bims-pideca Biomed News
on Class IA PI3K signalling in development and cancer
Issue of 2025–11–30
34 papers selected by
Ralitsa Radostinova Madsen, MRC-PPU



  1. Nat Commun. 2025 Nov 28. 16(1): 10731
      Most Epstein-Barr virus-associated gastric carcinoma (EBVaGC) harbor non-silent mutations that activate phosphoinositide 3 kinase (PI3K) to drive downstream metabolic signaling. To gain insights into PI3K/mTOR pathway dysregulation in this context, we perform a human genome-wide CRISPR/Cas9 screen for hits that synergistically blocked EBVaGC proliferation together with the PI3K antagonist alpelisib. Multiple subunits of carboxy terminal to LisH (CTLH) E3 ligase, including the catalytic MAEA subunit, are among top screen hits. CTLH negatively regulates gluconeogenesis in yeast, but not in higher organisms. The CTLH substrates MKLN1 and ZMYND19, which highly accumulated upon MAEA knockout, associate with one another and with lysosome outer membranes to inhibit mTORC1. Rather than perturbing mTORC1 lysosomal recruitment, ZMYND19 and MKLN1 block the interaction between mTORC1 and Rheb and also with mTORC1 substrates S6 and 4E-BP1. Thus, CTLH enables cells to rapidly tune mTORC1 activity at the lysosomal membrane via the ubiquitin/proteasome pathway.
    DOI:  https://doi.org/10.1038/s41467-025-65760-6
  2. Nat Commun. 2025 Nov 28. 16(1): 10761
      Lysosomes are essential organelles that regulate cellular homeostasis through complex membrane interactions. Phosphoinositide lipids play critical roles in orchestrating these functions by recruiting specific proteins to organelle membranes. The PIKfyve/Fig4/Vac14 complex regulates PI(3,5)P₂ metabolism, and intriguingly, while loss-of-function mutations cause neurodegeneration, acute PIKfyve inhibition shows therapeutic potential in neurodegenerative disorders. We demonstrate that PIKfyve/Fig4/Vac14 dysfunction triggers a compensatory response where reduced mTORC1 activity leads to ULK1-dependent trafficking of ATG9A and PI4KIIα from the TGN to lysosomes. This increases lysosomal PI(4)P, facilitating cholesterol and phosphatidylserine transport at ER-lysosome contacts to promote membrane repair. Concurrently, elevated lysosomal PI(4)P recruits ORP1L to ER-lysosome-mitochondria three-way contacts, enabling PI(4)P transfer to mitochondria that drives ULK1-dependent fragmentation and increased respiration. These findings reveal a role for PIKfyve/Fig4/Vac14 in coordinating lysosomal repair and mitochondrial homeostasis, offering insights into cellular stress responses.
    DOI:  https://doi.org/10.1038/s41467-025-65798-6
  3. Children (Basel). 2025 Oct 28. pii: 1460. [Epub ahead of print]12(11):
      Background: Lymphatic malformations (LM) are rare congenital vascular anomalies caused by abnormal development and growth of lymphatic vessels. These malformations can lead to a wide range of symptoms, from mild swelling to more severe complications. Treatment options remain limited, especially for complex cases. Recent research has suggested that PIK3CA mutations play a key role in the pathogenesis of LM, potentially offering new possibilities for targeted treatment strategies. Methods: In this study, a cohort of 36 patients diagnosed with LM, Klippel-Trenaunay syndrome (KTS), and Proteus syndrome was analyzed. PIK3CA mutations were assessed in tissue samples obtained from the LM during clinically indicated procedures using digital droplet polymerase chain reaction (ddPCR), targeting five hotspots. Results: PIK3CA mutations were found in 18 patients (50%). The most frequent mutation was p.E542K (c.1624G>A), found in 19.44% of patients, followed by p.H1047R (c.3149A>G), p.E545K (c.1633G>A), and p.H1047L (c.3140A>T) each occurring in 11.11% of the cases. Mutations were more common in isolated LMs, with 63.16% of patients exhibiting PIK3CA mutations. Conclusions: PIK3CA mutations are common in LM, supporting the potential for targeted therapies like PI3K inhibitors in treating complex cases. This research highlights the importance of genetic analysis in the management of LM and offers a new therapeutic approach.
