bims-moremu Biomed News
on Molecular regulators of muscle mass
Issue of 2025–06–22
twenty-six papers selected by
Anna Vainshtein, Craft Science Inc.



  1. iScience. 2025 Jun 20. 28(6): 112712
      Skeletal muscle development and regeneration requires the activities of myogenic and non-myogenic muscle stem cell populations. Non-myogenic muscle stem cells, such as fibro-adipogenic progenitors (FAPs), play important roles in muscle regeneration after injury. Activated FAPs promote myogenic muscle stem cell differentiation and contribute to the restoration of muscle architecture. In pathological conditions, FAPs can differentiate into adipocytes or fibroblasts, causing fatty infiltrations or muscle fibrosis, respectively. Here, we identified the extracellular matrix protein ADAMTS-like 2 (ADAMTSL2) as a regulator of adipogenic and fibrogenic FAP differentiation. In the context of fibrogenic FAP differentiation, ADAMTSL2 inhibited the differentiation of primary mouse and human FAPs into fibroblasts in a transforming growth factor β (TGF-β)-dependent manner. Together with our previous data, a model emerges where ADAMTSL2 has a dual role in skeletal muscle biology, a pro-myogenic role, where ADAMTSL2 promotes myogenic muscle stem cell differentiation, and a TGF-β-dependent anti-fibrotic role where ADAMTSL2 attenuates FAP-to-fibroblast differentiation.
    Keywords:  Biochemistry; Cell biology; Specialized functions of cells
    DOI:  https://doi.org/10.1016/j.isci.2025.112712
  2. Front Immunol. 2025 ;16 1597222
       Background: Age related muscle atrophy is associated with chronic inflammation and impaired autophagy. Resistance training serves as an effective intervention for enhancing skeletal muscle hypertrophy.
    Methods: This study utilized a naturally aged mouse model to investigate the role of the mammalian target of rapamycin complex 1 (mTORC1) pathway in mediating the effects of resistance training on chronic inflammation and autophagy in aged skeletal muscle.
    Results: Our findings demonstrate that resistance training increased the wet weight of the gastrocnemius (GAS) and quadriceps (Quad), absolute number of fibers and the cross-sectional areas (CSA) of skeletal muscles, as well as enhanced the maximum load and maximum grip strength. These findings indicate that resistance training improved the quality and strength of skeletal muscles in aging mice. Resistance training alleviated inflammation in aged skeletal muscle by promoting M2 macrophage polarization, reducing the mRNA levels of tumor necrosis factor alpha (TNF-α), nuclear factor-kappaB (NF-κB) and interleukin-1beta (IL-1β), and increasing the mRNA levels of interleukin-6 (IL-6) and interleukin-10 (IL-10). In aged skeletal muscle, resistance training decreased the protein expression of mTOR, regulatory-associated protein of mTOR (Raptor), p70 ribosomal protein s6 kinase (p70S6K), IL-1β, and hypoxia-inducible factor 1-alpha (HIF-1α) without affecting protein kinase B (AKT) activity. Moreover, autophagy, which is reduced in aged muscle, was increased by resistance training through increased AMP-activated protein kinase (AMPK) activity and increased BCL-2-interacting protein 1 (Beclin1) and transcriptional factor EB (TFEB) expression.
    Discussion: Our study suggests that resistance training was associated with alleviated inflammation and regulated autophagy, potentially involving the mTORC1-HIF-1α and mTORC1-AMPK pathways, which may contribute to improved skeletal muscle mass in aged mice.
    Keywords:  aging; autophagy; chronic inflammation; mTORC1; skeletal muscle
    DOI:  https://doi.org/10.3389/fimmu.2025.1597222
  3. FASEB J. 2025 Jun 30. 39(12): e70609
      In addition to the canonical metabolism-regulating function, Adenosine monophosphate-activated protein kinase (AMPK) has noncanonical functions, in which AMPK spatiotemporally phosphorylates specific sets of substrates. Recently, we identified LSMEM2, a novel substrate of AMPK in the heart. LSMEM2 is a membrane protein localized at the intercalated disc (ICD), whose function is currently under investigation. Interestingly, LSMEM2 is also expressed in the skeletal muscles. As skeletal muscles lack a homophilic intercellular junction corresponding to the ICD in the heart, predicting the role of LSMEM2 in skeletal muscles is difficult. In this study, we identified that LSMEM2 is expressed in skeletal muscles, specifically at the neuromuscular junction (NMJ). LSMEM2-knockout mice showed no histological abnormalities, suggesting that LSMEM2 is not essential for skeletal muscle development. The overexpression of full-length wild-type or C-del mutant of LSMEM2 led to the tubular aggregate formation with functional abnormality in male mice. RNA sequence analysis revealed that the gene sets of mitochondrial oxidative phosphorylation and vesicle-mediated transport are enriched in LSMEM2 overexpression. Furthermore, histological analysis demonstrated the accumulation of swollen subsarcolemmal mitochondria in LSMEM2-overexpressing skeletal muscles. The study findings suggest that LSMEM2 may play a role in the pathogenesis of skeletal muscle diseases.
