bims-musmir Biomed News
on microRNAs in muscle
Issue of 2025–10–12
six papers selected by
Katarzyna Agnieszka Goljanek-Whysall, University of Galway



  1. J Cachexia Sarcopenia Muscle. 2025 Oct;16(5): e70090
       BACKGROUND: The coupling of oxygen consumption to ATP synthesis via oxidative phosphorylation (OXPHOS) is central for cellular energy homeostasis. Some studies suggest exercise training increases the efficiency of ATP synthesis, but the molecular mechanisms are unclear. We have previously shown that exercise remodels the lipid composition of mitochondrial membranes, and some of these changes in mitochondrial lipids might influence OXPHOS efficiency (ATP produced per O2 consumed, referred to as P/O). Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a transcriptional co-activator that coordinately regulates exercise-induced adaptations, including mitochondria. We hypothesized that increased PGC-1α activity might remodel mitochondrial membrane lipids and promote energy efficiency.
    METHODS: Mice with skeletal muscle-specific overexpression of PGC-1α (MCK-PGC-1α) and their wildtype littermates were used for this study. Lipid mass spectrometry and quantitative PCR were used to assess muscle mitochondrial lipid composition and their biosynthesis pathway. The abundance of OXPHOS enzymes was determined by Western blotting. High-resolution respirometry and fluorometry analyses were performed to characterize mitochondrial bioenergetics (ATP production, O2 consumption and P/O) for permeabilized fibres and isolated mitochondria. Respiratory supercomplexes were assessed by blue native PAGE.
    RESULTS: Lipidomic analyses of skeletal muscle mitochondria from wildtype and MCK-PGC-1α mice revealed that PGC-1α increases the concentrations of cone-shaped lipids such as phosphatidylethanolamine (PE; +25%, p < 0.0001), cardiolipin (CL; +184%, p < 0.0001) and lysophospholipids (+34%-94%, all p < 0.01), while decreasing the concentrations of phosphatidylcholine (PC; -4%, p = 0.0020), phosphatidylinositol (PI; -17%, p < 0.0001) and phosphatidic acid (PA; -35%, p < 0.0001). However, while PGC-1α overexpression increased the abundance of OXPHOS enzymes (two- to fourfold, p < 0.0001), the rate of O2 consumption (1.5-fold, p = 0.0030), or the respiratory supercomplexes (~1.5-fold, p < 0.01), P/O values were unaffected by PGC-1α overexpression in permeabilized fibres or isolated mitochondria.
    CONCLUSIONS: Collectively, overexpression of PGC-1α promotes the biosynthesis of mitochondrial PE and CL, but neither PGC-1α nor the mitochondrial membrane lipid remodelling induced in MCK-PGC-1α mice is sufficient to increase the efficiency of mitochondrial ATP synthesis. These findings indicate that PGC-1α-dependent mechanisms or changes in mitochondrial membrane lipids may be insufficient to alter P/O. While muscles from MCK-PGC-1α mice are known not to completely phenocopy adaptations with exercise training, our findings also highlight that there is a need to examine whether exercise training indeed improves P/O in mouse skeletal muscle.
    Keywords:  exercise; mitochondria; phospholipids; skeletal muscle
    DOI:  https://doi.org/10.1002/jcsm.70090
  2. BMC Genomics. 2025 Oct 09. 26(1): 898
       BACKGROUND: Sarcopenic obesity (SO) is the combined condition of sarcopenia and obesity, which commonly occurs in the older adult population. SO is associated with poor physical function, increased risk of musculoskeletal injury, and reduced quality of life and independence. Information on the molecular underpinnings of this condition is limited. The aim of this study was to provide a transcriptomic analysis characterizing SO induced by lifelong obesity and to provide a deeper understanding of the etiology of the condition.
    RESULTS: Young (Y) (6 mo) and aged (A) (21-24 mo) mice were fed either normal chow (L) (12% kcal from fat) or high-fat (O) (60% kcal from fat) diets ad libitum. Through multiple analyses, we observed that genes related to ECM remodeling were downregulated in AO compared with YO, providing insight into the effects of age in a life-long obese condition. Furthermore, we observed that genes related to contraction in slow-twitch muscle fibers and fast-to-slow muscle fiber type transitions were upregulated with obesity in the aged condition.
    CONCLUSION: Taken together, our findings reveal specific pathways of dysregulation in SO skeletal muscle, offering molecular insights that enhance our understanding of the underlying mechanisms contributing to impaired muscle function in this condition. Further exploration of these dysregulations is critical to identifying therapeutic treatments targeting the source of muscle functional impairment in SO.
