bims-meract Biomed News
on Metabolic reprogramming and anti-cancer therapy
Issue of 2025–07–27
twenty-one papers selected by
Andrea Morandi, Università degli Studi di Firenze



  1. J Mol Histol. 2025 Jul 23. 56(4): 239
      
    Keywords:  Chemotherapy resistance; Colorectal cancer; Ferroptosis; Mitochondrial metabolism; Mitochondrial respiration
    DOI:  https://doi.org/10.1007/s10735-025-10533-0
  2. Funct Integr Genomics. 2025 Jul 19. 25(1): 158
      Colorectal cancer (CRC) is the third most common cancer globally and a leading cause of cancer-related death. Oxaliplatin, a key platinum-based chemotherapy, significantly improves outcomes in CRC patients. Nevertheless, oxaliplatin resistance often emerges, leading to worse prognosis. Exploring new biomarkers and mechanisms of resistance is crucial for overcoming oxaliplatin resistance and enhancing therapeutic efficacy. Through bioinformatics analysis, high NPC2 expression was found to be associated with oxaliplatin resistance and poor prognosis in CRC patients. Moreover, NPC2 was highly expressed in CRC tissues, especially in metastatic CRC tissues. Additionally, the expression of N6-methyladenosine (m6A) demethylase ALKBH5 was elevated in oxaliplatin-resistant colorectal cancer cells. Mechanically, ALKBH5 promotes m6A demethylation of NPC2 mRNA in a YTHDF2-dependent process, thereby enhancing the stability of NPC2 mRNA and making colorectal cancer cells oxaliplatin-resistant. Our results show that by inhibiting NPC2 or ALKBH5, we can re-sensitize resistant CRC cells to oxaliplatin in vitro and in vivo. In summary, ALKBH5-mediated m6A demethylation promotes the stability of NPC2 mRNA and plays a key role in promoting oxaliplatin resistance in colorectal cancer. Targeting the ALKBH5/NPC2 axis have important therapeutic potential for patients with oxaliplatin-resistant colorectal cancer.
    Keywords:  ALKBH5; Colorectal cancer; NPC2; Oxaliplatin resistance; m6A
    DOI:  https://doi.org/10.1007/s10142-025-01651-9
  3. EMBO Mol Med. 2025 Jul 24.
      Pleural mesothelioma (PM) is one of the deadliest cancers, with limited therapeutic options due to its therapeutically intractable genome, which is characterized by the functional inactivation of tumor suppressor genes (TSGs) and high tumor heterogeneity, including diverse metabolic adaptations. However, the molecular mechanisms underlying these metabolic alterations remain poorly understood, particularly how TSG inactivation rewires tumor metabolism to drive tumorigenesis and create metabolic dependencies. Through integrated multi-omics analysis, we identify for the first time that NF2 loss of function defines a distinct PM subtype characterized by enhanced de novo pyrimidine synthesis, which NF2-deficient PM cells are critically dependent on for sustained proliferation in vitro and in vivo. Mechanistically, NF2 loss activates YAP, a downstream proto-oncogenic transcriptional coactivator in the Hippo signalling pathway, which in turn upregulates CAD and DHODH, key enzymes in the de novo pyrimidine biosynthesis pathway. Our findings provide novel insights into metabolic reprogramming in PM, revealing de novo pyrimidine synthesis as a synthetic lethal vulnerability in NF2-deficient tumors. This work highlights a potential therapeutic strategy for targeting NF2-deficient mesothelioma through metabolic intervention.
    Keywords:  De Novo Pyrimidine Synthesis; Metabolic Diversity; Neurofibromin 2 (NF2); Pleural Mesothelioma (PM); Synthetic Lethality
    DOI:  https://doi.org/10.1038/s44321-025-00278-4
  4. Cell Commun Signal. 2025 Jul 24. 23(1): 351
       BACKGROUND: Metabolic and stress response adaptations in prostate cancer (PCa) mediate tumor resistance to radiation therapy (RT). Our study investigated the roles of glutamine (Gln) transporters SLC1A5, SLC7A5, and SLC38A1 in regulating NUPR1-mediated stress response, PCa cell survival, metabolic reprogramming, and response to RT.
