bims-stacyt Biomed News
on Metabolism and the paracrine crosstalk between cancer and the organism
Issue of 2025–08–10
five papers selected by
Cristina Muñoz Pinedo, L’Institut d’Investigació Biomèdica de Bellvitge



  1. J Clin Invest. 2025 Aug 01. pii: e191934. [Epub ahead of print]135(15):
      A central challenge in cancer therapy is the effective delivery of anticancer treatments while minimizing adverse effects on patient health. The potential dual impact of therapy is clearly illustrated in cancer-associated cachexia, a multifactorial syndrome characterized by involuntary weight loss, systemic inflammation, metabolic dysregulation, and behavioral alterations such as anorexia and apathy. While cachexia research often focuses on tumor-driven mechanisms, the literature indicates that cancer therapies themselves, particularly chemotherapies and targeted treatments, can initiate or exacerbate the biological pathways driving this syndrome. Here, we explore how therapeutic interventions intersect with the pathophysiology of cachexia, focusing on key organ systems including muscle, adipose tissue, liver, heart, and brain. We highlight examples such as therapy-induced upregulation of IL-6 and growth-differentiation factor 15, both contributing to reduced nutrient intake and a negative energy balance via brain-specific mechanisms. At the level of nutrient release and organ atrophy, chemotherapies also converge with cancer progression, for example, activating NF-κB in muscle and PKA/CREB signaling in adipose tissue. By examining how treatment timing and modality align with the natural trajectory of cancer cachexia, we underscore the importance of incorporating physiological endpoints alongside tumor-centric metrics in clinical trials. Such integrative approaches may better capture therapeutic efficacy while preserving patient well-being.
    DOI:  https://doi.org/10.1172/JCI191934
  2. J Extracell Vesicles. 2025 Aug;14(8): e70138
      Extracellular vesicles (EVs) are nano-sized, spherical, and lipid bilayered particles secreted by most types of cells. Tumor microenvironment is a complex system and highly regulated by dynamic interaction of cancer cells with stromal cells, such as immune cells and endothelial cells. Angiogenesis, a process of new capillary formation from pre-existing vasculatures, plays a critical role in tumorigenesis and cancer progression by providing oxygen and nutrients to proliferating cancer cells. Since EVs have emerged as important mediators of intercellular communication in pathophysiological circumstances, several studies have reported pro-angiogenic activities of EVs derived from in vitro cultured cancer cells and other cells. However, the angiogenic role of EVs directly isolated from in vivo tumor tissues has not been investigated. Here, we isolated EVs directly from mouse primary tumor tissues with high purity and investigated the angiogenic potential of in vivo tumor tissue-derived EVs (tEVs). The purified tumor tEVs showed EV-like features with nano-sized, spherical, and lipid bilayered structures, and enriched with EV marker proteins such as tetraspanins, while being de-enriched with proteins from Golgi apparatus and nucleus. Interestingly, tumor tEVs promoted extensive neovascularization and numerous macrophage infiltrations in vivo Matrigel plug assay. In addition, the angiogenic properties of tumor tEVs were mediated by the infiltrated macrophages through producing vascular endothelial growth factor (VEGF), a pro-angiogenic molecule. These results suggested that tumor tEVs have potent in vivo angiogenic activity by directly promoting macrophage recruitment and activating infiltrated macrophages to produce VEGF. Our study provides the first direct evidence for a key role of infiltrated macrophage VEGF production in tumor tEV-mediated neovascularization in the tumor microenvironment.
