bims-moremu Biomed News
on Molecular regulators of muscle mass
Issue of 2026–01–04
thirty-one papers selected by
Anna Vainshtein, Craft Science Inc.



  1. Autophagy. 2025 Dec 31. 1-2
      Macroautophagy/autophagy protects muscle from proteotoxic stress and maintains tissue homeostasis, yet skeletal muscle relies on it more than most organs. Adult fibers endure constant mechanical strain and require continuous turnover of long-lived proteins, while muscle stem cells (MuSCs) depend on autophagy to remain quiescent, activate after injury, and regenerate effectively. How autophagy is transcriptionally regulated in muscle has been unclear. We identified DEAF1 as a transcriptional brake on autophagy. In MuSCs, DEAF1 controls activation and regeneration and becomes aberrantly elevated with age, promoting protein aggregate formation and cell death. In muscle fibers, DEAF1 is chronically induced during aging, suppressing autophagy and driving functional decline. Exercise reverses DEAF1 induction, restoring autophagy and muscle function. These findings reveal DEAF1 as a key regulator linking autophagy to regeneration and aging, highlighting a therapeutically tractable axis for preserving muscle health.
    Keywords:  Autophagy; DEAF1; muscle; muscle stem cell; regeneration
    DOI:  https://doi.org/10.1080/15548627.2025.2610451
  2. Biochimie. 2025 Dec 29. pii: S0300-9084(25)00317-7. [Epub ahead of print]
      Skeletal muscle is fundamental to human health, serving as the primary effector of movement and a central regulator of systemic metabolism. Age-related declines in muscle mass and mitochondrial function contribute to frailty, metabolic dysfunction, and loss of independence in older adults. While these changes are often attributed to reduced physical activity, chronic inflammation, and impaired regenerative capacity, emerging evidence implicates environmental and metabolic sensing pathways in muscle degeneration. The aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor best known for mediating responses to environmental pollutants such as dioxins, has recently been recognized as a key regulator of endogenous metabolic and redox processes. AHR activation occurs not only through xenobiotic exposure but also via endogenous ligands derived from tryptophan metabolism-including kynurenine and indole derivatives-whose levels rise in aging, chronic kidney disease (CKD), and other pollutant exposures. Sustained AHR activation in skeletal muscle has been shown to impair mitochondrial oxidative phosphorylation, promote proteolysis, and disrupt neuromuscular junction integrity, linking AHR signaling to muscle pathology. Experimental studies in rodent models demonstrate that pharmacologic or genetic inhibition of AHR can preserve muscle mass, mitochondrial function, and regenerative capacity. This review summarizes the molecular biology of the AHR, its emerging roles in skeletal muscle physiology and pathology, and the growing experimental toolkit for interrogating its function. Understanding how AHR signaling integrates environmental, metabolic, and aging cues may reveal new therapeutic opportunities to preserve skeletal muscle health and physical function across the lifespan.
    Keywords:  AHR; aging; frailty; hydrocarbon; muscle
    DOI:  https://doi.org/10.1016/j.biochi.2025.12.012
  3. Cell Commun Signal. 2025 Dec 29. 23(1): 535
      Quiescence - the reversible growth-arrested G0 state - is critical for the long-term maintenance of many adult stem cells and hence the tissues' life-long regenerative capacity. Failure to maintain stem cell quiescence during aging leads to stem cell loss and reduced ability to regenerate after injury. However, our knowledge about the complex molecular signaling networks required for maintaining stem cells in quiescence is incomplete. Here, we discovered an essential role for FGFR1 and FGFR4 in regulating skeletal muscle stem cell, i.e. satellite cell (SC), quiescence via inhibiting their precocious differentiation. Moreover, SC-specific Fgfr1/4-deficiency results in impaired muscle regeneration and decreased SC self-renewal. Our data reveal that the activated Fgfr1/4-deficient SC-lineage prematurely exits the cell cycle and undergoes terminal differentiation, which is regulated via the ERK signaling axis. This study advances our foundational knowledge on the molecular mechanisms governing SC quiescence, and reveals FGFR1/4 as promising therapeutic targets with the potential to enhance regenerative outcomes in skeletal muscle repair.
    Keywords:  FGF signaling; Quiescence; Satellite cell
    DOI:  https://doi.org/10.1186/s12964-025-02533-0
  4. Front Physiol. 2025 ;16 1636766
       Background: Glutamate is the major excitatory neurotransmitter in the nervous system, common in neuromuscular junctions, and with abnormally reduced levels in several muscle diseases. Glutamate receptor AMPA GluR3, encoded by the GRIA3 gene, has important neurophysiological roles in regulation of neural networks, sleep, and breathing. GluR3 deletion or abnormal function increases the susceptibility to seizures and disrupts oscillatory networks of sleep, breathing, exploratory activity, and motor coordination.
    Questions: Do human skeletal muscle cells express GluR3? Are they activated by glutamate? Do autoimmune GluR3B antibodies of Nodding Syndrome (NS) patients, and/or other intractable epilepsy patients, that bind and damage neural cells, also bind and affect skeletal muscle cells?
    Results: We discovered several original findings: 1) Human primary skeletal muscle cells (myoblasts) express GluR3 RNA and protein, evident by PCR and immunostaining, 2) glutamate (10-8-10-5M) increases intracellular sodium in human skeletal muscle cells and increases muscle cell number (probably by inducing muscle cell proliferation), 3) AMPA and NMDA increase intracellular sodium in skeletal muscle cells, 4) GluR3B monoclonal antibody binds skeletal muscle cells and increases their number, 5) autoimmune affinity-purified GluR3B antibodies of epileptic NS patients, suffering from nodding due to loss of muscle tone and muscle wasting, bind skeletal muscle cells, 6) purified IgGs rich in autoimmune GluR3B antibodies of intractable epilepsy patients bind and kill skeletal muscle cells.
