bims-meract Biomed News
on Metabolic reprogramming and anti-cancer therapy
Issue of 2025–06–08
27 papers selected by
Andrea Morandi, Università degli Studi di Firenze



  1. bioRxiv. 2025 May 24. pii: 2025.05.20.655097. [Epub ahead of print]
       Background: The 1p/19q co-deletion is a hallmark of oligodendrogliomas. The goal of this study was to exploit metabolic vulnerabilities induced by the 1p/19q co-deletion for oligodendroglioma therapy and non-invasive imaging.
    Methods: We used stable isotope tracing, mass spectrometry, and genetic and pharmacological approaches to interrogate [U- 13 C]-glucose metabolism in patient-derived oligodendroglioma models (SF10417, BT88, BT54, TS603, NCH612). We examined whether tracing [6,6'- 2 H]-glucose metabolism using deuterium metabolic imaging (DMI) provided an early readout of treatment response.
    Results: The expression of the glycolytic enzyme enolase 1 (ENO1; chromosome 1p36.23) was reduced in patient-derived oligodendroglioma cells and patient biopsies due to the 1p/19q co-deletion and histone hypermethylation. Conversely, ENO2 was upregulated, an effect that was driven by mitogen-activated protein kinase (MAPK) signaling and ERK1-mediated phosphorylation and inactivation of the CIC transcriptional repressor in oligodendrogliomas. Genetic ablation of ENO2 or pharmacological inhibition using POMHEX inhibited proliferation with nanomolar potency but was not cytotoxic to oligodendroglioma cells or tumor xenografts. Mechanistically, ENO2 loss abrogated [U- 13 C]-glucose metabolism to lactate but shunted glucose towards biosynthesis of serine and purine nucleotides, an effect that was driven by phosphoglycerate dehydrogenase (PHGDH). Importantly, the PHGDH inhibitor D8 was synthetically lethal in combination with POMHEX, and the combination induced tumor regression in vivo. Furthermore, DMI of lactate production from [6,6'- 2 H]-glucose provided an early readout of response to combination therapy that preceded MRI-detectable alterations and reflected extended survival.
    Conclusions: We have identified ENO2 and PHGDH as 1p/19q co-deletion-induced metabolic vulnerabilities in oligodendrogliomas and demonstrated that DMI reports on early response to therapy.
    KEY POINTS: The 1p/19q co-deletion upregulates ENO2 in oligodendrogliomas.ENO2 inhibition inhibits glycolysis but upregulates serine and nucleotide biosynthesis via PHGDH.Combined inhibition of ENO2 and PHGDH is lethal, an effect that can be visualized by DMI.
    IMPORTANCE OF THE STUDY: Oligodendrogliomas are devastating primary brain tumors with long-lasting and life-altering effects on physical and cognitive function. The presence of a 1p/19q co-deletion defines oligodendrogliomas. Here, using clinically relevant patient-derived models and patient tissue, we show that the 1p/19q co-deletion leads to loss of the glycolytic enzyme ENO1 and upregulation of ENO2 in oligodendrogliomas. This provides a unique therapeutic opportunity since most cells rely on ENO1 for glycolysis. Targeting ENO2 using the brain-penetrant inhibitor POMHEX abrogates glycolysis but redirects glucose toward serine and nucleotide biosynthesis, an effect that is driven by PHGDH, the rate-limiting enzyme for serine biosynthesis. Importantly, combined treatment with POMHEX and the PHGDH inhibitor D8 is synthetically lethal in vitro and in vivo. Furthermore, visualizing glucose metabolism using DMI provides an early readout of response to therapy that predicts extended survival in vivo . In summary, we have developed a unique integrated metabolic therapy and imaging approach for oligodendrogliomas.
    DOI:  https://doi.org/10.1101/2025.05.20.655097
  2. Elife. 2025 Jun 04. pii: RP103953. [Epub ahead of print]14
      Trastuzumab resistance remains a challenge for HER2-positive breast cancer treatment. Targeting metabolic reprogramming would provide novel insights for therapeutic strategies. Here, we integrated metabolomics, transcriptomics, and epigenomics data of trastuzumab-sensitive and primary-resistant HER2-positive breast cancer to identify metabolic alterations. Aberrant cysteine metabolism was discovered in trastuzumab primary-resistant breast cancer at both circulating and intracellular levels. The inhibition of SLC7A11 and cysteine starvation could synergize with trastuzumab to induce ferroptosis. Mechanistically, increased H3K4me3 and decreased DNA methylation enhanced SLC7A11 transcription and cystine uptake in trastuzumab-resistant breast cancer. The regulation of epigenetic modifications modulated cysteine metabolism and ferroptosis sensitivity. These results revealed an innovative approach for overcoming trastuzumab resistance by targeting specific amino acid metabolism.
