bims-cagime Biomed News
on Cancer, aging and metabolism
Issue of 2025–06–01
29 papers selected by
Kıvanç Görgülü, Technical University of Munich



  1. JCI Insight. 2025 May 27. pii: e169105. [Epub ahead of print]
      Resistance to chemotherapy of pancreatic ductal adenocarcinoma (PDAC) is largely driven by intratumoral heterogeneity (ITH) due to tumor cell plasticity and clonal diversity. In order to develop novel strategies to overcome this defined mechanism of resistance, tools to monitor and quantify ITH in a rapid and scalable fashion are needed urgently. Here, we employed label-free digital holographic microscopy (DHM) to characterize ITH in PDAC. We established a robust experimental and machine learning analysis pipeline to perform single cell phenotyping based on DHM-derived phase images of PDAC cells in suspension. Importantly, we are able to detect dynamic changes in tumor cell differentiation and heterogeneity of distinct PDAC subtypes upon induction of epithelial-to-mesenchymal transition and under treatment-imposed pressure in murine and patient-derived model systems. This platform allows us to assess phenotypic ITH in PDAC on a single cell level in real-time. Implementing this technology into the clinical workflow has the potential to fundamentally increase our understanding of tumor heterogeneity during evolution and treatment response.
    Keywords:  Cancer; Gastroenterology; Oncology
    DOI:  https://doi.org/10.1172/jci.insight.169105
  2. EMBO Rep. 2025 May 28.
      Phase-separated p62 bodies activate NRF2, a key transcription factor for antioxidant response, by sequestering KEAP1, which targets NRF2 for degradation. Although p62 bodies containing KEAP1 are degraded by autophagy, they accumulate in various liver disorders. Their precise disease role remains unclear. We show that excessive KEAP1 retention in p62 bodies and NRF2 activation are major causes of liver damage when autophagy is impaired. In mice with weakened or blocked p62-KEAP1 interactions, KEAP1 retention and NRF2 activation under autophagy-deficient conditions were suppressed. Transcriptome and proteome analyses reveal that p62 mutants unable to bind KEAP1 normalize the expression of NRF2 targets induced by defective autophagy. Autophagy deficiency causes organelle accumulation, especially of the ER, regardless of p62 mutation. Liver damage and hepatomegaly resulting from autophagy suppression markedly improved in mice carrying p62 mutants, particularly those with blocked KEAP1 binding. These findings highlight excessive KEAP1 retention in p62 bodies and defective organelle turnover as key drivers of liver pathology, underscoring the significance of phase separation in vivo.
    Keywords:  KEAP1; Liquid–Liquid Phase Separation; NRF2; Stress Response; p62
    DOI:  https://doi.org/10.1038/s44319-025-00483-9
  3. Nat Commun. 2025 May 26. 16(1): 4866
      Basal pancreatic ductal adenocarcinoma (PDAC) has the worst overall survival and is the only subtype that serves as an independent poor prognostic factor. We identify elevated levels of LIN28B and its downstream target, HMGA2, in basal PDAC. Notably, LIN28B significantly accelerates KRAS-driven PDAC progression in a mouse model. Here, we show that HMGA2 promotes basal PDAC pathogenesis by enhancing mRNA translation downstream of LIN28B. Mechanistically, HMGA2 suppresses leucine carboxyl methyltransferase 1 (LCMT1) at the chromatin level, reducing PP2A methylation and activity. This leads to increased phosphorylation of S6K and eIF4B, boosting mRNA translation. Additionally, HMGA2 downregulates B56α (PPP2R5A), disrupting functional PP2A holoenzyme assembly and further sustaining phosphorylated S6K levels. Impaired PP2A function mimics HMGA2's effects, reinforcing increased mRNA translation and basal lineage features. This work uncovers a critical link between the LIN28B/HMGA2 axis, protein synthesis, and PDAC lineage specificity via LCMT1-mediated PP2A methylation and B56α-PP2A disruption.
    DOI:  https://doi.org/10.1038/s41467-025-60129-1
  4. Nat Cell Biol. 2025 May 26.
      The pathological role and mechanism of psychological stress in cancer progression are little known. Here we show in a mouse model that psychological stress drives pancreatic ductal adenocarcinoma (PDAC) progression by stimulating tumour nerve innervation. We demonstrate that nociception and other stressors activate sympathetic nerves to release noradrenaline, downregulating RNA demethylase alkB homologue 5 (Alkbh5) in tumour cells. Alkbh5 deficiency in these cancer cells causes aberrant N6-methyladenosine (m6A) modification of RNAs, which are packed into extracellular vesicles and delivered to nerves in the tumour microenvironment, enhancing hyperinnervation and PDAC progression. ALKBH5 levels are inversely correlated with tumour innervation and survival time in patients with PDAC. Animal experiments identify a natural flavonoid, fisetin, that prevents neurons from taking in extracellular vesicles containing m6A-modified RNAs, thus suppressing the excessive innervation and progression of PDAC tumours. Our study sheds light on a molecular mechanism by which crosstalk between the neuroendocrine system and cancer cells links psychological stress and cancer progression and raises a potential strategy for PDAC therapy.