    Keywords:  PIK3CA mutation; lymphatic malformation; pediatrics; targeted therapy; vascular malformation
    DOI:  https://doi.org/10.3390/children12111460
  4. bioRxiv. 2025 Nov 11. pii: 2025.11.11.687767. [Epub ahead of print]
      Imaging-based CRISPR screens enable high-content functional genomics by capturing phenotypic changes in cells after genetic perturbation. Protein barcodes provide cost-effective, easy-to-implement, and imaging-compatible barcoding for pooled perturbations, yet their scalability has been constrained by the need for arrayed cloning, lentiviral recombination between barcodes and guides, and difficulties in decoding barcodes with high confidence. Here, we introduce poolVis and cellPool, an integrated experimental and computational platform designed to address these limitations. poolVis uses Cre-lox-mediated reconfiguration to position barcode-sgRNA pairs in proximity during viral integration, which greatly reduces barcode shuffling during pooled cloning and delivery. cellPool leverages a scalable computational workflow and the unique aspects of protein barcodes to produce unpooled image galleries from multi-terabyte scale datasets. Applying this platform to single- and double-CRISPRi profiling of cell-cycle genes and chromokinesins in the MCF10A cells uncovered established and previously unrecognized phenotypes, including nuclear morphology changes and reciprocal sign epistasis in DNA damage.
    DOI:  https://doi.org/10.1101/2025.11.11.687767
  5. JCI Insight. 2025 Nov 24. pii: e187448. [Epub ahead of print]10(22):
      Therapeutics blocking PI3K/mTOR complex 1 (mTORC1) are commonly used for tumor treatment, and at times achieve major responses, yet minimal residual disease (MRD) persists, leading to tumor relapse. We developed multiple MRD models both in vitro (rapamycin persistent, RP) and in vivo after mTORC1 inhibition. All 11 RP/MRD cell lines showed complete growth and signaling insensitivity to rapamycin but variable sensitivity to bi-steric mTORC1 inhibitors, with MtorS2035 mutations identified in 4 of 7 RP cell lines. Multiomic analyses identified a pronounced shift toward oxidative phosphorylation and away from glycolysis with increased mitochondrial number in all RP/MRD models. MYC and SWI/SNF expression was significantly enhanced. Both the SWI/SNF inhibitor AU-15330 and the mitochondrial complex I oxidative phosphorylation inhibitor IACS-010759 showed pronounced synergy with bi-steric mTORC1 inhibitors to cause cuproptotic cell death in RP/MRD cells, suggesting these combinations as a potential patient treatment strategy for rapalog resistance.
    Keywords:  Cancer; Epigenetics; Metabolism; Mitochondria; Oncology
    DOI:  https://doi.org/10.1172/jci.insight.187448
  6. bioRxiv. 2025 Oct 25. pii: 2025.10.24.684471. [Epub ahead of print]
      Cells rely on precise metabolic control to adapt to environmental cues. The mechanistic target of rapamycin complex 1 (mTORC1) senses nutrient availability, with amino acids serving as key signals. Lysosomes, which act as nutrient recycling centers, maintain amino acid homeostasis by breaking down macromolecules and releasing amino acids for cellular use. SLC38A9, a lysosomal amino acid transporter, functions as both a transporter and a sensor in the mTORC1 pathway. Here, we investigated whether SLC38A9 activity is regulated by pH. We show that arginine uptake by SLC38A9 is pH-dependent, and that the histidine residue His544 serves as the pH sensor. Mutating His544 abolishes the pH dependence of arginine uptake without impairing overall transport activity, indicating that His544 is not directly involved in substrate binding. Instead, protonation or deprotonation of His544 appears to influence transport through SLC38A9. To explore this mechanism, we compared two structures of SLC38A9 that we determined, one at high pH and one at low pH, and proposed a working model for pH-induced activation. These findings highlight the role of local ionic changes in modulating lysosomal transporters and underscore the intricate regulatory mechanisms that govern SLC38A9 function and, ultimately, mTORC1 signaling.
    DOI:  https://doi.org/10.1101/2025.10.24.684471
  7. Science. 2025 Nov 27. eadv7111
      The mTOR protein kinase forms two multiprotein complexes, mTORC1 and mTORC2, that function in distinct signaling pathways. mTORC1 is regulated by nutrients, and mTORC2 is a central node in phosphoinositide-3 kinase (PI3K) and small guanosine triphosphate Ras signaling networks commonly deregulated in cancer and diabetes. Although mTOR phosphorylates many substrates in vitro, in cells, mTORC1 and mTORC2 have high specificity: mTORC2 phosphorylates the protein kinases Akt and PKC, but not closely related kinases that are mTORC1 substrates. To understand how mTORC2 recognizes substrates, we created semisynthetic probes to trap the mTORC2-Akt complex and determine its structure. Whereas most protein kinases recognize amino acids adjacent to the phosphorylation site, local sequence contributes little to substrate recognition by mTORC2. Instead, the specificity determinants were secondary and tertiary structural elements of Akt that bound the mTORC2 component mSin1 distal to the mTOR active site and were conserved amongst at least 18 related substrates. These results reveal how mTORC2 recognizes its canonical substrates and may enable the design of mTORC2-specific inhibitors.
