bims-merabr Biomed News
on Metabolic rewiring in aggressive breast cancer
Issue of 2025–04–27
five papers selected by
Barbara Mensah Sankofi, University of Oklahoma Health Sciences Center



  1. Med Oncol. 2025 Apr 23. 42(5): 178
      Chemotherapy resistance remains a major obstacle in breast cancer treatment. This study identifies DCAF13, a substrate recognition receptor of the CRL4 ubiquitin ligase complex, as a critical regulator of chemotherapy resistance through c-Myc-driven metabolic reprogramming. We found that DCAF13 expression was significantly upregulated in chemotherapy-resistant breast cancer cell lines compared to their parental counterparts. Inhibition of DCAF13 enhanced chemotherapy sensitivity, whereas its overexpression suppressed drug sensitivity. Mechanistically, DCAF13 upregulated c-Myc expression, driving metabolic reprogramming, characterized by increased glycolysis and oxidative phosphorylation. This metabolic shift promoted cell proliferation and resistance to chemotherapy. Clinically, DCAF13 expression correlated with poor prognosis in breast cancer patients, particularly in advanced stages and triple-negative breast cancer (TNBC). Our findings highlight the DCAF13-c-Myc axis as a critical mediator of chemotherapy resistance, suggesting that targeting this pathway could provide novel therapeutic strategies to overcome drug resistance in breast cancer. Further clinical studies are needed to explore the potential of DCAF13 as a therapeutic target.
    Keywords:  Breast cancer; Chemotherapy resistance; DCAF13; Metabolic reprogramming; c-Myc
    DOI:  https://doi.org/10.1007/s12032-025-02722-4
  2. BMC Cancer. 2025 Apr 22. 25(1): 747
       BACKGROUND: FOXO1 is known to act as a tumor suppressor gene in breast cancer, but its exact mechanism of action remains unclear.
    OBJECTIVE: This study aimed to clarify how FOXO1 suppresses breast cancer cell proliferation and induces apoptosis.
    METHODS: Breast cancer cell lines were generated with stable knockdown or overexpression of FOXO1. RT-qPCR and western blot assays were conducted to confirm transfection efficiency. CCK-8 and colony formation assays were used to assess cell proliferation, while flow cytometry measured apoptosis. The cells were subcutaneously injected into nude mice, and the volume and mass of the resulting tumors were evaluated. Immunohistochemistry was used to analyze Ki-67 expression in the tumors. A TUNEL assay examined apoptosis in the tumor cells. We performed bioinformatic analysis to identify FOXO1-targeted miRNAs and their downstream target mRNAs.
    RESULTS: Overexpression of FOXO1 inhibited breast cancer cell proliferation and promoted apoptosis. In contrast, knockdown of FOXO1 enhanced cell proliferation and reduced apoptosis. Among the downstream miRNAs we identified, miR-99a-5p was found to be downregulated in breast cancer tissue. FOXO1 binds to the miR-99a promoter, facilitating its transcription. Inhibition of miR-99a-5p partially reversed the effects of FOXO1 overexpression on cell proliferation and apoptosis. E2F7, a target mRNA of miR-99a-5p, showed a negative correlation with FOXO1 expression in breast cancer mRNAs we screened. Silencing E2F7 partially mitigated the inhibitory effects of miR-99a-5p on proliferation and apoptosis in FOXO1-overexpressing cells. E2F7 binds to the FOXO1 promoter, thus suppressing its transcription and reducing its expression.
    CONCLUSION: FOXO1 suppresses breast cancer cell proliferation and promotes apoptosis by enhancing the transcription and expression of miR-99a-5p, while inhibiting its target gene E2F7. E2F7, in turn, represses the transcription of FOXO1, lowering its expression.