    METHODS: The radiosensitizing potential of GLS inhibition with CB-839 was analyzed in prostate cancer xenograft models. The level of gene expression was analyzed by RNA sequencing and RT-qPCR in the established cell lines or patient-derived tumor and adjacent non-cancerous tissues. Phosphoproteomic analysis was employed to identify the underlying signaling pathways. The publicly available PCa patient datasets, and a dataset for the patients treated with RT were analyzed by SUMO software. The key parameters of mitochondrial functions were measured by Seahorse analysis. Analysis of the general oxidative stress level and mitochondrial superoxide detection were conducted using flow cytometry. γH2A.X foci analysis was used to assess the DNA double strand break. Relative cell sensitivity to RT was evaluated by radiobiological clonogenic assays. Aldefluor assay and sphere-forming analysis were used to determine cancer stem cell (CSC) phenotype.
    RESULTS: A siRNA-mediated knockdown of Gln transporters SLC1A5, SLC7A5, and SLC38A1 resulted in significant radiosensitization of PCa cells. Consistently, the first-in-clinic glutaminase (GLS) inhibitor CB-839, combined with RT, demonstrated a synergistic effect with radiotherapy in vivo, significantly delaying tumor growth. Inhibition of Gln metabolism or knockdown of Gln transporters SLC1A5, SLC7A5, or SLC38A1 induces expression of NUPR1, a stress response transcriptional regulator, but simultaneously uncouples the NUPR1-driven metabolic stress-adaptation program. Similarly to the effect from NUPR1 knockdown, depletion of these Gln transporters led to reduced cell viability, accumulation of mitochondrial ROS, and increased PCa radiosensitivity. This effect is more pronounced in PCa cells with high dependency on OXPHOS for energy production.
    CONCLUSIONS: Our work underscores the role of Gln transporters and the NUPR1-mediated stress response in PCa cell survival, oxidative stress, mitochondrial functions, and radioresistance. Our findings provide a potential therapeutic in vivo strategy to enhance the efficacy of RT and suggest a potential synergism between the depletion of Gln transporters or NUPR1 and OXPHOS inhibition.
    Keywords:  GLS; Glutamine transporters; Mitochondria; NUPR1; Oxidative stress; Prostate cancer; Radiation
    DOI:  https://doi.org/10.1186/s12964-025-02344-3
  5. Oncogene. 2025 Jul 22.
      Several mechanisms of resistance of cancer cells to cyclin-dependent kinase inhibitors (CDKi) have been identified, including the upregulation of metabolic regulators such as glutaminase. However, whether such resistance mechanisms represent optimal targets has not been determined. Here, we have systematically analyzed metabolic reprogramming in colorectal cancer cells exposed to Palbociclib, a CDKi selectively targeting CDK4/6, or Telaglenastat, a selective glutaminase inhibitor. Through multiple approaches, we show that Palbociclib and Telaglenastat elicit complementary metabolic responses and are thus uniquely suited to counter the metabolic reprogramming induced by the reciprocal drug. As such, while Palbociclib induced reduced tumor growth in vivo, and Telaglenastat did not show a significant effect, the drug combination displayed a strong synergistic effect on tumor growth. Likewise, initial responses to Palbociclib were followed by signs of adaptation and resistance, which were prevented by combining Palbociclib with Telaglenastat. In conclusion, combination with Telaglenastat optimally forestalls acquired resistance to Palbociclib in cancer cells.
    DOI:  https://doi.org/10.1038/s41388-025-03495-w
  6. Life Sci. 2025 Jul 22. pii: S0024-3205(25)00511-9. [Epub ahead of print] 123876
      Lenvatinib is one of first-line therapeutic agents for advanced hepatocellular carcinoma (HCC), yet lenvatinib resistance of tumor resulting in a weak response on many patients. Mitochondrial energy metabolism is environmentally adaptable and has been shown to play a crucial role in tumor resistance to therapy. Therefore, identification of the key regulator of mitochondrial energy metabolism during lenvatinib resistance provides a novel target for drug-resistant HCC. We found that POLRMT upregulated in lenvatinib-resistant HCC and is associated with poor patient prognosis. POLRMT increased oxidative phosphorylation levels and mitochondrial ATP production through transcriptional upregulation of respiratory chain complexes, which counteracted lenvatinib-induced cellular ATP decrease and the AMPK-caspase 3 signaling pathway. Furthermore, using IMT1B, a specific inhibitor of POLRMT, resensitized the resistant HCC to lenvatinib in vitro and in vivo. This study highlights the critical role of POLRMT in maintaining mitochondrial ATP production, and suggests that POLRMT could serve as a novel prognostic biomarker and potential therapeutic target for lenvatinib-resistant HCC.
