1
|
Tsitkanou S, Morena da Silva F, Cabrera AR, Schrems ER, Muhyudin R, Koopmans PJ, Khadgi S, Lim S, Delfinis LJ, Washington TA, Murach KA, Perry CGR, Greene NP. Mitochondrial antioxidant SkQ1 attenuates C26 cancer-induced muscle wasting in males and improves muscle contractility in female tumor-bearing mice. Am J Physiol Cell Physiol 2024; 327:C1308-C1322. [PMID: 39344417 PMCID: PMC11559642 DOI: 10.1152/ajpcell.00497.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
Mitochondrial dysfunction is a hallmark of cancer cachexia (CC). Mitochondrial reactive oxygen species (ROS) are elevated in muscle shortly after tumor onset. Targeting mitochondrial ROS may be a viable option to prevent CC. The aim of this study was to evaluate the efficacy of a mitochondria-targeted antioxidant, SkQ1, to mitigate CC in both biological sexes. Male and female Balb/c mice were injected bilaterally with colon 26 adenocarcinoma (C26) cells (total 1 × 106 cells) or PBS (equal volume control). SkQ1 was dissolved in drinking water (∼250 nmol/kg body wt/day) and administered to mice beginning 7 days following tumor induction, whereas control groups consumed normal drinking water. In vivo muscle contractility of dorsiflexors, deuterium oxide-based protein synthesis, mitochondrial respiration and mRNA content of mitochondrial, protein turnover, and calcium channel-related markers were assessed at endpoint (25 days following tumor induction). Two-way ANOVAs, followed by Tukey's post hoc test when interactions were significant (P ≤ 0.05), were performed. SkQ1 attenuated cancer-induced atrophy, promoted protein synthesis, and abated Redd1 and Atrogin induction in gastrocnemius of C26 male mice. In female mice, SkQ1 decreased muscle mass and increased catabolic signaling in the plantaris of tumor-bearing mice, as well as reduced mitochondrial oxygen consumption, regardless of tumor. However, in females, SkQ1 enhanced muscle contractility of the dorsiflexors with concurrent induction of Ryr1, Serca1, and Serca2a in TA. In conclusion, the mitochondria-targeted antioxidant SkQ1 may attenuate CC-induced muscle loss in males, while improving muscle contractile function in tumor-bearing female mice, suggesting sexual dimorphism in the effects of this mitochondrial therapy in CC.NEW & NOTEWORTHY Herein, we assess the efficacy of the mitochondria-targeted antioxidant SkQ1 to mitigate cancer cachexia (CC) in both biological sexes. We demonstrate that SkQ1 administration attenuates muscle wasting induced by C26 tumors in male, but not female, mice. Conversely, we identify that in females, SkQ1 improves muscle contractility. These phenotypic adaptations to SkQ1 are aligned with respective responses in muscle protein synthesis, mitochondrial respiration, and mRNA content of protein turnover, as well as mitochondrial and calcium handling-related markers.
Collapse
Affiliation(s)
- Stavroula Tsitkanou
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Francielly Morena da Silva
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ana Regina Cabrera
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ruqaiza Muhyudin
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Pieter J Koopmans
- Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, United States
| | - Sabin Khadgi
- Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Seongkyun Lim
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Luca J Delfinis
- Muscle Health Research Centre and the School of Kinesiology & Health Sciences, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Kevin A Murach
- Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, United States
| | - Christopher G R Perry
- Muscle Health Research Centre and the School of Kinesiology & Health Sciences, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Nicholas P Greene
- Cachexia Research Laboratory, Department of Health, Human Performance and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
2
|
Langston PK, Mathis D. Immunological regulation of skeletal muscle adaptation to exercise. Cell Metab 2024; 36:1175-1183. [PMID: 38670108 DOI: 10.1016/j.cmet.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
Exercise has long been acknowledged for its powerful disease-preventing, health-promoting effects. However, the cellular and molecular mechanisms responsible for the beneficial effects of exercise are not fully understood. Inflammation is a component of the stress response to exercise. Recent work has revealed that such inflammation is not merely a symptom of exertion; rather, it is a key regulator of exercise adaptations, particularly in skeletal muscle. The purpose of this piece is to provide a conceptual framework that we hope will integrate exercise immunology with exercise physiology, muscle biology, and cellular immunology. We start with an overview of early studies in the field of exercise immunology, followed by an exploration of the importance of stromal cells and immunocytes in the maintenance of muscle homeostasis based on studies of experimental muscle injury. Subsequently, we discuss recent advances in our understanding of the functions and physiological relevance of the immune system in exercised muscle. Finally, we highlight a potential immunological basis for the benefits of exercise in musculoskeletal diseases and aging.
Collapse
Affiliation(s)
- P Kent Langston
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Koopmans PJ, Williams‐Frey TD, Zwetsloot KA. Stuart has got the PoWeR! Skeletal muscle adaptations to a novel heavy progressive weighted wheel running exercise model in C57BL/6 mice. Exp Physiol 2024; 109:271-282. [PMID: 37974360 PMCID: PMC10988744 DOI: 10.1113/ep091494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
Murine exercise models are developed to study the molecular and cellular mechanisms regulating muscle mass. A progressive weighted wheel running model, named 'PoWeR', was previously developed to serve as a more translatable alternative to involuntary resistance-type exercise models in rodents, such as synergist ablation. However, mice still run great distances despite the added resistance as evidenced by a large glycolytic-to-oxidative shift in muscle fibre type. Thus, PoWeR reflects a blended resistance/endurance model. In an attempt to bias PoWeR further towards resistance-type exercise, we developed a novel heavy PoWeR model (hPoWeR) utilizing higher wheel loads (max of 12.5 g vs 6 g). Adult male C57BL/6 mice voluntarily performed an 8-week progressive loading protocol (PoWeR or hPoWeR). Running distance peaked at ∼5-6 km day-1 in both treatments and was maintained by PoWeR mice, but declined in the hPoWeR mice as load increased beyond 7.5 g. Peak isometric force of the gastrocnemius-soleus-plantaris complex tended to increase in wheel running treatments. Soleus mass increased by 19% and 24% in PoWeR and hPoWeR treatments, respectively, and plantaris fibre cross-sectional area was greater in hPoWeR, compared to PoWeR. There were fewer glycolytic and more oxidative fibres in the soleus and plantaris muscles in the PoWeR treatment, but not hPoWeR. Collectively, these data suggest hPoWeR may modestly alter skeletal muscle supporting the aim of better reflecting typical resistance training adaptations, in line with decreased running volume and exposure to higher resistance. Regardless, PoWeR remains an effective hypertrophic concurrent training model in mice.
Collapse
Affiliation(s)
- Pieter J. Koopmans
- Integrative Muscle Physiology LaboratoryAppalachian State UniversityBooneNorth CarolinaUSA
- Department of Public Health and Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
- Cell and Molecular Biology ProgramUniversity of ArkansasFayettevilleArkansasUSA
| | - Therin D. Williams‐Frey
- Integrative Muscle Physiology LaboratoryAppalachian State UniversityBooneNorth CarolinaUSA
- Department of BiologyAppalachian State UniversityBooneNorth CarolinaUSA
| | - Kevin A. Zwetsloot
- Integrative Muscle Physiology LaboratoryAppalachian State UniversityBooneNorth CarolinaUSA
- Department of Public Health and Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
- Department of BiologyAppalachian State UniversityBooneNorth CarolinaUSA
| |
Collapse
|
4
|
Schifino AG, Raymond‐Pope CJ, Heo J, McFaline‐Figueroa J, Call JA, Greising SM. Resistance wheel running improves contractile strength, but not metabolic capacity, in a murine model of volumetric muscle loss injury. Exp Physiol 2023; 108:1282-1294. [PMID: 37526646 PMCID: PMC10543535 DOI: 10.1113/ep091284] [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: 05/06/2023] [Accepted: 07/31/2023] [Indexed: 08/02/2023]
Abstract
The primary objective of this study was to determine if low- or high-resistance voluntary wheel running leads to functional improvements in muscle strength (i.e., isometric and isokinetic torque) and metabolic function (i.e., permeabilized fibre bundle mitochondrial respiration) after a volumetric muscle loss (VML) injury. C57BL/6J mice were randomized into one of four experimental groups at age 12 weeks: uninjured control, VML untreated (VML), low-resistance wheel running (VML-LR) and high-resistance wheel running (VML-HR). All mice, excluding the uninjured, were subject to a unilateral VML injury to the plantar flexor muscles and wheel running began 3 days post-VML. At 8 weeks post-VML, peak isometric torque was greater in uninjured compared to all VML-injured groups, but both VML-LR and VML-HR had greater (∼32%) peak isometric torque compared to VML. All VML-injured groups had less isokinetic torque compared to uninjured, and there was no statistical difference among VML, VML-LR and VML-HR. No differences in cumulative running distance were observed between VML-LR and VML-HR groups. Because adaptations in VML-HR peak isometric torque were attributed to greater gastrocnemius muscle mass, atrophy- and hypertrophy-related protein content and post-translational modifications were explored via immunoblot; however, results were inconclusive. Permeabilized fibre bundle mitochondrial oxygen consumption was 22% greater in uninjured compared to VML, but there was no statistical difference among VML, VML-LR and VML-HR. Furthermore, neither wheel running group demonstrated a change in the relative protein content of the mitochondrial biogenesis transcription factor, peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α). These results indicate that resistance wheel running alone only has modest benefits in the VML-injured muscle. NEW FINDINGS: What is the central question of the study? Does initiation of a resistance wheel running regimen following volumetric muscle loss (VML) improve the functional capacity of skeletal muscle? What is the main finding and its importance? Resistance wheel running led to greater muscle mass and strength in mice with a VML injury but did not result in a full recovery. Neither low- nor high-resistance wheel running was associated with a change in permeabilized muscle fibre respiration despite runners having greater whole-body treadmill endurance capacity, suggesting resilience to metabolic adaptations in VML-injured muscle. Resistance wheel running may be a suitable adjuvant rehabilitation strategy, but alone does not fully mitigate VML pathology.
Collapse
Affiliation(s)
| | | | - Junwon Heo
- Department of Physiology and PharmacologyUniversity of GeorgiaAthensGAUSA
| | | | - Jarrod A. Call
- Department of Physiology and PharmacologyUniversity of GeorgiaAthensGAUSA
- Regenerative Bioscience CenterUniversity of GeorgiaAthensGAUSA
| | | |
Collapse
|
5
|
Cabrera AR, Deaver JW, Lim S, Morena da Silva F, Schrems ER, Saling LW, Tsitkanou S, Rosa-Caldwell ME, Wiggs MP, Washington TA, Greene NP. Females display relatively preserved muscle quality compared with males during the onset and early stages of C26-induced cancer cachexia. J Appl Physiol (1985) 2023; 135:655-672. [PMID: 37535708 PMCID: PMC10642509 DOI: 10.1152/japplphysiol.00196.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/05/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023] Open
Abstract
Cancer cachexia is clinically defined by involuntary weight loss >5% in <6 mo, primarily affecting skeletal muscle. Here, we aimed to identify sex differences in the onset of colorectal cancer cachexia with specific consideration to skeletal muscle contractile and metabolic functions. Eight-weeks old BALB/c mice (69 males, 59 females) received subcutaneous C26 allografts or PBS vehicle. Tumors were developed for 10-, 15-, 20-, or 25 days. Muscles and organs were collected, in vivo muscle contractility, protein synthesis rate, mitochondrial function, and protein turnover markers were assessed. One-way ANOVA within sex and trend analysis between sexes were performed, P < 0.05. Gastrocnemius and tibialis anterior (TA) muscles became atrophic in male mice at 25 days, whereas female mice exhibited no significant differences in muscle weights at endpoints despite presenting hallmarks of cancer cachexia (fat loss, hepatosplenomegaly). We observed lowered muscle contractility and protein synthesis concomitantly to muscle mass decay in males, with higher proteolytic markers in muscles of both sexes. mRNA of Opa1 was lower in TA, whereas Bnip3 was higher in gastrocnemius after 25 days in male mice, with no significant effect in female mice. Our data suggest relative protections to skeletal muscle in females compared with males despite other canonical signs of cancer cachexia and increased protein degradation markers; suggesting we should place onus upon nonmuscle tissues during early stages of cancer cachexia in females. We noted potential protective mechanisms relating to skeletal muscle contractile and mitochondrial functions. Our findings underline possible heterogeneity in onset of cancer cachexia between biological sexes, suggesting the need for sex-specific approaches to treat cancer cachexia.NEW & NOTEWORTHY Our study demonstrates biological-sex differences in phenotypic characteristics of cancer cachexia between male and female mice, whereby females display many common characteristics of cachexia (gonadal fat loss and hepatosplenomegaly), protein synthesis markers alterations, and common catabolic markers in skeletal muscle despite relatively preserved muscle mass in early-stage cachexia compared with males. Mechanisms of cancer cachexia appear to differ between sexes. Data suggest need to place onus of early cancer cachexia detection and treatment on nonmuscle tissues in females.