    DOI:  https://doi.org/10.1038/s41556-025-01667-0
  5. Nat Cancer. 2025 May 26.
      Cachexia is the wasting of skeletal muscle in cancer and is a major complication that impacts a person's quality of life. We hypothesized that cachexia is mediated by dysfunction of the vascular system, which is essential for maintaining perfusion and tempering inappropriate immune responses. Using transparent tissue topography, we discovered that loss of muscle vascular density precedes muscle wasting in multiple complementary tumor models, including pancreatic adenocarcinoma, colon carcinoma, lung adenocarcinoma and melanoma models. We also observed that persons suffering from cancer cachexia exhibit substantial loss of muscle vascular density. As tumors progress, increased circulating activin A remotely suppresses the expression of peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α) in the muscle endothelium, thus inducing vascular leakage. Restoring endothelial PGC1α activity preserved vascular density and muscle mass in tumor-bearing mice. Our study suggests that restoring muscle endothelial function could be a valuable therapeutic approach for cancer cachexia.
    DOI:  https://doi.org/10.1038/s43018-025-00975-6
  6. Trends Cell Biol. 2025 May 26. pii: S0962-8924(25)00109-6. [Epub ahead of print]
      Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a dire prognosis. Standard-of-care chemotherapy regimens offer marginal survival benefit and carry risk of severe toxicity, while immunotherapy approaches have uniformly failed in clinical trials. Extensive desmoplasia in the PDAC tumor microenvironment (TME) disrupts blood flow to and from the tumor, thereby creating a nutrient-depleted, hypoxic, and acidic milieu that suppresses the function of antitumor immune cells and imparts chemotherapy resistance. Additionally, recent seminal studies have demonstrated crucial roles for metabolic crosstalk - the exchange of metabolites between PDAC cells and stromal cell populations in the TME - in establishing and maintaining core malignant behaviors of PDAC: tumor growth, metastasis, immune evasion, and therapy resistance. In this review, we provide a conceptual overview of metabolic crosstalk and how it evolves under various selection pressures in the TME, analyze the landscape of proposed tumorigenic metabolic crosstalk pathways, and highlight potentially druggable nodes.
    Keywords:  hypoxia; metabolic competition; pancreatic cancer; stromal diversity; therapy resistance; tumor–stromal metabolic crosstalk
    DOI:  https://doi.org/10.1016/j.tcb.2025.04.007
  7. Mol Biol Cell. 2025 May 28. mbcE25050208
      Michael P. Sheetz (1946-2025) advanced the field of mechanobiology through his creative experiments, new methodologies and keen insights. His research touched many fields of cell biology including membrane biophysics, motor proteins, the cytoskeleton, cell migration, and cellular senescence. In addition to his research, Sheetz was a leader who built vibrant academic departments and institutes and advanced the careers of many trainees.
    DOI:  https://doi.org/10.1091/mbc.E25-05-0208
  8. Nat Commun. 2025 May 30. 16(1): 5055
      Septins are cytoskeletal proteins that form filaments and higher-order structures, and remodel membranes in a variety of processes. Structural and cell biological studies provided atomic- and micro-scale details, but the understanding of septin assembly at the mesoscale is limited. Here, we used high-speed atomic force microscopy (HS-AFM) to analyze yeast septin assembly on yeast supported lipid bilayers (SLBs). We found the coexistence of three lipid phases in yeast membranes, where septin polymerized selectively on the liquid-disordered phase. Septin filaments adhered to membranes with a conserved face; and paired filaments, previously reported in less native environments, were not observed. Additionally, septin filaments exhibited lateral and longitudinal alignment. We used HS-AFM force-sweep experiments to disrupt septin structures and observe organizational recovery through self-templating. Finally, septin filaments stacked, where higher layer filament alignment was templated by the layer below. Thus, septins encode their 3D-structural organization, likely tunable by the membrane and bulk environment.
    DOI:  https://doi.org/10.1038/s41467-025-60344-w
  9. Cancers (Basel). 2025 May 15. pii: 1676. [Epub ahead of print]17(10):
      Pancreatic cancer is characterized by late diagnosis, therapy resistance, and poor prognosis, necessitating the exploration of early carcinogenesis and prevention methods. Preclinical mouse models have evolved from cell line-based to human tumor tissue- or organoid-derived xenografts, now to humanized mouse models and genetically engineered mouse models (GEMMs). GEMMs, primarily driven by oncogenic Kras mutations and tumor suppressor gene alterations, offer a realistic platform for investigating pancreatic cancer initiation, progression, and metastasis. The incorporation of inducible somatic mutations and CRISPR-Cas9 screening methods has expanded their utility. To better recapitulate tumor initiation triggered by inflammatory cues, common pancreatic risk factors are being integrated into model designs. This approach aims to decipher the role of environmental factors as secondary or parallel triggers of tumor initiation alongside oncogenic burdens. Emerging models exploring pancreatitis, obesity, diabetes, and other risk factors offer significant translational potential. This review describes current mouse models for studying pancreatic carcinogenesis, their combination with inflammatory factors, and their utility in evaluating pathogenesis, providing guidance for selecting the most suitable models for pancreatic cancer research.
