bims-meproc Biomed News
on Metabolism in Prostate Cancer
Issue of 2025–07–20
nine papers selected by
Grigor Varuzhanyan, UCLA



  1. Prostate Cancer. 2025 ;2025 8842203
      Background: Prostate cancer is the most common malignancy in men. Sestrin2 (SESN2) has antitumor activity against several types of cancers. However, the effect of SESN2 on prostate cancer is not well known. In this study, we showed that SESN2 inhibits human prostate cancer. Materials and Methods: To investigate the contribution of Sestrin2 to prostate cancer, we performed a bioinformatic analysis of the Cancer Genome Atlas database and Gene Expression Profiling Interactive Analysis. Using the Sestrin2 overexpression vector, we identified proliferation, migration, and invasion in prostate cancer cells. Furthermore, the effect of Sestrin2 on autophagy was confirmed by Western blot analysis and immunofluorescence staining. Results: We showed that expression of SESN2 was reduced in prostate cancer tissues and cell lines, and low expression of SESN2 correlated with decreased survival in prostate cancer patients. We have shown that SESN2 inhibits cell viability and cell proliferation-related protein levels in PC3 and DU145 prostate cancer cells. SESN2 inhibited EMT-related protein and migration and invasion levels. SESN2 promoted autophagy by increasing autophagy-related protein levels and LC3-positive cells. SESN2 increased pAMPK and decreased pmTOR protein levels. Furthermore, we used rapamycin, an mTOR inhibitor, to determine whether the AMPK/mTOR signaling pathway regulates autophagy in prostate cancer cells. Conclusion: Our study suggests that SESN2 inhibits prostate cancer cells by inducing autophagy through the AMPK/mTOR signaling pathway. These results indicate that SESN2 might be a novel target for prostate cancer.
    Keywords:  SESN2; autophagy; metastasis; proliferation; prostate cancer
    DOI:  https://doi.org/10.1155/proc/8842203
  2. bioRxiv. 2025 Jun 12. pii: 2025.06.09.658523. [Epub ahead of print]
      The bone microenvironment is abundant in adipocytes and fosters metastatic progression, but the underlying mechanisms are not fully understood. We hypothesize that Stearoyl-Coenzyme A Desaturase (SCD) acts as a tumor-promoting enzyme by modulating cellular stress to support the growth and survival of prostate cancer (PCa) in bone. We observe that SCD-amplified PCa cells are highly sensitive to SCD loss and show reduced PCa spheroid size, diminished mTOR signaling, and increased ER stress. Notably, SCD expression is further increased by adipocytes in SCD-amplified cell lines, and its loss increases DNA damage and activates repair pathways in PCa cells only when exposed to adipocytes. Furthermore, we observe PCa cell lines utilize SCD to regulate adipocyte-induced lipid peroxidation. Aligned with these results, pharmacological SCD inhibition in mice bearing SCD-amplified bone tumors reduces tumor size and reveals histochemical evidence of increased ER stress and DNA damage. Collectively, our data highlight the impact of SCD loss on SCD-amplified tumors and suggest germline characteristics of tumors may dictate their response to redox insult and the possibility of targeting DNA repair pathways in combination with SCD inhibition.
    DOI:  https://doi.org/10.1101/2025.06.09.658523
  3. Cancers (Basel). 2025 Jul 04. pii: 2243. [Epub ahead of print]17(13):
      Cancer is hallmarked by uncontrolled cell proliferation and enhanced cell survival, driven by a complex interplay of factors-including genetic and epigenetic changes-that disrupt metabolic and signaling pathways and impair organelle function. While the roles of mitochondria and the endoplasmic reticulum in cancer are widely recognized, emerging research is now drawing attention to the involvement of peroxisomes in tumor biology. Peroxisomes are essential for lipid metabolism, including fatty acid α- and β-oxidation, the synthesis of docosahexaenoic acid, bile acids, and ether lipids, as well as maintaining redox balance. Despite their critical functions, the role of peroxisomes in oncogenesis remains inadequately explored. Prostate cancer (PCa), the second most common cancer in men worldwide, exhibits a unique metabolic profile compared to other solid tumors. In contrast to the glycolysis-driven Warburg effect, primary PCa relies primarily on lipogenesis and oxidative phosphorylation. Peroxisomes are intricately involved in the metabolic adaptations of PCa, influencing both disease progression and therapy resistance. Key alterations in peroxisomal activity in PCa include the increased oxidation of branched-chain fatty acids, upregulation of α-methylacyl coenzyme A racemase (a prominent PCa biomarker), and downregulation of 1-alkyl-glycerone-3-phosphate synthase and catalase. This review critically examines the role of peroxisomes in PCa metabolism, progression, and therapeutic response, exploring their potential as biomarkers and targets for therapy. We also consider their relationship with androgen receptor signaling. A deeper understanding of peroxisome biology in PCa could pave the way for new therapies to improve patient outcomes.