    Keywords:  intercalated disc (ICD); membrane protein; neuromuscular junction (NMJ); skeletal muscle diseases; subsarcolemmal mitochondria; tubular aggregate
    DOI:  https://doi.org/10.1096/fj.202402152R
  4. Am J Physiol Cell Physiol. 2025 Jun 16.
      Although current treatments for Duchenne Muscular Dystrophy (DMD) have proven to be effective in delaying myopathy, there remains a strong need to identify novel targets to develop additional therapies. Mitochondrial dysfunction is an early pathological feature of DMD. A fine balance of mitochondrial dynamics (fission and fusion) is crucial to maintain mitochondrial function and skeletal muscle health. Excessive activation of Dynamin-Related Protein 1 (Drp1)-mediated mitochondrial fission was reported in animal models of DMD. However, whether Drp1-mediated mitochondrial fission is a viable target for treating myopathy in DMD remains unknown. Here, we treated a D2-mdx model of DMD (9-10 weeks old) with Mdivi-1, a selective Drp1 inhibitor, every other day (i.p. injection) for 5 weeks. We demonstrated that Mdivi-1 effectively improved skeletal muscle strength and reduced serum creatine kinase concentration. Mdivi-1 treatment also effectively inhibited mitochondrial fission regulatory protein markers, Drp1(Ser616) phosphorylation and Fis1 in skeletal muscles from D2-mdx mice, which resulted in reduced content of damaged and fragmented mitochondria. Furthermore, Mdivi-1 treatment attenuated lipid peroxidation product, 4-HNE, in skeletal muscle from D2-mdx mice, which was inversely correlated with muscle grip strength. Finally, we revealed that Mdivi-1 treatment downregulated expression of markers of fibrosis (Col1a1, MMP2 and MMP9) and inflammation (IL-6, MCP1, and CXCL12). In summary, these results demonstrate that inhibition of Drp1-mediated mitochondrial fission by Mdivi-1 is effective in improving muscle strength and alleviating muscle damage in D2-mdx mice. These improvements are associated with improved skeletal muscle mitochondrial integrity, leading to attenuated lipid peroxidation.
    Keywords:  Drp1; Mitochondrial Dynamics; Muscular Dystrophy; lipid peroxidation; mitochondria dynamics; muscle; muscular dystrophy
    DOI:  https://doi.org/10.1152/ajpcell.01009.2024
  5. Front Cell Dev Biol. 2025 ;13 1592512
       Introduction: Skeletal muscles are characterized by a significant ability to regenerate in response to injury. However, muscle repair is often inefficient and hindered by the development of fibrosis. The course of muscle repair is related to the type of skeletal muscle, i.e., fast- versus slow-twitch, and is controlled by various factors. Among them are TGFβ1 and two MMPs, i.e., MMP-2 and MMP-9 gelatinases that play a key role in the remodeling of the extracellular matrix (ECM). Although the role of TGFβ1 in the regulation of ECM protein synthesis is well established, its involvement in the regulation of enzymes, such as MMPs, is still not well understood. In this study, we investigated the relationship between TGFβ1 and MMP-9/MMP-2 in in vitro differentiating myoblasts isolated from rat slow-twitch Soleus or fast-twitch Extensor Digitorum Longus (EDL) muscles. We hypothesized that differences in the regulation of MMPs contribute to the varying repair efficiencies between muscle types.
    Methods: Using siRNA to silence TβR1 expression, suramin as a competitive inhibitor of the TβR1 receptor, and inhibitors of both the canonical and non-canonical TGFβ signaling pathways, we characterized the role of TGFβ1 in regulating MMP-9 and MMP-2 during differentiation of myoblasts derived from slow-twitch Soleus and fast-twitch EDL muscles in vitro.
    Results and discussion: Our results demonstrated that blocking TGFβ1 signaling pathway significantly improved regeneration in slow-twitch Soleus muscle, altered the activity of MMP-9 and MMP-2 in in vitro differentiating myoblasts, and Soleus and EDL-derived myoblasts differ in their response to inhibition of TGFβ-dependent signaling pathways.