    Keywords:  carcinogenesis; diabetes; genetically engineered mouse models; obesity; pancreatic cancer; pancreatitis; risk factors
    DOI:  https://doi.org/10.3390/cancers17101676
  10. Aging Cell. 2025 May 28. e70114
      Excess cellular senescence contributes to age-related increases in frailty and reductions in skeletal muscle strength. In the present study, we determined the efficacy of oral intermittent treatment (1 week on-2 weeks off-1 week on) with the natural flavonoid senolytic fisetin to improve frailty and grip strength in old mice. Further, the effects of fisetin on physical function were evaluated in young mice. We performed bulk RNA sequencing of quadricep skeletal muscle to determine the cell senescence-related signaling pathways modulated by fisetin. We also assessed the relative effects of fisetin on frailty and grip strength with aging in comparison with two other well-established approaches for the removal of senescent cells: (1) genetic-based clearance of excess senescent cells in old p16-3MR mice, a model that allows for clearance of p16-positive (p16+) senescent cells, and (2) oral intermittent treatment with the synthetic pharmacological senolytic ABT-263 in old mice. We found that fisetin mitigated the adverse changes in frailty and grip strength with aging. Fisetin had no effects in young mice. The improvements in frailty and grip strength in old mice were accompanied by favorable modulation of the skeletal muscle transcriptome, including lower abundance of cellular senescence-related genes (e.g., Cdkn1a and Ddit4). Improvements in frailty and grip strength with fisetin were comparable to those observed with genetic-based clearance of excess p16+ senescent cells and treatment with ABT-263. Taken together, our findings provide proof-of-concept support for fisetin as a senolytic strategy to improve physical function with aging.
    Keywords:  flavonoid; motor function; natural senolytic; senescence associated secretory phenotype; skeletal muscle senescence; transcriptome
    DOI:  https://doi.org/10.1111/acel.70114
  11. Autophagy. 2025 May 25.
      DDX11 is a DNA helicase involved in critical cellular functions, including DNA replication/repair/recombination as well as sister chromatid cohesion establishment. Bi-allelic mutations of DDX11 lead to Warsaw breakage syndrome (WABS), a rare genome instability disorder marked by significant prenatal and postnatal growth restriction, microcephaly, intellectual disability, and sensorineural hearing loss. The molecular mechanisms underlying WABS remain largely unclear. In this study, we uncover a novel role of DDX11 in regulating the macroautophagic/autophagic pathway. Specifically, we demonstrate that knockout of DDX11 in RPE-1 cells hinders the progression of autophagy. DDX11 depletion significantly reduces the conversion of MAP1LC3/LC3 (microtubule associated protein 1 light chain 3), suggesting a defect in autophagosome biogenesis. This is supported by imaging analysis with a LC3 reporter fused in tandem with the red and green fluorescent proteins (mRFP-GFP-LC3), which reveals fewer autophagosomes and autolysosomes in DDX11-knockout cells. Moreover, the defect in autophagosome biogenesis, observed in DDX11-depleted cells, is linked to an upstream impairment of the ATG16L1-precursor trafficking and maturation, a step critical to achieve the LC3 lipidation. Consistent with this, DDX11-lacking cells exhibit a diminished capacity to clear aggregates of a mutant HTT (huntingtin) N-terminal fragment fused to the green fluorescent protein (HTTQ74-GFP), an autophagy substrate. Finally, we demonstrate the occurrence of a functional interplay between DDX11 and SQSTM1, an autophagy cargo receptor protein, in supporting LC3 modification during autophagosome biogenesis. Our findings highlight a novel unprecedented function of DDX11 in the autophagy process with important implications for our understanding of WABS etiology.
    Keywords:  Autophagosome biogenesis; DDX11; DNA helicase; LC3; SQSTM1/p62; Warsaw breakage syndrome; neurodevelopmental disorders
    DOI:  https://doi.org/10.1080/15548627.2025.2507617
  12. Elife. 2025 May 27. pii: RP93180. [Epub ahead of print]12
      The ability of a single protein to trigger different functions is an assumed key feature of cell signaling, yet there are very few examples demonstrating it. Here, using an optogenetic tool to control membrane localization of RhoA nucleotide exchange factors (GEFs), we present a case where the same protein can trigger both protrusion and retraction when recruited to the plasma membrane, polarizing the cell in two opposite directions. We show that the basal concentration of the GEF prior to activation predicts the resulting phenotype. A low concentration leads to retraction, whereas a high concentration triggers protrusion. This unexpected protruding behavior arises from the simultaneous activation of Cdc42 by the GEF and sequestration of active RhoA by the GEF PH domain at high concentrations. We propose a minimal model that recapitulates the phenotypic switch, and we use its predictions to control the two phenotypes within selected cells by adjusting the frequency of light pulses. Our work exemplifies a unique case of control of antagonist phenotypes by a single protein that switches its function based on its concentration or dynamics of activity. It raises numerous open questions about the link between signaling protein and function, particularly in contexts where proteins are highly overexpressed, as often observed in cancer.