    Possible implications: Together, the novel findings in this study may have various important implications on muscle physiology and pathology and call for continuation studies on diverse physiological, pathological and therapeutic topics. Meanwhile, we raise few hypotheses: 1) GluR3 has an important physiological role in muscle cells and motor function, 2) impaired GluR3 function (due to genetic/epigenetic/autoimmune/infectious/inflammatory factors?) can cause muscle impairments and motor problems, 3) glutamate, by direct activation of GluR3 and/or other GluRs expressed in skeletal muscle cells, can beneficially affect muscle cell survival, growth, and function, 3) Glutamate, iGluR agonists, and/or GluR3B mAb may have therapeutic effects for muscle diseases, injuries, and age-related sarcopenia, 4) autoimmune GluR3B antibodies of NS patients and/or other epilepsy patients may bind GluR3 in muscle cells, damage these cells, and induce muscle dysfunction and motor problems.
    Keywords:  GluR3; GluR3B antibodies; autoimmune epilepsy; epilepsy; glutamate; glutamate receptor; human skeletal muscle; nodding syndrome
    DOI:  https://doi.org/10.3389/fphys.2025.1636766
  5. J Physiol. 2025 Dec 31.
      Resistance exercise training (RET) leads to marked interindividual heterogeneity in the hypertrophic response. Whether such heterogeneity is due to endogenous (i.e. inherent biological factors) or exogenous variables (i.e. external load) has not been firmly established. Twenty healthy young male participants completed thrice-weekly resistance exercise sessions for 10 weeks. Each participant had their legs and arms randomly assigned to perform unilateral bicep curls or knee extensions with either a higher (heavier) load (HL: 8-12 repetitions; ∼70%-80% of one-repetition maximum (1RM)) or, in the contralateral limb, lower load (LL: 20-25 repetitions at ∼30%-40% 1RM) for three sets to volitional fatigue during each session. Fat- and bone-free mass (dual-energy X-ray absorptiometry), muscle size (ultrasonography and muscle biopsies) and strength were measured pretraining and at 10 weeks. Skeletal muscle biopsies were obtained from the vastus lateralis, and we used ingested deuterated water to assess myofibrillar protein synthesis (MyoPS) at weeks 1 and 10 during training. Despite considerable interindividual variability in hypertrophic responses, we observed that muscle hypertrophy following RET was relatively well conserved within versus between subjects and was unaffected by load. Rates of MyoPS in weeks 1 and 10 of training were increased relative to rest (Week 1: Δ0.27 ± 0.11, P < 0.0001; Week 10: Δ0.10 ± 0.14%/d, P = 0.009); however, MyoPS was attenuated in week 10 versus week 1 (Δ0.16 ± 0.18%/d, P < 0.001). MyoPS rates were less heterogenous within versus between individuals. Variation in RET-induced muscle hypertrophy occurred independent of external load and was relatively well conserved (i.e., retention of the hypertrophic response) across different anatomical limbs within an individual. KEY POINTS: Considerable interindividual variability exists in resistance exercise training (RET)-induced muscle hypertrophy. However we observed that RET-induced muscle hypertrophy is relatively conserved within an individual (i.e. between the upper- and lower body) and is independent of external load when RET is performed to volitional fatigue. Changes in myofibrillar protein synthesis (MyoPS) rates are comparable with both higher and lower loads but are blunted following a period of RET despite progressive overload. There is negligible shared variance between RET-induced increases in muscle size and strength. Additionally, there are limited relationships between measures used to assess RET-induced muscle hypertrophy. We conclude that when effort is matched (i.e. working to volitional muscular fatigue), RET-induced hypertrophy is mediated to a far greater degree by inherent endogenous biological factors, which account for a large proportion of the heterogeneity between individuals.
    Keywords:  anabolism; exercise; human muscle; protein metabolism
    DOI:  https://doi.org/10.1113/JP289684
  6. FASEB J. 2026 Jan 15. 40(1): e71392
      Myozenin 1 (MYOZ1) is expressed in fast-twitch muscle fibers and functions as a calcineurin (CaN)-interacting protein. The deletion of MYOZ1 was reported to enhance the exercise capacity of mice. It would be prospective to explore the exact role of MYOZ1 in Duchenne muscular dystrophy pathology. The transfection of an adenoviral MYOZ1 shRNA in mdx mice was used to knock down MYOZ1 expression. Forelimb grip strength test, hanging wire test, and run-to-exhaustion test were conducted for assessing muscle strength and exercise ability of mice. Muscle tissue pathology was detected by HE and Masson staining. In addition, the myofiber composition of tibialis anterior muscle relied on detecting the markers of slow- fast-twitch muscle fibers. Then, the indicators of mitochondrial function, autophagy, and fission were also investigated. According to the results, in the tibialis anterior muscle of mdx mice, MYOZ1 inhibition facilitated CaN signal transduction and ameliorated muscle atrophy, also upregulated slow-twitch muscle fiber markers and downregulated fast-twitch muscle fiber markers. Besides, mitochondrial DNA and ATP content and mitochondrial membrane potential were increased in the MYOZ1 silenced group. The inhibition of MYOZ1 also promoted the expressions of mitochondrial autophagy and fission-associated proteins, including LC3I, LC3II, and p-DRP1 (Ser637). Cyclosporin A, a CaN signaling inhibitor, reversed the effect of MYOZ1 inhibition described above. In conclusion, MYOZ1 inhibition mitigated the pathological progression of tibialis anterior muscle in mdx mice, presenting as the improved mitochondrial function and increased slow-twitch muscle fibers.
    Keywords:  calcineurin; duchenne muscular dystrophy; mitochondria; muscle fiber; myozenin 1
    DOI:  https://doi.org/10.1096/fj.202402233RR
  7. Sci Rep. 2025 Dec 29. 15(1): 44863
      Unconventional myosin VI (MVI) is an ATP-dependent actin-binding molecular motor that participates in numerous cellular and tissue functions, including striated muscle physiology. Lack of MVI expression significantly aberrates myogenesis and skeletal muscle metabolism, and alters myoblast adhesion, fusion, and cytoskeletal organisation. Concomitantly, MVI knockout mice display functional and structural cardiac defects. Here, for the first time, we investigate the impact of MVI on neuromuscular junctions (NMJs), the peripheral synapses crucial for skeletal muscle contraction. We show that MVI is enriched at the postsynaptic machinery of developing and adult NMJs. We analyse the morphology of NMJs of MVI knockout mice (Snell's waltzer, SV) during early developmental remodelling and show that MVI deficiency delays NMJ maturation in fast- and slow-twitch muscles. It also reduces the NMJ size of the soleus muscle, as demonstrated by the decreased morphological parameters of both presynaptic and postsynaptic compartments. Simultaneously, synaptic elimination remains unaffected after MVI knockout, suggesting that the observed phenotypes are innervation-independent. Lastly, depletion of MVI impairs the grip strength of both female and male SV/SV mice. In summary, our studies show that MVI is an important regulator of NMJ size and maturation, controls muscle performance, and its impact is independent of innervation and sex.
