bims-merabr Biomed News
on Metabolic rewiring in aggressive breast cancer
Issue of 2025–03–02
eight papers selected by
Barbara Mensah Sankofi, University of Oklahoma Health Sciences Center



  1. Cell Death Dis. 2025 Feb 27. 16(1): 142
      Activating transcription factors (ATFs), members of the adaptive-response gene family, participate in cellular processes to aid adaptations in response to extra and/or intracellular changes. In this study, we observed that one of the ATFs, Activating transcription factor 3 (ATF3), is upregulated under hypoxia via alterations in the epigenetic landscape of its promoter, followed by transcriptional upregulation. Under hypoxic conditions, Hypoxia-inducible factor 1-alpha (HIF1ɑ) alleviates methylation at the ATF3 promoter by recruiting TET1 and induces ATF3 transcription. In addition, our RNA-seq analysis showed that ATF3 globally affects transcription under hypoxia and controls the processes of EMT and cancer invasion by stimulating the transcription of Prolyl 4-Hydroxylase Subunit Alpha 1 (P4HA1), an enzyme which enhances invasion-conducive extracellular matrix (ECM) under hypoxic conditions. Prolyl hydroxylases play a critical role in the hydroxylation and deposition of collagen in the extracellular matrix (ECM) during the evolution of cancer, which is necessary for metastasis. Importantly, P4HA1 undergoes alternative splicing under hypoxia, where the inclusion of exon 9a is increased. Interestingly, involvement of ATF3 in P4HA1 splicing was also evident, as binding of ATF3 at intron 9a led to demethylation of this DNA region via recruitment of TET1. Furthermore, we also show that the demethylated DNA region of intron 9a then becomes accessible to CCCTC-binding factor (CTCF). Thus, a cascade of demethylation via ATF3 recruited TET1, followed by increased RNA Pol II pause at intron 9a via CTCF, leads to inclusion of exon 9a. The P4HA1 9a isoform leads to enhanced invasion under hypoxic conditions by increasing deposition of collagen in the ECM. These results reveal a novel hypoxia-induced HIF1ɑ-ATF3-P4HA1 axis which can potentially be exploited as a therapeutic target to impede EMT and ultimately breast cancer invasion. Hypoxia induced ATF3 regulates P4HA1 expression and alternative splicing to promote breast cancer invasion.
    DOI:  https://doi.org/10.1038/s41419-025-07461-y
  2. Stem Cell Res Ther. 2025 Feb 23. 16(1): 83
       BACKGROUND: Considerable evidence suggests that tumor initiation, malignancy, metastasis and recurrence occur due to emergence of cancer stem cells (CSCs). Fas binding factor 1 (FBF1) is a multifunctional protein that plays essential roles in the regulation of development and cell fate decisions. However, the function in maintaining stem cell-like properties of breast cancer remains elusive.
    METHODS: Tissue microarray was used to evaluate FBF1 expression. Cancer stemness assays were performed in FBF1 silencing and overexpressing cells in vitro and in a xenograft model in vivo. RNA sequencing, immunofluorescence and immunoprecipitation assays were performed to explore the underlying mechanism. Clinical expression and significance of FBF1 and stemness-associated factors were explored by analyzing datasets.
    RESULTS: We report that FBF1 was highly expressed in breast cancer and significantly correlated with clinical progression. Silencing FBF1 in MDA-MB-231 cells restrained CSCs properties, including side population, sphere formation and migration, whereas ectopic FBF1 expression increased the side population proportion, enhanced the sphere formation ability, and promoted the expression of core stemness genes, such as SOX2, OCT4, KLF4 and NANOG, as well as facilitated metastasis of T47D breast cancer cells. Furthermore, mice bearing FBF1-overexpressed T47D xenografts had higher tumorigenic frequency and stronger metastasis potential. In addition, exploration of the underlying mechanism indicated that FBF1 binds PI3K which then activates PI3K-AKT phosphorylation cascades. Then the activated p-AKT interacts with stemness marker SOX2, elevates SOX2 and OCT4 activity, and finally forms PI3K/AKT/SOX2 axis, which mediates stem cell-like identities. Moreover, PI3K inhibitors abolished FBF1-mediated signaling pathway and diminished breast cancer stemness in vitro and in vivo. In 24 human breast cancer samples, we found a good positive correlation between the expression of FBF1 and p-AKT, as well as between FBF1 and SOX2 as determined by IHC. Clinical data showed that FBF1 expression was positively correlated with the expression of POU5F1 (OCT4), AKT1 and was negatively correlated with PTEN, which is a negative regulator of PI3K/AKT signaling.