    Keywords:  Lenvatinib resistance; Mitochondria; Oxidative phosphorylation; POLRMT
    DOI:  https://doi.org/10.1016/j.lfs.2025.123876
  7. Trends Pharmacol Sci. 2025 Jul 22. pii: S0165-6147(25)00139-7. [Epub ahead of print]
      Despite the development of new classes of therapeutics in oncology, patients with tumors harboring mutations in the tumor suppressor gene STK11/LKB1 continue to exhibit poor clinical response and therapeutic resistance. Recent advances in the understanding of LKB1-mutant tumor biology have illuminated how metabolism and the tumor microenvironment (TME) function as effectors of the aggressive nature of this tumor type. New findings have revealed how metabolic reprogramming, a hallmark of LKB1-mutant tumor biology, can be exploited as a potential targetable liability in these tumors. Characterization of the distinctly immunosuppressive LKB1-mutant TME has motivated multiple discoveries of new approaches for rewiring the microenvironment to overcome immunotherapy resistance. Indeed, overcoming therapeutic resistance in LKB1-deficient tumors continues to be a major research focus, and some preclinical studies have advanced to clinical trials. In this review, we critically analyze these findings and discuss therapies in development that aim to leverage this new understanding for clinical benefit.
    Keywords:  LKB1/STK11; cancer; kinase; lung cancer; metabolism; therapeutic resistance; tumor microenvironment; tumor suppressor
    DOI:  https://doi.org/10.1016/j.tips.2025.06.008
  8. Cell Death Dis. 2025 Jul 20. 16(1): 538
      Sorafenib is an antiangiogenic and antiproliferative chemotherapeutic drug that plays a crucial role in the treatment of patients with advanced hepatocellular carcinoma (HCC). However, resistance to sorafenib greatly limits its therapeutic efficacy. This highlights the importance of determining the mechanisms underlying resistance to antiangiogenic therapy. In this study, we found that the extracellular matrix (ECM) stiffness was closely related to the prognosis of HCC patients and chemotherapy resistance. Using atomic force microscopy, we assessed ECM stiffness in tumor samples from 30 HCC patients treated with sorafenib, and the ECM stiffness in sorafenib-resistant patients was significantly greater than that in those who responded to sorafenib treatment. In a liver orthotopic xenograft model, reducing tumor ECM stiffness by inhibiting LOX enzyme activity significantly enhanced the efficacy of sorafenib and suppressed tumor progression. We found that glucose-6-phosphate dehydrogenase (G6PD) is regulated by ECM stiffness and is involved in resistance to sorafenib. Further in vitro and in vivo experiments confirmed that ECM stiffness can upregulate G6PD expression through the ITGB1-PI3K/AKT pathway, mediating sorafenib resistance in HCC. Clinical tissue microarray analysis revealed that the expression of collagen I, α-SMA, ITGB1, p-AKT, and G6PD was associated with sorafenib resistance in HCC patients. These results indicated that reducing ECM stiffness can increase the sensitivity of HCC to sorafenib and that the ITGB1-PI3K/AKT-G6PD cascades may serve as potential therapeutic targets for reversing sorafenib resistance.
    DOI:  https://doi.org/10.1038/s41419-025-07842-3
  9. Biochim Biophys Acta Rev Cancer. 2025 Jul 21. pii: S0304-419X(25)00138-6. [Epub ahead of print]1880(5): 189396
      Breast cancer (BC), one of the most frequent causes of cancer-related death in women, is known to be a highly heterogeneous disease in regard to molecular subtypes, which seem to possess different metabolic profiles. Aberrant metabolism is well understood as one of the hallmarks of cancer and it contributes to BC progression, therapeutic resistance, and metastasis. Here, we analyze BC metabolism and how certain cancer types, such as hormone receptor-positive, HER2-positive, and triple-negative BC, use glycolysis, lipid metabolism, amino acid compulsion, and mitochondrial biogenesis to feed and proliferate. These metabolic hallmarks, in the context of the tumor microenvironments, are illustrated to highlight the metabolic byproducts that are derived from reprogrammed pathways and are vital to immunosuppression and tumor survival under low oxygen and nutrient availability. Furthermore, we emphasize novel trends in anticancer drugs designed to strike on these metabolic dependencies to suppress tumor growth. In addition to summing up current knowledge about metabolic reprogramming in BC, this review reveals new targets for specific treatments that might enhance prognosis in certain types of BC. This review aims to bridge basic scientific insights and clinical perspectives, guiding future metabolic interventions in BC toward clinically relevant, subtype-specific therapeutic strategies.