    Keywords:  androgen receptor; ether lipids; fatty acid oxidation; metabolic rewiring; peroxisomes; prostate cancer; redox homeostasis
    DOI:  https://doi.org/10.3390/cancers17132243
  4. Int J Mol Sci. 2025 Jun 23. pii: 6013. [Epub ahead of print]26(13):
      Prostate cancer (PCa) therapy faces challenges due to tumor heterogeneity, plasticity, and progression. Metabolic reprogramming, a dynamic process, has emerged as a key focus in PCa treatment. However, conventional therapies targeting cancer-specific metabolic pathways or employing chemosensitizers are often limited by compensatory mechanisms and metabolic complexity. This review highlights the roles of transcription factors, including AR, p53, c-Myc, HIF-1, Nrf2, and PPARγ, in regulating PCa metabolism by influencing signaling pathways, enzymes, and gene expression. Multi-target compounds, particularly natural products, show potential for disrupting multiple metabolic enzymes, opening up new research possibilities. Notable examples include β-elemene, juglone, tannic acid, and withaferin A, which target critical metabolic processes through enzyme inhibition, transcription factor modulation, epigenetic changes, and protein interaction disruption. Naturally derived metabolites can elicit transversal responses in diverse metabolic pathways, particularly in p53 and MYC transcription factors. Additionally, compounds such as pentacyclic terpenoids (ursolic acid with ursane skeleton), sulforaphane, and isothiocyanate-related moieties may induce metabolic and epigenetic changes through S-adenosyl methionine (SAM) and acetyl-CoA modulation, potentially affecting new areas of research through metabolic processes. We propose a cooperative crosstalk between metabolic reprogramming and transcription factors/epigenetic modulation in PCa. This approach holds potential for expanding PCa therapeutics and opening new avenues for research.
    Keywords:  cancer treatment; epigenetics; metabolic reprogramming; prostate cancer; transcription factor
    DOI:  https://doi.org/10.3390/ijms26136013
  5. Lipids Health Dis. 2025 Jul 14. 24(1): 238
       OBJECTIVE: This study aimed to investigate the regulatory role and mechanisms of chenodeoxycholic acid (CDCA) on lipid metabolism, and to evaluate its therapeutic potential in lipid metabolism-related diseases such as acne, seborrheic alopecia, and prostate cancer, with the aim of identifying safer and more sustainable treatment alternatives.
    METHODS: Lipid-overproduction models were established using RM-1 (murine prostate cancer cells) and primary sebaceous gland cells, stimulated by linoleic acid (LA) and dihydrotestosterone (DHT), respectively. CDCA's mechanisms were explored using transcriptomics, proteomics, and fatty acid-targeted metabolomics. In vivo validation was conducted via intradermal injection of CDCA into the sebaceous gland area of golden hamsters to assess its effect on sebaceous lipid metabolism.
    RESULTS: CDCA significantly reduced intracellular lipid accumulation in both murine prostate cancer cells (RM-1) and primary sebaceous gland cells, and suppressed the expression of the androgen receptor (AR), along with the downregulation of key lipogenic enzymes (SREBF1, FASN, FADS2). Mitochondrial membrane potential was restored in CDCA-treated cells. Multi-omics analyses revealed that CDCA modulated fatty acid biosynthesis and degradation, energy metabolism, mitochondrial function, and Peroxisome Proliferator-Activated Receptor Gamma (PPAR-γ) signaling. Lipidomic profiling demonstrated a shift from saturated to unsaturated fatty acid dominance after CDCA treatment. In vivo, CDCA decreased sebaceous lipid accumulation and downregulated PPAR-γ expression in golden hamsters.