    CONCLUSION: Collectively, we identified a potential CSCs regulator and suggested a novel mechanism by which FBF1 governs cancer cell stemness. This study thus introduces an effective target for the diagnosis and treatment of breast cancer.
    Keywords:  Breast cancer; Cancer stem cell; FBF1; SOX2; Stemness
    DOI:  https://doi.org/10.1186/s13287-025-04194-9
  3. Transl Oncol. 2025 Feb 21. pii: S1936-5233(25)00052-X. [Epub ahead of print]54 102321
       BACKGROUND: RING finger protein 135 (RNF135) is identified as a regulator in certain cancer types. However, its role and molecular mechanisms in breast cancer are still unclear.
    METHODS: Herein, we investigated the level of RNF135 in tumor tissues of breast patients using the online database and confirmed the data by real-time PCR and western blot analysis. The effects of RNF135 on stemness maintenance and migration/invasion capability of breast cells were investigated by sphere formation, flow cytometry, and transwell assays. Limiting dilution xenograft assay and metastatic model were applied to assess the implications of RNF135 in tumorigenesis, chemoresistance, and metastasis.
    RESULTS: Our results revealed that RNF135 was upregulated in tumor tissues of breast patients, especially in metastatic patients. Knockdown of RNF135 suppressed stemness, and migration/invasion capability of breast cancer cells. Conversely, RNF135 overexpression enhanced the stemness and migration/invasion ability of breast cancer cells. Limiting dilution xenograft and metastatic assays demonstrated that RNF135 was required for the self-renewal of CSCs to initiate breast cancer development and metastasis. Mechanistically, DDX58 was identified as the substrate of RNF135 and RNF135 could facilitated the ubiquitination and degradation of DDX58. Notably, overexpression of DDX58 rescued the promoting effects of RNF135 on the stemness and migration/invasion ability of breast cancer cells.
    CONCLUSIONS: Overall, our results implied that RNF135 promotes the stemness of breast cancer cells by ubiquitinating and degrading DDX58 and targeting of RNF135/DDX58 axis might be a feasible method to suppress tumorigenesis and metastasis of breast cancer patients.
    Keywords:  Breast cancer; DDX58; Metastasis; RNF135; Stemness
    DOI:  https://doi.org/10.1016/j.tranon.2025.102321
  4. Cells. 2025 Feb 15. pii: 286. [Epub ahead of print]14(4):
      In the breast tumor microenvironment (TME), adipocytes exert a selective pressure on the behavior of breast cancer stem cells (BCSCs), which are involved in endocrine therapy resistance. In obesity, adipocytes secrete reduced levels of adiponectin, which promotes the growth and progression of ERα-positive breast cancer (BC). Here, we examined how low adiponectin levels affect the enrichment of the BCSC subpopulation and the mechanisms contributing to the maintenance of endocrine therapy resistance in BC. Flow cytometry, qRT-PCR, and Western blotting analysis were performed to assess stemness, the cell cycle, and apoptosis markers in MCF-7 wild-type (WT) and tamoxifen-resistant (TR) mammospheres. nLC-MS/MS was employed to profile and compare the proteome of BCSCs. Differentially expressed proteins were intersected with data from the MetacoreTM dataset. Our study demonstrated that adiponectin increased the percentage of CD44+/CD24-/ALDH1+ stem-like cells in TR MCF-7 mammospheres. Specifically, adiponectin contributed to the maintenance of BCSC bulk in TR MCF-7 cells through a slow cycling rate, supported by decreased levels of Cyclin D1 and Ki67 and increased p21 and p27 expression, and through escape from apoptosis, sustained by reduced ROS production and preserved maintenance of mitochondrial membrane potential. Our results provide new insights into the contribution of adiponectin to poor ERα-positive BC outcomes. Deeply understanding adiponectin's role in stemness may disclose novel therapeutic approaches to treat hormone-resistant obese BC patients.