Collapse
Affiliation(s)
- Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - J William Deaver
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Seongkyun Lim
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Francielly Morena da Silva
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Landen W Saling
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Stavroula Tsitkanou
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Megan E Rosa-Caldwell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States
| | - Michael P Wiggs
- Department of Health, Human Performance and Recreation, Baylor University, Waco, Texas, United States
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
6
|
Chan KM, Griffith JL, Pacheco YC, Allen KD. Wheel Running Exacerbates Joint Damage after Meniscal Injury in Mice, but Does Not Alter Gait or Physical Activity Levels. Med Sci Sports Exerc 2023; 55:1564-1576. [PMID: 37144624 PMCID: PMC10524358 DOI: 10.1249/mss.0000000000003198] [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] [Indexed: 05/06/2023]
Abstract
PURPOSE Exercise and physical activity are recommended to reduce pain and improve joint function in patients with knee osteoarthritis (OA). However, exercise has dose effects, with excessive exercise accelerating OA development and sedentary behaviors also promoting OA development. Prior work evaluating exercise in preclinical models has typically used prescribed exercise regimens; however, in-cage voluntary wheel running creates opportunities to evaluate how OA progression affects self-selected physical activity levels. This study aimed to evaluate how voluntary wheel running after a surgically induced meniscal injury affects gait characteristics and joint remodeling in C57Bl/6 mice. We hypothesize that injured mice will reduce physical activity levels as OA develops after meniscal injury and will engage in wheel running to a lesser extent than the uninjured animals. METHODS Seventy-two C57Bl/6 mice were divided into experimental groups based on sex, lifestyle (physically active vs sedentary), and surgery (meniscal injury or sham control). Voluntary wheel running data were continuously collected throughout the study, and gait data were collected at 3, 7, 11, and 15 wk after surgery. At end point, joints were processed for histology to assess cartilage damage. RESULTS After meniscal injury, physically active mice showed more severe joint damage relative to sedentary mice. Nevertheless, injured mice engaged in voluntary wheel running at the same rates and distances as mice with sham surgery. In addition, physically active mice and sedentary mice both developed a limp as meniscal injury progressed, yet exercise did not further exacerbate gait changes in the physically active mice, despite worsened joint damage. CONCLUSIONS Taken together, these data indicate a discordance between structural joint damage and joint function. Although wheel running after meniscal injury did worsen OA-related joint damage, physical activity did not necessarily inhibit or worsen OA-related joint dysfunction or pain in mice.
Collapse
Affiliation(s)
- Kiara M. Chan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
- Department of Kinesiology, Indiana University, Bloomington, IN
| | - Jacob L. Griffith
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Yan Carlos Pacheco
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene
| | - Kyle D. Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
- Department of Orthopedics and Sports Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
7
|
Hanna BS, Yaghi OK, Langston PK, Mathis D. The potential for Treg-enhancing therapies in tissue, in particular skeletal muscle, regeneration. Clin Exp Immunol 2023; 211:138-148. [PMID: 35972909 PMCID: PMC10019136 DOI: 10.1093/cei/uxac076] [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: 05/11/2022] [Revised: 06/29/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Foxp3+CD4+ regulatory T cells (Tregs) are famous for their role in maintaining immunological tolerance. With their distinct transcriptomes, growth-factor dependencies and T-cell receptor (TCR) repertoires, Tregs in nonlymphoid tissues, termed "tissue-Tregs," also perform a variety of functions to help assure tissue homeostasis. For example, they are important for tissue repair and regeneration after various types of injury, both acute and chronic. They exert this influence by controlling both the inflammatory tenor and the dynamics of the parenchymal progenitor-cell pool in injured tissues, thereby promoting efficient repair and limiting fibrosis. Thus, tissue-Tregs are seemingly attractive targets for immunotherapy in the context of tissue regeneration, offering several advantages over existing therapies. Using skeletal muscle as a model system, we discuss the existing literature on Tregs' role in tissue regeneration in acute and chronic injuries, and various approaches for their therapeutic modulation in such contexts, including exercise as a natural Treg modulator.
Collapse
Affiliation(s)
- Bola S Hanna
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - Omar K Yaghi
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - P Kent Langston
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| |
Collapse
|
8
|
Su Y, Song Y. The new challenge of “exercise + X″ therapy for Duchenne muscular dystrophy—Individualized identification of exercise tolerance and precise implementation of exercise intervention. Front Physiol 2022; 13:947749. [PMID: 35991169 PMCID: PMC9389311 DOI: 10.3389/fphys.2022.947749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/07/2022] [Indexed: 12/05/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive fatal muscular disease. Gene therapy, cell therapy, and drug therapy are currently the most widely used treatments for DMD. However, many experiments on animals and humans suggested that appropriate exercise could improve the effectiveness of such precision medicine treatment, thereby improving patient’s muscle quality and function. Due to the striated muscle damage of DMD individuals, there are still many debates about whether DMD animals or patients can exercise, how to exercise, when to exercise best, and how to exercise effectively. The purpose of this review is to summarize and investigate the scientific basis and efficacy of exercise as an adjuvant therapy for DMD gene therapy, cell therapy and drug therapy, as well as to present the theoretical framework and optional strategies of “exercise + X″″ combination therapy.
Collapse
Affiliation(s)
- Yuhui Su
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
- Institute of Physical Education, Jilin Normal University, Siping, China
| | - Yafeng Song
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
- *Correspondence: Yafeng Song,
| |
Collapse
|
9
|
Dungan CM, Brightwell CR, Wen Y, Zdunek CJ, Latham CM, Thomas NT, Zagzoog AM, Brightwell BD, Nolt GL, Keeble AR, Watowich SJ, Murach KA, Fry CS. Muscle-Specific Cellular and Molecular Adaptations to Late-Life Voluntary Concurrent Exercise. FUNCTION 2022; 3:zqac027. [PMID: 35774589 PMCID: PMC9233305 DOI: 10.1093/function/zqac027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/06/2022] [Accepted: 05/08/2022] [Indexed: 01/07/2023] Open
Abstract
Murine exercise models can provide information on factors that influence muscle adaptability with aging, but few translatable solutions exist. Progressive weighted wheel running (PoWeR) is a simple, voluntary, low-cost, high-volume endurance/resistance exercise approach for training young mice. In the current investigation, aged mice (22-mo-old) underwent a modified version of PoWeR for 8 wk. Muscle functional, cellular, biochemical, transcriptional, and myonuclear DNA methylation analyses provide an encompassing picture of how muscle from aged mice responds to high-volume combined training. Mice run 6-8 km/d, and relative to sedentary mice, PoWeR increases plantarflexor muscle strength. The oxidative soleus of aged mice responds to PoWeR similarly to young mice in every parameter measured in previous work; this includes muscle mass, glycolytic-to-oxidative fiber type transitioning, fiber size, satellite cell frequency, and myonuclear number. The oxidative/glycolytic plantaris adapts according to fiber type, but with modest overall changes in muscle mass. Capillarity increases markedly with PoWeR in both muscles, which may be permissive for adaptability in advanced age. Comparison to published PoWeR RNA-sequencing data in young mice identified conserved regulators of adaptability across age and muscles; this includes Aldh1l1 which associates with muscle vasculature. Agrn and Samd1 gene expression is upregulated after PoWeR simultaneous with a hypomethylated promoter CpG in myonuclear DNA, which could have implications for innervation and capillarization. A promoter CpG in Rbm10 is hypomethylated by late-life exercise in myonuclei, consistent with findings in muscle tissue. PoWeR and the data herein are a resource for uncovering cellular and molecular regulators of muscle adaptation with aging.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Physical Therapy, University of Kentucky, Lexington 40536, KY, USA
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | - Camille R Brightwell
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Yuan Wen
- Department of Physical Therapy, University of Kentucky, Lexington 40536, KY, USA
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | | | - Christine M Latham
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Alyaa M Zagzoog
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Benjamin D Brightwell
- Kinesiology and Health Promotion Graduate Program, University of Kentucky, Lexington 40536, KY, USA
| | - Georgia L Nolt
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | - Alexander R Keeble
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Stanley J Watowich
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston 77555, TX, USA
| | - Kevin A Murach
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville 72701, AR, USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville 72701, AR, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| |
Collapse
|
10
|
Lim S, Deaver JW, Rosa-Caldwell ME, Lee DE, Morena da Silva F, Cabrera AR, Schrems ER, Saling LW, Washington TA, Fluckey JD, Greene NP. Muscle miR-16 deletion results in impaired insulin sensitivity and contractile function in a sex-dependent manner. Am J Physiol Endocrinol Metab 2022; 322:E278-E292. [PMID: 35068192 PMCID: PMC8897019 DOI: 10.1152/ajpendo.00333.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/22/2022]
Abstract
microRNAs (miRs) are linked to various human diseases including type 2 diabetes mellitus (T2DM) and emerging evidence suggests that miRs may serve as potential therapeutic targets. Lower miR-16 content is consistent across different models of T2DM; however, the role of miR-16 in muscle metabolic health is still elusive. Therefore, the purpose of this study was to investigate how deletion of miR-16 in mice affects skeletal muscle metabolic health and contractile function in both sexes. This study was conducted using both 1) in vitro and 2) in vivo experiments. In in vitro experiments, we used C2C12 myoblasts to test if inhibition or overexpression of miR-16 affected insulin-mediated glucose handling. In in vivo experiments, we generated muscle-specific miR-16 knockout (KO) mice fed a high-fat diet (HFD) to assess how miR-16 content impacts metabolic and contractile properties including glucose tolerance, insulin sensitivity, muscle contractile function, protein anabolism, and mitochondrial network health. In in vitro experiments, although inhibition of miR-16 induced impaired insulin signaling (P = 0.002) and glucose uptake (P = 0.014), overexpression of miR-16 did not attenuate lipid overload-induced insulin resistance using the diacylglycerol analog 1-oleoyl-2-acetyl-sn-glycerol. In in vivo experiments, miR-16 deletion induced both impaired muscle contractility (P = 0.031-0.033), and mitochondrial network health (P = 0.008-0.018) in both sexes. However, although males specifically exhibited impaired insulin sensitivity following miR-16 deletion (P = 0.030), female KO mice showed pronounced glucose intolerance (P = 0.046), corresponding with lower muscle weights (P = 0.015), and protein hyperanabolism (P = 0.023). Our findings suggest distinct sex differences in muscle adaptation in response to miR-16 deletion and miR-16 may serve as a key regulator for metabolic dysregulation in T2DM.NEW & NOTEWORTHY We set to investigate the role of miR-16 in skeletal muscle during diet-induced insulin resistance. Our data provide novel evidence that the lack of miR-16 induced multiple aberrations in insulin sensitivity, muscle contractility, mitochondrial network health, and protein turnover in a sex-dependent manner. Interestingly, miR-16 deletion leads to insulin resistance in males and exacerbated glucose intolerance in females, suggesting different mechanisms of metabolic dysregulation with a lack of miR-16 between sexes.
Collapse
Affiliation(s)
- Seongkyun Lim
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - J William Deaver
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Megan E Rosa-Caldwell
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - David E Lee
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Francielly Morena da Silva
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Landen W Saling
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - James D Fluckey
- Muscle Biology Laboratory, Department of Health and Kinesiology, Texas A&M University, College Station, Texas
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
11
|
Lim S, Deaver JW, Rosa-Caldwell ME, Haynie WS, Morena da Silva F, Cabrera AR, Schrems ER, Saling LW, Jansen LT, Dunlap KR, Wiggs MP, Washington TA, Greene NP. Development of metabolic and contractile alterations in development of cancer cachexia in female tumor-bearing mice. J Appl Physiol (1985) 2022; 132:58-72. [PMID: 34762526 PMCID: PMC8747017 DOI: 10.1152/japplphysiol.00660.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 01/03/2023] Open
Abstract
Cancer cachexia (CC) results in impaired muscle function and quality of life and is the primary cause of death for ∼20%-30% of patients with cancer. We demonstrated mitochondrial degeneration as a precursor to CC in male mice; however, whether such alterations occur in females is currently unknown. The purpose of this study was to elucidate muscle alterations in CC development in female tumor-bearing mice. Sixty female C57BL/6J mice were injected with PBS or Lewis lung carcinoma at 8 wk of age, and tumors developed for 1, 2, 3, or 4 wk to assess the time course of cachectic development. In vivo muscle contractile function, protein fractional synthetic rate (FSR), protein turnover, and mitochondrial health were assessed. Three- and four-week tumor-bearing mice displayed a dichotomy in tumor growth and were reassigned to high tumor (HT) and low tumor (LT) groups. HT mice exhibited lower soleus, tibialis anterior, and fat weights than PBS mice. HT mice showed lower peak isometric torque and slower one-half relaxation time than PBS mice. HT mice had lower FSR than PBS mice, whereas E3 ubiquitin ligases were greater in HT than in other groups. Bnip3 (mitophagy) and pMitoTimer red puncta (mitochondrial degeneration) were greater in HT mice, whereas Pgc1α1 and Tfam (mitochondrial biogenesis) were lower in HT mice than in PBS mice. We demonstrate alterations in female tumor-bearing mice where HT exhibited greater protein degradation, impaired muscle contractility, and mitochondrial degeneration compared with other groups. Our data provide novel evidence for a distinct cachectic development in tumor-bearing female mice compared with previous male studies.NEW & NOTEWORTHY Our study demonstrates divergent tumor development and tissue wasting within 3- and 4-wk mice, where approximately half the mice developed large tumors and subsequent cachexia. Unlike previous male studies, where metabolic perturbations precede the onset of cachexia, females appear to exhibit protections from the metabolic perturbations and cachexia development. Our data provide novel evidence for divergent cachectic development in tumor-bearing female mice compared with previous male CC studies, suggesting different mechanisms of CC between sexes.
Collapse
Affiliation(s)
- Seongkyun Lim
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - J William Deaver
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Megan E Rosa-Caldwell
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Wesley S Haynie
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Francielly Morena da Silva
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Landen W Saling
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Lisa T Jansen
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Kirsten R Dunlap
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Michael P Wiggs
- Mooney Laboratory for Exercise, Nutrition, and Biochemistry, Department of Health, Human Performance and Recreation, Baylor University, Waco, Texas
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
12
|
Bivona JJ, Poynter ME. An open-source, lockable mouse wheel for the accessible implementation of time- and distance-limited elective exercise. PLoS One 2021; 16:e0261618. [PMID: 34932607 PMCID: PMC8691618 DOI: 10.1371/journal.pone.0261618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/06/2021] [Indexed: 11/18/2022] Open
Abstract
Current methods of small animal exercise involve either voluntary (wheel running) or forced (treadmill running) protocols. Although commonly used, each have several drawbacks which cause hesitancy to adopt these methods. While mice will instinctively run on a wheel, the distance and time spent running can vary widely. Forced exercise, while controllable, puts animals in stressful environments in which they are confined and often shocked for "encouragement." Additionally, both methods require expensive equipment and software, which limit these experiments to well-funded laboratories. To counter these issues, we developed a non-invasive mouse running device aimed to reduce handler-induced stress, provide time- and distance-based stopping conditions, and enable investigators with limited resources to easily produce and use the device. The Lockable Open-Source Training-Wheel (LOST-Wheel) was designed to be 3D printed on any standard entry-level printer and assembled using a few common tools for around 20 USD. It features an on-board screen and is capable of tracking distances, running time, and velocities of mice. The LOST-Wheel overcomes the largest drawback to voluntary exercise, which is the inability to control when and how long mice run, using a servo driven mechanism that locks and unlocks the running surface according to the protocol of the investigator. While the LOST-Wheel can be used without a computer connection, we designed an accompanying application to provide scientists with additional analyses. The LOST-Wheel Logger, an R-based application, displays milestones and plots on a user-friendly dashboard. Using the LOST-Wheel, we implemented a timed running experiment that showed distance-dependent decreases in serum myostatin as well as IL-6 gene upregulation in muscle. To make this device accessible, we are releasing the designs, application, and manual in an open-source format. The implementation of the LOST-Wheel and future iterations will improve upon existing murine exercise equipment and research.
