bims-stacyt Biomed News
on Metabolism and the paracrine crosstalk between cancer and the organism
Issue of 2025–11–09
seven papers selected by
Cristina Muñoz Pinedo, L’Institut d’Investigació Biomèdica de Bellvitge



  1. Int J Mol Med. 2026 Jan;pii: 13. [Epub ahead of print]57(1):
      Amino acid (AA) sensing plays an important role in maintaining cellular metabolic homeostasis as well as tumorigenesis and progression. Studies on classic AA sensing pathways such as rapamycin complex 1 (mTORC1) and general control nonderepressible 2 (GCN2) have revealed their central position in cancer metabolic reprogramming. AA sensing pathways are often hijacked in tumors to adapt to the nutrient‑deprived microenvironment, promoting cell proliferation, anti‑apoptosis and treatment tolerance. In addition, the regulation of AA sensing and transport plays a crucial role in maintaining the metabolic flexibility of tumor cells. By targeting the AA sensing mechanism, it is expected to disrupt the metabolic homeostasis of cancer cells, providing new strategies for precision therapy. The present review summarized the latest advances in the research on the role of the mTORC1 and GCN2 AA sensing pathways in tumor metabolism, emphasizing their potential and the challenges faced in cancer diagnosis and treatment. Additionally, it provided novel insights into the therapeutic targeting of AA sensing pathways and proposes future research directions aimed at overcoming current limitations in cancer metabolism therapy.
    Keywords:  amino acid sensing; metabolic reprogramming; targeted cancer therapy; tumor microenvironment
    DOI:  https://doi.org/10.3892/ijmm.2025.5684
  2. Horm Metab Res. 2025 Nov 07.
      An independent association between insulin resistance and cancer has been consistently reported in humans. Patients with cancer display insulin resistance or its clinical manifestations, and this metabolic adaptation precedes the clinical diagnosis of cancer. Insulin resistance in cancer patients is associated with a metabolic switch from oxidative metabolism toward glycolysis that spares oxygen to be used in anabolic processes and facilitates the fast production of energy and intermediate metabolites required for the rapid proliferation of cancer cells. In malignant cells, glucose consumption via glycolysis occurs under normoxic conditions (aerobic glycolysis). Pathogenic mechanisms underlying insulin resistance in cancer patients include hypoxia-inducible factor-1 upregulation and overproduction of cytokines, such as interferon, interleukin-6, interleukin-18, and interleukin-1β. Deficit of 2-oxoglutarate (α-ketoglutarate) has been detected in cancer cells and may facilitate hypoxia-inducible factor-1 assembly and activity. Overproduction of cytokines in cancer patients follows activation of the immune system by abnormal nucleic acid variants. Anomalous DNA or RNA structures are recognized by immune sensors and stimulate signaling pathways that ultimately increase cytokine production. Likewise, interferon overproduction occurs in congenital disorders that feature ineffectively repaired DNA lesions, such as Werner syndrome, Bloom syndrome, mutations in DNA polymerase-δ1, and ataxia telangiectasia. These diseases cause simultaneous insulin resistance and a high tendency to develop cancer, highlighting the relationship between the two processes. Defectively repaired DNA injury endangers genomic integrity, predisposing to cancer, and activates the immune system to increase interferon production and subsequent insulin resistance. Hypoxia-inducible factor-1 and cytokines induce insulin resistance by suppressing peroxisome proliferator-activated-γ in the subcutaneous adipose tissue.
    DOI:  https://doi.org/10.1055/a-2715-0775
  3. Clin Nutr ESPEN. 2025 Nov 04. pii: S2405-4577(25)02989-4. [Epub ahead of print]
       INTRODUCTION: Malnutrition is a frequent and harmful side effect of anticancer therapy, and even though it is known that cancer patients require sufficient nutritional support, nutrition is still not one of the first factors considered in routine clinical practice. The MOUSEION-012 meta-analysis explored the incidence of decreased appetite events among cancer patients receiving immunotherapy as monotherapies or in combination with other anticancer agents.
    METHODS: The MOUSEION-012 meta-analysis was recorded with PROSPERO n. CRD420251004424 and performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA). Proportions were assessed for any grade of decreased appetite and for grade ≥ 3 and two separate subgroups of meta-analysis were performed.