    Keywords:  adiponectin; apoptosis; breast cancer; cancer stem cells; endocrine resistance; tamoxifen
    DOI:  https://doi.org/10.3390/cells14040286
  5. Front Immunol. 2025 ;16 1534936
       Introduction: Breast cancer is the most frequently diagnosed malignancy and a leading cause of cancer-related mortality among women worldwide. Although NADPH oxidase 4 (NOX4) has been implicated in various oncogenic processes, its exact function in breast cancer progression, metabolic reprogramming, and immune modulation remains unclear.
    Methods: We used murine 4T1 and EO771 breast cancer models to generate NOX4 knockout (KO) cell lines via CRISPR/Cas9. In vitro assays (cell proliferation, colony formation, wound healing, and Seahorse metabolic analyses) and in vivo orthotopic tumor studies assessed the impact of NOX4 loss. Transcriptomic changes were identified through RNA sequencing and gene set enrichment analysis. We performed MYC knockdown in NOX4 KO cells to investigate its mechanistic role. Flow cytometry characterized tumor-infiltrating immune cells. Finally, NOX4-overexpressing cells were tested for survival benefit and response to dual-checkpoint immunotherapy (anti-PD-1/anti-CTLA-4).
    Results: NOX4 deletion accelerated tumor growth in vivo and enhanced proliferation, colony formation, and migratory capacity in vitro. Metabolic profiling showed that NOX4 KO cells had elevated glycolysis and fatty acid oxidation, along with increased mitochondrial mass. Transcriptomic and enrichment analyses revealed MYC pathway activation in NOX4 KO cells; suppressing MYC reversed these hyperproliferative and metabolic changes. Immunologically, NOX4 KO reduced CD8+ T cell infiltration and function, partially due to lowered CCL11/CCL5 levels, while PD-L1 expression was upregulated. In contrast, NOX4 overexpression improved survival in mice and synergized with checkpoint blockade, demonstrating a positive effect on anti-tumor immunity.
    Discussion: These findings show that NOX4 constrains breast cancer aggressiveness by limiting MYC-driven metabolic adaptations and supporting CD8+ T cell-mediated immunity. Loss of NOX4 promotes a more malignant phenotype and dampens T cell responses, whereas its overexpression prolongs survival and enhances checkpoint inhibitor efficacy. Therapeutically targeting the NOX4-MYC axis and leveraging NOX4's immunomodulatory capacity could offer promising strategies for breast cancer management.
    Keywords:  MYC; NOX4; breast cancer; fatty acid oxidation; metabolic reprogramming
    DOI:  https://doi.org/10.3389/fimmu.2025.1534936
  6. bioRxiv. 2025 Feb 13. pii: 2025.02.10.637542. [Epub ahead of print]
      Obesity is an established risk factor for breast cancer (BC), yet the specific mechanisms driving this association remain unclear. Dysregulated lipid metabolism has emerged as a key factor in cancer cell biology. While obesity is often accompanied by hyperlipidemia, the isolated impact of elevated lipid levels on BC growth has not been experimentally tested. Using the E0771 orthotopic model of obesity-accelerated BC growth in immune-competent mice, we investigated the direct role of systemic lipids in tumor growth. Combining dietary and genetic mouse models, we show that elevated circulating lipids are sufficient to accelerate BC tumor growth even in the absence of obesity or alterations in blood glucose and/or insulin levels. Pharmacological lowering of systemic lipid levels attenuates BC growth in obese mice, suggesting a direct role for lipids in fueling tumor expansion. Notably, we also show that weight loss alone, without a corresponding reduction in lipid levels such as that induced by a ketogenic diet, fails to protect against BC, highlighting the necessity of targeting lipid metabolism in obesity-associated BC. Our findings establish hyperlipidemia as a critical driver of BC progression and suggest that lipid-lowering interventions may be a promising strategy to mitigate BC risk in obese individuals.