Collapse
Affiliation(s)
- Joseph J. Bivona
- Department of Medicine and Vermont Lung Center, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cellular, Molecular, and Biomedical Sciences Doctoral Program, University of Vermont, Burlington, Vermont, United States of America
| | - Matthew E. Poynter
- Department of Medicine and Vermont Lung Center, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| |
Collapse
|
13
|
Peng Y, Qing X, Shu H, Tian S, Yang W, Chen S, Lin H, Lv X, Zhao L, Chen X, Pu F, Huang D, Cao X, Shao Z, Yp, Zs, Xc, Yp, Yp, Xq, Hs, St, Wy, Yp, Xq, Hs, St, Hl, Xl, Lz, Xc, Fp, Sc, Yp, Xq, Hs, St, Yp, Xq, Wy, Hl, Xl, Lz, Xc, Fp, Sc, Hdh, Wy, Hl, Xl, Lz, Xc, Fp, Sc, Hdh, Zs, Xc. Proper animal experimental designs for preclinical research of biomaterials for intervertebral disc regeneration. BIOMATERIALS TRANSLATIONAL 2021; 2:91-142. [PMID: 35836965 PMCID: PMC9255780 DOI: 10.12336/biomatertransl.2021.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/09/2021] [Indexed: 01/17/2023]
Abstract
Low back pain is a vital musculoskeletal disease that impairs life quality, leads to disability and imposes heavy economic burden on the society, while it is greatly attributed to intervertebral disc degeneration (IDD). However, the existing treatments, such as medicines, chiropractic adjustments and surgery, cannot achieve ideal disc regeneration. Therefore, advanced bioactive therapies are implemented, including stem cells delivery, bioreagents administration, and implantation of biomaterials etc. Among these researches, few reported unsatisfying regenerative outcomes. However, these advanced therapies have barely achieved successful clinical translation. The main reason for the inconsistency between satisfying preclinical results and poor clinical translation may largely rely on the animal models that cannot actually simulate the human disc degeneration. The inappropriate animal model also leads to difficulties in comparing the efficacies among biomaterials in different reaches. Therefore, animal models that better simulate the clinical charateristics of human IDD should be acknowledged. In addition, in vivo regenerative outcomes should be carefully evaluated to obtain robust results. Nevertheless, many researches neglect certain critical characteristics, such as adhesive properties for biomaterials blocking annulus fibrosus defects and hyperalgesia that is closely related to the clinical manifestations, e.g., low back pain. Herein, in this review, we summarized the animal models established for IDD, and highlighted the proper models and parameters that may result in acknowledged IDD models. Then, we discussed the existing biomaterials for disc regeneration and the characteristics that should be considered for regenerating different parts of discs. Finally, well-established assays and parameters for in vivo disc regeneration are explored.
Collapse
Affiliation(s)
- Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shuo Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wenbo Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Songfeng Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lei Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xi Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Feifei Pu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Donghua Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xu Cao
- Department of Orthopaedic Surgery, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA,Corresponding authors: Zengwu Shao, ; Xu Cao,
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China,Corresponding authors: Zengwu Shao, ; Xu Cao,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Beneficial Role of Exercise in the Modulation of mdx Muscle Plastic Remodeling and Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10040558. [PMID: 33916762 PMCID: PMC8066278 DOI: 10.3390/antiox10040558] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/02/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive progressive lethal disorder caused by the lack of dystrophin, which determines myofibers mechanical instability, oxidative stress, inflammation, and susceptibility to contraction-induced injuries. Unfortunately, at present, there is no efficient therapy for DMD. Beyond several promising gene- and stem cells-based strategies under investigation, physical activity may represent a valid noninvasive therapeutic approach to slow down the progression of the pathology. However, ethical issues, the limited number of studies in humans and the lack of consistency of the investigated training interventions generate loss of consensus regarding their efficacy, leaving exercise prescription still questionable. By an accurate analysis of data about the effects of different protocol of exercise on muscles of mdx mice, the most widely-used pre-clinical model for DMD research, we found that low intensity exercise, especially in the form of low speed treadmill running, likely represents the most suitable exercise modality associated to beneficial effects on mdx muscle. This protocol of training reduces muscle oxidative stress, inflammation, and fibrosis process, and enhances muscle functionality, muscle regeneration, and hypertrophy. These conclusions can guide the design of appropriate studies on human, thereby providing new insights to translational therapeutic application of exercise to DMD patients.
Collapse
|
15
|
Hamm SE, Fathalikhani DD, Bukovec KE, Addington AK, Zhang H, Perry JB, McMillan RP, Lawlor MW, Prom MJ, Vanden Avond MA, Kumar SN, Coleman KE, Dupont JB, Mack DL, Brown DA, Morris CA, Gonzalez JP, Grange RW. Voluntary wheel running complements microdystrophin gene therapy to improve muscle function in mdx mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:144-160. [PMID: 33850950 PMCID: PMC8020351 DOI: 10.1016/j.omtm.2021.02.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
We tested the hypothesis that voluntary wheel running would complement microdystrophin gene therapy to improve muscle function in young mdx mice, a model of Duchenne muscular dystrophy. mdx mice injected with a single dose of AAV9-CK8-microdystrophin or vehicle at age 7 weeks were assigned to three groups: mdxRGT (run, gene therapy), mdxGT (no run, gene therapy), or mdx (no run, no gene therapy). Wild-type (WT) mice were assigned to WTR (run) and WT (no run) groups. WTR and mdxRGT performed voluntary wheel running for 21 weeks; remaining groups were cage active. Robust expression of microdystrophin occurred in heart and limb muscles of treated mice. mdxRGT versus mdxGT mice showed increased microdystrophin in quadriceps but decreased levels in diaphragm. mdx final treadmill fatigue time was depressed compared to all groups, improved in mdxGT, and highest in mdxRGT. Both weekly running distance (km) and final treadmill fatigue time for mdxRGT and WTR were similar. Remarkably, mdxRGT diaphragm power was only rescued to 60% of WT, suggesting a negative impact of running. However, potential changes in fiber type distribution in mdxRGT diaphragms could indicate an adaptation to trade power for endurance. Post-treatment in vivo maximal plantar flexor torque relative to baseline values was greater for mdxGT and mdxRGT versus all other groups. Mitochondrial respiration rates from red quadriceps fibers were significantly improved in mdxGT animals, but the greatest bioenergetic benefit was observed in the mdxRGT group. Additional assessments revealed partial to full functional restoration in mdxGT and mdxRGT muscles relative to WT. These data demonstrate that voluntary wheel running combined with microdystrophin gene therapy in young mdx mice improved whole-body performance, affected muscle function differentially, mitigated energetic deficits, but also revealed some detrimental effects of exercise. With microdystrophin gene therapy currently in clinical trials, these data may help us understand the potential impact of exercise in treated patients.
Collapse
Affiliation(s)
- Shelby E Hamm
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| | - Daniel D Fathalikhani
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| | - Katherine E Bukovec
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| | - Adele K Addington
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| | - Haiyan Zhang
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| | - Justin B Perry
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| | - Ryan P McMillan
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| | - Michael W Lawlor
- Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mariah J Prom
- Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mark A Vanden Avond
- Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Suresh N Kumar
- Department of Pathology and Laboratory Medicine and Children's Hospital of Wisconsin Research Institute Imaging Core, Milwaukee, WI 53226, USA
| | - Kirsten E Coleman
- Powell Gene Therapy Center Toxicology Core, University of Florida, Gainesville, FL 32610, USA
| | - J B Dupont
- Translational Gene Therapy for Genetic Diseases, INSERM UMR1089, IRS2 Nantes Biotech, Université de Nantes, Nantes 44200, France
| | - David L Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98104, USA.,Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98107, USA
| | - David A Brown
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| | | | | | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, VA 24060, USA
| |
Collapse
|
16
|
Ng SY, Ljubicic V. Recent insights into neuromuscular junction biology in Duchenne muscular dystrophy: Impacts, challenges, and opportunities. EBioMedicine 2020; 61:103032. [PMID: 33039707 PMCID: PMC7648118 DOI: 10.1016/j.ebiom.2020.103032] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/28/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common and relentless form of muscular dystrophy. The pleiotropic effects of dystrophin deficiency include remarkable impacts on neuromuscular junction (NMJ) structure and function. Some of these alterations contribute to the severe muscle wasting and weakness that distinguish DMD, while others attempt to compensate for them. Experimental approaches that correct NMJ biology in pre-clinical models of DMD attenuate disease progression and improve functional outcomes, which suggests that targeting the NMJ may be an effective therapeutic strategy for DMD patients. The objectives of this review are to 1) survey the distinctions in NMJ structure, function, and gene expression in the dystrophic context as compared to the healthy condition, and 2) summarize the efforts, opportunities and challenges to correct NMJ biology in DMD. This information will expand our basic understanding of neuromuscular biology and may be useful for designing novel NMJ-targeted drug or behavioural strategies to mitigate the dystrophic pathology and other disorders of the neuromuscular system.
Collapse
Affiliation(s)
- Sean Y Ng
- Department of Kinesiology, McMaster University, Hamilton L8S 4L8, Ontario, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton L8S 4L8, Ontario, Canada.
| |
Collapse
|
17
|
Reddy A, Bozi LHM, Yaghi OK, Mills EL, Xiao H, Nicholson HE, Paschini M, Paulo JA, Garrity R, Laznik-Bogoslavski D, Ferreira JCB, Carl CS, Sjøberg KA, Wojtaszewski JFP, Jeppesen JF, Kiens B, Gygi SP, Richter EA, Mathis D, Chouchani ET. pH-Gated Succinate Secretion Regulates Muscle Remodeling in Response to Exercise. Cell 2020; 183:62-75.e17. [PMID: 32946811 DOI: 10.1016/j.cell.2020.08.039] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/05/2020] [Accepted: 08/17/2020] [Indexed: 12/23/2022]
Abstract
In response to skeletal muscle contraction during exercise, paracrine factors coordinate tissue remodeling, which underlies this healthy adaptation. Here we describe a pH-sensing metabolite signal that initiates muscle remodeling upon exercise. In mice and humans, exercising skeletal muscle releases the mitochondrial metabolite succinate into the local interstitium and circulation. Selective secretion of succinate is facilitated by its transient protonation, which occurs upon muscle cell acidification. In the protonated monocarboxylic form, succinate is rendered a transport substrate for monocarboxylate transporter 1, which facilitates pH-gated release. Upon secretion, succinate signals via its cognate receptor SUCNR1 in non-myofibrillar cells in muscle tissue to control muscle-remodeling transcriptional programs. This succinate-SUCNR1 signaling is required for paracrine regulation of muscle innervation, muscle matrix remodeling, and muscle strength in response to exercise training. In sum, we define a bioenergetic sensor in muscle that utilizes intracellular pH and succinate to coordinate tissue adaptation to exercise.
Collapse
Affiliation(s)
- Anita Reddy
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Luiz H M Bozi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA; Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Omar K Yaghi
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Evanna L Mills
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hilary E Nicholson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Margherita Paschini
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ryan Garrity
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Julio C B Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Christian S Carl
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kim A Sjøberg
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Murach KA, McCarthy JJ, Peterson CA, Dungan CM. Making Mice Mighty: recent advances in translational models of load-induced muscle hypertrophy. J Appl Physiol (1985) 2020; 129:516-521. [PMID: 32673155 DOI: 10.1152/japplphysiol.00319.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The ability to genetically manipulate mice allows for gain- and loss-of-function in vivo, making them an ideal model for elucidating mechanisms of skeletal muscle mass regulation. Combining genetic models with mechanical muscle loading enables identification of specific factors involved in the hypertrophic response as well as the ability to test the requirement of those factors for adaptation, thereby informing performance and therapeutic interventions. Until recently, approaches for inducing mechanically mediated muscle hypertrophy (i.e., resistance-training analogs) have been limited and considered "nontranslatable" to humans. This mini-review outlines recent translational advances in loading-mediated strategies for inducing muscle hypertrophy in mice, and highlights the advantages and disadvantages of each method. The skeletal muscle field is poised for new breakthroughs in understanding mechanisms regulating load-induced muscle growth given the numerous murine tools that have very recently been described.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
19
|
Masschelein E, D'Hulst G, Zvick J, Hinte L, Soro-Arnaiz I, Gorski T, von Meyenn F, Bar-Nur O, De Bock K. Exercise promotes satellite cell contribution to myofibers in a load-dependent manner. Skelet Muscle 2020; 10:21. [PMID: 32646489 PMCID: PMC7346400 DOI: 10.1186/s13395-020-00237-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/15/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Satellite cells (SCs) are required for muscle repair following injury and are involved in muscle remodeling upon muscular contractions. Exercise stimulates SC accumulation and myonuclear accretion. To what extent exercise training at different mechanical loads drive SC contribution to myonuclei however is unknown. RESULTS By performing SC fate tracing experiments, we show that 8 weeks of voluntary wheel running increased SC contribution to myofibers in mouse plantar flexor muscles in a load-dependent, but fiber type-independent manner. Increased SC fusion however was not exclusively linked to muscle hypertrophy as wheel running without external load substantially increased SC fusion in the absence of fiber hypertrophy. Due to nuclear propagation, nuclear fluorescent fate tracing mouse models were inadequate to quantify SC contribution to myonuclei. Ultimately, by performing fate tracing at the DNA level, we show that SC contribution mirrors myonuclear accretion during exercise. CONCLUSIONS Collectively, mechanical load during exercise independently promotes SC contribution to existing myofibers. Also, due to propagation of nuclear fluorescent reporter proteins, our data warrant caution for the use of existing reporter mouse models for the quantitative evaluation of satellite cell contribution to myonuclei.
