bims-cagime Biomed News
on Cancer, aging and metabolism
Issue of 2025–07–20
forty-two papers selected by
Kıvanç Görgülü, Technical University of Munich



  1. Nature. 2025 Jul 16.
      Tumour necrosis is associated with poor prognosis in cancer1,2 and is thought to occur passively when tumour growth outpaces nutrient supply. Here we report, however, that neutrophils actively induce tumour necrosis. In multiple cancer mouse models, we found a tumour-elicited Ly6GHighLy6CLow neutrophil population that was unable to extravasate in response to inflammatory challenges but formed neutrophil extracellular traps (NETs) more efficiently than classical Ly6GHighLy6CHigh neutrophils. The presence of these 'vascular-restricted' neutrophils correlated with the appearance of a 'pleomorphic' necrotic architecture in mice. In tumours with pleomorphic necrosis, we found intravascular aggregates of neutrophils and NETs that caused occlusion of the tumour vasculature, driving hypoxia and necrosis of downstream vascular beds. Furthermore, we found that cancer cells adjacent to these necrotic regions (that is, in 'perinecrotic' areas) underwent epithelial-to-mesenchymal transition, explaining the paradoxical metastasis-enhancing effect of tumour necrosis. Blocking NET formation genetically or pharmacologically reduced the extent of tumour necrosis and lung metastasis. Thus, by showing that NETs drive vascular occlusion, pleomorphic necrosis and metastasis, we demonstrate that tumour necrosis is not necessarily a passive byproduct of tumour growth and that it can be blocked to reduce metastatic spread.
    DOI:  https://doi.org/10.1038/s41586-025-09278-3
  2. bioRxiv. 2025 Jun 27. pii: 2025.06.25.661602. [Epub ahead of print]
      The asymmetric resting distribution of the three major phospholipid classes on the mammalian plasma membrane, with phosphatidylserine and phosphatidylethanolamine mostly on the inner leaflet, and phosphatidylcholine mostly on the outer leaflet, is maintained by ATP-dependent flippases and floppases that exhibit headgroup selectivity. Upon signaling cues, this asymmetry can be dissipated by various phospholipid scramblases, allowing cells to respond to stimuli and adapt to different physiological contexts. The prevailing view in the field is that phospholipid scramblases on the plasma membrane act without headgroup preference. Here we report contrary experimental evidence based on a phospholipid scrambling assay that quantifies the fluorescence polarization of nitrobenzoxadiazole-labeled phospholipids for kinetic monitoring of phospholipid scrambling on the plasma membrane of living cells. Our experiments reveal that the plasma membrane-residing calcium-activated phospholipid scramblase TMEM16F preferentially acts on phosphatidylserine and phosphatidylcholine over phosphatidylethanolamine.
    DOI:  https://doi.org/10.1101/2025.06.25.661602
  3. bioRxiv. 2025 Jun 12. pii: 2025.06.10.656791. [Epub ahead of print]
      The transition from benign to malignant growth is a pivotal yet poorly understood step in cancer progression that marks the shift from a pathologically inert condition to a clinically lethal disease. Here, we integrate lineage tracing, single-cell and spatial transcriptomics to visualize the molecular, cellular and tissue-level events that promote or restrain malignancy during the tumor initiation in mouse models of pancreatic ductal adenocarcinoma (PDAC). We identify a discrete progenitor-like population of KRAS -mutant cells that co-activates oncogenic and tumor-suppressive programs-including p53, CDKN2A, and SMAD4-engaging senescence-like responses and remodeling their microenvironment, ultimately assembling a niche that mirrors invasive PDAC. KRAS inhibition depletes progenitor-like cells and dismantles their niche. Conversely, p53 suppression enables progenitor cell expansion, epithelial-mesenchymal reprogramming, and immune-privileged niche formation. These findings position the progenitor-like state as the convergence point of cancer-driving mutations, plasticity, and tissue remodeling-revealing a critical window for intercepting malignancy at its origin.
    DOI:  https://doi.org/10.1101/2025.06.10.656791
  4. bioRxiv. 2025 Jun 01. pii: 2025.05.29.656943. [Epub ahead of print]
      Preclinical models of pancreatic ductal adenocarcinoma (PDAC) can greatly benefit from non-invasive imaging for evaluating disease progression and therapeutic response. Imaging approaches that can accurately and simultaneously track primary tumor growth, metastatic dissemination, and host cachexia over time are lacking. Here, we report an optimized dual-contrast micro-computed tomography (microCT) protocol for longitudinal imaging in orthotopic murine models of PDAC. This method enables high-resolution, volumetric quantification of orthotopic primary tumors, liver and lung metastases, and paraspinal skeletal muscle, providing a dynamic view of both tumor and host physiology. MicroCT primary tumor measurements strongly correlated with endpoint tumor weights and outperformed 2D ultrasound in early detection and volumetric accuracy, particularly for small or irregularly shaped tumors. This platform revealed heterogeneous metastatic kinetics across PDAC models and uncovered early, heterogeneous onset of skeletal muscle wasting, a hallmark of cancer cachexia. Notably, this protocol mimics clinical CT surveillance by enabling opportunistic cachexia assessment from tumor imaging datasets and offers substantial advantages over destructive endpoint analyses. Further, microCT radiation had no effect on our model endpoints. By capturing the temporal dynamics of tumor progression and host response, dual contrast microCT serves as a powerful translational platform for preclinical PDAC research and therapeutic testing.
    Significance: Dual-contrast microCT provides high-resolution, whole-body, non-invasive imaging in orthotopic murine PDAC models, enabling simultaneous tracking of tumor growth, metastasis, and skeletal muscle wasting-offering a clinically relevant, translational imaging platform.
    DOI:  https://doi.org/10.1101/2025.05.29.656943
  5. bioRxiv. 2025 Jul 07. pii: 2025.07.07.663528. [Epub ahead of print]
      Pancreatic ductal adenocarcinoma (PDAC) evolves through non-invasive precursor lesions, yet its earliest molecular events remain unclear. We established the first spatially resolved proteomic atlas of these lesions using Deep Visual Proteomics (DVP). AI-driven computational pathology classified normal ducts, acinar-ductal metaplasia (ADM), and pancreatic intraepithelial neoplasia (PanIN) from cancer-free organ donors (incidental, "iPanINs") and PDAC patients (cancer-associated, "cPanINs"). Laser microdissection of 96 discrete regions containing as few as 100 phenotypically matched cells and ultrasensitive mass spectrometry quantified a total of 8,512 proteins from formalin-fixed tissues. Distinct molecular signatures stratifying cPanINs from iPanINs, and remarkably, many cancer-associated proteins already marked histologically normal epithelium. Four core programs - stress adaptation, immune engagement, metabolic reprogramming, mitochondrial dysfunction - emerged early and intensified during progression. By integrating DVP with AI-guided tissue annotation, we demonstrate that molecular reprogramming precedes histological transformation, creating opportunities for earlier detection and interception of a near-uniformly lethal cancer.
    Significance: Our spatially-resolved proteomics atlas uncovers distinct molecular signatures in pancreatic cancer adjacent precursor lesions, clearly diverging from those in incidental, cancer-free pancreatic lesions. Our deep proteomics dataset offers a valuable resource for identifying novel biomarkers and therapeutic targets, informed by the earliest cancer-associated molecular events in archival pancreatic tissues.