    DOI:  https://doi.org/10.1126/science.adv7111
  8. bioRxiv. 2025 Oct 30. pii: 2024.12.06.627299. [Epub ahead of print]
      Cell morphology and subcellular protein organization provide important insights into cellular function and behavior. These features of cells can be studied using large-scale protein fluorescence microscopy, and machine learning has become a powerful tool to interpret the resulting images for biological insights. Here, we introduce SubCell, a suite of self-supervised deep learning models for fluorescence microscopy designed to accurately capture cellular morphology, protein localization, cellular organization, and biological function beyond what humans can readily perceive. These models were trained on the proteome-wide image collection from the Human Protein Atlas with a novel proteome-aware learning objective. SubCell outperforms state-of-the-art methods across a variety of tasks relevant to single-cell biology and generalizes to other fluorescence microscopy datasets without any fine-tuning. Additionally, we construct the first proteome-wide hierarchical map of proteome organization that is directly learned from image data. This vision-based multiscale cell map defines cellular subsystems with high resolution of protein complexes, reveals proteins with similar functions, and distinguishes dynamic and stable behaviors within cellular compartments. Finally, Subcell enables a rich multimodal protein representation when integrated with a protein sequence model, allowing for a more comprehensive capture of gene function than either vision-only or sequence-only models alone. In conclusion, SubCell creates deep, image-driven representations of cellular architecture that are applicable across diverse biological contexts and datasets.
    DOI:  https://doi.org/10.1101/2024.12.06.627299
  9. Commun Biol. 2025 Nov 22.
      Mass spectrometry-based phosphoproteomic workflows enable routine study of signalling dynamics in contexts where cells are abundant. However, the analysis of cell signalling governed by phosphorylation requires an additional enrichment step which poses a significant challenge when dealing with limited material. The development of sensitive techniques that allow phosphoproteomic analysis of a few hundred cells is increasingly enabling researchers to disambiguate the complexity of protein signalling networks within heterogeneous cell populations. The imminent task at hand is to apply these techniques in research contexts guided by the available biological material to address complex questions about cellular function. Examples range from characterising differential treatment responses in distinct cell populations to investigating rare cell types from primary patient material or in vivo models. To achieve this, adapted protocols need to consider appropriate isolation of specific cells, simplified sample processing to avoid losses, labelling and multiplexing, and optimised analytical methodologies. Here, we discuss these aspects of the workflow, highlighting how innovations from low-input and single-cell proteomics can be adapted to drive low-input phosphoproteomics forward.
    DOI:  https://doi.org/10.1038/s42003-025-09131-3
  10. bioRxiv. 2025 Oct 06. pii: 2025.10.05.680536. [Epub ahead of print]
      Identifying the drivers of cellular senescence that contribute to the decline in tissue function related to aging- and disease is critical for developing restorative interventions. Here, we investigated how increased mechanical stress from extracellular matrix (ECM) stiffening shapes endothelial cell (EC) senescence. We developed a 3D human in vitro model that decouples mechanical stress from inflammatory or biochemical inputs, enabling the study of senescence responses to tissue stiffening alone. We found that matrix stiffening induces an EC senescence phenotype with elevated p16/p21 and an immunomodulatory senescence-associated secretory phenotype (SASP), in the absence of inflammatory signals. This mechano-induced senescence state engaged a Notch-JNK-FOS signaling axis, and pharmacologic inhibition of Notch attenuated stiffness-induced senescence. Supporting the translational relevance of this mechanism, analysis of fibrotic capsule tissue from patients with synthetic breast implants, a model of localized, mechanically driven fibrosis, revealed increased p16 + Notch1 + endothelial populations. Complementary single-cell RNA sequencing data confirmed their enrichment in Notch/JNK- and SASP-related gene programs. Together, these findings define vascular senescence as a mechanosensitive process and identify tissue stiffening as an upstream aging signal. Our work offers a human-relevant platform for studying targetable stages of endothelial mechanoaging.