    Keywords:  cell biology; cell migration; human; optogenetics; signaling
    DOI:  https://doi.org/10.7554/eLife.93180
  13. Mol Cell. 2025 May 21. pii: S1097-2765(25)00412-5. [Epub ahead of print]
      Coordination of adaptive metabolism through signaling networks is essential for cellular bioenergetics and homeostasis. Phosphorylation of metabolic enzymes provides a rapid, efficient, and dynamic mechanism to regulate metabolic networks. Our structural analysis stratified phosphosites on metabolic enzymes based on proximity to functional and dimerization domains. Most phosphosites occur on oxidoreductases and are enriched near substrate, cofactor, active sites, or dimer interfaces. Despite low stoichiometry, phosphotyrosine (pY) is overrepresented in functional domains. Using high-fat diet (HFD)-induced obesity in C57BL/6J mice and multiomics, we measured HFD-induced sex-specific dysregulation of pY and metabolites, which was reversible with the antioxidant butylated hydroxyanisole (BHA). Computational modeling revealed predictive pY sites for HFD- or BHA-induced metabolite changes. We characterized functional roles for predictive pY sites on glutathione S-transferase pi 1 (GSTP1), isocitrate dehydrogenase 1 (IDH1), and uridine monophosphate synthase (UMPS) using CRISPR interference (CRISPRi) rescue and stable isotope tracing. Our findings reveal mechanisms whereby cellular signaling fine-tunes enzyme activity and metabolism.
    Keywords:  GSTP1; IDH1; UMPS; cell signaling; computational modelling; metabolism; metabolomics; obesity; oxidative stress response; phosphoproteomics
    DOI:  https://doi.org/10.1016/j.molcel.2025.05.007
  14. Redox Biol. 2025 May 21. pii: S2213-2317(25)00207-1. [Epub ahead of print]84 103694
      Pharmacological ascorbate (P-AscH, high-dose, intravenous, vitamin C), is a pro-drug that generates hydrogen peroxide (H2O2) and is being investigated as a neoadjuvant treatment for pancreatic adenocarcinoma (PDAC). In a randomized, phase II clinical trial, P-AscH demonstrated encouraging results in terms of efficacy and safety. However, some patients do not respond to P-AscH suggesting that resistance occurs in a subset of patients. The aims of this study were two-fold: first to characterize PDAC cells resistant to P-AscH, and second, determine if these alterations enhance metastatic potential. Resistance to P-AscH increased the ability to detoxify H2O2, altered redox metabolism and cell cycle regulation, however mechanisms to P-AscH resistance were different in the cell lines studied. Transcriptomic analysis demonstrated a significant enrichment of the epithelial-to-mesenchymal gene expression pattern in the cell lines studied, suggesting that upregulation of metastatic phenotypes occur during acquisition of resistance to P-AscH. Cells resistant to P-AscH demonstrated increased invasive potential, more aggressive tumor colonization, and higher abundance of circulating tumor cells in vivo. Our data support that resistance to oxidative stress enhances metastatic disease and indicates a potential route for PDAC to tolerate high levels of P-AscH and may explain why some patients do not respond to this treatment regimen.
    Keywords:  Ascorbate; Metastasis; Oxidative stress and metabolism; Pancreatic cancer
    DOI:  https://doi.org/10.1016/j.redox.2025.103694
  15. Nat Methods. 2025 May 28.
      Recent advances in imaging and computation have enabled analysis of large three-dimensional (3D) biological datasets, revealing spatial composition, morphology, cellular interactions and rare events. However, the accuracy of these analyses is limited by image quality, which can be compromised by missing data, tissue damage or low resolution due to mechanical, temporal or financial constraints. Here, we introduce InterpolAI, a method for interpolation of synthetic images between pairs of authentic images in a stack of images, by leveraging frame interpolation for large image motion, an optical flow-based artificial intelligence (AI) model. InterpolAI outperforms both linear interpolation and state-of-the-art optical flow-based method XVFI, preserving microanatomical features and cell counts, and image contrast, variance and luminance. InterpolAI repairs tissue damages and reduces stitching artifacts. We validated InterpolAI across multiple imaging modalities, species, staining techniques and pixel resolutions. This work demonstrates the potential of AI in improving the resolution, throughput and quality of image datasets to enable improved 3D imaging.