    Keywords:  Maturation; Molecular motor; Myosin VI; Neuromuscular junction; Skeletal muscle
    DOI:  https://doi.org/10.1038/s41598-025-28650-x
  8. Cell Mol Life Sci. 2025 Dec 31.
      Skeletal muscle has an innate ability to restore damaged muscle fibers by contributing specific progenitor cells, called muscle satellite cells. Here we show that secretoglobin (SCGB) 3A1, a tumor suppressor gene in various malignancies including rhabdomyosarcoma, is induced just after muscle injury and contributes to damaged muscle fiber regeneration. Lineage tracing of SCGB3A1 in mice show that SCGB3A1-positive cells highly express myosin heavy chain (MyHC)-IIX in damaged fiber area. Scgb3a1-null and Pax7CreERT2;Scgb3a1f/f conditional-null mice exhibit defective IIX and IIB fiber regeneration, with a concomitant reduction in the expression of Notch3, a gene important for the maintenance of satellite cell self-renewal pools. Aged Scgb3a1-null mice show reduced size of muscle fibers and mass, resulting in compromised muscle performance as compared to the age-matched wild-type mice. This study reveals that SCGB3A1 is an unexpected novel molecule expressed in muscle satellite cells that contributes to fiber type specific muscle regeneration.
    Keywords:  Muscle regeneration; Muscle satellite cells; Myosin heavy chain; Notch signaling; PAX7; SCGB3A1
    DOI:  https://doi.org/10.1007/s00018-025-06045-5
  9. Antioxidants (Basel). 2025 Nov 30. pii: 1445. [Epub ahead of print]14(12):
      Skeletal muscle, which in men accounts for nearly half of the body's mass, stands out as the most adaptable and energetically demanding tissue [...].
    DOI:  https://doi.org/10.3390/antiox14121445
  10. Mol Biol Rep. 2025 Dec 29. 53(1): 224
      Sarcopenia is a prevalent and debilitating skeletal muscle disorder in the aging population, characterized by progressive loss of muscle mass, strength, and function. Despite its significant impact on mobility, independence, and healthcare systems worldwide, effective pharmacological treatments remain limited. Recent advances in the understanding of sarcopenia pathophysiology have identified myostatin-a potent negative regulator of muscle growth-as a promising therapeutic target. Myostatin inhibitors-comprising direct agents such as monoclonal antibodies and small molecules, as well as indirect modulators including follistatin-based strategies and other pathway regulators-have demonstrated encouraging results in preclinical and early clinical studies by increasing muscle mass and improving muscle function. This comprehensive review summarizes current knowledge of myostatin's molecular mechanisms in muscle homeostasis, evaluates the efficacy and safety of various myostatin-targeted therapies in sarcopenia, and discusses the translational challenges and future directions for clinical application. The integration of myostatin inhibition into therapeutic regimens offers the potential to address a critical unmet need in sarcopenia management and improve the quality of life for elderly individuals.
    Keywords:  Aging; Muscle atrophy; Muscle regeneration; Myostatin inhibitors; Sarcopenia; Therapeutic targets
    DOI:  https://doi.org/10.1007/s11033-025-11390-6
  11. Int J Mol Sci. 2025 Dec 18. pii: 12184. [Epub ahead of print]26(24):
      Myopathy encompasses a group of diseases characterized by abnormalities in both muscle function and structure. However, the underlying regulatory mechanisms of newly formed myofiber development remain poorly defined. No promising therapeutic approach has been developed, but numerous medication options are available to alleviate symptoms. Our previous studies demonstrated that adenosine kinase (ADK) is critical in regulating adenosine metabolism, pathological angiogenesis, pathological vascular remodeling, and vascular inflammatory diseases. Adenosine dynamically distributes between extracellular and intracellular, and adenosine concentration regulates ADK expression. However, the mechanism by which adenosine triggers an ADK-dependent intracellular signaling pathway to regulate skeletal muscle regeneration is not well defined. This study aimed to evaluate whether the adenosine-induced intracellular signaling pathway is involved in regulating myopathy, and how it regulates the development of newly formed myofibers. In this study, an intramuscular injection of cardiotoxin was used to induce a skeletal muscle injury model; satellite cells and C2C12 cells were employed. Whether adenosine regulates satellite cell activity, new myofiber formation and differentiation, as well as fusion of myofibers, were determined by H&E staining, BrdU incorporation assay, and spheroid sprouting assay. Interaction between ADK and PFKFB3 was evaluated by IF staining, PPI network analysis, molecular docking simulation, and CO-immunoprecipitation assay. The results demonstrated that adenosine dynamically distributes between extracellular and intracellular through concentrative nucleoside transports or equilibrative nucleoside transporters, and it rapidly induces an ADK-dependent intracellular signaling pathway, which interacts with PFKFB3-mediated glycolytic metabolism to promote satellite cell activity, new myofiber formation, differentiation, and fusion, and eventually enhances skeletal muscle regeneration after injury stress. The remarkable endogenous regeneration capacity of skeletal muscle, which is regulated by adenosine-triggered intracellular signaling, presents a promising therapeutic strategy for treating muscle trauma and muscular dystrophies.