    Keywords:  Biomarker driven therapies; Breast cancer; Metabolic pathway inhibition; Metabolic reprogramming; Therapeutic resistance; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.bbcan.2025.189396
  10. Sci Rep. 2025 Jul 19. 15(1): 26263
      Acquired and intrinsic resistance to sunitinib is a major obstacle to improving the therapeutic efficacy of treatment for clear cell renal cell carcinoma (ccRCC). This study aimed to identify novel therapeutic targets and the potential molecular mechanisms to overcome sunitinib resistance in ccRCC. Utilizing genome-wide CRISPR/Cas9 screening and resistant transcriptomics, we identified that prostaglandin reductase 2 (PTGR2) is a novel therapeutic target to overcome sunitinib resistance in ccRCC. The silencing of PTGR2 enhanced the cytotoxic effects of sunitinib in ccRCC cells, as measured by cell viability assays, and suppressed tumor growth in xenograft models. Mechanistically, PTGR2 physically interacts with lysine specific demethylase 6A (KDM6A) via endogenous/exogenous co-immunoprecipitation. PTGR2 knockdown reduced KDM6A protein expression, while KDM6A overexpression partially reversed the sensitization effect of PTGR2 silencing, suggesting KDM6A is a major downstream effector. Our findings establish the PTGR2-KDM6A axis as a potential target for overcoming sunitinib resistance in ccRCC. Pharmacological inhibition of PTGR2 or targeted modulation of KDM6A activity represents a promising combination strategy to overcome sunitinib resistance and improve patient outcomes.
    Keywords:  Drug resistance; Genome-wide CRISPR/Cas9 screening; KDM6A; PTGR2; Renal cell carcinoma; Sunitinib
    DOI:  https://doi.org/10.1038/s41598-025-12192-3
  11. Cell Death Dis. 2025 Jul 21. 16(1): 539
      In breast cancer, the inner mitochondrial membrane fusion protein Optic Atrophy 1 (OPA1) is upregulated and its inhibition reverses acquired chemoresistance. However, it remains unclear whether OPA1 inhibition also targets normal breast cells. We show that OPA1 upregulation is a hallmark of metastatic breast cancer cells, which are selectively susceptible to OPA1 inhibition compared to isogenic normal or localized tumor cells. In an isogenic model spanning normal, transformed, and metastatic breast cancer cells, levels of Mitofusin 1 (MFN1) progressively declined while dynamin related protein 1 (DRP1) became increasingly active, correlating with fragmented mitochondria during cancer progression. Meanwhile, OPA1 levels were elevated in invasive cells characterized by mitochondrial fragmentation, tight cristae, and high respiration. OPA1 deletion selectively reduced metastatic cells mitochondrial respiration, proliferation, and migration. Specific OPA1 inhibitors MYLS22 and Opitor-0 diminished migration and increased death of metastatic cells, underscoring OPA1 as a selective vulnerability of metastatic breast cancer.
    DOI:  https://doi.org/10.1038/s41419-025-07878-5
  12. J Exp Clin Cancer Res. 2025 Jul 18. 44(1): 211
       BACKGROUND: Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality globally, significantly impacting worldwide health. Hence, identifying key molecular drivers of HCC progression is crucial for enhancing treatment options and prognostic methods. This study explores the function of Small Nuclear Ribonucleoprotein Polypeptides B and B1 (SNRPB) in HCC, unveiling critical pathways that affect the progression of the disease.