    Keywords:  VEGF; angiogenesis; extracellular vesicles; macrophages; tumor tissue‐derived extracellular vesicles
    DOI:  https://doi.org/10.1002/jev2.70138
  3. bioRxiv. 2025 Jul 26. pii: 2025.07.22.666143. [Epub ahead of print]
      The JAK-STAT3 signaling pathway is a key driver of colorectal cancer (CRC) progression. While STAT3 is canonically activated by cytokines such as IL-6, this activation is typically transient due to negative feedback mechanisms. In CRC, however, STAT3 is aberrantly and persistently activated, promoting tumor cell proliferation and survival. Here, we demonstrate that glucose sustains STAT3 activation independent of cytokine availability. By manipulating glucose metabolism, we show that both glucose and its downstream metabolite, GlcNAc, are essential for maintaining STAT3 activation. Moreover, cells with high basal STAT3 activity produce glucose-dependent glycosylated proteins that can activate STAT3 in neighboring cells via paracrine signaling. Proteomic analysis identified multiple candidate proteins involved in this process; however, no single protein was sufficient to fully activate STAT3, suggesting that a combination of glycosylated proteins likely acts synergistically. In vivo, inhibition of glycolysis reduces STAT3 activation in tumors, and genetic deletion of STAT3 in subcutaneous tumor models significantly decreases tumor growth. Together, these findings uncover a novel mechanism by which glucose metabolism supports sustained STAT3 activation in CRC, highlighting a potential metabolic vulnerability for therapeutic targeting.
    DOI:  https://doi.org/10.1101/2025.07.22.666143
  4. Cell Metab. 2025 Jul 30. pii: S1550-4131(25)00333-X. [Epub ahead of print]
      Glucose is essential for T cell proliferation and function, yet its specific metabolic roles in vivo remain poorly defined. Here, we identify glycosphingolipid (GSL) biosynthesis as a key pathway fueled by glucose that enables CD8+ T cell expansion and cytotoxic function in vivo. Using 13C-based stable isotope tracing, we demonstrate that CD8+ effector T cells use glucose to synthesize uridine diphosphate-glucose (UDP-Glc), a precursor for glycogen, glycan, and GSL biosynthesis. Inhibiting GSL production by targeting the enzymes UDP-Glc pyrophosphorylase 2 (UGP2), UDP-Gal-4-epimerase (GALE), or UDP-Glc ceramide glucosyltransferase (UGCG) impairs CD8+ T cell expansion upon pathogen challenge. Mechanistically, we show that glucose-dependent GSL biosynthesis is required for plasma membrane lipid raft integrity and optimal T cell receptor (TCR) signaling. Moreover, UGCG-deficient CD8+ T cells display reduced granzyme expression, cytolytic activity, and tumor control in vivo. Together, our data establish GSL biosynthesis as a critical metabolic fate of glucose-beyond energy production-that is required for CD8+ T cell responses in vivo.
    Keywords:  CD8(+) T cells; UGCG; cytotoxic function; glucose; glycosphingolipids; immunometabolism; lipid rafts; lipidomics; metabolomics; nucleotide sugar metabolism
    DOI:  https://doi.org/10.1016/j.cmet.2025.07.006
  5. Cell. 2025 Jul 29. pii: S0092-8674(25)00805-0. [Epub ahead of print]
      Cancer-associated cachexia (CAC) is a multifactorial and currently incurable syndrome responsible for nearly one-third of cancer-related deaths. It contributes to therapy resistance and increases mortality among affected patients. In this study, we show that cancer-induced systemic inflammation alters vagal tone in CAC mouse models. This vagal dysregulation disrupts the brain-liver vagal axis, leading to a reprogramming of hepatic protein metabolism through the depletion of HNF4α, a key transcriptional regulator of liver function. The loss of HNF4α disrupts hepatic metabolism and promotes systemic inflammation, resulting in cachectic phenotypes. Interventions targeting the right cervical vagus nerve surgically, chemically, electrically, or through a non-invasive transcutaneous device attenuate CAC progression, alleviate its clinical manifestations, and synergize with chemotherapy to improve overall health and survival in mice.
    Keywords:  HNF4α; cancer-associated cachexia; liver; metabolism; neuromodulation; vagus nerve
    DOI:  https://doi.org/10.1016/j.cell.2025.07.016