    Keywords:  ADK-dependent intracellular signaling pathway; PFKFB3-mediated glycolytic metabolism; adenosine; newly myofiber development; skeletal muscle regeneration
    DOI:  https://doi.org/10.3390/ijms262412184
  12. J Appl Physiol (1985). 2025 Dec 29.
      Several studies have examined the association between resistance training (RT)-induced muscle hypertrophy and androgen signaling in men. However, only one recent study has reported that estrogen receptor alpha (ERα) protein content positively associates with myofiber hypertrophy following RT. Thus, we investigated the acute and chronic effects of RT on skeletal muscle ERα markers in women and men, and whether these outcomes predicted hypertrophic responses. Given the role of ERα in satellite cell (SC) regulation, we also examined fiber type-specific SC content and SC-related proteins (MyoD, myogenin [Myog], cyclin D1 [CycD1]). Thirty-eight young individuals (19 women) completed 10 weeks of RT. Vastus lateralis biopsies and ultrasound-derived muscle cross-sectional area (mCSA) were obtained at baseline, 24h after first session (acute, biopsy only), and post- intervention. Total ERα, cytoplasmic ERα (cERα), and nuclear ERα (nERα) protein contents were assessed via Western blotting, ERα-DNA binding activity by an oligo-ELISA kit, and myofiber characteristics using immunohistochemistry. Men showed higher baseline total ERα than women. Both sexes showed acute reductions in cERα, nERα, MyoD, Myog, and CycD1. RT increased type I and II SC content and decreased cERα and CycD1, with no changes in ERα-DNA binding. No correlations were observed between ERα markers and hypertrophy in women, whereas in men, an acute reduction in cERα was negatively correlated with chronic mCSA changes. Although we provide further evidence of skeletal muscle ERα markers being responsive to RT, our data suggest that ERα signaling markers may not be a primary driver in RT-induced muscle growth.
    Keywords:  Hormone receptors; exercise; muscle growth regulators; sex
    DOI:  https://doi.org/10.1152/japplphysiol.00946.2025
  13. Cell Rep. 2025 Dec 30. pii: S2211-1247(25)01540-2. [Epub ahead of print]45(1): 116768
      Myocytes are exceptionally long-lived cells that must maintain proteome integrity over decades while adjusting for changes in functional output and metabolic demand. We used in vivo stable isotope labeling combined with mass spectrometry proteomics and correlated multi-isotope imaging mass spectrometry to quantify and visualize protein turnover across cardiac, fast-twitch, and slow-twitch skeletal muscles, creating a resource of hundreds of individual protein turnover rates from each tissue. We found that cardiac muscle has the highest rate of protein turnover, followed by slow-twitch skeletal muscle and then fast-twitch skeletal muscle, and that these different rates of protein turnover are driven by different levels of muscle use, rather than myosin isoform composition. We also identified protein age heterogeneity at the myofiber and sarcomere levels. These findings uncover fundamental principles of muscle protein maintenance and have broad implications for understanding cellular aging, muscle disease, and the design of therapeutic strategies targeting muscle protein turnover.
    Keywords:  CP: Metabolism; CP: Molecular biology; actin; cardiac muscle; half-life; long-lived proteins; multi-isotope imaging; protein turnover; skeletal muscle; stable-isotope labeling
    DOI:  https://doi.org/10.1016/j.celrep.2025.116768
  14. Dis Model Mech. 2025 Dec 01. pii: dmm052462. [Epub ahead of print]18(12):
      Centronuclear myopathies (CNMs) are rare congenital muscle disorders with no effective treatment. Previous studies showed that tamoxifen improved muscle function in mice modeling CNMs caused by variants in MTM1, BIN1 and DNM2. Here, we investigated whether tamoxifen administration improves muscle function and pathology in the severe recessive Ryr1TM/indel mouse model of RYR1-related CNM. Contractile performance, histological analyses and protein levels were assessed in Ryr1TM/indel mice and control littermates (wild type) treated with either a tamoxifen-enriched diet (65 mg/kg of food) or a control diet for 5 weeks, beginning at 3 weeks of age. Ryr1TM/indel mice displayed muscle weakness, reduced myofiber size and a high number of fibers with nuclei in abnormal position, regardless of the treatment. Force production during repeated contractions was reduced in tamoxifen-treated Ryr1TM/indel mice compared to that in untreated Ryr1TM/indel mice. The levels of CNM proteins (DNM2 and BIN1) were unchanged following the treatment. Tamoxifen did not improve muscle dysfunction, atrophy or histological hallmarks in Ryr1TM/indel mice. Our data indicate that tamoxifen supplementation is not beneficial and may negatively impact muscle function in this model of CNM, suggesting limited therapeutic value for patients with RYR1 mutations.
    Keywords:  Congenital myopathy; Force production; Internalized nuclei; Mouse model; Muscle atrophy; Pharmacotherapy
    DOI:  https://doi.org/10.1242/dmm.052462
  15. Biol Res Nurs. 2025 Dec 27. 10998004251411301
      Background: Inflammation is essential for skeletal muscle repair following acute injury, and successful muscle regeneration requires a balance between pro- and anti-inflammatory signaling. Estrogen regulates inflammatory responses, suggesting a role in tissue repair. CD169+ macrophages are also associated with tissue repair. However, these cells and related interferon-inducible genes have not been investigated in regenerating muscle. Objective: To examine how 17β-estradiol treatment influences the expression and co-expression network of Siglec1 (encoding CD169), an interferon-inducible gene, during muscle regeneration. Methods: Ovariectomized mice received either 17β-estradiol or placebo before undergoing a standardized crush injury to hindlimb muscle groups and exposure to simulated flight. Mice were euthanized at 32-h, 96-h, or 192-h postinjury. Differential gene expression analysis was performed on injured muscles to assess treatment-related transcriptional changes, and additional muscles were evaluated for regeneration stage and CD169 protein expression. Results: At 192-h postinjury, Siglec1 was among the top upregulated genes in 17β-estradiol-treated mice relative to placebo mice, which reflected a decline in Siglec1 expression over time in the placebo group. Correlation analysis revealed that Siglec1 was strongly associated with interferon-related genes under placebo treatment, whereas the connectivity of Siglec1 under 17β-estradiol treatment was weakened. Immunohistochemistry confirmed stronger CD169 staining in regenerating muscle of 17β-estradiol-treated mice. Conclusion: Siglec1 expression is more stable in an estrogen state compared to an ovarian-hormone-deficient state. Understanding how estrogen deficiency alters inflammatory signaling after muscle injury may inform interventions to promote recovery in postmenopausal women, who may be at risk for impaired muscle repair.