    DOI:  https://doi.org/10.1101/2025.07.07.663528
  6. Genes Dev. 2025 Jul 15.
      Cellular senescence plays a dual role in tissue biology by promoting tumor suppression and wound healing when transient but driving inflammation, fibrosis, and age-related disease when persistent. The growing recognition that senescent cell clearance can reverse these pathologies has catalyzed efforts to develop therapeutics that preferentially kill senescent cells (also known as "senolytics"). However, clinical translation from bench to bedside remains challenging due to senescent state heterogeneity, limited biomarkers, off-target toxicities, and the frailty of aged patients. Small molecule senolytics, although promising, often lack defined mechanisms of action and pose safety concerns that may constrain their use in older adults. Emerging precision approaches, including those that exploit surface markers and leverage engineered immune therapies, offer a rational and potentially more selective path forward. Here we highlight recent advances in senescence profiling and targeted clearance strategies, emphasizing the need for therapies designed with both biological complexity and the needs of aging populations in mind.
    Keywords:  aging; senescence; senolytics
    DOI:  https://doi.org/10.1101/gad.353134.125
  7. Nat Biomed Eng. 2025 Jul 14.
      The efficacy of drug therapy in multiple myeloma is conventionally assessed by whole-cell-population methods, serum analysis of light chains and monoclonal antibodies, immunofixation electrophoresis, or by flow cytometry of bone marrow aspirates and biopsies. These methods provide relevant information on the presence of specific immunoglobulins at high sensitivity and specificity but require a large number of cells, involve long and laborious sample preparation steps, and provide only tumour bulk information. Here we develop a single-cell imaging technique requiring a reduced number of primary cells for longitudinal evaluation of patient-specific treatment and assessment of treatment heterogeneity. By exploiting the mechanistic action of proteasome inhibition and in synergy with the label-free protein-structure specificity of mid-infrared optoacoustic microscopy, we present a technology that facilitates longitudinal evaluation of myeloma treatment and a patient's heterogeneous response. Detecting optical-generated ultrasound waves that intensify with optical absorption, this technology allows observation of proteins in living cells with high sensitivity. Specifically, we use intermolecular β-sheet formation as a biomarker for cell viability during therapy and apply it to assess drug-treatment performance in multiple myeloma patients.
    DOI:  https://doi.org/10.1038/s41551-025-01443-3
  8. J Clin Invest. 2025 Jul 15. pii: e191943. [Epub ahead of print]135(14):
      Pancreatic ductal adenocarcinoma (PDAC) remains among the most lethal cancers, with metastasis as the primary driver of mortality. While metastatic mechanisms are shared across malignancies, PDAC metastasis poses unique therapeutic challenges due to the presence of extensive tumor heterogeneity, desmoplasia, and immunosuppression - features that enable diverse migratory behaviors and therapeutic resistance. Recent advances have shown that metastatic progression in PDAC emerges from dynamic interactions between tumor cell-intrinsic and microenvironmental factors, each adapting to evolving stressors throughout the metastatic cascade. In the primary tumor, genomic instability and epigenetic reprogramming generate subclones with heightened invasive potential, while dense stromal reactions and myeloid-dominated immune suppression facilitate escape. During circulation, PDAC cells employ distinctive survival strategies through homotypic clustering and heterotypic interactions with blood components. At distant sites, PDAC cells adapt to organ-specific microenvironments through context-dependent metabolic and immune modulation, resulting in phenotypes that diverge from the primary tumor. In this Review, we examine how tumor-stroma crosstalk mechanisms shape metastatic progression in PDAC, provide a framework for understanding why conventional therapies often fail against metastatic disease, and highlight emerging opportunities for stage- and site-specific therapeutic interventions that target these unique adaptations.
    DOI:  https://doi.org/10.1172/JCI191943
  9. J Clin Invest. 2025 Jul 15. pii: e191940. [Epub ahead of print]135(14):
      The tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) is composed of a dense stromal compartment and is poorly vascularized, resulting in limited nutrient delivery. As a result, PDAC cells must adapt to cope with the metabolic stresses brought on by TME nutrient limitation. In this article, we first review recent studies that have provided quantitative measurements of nutrient levels in the PDAC TME. These studies have provided a new understanding of the nutrient limitations and metabolic stresses that occur in PDAC. We next discuss the adaptive strategies employed by PDAC in response to TME nutrient limitation. We propose that PDAC adaptations to metabolic stress can be generalized into four categories: (a) cutting down on metabolic costs by recycling metabolites and suppressing nonessential processes, (b) upregulating biosynthetic pathways to meet TME metabolic demands, (c) supporting essential metabolic processes with alternative fuel sources, and (d) dampening antiproliferative and cell death responses that nutrient limitation normally triggers. Improving our understanding of the nutrient limitations within the TME, and the adaptations cells employ to cope with these stresses, provides a more complete picture of PDAC biology and reveals new opportunities for therapeutic targeting of this disease.
    DOI:  https://doi.org/10.1172/JCI191940
  10. bioRxiv. 2025 May 10. pii: 2025.05.08.652966. [Epub ahead of print]
      Aging is a critical yet understudied determinant in pancreatic ductal adenocarcinoma (PDAC). Despite a strong epidemiological association with age, conventional PDAC preclinical models fail to capture the histopathological and stromal complexities that emerge in older organisms. Using an age-relevant syngeneic orthotopic model, we demonstrate that organismal aging accelerates PDAC progression and metastasis. Through transcriptomic profiling, we identify a conserved extracellular matrix gene signature enriched in cancer-associated fibroblasts (CAFs) from aged tumors, consistent with an augmented fibrotic landscape that supports immunosuppression, metastatic tropism, and poor prognosis. To directly test the functional impact of stromal aging, we employed heterochronic co-implantation models, revealing that revitalizing the aged tumor stroma with young CAFs restores immune infiltration and attenuates metastasis in older hosts. Conversely, aged CAFs, while immunosuppressive, fail to enhance metastasis in young hosts, suggesting that a youthful microenvironment exerts dominant regulatory control over disease progression. These findings demonstrate that stromal age is a critical modulator of both immune exclusion and metastatic behavior in PDAC. Importantly, our work establishes a new conceptual framework for understanding how aging shapes the tumor microenvironment in PDAC and opens a fertile avenue of investigation into age-specific stromal regulation. Moreover, this work raises compelling questions about the underlying molecular mechanisms-questions now accessible through our models-and lays the foundation for future efforts to therapeutically target stromal aging in PDAC.
    Statement of Significance: Our study links aging, stromal remodeling, and PDAC aggressiveness, highlighting how age-dependent stromal changes drive progression and suggesting that rejuvenating the aged microenvironment may improve outcomes in older patients.