    DOI:  https://doi.org/10.1038/s41592-025-02712-4
  16. ACS Appl Bio Mater. 2025 May 24.
      Multidrug resistance (MDR) presents a substantial challenge to the therapeutic efficacy of cancer chemotherapy. A common trait of drug-resistant cells is decreased cell membrane permeability, hindering the uptake of therapeutic agents. Additionally, these cells frequently overexpress drug efflux pumps that actively expel the drugs, leading to reduced intracellular accumulation. In this study, we introduce a high spatially resolved, domain-specific, mild heating strategy to counteract drug resistance using DNA nanodevices. This strategy aims to manipulate the membrane heterogeneity by increasing cell membrane permeability and decreasing the expression of drug efflux pumps. The DNA nanodevices (termed DNA nanoheaters) with specific domain affinity anchor distinct cell membrane domains (raft/nonraft) and elevate the local lipid environmental temperature upon near-infrared (NIR) laser exposure. This elevation in local lipid temperature modifies key biophysical membrane features of Doxorubicin-resistant tumor cells, resulting in a two-order magnitude decrease in IC50. Notably, our approach diverges from conventional methods that depend on the delivery of pharmacological reversal agents. Instead, we emphasize modulating the membrane properties of drug-resistant cells through mild physical stimuli, offering a potential reduction in systemic toxicity associated with chemotherapy.
    Keywords:  DNA nanotechnology; cell membrane; multidrug resistance; photothermal; tumor cell
    DOI:  https://doi.org/10.1021/acsabm.5c00605
  17. Exerc Sport Mov. 2024 ;pii: e00027. [Epub ahead of print]2(4):
      Although improvements in prevention and screening have curbed the incidence of some cancers, the global burden of cancer is substantial and continues to grow. The sustained high prevalence of many cancers reveals the need for additional strategies to reduce occurrence. Observational studies have linked physical inactivity to the risk of 13 different cancers. Indeed, physical activity can reduce the occurrence of several cancers by more than 20%, whereas sedentary behavior can increase cancer risk. Thus, physical activity presents a viable lifestyle intervention to reduce the global burden of cancer, and current research efforts are focused on establishing the effective physical activity mode and intensity for cancer prevention. Preclinical cancer studies have provided insight into the mechanisms mediating these effects. There is growing evidence that physical activity can 1) reduce the risk of obesity and, by extension, metabolic dysregulation; 2) improve immune surveillance and reduce inflammation; 3) enrich the colonic environment by favoring beneficial microbes and reducing transit time; and 4) regulate sex hormones. This graphical review describes the current state of knowledge on the benefits of physical activity for cancer prevention and associated plausible mechanisms.
    Keywords:  colonic environment; exercise; immune function; inflammation; metabolic dysregulation; sex hormones
    DOI:  https://doi.org/10.1249/esm.0000000000000027
  18. Cell Syst. 2025 May 22. pii: S2405-4712(25)00129-2. [Epub ahead of print] 101296
      The rapid development of multiplexed imaging technologies has enabled the spatial cartography of various healthy and tumor tissues. However, adequate statistical models are still lacking to compare tissue compositions across sample groups. Here, we developed two statistical models that accurately describe the distributions of cell counts in an imaging mass cytometry dataset comprising tissues from a lymph node, COVID-19-affected lung samples, and Hashimoto disease. The parameters of these distributions are directly linked to the field of view size and to cellular properties, including density and spatial aggregation. We identified statistical tests that improved statistical power for differential abundance testing compared with the commonly used rank-based test. Our analysis revealed spatial aggregation as the main determinant of statistical power and that high numbers of fields of view are required when cells are highly aggregated. To overcome this challenge, we propose a stratified sampling strategy that considerably reduces the required sample size.
    Keywords:  experimental design; multiplexed imaging; statistical modeling
    DOI:  https://doi.org/10.1016/j.cels.2025.101296
  19. Cancer Res. 2025 May 29.
      The 5-year overall survival rate for pancreatic cancer remains ~13%, underscoring the urgent need for improved treatment strategies. TGFβ is a promising target due to its significant involvement in the desmoplasia, immune suppression, and chemoresistance characteristic of pancreatic cancer. Over 300 clinical trials targeting TGFβ have been conducted in unselected patient cohorts; however, none of the therapies have gained FDA approval. Nevertheless, TGFβ blockade may hold promise for a subset of cancers with non-functional TGFβ signaling. Greater than 25% of pancreatic cancers carry mutations in SMAD4, a key component of canonical TGFβ signaling. In this study, we investigated the potential for stratifying patients based on SMAD4 mutational status to identify tumors susceptible to TGFβ inhibition. Analysis of SMAD4 expression in human pancreatic tumors revealed that SMAD4 mutation or loss is associated with worse disease-free survival. Intriguingly, intratumoral SMAD4 expression displayed heterogeneity among human pancreatic cancer samples. SMAD4 deficient genetically engineered mouse models and orthotopic SMAD4 knockout tumor models exhibited reduced survival, increased metastasis, and alterations in the tumor microenvironment compared to SMAD4 wildtype controls, consistent with gene and protein expression changes in the absence of functional SMAD4. Importantly, treating mice bearing SMAD4 deficient tumors with a blocking TGFβ antibody reduced tumor weight and improved survival. These findings suggest that genomic stratification by TGFβ axis alterations, such as SMAD4 mutations, may be a promising approach to identifying patients likely to benefit from a TGFβ inhibitor.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-24-1970
  20. J Immunother Cancer. 2025 May 24. pii: e011404. [Epub ahead of print]13(5):
       BACKGROUND: Immune checkpoint blockers (ICBs) have revolutionized cancer therapy, yet they remain largely ineffective in treating pancreatic ductal adenocarcinoma (PDAC). Moreover, ICBs can cause severe immune-related adverse events (irAEs), including fatal cardiac toxicity. Finally, obesity is a risk factor in PDAC that may differentially modulate ICB efficacy in a malignancy-dependent manner.