    Keywords:  HER2-positive breast cancer; cancer biology; cysteine metabolism; epigenetic modifications; human; mouse; trastuzumab primary resistance
    DOI:  https://doi.org/10.7554/eLife.103953
  3. Adv Sci (Weinh). 2025 Jun 06. e06950
      Mutations in epidermal growth factor receptor (EGFR) are the key drivers of lung cancer initiation and recurrence. The cancer cells undergo transformation to a reversible drug-tolerant persister (DTP) state prior to the development of resistance against EGFR-tyrosine kinase inhibitors (TKIs). Two DTP lung cancer cells with different proliferative capacities are established and identified dipeptidyl peptidase 4 (DPP4) as a potential therapeutic target. The DTP cells primarily relied on oxidative phosphorylation, which is accompanied by the up-regulation of fatty acid metabolism. Mechanistically, DPP4 facilitates the uptake of fatty acids via carnitine palmitoyl transferase 1a (CPT1A, and enhances fatty acid oxidation. In addition, the DPP4-mitogen-activated protein kinase kinase (MEK)-Nuclear factor erythroid-2-related factor 2 (Nrf2) signaling pathway maintains mitochondrial function by activating the antioxidant pathway. The combination of osimertinib and sitagliptin, a DPP4 inhibitor, not only suppressed tumor progression but also reduced the number of residual tumor cells and minimal residual disease. Notably, this combination therapy significantly lowered recurrence rates and extended the survival of tumor-bearing mice compared to the monotherapies. The study provides new insights into the metabolic adaptations of DTP lung cancer cells in response to EGFR-TKIs, offering novel therapeutic strategies for targeting these persister cells.
    Keywords:  DPP4; DTP; EGFR‐Tki; OXPHOS; lung cancer
    DOI:  https://doi.org/10.1002/advs.202506950
  4. bioRxiv. 2025 May 24. pii: 2025.05.21.655274. [Epub ahead of print]
      Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to meet their increased biosynthetic and energetic demands. While cells possess the capacity for de novo serine biosynthesis, most transformed cancer cells heavily depend on exogenous serine uptake to sustain their growth, yet the regulatory mechanisms driving this metabolic dependency remain poorly understood. Here, we uncover a novel mechanism by which Polo-like kinase 1 (PLK1), often overexpressed in prostate cancer, orchestrates a metabolic shift in serine and lipid metabolism through the phosphorylation of phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme of the serine synthesis pathway (SSP). We demonstrate that PLK1 phosphorylates PHGDH at three specific sites (S512, S513, S517), leading to a marked reduction in its protein level and enzymatic activity. This downregulation of SSP forces cancer cells to increase their reliance on exogenous serine uptake via the ASCT2 transporter, which, in turn, fuels the biosynthesis of lipids, including sphingolipids essential for tumor growth and survival. Targeting the SSP, serine uptake, or downstream lipid biosynthetic pathways may offer promising therapeutic avenues in PLK1-high advanced cancers.
    DOI:  https://doi.org/10.1101/2025.05.21.655274
  5. Biomaterials. 2025 May 26. pii: S0142-9612(25)00356-4. [Epub ahead of print]323 123437
      Metabolic reprogramming characterized by mitochondrial dysfunction and increased glycolysis is associated with aggressive tumor biology and poor therapeutic response. The interplays among NADPH oxidase (NOX)-mediated reactive oxygen species, regulation of glycolysis and oxidative phosphorylation (OXPHOS) in cancer cells suggest an opportunity to develop a new cancer therapy. We found that treatment with a hyaluronic acid nanoparticle encapsulated with GKT831 (HANP/GKT831), a NOX1/4 inhibitor, markedly inhibited the proliferation and invasion of cancer cells. Treated tumor cells had reduced levels of mitochondrial ROS, glycolysis, and OXPHOS. The combination of HANP/GKT831 with radiation reduced colony formation and invasion of tumor cells. The combination therapy markedly inhibited the levels of molecules in glycolysis, OXPHOS, and DNA repairing pathways in tumor cells. Systemic administrations of HANP/GKT831 combined with radiotherapy significantly inhibited tumor growth by 84.7 % in a mouse colorectal tumor model. Tumors treated with HANP/GKT831 and radiation had increased DNA damage and apoptotic cell death. Furthermore, the combined therapy increased intratumoral infiltration of activated cytotoxic T cells and M1 macrophages but reduced the levels of immunosuppressive fibroblasts and M2 macrophages. Our results support HANP/GKT831 as a cancer nanotherapeutic agent that induces redox and bioenergy stresses in cancer cells for enhanced therapeutic response to radiotherapy.
    Keywords:  Colorectal cancer; Glycolysis; Hyaluronic acid nanoparticle; Mitochondrial oxidative phosphorylation; NADPH oxidase inhibitor; Radiotherapy; Tumor metabolism
    DOI:  https://doi.org/10.1016/j.biomaterials.2025.123437
  6. iScience. 2025 May 16. 28(5): 112404
      Gallium based therapeutic strategies are thought to be an effective means to promote tumor cell killing that are currently being investigated in glioblastoma. Gallium is a group IIIa metal that is redox inactive, however, its atomic similarity to iron allows it to serve as an iron mimic that may disrupt Fe metabolism. The current understanding of its mechanism of action is related to its ability to inhibit ribonucleotide reductase and the electron transport chain, however, its effects on other iron-dependent metabolic processes (e.g., lipid metabolism) are unknown. Ferroptosis is a unique iron-dependent form of cell death, thus, the goal of this study is to evaluate the effects of Ga(NO3)3 on the induction of ferroptosis. Despite its redox inactivity, Ga(NO3)3 promotes the formation of oxidized lipid droplets. Moreover, Ga(NO3)3 enhances the toxicity of the ferroptosis inducer, erastin, and decreases cell stiffness, indicating an exacerbation of ferroptosis. Ga(NO3)3 also enhances the toxicity of the stearoyl CoA desaturase (SCD) inhibition, and its toxicity can be reversed by oleic acid supplementation, suggesting that Ga(NO3)3 can potentially inhibit SCD. Lipidomic analysis revealed a significant increase in oxidizable triglycerides following Ga(NO3)3 treatment. Thus, it appears that Ga(NO3)3 exacerbates ferroptosis in glioblastoma cells by disrupting the di-ferric lipid metabolic regulator SCD and warrants further investigation as an alternate mechanism of action for Ga-based therapy.