    DOI:  https://doi.org/10.1101/2025.05.08.652966
  11. J Clin Invest. 2025 Jul 15. pii: e191936. [Epub ahead of print]135(14):
      
    DOI:  https://doi.org/10.1172/JCI191936
  12. bioRxiv. 2025 May 10. pii: 2025.05.09.653205. [Epub ahead of print]
      The cell NAD+/NADH ratio can constrain biomass synthesis and influence proliferation in nutrient-limited environments. However, which cell processes regulate the NAD+/NADH ratio is not known. Here, we find that some cancer cells elevate the NAD+/NADH ratio in response to serine deprivation by increasing mitochondrial respiration. Cancer cells that elevate mitochondrial respiration have higher serine production and proliferation in serine limiting conditions than cells with no mitochondrial respiration response, independent of serine synthesis enzyme expression. Increases in mitochondrial respiration and the NAD+/NADH ratio promote serine synthesis regardless of whether serine is environmentally limiting. Lipid deprivation can also increase the NAD+/NADH ratio via mitochondrial respiration in some cells, including cells that do not increase respiration following serine deprivation. Thus, in cancer cells where lipid depletion raises the NAD+/NADH ratio, proliferation in serine depleted environments improves when lipids are also depleted. Taken together, these data suggest that changes in mitochondrial respiration in response to nutrient deprivation can influence the NAD+/NADH ratio in a cell-specific manner to impact oxidative biomass synthesis and proliferation. Given the complexity of tumor microenvironments, this work provides a metabolic framework for understanding how levels of more than one environmental nutrient affects cancer cell proliferation.
    DOI:  https://doi.org/10.1101/2025.05.09.653205
  13. bioRxiv. 2025 Jun 25. pii: 2025.06.20.660757. [Epub ahead of print]
      The molecular pathways involved in the response to radiation therapy in pancreatic ductal adenocarcinoma (PDAC) remain poorly understood. We aimed to elucidate the adaptive mechanisms and cellular interactions within PDAC to radiation therapy (RT). We constructed a transcriptomic landscape of the cellular subtypes and spatially resolved neighborhoods from 50 patient samples, including 16 longitudinally matched single cell RNA sequencing and 34 spatial transcriptomics specimens. To resolve shortcomings of cell-type mixtures in spatial data, we developed a novel statistical method called SpaCCI (spatially aware analysis of cell-cell interactions) to profile cell-cell interactions and ligand-receptor enrichment. This revealed CXCL12/TGFβ-driven persister cell niches where activated fibroblasts reprogram tumor- associated macrophages and spatially exclude stress-response CD8 T cells after RT. Persister cancer cells displayed transcriptional evidence of recalcitrance to metal-induced cell death pathways of ferroptosis and cuproptosis which were recapitulated in preclinical models. Our study reveals the selective pressures experienced by PDAC following RT that may help provide insight for future multimodal therapeutic strategies.
    DOI:  https://doi.org/10.1101/2025.06.20.660757
  14. bioRxiv. 2025 Jun 25. pii: 2025.06.20.660715. [Epub ahead of print]
      Despite the availability of RAS inhibitors and the dependence of >90% of pancreatic ductal adenocarcinomas (PDAC) on oncogenic KRAS mutations, resistance to KRAS inhibition remains a serious obstacle. We show here that phosphoinositide 3-kinase (PI3K) plays a major role in this resistance through upstream activation of wild-type RAS signaling - beyond its known KRAS effector function. Combining proximity labeling, CRISPR screens, live-cell imaging, and functional assays we found that PI3K orchestrates phosphoinositide-mediated GAB1 recruitment to the plasma membrane, nucleating assembly of RAS signaling complexes that activate mitogen-activated protein kinase (MAPK) in an EGFR/SHP2/SOS1-dependent manner. We further demonstrate that inhibiting PI3K enhances sensitivity to mutant-specific KRAS inhibitors in PDAC cells, including cells with clinically identified PIK3CA mutations. Our findings refine RAS-PI3K signaling paradigms, reveal that PI3K-driven wild-type RAS activation drives resistance to KRAS inhibition, and illuminate new avenues for augmenting KRAS-targeted therapies in PDAC.
    DOI:  https://doi.org/10.1101/2025.06.20.660715
  15. bioRxiv. 2025 Jun 17. pii: 2025.06.12.659314. [Epub ahead of print]
      Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer, with liver metastases significantly worsening outcomes. However, distinct features of the tumor microenvironment (TME) between primary and metastatic sites remain poorly defined. Cellular neighborhoods within the TME are recognized as functional units that influence tumor behavior. Conventional spatial methods, which assign equal weights to all cells in a region, fail to capture the nuances of cellular interactions. To address this, we developed Functional Cellular Neighborhood (FunCN) quantification, which integrates both the proportion and proximity of surrounding cells. Applying FunCN to PDAC imaging mass cytometry data, we identified neutrophil-enriched interactions in liver metastases compared to primary tumors, correlating with elevated VISTA expression by tumor cells. Additionally, FunCN clusters around CD8 + T cells in pancreas and liver were associated with higher TIGIT and LAG3, respectively. These findings demonstrate the importance of spatial immune landscapes in PDAC and identify potential therapeutic opportunities.
    DOI:  https://doi.org/10.1101/2025.06.12.659314
  16. bioRxiv. 2025 May 03. pii: 2025.04.29.651251. [Epub ahead of print]
      Pancreatic ductal adenocarcinoma (PDAC) occurs as a complex, multifaceted event driven by the interplay of tumor permissive genetic mutations, nature of cellular origin and microenvironmental stress. In this study, using primary human pancreatic acinar 3D organoids, we performed CRISPR knockout screen targeting 199 previously underappreciated potential tumor suppressors curated from clinical PDAC samples. Our data revealed significant enrichment of a list of candidates, with NF2 emerging as the top target. Functional validation confirmed that loss of NF2 promotes the transition of PDAC to an invasive state, potentially through extracellular matrix modulation. NF2 inactivation was found to enhance PDAC cell fitness under nutrient starvation. This adaptation not only reinforces the oncogenic state but also confers therapeutical resistance. Additionally, we found that NF2 loss is associated with the fibroblast heterogeneity and cancer-stroma communications in tumor evolution. These findings establish NF2 as a critical tumor suppressor in PDAC and uncover its role in mediating nutrient adaptation and drug resistance. Importantly, this study provides new insights into drug resistance mechanisms and potential therapeutic targets in PDAC.
    DOI:  https://doi.org/10.1101/2025.04.29.651251
  17. Small. 2025 Jul 17. e04631
      One-carbon (1C) metabolism supports the de novo synthesis of purines and pyrimidines, which are essential for cancer growth. So far, the most employed assay for targeting 1C metabolism is using the inhibitors of specific enzymes. However, metabolic compensation undermines their efficacy, and the lack of cell selectivity limits their application. Herein, a bifunctional Pd nanocatalyst coated with the cancer cell membrane (Pd@M) has been constructed to target 1C metabolism in cancer cells, delivering precise and potent cancer therapy. On the one hand, the Pd@M can catalyze formate and cause its depletion to disrupt cytoplasmic 1C metabolism and suppress cell metastasis. On the other hand, the Pd@M activates prodrugs in situ by bioorthogonal catalysis to inhibit the compensatory pathway, which strongly decreases nucleotide synthesis flux, thus causing potent inhibition of cell proliferation. This work presents a new way to disrupt cytoplasmic 1C metabolism and suppress cell metastasis by combination of precise depletion of cancer cell formate and inhibition of the compensatory pathway by using bioorthogonal chemistry.