    Keywords:  Extracellular matrix; Fibrosis; Muscle contraction; Sarcopenic obesity; Slow-twitch fiber type transition
    DOI:  https://doi.org/10.1186/s12864-025-12080-0
  3. Cell Signal. 2025 Oct 06. pii: S0898-6568(25)00575-3. [Epub ahead of print]136 112160
       OBJECTIVE: MicroRNAs (miRNAs) post-transcriptionally regulate gene expression by targeting mRNA 3'UTRs, critically influencing skeletal muscle development. While miR-133a-3p is implicated in myogenesis, its regulatory interplay with RNA-binding proteins (RBPs) in porcine skeletal muscle satellite cells (PSCs) remains unexplored. This study elucidates the functional role and molecular mechanism of miR-133a-3p through a novel hnRNPK/miR-133a-3p/UCP2 axis in PSCs.
    METHODS: RT-qPCR and western blot were used for gene expression analysis. miR-133a-3p mimic and miR-133a-3p inhibitor were conducted to overexpression and knockdown the expression of miR-133a-3p in PSCs, respectively. The hnRNPK overexpression vector was designed using the pcDNA3.1(+) vector. 5-ethynyl-2'-deoxyuridine (EdU) assays were conducted for cell proliferation. Immunofluorescence detection was employed for cell differentiation analysis. The dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and biotin-labeled miRNA pull-down assays were utilized for regulatory mechanism analysis of hnRNPK/miR-133a-3p/ UCP2.
    RESULTS: Transcriptional profiling revealed miR-133a-3p upregulation during PSCs differentiation. Gain- and loss-of-function assays demonstrated that miR-133a-3p suppresses proliferation but promotes differentiation; its inhibition yielded opposite effects. Comprehensive mechanistic studies (dual-luciferase, RIP, and biotin pull-down assays) identified hnRNPK as a direct binding partner of miR-133a-3p. Subsequent validation confirmed UCP2 as a downstream target, with hnRNPK attenuating miR-133a-3p-mediated UCP2 repression. Notably, hnRNPK antagonized miR-133a-3p's pro-differentiation effects, revealing its role as a negative myogenesis regulator.
    CONCLUSION: Collectively, we report a novel hnRNPK/miR-133a-3p/UCP2 axis governing mitochondrial gene expression and PSCs differentiation. This work advances understanding of RBP-miRNA interaction in post-transcriptional myogenic regulation and provides new targets for muscle biology and regenerative medicine.
    Keywords:  RNA-binding protein; UCP2; hnRNPK; miR-133a-3p; myogenic differentiation; skeletal muscle satellite cells
    DOI:  https://doi.org/10.1016/j.cellsig.2025.112160
  4. Am J Physiol Cell Physiol. 2025 Oct 06.
      Cancer cachexia causes skeletal muscle wasting and metabolic dysfunction, worsening clinical outcomes in colorectal cancer (CRC). This study examines microscopic and macroscopic skeletal muscle fiber characteristics, and muscle volume in cachectic and non-cachectic CRC patients compared to healthy controls (HCs), and explores how these factors relate to physical performance. In total, 12 cachectic CRC patients, 25 non-cachectic CRC patients, and 25 HCs were included. Cachexia was determined by weight loss and Cachexia Staging Score. Biopsies from the vastus lateralis and erector spinae muscles were analyzed using immunohistochemistry for muscle fiber type cross-sectional area (CSA) and distribution, myonuclear content, and capillary density. Muscle volume was assessed using three-dimensional ultrasound, and CSA and density by computerized tomography scans. Physical function was evaluated with the Short Physical Performance Battery test, handgrip strength, and the Physical Activity Scale for Individuals with Physical Disabilities. Quality of life was assessed using the 36-item Short Form Survey. Cachectic CRC patients showed reduced type II muscle fiber cross-sectional area in the vastus lateralis compared to HCs and non-cachectic CRC patients. Non-cachectic CRC patients exhibited a slow-to-fast muscle fiber shift compared to HCs. Myonuclear content was lower in both cancer groups. Muscle volume and density were reduced in cachectic CRC patients. Positive correlations were found between microscopic and macroscopic skeletal muscle characteristics, muscle strength, physical performance, and quality of life, respectively. CRC patients, especially those with cachexia, showed type II muscle fiber atrophy, reduced myonuclear content, and impaired physical function, emphasizing the need for targeted prehabilitation interventions.
