bims-moremu Biomed News
on Molecular regulators of muscle mass
Issue of 2025–11–23
34 papers selected by
Anna Vainshtein, Craft Science Inc.



  1. Exp Physiol. 2025 Nov 19.
      Skeletal muscle plays an important role in whole-body health, quality of life and regulation of metabolism. The maintenance of a healthy mitochondrial pool is imperative for the preservation of skeletal muscle quality and is mediated through mitochondrial quality control consisting of mitochondrial turnover mediated by a balance between organelle synthesis and degradation. The selective tagging and removal of dysfunctional mitochondria is essential for maintaining mitochondrial quality control and is termed mitophagy. The mechanisms of the initial stages of mitophagy involving the recognition and tagging of mitochondria within skeletal muscle are well established, but our understanding of the terminal step involving organelle degradation mediated via lysosomes is in its infancy. An assessment of the proteolytic functions to facilitate the removal and breakdown of dysfunctional mitochondria is crucial for our understanding of the mechanisms of mitophagy, which is essential for maintaining skeletal muscle health. The aim of this review is to address the current knowledge surrounding mitophagy and lysosomal function, alongside distinct physiological conditions, such as ageing, exercise and disuse, that have varying effects on mitophagy and lysosomal adaptations within skeletal muscle.
    Keywords:  Parkin; adaptation; lysosomes; mitophagy; skeletal muscle; transcription factor EB
    DOI:  https://doi.org/10.1113/EP093041
  2. J Cachexia Sarcopenia Muscle. 2025 Dec;16(6): e70112
       BACKGROUND: Myotonic dystrophy type 1 (DM1) is a multisystemic neuromuscular disorder characterized by CTG repeat expansion in the 3' untranslated region of the dystrophia myotonica protein kinase coding gene. The presence of expanded CTG repeats in DMPK mRNAs leads to the sequestration of RNA-binding factors such as the Muscleblind-like (MBNL) proteins, resulting in widespread splicing defects that contribute to progressive muscle weakness and myotonia. Previously, we identified misregulation of SORBS1 exon 25 splicing in both DM1 and MBNL1/2 double-knockout human-induced pluripotent stem cells (hiPSC)-derived skeletal muscle cells, suggesting a potential role in DM1 physiopathology.
    METHODS: We investigated SORBS1 exon 25 splicing misregulation in human skeletal muscle biopsies from DM1 patients and healthy controls. The functional consequence of SORBS1 exon 25 exclusion was assessed in mice, zebrafish and hiPSC-derived skeletal muscle cells using an antisense oligonucleotide-mediated exon-skipping strategy.
    RESULTS: In human congenital DM1 fetal skeletal muscle biopsies, SORBS1 exon 25 inclusion was reduced by 52.6 ± 10% compared to controls (p < 0.001). Analysis of RNA sequencing data from the DMseq database further revealed significant misregulation in tibialis anterior biopsies from 40 adult DM1 patients, with a 15.8 ± 3.7% decrease in splice inclusion (p < 0.0001). In mice, forced exclusion of Sorbs1 exon 25 led to neuromuscular junction degeneration, with increased denervation (10.5% ± 3.4%, p < 0.01) and postsynaptic destabilization (5.7% ± 2.5%, p < 0.05). In zebrafish, sorbs1 exon 25 misregulation significantly impaired locomotion, reducing trajectory, distance (57.9% ± 12%, p < 0.0001) and velocity (14% ± 0.5%, p < 0.05), while also disrupting acetylcholine receptor cluster morphology. Similarly, forced SORBS1 exon 25 exclusion in hiPSC-derived skeletal muscle cells diminished the formation of large acetylcholine receptor clusters upon agrin stimulation by 34% ± 4.5% (p < 0.0001).
    CONCLUSION: Our study identifies SORBS1 alternative splicing as an essential MBNL-regulated event during skeletal muscle development, potentially involved in neuromuscular junction formation and maintenance. The aberrant splicing of SORBS1 exon 25 in DM1 expands our understanding of how splicing dysregulation compromises neuromuscular system communication, shedding light on the broader impact of mRNA splicing regulation on NMJ integrity.
    Keywords:  alternative splicing; myotonic dystrophy type 1; neuromuscular junction
    DOI:  https://doi.org/10.1002/jcsm.70112
  3. Nat Commun. 2025 Nov 21.
      Loss of Fibronectin (FN) from the skeletal muscle stem cell (MuSC) niche represents a root cause of regenerative failure in aging. While FN has pleiotropic functions during healthy skeletal muscle regeneration, it remains unclear how aging affects its spatiotemporal specificity for MuSCs. Here, we demonstrate that activated MuSCs secrete an autoregulatory FN splice variant containing the EDB extra domain (EDB(+) FN), which is not expressed by accessory cells in the niche. EDB(+) FN splicing in MuSCs depends on serine/arginine-rich splicing factor 1 (Srsf1) whose promoter is controlled by Smad3. EDB(+) FN knockdown or downregulation in aging affects MuSC proliferation through aberrant integrin signaling and impairs skeletal muscle regeneration. During a defined regeneration interval in aged mice, Smad3 activation using transforming growth factor-beta 1 (TGFβ1) improves MuSC function and skeletal muscle repair by stimulating EDB(+) FN secretion. Altogether, we identify and characterize the TGFβ1-Smad3-Srsf1-EDB(+) FN pathway as a therapeutic target for age-associated regenerative failure.
    DOI:  https://doi.org/10.1038/s41467-025-66582-2
  4. EMBO Mol Med. 2025 Nov 17.
      Cancer cachexia is a debilitating syndrome characterized by the progressive loss of skeletal muscle mass with or without fat loss. Recent studies have implicated dysregulation of the endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR) pathways in skeletal muscle under various conditions, including cancer. In this study, we demonstrate that the IRE1α/XBP1 branch of the UPR promotes activation of the ubiquitin-proteasome system, autophagy, JAK-STAT3 signaling, and fatty acid metabolism in the skeletal muscle of the KPC mouse model of pancreatic cancer cachexia. Moreover, we show that the IRE1α/XBP1 pathway is a key contributor to muscle wasting. Skeletal muscle-specific deletion of the XBP1 transcription factor significantly attenuates tumor-induced muscle atrophy. Mechanistically, transcriptionally active XBP1 binds to the promoter regions of genes such as Map1lc3b, Fbxo32, and Il6, which encode proteins known to drive muscle proteolysis. Pharmacological inhibition of IRE1α using 4µ8C in KPC tumor-bearing mice attenuates cachexia-associated molecular changes and improves muscle mass and strength. Collectively, our findings suggest that targeting IRE1α/XBP1 pathway may offer a therapeutic strategy to counteract muscle wasting during pancreatic cancer-induced cachexia.