    DOI:  https://doi.org/10.1101/2025.02.10.637542
  7. Front Endocrinol (Lausanne). 2025 ;16 1523589
       Introduction: We previously demonstrated that progesterone (P4) can promote breast cancer cell proliferation and migration through activating the P4 receptor (PR)/cSrc-mediated signaling pathway. It has been suggested that high level of Src homology region 2 domain-containing phosphatase-2 (SHP2) might be involved in breast oncogenesis. This study aimed to investigate whether SHP2 is involved in the P4-mediated cSrc activation in breast cancer cells.
    Methods: T47D, MCF-7 and BT-483 breast cancer cell lines were used in this study. Cell proliferation and migration were examined using MTT technique and wound healing assay, respectively. Immunoprecipitation assay and Western blot analysis were performed to evaluate protein-protein interaction and protein expression, respectively. Small interfering RNA (siRNA) technique was used to knock down protein expression.
    Results: Knockdown of SHP2 expression abolished the P4-promoted cell proliferation and migration in T47D, MCF and BT-483 cell lines, suggesting that presence of SHP2 is essential for the P4-increased proliferation and migration of breast cancer cell lines. P4 (50 nM) treatment increased the complex formations of PR-cSrc-SHP2-caveolin-1, SHP2-p140Cap, and SHP2-Csk, and the level of p-cSrcY416 (activated form of cSrc). However, knockdown of SHP2 expression increased the complex formations of PR-cSrc-caveolin-1-Csk-p140Cap and the levels of p-caveolin-1, p-Csk and p-cSrcY527 (inactivated form of cSrc).
    Discussion: Our data suggest that SHP2 can bind to cSrc-negative regulatory proteins (p140Cap and Csk), hence preventing the interaction between cSrc and cSrc-negative regulatory proteins, leading to decreased phosphorylation of cSrc Y527 and prolonged cSrc activation. These findings highlight the role of SHP2 in the P4-promoted breast cancer cell proliferation and migration.
    Keywords:  Csk; SHP2; breast cancer cell lines; cSrc; p-cSrcY416; p-cSrcY527
    DOI:  https://doi.org/10.3389/fendo.2025.1523589
  8. Sci Rep. 2025 Feb 24. 15(1): 6593
      Mesenchymal stem cells (MSCs) have emerged as crucial players within the tumor microenvironment (TME), contributing through their paracrine secretome. Depending on the context, the MSC-derived secretome can either support or inhibit tumor growth. This study investigates the role of MSC-derived secretome in modulating breast cancer (BC) cell behavior, with a focus on ten-eleven translocation 1 (TET1), a DNA demethylase with known oncogenic properties in triple-negative breast cancer (TNBC). We first isolated and characterized human bone marrow-derived MSCs, and then assessed the impact of their secretome on BC cells. Treatment with the MSC-derived secretome significantly inhibited the proliferation and migration of both MDA-MB-231 and MCF-7 BC cell lines, resulting in reduced cell viability and migration rates compared to control cells. Western blot analyses revealed downregulation of Cyclin D1 and c-Myc, along with decreased expression of N-cadherin and increased expression of E-cadherin, indicating potential inhibition of the epithelial-to-mesenchymal transition. Differential gene expression analyses highlighted TET1 as significantly upregulated in TNBC tissues compared to normal samples. Further experiments confirmed that the MSC-derived secretome downregulated TET1 expression in BC cells, as evidenced by RT-qPCR and western blot analyses. To explore TET1's functional role, we silenced TET1 with siRNAs, observing cell cycle arrest and enhanced apoptosis-effects that mirrored those seen with MSC-secretome treatment. Notably, TET1 knockdown also increased MDA-MB-231 cell sensitivity to cisplatin, suggesting a role for TET1 in chemoresistance. These findings provide insight into the ability of MSCs to modulate BC cell progression through their secretome, highlighting the involvement of TET1 downregulation in inhibiting BC cell progression and enhancing cisplatin chemosensitivity. The MSC-derived secretome thus holds promise as an innovative, cell-free therapeutic approach in BC treatment.
    Keywords:  Breast cancer; Mesenchymal stem cells; Secretome; TET1
    DOI:  https://doi.org/10.1038/s41598-025-91314-3