    Keywords:  SOCS7; Siglec1; contusion; estrogen; interferon-inducible; muscle regeneration
    DOI:  https://doi.org/10.1177/10998004251411301
  16. Biomolecules. 2025 Dec 07. pii: 1709. [Epub ahead of print]15(12):
      Muscle gradually loses its regenerative capacity with aging. Recent evidence highlights age-related immune dysregulation as a key driver of satellite cell dysfunction and reduced muscle regeneration. Timely elimination of apoptotic cells by phagocytes through efferocytosis is essential for tissue repair. Therefore, exploring age-related alterations in the molecular machinery of efferocytosis and their impact on muscle regeneration is of great relevance. This study examined the efferocytic machinery in the gastrocnemius muscle tissue of young and aged rats after doxorubicin-induced acute myotoxicity and assessed the potential of Vitamin B12-loaded chitosan nanoparticles (B12 CS NPS) to enhance efferocytosis and promote skeletal muscle injury repair in aged rats. Aged rats exhibited impaired efferocytosis with a significant reduction in MerTK, PPARγ, and miR-124 expression, and increased ADAM17 expression. B12 CS NPS administration significantly improved efferocytosis and reduced necrotic tissue areas, accompanied by increased MerTK, PPARγ, and miR-124, and reduced ADAM17 expression. Supplementation with B12 CS NPS significantly enhanced satellite cell proliferation and differentiation, which was indicated by upregulated expression of Pax7, Myog, and MyoD. These findings reveal that age-related alterations in regulatory molecules impair efferocytosis in aged muscle and demonstrate the potential of B12 CS NPs to enhance efferocytosis and improve skeletal muscle repair.
    Keywords:  aging; doxorubicin; efferocytosis; muscle regeneration; skeletal muscle; vitamin B12
    DOI:  https://doi.org/10.3390/biom15121709
  17. Mol Ther Nucleic Acids. 2026 Mar 12. 37(1): 102785
      Lipid nanoparticles (LNPs) are emerging as nonviral vectors for gene therapy; yet, their strong liver tropism and lack of tissue specificity remain limiting. Here, we developed, through rational design, a skeletal muscle-targeted delivery platform by functionalizing LNPs with MyomP1, an extracellular conserved peptide derived from the muscle-specific fusogenic protein Myomerger. MyomP1-LNPs were engineered to encapsulate plasmid DNA or mRNA. In vitro, MyomP1 conjugation significantly increased transduction efficiency in murine and human myoblasts and myotubes. In vivo, MyomP1-LNPs significantly enhanced muscle transduction when delivering DNA cargo, strongly reduced liver accumulation following intramuscular and intravenous mRNA delivery, and attenuated local immune activation. This work demonstrates a ligand-guided strategy to overcome organ-specific barriers in nonviral gene transfer, with improved safety and specificity. It suggests that MyomP1-engineered LNPs hold strong potential to improve therapeutic outcomes for patients with rare muscle diseases, offering a promising alternative to traditional viral gene therapy platforms.
    Keywords:  MT: Delivery Strategies; gene therapy; lipid nanoparticles; liver detargeting; mRNA delivery; peptides
    DOI:  https://doi.org/10.1016/j.omtn.2025.102785
  18. NFS J. 2025 Dec;pii: 100253. [Epub ahead of print]41
      The goal of this study was to examine benefit of omega-3 polyunsaturated fatty acids (PUFA) derived from fish oil or resolvin D1, metabolite of docosahexaenoic acid, on skeletal muscle disuse atrophy. Atrophy was induced of the left leg of C57Bl6/J male mice for 1 week. Three different treatment experimental protocols were used by feeding a diet enriched with menhaden oil or daily injections with resolvin D1 before or after immobilization. Evaluation of muscle atrophy was performed by determining the difference in weight of the tibialis anterior muscle between the immobilized (left) and mobile contralateral side (right) and by immunohistochemistry. A 2-week prevention protocol with menhaden oil enriched diet or daily injections of 2 ng/g resolvin D1 provided the greatest protection from muscle atrophy. Significant protection was also observed when dietary treatment of the mice was initiated at time of immobilization. This study demonstrated that omega-3 PUFA and metabolites provide protection toward skeletal muscle disuse atrophy.
    Keywords:  Fatty acid metabolism; Omega-3 polyunsaturated fatty acids; Resolvins; Skeletal muscle atrophy; eicosapentaenoic acid, docosahexaenoic acid
    DOI:  https://doi.org/10.1016/j.nfs.2025.100253
  19. Genomics. 2025 Dec 31. pii: S0888-7543(25)00196-X. [Epub ahead of print] 111180
      Transposable elements (TEs), once considered "junk" DNA, constitute nearly half of the mammalian genome and can replicate and reposition within the host genome. Advances in omics technologies have improved the capture and annotation of TEs, enabling functional studies. Here, we review TEs classification, structure, regulation, and annotation methods. TEs act as regulatory elements or non-coding RNAs, influencing gene networks and cell fate. While once thought inactive in somatic cells, recent evidence suggests that TEs remain transcriptionally active in various tissues, contributing to function. Focusing on skeletal muscle development, pathological regeneration, and aging, we discuss TEs expression patterns and their potential functional. TEs exhibit stage-specific expression during muscle development and are implicated in muscle-related diseases. Building on the transposon theory of aging, we summarize the increased TEs transcription and chromatin accessibility in aging muscle. Understanding TEs in skeletal muscle biology provides insights into muscle development and age-related functional decline.