    Keywords:  ER Stress; Fatty Acid Oxidation; JAK-STAT; Muscle Wasting; Unfolded Protein Response
    DOI:  https://doi.org/10.1038/s44321-025-00337-w
  5. J Physiol. 2025 Nov 16.
      The concept of inter-organ cross-talk, central to understanding physiological adaptations to exercise and disease progression, has been significantly advanced by identifying skeletal muscle as an endocrine organ. This article traces the historical quest for an 'exercise factor', beginning with early speculations in the 19th century and culminating in the discovery of interleukin (IL)-6 as the first myokine and exercise factor in 2003. Initially explored through the works of Julius Geppert and Nathan Zuntz, and later Erling Asmussen and Marius Nielsen, as well as Maurice Goldstein and Bengt Saltin, the search focused on substances released during muscle work, which could influence systemic physiological responses. Despite controversies and the challenge of isolating such factors, the concept gained clarity with the identification of IL-6 that is released from contracting skeletal muscles and plays a crucial role in glucose homeostasis and other processes across various organs, comprising a breakthrough that established skeletal muscle as an endocrine organ.
    Keywords:  exercise physiology; hormone; interleukin‐6; inter‐organ cross‐talk
    DOI:  https://doi.org/10.1113/JP289744
  6. Mol Med. 2025 Nov 21.
       BACKGROUND: Follistatin is a potent regulator of various TGF-β superfamily members, including myostatin (MSTN) and activin A. Previous studies have shown that follistatin is crucial in enhancing myogenesis during acute muscle injury. The mechanism by which fibro-adipogenic progenitors (FAPs)-specific follistatin influences muscle homeostasis in obese mice remains unknown. Therefore, we investigated the physiological role of follistatin in PDGFRα-positive FAPs in the regulation of muscle homeostasis and exercise in obese mice.
    METHODS: A PDGFRα-specific follistatin knockout (follistatin KO) mouse model was generated using PDGFRα-GFP-CreERT2 (PDGFRα-GCE) and follistatinflox/flox mice. These mice were fed a 60% high-fat diet (HFD) for 20 weeks, followed by a series of analyses, including exercise tolerance test, grip strength test, glucose and insulin tolerance assays, gene expression analysis, histology, western blotting, and immunohistochemistry.
    RESULTS: We showed that follistatin KO mice had reduced expression of Fst in skeletal muscle and white adipose tissue. We also showed that follistatin KO mice exhibited decreased exercise performance and altered skeletal homeostasis during obesity. Deletion of follistatin in FAPs activated the MSTN: Activin A/SMADs signaling pathways, which negatively impacted muscle homeostasis. Furthermore, follistatin KO mice showed reduced muscle mass, increased muscle degradation, and atrophic myofibers. Mitochondrial biogenesis, oxidative phosphorylation, and fatty acid oxidation were also altered in the skeletal muscles of follistatin KO mice.
    CONCLUSION: Follistatin plays a protective role in mice by maintaining the metabolic health of skeletal muscles; it restores muscle function during HFD challenge, thereby reducing diet-induced obesity-related complications.
    Keywords:  Exercise capacity; Fibro-adipogenic progenitors (FAPs); Follistatin; Muscle mass; Obesity
    DOI:  https://doi.org/10.1186/s10020-025-01393-1
  7. In Vitro Cell Dev Biol Anim. 2025 Nov 17.
      Skeletal muscle tissue consists of not only myofibers, i.e., muscle cells, but also intramuscular adipocytes. Our previous study demonstrated that adipocytes produce secretory factors during differentiation, leading us to hypothesize that soluble factors derived from adipocytes regulate gene expression and cellular function in muscle cells. Yet the mechanism by which coexisting adipocytes influence muscle cells remains unclear. Here, microarray analysis was used to examine transcriptional changes in muscle cells under two co-culture conditions: myoblasts co-cultured with differentiated adipocytes and myotubes co-cultured with preadipocytes. Gene Ontology terms related to cell adhesion, extracellular matrix (ECM) organization, and metabolic processes were significantly enriched in both conditions. We also assessed the influence of adipocyte co-culture on myogenic differentiation and fiber type-specific gene expression. In myoblasts, co-culture with differentiated adipocytes had no significant effect on the expression of myogenic regulatory factors, whereas Myh2 and Myh4 expression was markedly increased in myotubes co-cultured with preadipocytes. These results indicate that adipocyte-derived soluble factors alter the transcriptional landscape of muscle cells, especially genes involved in ECM remodeling and metabolic regulation. This intercellular communication likely contributes to structural and metabolic adaptations in skeletal muscle tissue in vivo.
    Keywords:  Adipocyte; Co-culture; Extracellular matrix; Metabolic adaptation; Skeletal muscle cell
    DOI:  https://doi.org/10.1007/s11626-025-01092-5
  8. J Appl Physiol (1985). 2025 Nov 20.
      Icing is a common acute intervention following skeletal muscle injuries in sports medicine; however, previous studies in rodent models indicated that icing impairs muscle regeneration and promotes collagen deposition in regenerating muscles. Because fibrosis decreases muscle function and increases the risk of re-injury, it poses a significant challenge for athletes. To address the fibrosis-related impairment of muscle function, we examined the mechanisms underlying icing-induced fibrosis. Specifically, we focused on the response of fibro/adipogenic progenitors (FAPs), key players in muscle fibrosis, during muscle regeneration. Using a crush-injury model in male rats, we investigated the effects of icing on the FAP response and fibrosis-related outcomes. Icing inhibited the early-phase increase in FAPs within injured muscles. Moreover, icing upregulated the expression of transforming growth factor-β1 (TGF-β1), which promotes the differentiation of FAPs into myofibroblasts, particularly 4 and 5 days post-injury, when FAP accumulation was abundant. Consequently, an increased number of α-smooth muscle actin (α-SMA)-positive myofibroblasts and an expanded collagen fiber area were observed during the later stages of icing-treated muscle regeneration, indicating excessive muscle fibrosis. Taken together, our results suggest that icing after muscle injury may promote fibrosis by altering FAP behavior and its surrounding environment, which warrants careful re-evaluation of its clinical effectiveness.