    Keywords:  Biochemistry; Immunology; Molecular biology; Neuroscience
    DOI:  https://doi.org/10.1016/j.isci.2025.112404
  7. J Cell Mol Med. 2025 Jun;29(11): e70572
      Triple-negative breast cancer (TNBC) is associated with a poor prognosis due to insufficient molecular subtyping precision and limited actionable targets. Although metabolic reprogramming underlies TNBC chemotherapy resistance, establishing metabolic subtyping systems and investigating drug sensitivity across distinct metabolic subgroups could provide novel therapeutic avenues for breast cancer management. GSVA (Gene Set Variation Analysis) analysis of metabolic pathways reveals significant differences in TNBC (Triple-Negative Breast Cancer) patients. TNBC patients are classified into four metabolic subtypes through consensus clustering, based on their GSVA values of metabolic pathways. These subtypes are: MS_1, characterised by increased lipogenic activity; MS_2, characterised by increased carbohydrate and nucleotide metabolism; MS_3, a metabolism-active subtype with activation of all types of metabolism; and MS_4, characterised by suppressed metabolic activity across all types of metabolism. We next propose a novel method called MODIN (Multiomics-Driven Drug-Cell Interaction Network), which embeds multi-omics gene information (mRNA expression, copy number variation and DNA methylation) and drug SMILES data into a latent space, and then employs a multi-head attention-based interaction module to accurately predict the LN_IC50 values of 621 drugs in TNBC. Based on MODIN, noteworthy disparities in drug sensitivity emerge between the patient cohorts categorised as MS_2 and MS_3. MS_3 patients show a significantly higher sensitivity to chemotherapy regimens, especially for doxorubicin and docetaxel, while the MS_2 cohort displays marked resistance to these drugs. Our study reveals the metabolic heterogeneity of TNBC, and TNBC patients with increased carbohydrate and nucleotide metabolism exhibit the poorest prognoses and greater resistance to doxorubicin and docetaxel.
    Keywords:  drug efficacy prediction; metabolic heterogeneity; multi‐omics; triple‐negative breast cancer
    DOI:  https://doi.org/10.1111/jcmm.70572
  8. Sci Rep. 2025 Jun 03. 15(1): 19445
      Clinical advancement of novel therapeutics is often hindered by variable patient responses. Therefore, clinically translatable biomarkers of response are urgently required to facilitate precision medicine trials. Endogenous volatile organic compounds (VOCs) can be non-invasively detected in exhaled breath and biofluids and have shown great potential for early cancer detection. Since emerging evidence suggests that cancer-associated VOCs may reflect alterations in the tumour lipidome, we speculated that the response to metabolically active therapies could be monitored through VOC measurement. In this proof-of-concept study, we investigated the lipidomic and volatilomic profiles of mTOR catalytic inhibitors (mTORci)-resistant and -sensitive colorectal cancer cells. Distinct lipid-derived VOC signatures, including upregulated alkenes, aldehydes, and fatty acids were observed in mTORci-resistant cells. These enriched VOCs correlated with phospholipid structure and desaturation positions, suggesting that they may be surrogates of dysregulated lipid metabolism. This novel association between VOCs and drug response establishes a precedent for further investigation into VOC biomarkers of treatment response. VOCs associated with therapy response in vitro need to be targeted in clinical trials to identify biomarkers that could be translated to monitor therapeutic response in a clinical setting.
    Keywords:  Colorectal cancer; Gas chromatography-mass spectrometry; Gastrointestinal cancers; Lipid peroxidation; Multi-omics; Volatile organic compounds
    DOI:  https://doi.org/10.1038/s41598-025-04886-5
  9. Sci Transl Med. 2025 Jun 04. 17(801): eadk7786
      Endocrine resistance is frequently encountered in estrogen receptor-positive (ER+) breast cancer, often because of somatic mutations such as neurofibromin 1 (NF1) loss. The mechanisms by which ER-directed proliferation is lost in such cases are unknown, limiting the potential use of additional endocrine treatments. Here, we performed CRISPR-Cas9 knockout (KO) screens and found that nuclear receptor subfamily 2 group F member 2 (NR2F2), an orphan nuclear receptor, was essential for NF1 loss-induced endocrine resistance. Induction of NR2F2 was observed in ER+ cell line models and patient samples and occurred via activation of the mitogen-activated protein kinase (MAPK) pathway upon NF1 loss or other MAPK pathway genetic alterations. Mechanistically, increased NR2F2 orchestrated a repressed ER transcriptional program by repartitioning the ER cistrome, altering the balance of its associated transcriptional coregulators, and modifying global chromatin accessibility. Accordingly, genetic depletion or pharmacologic inhibition of NR2F2 restored sensitivity to hormone therapies in multiple models, including ER+ cell lines, patient-derived xenografts, and patient-derived organoid-based xenografts harboring diverse endocrine-resistance mechanisms such as NF1, AT-rich interactive domain-containing protein 1A (ARID1A), phoshatase and tensin homolog (PTEN) loss, or Kirsten rat sarcoma virus (KRAS) overexpression. Together, these findings underscore NR2F2 as a critical modulator of the hormone response pathway and suggest its inhibition as a promising strategy to overcome endocrine resistance in breast cancer.
