ca(2+). Consistently, metal-free Gu-notexin markedly induced membrane fusion of red blood cells (RBCs) compared with metal-free notexin, and fusion activity of metal-free Gu-notexin on cholesterol-depleted RBCs notably reduced. Compared with notexin, Gu-notexin highly induced uptake of calcein-loaded phosphatidylcholine (PC)/cholesterol and PC/cholesterol sulfate vesicles by K562 cells in the presence of EDTA. Taken together, our data suggest that notexin and Gu-notexin could induce vesicle leakage and fusion via a PLA(2) activity-independent mechanism, and guanidination promotes PLA(2) activity-independent fusogenicity of notexin on vesicles with lipid-supplied negative curvature.
Collapse
Affiliation(s)
- Pei-Hsiu Kao
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | | | | | | | | |
Collapse
|
45
|
Long MA, Rossi FMV. Targeted cell fusion facilitates stable heterokaryon generation in vitro and in vivo. PLoS One 2011; 6:e26381. [PMID: 22039476 PMCID: PMC3200330 DOI: 10.1371/journal.pone.0026381] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 09/26/2011] [Indexed: 11/19/2022] Open
Abstract
Induced cell fusion has enabled several important discoveries, including the phenomenon of nuclear reprogramming and may yet be applied as a novel therapy for degenerative diseases. However, existing fusogens lack the efficiency required to enable investigation of the epigenetic modifications underlying nuclear reprogramming and the specificity required for clinical application. Here we present a chimeric measles hemagglutinin, Hα7, which specifically and efficiently mediates the fusion of diverse cell types with skeletal muscle both in vitro and in vivo. When compared directly to polyethylene glycol, Hα7 consistently generated a substantial increase in heterokaryon yield and exhibited insignificant levels of toxicity. Moreover, this increased fusion efficiency enabled detection of chromatin modifications associated with nuclear reprogramming following Hα7-mediated fusion of human fibroblasts and mouse myotubes. Finally, Hα7 was also capable of increasing the contribution of transplanted fibroblasts to skeletal muscle repair in vivo, suggesting that this strategy could be used for therapeutic gene delivery.
Collapse
Affiliation(s)
- Michael A. Long
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fabio M. V. Rossi
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
46
|
McNeill Ingham SJ, Pochini ADC, de Oliveira DA, Garcia Lisboa BC, Beutel A, Valero-Lapchik VB, Ferreira AM, Abdalla RJ, Cohen M, Han SW. Bupivacaine Injection Leads to Muscle Force Reduction and Histologic Changes in a Murine Model. PM R 2011; 3:1106-9. [DOI: 10.1016/j.pmrj.2011.05.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 05/11/2011] [Accepted: 05/22/2011] [Indexed: 12/13/2022]
|
47
|
Church JE, Gehrig SM, Chee A, Naim T, Trieu J, McConell GK, Lynch GS. Early functional muscle regeneration after myotoxic injury in mice is unaffected by nNOS absence. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1358-66. [PMID: 21849632 DOI: 10.1152/ajpregu.00096.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) is an important signaling molecule produced in skeletal muscle primarily via the neuronal subtype of NO synthase (NOS1, or nNOS). While many studies have reported NO production to be important in muscle regeneration, none have examined the contribution of nNOS-derived NO to functional muscle regeneration (i.e., restoration of the muscle's ability to produce force) after acute myotoxic injury. In the present study, we tested the hypothesis that genetic deletion of nNOS would impair functional muscle regeneration after myotoxic injury in nNOS(-/-) mice. We found that nNOS(-/-) mice had lower body mass, lower muscle mass, and smaller myofiber cross-sectional area and that their tibialis anterior (TA) muscles produced lower absolute tetanic forces than those of wild-type littermate controls but that normalized or specific force was identical between the strains. In addition, muscles from nNOS(-/-) mice were more resistant to fatigue than those of wild-type littermates (P < 0.05). To determine whether deletion of nNOS affected muscle regeneration, TA muscles from nNOS(-/-) mice and wild-type littermates were injected with the myotoxin notexin to cause complete fiber degeneration, and muscle structure and function were assessed at 7 and 10 days postinjury. Myofiber cross-sectional area was lower in regenerating nNOS(-/-) mice than wild-type controls at 7 and 10 days postinjury; however, contrary to our original hypothesis, no difference in force-producing capacity of the TA muscle was evident between the two groups at either time point. Our findings reveal that nNOS is not essential for functional muscle regeneration after acute myotoxic damage.
Collapse
Affiliation(s)
- Jarrod E Church
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
48
|
Orengo JP, Ward AJ, Cooper TA. Alternative splicing dysregulation secondary to skeletal muscle regeneration. Ann Neurol 2011; 69:681-90. [PMID: 21400563 DOI: 10.1002/ana.22278] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 09/06/2010] [Accepted: 09/17/2010] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Dysregulation of alternative splicing has become a molecular hallmark of myotonic dystrophy type 1 (DM1), in which neonatal splice variants are expressed in adult skeletal muscle. Splicing dysregulation is induced by RNA containing expanded CUG repeats expressed from the expanded mutant allele by sequestration of muscleblindlike 1 (MBNL1) protein within nuclear RNA foci and increased CUGBP, ELAV-like family member 1 (CELF1) protein levels. Dysregulated splicing has also been identified in other neuromuscular disorders, suggesting either that diseases with different molecular causes share a common pathogenic mechanism or that dysregulated splicing can also be a common secondary consequence of muscle degeneration and regeneration. METHODS In this study, we examined regulation of alternative splicing in 4 different mouse models of muscular dystrophy, including DM1, limb-girdle muscular dystrophy, congenital merosin-deficient muscular dystrophy, and Duchenne muscular dystrophy, and 2 myotoxin (cardiotoxin and notexin) muscle injury models. RESULTS We show that DM1-like alternative splicing dysregulation and altered expression of MBNL1 and CELF1 occur in non-DM1 mouse models of muscular dystrophy and muscle injury, most likely due to recapitulation of neonatal splicing patterns in regenerating fibers. In contrast, CELF1 was elevated in nuclei of mature myofibers of the DM1 model, consistent with a primary effect of pathogenic RNA expression. INTERPRETATION Splicing dysregulation in DM1 is a primary effect of RNA containing expanded CUG repeats. However, we conclude that splicing changes can also be observed secondary to muscle regeneration, and this possibility must be taken into account when evaluating cause-effect relationships between dysregulated splicing and disease processes.
Collapse
Affiliation(s)
- James P Orengo
- Department of Pathology and Immunology and, Baylor College of Medicine Houston, TX 77030, USA
| | | | | |
Collapse
|
49
|
Gehrig SM, Lynch GS. Emerging drugs for treating skeletal muscle injury and promoting muscle repair. Expert Opin Emerg Drugs 2011; 16:163-82. [DOI: 10.1517/14728214.2010.524743] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
50
|
Kao PH, Chiou YL, Lin SR, Chang LS. Guanidination of notexin alters its membrane-damaging activity in response to sphingomyelin and cholesterol. J Biosci 2010; 35:583-93. [DOI: 10.1007/s12038-010-0067-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|