    Keywords:  fibro/adipogenic progenitor; icing; skeletal muscle fibrosis; skeletal muscle regeneration
    DOI:  https://doi.org/10.1152/japplphysiol.00649.2025
  9. bioRxiv. 2025 Oct 04. pii: 2025.10.02.680066. [Epub ahead of print]
      Myogenic commitment is a decisive and irreversible step in skeletal muscle regeneration, necessitating proliferating myoblasts to integrate metabolic cues with nuclear transcriptional programs. Among amino acids, glutamine is uniquely positioned to influence this transition by coupling energy production to macromolecule biosynthesis and epigenetic regulation. We reasoned that myoblasts must sense glutamine availability to ensure orderly progression toward commitment, and we tested this by examining the molecular consequences of acute glutamine withdrawal. We find that continued glutamine oxidation is required to sustain glycolysis, maintain mitochondrial fission, and preserve a redox balance that supports progression towards myogenic commitment. In its absence, myoblasts undergo a reductive shift, characterized by mitochondrial elongation, membrane depolarization, and suppression of glycolysis, ultimately leading to growth arrest. Transcriptomic profiling reveals reduced MyoD and MKi67 , accompanied by increased Sprouty1 levels, defining a reversible non-proliferative state that resembles but is distinct from quiescent and reserve cells. We term this state Poised Metabolic Arrest (PMA), a cellular response to glutamine limitation during myogenic progression. Mechanistically, PMA is driven by Nrf2-dependent increased glutathione (GSH) biosynthesis and upregulation of mitochondrial GSH carrier Slc25a39 when glutamine is limited. Depleting mitochondrial glutathione or silencing Slc25a39 forces exit from PMA. However, this premature exit compromises subsequent differentiation potential, indicating PMA serves to preserve differentiation competence when glutamine is limited. Consistent with this, both loss and overexpression of Slc25a39 impair myoblast differentiation in vitro and disrupt regeneration in vivo. Together, these data suggest that a reciprocal Slc25a39-Nrf2 redox axis functions as a nutrient-dependent checkpoint, coupling glutamine availability to mitochondrial remodeling and metabolic reprogramming, necessary to establish irreversible myogenic commitment.
    DOI:  https://doi.org/10.1101/2025.10.02.680066
  10. Acta Physiol (Oxf). 2025 Dec;241(12): e70135
       BACKGROUND: Myotonic dystrophy type 1 (DM1) is caused by expanded CTG repeats in the DMPK gene, causing the accumulation of toxic RNA that sequesters RNA-binding proteins. Clinically, DM1 is characterized by progressive muscle weakness and atrophy, resulting in reduced physical capacity and quality of life. Recent evidence implicates mitochondrial dysfunction in DM1 pathophysiology. While aerobic exercise has been shown to improve skeletal muscle and mitochondrial health in individuals with DM1, the benefits of strength training remain unexplored.
    OBJECTIVES: We investigated the effects of a 12-week strength training program on mitochondrial respiration, reactive oxygen species (ROS) production and muscle integrity in women with DM1.
    METHODS: Vastus lateralis muscle biopsies were collected pre- and post-training in participants with DM1 and once in unaffected/untrained individuals. Mitochondrial respiration and hydrogen peroxide emission (marker of ROS production) were assessed in permeabilized myofibers, while OXPHOS protein contents were quantified by immunoblotting and immunofluorescence. Markers of myofiber denervation (NCAM+) and integrity (centrally located myonuclei, damaged laminin, nuclear clumps) were assessed on histological sections.
    RESULTS: At baseline, DM1 participants exhibited lower mitochondrial respiration compared to unaffected individuals. Strength training significantly improved mitochondrial respiration and content in DM1 participants. At baseline, absolute ROS production was lower, while ROS production normalized to oxygen consumption (free radical leak) was higher, in DM1. Histological signs of denervation and altered muscle integrity were observed. Strength training partially normalized mitochondrial free radical leak and restored some markers of myofiber integrity.
    CONCLUSION: Collectively, our results indicate that strength training enhances mitochondrial health and improves myofiber integrity in women with DM1.
    Keywords:  ROS; exercise; mitochondria; mitochondrial function; mitochondrial respiration; myotonic dystrophy; neuromuscular disease; oxidative phosphorylation defects; resistance training
    DOI:  https://doi.org/10.1111/apha.70135
  11. Ageing Res Rev. 2025 Nov 17. pii: S1568-1637(25)00292-2. [Epub ahead of print] 102946
      The gap between lifespan and healthspan is increasing globally, resulting in millions of individuals spending additional years burdened by frailty or disease. This disparity, paired with the increasingly aged populations of Western nations, poses a palpable predicament to public health and the economy. Deterioration of the skeletal muscle system is a key contributor to illness, loss of independence, and diminishing healthspan. Muscle quality correlates to longevity due to its significant role in metabolic homeostasis and autonomous mobility, reducing instances of adverse events such as falls and fractures. The age-related loss of muscle mass and function is termed sarcopenia, affecting older adults ubiquitously without intervention through regular resistance training. Although clinical manifestations of sarcopenia are well characterized, the molecular mechanisms underlying its pathogenesis remain incompletely understood, limiting the development of targeted, mechanism-based interventions. To identify interventions beyond exercise that delay sarcopenia, it is necessary to identify early onset physiological alterations defining this process. Genetically modified mouse models of accelerated or delayed aging offer valuable insight into the cellular mechanisms that drive or mitigate sarcopenia. The latter is often achieved by disrupting the somatotropic axis, as multiple models exist that either lack growth hormone (GH) production or a functional GH receptor (GHR) paired with a secondary deficiency in insulin like growth factor-1 (IGF-1), which reliably extends lifespan across various species. This review evaluates GH's paradoxical role in muscular maintenance and contrasts the skeletal muscle health of various murine models of aging in effort to better outline the molecular underpinnings of sarcopenia.
    Keywords:  growth hormone; healthspan; murine models; sarcopenia; skeletal muscle
    DOI:  https://doi.org/10.1016/j.arr.2025.102946
  12. Am J Physiol Cell Physiol. 2025 Nov 18.
      Frailty in patients with chronic kidney disease (CKD) greatly exacerbates disease comorbidities and increases probability of death. Prior research underscores molecular alterations in skeletal muscle physiology that may underly frailty and poor intervention response in this patient population. CKD can negatively affect satellite cell abundance and function, reducing skeletal muscle injury resilience and adaptive capacity. Pathogenic drivers of compromised satellite cell abundance and activity in patients with CKD remain largely unknown. To address this gap in knowledge, we isolated primary myogenic progenitor cells (MPCs) from patients with CKD and control participants. We also sought to define cell-extrinsic and intrinsic processes that may underlie myogenic deficits. We performed RNA sequencing on MPCs from control participants cultured in control serum, MPCs from control participants cultured in CKD serum, and MPCs from CKD participants cultured in control serum. We identified zinc mishandling as a shared pathway between control cells treated with CKD serum and CKD cells treated with control serum. Consistent with these observations, we found zinc deficiency and attenuated myogenesis in MPCs from patients with CKD. Finally, we showed that zinc supplementation partially restores the myogenic capacity of MPCs from patients with CKD. Together, these data highlight the importance of zinc metabolism in myogenesis and identify a novel mechanism whereby CKD pathogenesis impedes MPC differentiation.