    Keywords:  MMP-2 and MMP-9; TGFβ1; matrix metalloproteinases; satellite cells; skeletal muscle
    DOI:  https://doi.org/10.3389/fcell.2025.1592512
  6. J Physiol. 2025 Jun 15.
      Low cardiorespiratory fitness increases the risk for cardiometabolic disease. Endurance exercise training promotes cardiorespiratory fitness and improves cardiometabolic risk factors, but with great heterogeneity. Here, we tested the hypothesis that the metabolic phenotype imparted by low parental (inborn) cardiorespiratory fitness would be overcome by early-life exercise training, and that exercise adaptations would be influenced in part by inborn fitness. At 26 days of age, male and female rat low-capacity runners (LCR, n = 20) and high-capacity runners (HCR, n = 20) generated by artificial selection were assigned to either sedentary control (CTRL, n = 10) or voluntary wheel running (VWR, n = 10) for 6 weeks. Post-intervention, whole-body metabolic phenotyping was conducted, and the respiratory function of isolated skeletal muscle and liver mitochondria was assayed. Transcriptomic and proteomic profiling of these tissues was performed using RNA-sequencing and mass spectrometry, respectively. Daily VWR volume was 1.8-fold higher in HCR-VWR compared to LCR-VWR. In LCR, VWR reduced adiposity and enhanced glucose tolerance, coincident with elevated total energy expenditure. Although intrinsic skeletal muscle mitochondrial respiratory function was unchanged, estimated skeletal muscle oxidative capacity increased in VWR groups. In liver mitochondria, VWR increased both maximal oxidative capacity and ATP-linked respiration only in HCR. Transcriptomic and proteomic profiling revealed extensive remodelling of skeletal muscle and liver tissue by VWR, elements of which were both shared and distinct based on inborn fitness. Early-life exercise partly offsets the metabolic effects of low inborn fitness, but molecular adaptations to VWR are dependent on inborn fitness, with potential implications for personalized exercise medicine. KEY POINTS: Low cardiorespiratory fitness is a heritable trait associated with increased risk for cardiometabolic disease. Endurance exercise training promotes cardiorespiratory fitness and metabolic health but how genetic (inborn) fitness influences exercise-induced adaptations is unclear. We used rats selectively bred for low (LCR) or high running capacity (HCR) to test whether: (1) early-life voluntary wheel running (VWR) could offset poor metabolic health in LCR rats and (2) inborn fitness modulates adaptations to VWR. VWR improved body composition and glucose tolerance in LCR rats but did not alter mitochondrial respiratory function. Molecular analyses revealed that VWR induced shared and distinct changes in skeletal muscle and liver depending on inborn fitness, highlighting individualized biological responses. These findings suggest that genetic factors linked to fitness influence how the body adapts to exercise, with implications for personalized exercised medicine.
    Keywords:  bioenergetics; exercise physiology; lipid metabolism; mitochondria
    DOI:  https://doi.org/10.1113/JP288331
  7. Kobe J Med Sci. 2025 May 01. 71(1): E31-E40
      Piezo1, a mechanosensitive ion channel that opens in response to mechanical stimuli, is widely expressed among mammalian cell types, and regulates a diverse range of physiological processes. Although evidence has suggested potential clinical benefit of Piezo1 activation for various conditions, the safety and efficacy of such activation in living animals have remained unclear. To investigate the therapeutic potential of Piezo1 activation, we here generated genetically modified mouse models in which Piezo1 is overexpressed either specifically in skeletal muscle or systemically in response to tamoxifen treatment in adult animals. Cast immobilization induced a reduction in both muscle mass and the abundance of Piezo1 mRNA in skeletal muscle of the affected limbs in control mice. Overexpression of Piezo1 in skeletal muscle prevented the immobilization-induced reduction both in soleus muscle mass and in the corresponding cross-sectional area of myofibers, suggesting the potential benefit of Piezo1 activation for prevention of immobilization-induced muscle atrophy. Furthermore, mice with systemic overexpression of Piezo1 showed no apparent abnormalities in growth or general activity. Red blood cells from these mice manifested slight resistance to hypoosmolarity-induced hemolysis, and the animals did not develop apparent hemolytic anemia. Our findings demonstrate promising efficacy and safety of Piezo1 activation in living animals and thereby highlight the therapeutic potential of targeting the Piezo1 signaling pathway.
    Keywords:  Hemolytic anemia; Immobilization; Muscle atrophy; Overexpression; Piezo1
    DOI:  https://doi.org/10.24546/0100495773
  8. Mol Metab. 2025 Jun 12. pii: S2212-8778(25)00092-4. [Epub ahead of print] 102185
      Endurance exercise reduces the risk of metabolic diseases by improving skeletal muscle metabolism, particularly in individuals with overweight and obesity. As biological sex impacts glucose and fatty acid handling in skeletal muscle, we hypothesized sex differences at the transcriptomic and proteomic level in the acute response to exercise and after an 8-week exercise intervention. We analyzed skeletal muscle biopsies from 25 sedentary subjects (16f/9m) with overweight and obesity using transcriptomics and proteomics at baseline, after acute exercise, and following an 8-week endurance training program. Regulation of sex-specific differences was studied in primary myotubes from the donors. At baseline, differentially methylated CpG-sites potentially explain up to 59% of transcriptomic and 67% of proteomic sex differences. Differences were dominated by higher abundance of fast-twitch fiber type proteins, and transcripts and proteins regulating glycogen degradation and glycolysis in males. Females showed higher abundance of proteins regulating fatty acid uptake and storage. Acute exercise induced stress-responsive transcripts and serum myoglobin predominantly in males. Both sexes adapted to 8-week endurance training by upregulating mitochondrial proteins involved in TCA cycle, oxidative phosphorylation, and β-oxidation. Training equalized fast-twitch fiber type protein levels, mainly by reducing them in males. In vivo sex differences in autosomal genes were poorly retained in myotubes but partially restored by sex hormone treatment. In conclusion, our findings highlight sex-specific molecular signatures that reflect known differences in glucose and lipid metabolism between female and male skeletal muscle. After just 8 weeks of endurance training, these sex differences were attenuated, suggesting a convergence towards a shared beneficial adaptation at the molecular level.