    Keywords:  Cachexia; Colorectal cancer; Cross-sectional area; Muscle Atrophy; Muscle fiber typing
    DOI:  https://doi.org/10.1152/ajpcell.00533.2025
  5. Sci Rep. 2025 Oct 06. 15(1): 34810
      Sarcopenia and obesity, two prevalent metabolic disorders in aging populations, often coexist and share overlapping pathophysiological mechanisms, yet the molecular mechanisms underlying their comorbidity remain elusive. This study aimed to identify key gene expression signatures and pathways underlying their comorbidity through integrative transcriptomic and bioinformatics analyses. Gene expression datasets from sarcopenia (GSE111016, skeletal muscle) and obesity (GSE152991, adipose tissue) were downloaded from the GEO database. Differentially expressed genes (DEGs) were identified using the limma package, and 208 common differentially expressed genes (CDEGs) were selected via Venn diagram intersection. Functional enrichment analyses (GO and KEGG) were performed to explore shared biological processes and pathways. A protein-protein interaction (PPI) network was constructed using STRING and Cytoscape, and key CDEGs were identified via ten topological algorithms (e.g., MCC, Degree) in the CycloHubba plugin. Pearson correlation analysis and qPCR were used to validate gene co-expression patterns and expression levels in tissue samples. GO and KEGG analyses revealed that CDEGs were significantly enriched in mitochondrial oxidative phosphorylation, electron transport chain, and thermogenesis pathways, with overlap in neurodegenerative disease pathways. The PPI network and multi-algorithm integration identified four key CDEGs: SDHB, SDHD, ATP5F1A, and ATP5F1B, all of which are components of mitochondrial respiratory chain complexes. These genes exhibited strong positive correlations (r > 0.86, p < 10⁻¹²) in both datasets and were significantly downregulated in sarcopenia and obesity tissues, as validated by qPCR. This study confirms mitochondrial dysfunction, particularly impaired oxidative phosphorylation, as a common pathological mechanism linking sarcopenia and obesity. The key genes SDHB, SDHD, ATP5F1A, and ATP5F1B represent potential therapeutic targets for managing these comorbid metabolic disorders. Future research should explore their functional roles in energy metabolism and cross-tissue crosstalk to develop targeted interventions.
    Keywords:  Bioinformatics; Mitochondrial dysfunction; Obesity; Oxidative phosphorylation; Sarcopenia
    DOI:  https://doi.org/10.1038/s41598-025-18824-y
  6. J Physiol. 2025 Oct 09.
      The mitochondrial Ca2+ uniporter (MCU) links energy metabolism to cell excitability and signalling throughout the lifespan. However, whether neural metabolism responds to MCU impairments in a sex-specific manner has remained unknown, especially in models with partial MCU downregulation. Using hippocampal slices from adult heterozygous Mcu knock-out (hKO) mice, we observed sexually dimorphic changes in NAD(P)H autofluorescence dynamics following neuronal stimulation. In male mice, these signals were preserved despite decreased mitochondrial Ca2+ uptake, likely due to increased MDH2 levels and potentially other enzymes from the tricarboxylic acid cycle, the malate aspartate shuttle, and glycolysis. In contrast to males, neural tissue from female hKO mice showed delayed NAD(P)H production and limited NAD+ availability when compared to sex-matched controls, despite intact mitochondrial Ca2+ uptake. In addition, both male and female hKO mice exhibit decreased NADP+ levels and GSH/GSSG ratios (along with increased protein S-glutathionylation), indicating a weakened antioxidant capacity. Strikingly, markers of oxidative damage were also decreased (albeit more prominently in male mice), suggesting attenuated generation of reactive oxygen species. In addition, sex-specific changes in the hippocampal metabolome were manifested in hKO mice, along with a common decrease in spermidine levels. However, spermidine-dependent hypusination of eIF5A remained unaltered, suggesting further compensatory mechanisms at this age. In summary, our findings indicate that brain tissue can adapt to partial MCU deficits by salvaging most mitochondrial NADH production in active states, while compromising redox signalling and the polyamine pathway. The interplay between these molecular phenotypes likely impacts neurological conditions and potentially cognitive impairment with age. KEY POINTS: The inactivation of one Mcu allele (which encodes the mitochondrial Ca2+ uniporter) leads to altered neuronal excitability and attenuated mitochondrial Ca2+ elevations in active neurons from 6- to 12-months-old female and male mice, respectively. Tissue autofluorescence imaging reveals delayed mitochondrial NAD(P)H production in stimulated hippocampal tissue from female but not male heterozygous Mcu knockout mice. Mitochondrial Ca2+ uniporter haploinsufficiency is characterized by a sex-specific decrease in oxidative stress markers in the brain, despite a decline in NADP+ levels and the GSH/GSSG ratio in both male and female mice. Changes in the abundance of enzymes and polar metabolites in brain tissue reveal sexually dimorphic metabolic remodelling in the context of Mcu haploinsufficiency. Life-long downregulation of the mitochondrial Ca2+ uniporter results in decreased hippocampal spermidine levels in adult male and female mice.
    Keywords:  NAD(P)H; brain metabolism; calcium; hippocampus; mitochondria; sexual dimorphism; spermidine
    DOI:  https://doi.org/10.1113/JP287618