    Keywords:  bioorthogonal; cancer therapy; metastasis suppression; nanocatalysts; one‐carbon metabolism
    DOI:  https://doi.org/10.1002/smll.202504631
  18. Nat Aging. 2025 Jul 11.
      There is robust evidence that senescence can be propagated in vitro through mechanisms including the senescence-associated secretory phenotype, resulting in the non-cell-autonomous induction of secondary senescence. However, the induction, regulation and physiological role of secondary senescence in vivo remain largely unclear. Here we generated senescence-inducible mouse models expressing either the constitutively active form of MEK1 or MKK6 and mCherry, to map primary and secondary senescent cells. Our models recapitulate characteristic features of senescence and demonstrate that primary and secondary phenotypes are highly tissue- and inducer-dependent. Spatially resolved RNA expression analyses at the single-cell level reveal that each senescence induction results in a unique transcriptional profile-even within cells of the same cell type-explaining the heterogeneity of senescent cells in vivo. Furthermore, we show that interleukin-1β, primarily derived from macrophages, induces secondary phenotypes. Our findings provide insight into secondary senescence in vivo and useful tools for understanding and manipulating senescence during aging.
    DOI:  https://doi.org/10.1038/s43587-025-00917-y
  19. bioRxiv. 2025 May 01. pii: 2025.04.28.650414. [Epub ahead of print]
      Aggressive cancers, such as pancreatic ductal adenocarcinoma (PDAC), are often characterized by a complex and desmoplastic tumor microenvironment rich in stroma, a supportive connective tissue composed primarily of extracellular matrix (ECM) and non-cancerous cells. Desmoplasia, which is a dense deposition of stroma, is a major reason for therapy resistance, acting both as a physical barrier that interferes with drug penetration and as a supportive niche that protects cancer cells through diverse mechanisms. A precise understanding of spatial cell interactions within the tumor microenvironment in stroma-rich cancers is essential for optimizing therapeutic responses. It allows detailed mapping of stromal-tumor interfaces, comprehensive phenotyping of diverse cell types and their functional states, and insights into changes in cellular distribution and tissue architecture, thus leading to an improved assessment of drug responses. Recent advances in multiplexed immunofluorescence imaging have enabled the acquisition of large batches of whole-slide tumor images, but scalable and reproducible methods to analyze the spatial distribution of cell states relative to stromal regions remain limited. To address this gap, we developed an open-source computational pipeline that integrates QuPath (Bankhead et al. 2017), StarDist (Schmidt et al. 2018), and custom Python scripts to quantify biomarker expression at a single- and sub-cellular resolution across entire tumor sections. Our workflow includes: (i) automated nuclei segmentation using StarDist, (ii) machine learning-based cell classification using multiplexed marker expression, (iii) modeling of stromal regions based on fibronectin staining, (iv) sensitivity analyses on classification thresholds to ensure robustness across heterogeneous datasets, and (v) distance-based quantification of the proximity of each cell to the stromal border. To improve consistency across slides with variable staining intensities, we introduce a statistical strategy that translates classification thresholds by propagating a chosen reference percentile across the distribution of marker-related cell measurement in each image. We apply this approach to quantify spatial patterns of distribution of the phosphorylated form of the N-Myc downregulated gene 1 (NDRG1), a novel DNA repair protein that conveys signals from the ECM to the nucleus to maintain replication fork homeostasis, and a known cell proliferation marker Ki67 in fibronectin-defined stromal regions in PDAC xenografts. The pipeline is applicable for the analysis of various stroma-rich tissues and is publicly available: https://github.com/HMS-IAC/stroma-spatial-analysis-web .
    Summary paragraph: 2.Our study introduces a scalable and reproducible image analysis pipeline that quantifies spatial biomarker distributions relative to the stroma in tumor tissues using open-source tools. By modeling cell-level intensity distributions and calibrating classification thresholds across heterogeneous images, we uncover spatially organized patterns of stroma sensing, DNA damage, and proliferative response in pancreatic tumors. This approach enables robust, quantitative analysis of tumor-stroma interactions and is readily adaptable to other tumor types and biomarker panels, providing a valuable resource for spatial pathology and tumor microenvironment research.
    DOI:  https://doi.org/10.1101/2025.04.28.650414
  20. Nat Cell Biol. 2025 Jul;27(7): 1114-1124
      The aminoglycoside antibiotic neomycin has robust antibacterial properties, yet its clinical utility is curtailed by its nephrotoxicity and ototoxicity. The mechanism by which the polycationic neomycin enters specific eukaryotic cell types remains poorly understood. In budding yeast, NEO1 is required for neomycin resistance and encodes a phospholipid flippase that establishes membrane asymmetry. Here we show that mutations altering Neo1 substrate recognition cause neomycin hypersensitivity by exposing phosphatidylinositol-4-phosphate (PI4P) in the plasma membrane extracellular leaflet. Cryogenic electron microscopy reveals PI4P binding to Neo1 within the substrate translocation pathway. PI4P enters the lumen of the endoplasmic reticulum and is flipped by Neo1 at the Golgi to prevent PI4P secretion to the cell surface. Deficiency of the orthologous ATP9A in human cells also causes exposure of PI4P and neomycin sensitivity. These findings unveil conserved mechanisms of aminoglycoside sensitivity and phosphoinositide homoeostasis, with important implications for signalling by extracellular phosphoinositides.
    DOI:  https://doi.org/10.1038/s41556-025-01692-z
  21. J Chem Phys. 2025 Jul 21. pii: 034708. [Epub ahead of print]163(3):
      The physicochemical properties of lipid bilayers (membranes) are closely associated with various cellular functions and are often evaluated using absorption and fluorescence spectroscopies. For instance, by employing fluorescent probes that exhibit spectra reflective of the surrounding membrane environment, one can estimate the membrane polarity. Thus, elucidating how such probes are embedded within the membranes would be beneficial for enabling a deeper interpretation of the spectra. Here, we apply molecular dynamics simulation with an enhanced sampling method to investigate the embedded state of 6-propionyl-2-dimethylaminonaphthalene (Prodan) within a membrane composed of 1,2-dioleoyl-sn-glycero-3-phosphocholine, as well as its variation upon the addition of ethanol as a cosolvent to the aqueous phase. In the absence of ethanol, it is found that the bulky moieties of Prodan (propionyl and dimethylamine groups) prefer to be oriented toward the membrane center owing to the voids existing near the center. The structural change in the membrane induced by the addition of ethanol causes a reduction in the void population near the center, resulting in a diminished orientation preference of Prodan.