    Keywords:  Exercise; LC-MSMS based quantitative proteomics; Sex-specific; Skeletal muscle
    DOI:  https://doi.org/10.1016/j.molmet.2025.102185
  9. Physiol Rep. 2025 Jun;13(12): e70420
      Compared with 2D monolayers, 3D models more closely mimic native muscle tissue and allow functional measurements. A more complete understanding of how culture conditions and duration affect myotube maturity/function is crucial for validating the transition to 3D systems. Human skeletal muscle cells were cultured as 2D monolayers or within 3D hydrogels for up to 21 days. Quantitative proteomic analysis and functional measurements were conducted to evaluate muscle cell differentiation. Myoblasts differentiated into myotubes by 8 days in both environments; however, at Day 8, 3D constructs exhibited a predominantly slow-twitch phenotype, compared with the mixed fiber type of 2D monolayers. By Day 21, 3D constructs demonstrated enhanced mitochondrial maturity, extracellular matrix remodeling, and a fast-twitch phenotype, indicated by increased myosin-2 abundance (Log2(FC)>1.29, p <0.05). Passive tension increased by >20% following prolonged culture of 3D muscle constructs, but contractile forces reduced by >40%. This study provides a comprehensive proteomic profile of human skeletal muscle cells in 2D and 3D, demonstrating that 3D culture promoted myotube maturity and highlighting the importance of selecting appropriate culture conditions. Data suggest 8 days of differentiation as ideal for achieving peak contractile force in 3D constructs, providing optimal models for testing interventions aimed at preserving muscle function.
    Keywords:  bioengineering; hydrogel; maturation; proteomics; skeletal muscle
    DOI:  https://doi.org/10.14814/phy2.70420
  10. Trends Endocrinol Metab. 2025 Jun 12. pii: S1043-2760(25)00119-5. [Epub ahead of print]
      Exercise-induced inflammation is regarded as a response to muscle damage from mechanical stress, but controlled immune signaling can be beneficial by promoting metabolic adaptation which, for example, decreases obesity and lowers the risk of diabetes. In addition to oxidative metabolism, mitochondria play a central role in initiating innate immune signaling. We review recent work that has identified the cGAS-STING-NF-κB signaling pathway, activated by the downregulation of mitochondrial proteins CHCHD4 and TRIAP1, as mediating skeletal muscle adaptation to exercise training as well as potentially promoting cellular resilience to environmental stresses. Notably, CHCHD4 haploinsufficiency prevents obesity in aging mice; therefore, this innate immune signaling pathway could be targeted to achieve some of the health benefits of exercise.
    Keywords:  CHCHD4; TRIAP1; exercise; fiber type; innate immunity; metabolism; mtDNA; obesity
    DOI:  https://doi.org/10.1016/j.tem.2025.05.004
  11. Am J Physiol Cell Physiol. 2025 Jun 16.
      Aim: Within contracting human skeletal muscle (SKM), oxygen pressure significantly drops, which has been linked to the activation of a signaling cascade mediated by hypoxia-inducible factor 1α (HIF-1α). This cascade leads to SKM angiogenesis through vascular endothelial growth factor (VEGF). However, the role of HIF-1α in exercise-induced VEGF expression within SKM remains unclear. Methods: In this study, we systematically reviewed the literature to quantitatively synthesize all available evidence on HIF-1α activation in exercised human muscle. Results: We identified 21 studies providing 39 effect sizes of pre- and post-exercise SKM HIF-1α data from 235 subjects, with 15 of them also presenting data on VEGF mRNA levels. HIF-1α mRNA increased in response to high-intensity and resistance exercise, regardless of participants' physical fitness levels. Notably, meta-regression showed that exercise-induced VEGF expression is not modulated by HIF-1α mRNA levels. Similarly, when plotting exercise-induced fold changes of VEGF and HIF-1α no significant relationship was observed. Conclusion: Our findings demonstrate that HIF-1α is expressed in contracting SKM. However, the role of HIF-1α in the exercise-induced angiogenic response remains unclear, as most of the available evidence is limited to transcriptional data.
