bims-midomi Biomed News
on MDM2 and mitochondria
Issue of 2025–08–31
six papers selected by
Gavin McStay, Liverpool John Moores University



  1. Cancer Med. 2025 Sep;14(17): e71146
       BACKGROUND: This study investigates the mechanisms by which GNL3L influences ESCC progression.
    METHODS: GNL3L expression was analyzed via immunohistochemistry in ESCC tissues. Cell proliferation (EdU and CCK8 assays), migration, invasion (wound healing and Transwell assays), cell cycle, and apoptosis (flow cytometry) were assessed. Levels of GNL3L, MDM2, p53, and p21 were evaluated by qRT-PCR and western blot. Tumor growth was observed in nude mice injected with TE-1 cells.
    RESULTS: GNL3L was upregulated in ESCC specimens (p < 0.05) and knockdown reduced proliferation and migration while enhancing apoptosis (p < 0.01). GNL3L interacted with MDM2; knocking down GNL3L decreased MDM2 and increased p53 and p21 (p < 0.01). MDM2 overexpression enhanced malignant characteristics, reversible by GNL3L silencing (p < 0.01). Moreover, MDM2 knockdown inhibited malignant characteristics, reversible by GNL3L overexpression (p < 0.01). In vivo, the sh-GNL3L group exhibited the smallest tumor volumes after 5 weeks (p < 0.01).
    CONCLUSIONS: GNL3L correlates with ESCC malignancy, influencing the MDM2-p53-p21 axis. GNL3L-MDM2 interaction is critical in ESCC progression.
    Keywords:  GNL3L; MDM2; apoptosis; esophageal squamous cell carcinoma; invasion; proliferation
    DOI:  https://doi.org/10.1002/cam4.71146
  2. Colloids Surf B Biointerfaces. 2025 Aug 19. pii: S0927-7765(25)00565-X. [Epub ahead of print]256(Pt 2): 115058
      Lung cancer is a leading cause of cancer-related mortality worldwide due to increasing incidence and poor clinical outcomes. Over 50 % of human cancers involve alterations in the tumor suppressor protein p53, mostly resulting in loss of function. Idasanutlin (IDA), a hydrophobic, anionic molecule, is a potent MDM2 inhibitor capable of restoring p53 activity in cancers retaining wild-type (WT) p53. This study aimed to develop an IDA loaded lipid Nanocomplex (IDLIN) for enhancing solubility, stability and loading efficiency followed by systematically testing its effectiveness in non-small cell lung cancer (NSCLC). We hypothesized that incorporating a hydrophobic ion-pairing agent would enhance IDA encapsulation by forming a lipophilic complex. A self-nanoemulsifying drug delivery system (SNEDDS) was formulated using a hydrophobic nanocomplex of IDA and a cationic ionizable lipid, DLin-DMA, resulting in significantly improved physical stability and loading efficiency. IDLIN exhibited a mean droplet size of 81.17 ± 0.485 nm, polydispersity index of 0.122 ± 0.009, and zeta potential of -3.18 ± 0.956 mV at physiological pH and + 11.37 ± 0.404 mV at tumor microenvironment mimicking pH. IDLIN demonstrated potent anticancer activity in NSCLC cell lines A549, H460, and PC9. IDLIN resisted drug precipitation, inhibited colony formation, and was well tolerated for intravenous administration, showing negligible hemolysis. Western blot analysis revealed upregulation of p53 and MDM2 proteins in IDLIN-treated cells. In 3D tumor spheroid models, IDLIN significantly inhibited tumor growth compared to control. This study presents IDLIN as a promising nanoformulation for delivery of IDA, demonstrating therapeutic potential in NSCLC treatment.
    Keywords:  DLin-DMA; Hydrophobic ion-pairing complex; Idasanutlin; MDM2 inhibitor; Non-small cell lung cancer; P53
    DOI:  https://doi.org/10.1016/j.colsurfb.2025.115058
  3. Asian Pac J Cancer Prev. 2025 Aug 01. pii: 91796. [Epub ahead of print]26(8): 2899-2907
       OBJECTIVE: Ursolic acid (UA) has been proven to inhibit various cancer signaling pathways; however, the involvement of UA in overcoming tamoxifen resistance remains unclear and needs further investigation. This study aims to discover the potential gene targets and explore how ursolic acid interacts with those genes to restore sensitivity to tamoxifen.
    METHODS: Analyzing gene expression data from GeneCards and Swisstargetprediction for UA related genes, and the Gene Expression Omnibus (GEO) for tamoxifen resistance genes. DEGs were analyzed for functional annotation and molecular pathways using DAVID v6.8, continued with constructing a protein-protein interaction (PPI) network to highlight crucial genes associated with tamoxifen resistance using STRING-DB and Cytoscape. Genetic alteration analysis using cBioportal for target validation and consideration. Molecular docking was done using Autodock4 and PyMoL for visualisation.
    RESULTS: The KEGG pathway and PPI network suggest that MDM2, STAT3, TGFB1, and MAPK1 were indicated as potential target genes of UA. Genetic alteration analysis further confirms that MDM2 has the highest alteration, which becomes potentially targeted by UA. Molecular docking analysis confirms that UA can target MDM2 by targeting the N-terminus site on 4HBM and 5ZXF structure. The binding energy of UA is -5.36 for 4HBM and -8.71 for 5ZXF, with all RMSD values below 2. This result shows that UA has a lower docking score than the native ligand for the 5ZXF structure. Additionally, MDM2 is mainly involved in the PI3K-Akt pathway, which plays a role in the chemotherapy resistance mechanism.
    CONCLUSION: MDM2 has become a potential target for UA to reverse the tamoxifen resistance mechanism in breast cancer.
