Skip to main content

EDITORIAL article

Front. Physiol., 17 October 2022
Sec. Striated Muscle Physiology
This article is part of the Research Topic Skeletal Muscle in Age-Related Diseases: From Molecular Pathogenesis to Potential Interventions View all 5 articles

Editorial: Skeletal muscle in age-related diseases: From molecular pathogenesis to potential interventions

  • 1Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, United States
  • 2Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates

Introduction

Skeletal muscle is a dynamic organ that responds to various stimuli by altering its size, strength, and composition. Maintenance of skeletal muscle mass and strength is critical for well-being and overall health. Conversely, ageing, and age-related diseases are associated with the loss of muscle mass and strength, termed sarcopenia. The exact molecular mechanisms of sarcopenia are poorly understood, which hamper the development of effective interventions to boost muscle mass and strength in ageing.

Sarcopenia is accompanied by functional disability and decreased quality of life. Skeletal muscle exhibits several characteristic alterations in sarcopenia, including diminished mitochondrial respiratory activity (Short et al., 2005) contractile dysfunction (Joseph et al., 2012; Grevendonk et al., 2021) elevated oxidative stress (Meng and Yu, 2010), reduced satellite cell count (Zwetsloot et al., 2013), senescence phenotype (Saito and Chikenji, 2021), loss of motor units (Piasecki et al., 2016), neuromuscular junction degradation (Karim et al., 2022), dysregulated calcium homeostasis (Qaisar et al., 2020), and hormonal changes (Qaisar et al., 2013).

Sarcopenia may be physiological and accompanied by healthy ageing without comorbidities. Nevertheless, sarcopenia is associated with alterations in skeletal muscle gene expression involving damage accumulation and compensatory mechanisms. An ambitious study by Tumasian III et al. investigated this question by extracting RNAs from muscle biopsies of healthy individuals over a wide age range. Altogether, 1,134 RNAs changed significantly with advancing age out of the 57,205 protein-coding and non-coding RNAs (Tumasian et al., 2021).

After its onset, sarcopenia is difficult to treat because it involves complex molecular interactions and is a risk factor for developing other diseases. Consequently, developing pharmaceutical strategies for sarcopenia remains a critical challenge due to multifactorial etiology and interface of skeletal muscle with multiple organs in the context of ageing. In our Research Topic “Skeletal Muscle in Age-Related Diseases: From Molecular Pathogenesis to Potential Interventions” several studies address important questions in the field as well as examine potential molecular targets to treat ageing skeletal muscle.

During skeletal muscle ageing, mitochondrial dysfunction contributes to reactive oxygen species (ROS)-induced oxidative damage (Qaisar et al., 2018), while exercise can improve mitochondrial function and reduce oxidative stress (Holloszy, 1967). However, this therapeutic effect of exercise is gradually blunted with advancing age, partly due to limiting exercise capacity and related health constraints. Therefore, understanding the molecular underpinnings of oxidative stress pathways and developing strategies to circumvent them may provide new bedside therapeutic options that are not reliant on exercise alone.

Ruan et al. examined the role of the long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), which was found to have decreased expression in aged skeletal muscle. Previous studies have suggested that changes in lncRNA expression can influence muscle growth and differentiation by incompletely understood mechanisms (Butchart et al., 2016). While it is known that lncRNAs can function as sponges that regulate levels of free microRNAs (miRNAs), the study by Ruan et al. examined their role in the less understood realm of muscle ageing. Previously the authors showed that heightened ROS production associated with ageing is accompanied by increased senescence-associated miRNA-34a-5p levels (Fulzele et al., 2019). In the current study, skeletal muscles obtained from young and aged mice of both sexes were examined for reciprocal changes in MALAT1 and miR-34a-5p expression. In-vitro studies in C2C12 myoblasts demonstrated that H2O2 decreased MALAT1 expression, while siRNA-mediated MALAT1 knock-down significantly increased TGF-β1 expression. The crosstalk or interplay between oxidative conditions/MALAT1/miR-34a may represent a missing link connecting ageing phenomenon in skeletal muscle with the development of fibrosis and cellular senescence. Studies such as this may unlock new potential age-related therapeutic targets to counter the deterioration of muscle health.