Collapse
Affiliation(s)
- Evi Masschelein
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Gommaar D'Hulst
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Joel Zvick
- Department Health Sciences and Technology, Laboratory of Regenerative and Movement Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Laura Hinte
- Department Health Sciences and Technology, Laboratory of Nutrition and Metabolic Epigenetics, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Inés Soro-Arnaiz
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Tatiane Gorski
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ferdinand von Meyenn
- Department Health Sciences and Technology, Laboratory of Nutrition and Metabolic Epigenetics, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ori Bar-Nur
- Department Health Sciences and Technology, Laboratory of Regenerative and Movement Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Katrien De Bock
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
| |
Collapse
|
20
|
Lanza G, Pino M, Fisicaro F, Vagli C, Cantone M, Pennisi M, Bella R, Bellomo M. Motor activity and Becker's muscular dystrophy: lights and shadows. PHYSICIAN SPORTSMED 2020; 48:151-160. [PMID: 31646922 DOI: 10.1080/00913847.2019.1684810] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Becker's disease is an inherited muscular dystrophy caused by mutations in the gene coding for the dystrophin protein that leads to quantitative and/or qualitative protein dysfunction and consequent muscle degeneration. Studies in animal models demonstrate that, while eccentric or high-intensity training are deleterious for dystrophic muscles, low-intensity aerobic training may slowdown the disease process and progression. Based on these preclinical data, the available studies in patients with Becker's muscular dystrophy undergoing workout on a cycle ergometer or on a treadmill, at a heart rate ≤65% of their maximal oxygen uptake, showed that aerobic exercise counteracts physical deterioration and loss of functional abilities. These findings suggest an improvement of physical performance through an increase of muscle strength, fatigue resistance, and dexterity capacities, without substantial evidence of acceleration of muscular damage progression. Therefore, individually tailored mild-to-moderate intensity aerobic exercise should be considered as part of the management of these patients. However, further research is necessary to define specific and standardized guidelines for the prescription of type, intensity, frequency, and duration of motor activities. In this review, we provided a summary of the impact of physical activity both in animal models and in patients with Becker's muscular dystrophy, with the intent to identify trends and gaps in knowledge. The potential therapeutic implications and future research directions have been also highlighted.
Collapse
Affiliation(s)
- Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
- Department of Neurology IC, Oasi Research Institute - IRCCS, Troina, Italy
| | - Marcello Pino
- School of Human and Social Science, University Kore of Enna, Enna, Italy
| | - Francesco Fisicaro
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, Catania, Italy
| | - Carla Vagli
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, Catania, Italy
| | - Mariagiovanna Cantone
- Department of Neurology, Sant'Elia Hospital, ASP Caltanissetta, Caltanissetta, Italy
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, Catania, Italy
| | - Maria Bellomo
- School of Human and Social Science, University Kore of Enna, Enna, Italy
| |
Collapse
|
21
|
Muscular Dystrophy and Rehabilitation Interventions with Regenerative Treatment. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2020. [DOI: 10.1007/s40141-019-00255-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
22
|
Torres-Espín A, Beaudry E, Fenrich K, Fouad K. Rehabilitative Training in Animal Models of Spinal Cord Injury. J Neurotrauma 2019; 35:1970-1985. [PMID: 30074874 DOI: 10.1089/neu.2018.5906] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rehabilitative motor training is currently one of the most widely used approaches to promote moderate recovery following injuries of the central nervous system. Such training is generally applied in the clinical setting, whereas it is not standard in preclinical research. This is a concern as it is becoming increasingly apparent that neuroplasticity enhancing treatments require training or some form of activity as a co-therapy to promote functional recovery. Despite the importance of training and the many open questions regarding its mechanistic consequences, its use in preclinical animal models is rather limited. Here we review approaches, findings and challenges when training is applied in animal models of spinal cord injury, and we suggest recommendations to facilitate the integration of training using an appropriate study design, into pre-clinical studies.
Collapse
Affiliation(s)
- Abel Torres-Espín
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta , Edmonton, Alberta, Canada
| | - Eric Beaudry
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta , Edmonton, Alberta, Canada
| | | | - Karim Fouad
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta , Edmonton, Alberta, Canada
| |
Collapse
|
23
|
D'Hulst G, Palmer AS, Masschelein E, Bar-Nur O, De Bock K. Voluntary Resistance Running as a Model to Induce mTOR Activation in Mouse Skeletal Muscle. Front Physiol 2019; 10:1271. [PMID: 31636571 PMCID: PMC6787551 DOI: 10.3389/fphys.2019.01271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/19/2019] [Indexed: 12/30/2022] Open
Abstract
Long-term voluntary resistance running has been shown to be a valid model to induce muscle growth in rodents. Moreover, the mammalian target of rapamycin complex 1 (mTORC1) is a key signaling complex regulating exercise/nutrient-induced alterations in muscle protein synthesis. How acute resistance running affects mTORC1 signaling in muscle and if resistance applied to the wheel can modulate mTORC1 activation has not yet been fully elucidated. Here, we show that both acute resistance running and acute free running activated mTORC1 signaling in the m. gastrocnemius, m. soleus, and m. plantaris, but not in m. tibialis anterior of mice when compared to sedentary controls. Furthermore, only the low threshold oxidative part in the m. gastrocnemius showed increased mTORC1 signaling upon running and acute heavy-load resistance running evoked higher downstream mTORC1 signaling in both m. soleus and m. plantaris than free running without resistance, pointing toward mechanical load as an important independent regulator of mTORC1. Collectively, in this study, we show that voluntary resistance running is an easy-to-use, time-efficient and low stress model to study acute alterations in mTORC1 signaling upon high-load muscular contractions in mice.
Collapse
Affiliation(s)
- Gommaar D'Hulst
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Andrew S Palmer
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Evi Masschelein
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ori Bar-Nur
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Ng SY, Mikhail A, Ljubicic V. Mechanisms of exercise-induced survival motor neuron expression in the skeletal muscle of spinal muscular atrophy-like mice. J Physiol 2019; 597:4757-4778. [PMID: 31361024 PMCID: PMC6767691 DOI: 10.1113/jp278454] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/26/2019] [Indexed: 12/29/2022] Open
Abstract
Key points Spinal muscular atrophy (SMA) is a health‐ and life‐limiting neuromuscular disorder caused by a deficiency in survival motor neuron (SMN) protein. While historically considered a motor neuron disease, current understanding of SMA emphasizes its systemic nature, which requires addressing affected peripheral tissues such as skeletal muscle in particular. Chronic physical activity is beneficial for SMA patients, but the cellular and molecular mechanisms of exercise biology are largely undefined in SMA. After a single bout of exercise, canonical responses such as skeletal muscle AMP‐activated protein kinase (AMPK), p38 mitogen‐activated protein kinase (p38) and peroxisome proliferator‐activated receptor γ coactivator 1α (PGC‐1α) activation were preserved in SMA‐like Smn2B/− animals. Furthermore, molecules involved in SMN transcription were also altered following physical activity. Collectively, these changes were coincident with an increase in full‐length SMN transcription and corrective SMN pre‐mRNA splicing. This study advances understanding of the exercise biology of SMA and highlights the AMPK–p38–PGC‐1α axis as a potential regulator of SMN expression in muscle.
Abstract Chronic physical activity is safe and effective in spinal muscular atrophy (SMA) patients, but the underlying cellular events that drive physiological adaptations are undefined. We examined the effects of a single bout of exercise on molecular mechanisms associated with adaptive remodelling in the skeletal muscle of Smn2B/− SMA‐like mice. Skeletal muscles were collected from healthy Smn2B/+ mice and Smn2B/− littermates at pre‐ (postnatal day (P) 9), early‐ (P13) and late‐ (P21) symptomatic stages to characterize SMA disease progression. Muscles were also collected from Smn2B/− animals exercised to fatigue on a motorized treadmill. Intracellular signalling and gene expression were examined using western blotting, confocal immunofluorescence microscopy, real‐time quantitative PCR and endpoint PCR assays. Basal skeletal muscle AMP‐activated protein kinase (AMPK) and p38 mitogen‐activated protein kinase (p38) expression and activity were not affected by SMA‐like conditions. Canonical exercise responses such as AMPK, p38 and peroxisome proliferator‐activated receptor γ coactivator‐1α (PGC‐1α) activation were observed following a bout of exercise in Smn2B/− animals. Furthermore, molecules involved in survival motor neuron (SMN) transcription, including protein kinase B (AKT) and extracellular signal‐regulated kinases (ERK)/ETS‐like gene 1 (ELK1), were altered following physical activity. Acute exercise was also able to mitigate aberrant proteolytic signalling in the skeletal muscle of Smn2B/− mice. Collectively, these changes were coincident with an exercise‐evoked increase in full‐length SMN mRNA expression. This study advances our understanding of the exercise biology of SMA and highlights the AMPK–p38–PGC‐1α axis as a potential regulator of SMN expression alongside AKT and ERK/ELK1 signalling. Spinal muscular atrophy (SMA) is a health‐ and life‐limiting neuromuscular disorder caused by a deficiency in survival motor neuron (SMN) protein. While historically considered a motor neuron disease, current understanding of SMA emphasizes its systemic nature, which requires addressing affected peripheral tissues such as skeletal muscle in particular. Chronic physical activity is beneficial for SMA patients, but the cellular and molecular mechanisms of exercise biology are largely undefined in SMA. After a single bout of exercise, canonical responses such as skeletal muscle AMP‐activated protein kinase (AMPK), p38 mitogen‐activated protein kinase (p38) and peroxisome proliferator‐activated receptor γ coactivator 1α (PGC‐1α) activation were preserved in SMA‐like Smn2B/− animals. Furthermore, molecules involved in SMN transcription were also altered following physical activity. Collectively, these changes were coincident with an increase in full‐length SMN transcription and corrective SMN pre‐mRNA splicing. This study advances understanding of the exercise biology of SMA and highlights the AMPK–p38–PGC‐1α axis as a potential regulator of SMN expression in muscle.
Collapse
Affiliation(s)
- Sean Y Ng
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Andrew Mikhail
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
25
|
Bouglé A, Rocheteau P, Briand D, Hardy D, Verdonk F, Tremolada C, Hivelin M, Chrétien F. Beneficial role of adipose-derived mesenchymal stem cells from microfragmented fat in a murine model of duchenne muscular dystrophy. Muscle Nerve 2019; 60:328-335. [PMID: 31228273 DOI: 10.1002/mus.26614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/12/2019] [Accepted: 06/16/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION No etiologic therapy is available for Duchenne muscular dystrophy (DMD), but mesenchymal stem cells were shown to be effective in preclinical models of DMD. The objective of this study is to investigate the effect of microfragmented fat extracted on a murine model of DMD. METHODS Fat tissue was extracted from healthy human participants and injected IM into DMD mice. Histological analysis, cytokines, and force measurement were performed up to 4 weeks after injection. RESULTS Duchenne muscular dystrophy mice injected with microfragmented fat exhibited an improved muscle phenotype (decreased necrosis and fibrosis), a decrease of inflammatory cytokines, and increased strength. DISCUSSION Administration of microfragmented fat in key muscles may improve muscular phenotype in patients with DMD. Muscle Nerve, 2019.
Collapse
Affiliation(s)
- Adrien Bouglé
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France.,Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Department of Anesthesiology and Critical Care Medicine, Pitié-Salpêtrière Hospital, Paris, France.,Assistance Publique-Hôpitaux de Paris, Paris, France.,Department of Anesthesiology and Critical Care Medicine, Institute of Cardiology, Pitié-Salpêtrière Hospital, Paris, France
| | - Pierre Rocheteau
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France.,Service Hospitalo-Universitaire, Centre Hospitalier Sainte Anne, Paris, France.,Laboratoire Universitaire de Neuropathologie, Centre Hospitalier Sainte Anne, Paris, France
| | - David Briand
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France
| | - David Hardy
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France
| | - Franck Verdonk
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France.,Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Department of Anesthesiology and Critical Care Medicine, Pitié-Salpêtrière Hospital, Paris, France.,Assistance Publique-Hôpitaux de Paris, Paris, France.,Department of Anesthesiology and Critical Care Department, Saint-Antoine Hospital, Paris, France
| | | | - Mikael Hivelin
- Assistance Publique-Hôpitaux de Paris, Paris, France.,Descartes University, Assistance Publique - Hôpitaux de Paris, Department of Plastic Surgery, Hôpital Européen Georges Pompidou, Paris, France.,Department of Plastic Surgery, Hôpital Européen Georges Pompidou, Paris, France
| | - Fabrice Chrétien
- Infection and Epidemiology Department, Experimental Neuropathology Unit, Institut Pasteur, Paris, France.,Laboratoire Universitaire de Neuropathologie, Centre Hospitalier Sainte Anne, Paris, France.,Descartes University, Assistance Publique - Hôpitaux de Paris, Department of Plastic Surgery, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
26
|
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a neuromuscular disease caused by a dystrophin protein deficiency. Dystrophin functions to stabilize and protect the muscle fiber during muscle contraction; thus, the absence of functional dystrophin protein leads to muscle injury. DMD patients experience progressive muscle necrosis, loss of function, and ultimately succumb to respiratory failure or cardiomyopathy. Exercise is known to improve muscle health and strength in healthy individuals as well as positively affect other systems. Because of this, exercise has been investigated as a potential therapeutic approach for DMD. METHODS This review aims to provide a concise presentation of the exercise literature with a focus on dystrophin-deficient muscle. Our intent was to identify trends and gaps in knowledge with an appreciation of exercise modality. RESULTS After compiling data from mouse and human studies, it became apparent that endurance exercises such as a swimming and voluntary wheel running have therapeutic potential in limb muscles of mice and respiratory training was beneficial in humans. However, in the comparatively few long-term investigations, the effect of low-intensity training on cardiac and respiratory muscles was contradictory. In addition, the effect of exercise on other systems is largely unknown. CONCLUSIONS To safely prescribe exercise as a therapy to DMD patients, multisystemic investigations are needed including the evaluation of respiratory and cardiac muscle.