    Keywords:  CKD; metallothioneins; myogenic progenitor cells; satellite cells; skeletal muscle
    DOI:  https://doi.org/10.1152/ajpcell.00767.2025
  13. bioRxiv. 2025 Sep 29. pii: 2025.09.26.678822. [Epub ahead of print]
      For efficient regeneration, muscle stem cells (MuSCs) transition out of quiescence through a series of progressively more activated states. During MuSC aging, transition through the earliest steps is the slowest and delayed, with the molecular regulators that govern this transition not well characterized. By analyzing the dynamic changes of MuSCs at the molecular (scRNA-Seq and Cell Painting) and phenotypic (heteromotility) level at single cell resolution we found that the Integrated Stress Response (ISR) Pathway is a critical regulator of MuSC transition states. Aged MuSCs have increased baseline ISR activity in quiescence that does not increase during activation to levels observed in adult MuSCs. Rapid and transient pharmacological ISR activation in vitro was sufficient to increase aged MuSC activation rate and migratory behavior as well as alter the transcriptional states toward a younger phenotype. ISR activation also improved aged MuSC potency and aged mouse muscle regeneration in vivo. Therefore, pharmacological activation of the ISR has therapeutic potential to improve MuSC function and skeletal muscle repair during aging.
    Keywords:  Aging; ISRIB; Sal003; activation dynamics; integrated stress response; muscle stem cells; state transitions
    DOI:  https://doi.org/10.1101/2025.09.26.678822
  14. Nat Commun. 2025 Nov 18. 16(1): 10091
      The Rab GTPase activating protein (RabGAP) AS160 translocates from the cytosol into the nucleus acting as a transcriptional co-activator of Signal Transducer and Activator of Transcription 3 (STAT3) to regulate proliferation of muscle satellite cells (MuSCs). How this AS160-STAT3 complex is regulated remains largely unclear yet. Here, we show that TBC1D1, a RabGAP related to AS160, forms a super-complex with AS160 and STAT3 to retain the AS160-STAT3 complex in the cytosol. Phosphorylation of TBC1D1-Thr596 by protein kinase B dissociates TBC1D1 from AS160 thus releasing the cytosolic retention of the AS160-STAT3 complex. A non-phosphorylatable alanine substitution of Thr596 inhibits MuSC proliferation and impairs repair of injured muscle. In contrast, TBC1D1 deficiency, but not its GAP-inactive mutation, promotes MuSC proliferation and muscle regeneration. Thus, TBC1D1 is a negative regulator of MuSC proliferation through cytosolic retention of the AS160-STAT3 complex and might be a valuable therapeutic target for muscle regeneration.
    DOI:  https://doi.org/10.1038/s41467-025-65141-z
  15. FEBS Lett. 2025 Nov 17.
      It has been proposed that the regulatory Sit4-associated protein subunit 3 (SAPS3) of protein phosphatase 6 (PP6C) acts as an AMP-activated protein kinase (AMPK) inhibitor by recruiting PP6C to dephosphorylate AMPKα-T172. While we confirm this interaction in HEK293 cells, we find limited evidence for a SAPS3-AMPK interaction in metabolically perturbed liver and skeletal muscle from humans and mice. Across fasting, high-fat diet feeding and exercise conditions, co-immunoprecipitation assays failed to detect endogenous SAPS3-AMPK and PP6C-AMPK interactions. These findings challenge the physiological relevance of SAPS3/PP6C as regulators of AMPK in mature tissues and highlight the need for further investigation into the regulation of AMPK by protein phosphatases in vivo.
    Keywords:  AMPK; AMP‐activated protein kinase; PP6C; PPP6C; SAPS3; liver; metabolism; protein interaction; protein phosphatase; skeletal muscle
    DOI:  https://doi.org/10.1002/1873-3468.70222
  16. Curr Obes Rep. 2025 Nov 19. 14(1): 80
       PURPOSE OF REVIEW: This review comprehensively highlights the molecular and cellular pathways that link insulin resistance (IR) to muscle atrophy.
    RECENT FINDINGS: Skeletal muscle IR is a key driver of muscle atrophy in obesity. It disrupts metabolic homeostasis, leading to impaired glucose uptake and utilization. Crucially, IR shifts the balance in skeletal muscle from anabolic to catabolic processes by simultaneously inhibiting protein synthesis and promoting proteolysis, resulting in a progressive decline of muscle mass and function. This review summarizes how defective insulin signaling activates a cascade of intracellular events that accelerate muscle wasting in obese individuals. The nexus between IR and muscle atrophy in obesity involves multiple interconnected mechanisms, including mitochondrial dysfunction, elevated inflammation and oxidative stress, and compromised satellite cell function-essential for muscle repair and regeneration. The prevalence of IR, which rises with age and is exacerbated by sedentary behavior and poor nutrition, underscores the importance of understanding these signaling pathways. Elucidating these mechanisms is critical for developing effective interventions to combat muscle loss and enhance metabolic health in the obese population.
    Keywords:  Body fat; Endocrines; Muscle atrophy; Protein breakdown; Protein synthesis
    DOI:  https://doi.org/10.1007/s13679-025-00672-6
  17. Front Endocrinol (Lausanne). 2025 ;16 1700668
      Perilipins are essential structural proteins localized on the surface of lipid droplets, with perilipin 2, 3, and 5 exhibiting specific expression in skeletal muscle. Intramuscular lipids are predominantly stored within lipid droplets, tightly regulated by perilipins. Perilipin 3 primarily governs lipid droplet biogenesis, whereas Perilipin 2 and PLIN5 play critical roles in mediating lipolysis through lipid droplet-organelle interactions and in responding to exercise-induced signaling cascades. Acute exercise selectively depletes lipid droplets coated with Perilipin 2 and Perilipin 5 while inducing subcellular relocalization of perilipin proteins. In contrast, moderate-intensity continuous training and high-intensity interval training elicit adaptive alterations in skeletal muscle perilipins protein expression: Moderate-intensity continuous training significantly upregulates perilipin 2 and 5 expression, whereas high-intensity interval training specifically enhances perilipin5 expression, fostering enhanced physical interactions between lipid droplets and mitochondria, thereby mitigating ectopic lipid accumulation. This study elucidates the intricate regulatory mechanisms of perilipin-mediated organelle interactions under diverse exercise modalities and their contributions to optimizing skeletal muscle lipid metabolism, providing a robust theoretical framework for developing targeted exercise-based interventions and potential therapeutic targets for metabolic disorders.