The study by You and Chen directly addressed the effect of age-associated motor neuron loss on the development of sarcopenia. The authors observed that aged mice with obvious signs of sarcopenia were better protected from accelerated denervation-induced muscle atrophy seen in much younger mice. The overall skeletal mass is dictated by factors that determine the net balance between the availability of its building blocks. e.g., protein synthesis (anabolic) versus degradation (catabolic). Interestingly, in aged mice, the tibialis anterior (TA) muscle displayed resistance to denervation-induced atrophy. It was found that aging promotes protein synthesis rates and ribosomal RNA (rRNA) biogenesis in denervated TA, which is independent of mTORC1 regulation but via enhanced signaling through the Akt-GSK-3β pathway.

Another line of thought in treating sarcopenia in aged individuals is replacing molecular factors lost during aging. While iTRAQ analysis yielded a lengthy list of peptide changes, the study by Lo et al. cut to the chase and first examined whether fetal skeletal muscle extract could benefit muscle health of aged mice. After 8 weeks of treatment, the aged mice showed enhanced muscle performance with increased lean mass, heightened grip strength, and isometric force generation. While the authors have not yet identified the factors in the fetal serum providing beneficial effects on aged muscle, the study is intriguing. Further analysis may yield one or multiple curative factors that can be replaced by direct injection or other administration methods to treat sarcopenia.

Exercise training accompanied by nutritional approaches remains the best non-pharmaceutical treatment strategy to counter sarcopenia. However, in the elderly or immobilized, these may not be plausible treatment strategies. Falqueto et al. highlight the pros and cons of using anabolic-androgenic steroids to increase muscle function and mass. Sarcopenia is driven by diverse pathological processes in ageing patients, including elevated muscle catabolism than anabolism. The combination of anabolic hormones and exercise training can reverse muscle wasting in aged individuals. However, in some studies, the type of exercise mattered—with resistance training yielding favorable results by minimizing fatigue and exercise intolerance while increasing muscle strength, hypertrophy, and power. Although these above outcomes appear favorable, the discernable observed may depend on differences in participant exclusion criteria. Overall, it is promising that administration of anabolic hormones to some patients bolstered the exercise effect on sarcopenia, at least in some patients.

Experimental studies highlighted in this edition provide new and provocative findings that represent novel targets and potential as alternative therapies for sarcopenia. These studies further our understanding of the role of lncRNAs in protecting against muscle senescence, the effect of innervation and ageing, and beneficial properties of fetal muscle extract in sarcopenic muscle.

Author contributions

MP and RQ drafted and edited the editorial.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Butchart L. C., Fox A., Shavlakadze T., Grounds M. D. (2016). The long and short of non-coding RNAs during post-natal growth and differentiation of skeletal muscles: Focus on lncRNA and miRNAs. Differentiation. 92, 237–248. doi:10.1016/j.diff.2016.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Fulzele S., Mendhe B., Khayrullin A., Johnson M., Kaiser H., Liu Y., et al. (2019). Muscle-derived miR-34a increases with age in circulating extracellular vesicles and induces senescence of bone marrow stem cells. Aging (Albany NY) 11, 1791–1803. doi:10.18632/aging.101874

PubMed Abstract | CrossRef Full Text | Google Scholar

Grevendonk L., Connell N. J., McCrum C., Fealy C. E., Bilet L., Bruls Y. M. H., et al. (2021). Impact of aging and exercise on skeletal muscle mitochondrial capacity, energy metabolism, and physical function. Nat. Commun. 12, 4773. doi:10.1038/s41467-021-24956-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Holloszy J. O. (1967). Biochemical adaptations in muscle. J. Biol. Chem. 242, 2278–2282. doi:10.1016/s0021-9258(18)96046-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Joseph A. M., Adhihetty P. J., Buford T. W., Wohlgemuth S. E., Lees H. A., Nguyen L. M., et al. (2012). The impact of aging on mitochondrial function and biogenesis pathways in skeletal muscle of sedentary high- and low-functioning elderly individuals. Aging Cell 11, 801–809. doi:10.1111/j.1474-9726.2012.00844.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Karim A., Iqbal M. S., Muhammad T., Qaisar R. (2022). Evaluation of sarcopenia using biomarkers of the neuromuscular junction in Parkinson's disease. J. Mol. Neurosci. 72, 820–829. doi:10.1007/s12031-022-01970-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Meng S. J., Yu L. J. (2010). Oxidative stress, molecular inflammation and sarcopenia. Int. J. Mol. Sci. 11, 1509–1526. doi:10.3390/ijms11041509