    METHODS: We investigated the mechanisms underlying irAEs induced by dual ICB therapy and sought to identify strategies to mitigate them while improving ICB efficacy in the obese setting. To this end, we used a clinically relevant mouse model that integrated key features of human PDAC: (1) high-fat diet-induced obesity, (2) an orthotopic PDAC, and (3) a therapeutic regimen combining chemotherapy (FOLFIRINOX) with ICBs (α-programmed cell death protein-1 + α-cytotoxic T-lymphocyte associated protein-4 antibodies).
    RESULTS: Obese mice developed cardiac irAEs and had elevated serum interleukin (IL)-1β levels after chemoimmunotherapy. IL-1β blockade not only prevented myocarditis and reduced cardiac fibrosis but also enhanced the antitumor efficacy of the combination of chemotherapy plus dual ICB therapy and significantly improved the overall survival of PDAC-bearing obese mice.
    CONCLUSIONS: Our findings provide the rationale and compelling data to test a Food and Drug Administration-approved anti-IL-1β antibody in combination with chemotherapy and dual ICB therapy in patients with pancreatic cancer with obesity.
    Keywords:  Cardiotoxicity; Immune Checkpoint Inhibitor; Immune related adverse event - irAE
    DOI:  https://doi.org/10.1136/jitc-2024-011404
  21. Phys Rev E. 2025 Apr;111(4-1): 044405
      The self-organization of proteins into enriched compartments and the formation of complex patterns are crucial processes for life on the cellular level. Liquid-liquid phase separation is one mechanism for forming such enriched compartments. When phase-separating proteins are membrane-bound and locally disturb it, the mechanical response of the membrane mediates interactions between these proteins. How these membrane-mediated interactions influence the steady state of the protein density distribution is thus an important question to investigate in order to understand the rich diversity of protein and membrane-shape patterns present at the cellular level. This work starts with a widely used model for membrane-bound phase-separating proteins. We numerically solve our system to map out its phase space and perform a careful, systematic expansion of the model equations to characterize the phase transitions through linear stability analysis and free energy arguments. We observe that the membrane-mediated interactions, due to their long-range nature, are capable of qualitatively altering the equilibrium state of the proteins. This leads to arrested coarsening and length-scale selection instead of simple demixing and complete coarsening. In this study, we unambiguously show that long-range membrane-mediated interactions lead to pattern formation in a system that otherwise would not do so. This work provides a basis for further systematic study of membrane-bound pattern-forming systems.
    DOI:  https://doi.org/10.1103/PhysRevE.111.044405
  22. Biology (Basel). 2025 Apr 26. pii: 477. [Epub ahead of print]14(5):
      KRAS mutations are major drivers of human cancers, yet how distinct mutations rewire protein interactions and metabolic pathways to promote tumorigenesis remains poorly understood. To address this, we systematically mapped the protein interaction networks of wild-type KRAS and three high-frequency oncogenic mutants (G12C, G12D, and G12V) using TurboID proximity labeling coupled with quantitative proteomics. Bioinformatic analysis revealed mutant-specific binding partners and metabolic pathway alterations, including significant enrichment in insulin signaling, reactive oxygen species regulation, and glucose/lipid metabolism. These changes collectively drive tumor proliferation and immune evasion. Comparative analysis identified shared interactome shifts across all mutants: reduced binding to LZTR1, an adaptor for KRAS degradation, and enhanced recruitment of LAMTOR1, a regulator of mTORC1-mediated growth signaling. Our multi-dimensional profiling establishes the first comprehensive map of KRAS-mutant interactomes and links specific mutations to metabolic reprogramming. These findings provide mechanistic insights into KRAS-driven malignancy and highlight LZTR1 and LAMTOR1 as potential therapeutic targets. The study further lays a foundation for developing mutation-specific strategies to counteract KRAS oncogenic signaling.