    Keywords:  exercise; lipid metabolism; organelle interaction; perilipin; skeletal muscle
    DOI:  https://doi.org/10.3389/fendo.2025.1700668
  18. Adv Sci (Weinh). 2025 Nov 20. e16719
      Iron is critical to optimal athletic performance because of its role in energy metabolism, oxygen transport, and acid-base balance. However, the precise mechanism how skeletal muscle maintains iron homeostasis during exercise remains enigmatic. Here, it is demonstrated that the HECT-domain containing ubiquitin ligase E3 Huwe1 (also known as MULE or ARF-BP1) in skeletal muscle is suppressed upon exhausted exercise. Loss of Huwe1 in skeletal muscle restrains the exercise performance of Huwe1 conditional knockout (cKO) mice, accompanied with pronounced oxidative stress. Mechanistically, Huwe1 depletion stabilizes c-Myc protein, leading to upregulated sarcolipin (Sln) expression with inhibited SarcoEndoplasmic Reticulum Calcium ATPase (SERCA) activity, and downregulated ferroportin (Fpn, also known as Slc40a1) expression with iron overload. Silencing of c-Myc restores SERCA activity and iron export. Consistently, SERCA activator CDN1163, Sln silencing, or dietary iron restriction ameliorates the exercise performance of Huwe1 cKO mice. Of note, improved exercise performance is accompanied with diminished oxidative stress in Huwe1 cKO mice upon iron restriction. Taken together, the results unveil a key function for HUWE1 in skeletal muscle as a fundamental coordinator of iron and calcium homeostasis by regulating SERCA activity and iron metabolism. These findings reveal a regulatory pathway on controlling iron/calcium homeostasis and exercise capacity.
    Keywords:  Exercise; HUWE1; Iron overload; Oxidative stress; SERCA
    DOI:  https://doi.org/10.1002/advs.202516719
  19. J Cachexia Sarcopenia Muscle. 2025 Dec;16(6): e70119
       BACKGROUND: Mechanical inactivity rapidly induces diaphragm muscle fibres' contractile dysfunction and atrophy. Diaphragm weakness can impair respiratory function, quality of life and increase risks of morbidity and mortality. Muscle RING-finger protein-1 (MuRF1) expression is upregulated during denervation and muscle inactivity and is known to target key muscle proteins for degradation. We previously reported that the small-molecule targeting MuRF1 (MyoMed-205) protects against diaphragm contractile dysfunction and atrophy after 12 h of unilateral diaphragm denervation (UDD) in rats. In this study, we investigated the mechanisms by which MyoMed-205 protects the diaphragm structure and function during early UDD in rats.
    METHODS: Male Wistar rats were subjected to unilateral diaphragm denervation (UDD) for 12 h. Immediately after UDD, rats received either a placebo (vehicle) or small-molecule targeting MuRF1 (MyoMed-205, 50 mg/kg bw), and outcomes were compared with sham-operated controls. Diaphragm was used for histological, morphometric, transcriptomic (RNA-seq) and protein content (Western blot) analysis.
    RESULTS: UDD induced diaphragm slow- (Type I: p = 0.03) and fast-twitch (Type IIa: p = 0.04; Type IIb/x: p = 0.02) fibres atrophy after 12 h, which was prevented by MyoMed-205 (p < 0.05). Mechanistically, UDD perturbed mechanisms involved with myofibre ultrastructure and contractility, mitochondrial function, proteolysis and tissue remodelling in the diaphragm. MyoMed-205 enhanced the activation of mechanisms required for sarcomere integrity, calcium handling, antioxidant defence, chaperone-mediated unfolded protein response and muscle growth. MyoMed-205 also mitigated intramuscular fat deposition and pro-fibrotic responses triggered by UDD.
    CONCLUSIONS: Small-molecule targeting MuRF1 (MyoMed-205) protects against diaphragm muscle contractile dysfunction and atrophy after 12 h of UDD. Herein, we demonstrate that this protective effect involved augmented activation of signalling pathways controlling muscle structure and function, chaperone-mediated unfolded protein and muscle growth, while mitigating intramuscular fat deposition and pro-fibrotic responses triggered by UDD at the transcriptional and/or protein level.
    Keywords:  MyoMed‐205; PI3K‐Akt–mTOR pathway; TRIM63; mechanical unloading; skeletal muscle; ubiquitin‐proteasome system; unilateral diaphragm denervation
    DOI:  https://doi.org/10.1002/jcsm.70119
  20. Sci Rep. 2025 Nov 22.
      Duchenne muscular dystrophy (DMD) is characterized by progressive skeletal muscle degeneration and weakness. DMD is the most common muscular dystrophy; commonly diagnosed in childhood, it has a deadly outcome, typically for respiratory or cardiac failure. In spite of a longer disease course, obtained in recent years thanks to palliative pharmacological treatments, DMD remains one of the foremost public health challenges. The intricate interaction between dysfunctional skeletal muscle fibers and the resident different cell types, including inflammatory and fibroadipogenic progenitors, directly contributes to the progression of the disease, affecting its severity. In this study, we used the remarkable precision of X-ray phase-contrast tomography (XPCT) to conduct an unprecedented three-dimensional (3D) examination of skeletal muscle architecture, comparing healthy and dystrophic mdx mice. The morphological features observed in XPCT images were compared to conventional histological sections, corroborated by morphometric evaluation of bidimensional parameters. Through XPCT, we followed the spatial disposition of degenerating or distorted dystrophic myofibers along a length of approximately 1.3 mm. In addition, the exploitation of micro-XPCT, unveiled significant quantitative differences between healthy and mdx muscles in numerous 3D parameters, such as myofiber length, variability in myofiber caliber, myofiber volume, and the volume of interstitial tissues and cells. We propose XPCT as a novel imaging tool for the ex vivo characterization of the fine architecture of dystrophic muscles: this approach is particularly relevant to highlight the outcomes of a treatment in pre-clinical models and provides the structural bases underlying the functional features of the diseased skeletal muscle.