    CONCLUSION: CDCA exerted multifaceted regulatory effects on lipid metabolism, hormone signaling, and mitochondrial dynamics. These effects contributed to the maintenance of sebaceous gland homeostasis and supported the development of innovative and potentially more biocompatible therapies for lipid-related disorders.
    Keywords:  Chenodeoxycholic acid (CDCA); Fatty acid metabolism; Lipid homeostasis
    DOI:  https://doi.org/10.1186/s12944-025-02656-w
  6. Int J Mol Sci. 2025 Jun 20. pii: 5939. [Epub ahead of print]26(13):
      The treatment of choice for prostate cancer is androgen deprivation (ADT) and novel hormonal agents such as Abiraterone, Enzalutamide, or Apalutamide. Initially, this therapy is highly effective, but a significant challenge arises as most patients eventually develop resistance, resulting in castration-resistant prostate cancer (CRPC). Furthermore, the sequential use of these drugs can lead to cross-resistance, diminishing their efficacy. Tumor heterogeneity plays a pivotal role in the development of resistance to different treatments. This study utilized cellular models of CRPC to assess the response to Apalutamide when it was administered as a second- or third-line treatment. Functional and genetic analyses were conducted in various CRPC cell models exposed to Apalutamide. These analyses included real-time cell monitoring assays, flow cytometry, clonogenicity assays, and RT-qPCR. CRPC cell models were capable of continued proliferation, maintained cell cycle profiles similar to those of untreated cells, and retained their clonogenic potential. Cross-resistance to Apalutamide in models of ADT, ADT plus Enzalutamide, or Abiraterone resistance did not correlate with the expression levels of AR-V7 and AR-V9 variants. Gene expression analysis of resistant prostate cancer cell lines revealed that treatment with Apalutamide induced the emergence of more aggressive phenotypes, including cancer stem cells or neuroendocrine differentiation profiles. Most CRPC cell models developed cross-resistance to Apalutamide and were able to proliferate and retain their clonogenic capability. Apalutamide resistance was not linked to the expression of AR-V7 or AR-V9 variants but was instead associated to bypass of AR signaling pathway and the emergence of more aggressive expression profiles.
    Keywords:  Abiraterone; Apalutamide; Enzalutamide; androgen deprivation therapy (ADT); castration-resistant prostate cancer (CRPC); prostate cancer
    DOI:  https://doi.org/10.3390/ijms26135939
  7. Front Cell Dev Biol. 2025 ;13 1551010
       Introduction: Drug repurposing is gaining consideration in cancer due to the challenges of poor outcomes and resistance associated with the current conventional modalities. Non-steroidal anti-inflammatory drugs (NSAIDs), widely used for treating inflammation, are being explored for their potential efficacy in cancer treatment, including prostate cancer (PCa). This study aims to evaluate the efficacy of Piroxicam (PXM), an NSAID, in enhancing the sensitivity of PCa cells to chemotherapy and hormonal drugs.
    Methods: Computational analysis was conducted to identify differentially expressed genes between our established murine PCa cell models, PLum-AD (androgen-dependent) and PLum-AI (androgen-independent), to uncover potential therapeutic targets. In two-dimensional (2D) cell culture, cell proliferation, viability, and migration assays were performed on PLum-AD and PLum-AI cells treated with PXM alone or in combination with docetaxel (Doc) or enzalutamide (Enz). Additionally, the impact of these treatments on stem-like progenitor cells was assessed using three-dimensional (3D)-Matrigel™-based sphere-forming and organoid formation assays.
    Results: Transcriptomic analysis revealed that inflammatory pathways are enriched during PCa progression, making them viable targets for NSAID-based interventions. Single treatment of PXM demonstrated significant anti-cancer effects on PLum-AD and PLum-AI cells, evidenced by reduced cell proliferation, viability, migration, sphere growth, and organoid growth.
    Discussion: Importantly, PXM treatment in combination with Doc or Enz resulted in more pronounced antineoplastic effects compared to single-drug exposure. Our work suggests PXM as a potential adjunctive therapy to enhance the efficacy of conventional treatments in PCa patients.