    Keywords:  In Silico; MDM2; Tamoxifen Resistant; breast cancer; ursolic acid
    DOI:  https://doi.org/10.31557/APJCP.2025.26.8.2899
  4. Biomolecules. 2025 Jul 27. pii: 1088. [Epub ahead of print]15(8):
      The protein p53, often referred to as the "guardian of the genome," is essential for preserving cellular balance and preventing cancerous transformations. As one of the most commonly altered genes in human cancers, its impaired function is associated with tumor initiation, development, and resistance to treatment. Exploring the diverse roles of p53, which include regulating the cell cycle, repairing DNA, inducing apoptosis, reprogramming metabolism, and modulating immunity, provides valuable insights into cancer mechanisms and potential treatments. This review integrates recent findings on p53's dual nature, functioning as both a tumor suppressor and an oncogenic promoter, depending on the context. Wild-type p53 suppresses tumors by inducing cell cycle arrest or apoptosis in response to genotoxic stress, while mutated variants often lose these functions or gain novel pro-oncogenic activities. Emerging evidence highlights p53's involvement in non-canonical pathways, such as regulating tumor microenvironment interactions, metabolic flexibility, and immune evasion mechanisms. For instance, p53 modulates immune checkpoint expression and influences the efficacy of immunotherapies, including PD-1/PD-L1 blockade. Furthermore, advancements in precision diagnostics, such as liquid biopsy-based detection of p53 mutations and AI-driven bioinformatics tools, enable early cancer identification and stratification of patients likely to benefit from targeted therapies. Therapeutic strategies targeting p53 pathways are rapidly evolving. Small molecules restoring wild-type p53 activity or disrupting mutant p53 interactions, such as APR-246 and MDM2 inhibitors, show promise in clinical trials. Combination approaches integrating gene editing with synthetic lethal strategies aim to exploit p53-dependent vulnerabilities. Additionally, leveraging p53's immunomodulatory effects through vaccine development or adjuvants may enhance immunotherapy responses. In conclusion, deciphering p53's complex biology underscores its unparalleled potential as a biomarker and therapeutic target. Integrating multi-omics analyses, functional genomic screens, and real-world clinical data will accelerate the translation of p53-focused research into precision oncology breakthroughs, ultimately improving patient outcomes.
    Keywords:  cancer; molecular biology; p53; precision diagnosis; therapeutic breakthroughs
    DOI:  https://doi.org/10.3390/biom15081088
  5. Biomark Res. 2025 Aug 27. 13(1): 111
      The evolution of targeted protein degradation (TPD) has been significantly propelled by the advent of proteolysis-targeting chimeras (PROTACs), which utilize heterobifunctional molecules to facilitate the ubiquitination-mediated degradation of previously "undruggable" proteins. Mouse double minute 2 (MDM2), which is often overexpressed in various diseases and plays a crucial role in regulating key pathways like p53, emerges as an exemplary candidate for therapeutic exploitation within the TPD realm, serving both as an intrinsic E3 ligase and as a direct protein of interest (POI). By harnessing MDM2's inherent E3 ligase activity, PROTACs have been designed to efficiently degrade specific POIs, achieving substantial success in both in vitro and in vivo studies. Alternatively, PROTACs have been developed to directly target MDM2 itself, offering new approaches for therapeutic intervention. Recent research has yielded valuable strategies for optimizing MDM2-harnessing and MDM2-targeted PROTAC designs, concentrating on warhead selection of POI, linker length and composition optimization, and the choice among various E3 ligases and their corresponding recruiters. These advancements not only broaden the scope of PROTAC technologies but also expedite the development of MDM2-based therapies, inspiring approaches for disease treatment.
    Keywords:  Bridged PROTAC; Homo-PROTAC; MDM2; PROTAC; TPD
    DOI:  https://doi.org/10.1186/s40364-025-00826-7
  6. Cancer Genet. 2025 Aug 14. pii: S2210-7762(25)00099-7. [Epub ahead of print]298-299 10-15
      YEATS4 resides within the 12q13-15 chromosomal region, where it is frequently co-amplified with MDM2 and CDK4 in liposarcomas (LPS). However, its independent role in LPS progression and dedifferentiation remains poorly defined. In this study, YEATS4 expression was analyzed in 57 formalin-fixed paraffin-embedded (FFPE) LPS samples using quantitative real-time PCR and compared across histological subtypes. MDM2 amplification status was determined by fluorescence in situ hybridization (FISH). The functional relevance of YEATS4 was assessed via siRNA-mediated knockdown in two well-differentiated LPS (WDLPS) cell lines, GOT-3 and 93T449. Relative YEATS4 mRNA expression was significantly higher in MDM2-positive compared to MDM2-negative tumors (median = 0.413 vs. 0.007; p = 0.008). Using the median YEATS4 expression value (0.227) - calculated from WDLPS and DDLPS cases only - as a threshold, high YEATS4 expression was observed in 64% of high-grade dedifferentiated LPS (DDLPS), 54% of low-grade DDLPS, and 29% of WDLPS cases (p = 0.302). Functionally, YEATS4 silencing significantly reduced cell viability in 93T449 cells at Days 5 (24.1%) and Day 7 (22.1%) compared to control (p < 0.001). In GOT3 cells, a slight reduction was noted at Day 3 (7.6%) which was not sustained. In summary, YEATS4 could contribute to LPS progression in a subset of MDM2-amplified tumors, particularly in high-grade DDLPS. Its variable functional impact across models highlights the complexity of the 12q13-15 amplicon and supports further investigation into YEATS4 as a potential molecular marker and therapeutic target in LPS.
    Keywords:  MDM2; YEATS4; liposarcoma; real-time PCR
    DOI:  https://doi.org/10.1016/j.cancergen.2025.08.005