    DOI:  https://doi.org/10.1063/5.0279530
  22. bioRxiv. 2025 May 04. pii: 2025.04.29.650640. [Epub ahead of print]
      Recent advances in spatial proteomics have enabled high-dimensional protein analysis within tissue samples, yet few methods accurately detect low-abundance functional proteins. Spatial MIST (Multiplex In Situ Tagging) is one such technique, capable of profiling over 100 protein markers spatially at single-cell resolution on tissue sections and cultured cells. However, despite the availability of various open-source tools for image registration and visualization, no dedicated software exists to align the images and analyze spatial MIST data effectively. To address this gap, we present MIST-Explorer, a comprehensive, user-friendly toolkit for the visualization and analysis of single-cell spatial MIST array data. Developed in Python with a PyQt6-based graphical interface, MIST-Explorer streamlines the spatial omics workflow-from image preprocessing and registration to cell segmentation and protein quantification. The software supports two workflows: one for preprocessed datasets and another for raw image inputs, ensuring broad compatibility across experimental designs. Key features include tile-based image registration using Astroalign and PyStackReg, deep learning-based segmentation with StarDist, multi-channel visualization with layer controls, and an interactive analysis module offering ROI selection along with histograms, heatmaps, and UMAP plots. MIST-Explorer generates spatially resolved expression tables readily compatible with downstream single-cell analysis pipelines. By integrating all major steps into a single platform, MIST-Explorer empowers researchers to derive biological insights from complex spatial omics datasets without requiring extensive computational expertise.
    Availability and implementation: Freely available at https://github.com/MIST-Explorer/MIST-Explorer .
    DOI:  https://doi.org/10.1101/2025.04.29.650640
  23. bioRxiv. 2025 Jul 11. pii: 2025.07.06.663397. [Epub ahead of print]
      A burgeoning approach to treat cancer is the pharmacological induction of ferroptotic cell death of tumor cells. However, the impact of disrupting anti-ferroptotic pathways in the broader tumor microenvironment (TME), such as in immune cells, is still undefined and may complicate treatments. Here, we show that Ferroptosis Suppressor Protein 1 (FSP1 /Aifm2 ) is critically required for regulatory T cell (Treg) resistance to ferroptosis and their immunosuppressive function within the TME. Compared to other canonical ferroptosis regulators such as GPX4, GCH1, and NRF2, only FSP1 was induced upon T cell activation. Deletion of Aifm2 in all T cells, or Tregs specifically, enhanced tumor control by selectively disrupting Treg immunosuppression within tumors without inciting autoimmune pathology in mice. As opposed to deletion of Gpx4 in all T cells, T cell deletion of Aifm2 did not impair antigen-specific CD8 + T cell responses. These results reveal a unique opportunity for targeting a regulator of ferroptosis that can not only directly target cancer cells, but also simultaneously enhance anti-cancer immune responses without inciting autoimmunity.
    DOI:  https://doi.org/10.1101/2025.07.06.663397
  24. bioRxiv. 2025 May 09. pii: 2025.05.04.652145. [Epub ahead of print]
      Spatial proteomics via multiplexed tissue imaging is transforming how we study biology, enabling researchers to investigate dozens of markers in a single tissue section and explore how cells behave in their native habitat. While imaging technologies have advanced rapidly, data analyses remain a bottleneck. To address this, we developed PIPΣX (Pipeline for Image Processing and EXploration), a user-friendly, end-to-end open-source software designed to make complex image analysis approachable, even for those with little or no programming skills. PIPΣX combines robust automation with an intuitive graphical user interface, guiding users through each step of the analysis, from image preprocessing and membrane-aware cell segmentation to signal quantification and spatial data exploration. Each feature includes built-in explanations, recommendations, and quality controls to help users make confident choices throughout the process. PIPΣX is compatible with a wide range of multiplexed imaging platforms, and its outputs integrate seamlessly with visualization tools like TissUUmaps and QuPath. Also, it supports downstream applications by enabling direct export of selected cell coordinates for laser microdissection. This functionality facilitates precise isolation of target cell populations for deep proteomic or transcriptomic profiling. With PIPΣX, researchers can extract meaningful biological insights from multiplexed images more easily and robustly, helping to bridge the gap between powerful imaging technologies and real-world scientific discovery.
    Highlights: PIPΣX offers a user-friendly "one-stop shop" pipeline for multiplexed tissue image analysiswithout codingWorks across diverse tissue types and imaging platforms at whole-slide scaleIncludes membrane-aware segmentation and quality control featuresSeamlessly integrates with visualization platforms like TissUUmaps and QuPath for data explorationEnables export for automated laser microdissection and spatial single-cell profiling.
    DOI:  https://doi.org/10.1101/2025.05.04.652145
  25. Res Sq. 2025 Jul 10. pii: rs.3.rs-6986228. [Epub ahead of print]
      Metastasis remains the leading cause of cancer-related mortality. Disseminated tumor cells (DTCs) colonize distant organs where they enter a prolonged state of quiescence, named cellular dormancy, within collagen-rich extracellular matrix (ECM) niches. How dormant cells regulate the formation of collagen-rich niches and the mechanisms maintaining collagen proteostasis during dormancy and reactivation are not understood. Here, we identify prolyl hydroxylase P4HA2 as a key regulator of tumor dormancy through its dual role in collagen proline hydroxylation and mitochondrial function. We demonstrate that P4HA2-mediated proline hydroxylation of collagens balances the NAD+/NADH ratio, sustaining dormancy by limiting mitochondrial activity. Loss of P4HA2 disrupts collagen proteostasis, induces autophagy, and activates the proline catabolism enzyme ALDH4A1, lowering the NAD+/NADH ratio, which fuels mitochondrial energetics and triggers DTC awakening. Notably, ALDH4A1 is essential for the survival of these reactivated dormant cells, and its depletion induces apoptosis upon awakening, revealing a metabolic vulnerability in reactivated dormant cells. Our findings establish a previously unrecognized link between collagen homeostasis, NADH metabolism and tumor cell dormancy, unveiling a mechanistic framework for identifying actionable targets to eliminate DTCs and prevent metastatic relapse.
    DOI:  https://doi.org/10.21203/rs.3.rs-6986228/v1
  26. bioRxiv. 2025 May 01. pii: 2025.04.30.651574. [Epub ahead of print]
      Lipid bilayers form the basis of organellar architecture, structure, and compartmentalization in the cell. Decades of biophysical, biochemical, and imaging studies on purified or in vitro reconstituted liposomes have shown that variations in lipid composition influence the physical properties of membranes, such as thickness and curvature. However, similar studies characterizing these membrane properties within the native cellular context have remained technically challenging. Recent advancements in cellular cryo-electron tomography (cryo-ET) imaging enable high-resolution, three-dimensional views of native organellar membrane architecture preserved in near-native conditions. We previously developed a 'Surface Morphometrics' pipeline that generates surface mesh reconstructions to model and quantify cellular membrane ultrastructure from cryo-ET data. Here, we expand this pipeline to measure the distance between the phospholipid head groups (PHG) of the membrane bilayer as a readout of membrane thickness. Using this approach, we demonstrate thickness variations both within and between distinct organellar membranes. We also demonstrate that membrane thickness positively correlates with other features, such as membrane curvedness. Further, we show that subcompartments of the mitochondrial inner membrane exhibit varying membrane thicknesses that are independent of network morphology (i.e., fragmented versus elongated networks). Finally, we demonstrate that large membrane-associated macromolecular complexes exhibit distinct density profiles that correlate with local variations in membrane thickness. Overall, our updated Surface Morphometrics pipeline provides a framework for investigating how changes in membrane composition in various cellular and disease contexts affect organelle ultrastructure and function.