    Keywords:  docetaxel; drug repurposing; enzalutamide; organoids; piroxicam; prostate cancer; tumor spheres
    DOI:  https://doi.org/10.3389/fcell.2025.1551010
  8. Mol Immunol. 2025 Jul 16. pii: S0161-5890(25)00177-4. [Epub ahead of print]185 27-38
      Drug resistance in cancer treatment is a major challenge, and macrophage polarization plays a key role in the development of prostate cancer (PCa). Growth differentiation factor 15 (GDF15) is highly expressed in most cancers and is induced during anticancer treatment. The aim of this study was to investigate the regulatory mechanism of GDF15 in macrophage polarization and resistance to docetaxel (DTX) in PCa patients. We collected clinical samples from PCa patients to evaluate the expression level of GDF15 and its correlation with M2-type macrophage polarization. In this study, CCK-8, RTqPCR, flow cytometry and western blotting were used to investigate the mechanisms by which GDF15 regulates macrophage M2 polarization and PCa chemotherapy resistance. The results showed that GDF15 was significantly upregulated in PCa samples and was closely related to the level of M2 macrophage polarization. Further experiments revealed that M2 macrophages synthesize GDF15, which is involved in the regulation of DTX resistance in PCa cells. Following knockdown of GDF15 expression in M2-type macrophages, we observed that the resistance of PCa cells to DTX was significantly attenuated. This regulatory mechanism was achieved mainly through the inhibition of the PI3K/AKT signaling pathway, preventing the M2 polarization of macrophages. In conclusion, the upregulation of GDF15 in M2 macrophages can activate the PI3K/AKT signaling pathway, enhancing the DTX resistance of PCa cells. These findings provide new insights and potential targets for treatment strategies against PCa chemotherapy resistance.
    Keywords:  Docetaxel resistance; GDF15; Macrophage polarization; PI3K/AKT; Prostate cancer
    DOI:  https://doi.org/10.1016/j.molimm.2025.07.006
  9. Nucl Med Biol. 2025 Jun 30. pii: S0969-8051(25)00057-5. [Epub ahead of print]148-149 109048
       INTRODUCTION: Cancer cells often exhibit aberrant cellular metabolism, with a common characteristic being their reliance on anaerobic glucose utilization. Prostate cancer (PC), however, displays unique metabolic profiles that extend beyond glycolysis, notably incorporating amino acid metabolism. This divergence in metabolic patterns opens potential avenues for targeted therapeutic strategies using D-amino acids. In this work, we investigated the biodistribution and tumour accumulation of D- and L-[methyl-11C]alanine using positron emission tomography (PET) imaging in a genetic mouse model of prostate cancer.
    METHODS: D- and L-[methyl-11C]alanine were synthesized according to established protocols. Conditional PTEN knockout mice (n = 6) were used as a model for prostate cancer. Dynamic PET-CT scans were performed for 60 min immediately following intravenous administration of the radiolabelled amino acids at the ages of 4 and 7 months. PET images were reconstructed, and volumes of interest (VOIs) were delineated in major organs, including the prostate and bladder. Dynamic time-activity curves (TACs) were analysed in terms of Standardized Uptake Values (SUV). After imaging, samples of the reproductive system were collected for static PET-CT imaging and subsequent histological analysis.
    RESULTS AND DISCUSSION: L- and D-[methyl-11C]alanine were synthesized with good radiochemical yields and high enantiomeric excess using enantioselective alkylation with [11C]methyl iodide in the presence of a chiral phase transfer catalysts. PET imaging of a genetic faithful mouse model of prostate cancer demonstrated that D-[methyl-11C]alanine exhibited faster blood clearance, higher age-dependent kidney retention, and greater prostate lesion uptake at 7 months compared to L-[methyl-11C]alanine. Histological analysis confirmed malignant lesions in the prostate of PTEN knockout mice, corroborating the PET imaging findings.
    CONCLUSIONS: Our study offers valuable insights into the metabolic landscape of prostate cancer using a genetic mouse model that closely mimics human disease pathogenesis. The significantly greater accumulation of D-[methyl-11C]alanine compared to its L-enantiomer underscores the potential of D-amino acids as biomarkers, and their potential use to interfere with cancer cell metabolism.
    Keywords:  D-amino acid; Metabolism; Positron emission tomography; Prostate cancer
    DOI:  https://doi.org/10.1016/j.nucmedbio.2025.109048