    DOI:  https://doi.org/10.1101/2025.10.05.680536
  11. bioRxiv. 2025 Nov 05. pii: 2025.11.04.686676. [Epub ahead of print]
      High-throughput pooled screening has advanced functional genomics, but most existing methods rely on endpoint sequencing and are blind to dynamic, time-resolved phenotypes. We developed RainBar (Rainbow Barcodes), an optical barcoding system that supports pooled live-cell imaging with single-cell resolution. RainBar uses lentiviral co-delivery of spectrally distinct nuclear and cytoplasmic fluorescent proteins to encode up to 64 unique perturbations per well. To mitigate barcode recombination and improve decoding accuracy, we employed single-template viral production, codon diversification, and a ratio-based spectral unmixing pipeline tailored to overlapping fluorophores. An inverted cytoplasmic segmentation approach and multilayer perceptron classifier enabled accurate barcode identification in both arrayed and pooled formats. As a proof of concept, we applied RainBar to dissect NF-κB signaling dynamics in epithelial cells. Live imaging of RelA translocation uncovered stimulus-specific kinetics: IL-1β triggered rapid recovery, while TNF induced prolonged nuclear localization. In pooled CRISPRi screens, RainBar recovered known NF-κB regulators (e.g., IL1R1, MYD88, TNFRSF1A) and highlighted additional modulators, including the Ino80 chromatin remodeling complex subunits and KAT2A acetyltransferase. Together, these results position RainBar as a flexible platform for multiplexed, image-based functional genomics, with potential to reveal dynamic signaling architectures across diverse cellular contexts in live cells.
    DOI:  https://doi.org/10.1101/2025.11.04.686676
  12. Am J Med Genet C Semin Med Genet. 2025 Nov 27.
      Vascular anomalies represent a broad spectrum of disorders characterized by aberrant blood or lymphatic vessel development, which can lead to complex clinical phenotypes. Historically, vascular anomalies were classified solely on the basis of their clinical and histopathologic features. However, the last two decades have witnessed significant advances in our understanding of the genetic basis of these lesions. It is now recognized that many vascular anomalies arise from somatic pathogenic variants in key growth signaling pathways, including the PI3K-AKT-mTOR and RAS-MAPK pathways. These insights have catalyzed the development of targeted therapies designed to address the molecular underpinnings of disease. mTOR inhibitors, originally developed and widely used as anticancer agents, have also demonstrated significant efficacy in improving outcomes for patients with low-flow vascular malformations such as lymphatic malformations and venous malformations. Similarly, MEK inhibitors and other oncology drugs are being repurposed as promising therapeutic options for complex lymphatic anomalies and arteriovenous malformations, conditions that historically have had limited medical therapeutic options. Clinical trials for vascular anomalies are emerging, but questions remain about how to best measure response in these patients, as well as the optimal duration of treatment. This case-based review explores recent developments in precision medicine for vascular anomalies, highlighting a paradigm shift in the management of these complex and often therapeutically challenging disorders.
    Keywords:  PIK3CA‐related overgrowth spectrum; alpelisib; arteriovenous malformation; sirolimus; vascular anomalies
    DOI:  https://doi.org/10.1002/ajmgc.70000
  13. bioRxiv. 2025 Nov 07. pii: 2025.11.06.686987. [Epub ahead of print]
      Cell state plasticity drives metastasis and therapy resistance in cancers. In melanoma, these behaviors map onto a melanocytic-to-mesenchymal-like continuum regulated by AP-1 transcription factors. However, how the AP-1 network encodes a limited set of discrete states, why their distributions vary across tumors, and what drives phenotypically consequential AP-1 state transitions remain unclear. We develop a mechanistic ODE model of the AP-1 network capturing their dimerization-controlled, co-regulated, competitive interactions. Calibrated to heterogeneous single-cell data across genetically diverse melanoma populations and combined with statistical learning, the model reveals network features explaining population-specific AP-1 state distributions. These features correlate with MAPK activity across tumor lines and with variability within clones, linking MAPK signaling to AP-1 states. The model predicts and experiments validate adaptive AP-1 reconfiguration under MAPK inhibition, inducing a dedifferentiated, therapy-resistant state that can be blocked by model-guided AP-1 perturbations. These results establish AP-1 as a configurable network and provide a computational framework for predicting and modulating AP-1 driven cell state plasticity.