Collapse
|
27
|
Graber TG, Fandrey KR, Thompson LV. Novel individualized power training protocol preserves physical function in adult and older mice. GeroScience 2019; 41:165-183. [PMID: 31076998 PMCID: PMC6544743 DOI: 10.1007/s11357-019-00069-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/16/2019] [Indexed: 11/27/2022] Open
Abstract
Sarcopenia, the age-related loss of muscle mass and strength, contributes to frailty, functional decline, and reduced quality of life in older adults. Exercise is a recognized therapy for sarcopenia and muscle dysfunction, though not a cure. Muscle power declines at an increased rate compared to force, and force output declines earlier than mass. Thus, there is a need for research of exercise focusing on improving power output and functionality in older adults. Our primary purpose was proof-of-concept that a novel individualized power exercise modality would induce positive adaptations in adult mice, before the exercise program was applied to an aged cohort. We hypothesized that after following our protocol, both adult and older mice would show improved function, though there would be evidence of anabolic resistance in the older mice. Male C57BL/6 mice (12 months of age at study conclusion) were randomized into control (n = 9) and exercise (n = 6) groups. The trained group used progressive resistance (with a weighted harness) and intensity (~ 4-10 rpm) on a custom motorized running wheel. The mice trained similarly to a human workout regimen (4-5 sets/session, 3 sessions/week, for 12 weeks). We determined significant (p < 0.05) positive adaptations post-intervention, including: neuromuscular function (rotarod), strength/endurance (inverted cling grip test), training physiology (force/power output per session), muscle size (soleus mass), and power/velocity of contraction (in vitro physiology). Secondly, we trained a cohort of older male mice (28 months old at conclusion): control (n = 12) and exercised (n = 8). While the older exercised mice did preserve function and gain benefits, they also demonstrated evidence of anabolic resistance.
Collapse
Affiliation(s)
- Ted G. Graber
- Department of Nutrition and Metabolism, Division of Rehabilitation Science, University of Texas Medical Branch, Galveston, TX USA
| | - Katie R. Fandrey
- Program in Physical Therapy, University of Minnesota-Twin Cities, Minneapolis, MN USA
| | - LaDora V. Thompson
- Department of Physical Therapy & Athletic Training, Boston University, Boston, MA USA
| |
Collapse
|
28
|
Manta A, Stouth DW, Xhuti D, Chi L, Rebalka IA, Kalmar JM, Hawke TJ, Ljubicic V. Chronic exercise mitigates disease mechanisms and improves muscle function in myotonic dystrophy type 1 mice. J Physiol 2019; 597:1361-1381. [PMID: 30628727 DOI: 10.1113/jp277123] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 01/04/2019] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Myotonic dystrophy type 1 (DM1), the second most common muscular dystrophy and most prevalent adult form of muscular dystrophy, is characterized by muscle weakness, wasting and myotonia. A microsatellite repeat expansion mutation results in RNA toxicity and dysregulation of mRNA processing, which are the primary downstream causes of the disorder. Recent studies with DM1 participants demonstrate that exercise is safe, enjoyable and elicits benefits in muscle strength and function; however, the molecular mechanisms of exercise adaptation in DM1 are undefined. Our results demonstrate that 7 weeks of volitional running wheel exercise in a pre-clinical DM1 mouse model resulted in significantly improved motor performance, muscle strength and endurance, as well as reduced myotonia. At the cellular level, chronic physical activity attenuated RNA toxicity, liberated Muscleblind-like 1 protein from myonuclear foci and improved mRNA alternative splicing. ABSTRACT Myotonic dystrophy type 1 (DM1) is a trinucleotide repeat expansion neuromuscular disorder that is most prominently characterized by skeletal muscle weakness, wasting and myotonia. Chronic physical activity is safe and satisfying, and can elicit functional benefits such as improved strength and endurance in DM1 patients, but the underlying cellular basis of exercise adaptation is undefined. Our purpose was to examine the mechanisms of exercise biology in DM1. Healthy, sedentary wild-type (SED-WT) mice, as well as sedentary human skeletal actin-long repeat animals, a murine model of DM1 myopathy (SED-DM1), and DM1 mice with volitional access to a running wheel for 7 weeks (EX-DM1), were utilized. Chronic exercise augmented strength and endurance in vivo and in situ in DM1 mice. These alterations coincided with normalized measures of myopathy, as well as increased mitochondrial content. Electromyography revealed a 70-85% decrease in the duration of myotonic discharges in muscles from EX-DM1 compared to SED-DM1 animals. The exercise-induced enhancements in muscle function corresponded at the molecular level with mitigated spliceopathy, specifically the processing of bridging integrator 1 and muscle-specific chloride channel (CLC-1) transcripts. CLC-1 protein content and sarcolemmal expression were lower in SED-DM1 versus SED-WT animals, but they were similar between SED-WT and EX-DM1 groups. Chronic exercise also attenuated RNA toxicity, as indicated by reduced (CUG)n foci-positive myonuclei and sequestered Muscleblind-like 1 (MBNL1). Our data indicate that chronic exercise-induced physiological improvements in DM1 occur in concert with mitigated primary downstream disease mechanisms, including RNA toxicity, MBNL1 loss-of-function, and alternative mRNA splicing.
Collapse
Affiliation(s)
- Alexander Manta
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Derek W Stouth
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Donald Xhuti
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Leon Chi
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Jayne M Kalmar
- Department of Kinesiology & Physical Education, Wilfred Laurier University, Waterloo, ON, Canada, N2L 3C5
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| |
Collapse
|
29
|
Lindsay A, Larson AA, Verma M, Ervasti JM, Lowe DA. Isometric resistance training increases strength and alters histopathology of dystrophin-deficient mouse skeletal muscle. J Appl Physiol (1985) 2018; 126:363-375. [PMID: 30571283 DOI: 10.1152/japplphysiol.00948.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mutation to the dystrophin gene causes skeletal muscle weakness in patients with Duchenne muscular dystrophy (DMD) or Becker muscular dystrophy (BMD). Deliberation continues regarding implications of prescribing exercise for these patients. The purpose of this study was to determine whether isometric resistance exercise (~10 tetanic contractions/session) improves skeletal muscle strength and histopathology in the mdx mouse model of DMD. Three isometric training sessions increased in vivo isometric torque (22%) and contractility rates (54%) of anterior crural muscles of mdx mice. Mice expressing a BMD-causing missense mutated dystrophin on the mdx background showed comparable increases in torque (22%), while wild-type mice showed less change (11%). Increases in muscle function occurred within 1 h and peaked 3 days posttraining; however, the adaptation was lost after 7 days unless retrained. Six isometric training sessions over 4 wk caused increased isometric torque (28%) and contractility rates (22-28%), reduced fibrosis, as well as greater uniformity of fiber cross-sectional areas, fewer embryonic myosin heavy-chain-positive fibers, and more satellite cells in tibialis anterior muscle compared with the contralateral untrained muscle. Ex vivo functional analysis of isolated extensor digitorum longus (EDL) muscle from the trained hindlimb revealed greater absolute isometric force, lower passive stiffness, and a lower susceptibility to eccentric contraction-induced force loss compared with untrained EDL muscle. Overall, these data support the concept that exercise training in the form of isometric tetanic contractions can improve contractile function of dystrophin-deficient muscle, indicating a potential role for enhancing muscle strength in patients with DMD and BMD. NEW & NOTEWORTHY We focused on adaptive responses of dystrophin-deficient mouse skeletal muscle to isometric contraction training and report that in the absence of dystrophin (or in the presence of a mutated dystrophin), strength and muscle histopathology are improved. Results suggest that the strength gains are associated with fiber hypertrophy, reduced fibrosis, increased number of satellite cells, and blunted eccentric contraction-induced force loss in vitro. Importantly, there was no indication that the isometric exercise training was deleterious to dystrophin-deficient muscle.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota , Minneapolis, Minnesota.,Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota , Minneapolis, Minnesota
| | - Alexie A Larson
- Department of Integrative Biology and Physiology, University of Minnesota , Minneapolis, Minnesota
| | - Mayank Verma
- Department of Integrative Biology and Physiology, University of Minnesota , Minneapolis, Minnesota.,Medical Scientist Training Program, University of Minnesota Medical School , Minneapolis, Minnesota
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota , Minneapolis, Minnesota
| | - Dawn A Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
30
|
Heutinck L, Jansen M, van den Elzen Y, van der Pijl D, de Groot IJM. Virtual Reality Computer Gaming with Dynamic Arm Support in Boys with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2018; 5:359-372. [PMID: 29991140 DOI: 10.3233/jnd-180307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND In boys with Duchenne muscular dystrophy (DMD), loss of upper limb function becomes more evident after the onset of wheelchair-dependency, because of the inability to lift the arms against gravity. With an increasing population of older wheelchair-dependent boys with DMD it is worthwhile to know whether training can delay the loss of upper limb functions. Dynamic arm supports may enable boys with impaired arm function to train their muscles without becoming exhausted by providing external mechanical compensation for muscle weakness. OBJECTIVE This study investigated the effect of gravity-compensated 3D-training for the arms on the functional abilities in boys with DMD. METHODS An explorative RCT was conducted among boys with DMD with impaired arm function (n = 16). Boys in the intervention group (n = 7) trained their arms by playing virtual reality games while using dynamic arm support during 20 weeks. The primary endpoint was the difference in change in Performance of the Upper Limb (PUL) score between the intervention and control group (n = 9) after 20 weeks. Secondary outcome measures were at the different ICF-CY levels. RESULTS No significant group differences were found for the PUL. Elbow range of motion (p = 0.018) and extension strength (p = 0.038) improved in the intervention group and worsened in the control group. CONCLUSIONS Although this study did not show a significant effect of training on the primary outcome measure, there are indications that training may decline the loss of range of motion and strength. This may prolong the functional abilities on long-term. TRIAL REGISTRATION Netherlands Trial Register 3857.
Collapse
Affiliation(s)
- Lotte Heutinck
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Merel Jansen
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yolanda van den Elzen
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Imelda J M de Groot
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
31
|
Voluntary exercise improves muscle function and does not exacerbate muscle and heart pathology in aged Duchenne muscular dystrophy mice. J Mol Cell Cardiol 2018; 125:29-38. [PMID: 30336143 DOI: 10.1016/j.yjmcc.2018.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/21/2018] [Accepted: 10/08/2018] [Indexed: 12/31/2022]
Abstract
Duchenne muscular dystrophy is a severe muscle wasting disease, characterized by a severely reduced lifespan in which cardiomyopathy is one of the leading causes of death. Multiple therapies aiming at dystrophin restoration have been approved. It is anticipated that these therapies will maintain muscle function for longer and extend the ambulatory period, which in turn will increase the cardiac workload which could be detrimental for cardiac function. We investigated the effects of voluntary running exercise in combination with low dystrophin levels on function and pathology of skeletal muscle and heart. We divided 15.5-month old female mdx (no dystrophin), mdx-XistΔhs (varying low dystrophin levels) and wild type mice (BL10-WT and XistΔhs-WT) to either a sedentary or voluntary wheel running regime and assessed muscle function at 17.5 months of age. Thereafter, a cardiac MRI was obtained, and muscle and heart histopathology were assessed. We show that voluntary exercise is beneficial to skeletal muscle and heart function in dystrophic mice while not affecting muscle pathology. Low amounts of dystrophin further improve skeletal muscle and cardiac function. These findings suggest that voluntary exercise may be beneficial for skeletal muscle and heart in DMD patients, especially in conjunction with low amounts of dystrophin.
Collapse
|
32
|
Delacroix C, Hyzewicz J, Lemaitre M, Friguet B, Li Z, Klein A, Furling D, Agbulut O, Ferry A. Improvement of Dystrophic Muscle Fragility by Short-Term Voluntary Exercise through Activation of Calcineurin Pathway in mdx Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2662-2673. [PMID: 30142334 DOI: 10.1016/j.ajpath.2018.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 06/23/2018] [Accepted: 07/16/2018] [Indexed: 01/01/2023]
Abstract
Dystrophin deficiency in mdx mice, a model for Duchenne muscular dystrophy, leads to muscle weakness revealed by a reduced specific maximal force as well as fragility (ie, higher susceptibility to contraction-induced injury, as shown by a greater force decrease after lengthening contractions). Both symptoms could be improved with dystrophin restoration-based therapies and long-term (months) voluntary exercise. Herein, we evaluated the effect of short-term (1-week) voluntary wheel running. We found that running improved fragility of tibialis anterior muscle (TA), but not plantaris muscle, independently of utrophin up-regulation, without affecting weakness. Moreover, TA muscle excitability was also preserved by running, as shown by compound muscle action potential measurements after lengthening contractions. Of interest, the calcineurin inhibitor cyclosporin A prevented the effect of running on both muscle fragility and excitability. Cyclosporin also prevented the running-induced changes in expression of genes involved in excitability (Scn4a and Cacna1s) and slower contractile phenotype (Myh2 and Tnni1) in TA muscle. In conclusion, short-term voluntary exercise improves TA muscle fragility in mdx mice, without worsening weakness. Its effect was related to preserved excitability, calcineurin pathway activation, and changes in the program of genes involved in excitability and slower contractile phenotype. Thus, remediation of muscle fragility of Duchenne muscular dystrophy patients through appropriate exercise training deserves to be explored in more detail.