    Keywords:  KRAS G12 mutants; KRAS interactome; TurboID proximity-labeling; metabolic reprogramming; quantitative proteomics
    DOI:  https://doi.org/10.3390/biology14050477
  23. Nature. 2025 May 28.
      The multicellular coordination that underlies tissue homeostasis and disease progression is of fundamental interest1-5. However, how diverse cell types are organized within tissue niches for cohesive functioning remains largely unknown. Here we systematically characterized cross-tissue coordinated cellular modules in healthy tissues, uncovering their spatiotemporal dynamics and phenotypic associations, and examined their rewiring in cancer. We first compiled a comprehensive single-cell transcriptomic atlas from 35 human tissues, revealing substantial inter-tissue variability in cellular composition. By leveraging covariance in cellular abundance, we identified 12 cellular modules with distinct cellular compositions, tissue prevalences and spatial organizations, and demonstrated coordinated intercellular communication within cellular modules using in situ spatial and in vivo perturbation data. Among them, two immune cellular modules in the spleen showed contrasting chronological dynamics with ageing. Analysis of multicellular changes in the breast revealed a menopausal trajectory associated with fibroblast dynamics. Furthermore, interrogation across cancer types uncovered simultaneous rewiring of two types of multicellular ecosystem during tumour progression, including the loss of tissue-specific healthy organization and the emergence of a convergent cancerous ecosystem. These findings reveal fundamental organizing principles of multicellular ecosystems in health and cancer, laying a foundation for further investigations into tissue-level functional coordination across diverse contexts.
    DOI:  https://doi.org/10.1038/s41586-025-09053-4
  24. PLoS Biol. 2025 May 27. 23(5): e3003196
      The study of ubiquitous circadian rhythms in human physiology requires regular measurements across time. Repeated sampling of the different internal tissues that house circadian clocks is both practically and ethically infeasible. Here, we present a novel unsupervised machine learning approach (COFE) that can use single high-throughput omics samples (without time labels) from individuals to reconstruct circadian rhythms across cohorts. COFE can simultaneously assign time labels to samples and identify rhythmic data features used for temporal reconstruction, while also detecting invalid orderings. With COFE, we discovered widespread de novo circadian gene expression rhythms in 11 different human adenocarcinomas using data from The Cancer Genome Atlas (TCGA) database. The arrangement of peak times of core clock gene expression was conserved across cancers and resembled a healthy functional clock except for the mistiming of a few key genes. Moreover, rhythms in the transcriptome were strongly associated with the cancer-relevant proteome. The rhythmic genes and proteins common to all cancers were involved in metabolism and the cell cycle. Although these rhythms were synchronized with the cell cycle in many cancers, they were uncoupled with clocks in healthy matched tissue. The targets of most of FDA-approved and potential anti-cancer drugs were rhythmic in tumor tissue with different amplitudes and peak times. These findings emphasize the utility of considering "time" in cancer therapy, and suggest a focus on clocks in healthy tissue rather than free-running clocks in cancer tissue. Our approach thus creates new opportunities to repurpose data without time labels to study circadian rhythms.
    DOI:  https://doi.org/10.1371/journal.pbio.3003196
  25. Cancer Cell. 2025 May 27. pii: S1535-6108(25)00212-0. [Epub ahead of print]
      Most cancer proteomics studies to date have focused on a single cancer type. We report The Pan-Cancer Proteome Atlas (TPCPA) based on data-independent acquisition mass spectrometry, to better understand cancer biology and identify therapeutic targets and biomarkers. TPCPA includes 9,670 proteins derived from 999 primary tumors representing 22 cancer types. We describe pan-cancer and cancer type-enriched proteins with extensive external annotation, prioritizing candidate drug targets and biomarkers. Relevant for proteolysis-targeting chimeras, we identify E3-ubiquitin ligases highly expressed in specific tumor types, including HERC5 (esophageal cancer) and RNF5 (liver cancer). Co-expression analysis reveals 13 modules, including unexpected hub proteins as potential drug targets (e.g., GFPT1, LRPPRC, PINK1, DOCK2, and PTPN6). Analysis of 195 colorectal cancers identifies protein markers for RNA-based consensus molecular subtypes (CMSs) and two immune subtypes with prognostic value. We report a cancer type classifier for identification of cancers of unknown primary origin. All TPCPA data can be queried in a dedicated web resource.
    Keywords:  bioinformatics; biomarker/ target; colorectal cancer subtypes; mass spectrometry; multi-cancer (sub)type classification; pan-cancer; proteome
    DOI:  https://doi.org/10.1016/j.ccell.2025.05.003
  26. J Biochem. 2025 May 29. pii: mvaf027. [Epub ahead of print]
      Autophagy suppresses tumorigenesis in normal cells, but in established tumors, it can promote tumor progression, particularly by enhancing resistance to stress. However, the mechanism underlying this tumor-promoting function remains unclear. To investigate this, we adopted an interdisciplinary approach combining database analysis with experimental validation. Specifically, by classifying the autophagy-related genes using AutoML analysis on their expression patterns in the COSMIC database, we identified an autophagy subnetwork that correlated with the PLK1-RAD9A axis, a pathway we had previously linked to genotoxic resistance. Cell-based experiments confirmed that autophagy enhanced PLK1 expression at both the transcriptional and translational levels, facilitating genotoxic resistance. Notably, in stressed S-phase cells, we found that PLK1 expression levels varied among individual cells, yet overall cell population acquired genotoxin resistance. The genotoxin resistance in the cell population with heterogeneous PLK1 expression was driven by autophagy by facilitating the secretion of currently unidentified factors, likely by switching funtion of RAD9A from DNA checkpoint to substance secretion. Together our data demonstrate that intra-tumor heterogeneity contributes to the malignant features of tumors through an autophagy-PLK-RAD9A axis that promotes intercellular communication via secretion. (180 words).