    Keywords:  Aging; Development; Regeneration; Skeletal muscle; Transposable elements
    DOI:  https://doi.org/10.1016/j.ygeno.2025.111180
  20. J Med Genet. 2025 Dec 30. pii: jmg-2025-111261. [Epub ahead of print]
      Duchenne muscular dystrophy (DMD) is a severe X-linked myopathy characterised by progressive skeletal and cardiac muscle degeneration, loss of ambulation, respiratory failure and premature mortality. Although corticosteroids and gene therapies have improved disease management, they are limited by significant side effects, mutation specificity and delivery challenges, underscoring the need for an alternative or an adjunctive strategy. Emerging evidence identifies autophagy dysregulation as a critical secondary pathological mechanism in DMD, contributing to impaired clearance of damaged organelles and toxic protein aggregates, exacerbating muscle atrophy and fibrosis.This review aims to acknowledge current insights into autophagy regulation in healthy muscle and its disruption in DMD, explore its crosstalk with key pathological pathways such as nuclear factor kappa B signalling, mitochondrial dysfunction and endoplasmic reticulum stress and critically evaluate emerging therapeutic strategies targeting autophagy.Autophagy, a fundamental cellular recycling process, is suppressed in DMD by hyperactivation of the Akt-mTOR pathway and dysregulated calcium homeostasis. This leads to mitochondrial dysfunction, oxidative stress and activation of inflammatory cascades. Recent preclinical studies highlight the therapeutic potential of pharmacological and dietary autophagy modulators, including rapamycin, 5-aminoimidazole-4-carboxamide ribonucleotide, low protein diets, SRT2104 and Givinostat, which improve autophagic flux, restore mitochondrial integrity and attenuate fibrosis. Lifestyle interventions and combinatorial approaches further underscore the importance of integrating multimodal strategies.Further research should focus on longitudinal studies to optimise therapeutic timing, validate dynamic biomarkers (LC-II, p62, miRNAs) and leverage artificial intelligence with multiomics integration for precision therapies. Targeting autophagy and its interconnected pathways holds promise for transforming DMD management and improving patient outcomes.
    Keywords:  Molecular Biology; Musculoskeletal Diseases; Neuromuscular Diseases; X-Linked Genetic Diseases
    DOI:  https://doi.org/10.1136/jmg-2025-111261
  21. Cell Biosci. 2025 Dec 27.
      Peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) is a crucial coactivator that regulates mitochondrial biogenesis and function across diverse tissues, including the brain, heart, skeletal muscle, bone marrow, and liver. The diversity of PGC-1α isoforms in distinct tissues allows this co-transcription factor to exert wide-ranging biological effects, including regulating mitochondrial functions, oxidative stress, and endoplasmic reticulum homeostasis. Here, we focus on the key roles of PGC-1α in cell differentiation. Initially identified in brown adipose tissue in response to cold exposure, PGC-1α regulates cell differentiation by modulating gene expression networks involved in mitochondrial biogenesis. PGC-1α influences cell fate in several cell types, including adipocytes, skeletal muscle cells, and bone marrow-derived cells. A deeper understanding of PGC-1α provides valuable insights into developmental biology, tissue formation, and potential therapeutic targets for regenerative medicine and disease treatment. This review explores recent progress in understanding the roles of PGC-1α in cell differentiation, offering an integrated perspective on its significance in tissue and organism development.
    Keywords:  Cell differentiation; Metabolic reprogramming; PGC-1α; Tissue regeneration
    DOI:  https://doi.org/10.1186/s13578-025-01519-2
  22. Handb Exp Pharmacol. 2025 Dec 30.
      Hydrogen sulfide (H₂S) is increasingly recognized as gaseous endogenous molecule for its significant role in various physiological processes, behind its historical association with toxicity. Recent studies have highlighted H₂S's cytoprotective properties, including antioxidant, anti-inflammatory, and antifibrotic effects, particularly in the context of skeletal muscle (SKM) health. SKM disorders, such as muscular dystrophy, human malignant hyperthermia, and sarcopenia, lead to severe structural and functional impairments that adversely affect the quality of life. Although limited literature is available on the role of H2S in SKM physiopathology, it is gaining special interest. Emerging evidence suggests that H₂S may have a protective role in mitigating muscle damage and dysfunction. This chapter explores the dual functions of H₂S in SKM physiology and pathophysiology, emphasizing its potential therapeutic applications. We propose that H₂S-based strategies may offer promising avenues for alleviating the progression of muscle-related disorders and warrant further investigation to fully elucidate its mechanisms of action.
    Keywords:  H2S donors; Myopathies; Persulfidation
    DOI:  https://doi.org/10.1007/164_2025_779
  23. Spectrochim Acta A Mol Biomol Spectrosc. 2025 Dec 29. pii: S1386-1425(25)01711-1. [Epub ahead of print]350 127403
      Regeneration of skeletal muscle involves tightly coupled structural and redox processes that remain difficult to characterize in vivo or in situ with molecular specificity. Here, we introduce a label-free spectroscopic imaging framework that integrates multiphoton imaging-including second-harmonic generation (SHG) and two-photon-excited fluorescence (TPEF)-with steady-state and time-resolved fluorescence spectroscopy to visualize sarcomere remodeling and redox dynamics in living zebrafish muscle. SHG imaging revealed a transient disruption and gradual restoration of sarcomeric order, indicating an asynchronous, sequential reassembly of myofibrillar structures following localized femtosecond laser ablation. In parallel, TPEF imaging detected a pronounced and spatially confined increase in intrinsic emission at the injury site. Single-photon fluorescence spectroscopy and lifetime analysis identified this emission as protein-bound, oxidized flavin adenine dinucleotide (FAD), matching the photophysical signature of mitochondrial flavoproteins and revealing localized redox activation during the early phase of repair. Complementary reactive‑oxygen-species (ROS) imaging confirmed localized oxidative stress associated with elevated electron-transport activity, linking mitochondrial metabolism to structural recovery. Together, these multimodal observations establish a quantitative, label-free framework for correlating mitochondrial FAD-mediated redox activity with sarcomeric remodeling, establishing a versatile optical platform for investigating flavin photophysics, oxidative metabolism, and regenerative physiology in living muscle tissues.
    Keywords:  Femtosecond-laser ablation; Flavin adenine dinucleotide (FAD); Fluorescence lifetime spectroscopy; Mitochondrial redox metabolism; Second-harmonic generation (SHG); Skeletal muscle regeneration; Two-photon excited fluorescence (TPEF)
    DOI:  https://doi.org/10.1016/j.saa.2025.127403
  24. Front Immunol. 2025 ;16 1657015
      The repair of functional innervated and vascularized skeletal muscle from severe injuries, such as critical limb ischemia, denervation, and volumetric muscle loss, remains a critical clinical challenge. Regenerative cell therapies are often hindered by donor site morbidities and rapid clearance from injured tissue. Furthermore, emerging tissue engineering and biomaterials approaches are often stifled by-and may even worsen-the chronic, inflammatory microenvironment that debilitates these sites of muscle injury, as well as the underlying peripheral nerves and microvessels. Consequently, the role of the immune system in tissue repair has been increasingly studied and capitalized upon in the design of regenerative biomaterials to overcome these challenges. In this review, recent strategies for the development of immunomodulatory biomaterials for vascularized and innervated skeletal muscle repair will be discussed within the context of muscle, nervous, and vascular tissues, as well as the respective roles of immune cells and tissue progenitors during these repair processes. These strategies span chemical functionalization, sustained presentation of immunomodulatory cues, and inflammatory responses to natural and synthetic biomaterials, among other approaches.