    DOI:  https://doi.org/10.1126/scitranslmed.adk7786
  10. Adv Sci (Weinh). 2025 Jun 05. e10386
      Tumor cells reprogram their energy metabolism patterns to meet the needs of rapid growth and metastasis. The underlying mechanisms of long noncoding RNAs (lncRNAs) in glucose metabolism remodeling in breast cancer (BC) are still not well understood. Herein, the expression of a tumorigenic lncRNA, LINC01094 are demonstrated that, is significantly increased in BC tissues and is associated with poorer patient survival. METTL14-mediated m6A modification stabilized LINC01094 by recruiting the reader protein IGF2BP2, which contributed to the upregulation of LINC01094 expression in BC. Gain- and loss-of-function assays validated that LINC01094 triggered a switch in glucose metabolism from mitochondrial respiration to glycolysis, promoting BC progression both in vitro and in vivo. LINC01094 promoted the dimeric assembly and nuclear translocation of PKM2 by acting as a "molecular scaffold" for the PKM2/JMJD5 complex. This, in turn, facilitated energy metabolic reprogramming and cell proliferation induced by HIF1-α/β-catenin. Furthermore, the therapeutic potential of LINC01094 is evaluated through the administration of the PKM2 activator TEPP-46 in mouse xenografts. These findings highlight the critical roles of LINC01094 in cellular glucose metabolism and tumorigenesis in BC, suggesting that it is a potential therapeutic target.
    Keywords:  LINC01094; PKM2; breast cancer; glucose metabolic reprogramming; m6A modification
    DOI:  https://doi.org/10.1002/advs.202410386
  11. Free Radic Biol Med. 2025 Jun 02. pii: S0891-5849(25)00745-2. [Epub ahead of print]237 195-209
      Pancreatic cancer is a highly aggressive malignancy with a significant unmet medical need, as current treatments often yield poor responses. Ferroptosis, a recently recognized form of regulated cell death, has garnered increasing attention for its potential in cancer therapy. However, the molecular links connecting autophagy to ferroptosis remain largely unclear. In this study, we identified that the redox related protein glucose-6-phosphate dehydrogenase (G6PD) is overexpressed in pancreatic cancer and correlates with poor prognosis, promoting cancer cell proliferation and migration. Using PANC-1 and MiaPaCa-2 pancreatic cancer cell lines, we demonstrated that the treatment with ferroptosis-inducing compound RSL3 induced glycolytic dysfunction and significantly downregulated G6PD expression. Moreover, G6PD knockdown in these cell lines impaired the cellular antioxidant defence capability by decreasing the NADPH and GSH contents, leading to increased lipid peroxidation and malondialdehyde (MDA) accumulation. Particularly, G6PD depletion exacerbated RSL3 induced oxidative stress and synergistically promoted autophagy-dependent ferroptosis. Mechanistically, we found that G6PD knockdown disrupted redox homeostasis, triggering the activation of AMPK-mTOR pathway to induce autophagy. Furthermore, pharmacological inhibition of AMPK (with Compound C) rescued ferroptosis induced by G6PD knockdown and RSL3, whereas mTOR inhibition (with Rapamycin) further augmented cell death. Altogether, these findings suggest that G6PD contributes to ferroptosis resistance in pancreatic cancer cells by modulating oxidative balance and autophagy via the AMPK-mTOR pathway, highlighting its potential as a therapeutic target.
    Keywords:  AMPK-mTOR pathway; Autophagy; Ferroptosis; G6PD; Redox balance
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2025.06.002
  12. NPJ Precis Oncol. 2025 Jun 04. 9(1): 162
      Breast cancer, the leading cause of death in females, is profoundly affected by cooperative changes of molecular and pathological characters in fatty acid metabolism. To investigate the core molecule of fatty acid metabolism in breast cancer progression, we performed in-silico analyses and found 2,4-Dienoyl-CoA reductase (DECR1) was overexpressed in breast cancer tissues with impaired prognoses of patients. In vitro experiments showed that the proliferation and migration of breast cancer cells were significantly inhibited after DECR1 knockdown. Multi-omics analyses and metabolite detection indicated that DECR1 knockdown induced cell ferroptosis, and inhibited the production of arachidonic acid (AA) with phosphatidylcholine (PC) accumulation, AA was previously identified as ferroptosis inducer via up-regulating ACSL4, accordingly we found that knocking-down DECR1 increased the expression of ACSL4 with ferroptosis inhibitor GPX4 and SCL7A11downregulation. Additionally, we found the expression of phospholipase A2, group XIIA (PLA2G12A), a key enzyme in metabolic regulation of PC to AA, was elevated in DECR1 knocking-down cells. Further, by computer aided virtual screening, we identified various compounds with DECR1 targeting properties, and verified-Erigoster B, exerts antitumor effects by promoting ferroptosis. In conclusion, DECR1 could be a candidate target for breast cancer, and the DECR1 inhibitor Erigoster B can be a potential drug to exert therapeutic effect on breast cancer.