    METHODS: Utilizing multi-dimensional data that integrates bulk RNA sequencing (bulk RNA-seq), single-cell RNA sequencing (scRNA-seq), and spatial transcriptomics (ST) from HCC patients, we have identified SNRPB as a pivotal gene associated with the spliceosome, playing a central role in both tumor initiation and progression. We also investigated the intricate process by which SNRPB influences cyclin B1 (CCNB1) expression through FOXM1-mediated activation, using a combination of bioinformatics, functional assays, Chromatin Immunoprecipitation (ChIP), and Co-Immunoprecipitation (Co-IP) studies. Complementary in vivo experiments and metabolic assays were conducted to explore the relationship between tumor growth and lipid metabolism further. Additionally, evaluations of cisplatin sensitivity were performed, providing an in-depth analysis of influence of SNRPB on HCC.
    RESULTS: Across multiple cohorts, SNRPB exhibited a marked upregulation within tumors, correlating significantly with poor prognosis. Knockdown of SNRPB suppressed HCC cell proliferation and migration, while promoting apoptosis. Mechanistically, SNRPB regulated CCNB1 expression via FOXM1-mediated transcription, and SNRPB overexpression enhanced lipid metabolism and cisplatin resistance. This increase in drug sensitivity was mediated through alterations in lipid metabolism and the regulatory effects on CCNB1, providing a comprehensive insight into multifaceted role of SNRPB in HCC pathology and potential therapeutic targets. Finally, CCNB1 knockdown reversed the proliferative and tumorigenic effects of SNRPB overexpression in a preclinical HCC model.
    CONCLUSIONS: SNRPB promoted HCC progression by modulating the FOXM1-CCNB1 axis and lipid metabolism, and could act as a potential therapeutic target to augment chemotherapy sensitivity in HCC.
    Keywords:  Drug resistance; Hepatocellular carcinoma; Lipid metabolism; SNRPB
    DOI:  https://doi.org/10.1186/s13046-025-03463-y
  13. Cell Commun Signal. 2025 Jul 18. 23(1): 346
       BACKGROUND: Despite many new drugs, multiple myeloma (MM) remains an incurable plasma cell malignancy, and drug resistance is a long-standing topic in this field. Characterized by efficient transcription without being limited by the double helix structure and promoter, extrachromosomal circular DNA (EccDNA) has been proven to be widely involved in cancer development and drug resistance.
    METHODS: We performed circle-seq and mRNA-seq on samples from three MM patients at the time of complete response and relapse to screen EccDNA candidate molecules. Outward PCR and Sanger sequencing were used to identify EccDNA molecules. RT‒qPCR and WB were performed to detect gene expression levels. Fluorescence in situ hybridization (FISH) was carried out to detect the deletion of chromosome 17p (del (17p)). Transmission electron microscopy (TEM) was conducted to observe autophagosomes. Luciferase reporter assays were performed to validate the binding of microRNAs to target genes. Cell viability assays and apoptosis assays were employed to assess drug resistance. Xenograft tumor mouse models were established for in vivo experiments. Immunohistochemistry (IHC) was used to detect protein expression levels.
    RESULTS: We successfully identified an EccDNA molecule (EccDNAchr17:38719676-38719812) in one relapsed MM patient with del(17p) and named it MIR4726EccDNA. We demonstrated that the overexpression of MIR4726EccDNA in MM cells can increase bortezomib resistance. We further confirmed that the precursor miRNA carried by MIR4726EccDNA can be efficiently transcribed in MM cells and that MIR4726EccDNA drives bortezomib resistance via the MIR4726-5p/NXF1/NKIRAS2 axis. We further revealed that downregulation of NFKB inhibitor interacting Ras like 2 (NKIRAS2) activated the NF-κB pathway and increased autophagy. Moreover, we established a xenograft model of human MM via subcutaneous inoculation. We administered intra-tumoral injection of AgoMIR4726-5p and intraperitoneal injection of bortezomib and found that AgoMIR4726-5p promoted tumor progression and partially drove bortezomib resistance.
    CONCLUSIONS: In summary, our findings indicate that artificially synthesized MIR4726EccDNA is functional in cells and that MIR4726EccDNA enhances tumor progression and partially mediates drug resistance by enhancing MIR4726-5p/NXF1/NKIRAS2 axis dependent autophagy.