    Keywords:  DNA replication checkpoint; autophagy; cancer; genotoxic resistance; radiation
    DOI:  https://doi.org/10.1093/jb/mvaf027
  27. Entropy (Basel). 2025 Apr 29. pii: 483. [Epub ahead of print]27(5):
      The intricate multi-scale phenomenon of entropy generation, resulting from the inhomogeneous and anisotropic rearrangement of cells during their collective migration, is examined across three distinct regimes: (i) convective, (ii) conductive (diffusion), and (iii) sub-diffusion. The collective movement of epithelial monolayers on substrate matrices induces the accumulation of mechanical stress within the cells, which subsequently influences cell packing density, velocity, and alignment. Variations in these physical parameters affect cell-cell interactions, which play a crucial role in the storage and dissipation of energy within multicellular systems. The internal dynamics of entropy generation, as a consequence of energy dissipation, are characterized in each regime using viscoelastic constitutive models and the surface properties at the cell-matrix biointerface. The focus of this theoretical review is to clarify how cells can modulate their rate of energy dissipation by altering cell-cell and cell-matrix adhesion interactions, undergoing changes in shape, and re-establishing polarity due to the contact inhibition of locomotion. We approach these questions by discussing the physical aspects of these complex phenomena.
    Keywords:  accumulation of mechanical stress; cell jamming state transition; cell-cell interactions; contact inhibition of locomotion; viscoelasticity caused by collective cell migration
    DOI:  https://doi.org/10.3390/e27050483
  28. Am J Physiol Cell Physiol. 2025 May 30.
      Cells rely on substrate adhesion to activate diverse signaling pathways essential for proliferation and survival. In the absence of proper adhesion to extracellular matrix, cells undergo anoikis, a form of programmed cell death. Poorly attached cells often exhibit rounded morphology and form dynamic protrusions called blebs. While the role of blebs in amoeboid migration is well-documented, recent studies have highlighted their role in anoikis resistance. However, little is known about whether the most abundant membrane components-phospholipids- function in blebs-facilitated anoikis resistance. Here, we report an enrichment of PI(3,4,5)P3 and a depletion of PI(4,5)P2 at bleb membrane, compared to non-bleb regions of the plasma membrane. Our results have shown that both PI(3,4,5)P3 and PI(4,5)P2 have restricted diffusion pattern between the bleb and non-bleb membrane regions. Subsequently, we reveal that PI3K is recruited by SH3KBP1 via liquid-liquid phase separation (LLPS) and interacted with Septin at the bleb necks. This Septin-SH3KBP1-PI3K axis then contributes to differential phosphoinositides (PIs) distribution and anoikis resistance. These novel insights into PIs dynamics and the associated molecular scaffolding not only elucidate the mechanisms of blebs formation and anoikis resistance, but also highlight potential targets for therapeutic interventions in anchorage-independent cancers.
    Keywords:  anoikis; bleb; phosphoinositides
    DOI:  https://doi.org/10.1152/ajpcell.00096.2025
  29. Proc Natl Acad Sci U S A. 2025 Jun 03. 122(22): e2502876122
      Ferroptosis is a cell death mechanism distinguished by its dependence on iron-mediated lipid oxidation. Cancer cells highly resistant to conventional therapies often demonstrate lipid metabolic and redox vulnerabilities that sensitize them to cell death by ferroptosis. These include a unique dependency on the lipid antioxidant selenoenzyme, glutathione peroxidase 4 (GPx4), that acts as a ferroptosis inhibitor. Synthetic high-density lipoprotein-like nanoparticle (HDL NP) targets the high-affinity HDL receptor scavenger receptor class B type 1 (SR-B1) and regulates cell and cell membrane lipid metabolism. Recently, we reported that targeting cancer cell SR-B1 with HDL NP depleted cell GPx4, which is accompanied by increased cell membrane lipid peroxidation and cancer cell death. These data suggest that HDL NP may induce ferroptosis. Thus, we conducted an unbiased CRISPR-based positive selection screen and target validation studies in ovarian clear cell carcinoma (OCCC) cell lines to ascertain the mechanism through which HDL NP regulates GPx4 and kills cancer cells. The screen revealed two genes, acyl-CoA synthetase long chain family member 4 (ACSL4) and thioredoxin reductase 1 (TXNRD1), whose loss conferred resistance to HDL NP. Validation of ACSL4 supports that HDL NP induces ferroptosis as the predominant mechanism of cell death, while validation of TXNRD1 revealed that HDL NP reduces cellular selenium and selenoprotein production, most notably, GPx4. Accordingly, we define cancer cell metabolic targets that can be simultaneously actuated by a multifunctional, synthetic HDL NP ligand of SR-B1 to kill cancer cells by ferroptosis.
    Keywords:  cancer; cell death; ferroptosis; lipids; nanoparticles
    DOI:  https://doi.org/10.1073/pnas.2502876122