    DOI:  https://doi.org/10.1038/s41698-025-00945-2
  13. Gut. 2025 Jun 04. pii: gutjnl-2024-334361. [Epub ahead of print]
       BACKGROUND: Pancreatic cancer exhibits limited clinical responses to immunotherapy, highlighting the need for new strategies to counteract its immunosuppressive microenvironment. Although metabolic reprogramming and epigenetic changes contribute to malignancy, the impact of lactate-driven histone lactylation on the tumour microenvironment (TME) has not been fully explored.
    OBJECTIVE: This study aims to investigate the role of histone lactylation in pancreatic cancer, focusing on its effects on cholesterol metabolism and antitumour immunity.
    DESIGNS: Global lactylome profiling was conducted to identify novel epigenetic mechanisms driven by lactate-induced histone lactylation. Mechanisms were investigated via RNA sequencing, CUT&Tag, immunoprecipitation-mass spectrometry and GST-pull down. Mass cytometry by time-of-flight, in vitro co-culture system, orthotopic pancreatic cancer models and flow cytometry were used to explore Acetyl-CoA acetyltransferase (ACAT2) functions. A proteolysis-targeting chimaera (PROTAC) was developed to degrade ACAT2.
    RESULTS: Global lactylome profiling revealed that lactate-driven histone lactylation, particularly H3K18la, promotes the transcriptional activation of ACAT2. ACAT2 acetylates mitochondrial carrier homolog 2 (MTCH2), stabilising it and disrupting oxidative phosphorylation, which increases lactate production and fuels a positive feedback loop in pancreatic cancer. This loop facilitates the delivery of cholesterol via small extracellular vesicles (sEVs), polarising tumour-associated macrophages toward an immunosuppressive M2 phenotype. Additionally, the PROTAC targeting ACAT2 enhanced the efficacy of immune checkpoint blockade therapy in vivo.
    CONCLUSIONS: Our findings highlight the critical role of the H3K18la/ACAT2/sEV-cholesterol axis in TME reprogramming. Targeting this pathway may improve anti-PD-1 therapy response in pancreatic cancer, providing a novel therapeutic strategy by linking histone lactylation, cholesterol metabolic reprogramming and immune modulation.
    Keywords:  GENE TARGETING; IMMUNE RESPONSE; LIPID METABOLISM; MACROPHAGES; PANCREATIC CANCER
    DOI:  https://doi.org/10.1136/gutjnl-2024-334361
  14. ACS Nano. 2025 Jun 05.
      Aberrantly elevated lactate flux in tumors is increasingly recognized as a key driver of metabolic symbiosis, immunosuppression, and, ultimately, immunogenic chemotherapy resistance. Here, we propose a precise lactate homeostasis modulation strategy that selectively intercepts intracellular lactate molecules in highly glycolytic tumor cells. Targeting monocarboxylate transporter 4 (MCT4), a key lactate efflux transporter overexpressed in tumor cells, we developed a glucose-disguised delivery system for precise transport of regulatory molecules into glycolysis-dependent tumor cells. By modulating lactate-mediated crosstalk between heterogeneous tumor subpopulations (glycolysis-dependent and lactate-consuming cells) and immune cells, this strategy effectively disrupts lactate-driven metabolic cooperation within the tumor niche, which may contribute to overcoming lactate-associated resistance to chemo-immunotherapy.
    Keywords:  chemo-immunotherapy sensitization; immune activation; lactate; metabolic regulation; metabolic symbiosis; tumor microenvironment
    DOI:  https://doi.org/10.1021/acsnano.5c03545
  15. Free Radic Biol Med. 2025 May 29. pii: S0891-5849(25)00720-8. [Epub ahead of print]237 110-130
      Acetyl-CoA carboxylase (ACC) is a rate-limiting enzyme in de novo lipogenesis. Here, we show a unique function of ACC in disrupting cellular iron homeostasis to drive ferroptosis, an iron-dependent, lipid peroxidation-induced form of cell death. We observed neuronal lipid accumulation and elevated labile iron pool associated with ACC dephosphorylation in mouse models of obstructive sleep apnea (OSA), a highly prevalent neurodegenerative disorder. ACC gene (Acaca) knockout (KO) or inhibition of its enzymatic activity rescued cellular iron metabolism through restoring lysosomal integrity and function, suppressing neuronal ferroptosis. ACC inactivation-driven lysosomal iron homeostasis requires the NFE2L2/NRF2-TFEB axis. Empagliflozin mitigates cellular iron overload via the ACC-NRF2-TFEB-lysosome pathway to alleviate neuronal ferroptosis, cognitive impairment, and mood dysfunction in OSA mice. Furthermore, inhibiting neuronal ACC reduces microglial activation, characterized by elevated complement proteins and pro-inflammatory cytokines, while microglia-specific C1qa KO prevents neuronal injury in OSA mice. Our findings identify a unique coupling between iron homeostasis and lipogenic signaling, suggesting ACC as a potential therapeutic target for neuronal ferroptosis and the resultant microgliosis in neurodegenerative diseases.