    DOI:  https://doi.org/10.1038/s41598-025-28961-z
  21. Am J Physiol Cell Physiol. 2025 Nov 19.
      At the neuromuscular junction, nicotinic acetylcholine receptor (nAChR) dynamics is regulated in a nerve- and activity-dependent manner. Correlated local alterations in myoplasmic [Ca2+]i, induced by IP3-sensitive subsynaptic Ca2+ stores, have been proposed to signal motor endplate adaptation to motor neuron stimulation. Accordingly, there is evidence for a modulatory role of Ca2+/calmodulin-dependent protein kinase IIβ (CaMKIIβ) in the sorting, targeting, and/or incorporation of nAChRs into the postsynaptic membrane. As the scaffold protein Homer 2 emerges as a key player in integrating downstream postsynaptic signaling pathways, this study investigated the possible involvement of Homer 2 in the molecular mechanism controlling nAChR dynamics. Using Homer 2-/- transgenic mice, it was found that Homer 2 ablation leads to a chronic adaptation of the endplate characterized by: 1) reduction in nAChR activity due to slower insertion of nAChRs into the endplate; 2) reduced subsynaptic IP3R1 content and IP3-releasable Ca2+; and 3) impaired colocalization of CaMKIIβ with nAChRs. Overall, the present results demonstrate that Homer 2 ablation produces a significant alteration in endplate nAChR dynamics, which is associated with impaired organization of the subsynaptic IP3-driven Ca2+ signaling mechanism.
    Keywords:  Homer 2; endplate; neuromuscular junction; plasticity
    DOI:  https://doi.org/10.1152/ajpcell.00419.2025
  22. Skelet Muscle. 2025 Nov 21. 15(1): 32
       BACKGROUND: Skeletal muscle is a dynamic tissue capable of structural and metabolic remodeling in response to physiological and pathological stimuli. These adaptations are central to understanding the mechanisms underlying conditions such as genetic myopathies, cancer, aging, and recovery from injury. Muscle fiber characterization-assessing fiber type, size, and metabolic profile-is essential for such studies. However, conventional histological methods often rely on serial tissue sections and multiple staining protocols, which are time-consuming, require significant biological material, and introduce methodological bias.
    METHODS: We developed FLASH (Fluorescence-based Labeling for Assessing Skeletal muscle Histology), a novel methodology combining enzymatic (SDH or GPDH) and quadruple fluorescent labeling (Laminin, MYH4, MYH2, MYH7) on a single muscle section. The resulting images were analyzed using a custom macro in Fiji/ImageJ, integrating the Cellpose segmentation algorithm. This automated pipeline detects individual muscle fibers, quantifies their cross-sectional area (CSA), identifies fiber types based on myosin isoform expression, and measures enzymatic staining intensity. Batch analysis was implemented to process entire image folders automatically. Validation was performed by comparing automated fiber detection with expert manual segmentation using correlation analysis and Bland-Altman plots.
    RESULTS: The FLASH method allowed simultaneous assessment of both contractile and metabolic properties within individual fibers on the same section, removing the need for serial cuts. The automated image analysis achieved high accuracy in fiber detection (r > 0.95 compared to manual annotation) and produced consistent CSA and fiber-type quantification, even under suboptimal staining conditions. The macro enabled significant time savings by automating the complete analysis workflow, including ROI generation and Excel data export for each image.
    CONCLUSIONS: FLASH provides an efficient and robust tool for high-throughput skeletal muscle histology. By combining enzymatic and fluorescent co-labeling with machine learning-based image analysis, this method improves reproductibility, reduces experimental complexity, and minimizes user bias. FLASH is particularly well-suited for large-scale or longitudinal studies investigating muscle adaptation in health and disease.
    DOI:  https://doi.org/10.1186/s13395-025-00401-6
  23. Am J Physiol Cell Physiol. 2025 Nov 19.
      Rest is generally required for full muscle regeneration after an injury; however, rehabilitative activity is often used after injury to attempt a faster recovery. While rehabilitative activity can enhance muscle regeneration, there is also a risk that returning to vigorous muscle contractions too early after sustaining an injury, could reinjure the muscle, and negatively impact full muscle regeneration. It is not known whether MT added to rehabilitative muscle activity would speed regeneration of muscle morphology more rapidly than resting during the recovery period. Therefore, submaximal electrically evoked isometric contractions (EC) were given to injured muscles of MT treated mice, to test the hypothesis that MT would attenuate the negative regenerative effects of EC and improve the restoration of muscle mass and morphology after muscle injury. Cardiotoxin (CTX) was injected into the tibialis anterior (TA) muscle of one limb of C57BL/6 mice at 8-12 weeks of age to induce muscle injury. Systemic delivery MT or PBS was administered to the mice 48 h after injury. The TA received EC at 40Hz every other day for up to 14-days after CTX injury. While EC-induced mechanical injury slowed muscle repair, muscle fiber regeneration and nuclear domain size was improved by MT. The percentage of collagen and other non-contractile tissue was elevated in CTX-injured and EC treated muscles; however, MT reduced fibrosis/non-contractile tissue deposition in regenerating muscles. Our results provide evidence that systemic mitochondria delivery can improve muscle repair and can attenuate contraction-suppressed muscle fiber regeneration during recovery after injury.
    Keywords:  fibrosis; mitochondria; muscle fiber types; muscle injury; regeneration
    DOI:  https://doi.org/10.1152/ajpcell.00744.2025
  24. bioRxiv. 2025 Oct 03. pii: 2025.10.02.679855. [Epub ahead of print]
      Cardiorespiratory fitness (CRF) is a heritable trait associated with improved metabolic health and reduced mortality. To identify regulatory mechanisms associated with CRF variation, we generated and integrated 546 transcriptomic and epigenomic (chromatin accessibility and histone marks) profiles from 128 genetically heterogeneous rats selectively bred for high and low running capacity, a model that mirrors CRF-associated traits in humans. We found that selection for high CRF led to genetic convergence in skeletal muscle regulatory regions linked to genes involved in lipid metabolism and angiogenesis. We validated thousands of these genetic effects through generation and integration of 426 genotype, gene expression, and chromatin accessibility profiles in an independent HCR×LCR F2 population (n=147). Together, these 972 omics profiles show that genetic variation reshapes the chromatin landscape to support energy metabolism and oxygen delivery in skeletal muscle and overall CRF, offering a molecular framework to identify targets that reduce cardiometabolic disease risk.
    DOI:  https://doi.org/10.1101/2025.10.02.679855
  25. Am J Physiol Cell Physiol. 2025 Nov 20.
      Acute exercise increases energy demand in skeletal muscle and releases metabolic intermediates into circulation, yet the serum kinetics of exercise-mobilized metabolites remain poorly characterized. By applying high-frequency serial blood sampling and targeted metabolomics in a longitudinal exercise trial with 12 young, healthy adults (6 female, 6 male), we assessed temporal alterations in energy-related metabolites during acute aerobic exercise and after 1 hour of recovery. We provide evidence for 42 exercise-responsive metabolites, including end products of glycolysis, tricarboxylic acid cycle intermediates, ketone bodies and amino acids. Overall, the observed metabolic alterations closely resembled skeletal muscle energy metabolism, thereby refining fundamental principles of exercise biochemistry through detailed serum kinetics, including novel, so far uncharacterized responses in systemic energy homeostasis and inter-organ crosstalk.