    Keywords:  HIF-1; VEGF; angiogenesis; exercise; hypoxia; skeletal muscle; training
    DOI:  https://doi.org/10.1152/ajpcell.00297.2025
  12. Biogerontology. 2025 Jun 18. 26(4): 120
      Muscle atrophy, resulting from physical inactivity or protein deficiency, is a significant health concern. β-hydroxy-β-methylbutyrate (HMB) has potential in preserving muscle mass, but its mechanisms in various atrophy-inducing conditions are not fully understood. This study aimed to investigate HMB's effects on muscle atrophy induced by inactivity and protein deprivation, and to elucidate the underlying molecular mechanisms. Rats were subjected to inactivity or protein-deficient diets with or without HMB supplementation. Muscle morphology, strength, and biochemical parameters were assessed. In vitro studies using C2C12 myoblasts and mouse skeletal muscle satellite cells exposed to interleukin-6 (IL-6) explored molecular pathways involved in HMB's protective effects. Inactivity and protein deprivation led to muscle atrophy, reduced strength, and altered biochemical markers. HMB supplementation partially mitigated these effects, preserving muscle mass and function. HMB attenuated atrophy markers (Muscle Atrophy F-box and Muscle RING Finger 1 (MuRF1)) and maintained myogenic factor (Myogenin (MyoG)) levels. In vitro studies revealed that HMB's protective effects were mediated through the AKT/mTOR pathway, with concurrent regulation of autophagy pathways and preservation of mitochondrial function in both myoblasts and satellite cells. HMB specifically protected satellite cell viability and function through AKT-dependent mechanisms, maintaining protein synthesis and reducing apoptosis under IL-6-induced stress conditions. HMB supplementation shows protective effects against muscle atrophy induced by inactivity and protein deprivation, through multiple mechanisms including AKT/mTOR pathway activation, autophagy regulation, and maintenance of mitochondrial function in both myoblasts and satellite cells. These findings suggest HMB as a potential therapeutic strategy for preventing muscle atrophy in various clinical scenarios.
    Keywords:  AKT/mTOR pathway; HMB supplementation; Muscle atrophy; Myogenesis; Physical inactivity; Protein deprivation
    DOI:  https://doi.org/10.1007/s10522-025-10262-7
  13. Aging Dis. 2025 Jun 07.
      Sarcopenic obesity (SO), a geriatric syndrome characterized by the coexistence of progressive skeletal muscle atrophy and excessive adipose tissue accumulation, represents a growing public health challenge associated with aging populations. While multifactorial pathogenesis involves chronic inflammation, hormonal changes, and mitochondrial dysfunction, sedentary lifestyles and aging remain primary modifiable and non-modifiable risk factors, respectively. Mechanistically, exercise exerts dual therapeutic effects: (1) hypertrophy of type II muscle fibers through IGF-1/Akt/mTORC1 signaling activation, and (2) enhanced lipid β-oxidation via AMPK/PGC1α axis stimulation, thereby mitigating both sarcopenia and adiposity. The autophagy-lysosome system, a conserved cellular quality-control mechanism, orchestrates organelle turnover and nutrient recycling through three distinct pathways: macroautophagic, chaperone-mediated autophagy, and mitophagy. In SO, impaired proteolytic and lipolytic processes converge to induce autophagic flux blockade, manifested by accumulated p62/SQSTM1 and reduced LC3-II/LC3-I ratio. Targeting the AMPK/mTOR signaling nexus, which senses cellular energy status, emerges as a strategic intervention. Exercise-mediated ATP depletion activates AMPK while suppressing mTORC1, thereby synchronously inducing autophagy initiation (ULK1 phosphorylation) and lysosomal biogenesis (TFEB nuclear translocation). This metabolic reprogramming ultimately restores proteostasis and lipid homeostasis in myocytes and adipocytes.
    DOI:  https://doi.org/10.14336/AD.2025.0419
  14. Mater Today Bio. 2025 Aug;33 101924
      Skeletal muscle, the largest organ in the human body, plays vital roles in movement, heat generation, and internal organ protection. While healthy muscle can regenerate effectively, its regenerative capacity declines in conditions like congenital muscular dystrophy, severe trauma, or aging. Two-dimensional (2D) nanomaterials, with unique physicochemical properties such as high surface area, excellent biocompatibility, and tunable mechanical and electrical properties, have shown great promise in different forms of muscle injury, particularly in volumetric muscle loss (VML). Recent studies highlight their diverse applications in muscle regeneration, acting as cell recruitment platforms, drug delivery carriers, structural scaffolds, and anti-inflammatory agents. Additionally, their biological effects and intelligent responsiveness are emerging as key features. Despite these advances, safety concerns regarding toxicity and biodegradability remain a challenge for clinical application. To unlock the full potential of 2D materials, further research is needed, especially through interdisciplinary collaboration to better understand their biological effects. By addressing safety issues and harnessing their multifunctional and intelligent characteristics, 2D nanomaterials can offer a more effective and sustainable approach to skeletal muscle repair, paving the way for next-generation therapies in regenerative medicine.