    Keywords:  Acetyl-CoA carboxylase; Cognitive impairment; Ferroptosis; Iron homeostasis; Lysosome
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2025.05.421
  16. bioRxiv. 2025 May 14. pii: 2025.05.14.653978. [Epub ahead of print]
      Iron is vital to living cells, playing a key role in cellular respiration, DNA synthesis, and various metabolic functions. Importantly, cancer cells have a higher dependency on iron compared to normal cells to support their rapid growth and survival. Due to this fact, tumors are more vulnerable to ferroptosis, an iron-dependent form of regulated cell death. Radiation therapy (RT), a standard treatment for many cancer patients, is known to induce ferroptosis. Ultra-high dose rate FLASH RT offers an improved therapeutic window by minimizing damage to normal tissues while preserving tumor control. However, the precise biological mechanisms behind the protective effects of FLASH RT on normal tissues remain unclear. In this study, we propose that variations in lipid peroxidation and ferroptosis, driven by intrinsic differences in iron levels between normal and cancerous tissues, contribute to this effect. Our findings show that FLASH RT increases lipid peroxidation and induces ferroptosis in tumor cells but does not significantly elevate lipid peroxidation in normal tissues compared to conventional RT. To determine whether raising iron levels in normal tissues could abrogate the protective effects of FLASH, mice were fed a high-iron diet before RT. A high-iron diet before and after RT reversed the protective effect of FLASH, resulting in increased intestinal damage and lipid peroxidation. This suggests that baseline iron levels and iron-driven lipid peroxidation are critical factors in mediating the protective outcomes of FLASH RT. Overall, our study sheds light on the role of iron in modulating RT responses and provides new mechanistic insights into how FLASH RT influences normal and cancerous tissues.
    DOI:  https://doi.org/10.1101/2025.05.14.653978
  17. Cancer Sci. 2025 Jun 02.
      The medium-chain fatty acyl-CoA synthetase-5 (ACSM5) plays a crucial role in the development of some cancers. However, its impact on liver cancer is still not clear. In this study, we found that the proliferation ability of LM3 and HepG2 cells was significantly inhibited after ACSM5 was overexpressed, and this change was blocked by the ferroptosis inhibitor deferoxamine. ACSM5 increased the levels of malondialdehyde (MDA) and lipid reactive oxygen species (ROS), reduced the level of glutathione (GSH), and thus triggered ferroptosis. Furthermore, ACSM5 promoted the upregulation of cytochrome P450 oxidoreductase (POR). Knocking down POR blocked the promoting effect of ACSM5 on ferroptosis in HCC. Moreover, ACSM5 promoted the generation of arachidonic acid and thus increased the sensitivity to ferroptosis. In summary, our findings indicate that ACSM5 induces ferroptosis in hepatocellular carcinoma (HCC) by upregulating POR. The metabolic transformation of linoleic acid to arachidonic acid was also promoted by ACSM5; therefore, sensitivity to ferroptosis was increased.
    Keywords:  ACSM5; POR; ferroptosis; hepatocellular carcinoma; lipid metabolism; lipid peroxidation
    DOI:  https://doi.org/10.1111/cas.70115
  18. Nat Commun. 2025 Jun 05. 16(1): 5226
      Metastasis is the main cause of cancer-related deaths, yet the underlying mechanisms remain elusive. Here, using clear cell renal cell carcinoma (ccRCC), a tumor type with frequent lung metastases, we conduct an in vivo genome-wide CRISPR-Cas9 screen and identify HLF as a potent suppressor of lung metastasis. HLF depletion enhances ccRCC cell migration and lung metastasis, whereas HLF overexpression abrogates these effects. In ccRCC patients, HLF expression is reduced at metastatic sites and associates with epigenetic silencing mediated by the SWI/SNF ATPase subunit BRG1. HLF levels negatively correlate with migration potential in collagen. Mechanistically, HLF regulates LPXN expression, modulating the integration of collagen's mechanical cues with the actin cytoskeleton through Paxillin, thereby suppressing cancer cell migration and lung metastasis. Overexpression of HLF or pharmacological inhibition of BRG1 reduces cell invasion across multiple cancer types. Our findings suggest that targeting the BRG1-HLF axis offers a promising therapeutic strategy for combating metastatic cancers.
    DOI:  https://doi.org/10.1038/s41467-025-60329-9
  19. J Exp Clin Cancer Res. 2025 Jun 03. 44(1): 170
       BACKGROUND: Drug resistance and relapse are still major challenges in acute myeloid leukemia (AML) because of the inability to effectively eradicate leukemia stem cells (LSCs). Senescence induction combined with immune killing may offer promising strategies for LSC eradication. However, whether and how drug-resistant LSCs retain stemness via senescence and immune regulation remains unknown.
    METHODS: The immunoproteasome subunit PSMB10 expression levels were analyzed by single-cell RNA-seq data, along with the bioinformatics analysis of publicly available AML datasets, and quantified using RT-qPCR and flow cytometry (FCM) analysis on clinical samples from AML patients. The cellular senescence was evaluated by the assays of cell proliferation, cell cycle, senescence-associated β-galactosidase activity, and senescence-associated secretory phenotype factors. In vitro T-cell killing assay was played to determine immune escape reprogramming of AML cells. FCM was conducted to estimate intracellular drug concentration and cellular apoptosis rates. Human AML xenografts and PSMB10 knockout syngeneic mouse bone marrow transplantation models were utilized to investigate the function of PSMB10. Various techniques were employed for mechanism studies, including Lentivirus transduction or siRNA transfection, western blotting, co-immunoprecipitation assays, luciferase reporter assays, polysome profiling assays, quantitative proteomics, etc. RESULTS: PSMB10 mRNA was significantly upregulated in the surviving nonsenescent LSCs, exhibiting a 13-fold increase compared to senescent LSCs following chemotherapy. The specific high expression of PSMB10 in post-chemotherapy nonsenescent LSCs predicts a poor AML prognosis. The genetic inactivation of PSMB10 resulted in increased senescence and cytotoxic T lymphocyte (CTL) killing, as well as increased intracellular drug concentrations and drug-induced cellular senescence in different types of human AML cells, which also impeded human and murine leukemia initiation and stemness maintenance in vivo with a 19-fold decrease in the frequency of human LSCs and a 7.6-fold decrease of drug-resistant mouse LSCs, while normal hematopoietic cells remained unaffected. Mechanistically, the downregulation of PSMB10 boosted SLC22A16-mediated drug endocytosis and further induced chemotherapy drug-mediated senescence through the RPL6/RPS6-MDM2-P21 pathway in AML cells. Additionally, downregulating PSMB10 also impeded MHC-I protein degradation-induced escape of CTL killing.