    Keywords:  energy metabolism; exercise; inter-organ crosstalk; metabolomics; systemic communication
    DOI:  https://doi.org/10.1152/ajpcell.00715.2025
  26. Front Microbiol. 2025 ;16 1695448
      Skeletal muscle wasting disorders, such as sarcopenia and cachexia, pose a significant clinical challenge. The gut-muscle axis, a bidirectional signaling network, is now understood to be a critical regulator of muscle homeostasis, with the gut microbiota functioning as a key metabolic organ. Physical activity is a cornerstone intervention, exerting benefits by directly stimulating muscle and by favorably modulating the composition and metabolic output of the gut microbiota. This review synthesizes the molecular mechanisms of muscle wasting and the pathways of the gut-muscle axis, with a specific focus on microbial metabolites like short-chain fatty acids (SCFAs). We analyze how different exercise modalities modulate this system and critically evaluate evidence from human trials. By identifying key research gaps, this review argues for a paradigm shift toward integrated, personalized interventions that combine targeted exercise with nutritional and microbial strategies to more effectively combat muscle wasting disorders.
    Keywords:  cachexia; gut microbiota; gut-muscle axis; physical activity; sarcopenia; short-chain fatty acids; skeletal muscle wasting
    DOI:  https://doi.org/10.3389/fmicb.2025.1695448
  27. Sci Rep. 2025 Nov 19. 15(1): 40805
      Excessive muscle protein synthesis causes skeletal muscle hypertrophy. Essential amino acids are substrates for muscle proteins and stimulate muscle protein synthesis. Several essential amino acids are taken up into muscle cells through L-type amino acid transporter 1 (LAT1). However, LAT1 may influence protein synthesis in an amino acid uptake-independent manner. Here, we investigated the effects of LAT1 inhibition on protein synthesis in C2C12 myotubes and the associated mechanisms. JPH203 (50 μM), a selective inhibitor of LAT1, stimulated protein synthesis without changing expression of phosphorylated p70S6K (T389) and 4EBP1 (T37/46), an indicator of mTORC1 activity. Culturing in amino acid-free media did not suppress JPH203-induced protein synthesis. The mTORC1 inhibitor rapamycin (100 nM) did not suppress JPH203-induced protein synthesis. ATP-competitive mTOR inhibitor AZD8055 (1 μM) suppressed JPH203-induced protein synthesis. JPH203 treatment increased intracellular glutamine concentration. These results suggest that inhibition of LAT1 function augments muscle protein synthesis, possibly through the activation of rapamycin-insensitive mTOR signaling; elevated intracellular glutamine levels may contribute to the enhancement of muscle protein synthesis induced by LAT1 inhibition.
    Keywords:  L-type amino acid transporter 1; Leucine; Mechanistic target of rapamycin; Muscle protein synthesis; Myotubes
    DOI:  https://doi.org/10.1038/s41598-025-24534-2
  28. Int J Sport Nutr Exerc Metab. 2025 Nov 20. 1-8
      High-intensity interval training (HIIT) may elicit different skeletal muscle responses compared to work-matched moderate-intensity continuous training (MICT). The effect of work-matched HIIT versus MICT on myofibrillar protein synthesis remains to be determined. In the present study, we assessed the effect of short-term HIIT versus MICT on myofibrillar protein synthesis rates using a single-leg within-participant design. Ten healthy young men (age: 20 ± 1 years) performed six to eight training sessions with each leg over 2 weeks while ingesting deuterated water to assess myofibrillar protein synthesis. One leg was randomly assigned to perform HIIT and the other MICT. Skeletal muscle biopsies were collected at rest from one leg before and after a 2-week habituation period and from both legs after the training period to assess myofibrillar protein synthesis rates. HIIT and MICT increased single-leg maximal power output (main effect, p < .01), with no differences between legs (interaction: p = .61). Myofibrillar protein synthesis rates did not differ between the habituation period, MICT, or HIIT (1.39 ± 0.16%, 1.24 ± 0.30%, and 1.42 ± 0.31% per day, respectively; p = .29). In conclusion, we observed no detectable differences in daily myofibrillar protein synthesis rates between HIIT or work-matched MICT when assessed over a 2-week exercise training period in recreationally active young adult men.
    Keywords:  aerobic exercise; deuterated water; exercise intensity; heavy water; muscle protein synthesis
    DOI:  https://doi.org/10.1123/ijsnem.2025-0074
  29. J Physiol. 2025 Nov 18.
      
    Keywords:  GLP‐1 receptor agonists; adipose tissue; obesity; skeletal muscle
    DOI:  https://doi.org/10.1113/JP290350
  30. Biomed Pharmacother. 2025 Nov 19. pii: S0753-3322(25)01008-X. [Epub ahead of print]193 118814
      Sarcopenia is currently considered a systemic condition that goes beyond muscle atrophy to include multifunctional metabolic and cardiovascular dysfunction. The mediators between skeletal muscle loss and entire body insulin resistance and increased vulnerability to cardiotoxicity caused by chemotherapy are not clear. We hypothesise that mitochondrial-enriched, muscle-secreted extracellular vesicles (EVs) of mtDNA/mitoproteins, stress-regulated microRNAs (miR-1/133/206; miR-29 family), and ROS-modified damage-associated molecular patterns (DAMPs) are a mitochondrial stress bridge that secretes danger signals from sarcopenic muscle to the liver/adipose and heart. EV cargo mechanistically impairs insulin signaling (IRS-1 → PI3K-AKT → GLUT4) and cardiomyocyte pre-injury (loss of Δpsm, antioxidant repression, apoptosis), increasing the toxicity of doxorubicin. Should this framework be valid, it describes the clustering of sarcopenic patients with metabolic dysfunction and disproportional cardiotoxic incidents throughout cancer therapy and places circulating EV cargo as an indicator of outcomes and therapeutic interventions.