    Keywords:  Autophagy; Bortezomib; Extrachromosomal circular DNA (EccDNA); MIR4726-5p; Multiple myeloma; NKIRAS2; NXF1
    DOI:  https://doi.org/10.1186/s12964-025-02340-7
  14. iScience. 2025 Aug 15. 28(8): 113031
      For cancer patients, metastasis is a life-threatening event limiting therapeutic options. Molecularly, the metastatic phenotype can be conferred by mitochondrial reactive oxygen species (mtROS) generated upon metabolic stress. Mitochondrial damage can also trigger mtROS production, which is particularly well illustrated for anthracyclines. Here, we tested in mouse models of murine and human breast cancer whether this type of chemotherapy can trigger metastasis. We report that subcytotoxic doses of doxorubicin mimicking the clinical situation in poorly perfused tumor areas sequential trigger mtROS production, activate TGFβ pathway effector Pyk2, and increase cancer cell migration and invasion. Fortunately, the metastatic switch was incompletely induced, and doxorubicin did not promote breast cancer metastasis in immunocompetent mice. Yet, MitoTEMPO fully prevented metastatic dissemination and did not interfere with doxorubicin cytotoxicity, making it attractive to combine anthracyclines with mitochondria-targeted antioxidants.
    Keywords:  Cancer systems biology; Molecular network; Therapeutic procedure
    DOI:  https://doi.org/10.1016/j.isci.2025.113031
  15. Nat Commun. 2025 Jul 23. 16(1): 6770
      The functional programs adopted by cancer cells and their impact on the tumor microenvironment are complex and remain unclear. Here, we identify three distinct single-cell archetypes (i.e. metabolism, stemness and inflammation) in hepatocellular carcinoma (HCC) cells, each exhibiting unique spatial distribution. Further analysis shows an immune-suppressive niche populated by metabolism archetype cancer cells and TREM2-positive tumor-associated macrophages (TREM2+ TAMs), which exacerbates immune exclusion and compromises patient outcomes. Mechanistically, we demonstrate that the upregulated squalene epoxidase (SQLE) expression in metabolism archetype cancer cells facilitates the generation of oxidized LDL (oxLDL). OxLDL induces TREM2+ TAM polarization through the TREM2-SYK-CEBPα axis, enabling these TAMs to promote cancer cell invasion, resistance to effector cytokines and CD8+ T cell dysfunction. Importantly, cancer cell-intrinsic SQLE and TREM2+ TAMs are associated with inferior immunotherapy response in human and mouse HCC. Our results highlight an oxLDL-mediated metabolic interplay between cancer cells and TREM2+ TAMs, offering a promising therapeutic avenue for HCC immunotherapies.
    DOI:  https://doi.org/10.1038/s41467-025-62132-y
  16. J Ovarian Res. 2025 Jul 24. 18(1): 162
       BACKGROUD: Ovarian cancer (OC) is the most lethal gynecological tumor, primarily due to resistance to chemotherapy. Cyclin L2 (CCNL2) is a novel member of the cyclin family and mainly localized in nucleus. It regulates transcription and alternative splicing by interacting with cyclin-dependent kinases. However, its role in OC chemoresistance remains unknown.
    RESULTS: Here, we demonstrated that the expression level of CCNL2 was higher in OC tissues as well as in various other tumor types. Furthermore, elevated expression of CCNL2 indicated a poor prognosis in ovarian cancer. Functionally, CCNL2 promoted OC cell proliferation and xenograft growth. Depletion of CCNL2 enhanced chemotherapy sensitivity in OC cells. Mechanistically, YBX1 directly bound to CCNL2 mRNA, and its depletion reduced CCNL2 mRNA stability and protein expression. MeRIP assays revealed that YBX1 regulated CCNL2 via 5-methylcytosine (m⁵C) modification. Mutation of the key residue of YBX1 required for m5C function led to decreased CCNL2 expression. Further investigation of the YBX1 regulatory network identified a direct interaction between YBX1 and MATR3, which cooperatively modulated downstream targets. Notably, MATR3 knockdown reversed the YBX1-induced upregulation of CCNL2. Virtual screening identified YB-B1 as a YBX1 inhibitor that effectively downregulated both YBX1 and CCNL2 expression. In vitro, YB-B1 suppressed ovarian cancer cell proliferation and enhanced cisplatin cytotoxicity. Furthermore, patient-derived tumor xenograft (PDX) model also confirmed its chemosensitizing effect.