    Keywords:  Apoptosis; Breast cancer; Cell proliferation; E2F7; FOXO1; miR-99a-5p
    DOI:  https://doi.org/10.1186/s12885-025-14111-1
  3. Neoplasia. 2025 Apr 18. pii: S1476-5586(25)00044-2. [Epub ahead of print]65 101165
      Approximately 70-80% of breast cancers rely on estrogen receptor alpha (ERα) for growth. The unfolded protein response (UPR), a cellular response to endoplasmic reticulum stress (ERS), is an important process crucial for oncogenic transformation. The effect of ERS on ERα expression and signaling remains incompletely elucidated. Here, we focused on the regulatory mechanisms of ERS on ERα expression in ER-positive breast cancer (ER+ BC). Our results demonstrate that ERα protein and mRNA levels in ER+ BC cells are considerably reduced by the ERS inducers thapsigargin (TG) and brefeldin A (BFA) via the PERK/eIF2α/ATF4 signaling pathway. ChIP-qPCR and luciferase reporter gene analysis revealed that ERS induction facilitated ATF4 binding to the ESR1 (the gene encoding ERα) promoter region, thereby suppressing ESR1 promoter activity and inhibiting ERα expression. Furthermore, selective activation of PERK signaling or ATF4 overexpression attenuated ERα expression and tumor cell growth both in vitro and in vivo. In conclusion, our results demonstrate that ERS suppresses ERα expression transcriptionally via the PERK/eIF2α/ATF4 signaling. Our study provides insights into the treatment of ER+ BC by targeting ERα signaling through selective activation of the PERK branch of the UPR.
    Keywords:  ATF4; Breast cancer; ERα; ESR1; Endoplasmic reticulum stress; Unfolded protein response
    DOI:  https://doi.org/10.1016/j.neo.2025.101165
  4. Am J Physiol Endocrinol Metab. 2025 Apr 22.
      The transcription factor forkhead box (FOX)M1 regulates β-cell proliferation and insulin secretion. Our previous work demonstrates that expressing a constitutively active form of FOXM1 (FOXM1*) in β cells increases β-cell function, proliferation and mass in male mice. However, in contrast to what is observed in males, we demonstrate here that in female mice, expression of FOXM1* in β cells does not affect β-cell proliferation or glucose tolerance. Similarly, FOXM1* transduction of male but not female human islets enhances insulin secretion in response to elevated glucose. We, therefore, examined the mechanism behind this sexual dimorphism. Estrogen contributes to diabetes susceptibility differences between males and females, and the estrogen receptor (ER)α is the primary mediator of β-cell estrogen signaling. Moreover, in breast cancer cells, ERα and FOXM1 work together to drive gene expression. We, therefore, examined whether FOXM1 and ERα functionally interact in β cells. FOXM1* rescued elevated fasting glucose, glucose intolerance, and HOMA-B in female mice with a β-cell-specific ERα deletion. Furthermore, in the presence of estrogen, the FOXM1 and ERα cistromes exhibit significant overlap in βTC6 β cells. In addition, FOXM1 and ERα binding sites frequently occur in complex enhancers co-occupied by other islet transcription factors. These data indicate that FOMX1 and nuclear ERα cooperate to regulate β-cell function and suggest a general mechanism contributing to the lower incidence of diabetes observed in women.
    Keywords:  Beta cells; Estrogen receptor; Foxm1
    DOI:  https://doi.org/10.1152/ajpendo.00438.2024
  5. Cells. 2025 Apr 13. pii: 590. [Epub ahead of print]14(8):
      High-mobility group box 1 (HMGB1) is a nuclear chromatin protein overexpressed in various cancers and linked to tumor progression. Outside the cell, HMGB1 binds to receptors such as the receptor for advanced glycation end products (RAGE), promoting metastasis. Targeting this signaling pathway may provide a new therapeutic strategy for aggressive cancers. Metformin, a well-established antidiabetic drug, directly interacts with HMGB1, inhibiting its pro-inflammatory functions. This study investigates metformin's effects on the HMGB1/RAGE signaling pathway in triple-negative breast cancer (TNBC) cells. Using wound-healing and colony formation assays, we demonstrate that metformin reduces HMGB1-induced cell migration and proliferation. Immunoblotting and immunofluorescence analyses reveal that metformin decreases RAGE stabilization on the cell membrane, disrupts NF-κB signaling, and reverses the epithelial-to-mesenchymal transition (EMT) by increasing E-cadherin, reducing vimentin, and stabilizing β-catenin at the cell membrane. Furthermore, metformin lowers HMGB1 and RAGE protein levels, disrupting the positive feedback loop that promotes cancer aggressiveness. These findings highlight metformin's potential as a therapeutic agent in TNBC by inhibiting HMGB1/RAGE-driven metastasis.
    Keywords:  EMT; HMGB1; breast cancer; metastasis; metformin
    DOI:  https://doi.org/10.3390/cells14080590