    CONCLUSIONS: PSMB10 is a key candidate molecular target for eradicating drug-resistant LSCs via senescence and immune regulation.
    Keywords:  Acute myeloid leukemia; Chemotherapeutic drug resistance; Immune escape; Immunoproteasome; Leukemia stem cells; PSMB10; Senescence regulation
    DOI:  https://doi.org/10.1186/s13046-025-03420-9
  20. J Exp Clin Cancer Res. 2025 Jun 05. 44(1): 172
       BACKGROUND: Colorectal cancer (CRC) is a common and aggressive malignancy that frequently metastasizes to the liver, presenting significant therapeutic challenges. Despite its clinical importance, the mechanisms underlying CRC liver metastasis and resistance to immune therapy remain poorly understood. In this study, we aimed to investigate the molecular mechanisms driving CRC metastasis using a novel approach, which includes the establishment of highly metastatic CRC cell lines.
    METHODS: To explore the role of specific genes in CRC liver metastasis, we developed two highly metastatic CRC cell lines (LoVo-Hm and HCT116-Hm) by applying sustained selective pressure to primary CRC cells. RNA sequencing was performed to identify differentially expressed genes in these metastatic cells. Additionally, we conducted assays for cell migration, invasion, angiogenesis, and ELISA to evaluate VEGFA production, all to confirm the functional role of SLC9A2. Our findings were further validated in human CRC tissue samples and publicly available datasets to assess the clinical relevance of the identified targets.
    RESULTS: Our analysis revealed a significant downregulation of SLC9A2 in the highly metastatic CRC cell lines. Mechanistically, we found that SLC9A2 inhibits epithelial-mesenchymal transition (EMT) and metastasis by suppressing the STAT3 signaling pathway. Moreover, SLC9A2 reduces VEGFA secretion, normalizing tumor vasculature and reshaping the tumor microenvironment (TME), which ultimately enhances anti-tumor immunity. Comparative analysis of CRC tissue samples showed reduced SLC9A2 expression in tumor tissues compared to adjacent normal tissues, with a negative correlation to TNM staging. Importantly, higher SLC9A2 expression was associated with better treatment responses in immunotherapy cohorts.
    CONCLUSION: These findings highlight the critical role of SLC9A2 in regulating metastasis, angiogenesis, and TME remodeling in CRC. By modulating the STAT3 pathway and tumor vasculature, SLC9A2 emerges as a potential prognostic biomarker and therapeutic target. Targeting SLC9A2 may enhance immune responses and improve treatment outcomes in CRC, offering a promising avenue for future therapeutic strategies.
    Keywords:  Angiogenesis; Colorectal Cancer; Liver metastasis, immunotherapy; SLC9A2
    DOI:  https://doi.org/10.1186/s13046-025-03422-7
  21. Nat Cancer. 2025 Jun 04.
      Resistance to tyrosine kinase inhibitors (TKIs) dampens their clinical efficacy in hepatocellular carcinoma (HCC). Stress granules formed by phase separation are essential to stress response and can be involved in therapy resistance, but their mechanisms in HCC are unclear. Here our screen shows that the atypical serine/threonine kinase RIOK1 is highly expressed in HCC, linked to poor prognosis, and transcriptionally activated by NRF2 under various stress conditions. RIOK1 undergoes liquid-liquid phase separation by incorporating IGF2BP1 and G3BP1 into stress granules that sequestrate PTEN messenger RNA reducing its translation. This process activates the pentose phosphate pathway, facilitating stress resolution and cytoprotection against TKI. We further show that the small-molecule inhibitor chidamide downregulates RIOK1 and enhances TKI efficacy. RIOK1-positive stress granules are found in donafenib-resistant tumors from patients with HCC. These findings reveal a link between stress granule dynamics, metabolic reprogramming and HCC progression, offering the potential means to improve TKI efficacy.
    DOI:  https://doi.org/10.1038/s43018-025-00984-5
  22. bioRxiv. 2025 May 22. pii: 2025.05.17.654633. [Epub ahead of print]
      TP53 mutations confer treatment resistance across multiple cancers. Mechanisms of therapy resistance, beyond affecting transactivation of BCL-2 family genes, remain a mystery. Here, we report that TP53 mutated AML, triple negative breast cancer, and colorectal cancer escape therapy-induced apoptosis due to inability to activate caspase-3/7, despite having normal mitochondrial outer membrane permeabilization (MOMP) induction. To identify post-MOMP determinants of therapy resistance in TP53 mutated AML, we applied a multiomics approach - whole-genome CRISPR screen, bulk/single-cell RNAseq, and high-throughput drug screen. BIRC5 , encoding survivin, was selectively upregulated in paired hematopoietic stem/multipotent progenitor cells from TP53 mutant AML patients, with further enrichment after venetoclax-azacitidine (VenAza) relapse. Critically, BIRC5 was also upregulated in 17 of 26 TP53 mutant TCGA cancers. Genetic ablation of BIRC5 resensitized TP53 mutated AML to standard therapy by restoring caspase activation, validating therapeutic relevance. Importantly, targeting IAPs and survivin using clinically relevant inhibitors overcame VenAza resistance of TP53 mutant tumors in vivo , achieving sustained AML suppression. Combination with survivin inhibitors also overcame chemotherapy resistance in TP53 deficient solid cancers. Together, we discovered that wild-type TP53 is required in post-MOMP signaling and that BIRC5 dependency is an effective therapeutic target for poor prognosis, TP53 mutated cancers.