    RESULTS: A total of 36 manuscripts were included, specifically, 23 were observational studies and 13 were RCTones. A total of 11233 observations and 2317 events were recorded and heterogeneity among the studies was high and significant (P<0.001; tau2 = 1.23; I2 = 95.3% with a 95%-CI: [94.5%; 96.0%]) in Any Grade group. Considering Grade ≥ 3, a total of 11233 observations and 146 events were registered. The high heterogeneity among studies was significant (P<0.001; tau2 = 5.63; I2 = 77% with a 95%-CI: [70.2%; 82.2%]). Considering RCTstudies, a total of 15 studies were included, with a total of 10216 observations (5331 in the experimental group and 4885 in the control group) and 1920 events. Heterogeneity among the studies was high and significant (P<0.001; tau2 = 0.25; I2 = 85.4% with a 95%-CI: [77.4%; 90.6%]) for Any Grade group. Considering Grade ≥ 3, a total of 10216 observations (5331 in the experimental group and 4885 in the control group) and 124 events were reported. The heterogeneity across studies was moderate and significant (P=0.009; tau2 =075; I2 = 53.9% with a 95%-CI: [15.5%; 74.8%]).
    CONCLUSION: Appetite loss in cancer patients continues to be a significant clinical and research concern, and even while we have more options than ever before for managing cancer-related appetite loss, several challenges remain regarding the best ways to manage this symptom and enhance the quality of life for patients who experience it. In fact, appetite loss may be strictly linked to cancer cachexia, a multifactorial syndrome characterized by a progressive loss of skeletal muscle mass, along with adipose tissue wasting, systemic inflammation and other metabolic abnormalities leading to functional impairment.
    Keywords:  Anorexia; Appetite; Cancer; Immune checkpoint inhibitors; Immunotherapy; Neoplasms
    DOI:  https://doi.org/10.1016/j.clnesp.2025.10.031
  4. Nature. 2025 Nov 05.
      The endoplasmic reticulum (ER) is a highly interconnected membrane network that serves as a central site for protein synthesis and maturation1. A crucial subset of ER-associated transcripts, termed secretome mRNAs, encode secretory, lumenal and integral membrane proteins, representing nearly one-third of human protein-coding genes1. Unlike cytosolic mRNAs, secretome mRNAs undergo co-translational translocation, and thus require precise coordination between translation and protein insertion2,3. Disruption of this process, such as through altered elongation rates4, activates stress response pathways that impede cellular growth, raising the question of whether secretome translation is spatially organized to ensure fidelity. Here, using live-cell single-molecule imaging, we demonstrate that secretome mRNA translation is preferentially localized to ER junctions that are enriched with the structural protein lunapark and in close proximity to lysosomes. Lunapark depletion reduced ribosome density and translation efficiency of secretome mRNAs near lysosomes, an effect that was dependent on eIF2-mediated initiation and was reversed by the integrated stress response inhibitor ISRIB. Lysosome-associated translation was further modulated by nutrient status: amino acid deprivation enhanced lysosome-proximal translation, whereas lysosomal pH neutralization suppressed it. These findings identify a mechanism by which ER junctional proteins and lysosomal activity cooperatively pattern secretome mRNA translation, linking ER architecture and nutrient sensing to the production of secretory and membrane proteins.
    DOI:  https://doi.org/10.1038/s41586-025-09718-0
  5. Cell Death Dis. 2025 Nov 07. 16(1): 807
      Gastric cancer (GC) metastasis remains a major cause of poor prognosis, yet its molecular drivers are poorly understood. Here, we integrated single-cell RNA sequencing (scRNA-seq) of primary tumors and matched metastatic lymph nodes from six GC patients to identify a metastatic epithelial subpopulation characterized by EGR4 overexpression. Kaplan-Meier analysis revealed that high EGR4 expression correlated with reduced survival in GC patients. Mechanistically, chromatin immunoprecipitation sequencing (ChIP-seq) and luciferase assays demonstrated that EGR4 directly bound to the GDF15 promoter, driving its transcriptional activation. Functional studies showed that EGR4 promoted migration and metastasis via GDF15-mediated ErbB3/ErbB1 hetero-dimerization, which activated PI3K/AKT and MAPK/ERK pathways. Furthermore, CellChat analysis identified robust interactions between EGR4+ GC cells and cancer-associated fibroblasts (CAFs), particularly extracellular matrix (ECM)-remodeling eCAFs. Secreted GDF15 induced CAF activation through TGF-β receptor signaling, creating a pro-metastatic niche. Collectively, our study establishes the EGR4/GDF15 axis as a critical driver of GC metastasis, offering possible therapeutic targets for intervention.