Collapse
Affiliation(s)
- Clement Delacroix
- Research Center in Myology, Association Institute of Myology, Sorbonne University, INSERM, UMRS974, Paris, France
| | - Janek Hyzewicz
- Biological Adaptation and Aging, Institute of Biology Paris-Seine, UMR CNRS 8256, INSERM ERL U1164, Sorbonne University, Paris, France
| | - Megane Lemaitre
- Research Center in Myology, Association Institute of Myology, Sorbonne University, INSERM, UMRS974, Paris, France
| | - Bertrand Friguet
- Biological Adaptation and Aging, Institute of Biology Paris-Seine, UMR CNRS 8256, INSERM ERL U1164, Sorbonne University, Paris, France
| | - Zhenlin Li
- Biological Adaptation and Aging, Institute of Biology Paris-Seine, UMR CNRS 8256, INSERM ERL U1164, Sorbonne University, Paris, France
| | - Arnaud Klein
- Research Center in Myology, Association Institute of Myology, Sorbonne University, INSERM, UMRS974, Paris, France
| | - Denis Furling
- Research Center in Myology, Association Institute of Myology, Sorbonne University, INSERM, UMRS974, Paris, France
| | - Onnik Agbulut
- Biological Adaptation and Aging, Institute of Biology Paris-Seine, UMR CNRS 8256, INSERM ERL U1164, Sorbonne University, Paris, France
| | - Arnaud Ferry
- Research Center in Myology, Association Institute of Myology, Sorbonne University, INSERM, UMRS974, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
33
|
Ruehle MA, Stevens HY, Beedle AM, Guldberg RE, Call JA. Aggregate mesenchymal stem cell delivery ameliorates the regenerative niche for muscle repair. J Tissue Eng Regen Med 2018; 12:1867-1876. [PMID: 29774991 DOI: 10.1002/term.2707] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 04/18/2018] [Accepted: 05/08/2018] [Indexed: 01/02/2023]
Abstract
Duchenne muscular dystrophy is a severe muscle wasting disease due to the absence of the dystrophin protein from the muscle cell membrane, which renders the muscle susceptible to continuous damage. In Duchenne muscular dystrophy patients, muscle weakness, together with cycles of degeneration/regeneration and replacement with noncontractile tissue, limit mobility and lifespan. Because the loss of dystrophin results in loss of polarity and a reduction in the number of self-renewing satellite cells, it is postulated that these patients could achieve an improved quality of life if delivered cells could restore satellite cell function. In this study, we used both an established myotoxic injury model in wild-type (WT) mice and mdx mice alone (spontaneous muscle damage). Single (SC) and aggregated (AGG) mesenchymal stem cells (MSCs) were injected into the gastrocnemius muscles 4 hr after injury (WT mice). The recovery of peak isometric torque was longitudinally assessed over 5 weeks, with earlier takedowns for histological assessment of healing (fibre cross-section area and central nucleation) and MSC retention. AGG-treated WT mice had significantly greater torque recovery at Day 14 than SC or saline-treated mice and a greater CSA at Day 10, compared with SC/saline. AGG-treated mdx mice had a greater peak isometric torque compared with SC/saline. In vitro immunomodulatory factor secretion of AGG-MSCs was higher than SC-MSCs for all tested growth factors with the largest difference observed in hepatocyte growth factor. Future studies are necessary to pair immunomodulatory factor secretion with functional attributes, to better predict the potential therapeutic value of MSC treatment modalities.
Collapse
Affiliation(s)
- Marissa A Ruehle
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hazel Y Stevens
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Aaron M Beedle
- Department of Pharmaceutical Sciences, SUNY Binghamton University, Binghamton, NY, USA
| | - Robert E Guldberg
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jarrod A Call
- Department of Kinesiology, University of Georgia, Athens, GA, USA.,Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| |
Collapse
|
34
|
Heydemann A. Skeletal Muscle Metabolism in Duchenne and Becker Muscular Dystrophy-Implications for Therapies. Nutrients 2018; 10:nu10060796. [PMID: 29925809 PMCID: PMC6024668 DOI: 10.3390/nu10060796] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 02/06/2023] Open
Abstract
The interactions between nutrition and metabolism and skeletal muscle have long been known. Muscle is the major metabolic organ—it consumes more calories than other organs—and therefore, there is a clear need to discuss these interactions and provide some direction for future research areas regarding muscle pathologies. In addition, new experiments and manuscripts continually reveal additional highly intricate, reciprocal interactions between metabolism and muscle. These reciprocal interactions include exercise, age, sex, diet, and pathologies including atrophy, hypoxia, obesity, diabetes, and muscle myopathies. Central to this review are the metabolic changes that occur in the skeletal muscle cells of muscular dystrophy patients and mouse models. Many of these metabolic changes are pathogenic (inappropriate body mass changes, mitochondrial dysfunction, reduced adenosine triphosphate (ATP) levels, and increased Ca2+) and others are compensatory (increased phosphorylated AMP activated protein kinase (pAMPK), increased slow fiber numbers, and increased utrophin). Therefore, reversing or enhancing these changes with therapies will aid the patients. The multiple therapeutic targets to reverse or enhance the metabolic pathways will be discussed. Among the therapeutic targets are increasing pAMPK, utrophin, mitochondrial number and slow fiber characteristics, and inhibiting reactive oxygen species. Because new data reveals many additional intricate levels of interactions, new questions are rapidly arising. How does muscular dystrophy alter metabolism, and are the changes compensatory or pathogenic? How does metabolism affect muscular dystrophy? Of course, the most profound question is whether clinicians can therapeutically target nutrition and metabolism for muscular dystrophy patient benefit? Obtaining the answers to these questions will greatly aid patients with muscular dystrophy.
Collapse
Affiliation(s)
- Ahlke Heydemann
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA.
- Center for Cardiovascular Research, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
35
|
Martins CEC, Lima VBDS, Schoenfeld BJ, Tirapegui J. Effects of leucine supplementation and resistance training on myopathy of diabetic rats. Physiol Rep 2018; 5:e13273. [PMID: 28536139 PMCID: PMC5449559 DOI: 10.14814/phy2.13273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 11/24/2022] Open
Abstract
Leucine supplementation and resistance training positively influence the protein translation process and the cell signaling mTOR (mammalian target of rapamycin) pathway that regulates muscle protein balance and muscle remodeling, and thus may be therapeutic to diabetic myopathy. However, the effect of a combined intervention has not been well studied. Forty male Wistar rats were divided into five groups, control (C), diabetic control (D), diabetic + trained (DT), diabetic + L-leucine (DL), diabetic + L-leucine + trained (DLT). The supplementation of 5% leucine in chow, and resistance training were conducted for 8 weeks postweaning of rats. The extensor digitorum longus was used to assess signaling proteins involved in muscle protein synthesis, and the gastrocnemius and soleus were used for determination of muscle weight. Blood samples were collected for biochemical assays. Strength and ambulation tests were employed to evaluate motor performance. Results showed that both leucine supplementation and resistance training elevated the activity of mTOR-p70S6K in diabetic rats (P < 0.05). Moreover, though leucine supplementation in combination with resistance training demonstrated synergistic effects on p70S6K (P < 0.05), both treatments were capable of recovering motor performance (P < 0.05). In conclusion, 5% leucine supplementation combined with resistance training has the potential to attenuate muscle loss and motor performance decrements in diabetic rats, at least in part through increased protein synthesis.
Collapse
Affiliation(s)
- Carlos Eduardo C Martins
- Department of Food Science and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vanessa B de S Lima
- Department of Food Science and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Julio Tirapegui
- Department of Food Science and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Abstract
Duchenne muscular dystrophy is a lethal genetic disease of muscle wasting for which there is no cure. In healthy muscle, structure and function improve dramatically with exercise. In patients with dystrophy, little is known about the effects of exercise. As contemporary therapies rapidly progress and patients become more active, there is a need to understand the effects of exercise.
Collapse
|
37
|
Lee M, Soya H. Effects of acute voluntary loaded wheel running on BDNF expression in the rat hippocampus. J Exerc Nutrition Biochem 2017; 21:52-57. [PMID: 29370674 PMCID: PMC5772069 DOI: 10.20463/jenb.2017.0034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/28/2017] [Indexed: 12/13/2022] Open
Abstract
[Purpose] Voluntary loaded wheel running involves the use of a load during a voluntary running activity. A muscle-strength or power-type activity performed at a relatively high intensity and a short duration may cause fewer apparent metabolic adaptations but may still elicit muscle fiber hypertrophy. This study aimed to determine the effects of acute voluntary wheel running with an additional load on brain-derived neurotrophic factor (BDNF) expression in the rat hippocampus. [Methods] Ten-week old male Wistar rats were assigned randomly to a (1) sedentary (Control) group; (2) voluntary exercise with no load (No-load) group; or (3) voluntary exercise with an additional load (Load) group for 1-week (acute period). The expression of BDNF genes was quantified by real-time PCR. [Results] The average distance levels were not significantly different in the No-load and Load groups. However, the average work levels significantly increased in the Load group. The relative soleus weights were greater in the No-load group. Furthermore, loaded wheel running up-regulated the BDNF mRNA level compared with that in the Control group. The BDNF mRNA levels showed a positive correlation with workload levels (r=0.75), suggesting that the availability of multiple workload levels contributes to the BDNF-related benefits of loaded wheel running noted in this study. [Conclusion] This novel approach yielded the first set of findings showing that acute voluntary loaded wheel running, which causes muscular adaptation, enhanced BDNF expression, suggesting a possible role of high-intensity short-term exercise in hippocampal BDNF activity.
Collapse
|
38
|
Gaiad TP, Oliveira MX, Lobo AR, Libório LR, Pinto PAF, Fernandes DC, Santos AP, Ambrósio CE, Machado ASD. Low-intensity training provokes adaptive extracellular matrix turnover of a muscular dystrophy model. J Exerc Rehabil 2017; 13:693-703. [PMID: 29326902 PMCID: PMC5747205 DOI: 10.12965/jer.1735094.547] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/05/2017] [Indexed: 11/22/2022] Open
Abstract
Recommendations of therapeutic exercise in Duchenne muscular dystrophy are still controversial. The hypothesis that a low-intensity training (LIT) protocol leads to muscle adaptations on mdx mice model was tested. Dystrophic male mice with 8 weeks old were separated in exercised (mdxE, n= 8) and sedentary (mdxC, n= 8) groups. Wild-type mice were used as control (WT, n= 8) group. Exercised group underwent a LIT protocol (9 m/min, 30 min, 3 days/wk, 60 days) on a horizontal treadmill. At day 60 all animals were analyzed regarding parameters of markers of muscle lesion and extracellular matrix turnover of muscle tissue by collagens fibers on tibial anterior muscle. Histomorphometry attested that centrally located nuclei fibers and the coefficient of variance of minimal Feret’s diameter was similar in mdxE and mdxC groups (P= 1.000) and both groups presented higher mean values than WT group (P< 0.001). Fraction area of collagen fibers of mdxE group was lower than mdxC group (P= 0,027) and similar to WT group (P= 0,751). Intramuscular area of Col3 of the mdxE group was higher than mdxC and WT groups (P<0.001). Intramuscular area of Col1 on the mdxE group was similar to the mdxC group (P= 1.000) and both groups were higher than WT group (P< 0.001). LIT protocol had not influenced muscle injuries resulting from the dystrophin-deficiency membrane fragility. Although, LIT had provoked adaptations on extracellular matrix bringing higher elastic feature to dystrophic muscle tissue.
Collapse
Affiliation(s)
- Thaís P Gaiad
- Department of Physical Therapy, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | - Murilo X Oliveira
- Department of Physical Therapy, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | - Adalfredo R Lobo
- Institute of Agriculture Scinces, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Unaí, Brazil
| | - Lívia R Libório
- Department of Physical Therapy, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | - Priscilla A F Pinto
- Department of Physical Therapy, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | - Danielle C Fernandes
- Department of Physical Therapy, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | - Ana Paula Santos
- Department of Physical Therapy, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | | | - Alex Sander D Machado
- Faculty of Medicine FAMED, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| |
Collapse
|
39
|
Seward T, Harfmann BD, Esser KA, Schroder EA. Reinventing the wheel: comparison of two wheel cage styles for assessing mouse voluntary running activity. J Appl Physiol (1985) 2017; 124:923-929. [PMID: 29357507 DOI: 10.1152/japplphysiol.00880.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Voluntary wheel cage assessment of mouse activity is commonly employed in exercise and behavioral research. Currently, no standardization for wheel cages exists resulting in an inability to compare results among data from different laboratories. The purpose of this study was to determine whether the distance run or average speed data differ depending on the use of two commonly used commercially available wheel cage systems. Two different wheel cages with structurally similar but functionally different wheels (electromechanical switch vs. magnetic switch) were compared side-by-side to measure wheel running data differences. Other variables, including enrichment and cage location, were also tested to assess potential impacts on the running wheel data. We found that cages with the electromechanical switch had greater inherent wheel resistance and consistently led to greater running distance per day and higher average running speed. Mice rapidly, within 1-2 days, adapted their running behavior to the type of experimental switch used, suggesting these running differences are more behavioral than due to intrinsic musculoskeletal, cardiovascular, or metabolic limits. The presence of enrichment or location of the cage had no detectable impact on voluntary wheel running. These results demonstrate that mice run differing amounts depending on the type of cage and switch mechanism used and thus investigators need to report wheel cage type/wheel resistance and use caution when interpreting distance/speed run across studies. NEW & NOTEWORTHY The results of this study highlight that mice will run different distances per day and average speed based on the inherent resistance present in the switch mechanism used to record data. Rapid changes in running behavior for the same mouse in the different cages demonstrate that a strong behavioral factor contributes to classic exercise outcomes in mice. Caution needs to be taken when interpreting mouse voluntary wheel running activity to include potential behavioral input and physiological parameters.