    Keywords:  Cardiotoxicity; Chemotherapy; Extracellular vesicles; Insulin resistance; Mitochondrial stress; Sarcopenia
    DOI:  https://doi.org/10.1016/j.biopha.2025.118814
  31. J Appl Physiol (1985). 2025 Nov 20.
      This study aimed to elucidate the effects of repetition duration (contraction duration for each repetition) of resistance exercise on muscle hypertrophy and its underlying mechanisms using a rat exercise model. Male Sprague-Dawley rats were randomly assigned to three groups trained with short (S), medium (M), and long (L) repetition durations. During resistance exercise, the right gastrocnemius muscles were electrically stimulated to induce maximal tetanic contractions, each lasting for 1, 3, and 9- sec in S, M, and L groups, respectively. The number of contractions in each set and the interset rest duration were the same across groups, while the number of sets was adjusted to match the total torque-time integral of plantar flexion. The left (untrained) and right (trained) gastrocnemius muscles were sampled 48 h after 12 exercise bouts. The average and peak torque during each exercise were consistently highest in S and lowest in L. The muscle mass and fiber cross-sectional area significantly increased in S and M but not in L. The concentrations of total RNA and 18S+28S rRNA increased only in S and were correlated with muscle mass when the three groups were combined. We also sampled muscle tissues 6 h after a single exercise bout and found no significant difference in muscle protein synthesis, mTOR signaling activity, ribosome biogenesis, or protein degradation between the groups. These results suggest that long repetition duration does not promote but rather diminishes the hypertrophic effects of resistance exercise and that acute molecular responses to resistance exercise cannot predict such effects.
    Keywords:  electrical stimulation; mechanistic target of rapamycin; protein synthesis; resistance training; ribosome
    DOI:  https://doi.org/10.1152/japplphysiol.00956.2024
  32. J Physiol. 2025 Nov 16.
      Training with low carbohydrate availability can increase AMP-activated protein kinase (AMPK) activation, but whether increased AMPK activation is the result of low carbohydrate availability per se or concurrent increases in fatty acid availability/oxidation is unclear. This study assessed the independent effects of carbohydrate and fatty acid availability on exercise-induced skeletal muscle AMPK activation and downstream signalling. Eight active males who were aged between 18 and 60 years with a body mass index in the range 18.0-30.0 kg m-2 cycled on three occasions for 60 min at 95% of lactate threshold 1 with ingestion of either carbohydrate (CARB), niacin (NIACIN) or placebo (FAST) in a crossover design (11 ± 6 days washout). Blood and exhaled breath were sampled throughout exercise and muscle was sampled pre- and post-exercise. Fat oxidation and plasma non-esterified fatty acid concentrations were both lower in CARB vs. FAST with negligible difference between CARB vs. NIACIN. Plasma insulin concentrations were higher in CARB compared with both FAST and NIACIN. Net muscle glycogen use was greater with NIACIN vs. CARB. Although no evidence for differences were observed for phosphorylated AMPK, the downstream target, phosphorylated acetyl-CoA carboxylase was decreased with CARB vs. both FAST (-0.7 ± 0.6 fold, P = 0.04) and NIACIN (-1.0 ± 0.8 fold, P = 0.02). RNA-sequencing displayed several canonical changes with exercise but little difference between conditions. These data suggest carbohydrate ingestion suppresses exercise-induced phosphorylation of acetyl-CoA carboxylase independent of fatty acid availability. KEY POINTS: It is currently unknown whether the enhanced physiological adaptation to regularly exercising in a fasted-state are explained by low carbohydrate availability and/or the concomitant increase in fatty acid availability. This study used fasted exercise with niacin ingestion to reduce the lipaemic response associated with fasted exercise to isolate the effects of carbohydrate vs. fatty acid availability on exercise-induced skeletal muscle signalling. Our data show niacin ingestion increases muscle glycogen utilisation compared to carbohydrate ingestion during exercise, but both niacin and carbohydrate ingestion suppress fatty acid availability and fat oxidation to a similar extent. Our data demonstrate carbohydrate ingestion during exercise suppresses acetyl-CoA carboxylase phosphorylation compared to both niacin ingestion and extended overnight fasting. These data suggest that high carbohydrate availability inhibits exercise-induced acetyl-CoA carboxylase phosphorylation in human skeletal muscle, independent of circulating fatty acid concentrations.
    Keywords:  carbohydrate metabolism; cell signalling; exercise metabolism; lipid metabolism
    DOI:  https://doi.org/10.1113/JP289864
  33. Development. 2025 Nov 20. pii: dev.204933. [Epub ahead of print]
      The capacity to detect and respond to injury is critical for the recovery and long-term survival of many organisms. Wnts are commonly induced by tissue damage but how they become activated transcriptionally is not well understood. Here, we report that mouse Wnt1 and Wnt10b are induced following injury in both lung and muscle. These Wnts occupy the same chromosome and are transcribed in opposite directions with 12kb between them. We identified a highly conserved cis-acting regulatory region (enhancer) residing between Wnt1 and Wnt10b that, when fused to a LacZ reporter, is activated post-injury. This enhancer harbors putative AP-1 binding sites that are required for reporter activity, a feature observed in other injury-responsive enhancers. Injured muscles in mice carrying a germ-line deletion of the enhancer region display reduced Wnt1 and Wnt10b expression and show elevated intramuscular adipogenesis which can be a hallmark of impaired muscle regeneration or tissue maintenance. Enhancer redundancy is common in development, but our in vivo analysis shows that loss of a single injury-responsive regulatory region in adult tissues can produce a detectable phenotype.
    Keywords:  Enhancer; Injury; Muscle; Regeneration; Wnt
    DOI:  https://doi.org/10.1242/dev.204933
  34. Front Physiol. 2025 ;16 1711795
      In May 2025, the University of Houston (UH) hosted the inaugural Muscle Biology and Cachexia conference, organized by Drs. Ashok Kumar and Radbod Darabi. The conference attracted nearly 300 participants, including established scientists, early-career researchers, and students from across the United States, Canada, Italy, Singapore, and Turkey. Research was presented through a combination of oral presentations and poster sessions. The conference was driven by the increasing interest in skeletal and cardiac muscle biology and cancer cachexia among institutions at the Texas Medical Center and surrounding universities. It served as a platform to promote knowledge exchange and foster collaboration within this growing scientific community. The conference was supported by the UH College of Pharmacy (UHCOP), Division of Research (DOR), Drug Discovery Institute (DDI), and the Department of Pharmacological and Pharmaceutical Sciences (PPS). In conjunction with the conference, UH announced the formation of the Institute of Muscle Biology and Cachexia (IMBC). The IMBC aims to strengthen collaborative research efforts and enhance understanding of the molecular and signaling pathways that regulate muscle physiology and disease.
    Keywords:  bone; bone-muscle crosstalk; cachexia; molecular and signaling pathways; muscle; muscle physiology and disease
    DOI:  https://doi.org/10.3389/fphys.2025.1711795