98%
921
2 minutes
20
Background: Skeletal muscle atrophy, which is a debilitating condition exacerbated by estrogen deficiency, lacks effective therapeutic interventions. Although ferroptosis (an iron-dependent form of cell death driven by lipid peroxidation) has emerged as a contributor to muscle degeneration, its regulatory mechanisms remain poorly defined. In this study, we identified luteolin, which is a natural flavonoid, as a potent inhibitor of ferroptosis that mitigates estrogen deficiency-induced muscle atrophy by targeting SLC7A11.
Purpose: The aim of this study was to investigate the role of ferroptosis in the anti-muscle atrophy effects of luteolin.
Methods: Via database screening, luteolin was identified as a potential drug for improving muscle atrophy, and the promotion of C2C12 myogenic differentiation by luteolin was detected by using immunofluorescence (IF), quantitative reverse transcription PCR (RT-qPCR) and western blot (WB). The mechanism of luteolin-mediated ferroptosis in muscle atrophy was confirmed by RNA-seq, transmission electron microscopy (TEM), and GSH, MDA, SOD and Fe assays. Molecular docking, molecular dynamics simulation, surface plasmon resonance (SPR), cellular thermal shift assay (CETSA), drug affinity responsive target stability (DARTS) and siRNA-mediated gene knockout were applied to validate the notion that the mechanism of luteolin treatment of muscle atrophy involves target binding to SLC7A11. In addition, this study confirmed the role of luteolin in ameliorating muscle atrophy via the modulation of the SLC7A11-mediated ferroptosis pathway in vivo. Finally, the effect of luteolin on the myogenic differentiation of HsKMCs was investigated.
Results: Luteolin promotes myogenic differentiation and significantly inhibits myotube atrophy, with the main mechanism of these effects involving the direct binding of luteolin to the SLC7A11 protein to inhibit the occurrence of ferroptosis. We confirmed that luteolin can inhibit ferroptosis in muscle tissue and improve the loss of muscle mass and strength due to muscle atrophy in vivo. In addition, luteolin significantly inhibited myotube atrophy in HsKMCs and promoted their myogenic differentiation by modulating the SLC7A11-mediated ferroptosis.
Conclusions: Our findings demonstrate that luteolin regulates myogenesis and prevents muscle atrophy through binding to SLC7A11 and subsequently inhibiting ferroptosis. This study elucidates the critical role of the SLC7A11-ferroptosis axis in preserving muscle physiology during atrophy, while identifying luteolin as a therapeutic agent capable of targeting SLC7A11 to suppress ferroptosis and alleviate muscle atrophy.
Download full-text PDF |
Source |
---|---|
http://dx.doi.org/10.1016/j.phymed.2025.156799 | DOI Listing |
World Neurosurg
September 2025
Swedish Neuroscience Institute, Seattle, WA; Seattle Science Foundation, Seattle, WA.
Introduction: Lateral Lumbar Interbody Fusion (LLIF) is based on a less-invasive access corridor through the retroperitoneum and psoas muscle, though concerns persist over postoperative weakness and neuropathy on the surgical side. This study investigates if the trans-psoas LLIF approach is associated with long-term changes in psoas morphology, hip flexor (HF) weakness, and lower extremity dysesthesia.
Methods: The authors retrospectively reviewed all LLIF cases at a single institution from January 2016 to June 2024.
Neuromuscul Disord
August 2025
Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. Electronic address:
Spinal muscular atrophy (SMA) types 2 and 3 are chronic neuromuscular disorders characterized by progressive motor impairment. Although disease-modifying therapies such as risdiplam and nusinersen have shown clinical efficacy, real-world data in pediatric populations remain limited. This prospective observational study evaluated motor function outcomes in 20 children with SMA (aged 3 to 13 years; 12 with type 2, 8 with type 3) receiving either risdiplam or nusinersen in Northwestern Iran.
View Article and Find Full Text PDFInt Immunopharmacol
September 2025
Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China.
Peripheral nerve injury (PNI) is notoriously difficult to repair due to impaired axonal regeneration and dysregulated inflammatory microenvironments. This study demonstrates that crocin facilitates peripheral nerve regeneration by modulating the STAT3/Bcl-2/Beclin-1 signaling axis, enhancing autophagy while suppressing NLRP3 inflammasome-mediated pyroptosis. In a rat model of sciatic nerve crush injury, crocin treatment improved axonal regrowth and ultrastructural remyelination, as evidenced by upregulated expression of β3-Tubulin, neurofilament-200 (NF200), and myelin basic protein (MBP), alongside significantly elevated sciatic functional index (SFI) scores, reduced muscle atrophy, and diminished collagen deposition.
View Article and Find Full Text PDFJ Med Ultrason (2001)
September 2025
Department of Emergency Medicine, Faculty of Medicine, Kindai University, Osaka, Japan.
Purpose: This study aimed to investigate muscle atrophy in critically ill patients using ultrasonography. We compared the rectus femoris (a major muscle of the lower limbs) with the sternocleidomastoid (an accessory respiratory muscle).
Methods: Thirty-four patients hospitalized at the Critical Care Medical Center of Kindai University Hospital between January 2022 and March 2023 were enrolled.
J Neurochem
September 2025
Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy.
Patients with Duchenne muscular dystrophy (DMD) may experience neurobehavioral and cognitive concerns, including psychiatric symptoms, due to the absence of full-length dystrophin (Dp427), frequently accompanied by deficiencies in shorter isoforms. The lack of dystrophin affects neurophysiological processes from the uterine phase, impacting neural circuitry in brain regions such as the prefrontal cortex, hippocampus, and cerebellum. This leads to reduced inhibitory GABAergic transmission and altered hippocampal glutamatergic signaling.
View Article and Find Full Text PDF