    DOI:  https://doi.org/10.1038/s41419-025-08095-w
  6. Sci Rep. 2025 Nov 06. 15(1): 38885
      Neuroinflammation plays a central role in a wide spectrum of neurological diseases, driven generally by reactive microglia and astrocytes. Inflammatory stimulation of microglia and astrocytes leads to a metabolic shift from oxidative phosphorylation (OXPHOS) to glycolysis, which is required to support pro-inflammatory effector functions. This metabolic reprogramming is associated with impaired mitochondrial dynamics, including reduced biogenesis, increased fragmentation, and loss of membrane potential. Targeting microglia and astrocyte metabolism may offer a novel therapeutic approach for modulating neuroinflammation and restoring homeostatic immune functions. Here, we examined the potential of 2-Deoxy-D-Glucose (2DG), a glycolysis inhibitor, to attenuate neuroinflammation by restoring mitochondrial dynamics. In BV2 and primary glial cultures, low-dose 2DG reversed LPS-induced metabolic reprogramming, restoring OXPHOS, reducing mitochondrial fragmentation, and enhancing biogenesis. In vivo, it preserved spare respiratory capacity and increased complex-V activity in brain mitochondria from LPS-treated mice without affecting oxidative stress. At a mechanistic level, 2DG restored activation of AMP-activated protein kinase, a master regulator of mitochondrial dynamics. In conjunction with these metabolic effects, 2DG suppressed LPS-induced pro-inflammatory gene expression while enhancing markers associated with the resolution of inflammation and tissue repair. Critically, systemic low-dose 2DG reduced neuroinflammation and restored immune homeostasis in two LPS-induced mouse models, highlighting its therapeutic potential in neurological disorders.
    Keywords:  2-Deoxy-D-Glucose; Immunometabolism; Mitochondrial dynamics; Mitochondrial function; Neuroinflammation
    DOI:  https://doi.org/10.1038/s41598-025-22677-w
  7. Biochem Biophys Res Commun. 2025 Oct 30. pii: S0006-291X(25)01610-9. [Epub ahead of print]790 152894
      T-cell dysfunction is a critical obstacle to effective anti-cancer immunotherapy. Despite the fact that hypoxia is a common feature of the tumor microenvironment, the mechanisms driving T-cell senescence under hypoxic conditions remain largely unexplored. This study investigated the impact of hypoxic conditions on CD4+ T-cell senescence using Jurkat T cell line. Under hypoxia, T cell exhaustion was not induced; however, senescence-associated β-galactosidase (SA-β-gal) activity and lipid accumulation were increased. Tumor immune estimation resource analysis revealed that PLA2G4A that encodes cPLA2α protein is associated with reduced tumor purity and increased CD4+ T-cell infiltration in various cancers, suggesting a role in immune modulation. Hypoxia-induced activation of cPLA2α promoted lipid accumulation. Inhibition of cPLA2α under hypoxia led to a reduction in SA-β-gal activity and p27 expression, along with decreased levels of cyclin B1 and CDC2, indicative of G2/M cell cycle arrest. Moreover, cPLA2α inhibition restored CD28 expression and suppressed TGF-β1 levels, both of which are associated with T-cell senescence. Hypoxia-induced cPLA2α activation upregulated Cox-2-mediated prostaglandin E2 (PGE2) production, and exogenous PGE2 treatment further increased SA-β-gal activity. These findings illustrated that cPLA2α-driven lipid metabolism under hypoxia contributes to the T-cell senescence and may represent a therapeutic target to enhance anti-cancer immunity.
    Keywords:  Hypoxia; Immunotherapy; Senescence; T cell; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.bbrc.2025.152894