    DOI:  https://doi.org/10.1101/2025.05.17.654633
  23. Nat Commun. 2025 May 31. 16(1): 5068
      Lipid synthesis plays a central role in cell structure, signaling, and metabolism. A general method for the abiogenesis of natural lipids could transform the development of lifelike artificial cells and unlock new ways to explore lipid functions in living cells. Here, we demonstrate the abiotic formation of natural lipids in water using visible-light-driven photoredox chemistry. Radical-mediated coupling of hydrocarbon tails to polar single-chain precursors yields lipids identical to those enzymatically formed. Spatiotemporally controlled lipid generation promotes de novo vesicle formation, growth, and division. Lipid synthesis can be driven by RNA aptamers that specifically bind and activate photocatalysts, establishing a direct link between abiotic lipid metabolism and nucleic acid sequence. Light-mediated assembly of bioactive lipids can take place in living cells, triggering signaling events such as apoptosis and protein kinase C (PKC) activation. Our finding that photochemical lipid synthesis can be driven by simple genetic elements could be the starting point for developing protocells capable of Darwinian evolution. Additionally, the ability to generate specific membrane lipids in living cells with precise spatiotemporal control will advance studies on how lipid structure influences cellular function.
    DOI:  https://doi.org/10.1038/s41467-025-60358-4
  24. Redox Biol. 2025 May 22. pii: S2213-2317(25)00206-X. [Epub ahead of print]84 103693
       AIMS: This study investigates the mechanisms underlying acquired resistance to FGFR tyrosine kinase inhibitor (FGFR-TKI) in gastric cancer (GC), focusing on the interplay between ferroptosis and lipid metabolism of tumor cells.
    METHODS: We constructed FGFR-TKI-resistant cell lines from GC cells. RNA sequencing was performed to identify differentially expressed genes (DEGs) related to ferroptosis and assess lipid metabolism in resistant cells. GC microenvironment lipid profile was characterized by HPLC-MS/MS lipidomics. The effects of CHAC1 and cholesterol synthesis modulation on ferroptosis and FGFR-TKI resistance were assessed using in vitro and in vivo models.
    RESULTS: We found that FGFR-TKI can induce ferroptosis in FGFR-TKI-sensitive cells, while resistant cells exhibit decreased sensitivity to ferroptosis due to reduced CHAC1 expression, a key glutathione-specific degrading enzyme. Overexpression of CHAC1 enhances FGFR-TKI cytotoxicity. Additionally, cholesterol accumulation in resistant cells, associated with diminished stearic acid (SA) uptake, confers FGFR-TKI-induced ferroptosis resistance. In vivo studies show that CHAC1 overexpression or cholesterol synthesis inhibition can reverse FGFR-TKI resistance, which is dependent on ferroptosis.
    CONCLUSIONS: Dysregulated lipid homeostasis downregulated CHAC1-mediated ferroptosis, leading to FGFR-TKI resistance in gastric cancer. Overexpression of CHAC1 or inhibiting cholesterol synthesis presents promising therapeutic strategies to overcome FGFR-TKI resistance in GC.
    Keywords:  CHAC1; Cholesterol metabolism; FGFR-TKI resistance; Ferroptosis; Gastric cancer; Lipidomics
    DOI:  https://doi.org/10.1016/j.redox.2025.103693
  25. Cancer Res Commun. 2025 Jun 06.
      Resistance to chemotherapy is a critical challenge in triple negative breast cancer (TNBC). In this study, the proteomes of pretreatment core biopsy samples from 16 TNBC patients with differential response to neoadjuvant chemotherapy (NAC) were analyzed by nanoLC-MS/MS to identify biomarkers of intrinsic chemotherapy resistance. This led to the identification of cytochrome P450, family 1, subfamily B, polypeptide 1 (CYP1B1) and 71 additional proteins as significantly more abundant in chemoresistant than chemosensitive TNBC. Immunohistochemical analysis of 80 TNBC confirmed an association between elevated tumor cell CYP1B1 and residual cancer burden class 2/3 disease after NAC in T cell-excluded (TCE) TNBC (P <0.01), but not in T cell-infiltrated TNBC. The frequency of complete pathologic response in TCE-TNBC with elevated CYP1B1 was 18% versus 56% in TCE-TNBC with low CYP1B1 and 75% in T cell-infiltrated TNBC. Retrospective review of the chemotherapy regimens suggested that TCE-TNBC with elevated CYP1B1 were particularly resistant to doxorubicin/cyclophosphamide. This study is the first to associate resistance to neoadjuvant chemotherapy in TNBC with elevated CYP1B1.
    DOI:  https://doi.org/10.1158/2767-9764.CRC-25-0034