PubMed Abstract | CrossRef Full Text | Google Scholar

Nelke C., Dziewas R., Minnerup J., Meuth S. G., Ruck T. (2019). Skeletal muscle as potential central link between sarcopenia and immune senescence. EBioMedicine 49, 381–388. doi:10.1016/j.ebiom.2019.10.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Piasecki M., Ireland A., Stashuk D., Hamilton-Wright A., Jones D. A., McPhee J. S., et al. (2016). Age-related neuromuscular changes affecting human vastus lateralis. J. Physiol. 594, 4525–4536. doi:10.1113/JP271087

PubMed Abstract | CrossRef Full Text | Google Scholar

Qaisar R., Renaud G., Hedstrom Y., Pollanen E., Ronkainen P., Kaprio J., et al. (2013). Hormone replacement therapy improves contractile function and myonuclear organization of single muscle fibres from postmenopausal monozygotic female twin pairs. J. Physiol. 591, 2333–2344. doi:10.1113/jphysiol.2012.250092

PubMed Abstract | CrossRef Full Text | Google Scholar

Qaisar R., Bhaskaran S., Premkumar P., Ranjit R., Natarajan K. S., Ahn B., et al. (2018). Oxidative stress-induced dysregulation of excitation-contraction coupling contributes to muscle weakness. J. Cachexia Sarcopenia Muscle 9, 1003–1017. doi:10.1002/jcsm.12339

PubMed Abstract | CrossRef Full Text | Google Scholar

Qaisar R., Pharaoh G., Bhaskaran S., Xu H., Ranjit R., Bian J., et al. (2020). Restoration of sarcoplasmic reticulum Ca(2+) ATPase (SERCA) activity prevents age-related muscle atrophy and weakness in mice. Int. J. Mol. Sci. 22, E37. doi:10.3390/ijms22010037

PubMed Abstract | CrossRef Full Text | Google Scholar

Saito Y., Chikenji T. S. (2021). Diverse roles of cellular senescence in skeletal muscle inflammation, regeneration, and therapeutics. Front. Pharmacol. 12, 739510. doi:10.3389/fphar.2021.739510

PubMed Abstract | CrossRef Full Text | Google Scholar

Short K. R., Bigelow M. L., Kahl J., Singh R., Coenen-Schimke J., Raghavakaimal S., et al. (2005). Decline in skeletal muscle mitochondrial function with aging in humans. Proc. Natl. Acad. Sci. U. S. A. 102, 5618–5623. doi:10.1073/pnas.0501559102

PubMed Abstract | CrossRef Full Text | Google Scholar

Tumasian R. A., Harish A., Kundu G., Yang J. H., Ubaida-Mohien C., Gonzalez-Freire M., et al. (2021). Skeletal muscle transcriptome in healthy aging. Nat. Commun. 12, 2014. doi:10.1038/s41467-021-22168-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Zwetsloot K. A., Childs T. E., Gilpin L. T., Booth F. W. (2013). Non-passaged muscle precursor cells from 32-month old rat skeletal muscle have delayed proliferation and differentiation. Cell Prolif. 46, 45–57. doi:10.1111/cpr.12007

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: sarcopenia, skeletal muscle, aging, oxidative stress, senescence

Citation: Parvatiyar MS and Qaisar R (2022) Editorial: Skeletal muscle in age-related diseases: From molecular pathogenesis to potential interventions. Front. Physiol. 13:1056479. doi: 10.3389/fphys.2022.1056479

Received: 28 September 2022; Accepted: 05 October 2022;
Published: 17 October 2022.

Edited and reviewed by:

Paul M. L. Janssen, The Ohio State University, United States

Copyright © 2022 Parvatiyar and Qaisar. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Rizwan Qaisar, rqaisar@sharjah.ac.ae

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.