    DOI:  https://doi.org/10.1101/2025.04.30.651574
  27. bioRxiv. 2025 May 05. pii: 2025.05.01.651678. [Epub ahead of print]
      Pancreatic cancer remains as one of the most challenging malignancies to diagnose and treat due to the late development of symptoms and limited early diagnostic options. Intraductal papillary mucinous neoplasms (IPMNs) are non-invasive precursors to invasive pancreatic ductal adenocarcinoma (PDAC)and an understanding of the changes in patterns of protein expression that accompany the progression from normal ductal (ND) cell, to IPMN to PDAC may provide avenues for improved earlier detection. In this study, we present an optimized spatial tissue proteomics workflow, termed SP-Max (Spatial Proteomics Optimized for Maximum Sensitivity and Reproducibility in Minimal Sample), designed to maximize protein recovery and quantification from limited laser micro dissected (LMD) samples. Our workflow enabled the identification of more than 6,000 proteins and the quantification of over 5,200 protein groups from FFPE tissue contours of pancreatic tissues. Comparative analyses across ND, IPMN, and PDAC revealed critical molecular differences in protein pathways and potential markers of progression. SP-Max provides a systematic, reproducible approach that significantly enhances our ability to study precancerous lesions and cancer progression in pancreatic tissues at unprecedented resolution.
    DOI:  https://doi.org/10.1101/2025.05.01.651678
  28. J Clin Invest. 2025 Jul 15. pii: e191937. [Epub ahead of print]135(14):
      Pancreatic ductal adenocarcinoma (PDAC) is known to progress from one of two main precursor lesions: pancreatic intraepithelial neoplasia (PanIN) or intraductal papillary mucinous neoplasm (IPMN). The poor survival rates for patients with PDAC, even those diagnosed with localized disease, highlight the need for pancreatic cancer interception at the precursor stage. Although their basic biological drivers are well characterized, practical strategies for PanIN and IPMN interception remain elusive due to difficulties with detection, risk stratification, and low-morbidity intervention. Recently, advances in liquid biopsy, spatial multiomics analysis, and machine learning technology have provided deeper understanding of the molecular landscapes underlying pancreatic precursor development and progression. In this Review, we outline the different histologic phenotypes, clinical characteristics, and neoplastic cell-intrinsic and -extrinsic drivers of PanINs and IPMNs, with particular focus on current and potential future opportunities for pancreatic precancer interception.
    DOI:  https://doi.org/10.1172/JCI191937
  29. bioRxiv. 2025 May 11. pii: 2025.04.22.650048. [Epub ahead of print]
      The oligomerization of the transmembrane helices of single-pass membrane proteins is crucial to biological function and its misregulation can lead to many diseases. The study of transmembrane helix oligomerization is facilitated by the availability of genetic reporter assays, which are essential tools for understanding the organization and biology of single-pass systems. In particular, reporter assays are crucial for mapping the oligomerization interfaces of transmembrane helices through scanning mutagenesis but their application is limited by the need to clone and measure each construct individually. Here, we present "TOXGREEN sort-seq", a high-throughput version of the TOXGREEN assay that enables the direct measurement of transmembrane helix oligomerization in large libraries using fluorescence-activated cell sorting and next-generation sequencing. We show that TOXGREEN sort-seq is robust and reproduce the direct measurements of individual constructs with good accuracy and sensitivity. The method produced high-quality mutational profiles from a library of 17,400 constructs designed to probe the interface of 100 potential GAS right dimers predicted from sequences of human single-pass membrane proteins. We report the validated structural model of twelve dimers involved in a variety of biological functions, including immune response (interleukin-22 receptor subunit alpha-1, butyrophilin-like protein 3, hepatitis A virus cellular receptor 2), transport (transferrin receptor protein 1), and cell-surface signaling and proliferation (syndecan-3; semaphorins 5A, 6B and 6D). Remarkably, all three semaphorins in the dataset formed strong dimers and produced mutational profiles consistent with the computational structure. These findings open the possibility that dimerization may be relevant to these proteins' activity and provide a validated interface for assessing their biological role.
    DOI:  https://doi.org/10.1101/2025.04.22.650048
  30. Anal Chem. 2025 Jul 16.
      Endocytosis plays a critical role in regulating protein dynamics, and cancer cells often exploit this process to enhance their survival capacity. However, the development of reliable tools for high-fidelity visualization of endocytosis under cellular stress remains a significant challenge. In this study, we introduce a fluorescent probe, o-IP-IP, based on a dimeric dual-anchoring strategy, to investigate plasma membrane dynamics and endocytosis processes. The probe incorporates two alkyl chains for targeted binding to the plasma membrane while maintaining water solubility through two negatively charged units. This design enables o-IP-IP to exhibit target-activated fluorescence enhancement, broad applicability across various cell types, and the ability to distinguish between normal and cancerous cells based on membrane viscosity. The probe's dual anchoring minimizes passive transport-induced internalization, allowing prolonged labeling of the plasma membrane and enabling high-fidelity imaging of both membranes and endocytosis processes under endocytosis inhibition. This feature facilitated real-time observation of endocytosis in living cells under osmotic stress. Furthermore, o-IP-IP enabled visualization of endocytosis induced by different extracellular calcium ion concentrations, confirming its utility as a simple and effective tool for high-fidelity endocytosis tracking in cellular stress and cell death studies. The probe was also applied to study dynamic surface protein distribution under specific external stress, highlighting endocytosis as the dominant mechanism for adjusting surface proteins. These findings underscore endocytosis' central role in maintaining cellular homeostasis and provide effective strategies for designing targeted probes and drugs based on membrane-impermeable substances' endocytosis mechanisms.
    DOI:  https://doi.org/10.1021/acs.analchem.5c01629
  31. bioRxiv. 2025 Jun 17. pii: 2025.06.11.659098. [Epub ahead of print]
      Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense extracellular matrix (ECM) that sustains an immunosuppressive tumor microenvironment (TME). While this protective niche has been described, the molecular determinants orchestrating its formation and dictating its immune interactions are not well defined. Using Perturb-map, we determine how dozens of different gene perturbations shape the growth and cellular environments of PDAC clones through space and time. Our study reveals dynamic, gene-specific adaptations of immune neighborhoods during clonal selection. We identified Serpinb2 (PAI2) and Serpine1 (PAI1) as key cancer-derived mediators of TME remodeling and immune evasion. These factors promote the deposition of a fibrin-rich ECM that shapes immune cell composition, locally retains and polarizes immunosuppressive macrophages and excludes cytotoxic T cells. Deletion of either Serpinb2 or Serpine1 greatly enhanced tumor response to anti-PD1 immunotherapy in an aggressive PDAC model. Transcriptomic analysis further linked their expression to distinct PDAC subtypes and poor patient survival. Our findings demonstrate that Serpinb2 and Serpine1 establish a permissive niche for tumor progression and show how PDAC cells exploit components of the fibrinolysis pathway to remodel the ECM, alter macrophage composition, and protect themselves from immune editing, ultimately reinforcing the role of extracellular factors in shaping an immune-privileged tumor niche.