    Keywords:  biomaterials; immunomodulation; innervation; muscle regeneration; vascularization
    DOI:  https://doi.org/10.3389/fimmu.2025.1657015
  25. Exp Gerontol. 2025 Dec 30. pii: S0531-5565(25)00347-X. [Epub ahead of print] 113018
      Age-related muscle dysfunction is a major contributor to disability, frailty, and poor clinical outcomes in older adults. Skeletal Muscle Function Deficit (SMFD) framework integrates multiple domains as: muscle mass, muscle density, strength, and power to capture a broader spectrum of age-related muscle dysfunction. The primary aims of these analyses are to develop and validate a composite SMFD score and evaluate its association with key geriatric outcome. This study used data from the InCHIANTI follow-up study, involving an initial cohort of 1035 older participants, with a total of 3196 assessments. The SMFD score was computed by assigning quintile-based values of muscle area, density, strength, and lower limb power. Associations with adverse health outcomes, and major chronic diseases were analyzed using mixed-effects models. The SMFD score declined over time from baseline to the third follow-up was: β ± SE:-0.64 ± 0.12 (p-value < 0.001), β ± SE:-1.94 ± 0.13 (p-value < 0.001), and β ± SE:-4.43 ± 0.14 (p-value < 0.001), respectively, and was associated with: BADL (OR = 0.57; 95 %CI: 0.46-0.69), IADL (OR = 0.70; 95 %CI: 0.66-0.75), poor physical performance (SPPB < 7) (OR = 0.68; 95 %CI: 0.64-0.73), Fried's frailty phenotype (OR = 0.72; 95 % CI: 0.68-0.76), hospitalization (OR = 0.96; 95 %CI: 0.93-0.99), and falls' number (OR = 0.96; 95 %CI: 0.92-0.99). Whereas higher SMFD scores were negatively associated with Parkinson's disease, stroke, and hip osteoarthritis. The SMFD score is a valid, multidimensional measure that predicts adverse outcomes in older adults. It holds promise for use in clinical assessment, risk stratification, and targeted interventions.
    Keywords:  Aging muscle; Disability; Dynapenia; Falls; Frailty; InCHIANTI; Muscle strength; Powerpenia; Sarcopenia
    DOI:  https://doi.org/10.1016/j.exger.2025.113018
  26. Biomedicines. 2025 Nov 25. pii: 2876. [Epub ahead of print]13(12):
      Spinal muscular atrophy (SMA) has transitioned from a uniformly fatal disease to a treatable condition, yet incomplete neuromuscular recovery underscores the limits of current SMN-restorative therapies. Emerging data implicate disrupted axon-to-muscle exosomal signaling as an important, overlooked driver of residual dysfunction. Exosomes, nanovesicles mediating bidirectional neuronal-muscular communication, carry synaptic organizers, trophic factors, and microRNAs essential for neuromuscular junction integrity. SMN deficiency alters exosomal biogenesis and cargo, leading to loss of agrin-MuSK signaling, impaired β-actin transport, and muscle atrophy. Comparative insights from amyotrophic lateral sclerosis and muscular dystrophy reveal that stem-cell-derived or engineered exosomes restore synaptic stability, enhance regeneration, and cross biological barriers safely. Thus, we speculate herein on a translational model integrating exosome-based therapies with existing genetic interventions to achieve durable, systems-level recovery in SMA. Exosomal profiling may further yield minimally invasive biomarkers for disease monitoring and treatment optimization, establishing vesicle-mediated communication as a novel therapeutic axis in neuromuscular medicine.
    Keywords:  axon-to-muscle signaling; exosomes; neuromuscular junction; spinal muscular atrophy; translational therapy
    DOI:  https://doi.org/10.3390/biomedicines13122876
  27. Front Microbiol. 2025 ;16 1638880
      The interplay between gut microbiota and sarcopenia has emerged as a cutting-edge research topic in the medical field, garnering significant attention. Sarcopenia is an age-related syndrome characterized by a progressive decline in skeletal muscle mass, strength, and function, which profoundly impacts the quality of life in older adults and imposes substantial socioeconomic burdens on many counties. Accumulating evidence indicates that alterations in the gut microbiota are not only linked to various intestinal disorders but also to aging-associated conditions, such as sarcopenia. The gut microbiota plays a pivotal role in regulating skeletal muscle homeostasis via its metabolic products and is increasingly recognized as a potential pathophysiological factor contributing to sarcopenia development. Skeletal muscle, functioning as both a motor and endocrine organ, secretes myokines that exert critical regulatory effects on the gut microbiota. In sarcopenic individuals, reduced secretion of myokines correlates with decreased microbial diversity and compositional shifts, marked by diminished beneficial microbes and increased potentially harmful species. This establishes a vicious cycle of gut dysbiosis-sarcopenia-gut dysbiosis. Modulation of the gut microbiota has been demonstrated to enhance muscle mass and function in elderly patients with sarcopenia. Metabolites derived from the gut microbiota, such as amino acids, lipopolysaccharides, and short-chain fatty acids, are known to modulate skeletal muscle protein metabolism by influencing anabolic and catabolic pathways. Nevertheless, the bidirectional mechanisms underlying the relationship between gut microbiota and age-related sarcopenia remain incompletely understood. In this review, we aim to: (1) integrate current knowledge regarding the bidirectional interaction between sarcopenia and gut microbiota; (2) summarize existing management strategies for age-related sarcopenia based on this interaction.