    Keywords:  Regenerative medicine; Skeletal muscle repair; Two-dimensional nanomaterials
    DOI:  https://doi.org/10.1016/j.mtbio.2025.101924
  15. J Physiol. 2025 Jun 15.
      
    Keywords:  mitochondrial function; muscular dystrophy; voluntary exercise
    DOI:  https://doi.org/10.1113/JP289057
  16. J Cell Physiol. 2025 Jun;240(6): e70054
      Exercise and diet are the best-known methods for attenuating aging-related health decline. However, exercise in older age has diminished gains of strength and agility, and a danger of unrepaired muscle damage. Improving the understanding of age-related differences in response to exercise, our results demonstrate that in old mice, downhill treadmill (eccentric) exercise causes increased influx of CD45+ cells (inflammation) and fibrotic index (fibrosis) in the heart and skeletal muscles. To explain these changes, we identified newly synthesized proteins through bio-orthogonal noncanonical amino acid tagging (BONCAT) and established that exercise exacerbated age-associated protein patterns through a dysregulated transforming growth factor (TGF)-β, Ras/MAPK/PI3Akt, and JAK/STAT pathways. Testing causality, we found that an inhibitor of TGF-β (Alk5 inhibitor, A5i) in combination with the age-diminished peptide oxytocin, previously shown to rejuvenate muscle and brain in sedentary animals, allowed aged mice to exercise without pathologies of skeletal and heart muscles and youthfully restored their de novo proteomes.
    Keywords:  TGF‐beta; aging; exercise; fibrosis; inflammation; oxytocin; proteome
    DOI:  https://doi.org/10.1002/jcp.70054
  17. Cell Stem Cell. 2025 Jun 10. pii: S1934-5909(25)00192-4. [Epub ahead of print]
      Repair of muscle damage declines with age due to the accumulation of dysfunctional muscle stem cells (MuSCs). Here, we uncover that aged MuSCs have blunted prostaglandin E2 (PGE2)-EP4 receptor signaling, which causes precocious commitment and mitotic catastrophe. Treatment with PGE2 alters chromatin accessibility and overcomes the dysfunctional aged MuSC fate trajectory, increasing viability and triggering cell cycle re-entry. We employ neural network models to learn the complex logic of transcription factors driving the change in accessibility. After PGE2 treatment, we detect increased transcription factor binding at sites with CRE and E-box motifs and reduced binding at sites with AP1 motifs, overcoming the changes that occur with age. We find that short-term exposure of aged MuSCs to PGE2 augments their long-term regenerative capacity upon transplantation. Strikingly, PGE2 injections following myotoxin- or exercise-induced injury overcome the aged niche, leading to enhanced regenerative function of endogenous tissue-resident MuSCs and an increase in strength.
    Keywords:  Prostaglandin E2; aging; epigenetic remodeling; inflammaging; molecular memory; muscle stem cells; neural network analysis; regeneration; rejuvenation; sarcopenia
    DOI:  https://doi.org/10.1016/j.stem.2025.05.012
  18. Proc Natl Acad Sci U S A. 2025 Jun 24. 122(25): e2426081122
      The utility of a pure population of highly regenerative satellite stem cells (SSCs) is a prerequisite for successful cell-based muscle therapies. Previous works have reported several methods for the SSC isolation. However, the majority of cells isolated using previous methods are fibroblasts and other nonmyogenic cell types, necessitating further expensive and time-consuming purification steps often affecting the regenerative quality of the isolated SSCs. Here, we describe a simple, time-effective, and robust protocol for the isolation of a pure population of SSCs in a single direct step, eliminating the need for further purification steps. By separating the muscle fascicles from the adjacent connective tissues (i.e., epimysium and perimysium) and utilizing a defined dissociation medium, a cell pool enriched in SSCs was successfully obtained. Immunofluorescent staining confirmed the stemness and the myogenic purity of the isolated cells (~97%). Upon myogenic induction, SSCs gave rise to multinucleated myofibers that exhibited spontaneous contraction in the culture dish for up to 21 d. Efforts to optimize the culture conditions revealed that tissue culture plates (TCPs) coated with a tissue-specific extract significantly enhanced SSCs' attachment, growth, and differentiation compared to collagen I, Matrigel-coated TCPs, or noncoated TCPs. Further studies confirmed the robust myogenic regenerative capacity of the isolated cells, as evidenced by their ability to display key regenerative characteristics, demonstrating the mild effects of our isolation protocol on their regenerative capacity. The isolation protocol presented herein can potentially be used to obtain SSCs with high myogenic purity for skeletal muscle regenerative engineering and clinical indications.