    DOI:  https://doi.org/10.1101/2025.06.11.659098
  32. bioRxiv. 2025 Jun 15. pii: 2025.06.15.659774. [Epub ahead of print]
      Charting the spatiotemporal dynamics of cell fate determination in development and disease is a long-standing objective in biology. Here we present the design, development, and extensive validation of PEtracer, a prime editing-based, evolving lineage tracing technology compatible with both single-cell sequencing and multimodal imaging methodologies to jointly profile cell state and lineage in dissociated cells or while preserving cellular context in tissues with high spatial resolution. Using PEtracer coupled with MERFISH spatial transcriptomic profiling in a syngeneic mouse model of tumor metastasis, we reconstruct the growth of individually-seeded tumors in vivo and uncover distinct modules of cell-intrinsic and cell-extrinsic factors that coordinate tumor growth. More generally, PEtracer enables systematic characterization of cell state and lineage relationships in intact tissues over biologically-relevant temporal and spatial scales.
    DOI:  https://doi.org/10.1101/2025.06.15.659774
  33. Anal Chim Acta. 2025 Sep 22. pii: S0003-2670(25)00722-6. [Epub ahead of print]1368 344328
       BACK GROUND: Lipid peroxidation (LPO) and abnormal accumulation of lipid droplets (LDs) are key pathogenic factors contributing to the onset and progression of multiple diseases. Fluorescent probes for monitoring LPO and LDs accumulation are promising tools for the early diagnosis of related diseases, providing valuable insights that could lead to improved disease management and therapeutic strategies. However, to the best of our knowledge, a dual-channel fluorescent probe capable of simultaneously monitoring LPO and LD accumulation has not yet been reported.
    RESULTS: We rationally designed and synthesized a dual-responsive fluorescent probe, Cou-LPO4, for simultaneous visualization of LPO and LDs in living cells. Cou-LPO4 exhibits green fluorescence at 560 nm in response to lipid peroxyl radicals (LOO•) with excellent selectivity and sensitivity, and red fluorescence at 640 nm in response to LDs accumulation. Cou-LPO4 was successfully applied to evaluate LPO during RSL3-and erastin-induced ferroptosis by monitoring fluctuations in LOO• levels. Subsequently, using Cou-LPO4, we revealed a significant upregulation of LPO, accompanied by a marked accumulation of LDs in nonalcoholic fatty liver disease (NAFLD) cell model.
    SIGNIFICANCE: Cou-LPO4 is the first dual-channel fluorescent probe enabling simultaneous monitoring of LPO and LDs accumulation in living cells. It provides a powerful tool for studying LPO and LDs accumulation-related diseases like NAFLD, offering new insights for early diagnosis and therapeutic strategies.
    Keywords:  Dual-responsive fluorescent probe; Lipid droplets accumulation; Lipid peroxidation; Lipid peroxyl radical; Live cell imaging
    DOI:  https://doi.org/10.1016/j.aca.2025.344328
  34. JCSM Commun. 2025 Jan-Jun;8(1):pii: e117. [Epub ahead of print]8(1):
       Background: Cancer cachexia is a debilitating syndrome characterized by irreversible losses in skeletal muscle mass, with or without losses in adipose tissue. Cancer cachexia is an underrecognized syndrome that impacts ~50% of all cancer patients and accounts for up to ~20% of all cancer deaths [1, 2]. Lung cancer remains one of the deadliest cancers in the United States with an estimated 137,000 deaths in the year 2021 alone [3]. Lung cancer is highly comorbid with cancer cachexia [4]. Pre-clinical models are heavily relied upon to study both lung cancer and cancer cachexia, however there is a need to develop novel models to study the relationship between the two diseases. We therefore characterized the cachexia phenotype in the CMT-167 syngeneic lung cancer model.
    Methods: Male C57BL6/J mice, aged 8-10 weeks, were administered an intramuscular (IM) injection of either 0.5x106 CMT-167 cells or vehicle. Clinically relevant features of cancer cachexia were assessed 23 days after CMT-167 cell administration in tumor bearing mice by assessment of terminal skeletal muscle and adipose tissue mass, gastrocnemius myofiber cross sectional area (CSA), circulating biomarkers of cachexia, and skeletal muscle E3 ubiquitin ligase mRNA. A single intravenous dose pharmacokinetic study of pembrolizumab was completed to assess tumor status influence upon antibody pharmacokinetics.
    Results: Compared to tumor free (TF) mice, we observed lower terminal tumor-adjusted bodyweight, adipose tissue mass, gastrocnemius mass, quadriceps mass, and gastrocnemius myofiber CSA. CMT-167 tumor bearing (TB) mice did not lose bodyweight relative to starting weight, but instead failed to gain as much weight as TF controls. CMT-167 TB mice exhibited increased concentrations of circulating markers of cachexia and muscle wasting, such as IL-6 and TNF-α, although there was no difference in transcription of E3 ubiquitin ligases Trim63 (MuRF-1) and Fbxo32 (atrogin-1) in skeletal muscle compared to TF mice. CMT-167 TB mice exhibited increased catabolic clearance (CL) of the human IgG4 anti-PD-1, pembrolizumab, agreeing with published literature showing increased CL of immune checkpoint inhibitors in cachectic populations [5, 6]. Comparing the IM CMT-167 model to historical data with the well-established IM Lewis Lung Carcinoma model, CMT-167 TB mice displayed a less severe cachectic phenotype in terms of bodyweight and skeletal muscle effects.
    Conclusion: The IM CMT-167 model is a syngeneic lung cancer model of mild cachexia. CMT-167 TB mouse is a novel model in which to study cancer cachexia induction, skeletal muscle atrophy, and immune checkpoint inhibitor clearance mechanisms in the context of lung cancer.
    Keywords:  Antibody therapies; Cancer Cachexia; Lung Cancer; Muscle Atrophy; Pharmacokinetics
    DOI:  https://doi.org/10.1002/rco2.117
  35. bioRxiv. 2025 Jun 17. pii: 2025.06.13.659540. [Epub ahead of print]
      Metabolic adaptation to fasting may have conferred survival advantage to early humans and predicts weight gain caused by overnutrition in modern societies. Fasting suppresses brown adipose tissue (BAT) thermogenesis; however, it is unclear how BAT rewires cellular metabolism to balance between energy conservation and heat generation. Here, we report that BAT in mice under fasting and cold challenge consumed ketone bodies, specifically acetoacetate (AcAc). Ablating liver ketogenesis decreased, while enhancing hepatic AcAc output defended, body temperature in mice facing the dual challenge. Using stable isotope tracing in brown adipocytes in vitro combined with quantitative analysis of metabolic fluxes and lipidomics in BAT from genetic mouse models, we disentangled the two metabolic fates of AcAc - terminal oxidation in the mitochondria and lipid biosynthesis in the cytosol. Notably, AcAc-sourced carbon preferentially supported polyunsaturated fatty acid synthesis in BAT, linking to the positive impact of intermittent fasting on lipid profiles in both mice and humans. Therefore, ketone body utilization by thermogenic adipocytes contributes to metabolic resilience of mammals and can be targeted to optimize benefits of dietary regimens.