    Keywords:  age-related sarcopenia; crosstalk; gut-muscle axis; inflammation; microbial metabolites; myokines; neuroendocrine system; therapeutic targets
    DOI:  https://doi.org/10.3389/fmicb.2025.1638880
  28. Front Endocrinol (Lausanne). 2025 ;16 1717134
      Cachexia remains a major complication in cancer, with limited therapeutic options. Peroxisome proliferator-activated receptor gamma (PPARγ) has emerged as a key regulator of adipogenesis, lipid metabolism, and inflammation, but its role in cachexia is paradoxical. PPARγ activation can promote lipid storage, suppress inflammation, and modulate muscle-adipose crosstalk, potentially alleviating tissue wasting. Conversely, PPARγ agonists may enhance tumor growth in certain cancers, raising safety concerns. This review examines the dual functions of PPARγ in cancer cachexia, focusing on its regulation of adipose tissue remodeling (including browning and lipid metabolism), skeletal muscle homeostasis, and systemic inflammation, alongside tumor-promoting mechanisms that complicate its therapeutic use. Finally, emerging approaches such as selective PPARγ modulators (SPPARγMs) and tissue-targeted strategies are discussed to maximize anti-cachectic effects while minimizing oncogenic risks. Understanding these context-dependent actions is essential for translating PPARγ modulation into safe, effective cachexia therapies.
    Keywords:  PPARγ; TZDs; cancer cachexia; inflammation; lipolysis; muscle wasting
    DOI:  https://doi.org/10.3389/fendo.2025.1717134
  29. Antioxidants (Basel). 2025 Dec 12. pii: 1491. [Epub ahead of print]14(12):
      Background: Aging elevates reactive oxygen species (ROS) and weakens antioxidant defenses, contributing to cardiac dysfunction. The objective of this study was to determine whether sustained activation of skeletal muscle (SkM) Nrf2 preserves cardiac function during aging and to explore the underlying mechanisms, focusing on myocardial antioxidant pathways. Methods: Tamoxifen-induced SkM-specific Keap1 knockout male mice (iMS-Keap1flox/flox; SkM-Nrf2 overexpression) were divided into young wild-type (Y-WT), aged wild-type (A-WT), and aged knockout (A-KO) groups. Cardiac performance was evaluated by echocardiography and invasive hemodynamics. Myocardial proteomics identified differentially expressed proteins (DEPs) and enriched biological pathways. Results: Compared with Y-WT, A-WT mice showed impaired left ventricular function, including reduced ejection fraction, prolonged isovolumic relaxation time, blunted inotropic response to dobutamine, and elevated Tau index. These age-related deficits were partially reversed in A-KO mice. Proteomic analysis revealed 561 DEPs between A-WT and Y-WT, and 741 DEPs between A-KO and A-WT, enriched in calcium signaling, Nrf2-mediated oxidative stress response, oxidative phosphorylation, ROS detoxification, and cardiac-specific processes, such as hypertrophy, conduction, and dilated cardiomyopathy. Conclusions: Lifelong SkM-Nrf2 activation strengthens myocardial antioxidant capacity and alleviates age-related cardiac dysfunction. These data support an antioxidant crosstalk between skeletal muscle and the heart, highlighting a potential therapeutic target for aging-associated heart failure.
    Keywords:  Keap1 knockout; aging; cardiac dysfunction; interorgan antioxidant crosstalk; myocardial proteomics; nuclear factor erythroid 2-related factor
    DOI:  https://doi.org/10.3390/antiox14121491
  30. Imeta. 2025 Dec;4(6): e70093
      We used snRNA-seq to construct a high-resolution atlas of pectoral muscle development in broiler chickens from neonatal to adult stages. This analysis revealed pronounced molecular heterogeneity among satellite cells across developmental phases and uncovered a previously uncharacterized Runx1 + satellite cell subpopulation. By integrating pseudotime trajectory reconstruction, gene set enrichment analysis, dynamic expression profiling and loss-of-function assays, we established a critical regulatory role for RUNX1 in muscle hypertrophy. Mechanistically, RUNX1 promotes myotube hypertrophy by transcriptionally repressing Pik3r1, thereby reducing PI3K p85α levels, destabilizing PTEN, and activating the PI3K/AKT/mTOR signaling cascade, which enhances protein synthesis and drives myotube growth.
    DOI:  https://doi.org/10.1002/imt2.70093
  31. Biomater Sci. 2026 Jan 02.
      Sarcopenia and muscle atrophy are major health challenges associated with aging and various pathologies, characterized by progressive loss of muscle mass and function. These conditions severely diminish patient quality of life and impose a significant healthcare burden. Traditional interventions, such as exercise therapy and nutritional supplementation, have demonstrated limited efficacy, creating an urgent need for innovative therapeutic strategies. In recent years, the application of nanotechnology in biomedicine has provided novel therapeutics for these debilitating conditions. This article reviews the latest advancements in nanotechnology for the treatment of sarcopenia and muscle atrophy, with a focus on the applications of nanocarrier drug delivery systems (such as exosomes and lipid nanoparticles), nanoimmunomodulators, wearable nanobiosensors, nano-tissue-engineered muscles, and gene editing tools based on nanotechnology (such as CRISPR-Cas9). These technologies demonstrate significant clinical potential by improving drug targeting, enhancing bioavailability, promoting muscle regeneration, and enabling real-time monitoring of disease progression. For instance, drug delivery systems based on lipid nanoparticles (LNPs) have demonstrated approximately 30% higher bioavailability compared to traditional delivery systems in murine models, while the use of exosomes has also effectively promoted the repair and regeneration of muscle tissue in preclinical trials. However, the clinical translation of nanotechnology still faces several challenges. These include uncertainties regarding nanoparticle toxicity, immunogenicity, and clearance mechanisms, issues with the scalability and reproducibility of nanocarrier manufacturing, and ethical and regulatory concerns associated with the long-term use of gene editing and nanobiosensors. Consequently, future research should not only focus on further optimizing nanomaterial design and validating therapeutic efficacy but also address aspects such as biocompatibility, safety, ethical review, and regulatory policies. This comprehensive approach is essential to facilitate the clinical translation of nanotechnology for treating muscle degenerative diseases and to catalyze the development of personalized medicine.
    DOI:  https://doi.org/10.1039/d5bm01061f