    Keywords:  isolation; muscle regeneration; regenerative engineering; satellite stem cells; skeletal muscle
    DOI:  https://doi.org/10.1073/pnas.2426081122
  19. Mol Ther Methods Clin Dev. 2025 Jun 12. 33(2): 101488
      Muscles, traditionally recognized for their role in locomotion and breathing, also participate in tissue communication. Extracellular microRNAs (miRNA) have been identified as key players in intercellular and inter-organ communication in muscle and other tissues. We have previously shown that intramuscular administration of an antagomiR led to the repression of target miRNA in neighboring skeletal muscles. This study investigated whether antagomiRs could be delivered to distant muscle and other tissues following intramuscular administration. We designed antagomiRs targeting a muscle-specific miRNA, miR-133b; a ubiquitously expressed miRNA, miR-16; and a scrambled oligonucleotide. Although all sequences were detected in neighboring skeletal muscles and distant tissues following intramuscular administration, antagomiR-133b showed the highest accumulation and efficacy in various tissues. This is the first study to provide evidence that intramuscular administration of antagomiRs could be utilized to achieve efficient and widespread distribution in tissues. This in turn could form the basis for alternative future therapeutic approaches.
    Keywords:  antagomiR; biodistribution; gene therapy; in vivo, oligonucleotide delivery; intramuscular injection; miRNA; oligonucleotides; skeletal muscle; systemic administration
    DOI:  https://doi.org/10.1016/j.omtm.2025.101488
  20. Sci Transl Med. 2025 Jun 18. 17(803): eado2112
      In muscles of patients with Duchenne muscular dystrophy (DMD), expression of extracellular matrix proteases and proteoglycans, including A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS-5) and its substrates versican and fibronectin, is increased. Elevated ADAMTS-5 protease activity results in proinflammatory matrikines, including versikine. Here, we hypothesized that targeting ADAMTS-5 with the small-molecule inhibitor GLPG1972 could decrease dystrophic pathology. Using versikine as a marker of target engagement, we observed a robust reduction in human serum after up to 52 weeks of treatment with GLPG1972, confirming inhibition of ADAMTS-5 catalytic activity. Treatment of C57.muscular dystrophy x-linked (mdx) mice with GLPG1972, alone or in combination with the standard-of-care prednisolone, increased forelimb grip strength and force output of diaphragm muscles ex vivo when compared with vehicle treatment, and improvements in function were associated with decreased diaphragm inflammation and fibrosis. Treatment of C57.mdx mice with prednisolone also decreased diaphragm inflammation; however, grip strength, diaphragm force output, and fibrosis did not significantly differ from vehicle treatment. In D2.mdx mice, GLPG1972 improved forelimb grip strength and wire hang time compared with vehicle. Furthermore, GLPG1972 increased the strength of fast tibialis anterior muscles in situ and slow soleus muscles ex vivo, and this was associated with decreased inflammation, sarcolemma damage, and fibronectin deposition as assessed by immunohistochemistry. Together, these findings position ADAMTS-5 inhibition with GLPG1972 for further study as a disease-modifying strategy for DMD that may achieve both anti-inflammatory and antifibrotic effects on muscle, resulting in improved contractile function.
    DOI:  https://doi.org/10.1126/scitranslmed.ado2112
  21. Cell Metab. 2025 Jun 17. pii: S1550-4131(25)00269-4. [Epub ahead of print]
      Radiotherapy reduces the risk of cancer recurrence and death, but the fact that it's accompanied by multiple side effects including muscle fibrosis and weakness, seriously affects the life quality of patients. However, the underlying mechanism is poorly defined. Here, we identify that cancer cells secrete more spermidine synthase (SRM) enzyme through small extracellular vesicles to trigger skeletal muscle weakness upon radiotherapy. Mechanistically, irradiation-triggered arachidonic acid (ArA) accumulation elevates the ISGylation of the SRM protein, facilitating SRM packaging into extracellular vesicles from the primary tumor. Circulating SRM results in spermidine accumulation in skeletal muscle and type I collagen fiber biosynthesis in an eIF5A-dependent manner. However, losartan treatment blocks the ISGylation of SRM and its subsequent secretion. Collectively, our findings determine that ArA functions in concert for circulating SRM secretion upon radiotherapy, which aggravates skeletal muscle fibrosis through rewiring polyamine metabolism, shedding light on the alleviation of radiotherapy-mediated muscle weakness when combined with losartan treatment.
    Keywords:  extracellular vesicles; fibrosis; polyamine metabolism; radiotherapy; skeletal muscle