    DOI:  https://doi.org/10.1101/2025.06.13.659540
  36. bioRxiv. 2025 May 01. pii: 2025.04.28.650893. [Epub ahead of print]
      Advances in mass spectrometry (MS)-based lipidomics have led to a surge in data volume, underscoring a need for robust tools to evaluate and visualize these data comprehensively. Current workflows are often hampered by manual spreadsheet handling and insufficient assessment of data quality prior to analysis. Here, we introduce LipidCruncher , an open-source, web-based platform designed to process, visualize, and analyze lipidomic data with high efficiency and rigor. LipidCruncher consolidates key steps of the workflow, including data standardization, normalization, and stringent quality control to identify anomalies. The platform also provides advanced visualization and analysis tools, such as volcano plots, lipid saturation profiles, pathway mapping, and lipid heatmaps, that enable detailed and holistic data exploration. To demonstrate LipidCruncher 's utility, we analyzed lipidomic data from adipose tissue of mice lacking the triacylglycerol synthesis enzymes DGAT1 and DGAT2. We anticipate that LipidCruncher will be a valuable and user-friendly tool for standardizing and analyzing lipidomics data.
    DOI:  https://doi.org/10.1101/2025.04.28.650893
  37. Cell. 2025 Jul 14. pii: S0092-8674(25)00730-5. [Epub ahead of print]
      Fluorescent genetically encoded voltage indicators report transmembrane potentials of targeted cell types. However, voltage-imaging instrumentation has lacked the sensitivity to track spontaneous or evoked high-frequency voltage oscillations in neural populations. Here, we describe two complementary TEMPO (transmembrane electrical measurements performed optically) voltage-sensing technologies that capture neural oscillations up to ∼100 Hz. Fiber-optic TEMPO achieves ∼10-fold greater sensitivity than prior photometric voltage sensing, allows hour-long recordings, and monitors two neuron classes per fiber-optic probe in freely moving mice. With it, we uncovered cross-frequency-coupled theta- and gamma-range oscillations and characterized excitatory-inhibitory neural dynamics during hippocampal ripples and visual cortical processing. The TEMPO mesoscope images voltage activity in two cell classes across an ∼8-mm-wide field of view in head-fixed animals. In awake mice, it revealed sensory-evoked excitatory-inhibitory neural interactions and traveling gamma and 3-7 Hz waves in visual cortex and bidirectional propagation directions for both hippocampal theta and beta waves. These technologies have widespread applications probing diverse oscillations and neuron-type interactions in healthy and diseased brains.
    Keywords:  beta oscillations; fluorescence imaging; gamma oscillations; local field potentials; neural dynamics; sharp wave ripples; theta oscillations; voltage imaging; voltage indicators; voltage waves
    DOI:  https://doi.org/10.1016/j.cell.2025.06.028
  38. Animal Model Exp Med. 2025 Jul 15.
       BACKGROUND: Cancer-associated cardiac cachexia (CACC) refers to cardiac injury in cancer patients in a malignant state, but preclinical animal models remain inadequately developed.
    METHODS: This study established CACC models in C57BL/6J and BALB/c mice using orthotopic, intra-abdominal, and hematogenous metastatic tumor induction. Multimodal cardiac assessments, including echocardiography, transmission electron microscopy for myocardial ultrastructural and mitochondrial analysis, and ex vivo cardiomyocyte contractility assays, were systematically applied.
    RESULTS: Metastatic burden triggered CACC characterized by cardiac mass reduction, epicardial fat depletion, interstitial fibrosis, and electrocardiographic abnormalities. Histopathological analysis revealed cardiomyocyte atrophy, myofibrillar disarray, mitochondrial dysfunction, and ubiquitin-mediated Myh6 degradation via MuRF-1, accompanied by compensatory Myh7 upregulation. These findings mechanistically link tumor-induced cachexia to cardiac dysfunction through contractile protein remodeling.
    CONCLUSION: This work establishes a preclinical framework for targeting ubiquitin pathways to mitigate the morbidity of cancer-related cardiopathy. Our integrated approach delineates a hierarchical progression from subcellular dysfunction to macroscopic cardiac deterioration.
    Keywords:  cachexia; cardiac atrophy; contractility; myocardial myosin; onco‐cardiology
    DOI:  https://doi.org/10.1002/ame2.70060
  39. Nano Lett. 2025 Jul 14.
      The discovery of nanomaterials with enzyme-like activities, termed nanozymes, holds the potential to revolutionize traditional inhibitor-based therapies. However, precise identification of nanozymes for specific substrates and uncovering their catalytic domains remain significant challenges. Here, we developed a "fishing" method that utilizes AFM probes coated with substrate baits, functioning as fishing rods to enable in situ visualization of enzymatic reactions on nanomaterial surfaces. Through this approach, we identified four nanozymes, including graphene oxides (GOs) exhibiting lipoxygenase (LOX)-like activity. Notably, mesopores were identified as key catalytic domains on GO surfaces, guiding the engineering of tailored porous GOs (tp-GOs) with enhanced LOX-like activity. The optimized tp-GOs catalyzed ferroptosis in cancer cells and suppressed hepatic tumor growth and metastasis in mice. Our findings open new avenues in drug discovery, shifting the paradigm from enzyme inhibition to promoting specific biochemical reactions using nanozymes.
    Keywords:  Artificial Enzyme; Catalysis; Cell death; Ferroptosis; Nanomedicine; Reactive oxidative stress
    DOI:  https://doi.org/10.1021/acs.nanolett.5c02476
  40. Genes Dev. 2025 Jul 11.
      Mitochondria are no longer viewed solely as ATP- or metabolite-generating organelles but as key regulators of cellular signaling that shape physiologic aging. Contrary to earlier theories linking aging to mitochondrial DNA mutations and oxidative damage, current evidence shows that these factors do not causally limit physiologic aging. Instead, an evolving literature links age-related loss of mitochondrial signaling and function to important physiologic changes of aging. Moreover, mild inhibition of mitochondrial respiratory function with drugs like metformin promote health span. These findings open new paths for pharmacologically reprogramming mitochondrial signaling to extend healthy aging.
    Keywords:  aging; mitochondria; senescence
    DOI:  https://doi.org/10.1101/gad.353106.125
  41. bioRxiv. 2025 Jun 16. pii: 2025.06.11.659168. [Epub ahead of print]
      Protein quality control (PQC) systems are essential for cellular resilience to proteotoxic stress. Despite intensive study for decades, functional redundancies in the system obscure the contributions of the collectively important individual genes. Here, we leverage transposon sequencing across bacteria strains lacking key chaperones and proteases to reveal hidden determinants of stress response in protein homeostasis. By profiling fitness under multiple proteotoxic stresses, we uncover stress-specific vulnerabilities and reveal how major players of PQC mask correlations between transcriptomic responses and gene fitness. As an illustration of unexpected connections, we identify a heat-specific synthetic lethality between the disaggregase ClpB and DNA Polymerase I (PolA) mediated by persistent aggregation of the RecA recombinase and toxic persistence of the heat shock regulon. Our findings reveal that stress-induced aggregation is not broadly toxic. Rather, it becomes lethal in specific genetic or environmental contexts due to the depletion of components only needed in those specific circumstances. This work presents a framework to reveal normally hidden fragility in stress responses using gene fitness scores adaptable to a variety of systems.
    DOI:  https://doi.org/10.1101/2025.06.11.659168