Collapse
Affiliation(s)
- T Seward
- Department of Physiology, University of Kentucky , Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky , Lexington, Kentucky
| | - B D Harfmann
- Department of Integrative Physiology and Health Science, Alma College, Alma, Michigan
| | - K A Esser
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida.,Institute of Myology, University of Florida , Gainesville, Florida
| | - E A Schroder
- Department of Physiology, University of Kentucky , Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky , Lexington, Kentucky
| |
Collapse
|
40
|
Schill KE, Altenberger AR, Lowe J, Periasamy M, Villamena FA, Rafael-Fortney JIA, Devor ST. Muscle damage, metabolism, and oxidative stress in mdx mice: Impact of aerobic running. Muscle Nerve 2017; 54:110-7. [PMID: 26659868 DOI: 10.1002/mus.25015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 12/08/2015] [Accepted: 12/10/2015] [Indexed: 01/10/2023]
Abstract
INTRODUCTION We tested how a treadmill exercise program influences oxygen consumption, oxidative stress, and exercise capacity in the mdx mouse, a model of Duchenne muscular dystrophy. METHODS At age 4 weeks mdx mice were subjected to 4 weeks of twice-weekly treadmill exercise. Sedentary mdx and wild-type mice served as controls. Oxygen consumption, time to exhaustion, oxidative stress, and myofiber damage were assessed. RESULTS At age 4 weeks, there was a significant difference in exercise capacity between mdx and wild-type mice. After exercise, mdx mice had lower basal oxygen consumption and exercise capacity, but similar maximal oxygen consumption. Skeletal muscle from these mice displayed increased oxidative stress. Collagen deposition was higher in exercised versus sedentary mice. CONCLUSIONS Exercised mdx mice exhibit increased oxidative stress, as well as deficits in exercise capacity, baseline oxygen consumption, and increased myofiber fibrosis. Muscle Nerve 54: 110-117, 2016.
Collapse
Affiliation(s)
- Kevin E Schill
- Department of Human Sciences, Kinesiology Program, The Ohio State University, Columbus, Ohio, USA
| | - Alex R Altenberger
- Department of Human Sciences, Kinesiology Program, The Ohio State University, Columbus, Ohio, USA
| | - Jeovanna Lowe
- Department of Molecular & Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Muthu Periasamy
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Frederick A Villamena
- Deparment of Pharmacology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - JIll A Rafael-Fortney
- Department of Molecular & Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Steven T Devor
- Department of Human Sciences, Kinesiology Program, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
41
|
Roemers P, Mazzola PN, De Deyn PP, Bossers WJ, van Heuvelen MJG, van der Zee EA. Burrowing as a novel voluntary strength training method for mice: A comparison of various voluntary strength or resistance exercise methods. J Neurosci Methods 2017; 300:112-126. [PMID: 28587894 DOI: 10.1016/j.jneumeth.2017.05.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/01/2017] [Accepted: 05/30/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Voluntary strength training methods for rodents are necessary to investigate the effects of strength training on cognition and the brain. However, few voluntary methods are available. NEW METHOD The current study tested functional and muscular effects of two novel voluntary strength training methods, burrowing (digging a substrate out of a tube) and unloaded tower climbing, in male C57Bl6 mice. To compare these two novel methods with existing exercise methods, resistance running and (non-resistance) running were included. Motor coordination, grip strength and muscle fatigue were measured at baseline, halfway through and near the end of a fourteen week exercise intervention. Endurance was measured by an incremental treadmill test after twelve weeks. RESULTS Both burrowing and resistance running improved forelimb grip strength as compared to controls. Running and resistance running increased endurance in the treadmill test and improved motor skills as measured by the balance beam test. Post-mortem tissue analyses revealed that running and resistance running induced Soleus muscle hypertrophy and reduced epididymal fat mass. Tower climbing elicited no functional or muscular changes. COMPARISON WITH EXISTING METHODS As a voluntary strength exercise method, burrowing avoids the confounding effects of stress and positive reinforcers elicited in forced strength exercise methods. Compared to voluntary resistance running, burrowing likely reduces the contribution of aerobic exercise components. CONCLUSIONS Burrowing qualifies as a suitable voluntary strength training method in mice. Furthermore, resistance running shares features of strength training and endurance (aerobic) exercise and should be considered a multi-modal aerobic-strength exercise method in mice.
Collapse
Affiliation(s)
- P Roemers
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands.
| | - P N Mazzola
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - P P De Deyn
- Department of Neurology and Alzheimer Research Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - W J Bossers
- Center for Human Movement Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M J G van Heuvelen
- Center for Human Movement Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - E A van der Zee
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
42
|
Grassi B, Majerczak J, Bardi E, Buso A, Comelli M, Chlopicki S, Guzik M, Mavelli I, Nieckarz Z, Salvadego D, Tyrankiewicz U, Skórka T, Bottinelli R, Zoladz JA, Pellegrino MA. Exercise training in Tgα q*44 mice during the progression of chronic heart failure: cardiac vs. peripheral (soleus muscle) impairments to oxidative metabolism. J Appl Physiol (1985) 2017; 123:326-336. [PMID: 28522765 DOI: 10.1152/japplphysiol.00342.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 12/16/2022] Open
Abstract
Cardiac function, skeletal (soleus) muscle oxidative metabolism, and the effects of exercise training were evaluated in a transgenic murine model (Tgαq*44) of chronic heart failure during the critical period between the occurrence of an impairment of cardiac function and the stage at which overt cardiac failure ensues (i.e., from 10 to 12 mo of age). Forty-eight Tgαq*44 mice and 43 wild-type FVB controls were randomly assigned to control groups and to groups undergoing 2 mo of intense exercise training (spontaneous running on an instrumented wheel). In mice evaluated at the beginning and at the end of training we determined: exercise performance (mean distance covered daily on the wheel); cardiac function in vivo (by magnetic resonance imaging); soleus mitochondrial respiration ex vivo (by high-resolution respirometry); muscle phenotype [myosin heavy chain (MHC) isoform content; citrate synthase (CS) activity]; and variables related to the energy status of muscle fibers [ratio of phosphorylated 5'-AMP-activated protein kinase (AMPK) to unphosphorylated AMPK] and mitochondrial biogenesis and function [peroxisome proliferative-activated receptor-γ coactivator-α (PGC-1α)]. In the untrained Tgαq*44 mice functional impairments of exercise performance, cardiac function, and soleus muscle mitochondrial respiration were observed. The impairment of mitochondrial respiration was related to the function of complex I of the respiratory chain, and it was not associated with differences in CS activity, MHC isoforms, p-AMPK/AMPK, and PGC-1α levels. Exercise training improved exercise performance and cardiac function, but it did not affect mitochondrial respiration, even in the presence of an increased percentage of type 1 MHC isoforms. Factors "upstream" of mitochondria were likely mainly responsible for the improved exercise performance.NEW & NOTEWORTHY Functional impairments in exercise performance, cardiac function, and soleus muscle mitochondrial respiration were observed in transgenic chronic heart failure mice, evaluated in the critical period between the occurrence of an impairment of cardiac function and the terminal stage of the disease. Exercise training improved exercise performance and cardiac function, but it did not affect the impaired mitochondrial respiration. Factors "upstream" of mitochondria, including an enhanced cardiovascular O2 delivery, were mainly responsible for the functional improvement.
Collapse
Affiliation(s)
- Bruno Grassi
- Department of Medicine, University of Udine, Udine, Italy; .,Institute of Bioimaging and Molecular Physiology, National Research Council, Milan, Italy
| | - Joanna Majerczak
- Department of Muscle Physiology, Faculty of Rehabilitation, University School of Physical Education, Krakow, Poland
| | - Eleonora Bardi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Alessia Buso
- Department of Medicine, University of Udine, Udine, Italy
| | - Marina Comelli
- Department of Medicine, University of Udine, Udine, Italy
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University Medical College, Krakow, Poland.,Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Guzik
- Department of Muscle Physiology, Faculty of Rehabilitation, University School of Physical Education, Krakow, Poland
| | - Irene Mavelli
- Department of Medicine, University of Udine, Udine, Italy
| | - Zenon Nieckarz
- Institute of Physics, Jagiellonian University, Krakow, Poland; and
| | - Desy Salvadego
- Department of Medicine, University of Udine, Udine, Italy
| | - Urszula Tyrankiewicz
- Department of Magnetic Resonance Imaging, Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | - Tomasz Skórka
- Department of Magnetic Resonance Imaging, Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | | | - Jerzy A Zoladz
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University Medical College, Krakow, Poland
| | | |
Collapse
|
43
|
Ward LM, Kinnett K, Bonewald L. Proceedings of a Parent Project Muscular Dystrophy Bone Health Workshop: Morbidity due to osteoporosis in DMD: The Path Forward May 12-13, 2016, Bethesda, Maryland, USA. Neuromuscul Disord 2017; 28:64-76. [PMID: 28756052 DOI: 10.1016/j.nmd.2017.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/04/2017] [Accepted: 05/11/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Leanne M Ward
- Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada.
| | - Kathi Kinnett
- Parent Project Muscular Dystrophy, Middletown, OH, USA
| | - Lynda Bonewald
- Indiana Center for Musculoskeletal Health, Departments of Anatomy and Cell Biology and Orthopaedic Surgery, Indiana University, Indianapolis, IN, USA
| | | |
Collapse
|
44
|
Morici G, Frinchi M, Pitruzzella A, Di Liberto V, Barone R, Pace A, Di Felice V, Belluardo N, Cappello F, Mudò G, Bonsignore MR. Mild Aerobic Exercise Training Hardly Affects the Diaphragm of mdx Mice. J Cell Physiol 2017; 232:2044-2052. [PMID: 27576008 DOI: 10.1002/jcp.25573] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/29/2016] [Indexed: 12/22/2022]
Abstract
In the mdx mice model of Duchenne Muscular Dystrophy (DMD), mild endurance exercise training positively affected limb skeletal muscles, whereas few and controversial data exist on the effects of training on the diaphragm. The diaphragm was examined in mdx (C57BL/10ScSn-Dmdmdx) and wild-type (WT, C57BL/10ScSc) mice under sedentary conditions (mdx-SD, WT-SD) and during mild exercise training (mdx-EX, WT-EX). At baseline, and after 30 and 45 days (training: 5 d/wk for 6 weeks), diaphragm muscle morphology and Cx39 protein were assessed. In addition, tissue levels of the chaperonins Hsp60 and Hsp70 and the p65 subunit of nuclear factor-kB (NF-kB) were measured in diaphragm, gastrocnemius, and quadriceps in each experimental group at all time points. Although morphological analysis showed unchanged total area of necrosis/regeneration in the diaphragm after training, there was a trend for larger areas of regeneration than necrosis in the diaphragm of mdx-EX compared to mdx-SD mice. However, the levels of Cx39, a protein associated with active regeneration in damaged muscle, were similar in the diaphragm of mdx-EX and mdx-SD mice. Hsp60 significantly decreased at 45 days in the diaphragm, but not in limb muscles, in both trained and sedentary mdx compared to WT mice. In limb muscles, but not in the diaphragm, Hsp70 and NF-kB p65 levels were increased in mdx mice irrespective of training at 30 and 45 days. Therefore, the diaphragm of mdx mice showed little inflammatory and stress responses over time, and appeared hardly affected by mild endurance training. J. Cell. Physiol. 232: 2044-2052, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Giuseppe Morici
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy.,Istituto di Biomedicina e Immunologia Molecolare (IBIM), Consiglio Nazionale Delle Ricerche (CNR), Palermo, Italy
| | - Monica Frinchi
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy
| | - Alessandro Pitruzzella
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy.,Istituto Euro-Mediterraneo di Scienza e Tecnologia, Palermo, Italy
| | - Valentina Di Liberto
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy
| | - Rosario Barone
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy.,Istituto Euro-Mediterraneo di Scienza e Tecnologia, Palermo, Italy
| | - Andrea Pace
- Istituto Euro-Mediterraneo di Scienza e Tecnologia, Palermo, Italy.,Dipartimento di Scienze e Tecnologie Molecolari e Biomolecolari (STEMBIO)-University of Palermo, Palermo, Italy
| | - Valentina Di Felice
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy.,Istituto Euro-Mediterraneo di Scienza e Tecnologia, Palermo, Italy
| | - Natale Belluardo
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy
| | - Francesco Cappello
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy.,Istituto Euro-Mediterraneo di Scienza e Tecnologia, Palermo, Italy
| | - Giuseppa Mudò
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (BioNeC), University of Palermo, Palermo, Italy
| | - Maria R Bonsignore
- Istituto di Biomedicina e Immunologia Molecolare (IBIM), Consiglio Nazionale Delle Ricerche (CNR), Palermo, Italy.,Dipartimento Biomedico di Medicina Interna e Specialistica (DiBiMIS), University of Palermo, Palermo, Italy
| |
Collapse
|
45
|
Hyzewicz J, Tanihata J, Kuraoka M, Nitahara-Kasahara Y, Beylier T, Ruegg UT, Vater A, Takeda S. Low-Intensity Training and the C5a Complement Antagonist NOX-D21 Rescue the mdx Phenotype through Modulation of Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1147-1161. [PMID: 28315675 DOI: 10.1016/j.ajpath.2016.12.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 12/19/2016] [Accepted: 12/27/2016] [Indexed: 12/17/2022]
Abstract
Inflammatory events occurring in dystrophic muscles contribute to the progression of Duchenne muscular dystrophy (DMD). Low-intensity training (LIT) attenuates the phenotype of mdx mice, an animal model for DMD. Therefore, we postulated that LIT could have anti-inflammatory properties. We assessed levels of inflammatory cytokines and infiltrated immune cells in gastrocnemius muscle of mdx mice after LIT. We detected high levels of complement component C5a, chemokine ligand (CCL) 2, CD68+ monocytes/macrophages, and proinflammatory M1 macrophages in muscles of mdx mice. LIT decreased CCL2 levels, increased CD68+ cell numbers, and shifted the macrophage population to the regenerative M2 type. We investigated whether inhibition of C5a or CCL2 with L-aptamers could mimic the effects of LIT. Although no effect of CCL2 inhibition was detected, treatment with the C5a inhibitor, NOX-D21, rescued the phenotype of nonexercised mdx mice, but not of exercised ones. In both cases, the level of CD68+ cells increased and macrophage populations leaned toward the inflammatory M1 type. In muscles of nonexercised treated mice, the level of IL-1 receptor antagonist increased, damage decreased, and fibers were switched toward the glycolytic fast type; in muscles of exercised mice, fibers were switched to the oxidative slow type. These results reveal the effects of LIT on the inflammatory status of mdx mice and suggest that NOX-D21 could be an anti-inflammatory drug for DMD.