    CONCLUSIONS: In summary, we demonstrated that CCNL2 promoted OC cell proliferation and chemoresistance, with its expression regulated by YBX1 via m5C methylation. The small molecule inhibitor YB-B1 was identified as a promising solution to overcome chemotherapy resistance.
    CLINICAL TRIAL NUMBER: Not applicable.
    Keywords:  5-methylcytosine; CCNL2; Chemoresistance; Ovarian cancer
    DOI:  https://doi.org/10.1186/s13048-025-01753-9
  17. Cancer Lett. 2025 Jul 17. pii: S0304-3835(25)00485-9. [Epub ahead of print]631 217917
      Oxaliplatin is widely used in chemotherapy for patients with advanced colorectal cancer (CRC). However, frequent drug resistance limits its therapeutic efficacy in patients. Here, we found that a subset of cancer associated fibroblasts (CAFs) with activated glycolysis induced CRC resistance to oxaliplatin. Lactate derived from CAFs promoted the transcription of ANTXR1 through histone lactylation and induced ANTXR1 lactylation at lysine 453 residue. The increased expression of ANTXR1 and ANTXR1 K453la in CRC cells was correlated with oxaliplatin resistance in CRC cells and the poor prognosis of CRC patients. Mechanistically, lactylation promoted ANTXR1 stability and activated the RhoC/ROCK1/SMAD5 signal pathway, subsequently contributed to CRC stemness and oxaliplatin resistance. Genetic or pharmacologic inhibition of the lactate shuttle between CAFs and cancer cells improved chemotherapy efficiency in vitro and in cell/patient-derived xenograft models. These findings contribute to a better understanding of oxaliplatin resistance and indicates that inhibition of tumor-stromal interactions might be an attractive strategy for enhancing the efficacy of oxaliplatin.
    Keywords:  ANTXR1; Cancer associated fibroblasts; Colorectal cancer; Lactylation; Oxaliplatin
    DOI:  https://doi.org/10.1016/j.canlet.2025.217917
  18. Mol Cell. 2025 Jul 17. pii: S1097-2765(25)00548-9. [Epub ahead of print]85(14): 2654-2672.e7
      Metabolic reprogramming and DNA damage repair are essential in tumorigenesis and chemoresistance, yet their link remains elusive. Here, we show that LDHA deficiency impairs NHEJ and class switch recombination. Additionally, glycolysis-derived lactate promotes XLF lactylation at K288 within its Ku-binding motif (X-KBM) to regulate NHEJ. Mechanistically, DNA damage triggers ATM-mediated GCN5 phosphorylation to increase GCN5-XLF interaction and XLF lactylation, enhancing XLF-Ku80 binding, XLF recruitment to DSBs, and NHEJ efficiency. Cryo-EM structural analysis demonstrates that lactylated X-KBM (laX-KBM) forms a more extensive interface with Ku70/80, inducing conformational changes in the Ku80 vWA domain. XLF lactylation deficiency impairs NHEJ and sensitizes cancer cells to chemotherapy. A specific XLF K288 lactylation peptide inhibitor plus 5-fluorouracil synergistically kills colorectal cancer cells in PDX models with XLF hyperlactylation. These findings highlight that the GCN5-XLF lactylation axis is a critical NHEJ regulator and that targeting XLF lactylation can improve chemotherapy efficiency.
    Keywords:  Warburg effect; XLF; cell-penetrating peptide; chemoresistance; lactylation; non-homologous end-joining repair
    DOI:  https://doi.org/10.1016/j.molcel.2025.06.019
  19. Cell Metab. 2025 Jul 14. pii: S1550-4131(25)00324-9. [Epub ahead of print]
      Metabolic adaptations involved in tumor metastasis and immune evasion merit investigation. Here, using in vivo metabolic CRISPR/Cas9 knockout screening, we identified xylulokinase (XYLB) as a tumor suppressor that impairs lung colonialization by producing xylulose 5-phosphate (Xu5P), which promotes CD8+ T cell cytotoxicity. Mechanistically, CD8+ T cells express relatively high levels of solute carrier family 35 member E2 (SLC35E2), a homolog of the plant Xu5P transporter, to facilitate Xu5P uptake and subsequently intensify the pentose phosphate pathway and glycolysis for energy/redox balance. Furthermore, we revealed that Xu5P potentiates CD8+ T cell response by promoting Xu5P-responsive progenitor-like SLC35E2+ CD8+ exhausted T cells via tet methylcytosine dioxygenase 3 (TET3)-mediated DNA demethylation of the Tcf7 promoter. Clinically, elevated XYLB or blood Xu5P correlates with enhanced CD8+ T cell efficacy and reduced metastasis. In murine models, Xu5P supplementation or adopting Xu5P-rich diets synergizes with anti-PD-1 therapy to enhance antitumor immunity. These findings offer insights into the potentiality of dietary interventions for metastatic cancer.