    DOI:  https://doi.org/10.1016/j.cmet.2025.05.013
  22. Aging Dis. 2025 Jun 13.
      Osteosarcopenia the concurrent deterioration of bone (osteoporosis) and muscle (sarcopenia) represents a critical yet understudied geriatric syndrome that synergistically amplifies frailty, fractures, and loss of independence in aging populations. This dual pathology imposes a staggering socioeconomic burden through increased disability, prolonged hospitalization, and elevated mortality. Despite its clinical urgency, therapeutic advances remain stagnant, as current interventions e.g., bisphosphonates, vitamin D supplementation are palliative and fail to address the shared molecular drivers of bone-muscle crosstalk. Emerging evidence implicates cellular senescence and epigenetic dysregulation as convergent mechanisms driving osteosarcopenia. Senescent osteocytes, burdened by oxidative stress and mitochondrial dysfunction, secrete pro-inflammatory cytokines e.g., IL-6, TNF-α and matrix-degrading enzymes (e.g., MMPs) via the senescence-associated secretory phenotype (SASP), which erodes bone integrity and propagates muscle atrophy. Simultaneously, epigenetic alterations DNA hypermethylation of osteogenic genes RUNX2, histone deacetylation repressing myogenesis MYOD1, and dysregulated non-coding RNAs (miR-133, miR-214) lock musculoskeletal tissues into a degenerative state. These processes are exacerbated by age-related inflammaging and metabolic disturbances e.g., NAD+ depletion, which amplify oxidative stress and chromatin instability. The synergy between senescence and epigenetics in perpetuating osteosarcopenia remains poorly defined. Most preclinical models overlook comorbidities (e.g., diabetes, chronic inflammation) that accelerate musculoskeletal decline. Current therapies senolytics, histone deacetylase (HDAC) inhibitors lack tissue specificity and exhibit pleiotropic effects. This review addresses these gaps by synthesizing cutting-edge insights into the senescence-epigenetics axis as a unifying driver of osteosarcopenia. By elucidating how SASP factors (e.g., myostatin) and epigenetic reprogramming e.g., sirtuin 1 (SIRT1) hypermethylation disrupt bone-muscle crosstalk, we propose novel strategies to break the self-sustaining cycle of tissue degeneration. We highlight the promise of precision geroscience leveraging CRISPR-engineered organoids, multi-omics profiling, and AI-driven biomarkers to decode tissue-specific vulnerabilities and design dual-target therapies e.g., senolytics ++ bromodomain and extra-terminal (BET) inhibitors.
    DOI:  https://doi.org/10.14336/AD.2025.0370
  23. Aging Cell. 2025 Jun 17. e70124
      The second Biomarkers of Aging Symposium, jointly hosted by the National Institute on Aging (NIA) Intramural Research Program and the Biomarkers of Aging Consortium (BAC) on September 12, 2024, in Baltimore, MD, convened leading researchers, clinicians, and stakeholders in the aging field to share new developments and discuss roadmaps to advance biomarkers of aging. This meeting report summarizes the highlights of this symposium and underscores the urgent need to understand longitudinal, complex, and heterogeneous processes of aging to unlock the full potential of aging biomarkers.
    DOI:  https://doi.org/10.1111/acel.70124
  24. Mol Ther Nucleic Acids. 2025 Jun 10. 36(2): 102569
      Duchenne muscular dystrophy (DMD) is a fatal X-linked, recessive disease caused by mutations in the DMD gene encoding dystrophin, a membrane-associated protein necessary for maintaining muscle structure and function. One of the common DMD mutations is the deletion of exon 52 (Δ52), which introduces a premature stop codon in exon 53, preventing the expression of functional dystrophin protein. Patients with this mutation could benefit from skipping or reframing exon 53 to restore the dystrophin open reading frame. In this study, we investigated the efficacy of single-cut CRISPR gene editing with Staphylococcus pyogenes Cas9 (SpCas9)-LRVQR to restore dystrophin expression in patient-derived induced pluripotent stem cells (iPSCs) and a newly generated humanized DMD mouse model. We compared two injection routes for adeno-associated virus (AAV) serotype 9 to deliver gene-editing components to neonatal mice: intraperitoneal (IP) and facial vein (FV) injection. We observed efficient restoration of dystrophin protein expression across multiple skeletal muscle groups and the heart. The AAV9-mediated CRISPR single-cut approach ameliorated key DMD hallmarks, including histopathological phenotypes, impaired grip strength, and elevated serum creatine kinase levels. Our optimized strategies for dystrophin restoration in humanized DMD mice with exon 52 deletion represent a promising treatment for DMD.
    Keywords:  AAV; CRISPR; Duchenne muscular dystrophy; MT: RNA/DNA Editing; gene editing; humanized mouse model; single-cut
    DOI:  https://doi.org/10.1016/j.omtn.2025.102569
  25. Int J Nanomedicine. 2025 ;20 7251-7270
      The Klotho protein, encoded by the KL gene, has garnered significant attention as a pivotal biomolecule in the field of aging research. Its expression levels are closely correlated with both lifespan and overall health status, exerting influence over critical physiological processes such as metabolic homeostasis, oxidative stress response, and inflammation modulation. This review aims to systematically examine the multifaceted roles of Klotho within the context of aging and its implications for various age-associated disorders. We highlight the emerging evidence suggesting that Klotho may serve as a key regulator in age-related pathologies, including cardiovascular diseases, neurodegenerative disorders, and metabolic syndromes. Furthermore, we explore the potential of Klotho as a therapeutic target, positing that interventions aimed at enhancing Klotho activity could offer novel strategies for alleviating the health burdens experienced by the aging population.
    Keywords:  Klotho; aging; inflammation; oxidative stress; population aging
    DOI:  https://doi.org/10.2147/IJN.S514516