Collapse
Affiliation(s)
- Janek Hyzewicz
- Department of Molecular Therapy, National Institute of Neuroscience, National Centre of Neurology and Psychiatry, Tokyo, Japan
| | - Jun Tanihata
- Department of Molecular Therapy, National Institute of Neuroscience, National Centre of Neurology and Psychiatry, Tokyo, Japan
| | - Mutsuki Kuraoka
- Department of Molecular Therapy, National Institute of Neuroscience, National Centre of Neurology and Psychiatry, Tokyo, Japan
| | - Yuko Nitahara-Kasahara
- Department of Molecular Therapy, National Institute of Neuroscience, National Centre of Neurology and Psychiatry, Tokyo, Japan
| | - Teiva Beylier
- Department of Molecular Therapy, National Institute of Neuroscience, National Centre of Neurology and Psychiatry, Tokyo, Japan
| | - Urs T Ruegg
- Laboratory of Pharmacology, University of Geneva, Geneva, Switzerland
| | - Axel Vater
- Drug Discovery and Preclinical Development, NOXXON Pharma, Berlin, Germany
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Centre of Neurology and Psychiatry, Tokyo, Japan.
| |
Collapse
|
46
|
Southern WM, Nichenko AS, Shill DD, Spencer CC, Jenkins NT, McCully KK, Call JA. Skeletal muscle metabolic adaptations to endurance exercise training are attainable in mice with simvastatin treatment. PLoS One 2017; 12:e0172551. [PMID: 28207880 PMCID: PMC5313210 DOI: 10.1371/journal.pone.0172551] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/06/2017] [Indexed: 01/02/2023] Open
Abstract
We tested the hypothesis that a 6-week regimen of simvastatin would attenuate skeletal muscle adaptation to low-intensity exercise. Male C57BL/6J wildtype mice were subjected to 6-weeks of voluntary wheel running or normal cage activities with or without simvastatin treatment (20 mg/kg/d, n = 7-8 per group). Adaptations in in vivo fatigue resistance were determined by a treadmill running test, and by ankle plantarflexor contractile assessment. The tibialis anterior, gastrocnemius, and plantaris muscles were evaluated for exercised-induced mitochondrial adaptations (i.e., biogenesis, function, autophagy). There was no difference in weekly wheel running distance between control and simvastatin-treated mice (P = 0.51). Trained mice had greater treadmill running distance (296%, P<0.001), and ankle plantarflexor contractile fatigue resistance (9%, P<0.05) compared to sedentary mice, independent of simvastatin treatment. At the cellular level, trained mice had greater mitochondrial biogenesis (e.g., ~2-fold greater PGC1α expression, P<0.05) and mitochondrial content (e.g., 25% greater citrate synthase activity, P<0.05), independent of simvastatin treatment. Mitochondrial autophagy-related protein contents were greater in trained mice (e.g., 40% greater Bnip3, P<0.05), independent of simvastatin treatment. However, Drp1, a marker of mitochondrial fission, was less in simvastatin treated mice, independent of exercise training, and there was a significant interaction between training and statin treatment (P<0.022) for LC3-II protein content, a marker of autophagy flux. These data indicate that whole body and skeletal muscle adaptations to endurance exercise training are attainable with simvastatin treatment, but simvastatin may have side effects on muscle mitochondrial maintenance via autophagy, which could have long-term implications on muscle health.
Collapse
Affiliation(s)
- William M. Southern
- Department of Kinesiology, University of Georgia, Athens, Georgia, United States of America
| | - Anna S. Nichenko
- Department of Kinesiology, University of Georgia, Athens, Georgia, United States of America
| | - Daniel D. Shill
- Department of Kinesiology, University of Georgia, Athens, Georgia, United States of America
| | - Corey C. Spencer
- Department of Kinesiology, University of Georgia, Athens, Georgia, United States of America
| | - Nathan T. Jenkins
- Department of Kinesiology, University of Georgia, Athens, Georgia, United States of America
| | - Kevin K. McCully
- Department of Kinesiology, University of Georgia, Athens, Georgia, United States of America
| | - Jarrod A. Call
- Department of Kinesiology, University of Georgia, Athens, Georgia, United States of America
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
47
|
Puolakkainen T, Ma H, Kainulainen H, Pasternack A, Rantalainen T, Ritvos O, Heikinheimo K, Hulmi JJ, Kiviranta R. Treatment with soluble activin type IIB-receptor improves bone mass and strength in a mouse model of Duchenne muscular dystrophy. BMC Musculoskelet Disord 2017; 18:20. [PMID: 28103859 PMCID: PMC5244551 DOI: 10.1186/s12891-016-1366-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/14/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Inhibition of activin/myostatin pathway has emerged as a novel approach to increase muscle mass and bone strength. Duchenne muscular dystrophy (DMD) is a neuromuscular disorder that leads to progressive muscle degeneration and also high incidence of fractures. The aim of our study was to test whether inhibition of activin receptor IIB ligands with or without exercise could improve bone strength in the mdx mouse model for DMD. METHODS Thirty-two mdx mice were divided to running and non-running groups and to receive either PBS control or soluble activin type IIB-receptor (ActRIIB-Fc) once weekly for 7 weeks. RESULTS Treatment of mdx mice with ActRIIB-Fc resulted in significantly increased body and muscle weights in both sedentary and exercising mice. Femoral μCT analysis showed increased bone volume and trabecular number (BV/TV +80%, Tb.N +70%, P < 0.05) in both ActRIIB-Fc treated groups. Running also resulted in increased bone volume and trabecular number in PBS-treated mice. However, there was no significant difference in trabecular bone structure or volumetric bone mineral density between the ActRIIB-Fc and ActRIIB-Fc-R indicating that running did not further improve bone structure in ActRIIB-Fc-treated mice. ActRIIB-Fc increased bone mass also in vertebrae (BV/TV +20%, Tb.N +30%, P < 0.05) but the effects were more modest. The number of osteoclasts was decreased in histological analysis and the expression of several osteoblast marker genes was increased in ActRIIB-Fc treated mice suggesting decreased bone resorption and increased bone formation in these mice. Increased bone mass in femurs translated into enhanced bone strength in biomechanical testing as the maximum force and stiffness were significantly elevated in ActRIIB-Fc-treated mice. CONCLUSIONS Our results indicate that treatment of mdx mice with the soluble ActRIIB-Fc results in a robust increase in bone mass, without any additive effect by voluntary running. Thus ActRIIB-Fc could be an attractive option in the treatment of musculoskeletal disorders.
Collapse
Affiliation(s)
- Tero Puolakkainen
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Hongqian Ma
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland.,Institute of Dentistry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Heikki Kainulainen
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| | - Arja Pasternack
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Timo Rantalainen
- Centre for Physical Activity and Nutrition Research, Deakin University, Melbourne, Australia
| | - Olli Ritvos
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Kristiina Heikinheimo
- Department of Oral and Maxillofacial Surgery, Institute of Dentistry, University of Turku, Turku, Finland.,Department of Oral Diagnostic Sciences, Institute of Dentistry, University of Eastern Finland, Kuopio, Finland.,Kuopio University Hospital, Kuopio, Finland
| | - Juha J Hulmi
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riku Kiviranta
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland. .,Department of Endocrinology, Turku University Hospital, Turku, Finland.
| |
Collapse
|
48
|
Greising SM, Dearth CL, Corona BT. Regenerative and Rehabilitative Medicine: A Necessary Synergy for Functional Recovery from Volumetric Muscle Loss Injury. Cells Tissues Organs 2016; 202:237-249. [PMID: 27825146 DOI: 10.1159/000444673] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2016] [Indexed: 01/04/2023] Open
Abstract
Volumetric muscle loss (VML) is a complex and heterogeneous problem due to significant traumatic or surgical loss of skeletal muscle tissue. The consequences of VML are substantial functional deficits in joint range of motion and skeletal muscle strength, resulting in life-long dysfunction and disability. Traditional physical medicine and rehabilitation paradigms do not address the magnitude of force loss due to VML and related musculoskeletal comorbidities. Recent advancements in regenerative medicine have set forth encouraging and emerging therapeutic options for VML injuries. There is significant potential that combined rehabilitative and regenerative therapies can restore limb and muscle function following VML injury in a synergistic manner. This review presents the current state of the VML field, spanning clinical and preclinical literature, with particular focus on rehabilitation and regenerative medicine in addition to their synergy. Moving forward, multidisciplinary collaboration between clinical and research fields is encouraged in order to continue to improve the treatment of VML injuries and specifically address the encompassing physiology, pathology, and specific needs of this patient population. 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
|
49
|
Foltz SJ, Luan J, Call JA, Patel A, Peissig KB, Fortunato MJ, Beedle AM. Four-week rapamycin treatment improves muscular dystrophy in a fukutin-deficient mouse model of dystroglycanopathy. Skelet Muscle 2016; 6:20. [PMID: 27257474 PMCID: PMC4890530 DOI: 10.1186/s13395-016-0091-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/04/2016] [Indexed: 12/13/2022] Open
Abstract
Background Secondary dystroglycanopathies are a subset of muscular dystrophy caused by abnormal glycosylation of α-dystroglycan (αDG). Loss of αDG functional glycosylation prevents it from binding to laminin and other extracellular matrix receptors, causing muscular dystrophy. Mutations in a number of genes, including FKTN (fukutin), disrupt αDG glycosylation. Methods We analyzed conditional Fktn knockout (Fktn KO) muscle for levels of mTOR signaling pathway proteins by Western blot. Two cohorts of Myf5-cre/Fktn KO mice were treated with the mammalian target of rapamycin (mTOR) inhibitor rapamycin (RAPA) for 4 weeks and evaluated for changes in functional and histopathological features. Results Muscle from 17- to 25-week-old fukutin-deficient mice has activated mTOR signaling. However, in tamoxifen-inducible Fktn KO mice, factors related to Akt/mTOR signaling were unchanged before the onset of dystrophic pathology, suggesting that Akt/mTOR signaling pathway abnormalities occur after the onset of disease pathology and are not causative in early dystroglycanopathy development. To determine any pharmacological benefit of targeting mTOR signaling, we administered RAPA daily for 4 weeks to Myf5/Fktn KO mice to inhibit mTORC1. RAPA treatment reduced fibrosis, inflammation, activity-induced damage, and central nucleation, and increased muscle fiber size in Myf5/Fktn KO mice compared to controls. RAPA-treated KO mice also produced significantly higher torque at the conclusion of dosing. Conclusions These findings validate a misregulation of mTOR signaling in dystrophic dystroglycanopathy skeletal muscle and suggest that such signaling molecules may be relevant targets to delay and/or reduce disease burden in dystrophic patients. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0091-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Steven J Foltz
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W. Green St., Athens, GA 30602 USA
| | - Junna Luan
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W. Green St., Athens, GA 30602 USA
| | - Jarrod A Call
- Department of Kinesiology, University of Georgia, Athens, GA 30602 USA ; Regenerative Bioscience Center, University of Georgia, Athens, GA 30602 USA
| | - Ankit Patel
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W. Green St., Athens, GA 30602 USA
| | - Kristen B Peissig
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W. Green St., Athens, GA 30602 USA
| | - Marisa J Fortunato
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W. Green St., Athens, GA 30602 USA
| | - Aaron M Beedle
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W. Green St., Athens, GA 30602 USA
| |
Collapse
|
50
|
Morici G, Rappa F, Cappello F, Pace E, Pace A, Mudò G, Crescimanno G, Belluardo N, Bonsignore MR. Lack of Dystrophin Affects Bronchial Epithelium inmdxMice. J Cell Physiol 2016; 231:2218-23. [DOI: 10.1002/jcp.25339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 02/09/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Giuseppe Morici
- Dipartimento di Biomedicina e Neuroscienze Cliniche (BioNeC); University of Palermo; Palermo Sicilia Italy
- Istituto di Biomedicina e Immunologia Molecolare (IBIM); Consiglio Nazionale delle Ricerche (CNR); Palermo Sicilia Italy
| | - Francesca Rappa
- Dipartimento di Biomedicina e Neuroscienze Cliniche (BioNeC); University of Palermo; Palermo Sicilia Italy
- Dipartimento di Scienze Giuridiche della Società e dello Sport; University of Palermo; Palermo Sicilia Italy
- Istituto Euro-Mediterraneo di Scienza e Tecnologia; Palermo Sicilia Italy
| | - Francesco Cappello
- Dipartimento di Biomedicina e Neuroscienze Cliniche (BioNeC); University of Palermo; Palermo Sicilia Italy
- Istituto Euro-Mediterraneo di Scienza e Tecnologia; Palermo Sicilia Italy
| | - Elisabetta Pace
- Istituto di Biomedicina e Immunologia Molecolare (IBIM); Consiglio Nazionale delle Ricerche (CNR); Palermo Sicilia Italy
| | - Andrea Pace
- Istituto Euro-Mediterraneo di Scienza e Tecnologia; Palermo Sicilia Italy
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF); University of Palermo; Palermo Sicilia Italy
| | - Giuseppa Mudò
- Dipartimento di Biomedicina e Neuroscienze Cliniche (BioNeC); University of Palermo; Palermo Sicilia Italy
| | - Grazia Crescimanno
- Istituto di Biomedicina e Immunologia Molecolare (IBIM); Consiglio Nazionale delle Ricerche (CNR); Palermo Sicilia Italy
| | - Natale Belluardo
- Dipartimento di Biomedicina e Neuroscienze Cliniche (BioNeC); University of Palermo; Palermo Sicilia Italy
| | - Maria R. Bonsignore
- Istituto di Biomedicina e Immunologia Molecolare (IBIM); Consiglio Nazionale delle Ricerche (CNR); Palermo Sicilia Italy
- Dipartimento Biomedico di Medicina Interna e Specialistica (DiBiMIS); University of Palermo; Palermo Sicilia Italy
| |
Collapse
|