    Keywords:  Xu5P; dietary metabolite; immunotherapy; progenitor-like CD8(+) T cells; tumor metastasis
    DOI:  https://doi.org/10.1016/j.cmet.2025.06.011
  20. J Biol Chem. 2025 Jul 16. pii: S0021-9258(25)02336-1. [Epub ahead of print] 110486
      Reprogrammed metabolism of cancer cells offers a unique target for pharmacological intervention. The mitochondrial pyruvate carrier (MPC) plays important roles in cancer progression by transporting cytosolic pyruvate into the mitochondria for use in the TCA cycle. In the current study, a series of novel fluoro-substituted aminocarboxycoumarin derivatives have been evaluated for their mitochondrial pyruvate carrier (MPC) inhibition properties. Our studies indicate that the aminocarboxycoumarin template elicits potent MPC inhibitory characteristics, and specifically, structure activity relationship studies show that the N-methyl-N-benzyl structural template provides the optimal inhibitory capacity. Further respiratory experiments demonstrate that candidate compounds specifically inhibit pyruvate driven respiration without substantially affecting other metabolic fuels, consistent with MPC inhibition. Further, computational inhibitor docking studies illustrate that aminocarboxycoumarin binding characteristics are nearly identical to that of classical MPC inhibitor UK5099 bound to human MPC, recently determined by cryoEM. The lead candidate C5 elicits cancer cell proliferation inhibition specifically in monocarboxylate transporter 1 (MCT1) expressing murine breast cancer cells 4T1 and 67NR, consistent with its ability to accumulate intracellular lactate. In vivo tumor growth studies illustrate that C5 significantly reduces the tumor burden in two syngeneic murine tumor models with 4T1 and 67NR cells. These studies provide novel MPC inhibitors with potential for anticancer applications in MCT1 expressing breast cancer tumor models.
    Keywords:  aminocarboxycoumarin; breast cancer; mitochondrial pyruvate carrier; tumor metabolism
    DOI:  https://doi.org/10.1016/j.jbc.2025.110486
  21. Blood. 2025 Jul 23. pii: blood.2024026919. [Epub ahead of print]
      Bruton's tyrosine kinase inhibitors (BTKi) and cell therapy have successfully been used to treat mantle cell lymphoma (MCL); however, therapy resistance inevitably emerges. Cancer cells can progressively develop stable resistance by traversing through a transient drug-tolerant persister (DTP) state. The mechanisms enabling DTP cells to reversibly adapt to therapies and evolve to acquire heterogeneity remain poorly understood, and characterizing DTP cells in MCL continues to pose a challenge for clinic translation. Here using pirtobrutinib, a recently FDA-approved non-covalent BTKi, we identified pirtobrutinib-tolerant persister cells exhibiting morphological variability by presenting a unique population of enlarged cells (Giant cells) with reversible fate transitions. During treatment, Giant cells enter a non-proliferative, dedifferentiated state, addicted to an activated cytosolic tricarboxylic acid (TCA) cycle coupled with the malate-aspartate shuttle to engage in biosynthesis. Upon drug removal, the TCA cycle shifts to oxidative catabolism, promoting Giant cells to differentiate into regular-sized cells. Throughout the transition, acetyl-CoA modulates cell fate by fine-tuning stemness. Our biphasic model demonstrates that the metabolic switch governs the phenotypic plasticity of DTP cells in MCL, resulting a dynamic presence of DTP cells across various developmental states in response to systemic therapies. Targeting Giant cells prior to their differentiation offers a promising strategy to overcoming therapy resistance in MCL.
    DOI:  https://doi.org/10.1182/blood.2024026919