    DOI:  https://doi.org/10.1101/2025.11.06.686987
  14. bioRxiv. 2025 Oct 12. pii: 2025.10.11.681838. [Epub ahead of print]
      We map human artery and vein endothelial cell (EC) differentiation from pluripotent stem cells, and employ this roadmap to discover new mechanisms of vascular development (vein differentiation) and disease (viral infection). We discovered vein development unfolds in two steps driven by opposing signals: VEGF differentiates mesoderm into "pre-vein" ECs, but surprisingly, VEGF/ERK inhibition subsequently specifies vein ECs. Pre-vein ECs co-expressed certain arterial ( SOX17 ) and venous ( APLNR ) markers, harbored poised chromatin at future venous genes, but completed venous differentiation only upon VEGF inhibition. Intersectional lineage tracing revealed that early Sox17 + Aplnr + ECs also formed veins in vivo . Next, we compared how Ebola, Andes, and Nipah viruses infect artery and vein ECs under biosafety-level-4 containment. Each virus distinctly affected ECs. Interestingly, artery and vein ECs also responded divergently to the same virus, thus revealing that developmentally-specified cell identity impacts viral infection. Collectively, this arteriovenous differentiation roadmap illuminates vascular development and disease.
    DOI:  https://doi.org/10.1101/2025.10.11.681838
  15. Proc Natl Acad Sci U S A. 2025 Dec 02. 122(48): e2508893122
      Skeletal muscle is essential for movement, respiration, and metabolism, with mTORC1 acting as a key regulator of protein synthesis and degradation. In aging muscle, mTORC1 becomes overactivated, contributing to sarcopenia, though the mechanisms remain unclear. Here, we identify DEAF1, a FOXO-regulated transcription factor, as a key upstream driver of mTORC1 in aged muscle. Elevated Deaf1 expression increases mTOR transcription, leading to heightened mTORC1 activity, impaired proteostasis, and muscle senescence. Remarkably, exercise suppresses Deaf1 expression via FOXO activation, restoring mTORC1 balance and alleviating muscle aging. Conversely, FOXO inhibition or Deaf1 overexpression blocks exercise benefits on muscle health. These findings highlight DEAF1 as a critical link between FOXO and mTORC1 and suggest that targeting the FOXO-DEAF1-mTORC1 axis may offer therapeutic potential to preserve muscle function during aging.
    Keywords:  autophagy; mTORC1; muscle; proteostasis; sarcopenia
    DOI:  https://doi.org/10.1073/pnas.2508893122
  16. bioRxiv. 2025 Oct 31. pii: 2025.10.31.685785. [Epub ahead of print]
      Fluorescent in situ sequencing involves imaging-based sequencing by synthesis in intact cells or tissues to reveal target nucleotide sequences inside each cell. Often, the target sequences are barcodes that indicate a perturbation (e.g. CRISPR guide or genetic variant) delivered to the cell. However, processing in situ sequencing data presents a considerable challenge, requiring stitching and aligning tens of thousands of images with millions of cells, detecting small amplicon colonies across sequencing cycles, and calling reads. To address these challenges, we introduce STARCall: STitching, Alignment and Read Calling for in situ sequencing, a software package that analyzes raw in situ sequencing images to produce a genotype-to-phenotype mapping for each cell. STAR-Call improves upon previous solutions by combining stitching and alignment of images into a single step that minimizes both inter-cycle and intra-cycle alignment error. STARCall also improves detection and extraction of sequencing reads, incorporating filters and normalization to combat background fluorophore signal. We compare STARCall to other methods using a diverse set of images that include commonly encountered imaging problems such as variable intensity across channels and cycles and high levels of background. Specifically, this comprises ∼250,000 images from a pooled screen of ∼3,500 barcoded LMNA variants expressed in U2OS cells and ∼1,200 bar-coded PTEN variants in induced pluripotent stem cells (iPSC) and iPSC-derived neurons. Overall, STARCall aligned more than 50% of tiles with <1 pixel error on all nine image sets while alternative packages had higher error on four. STARCall also yielded an 8-40% increase in genotyped cells due to improved filtering and normalization methods that address background fluorescence. STARCall can call tools like CellPose to segment cells and CellProfiler to compute cell features from the phenotyping images. STARcall is open-source and freely available, providing a robust solution for the analysis of in situ sequencing data.
    Author summary: Short regions of RNA or DNA can be sequenced inside intact cells or tissues (i.e. in situ ) by combining a microscope and sequencing by DNA synthesis. Multiple cycles of sequencing are performed, in which incorporation of a single fluorescently labeled nucleotide is imaged, and the corresponding base detected. Recently, in situ sequencing has proved useful in optical pooled screens, where a library of perturbations such as CRISPR-mediated gene knockouts or genetic variants is introduced into cells, and in situ sequencing is used to reveal the specific perturbation in each cell. However, processing in situ sequencing data presents a considerable challenge, requiring stitching and aligning tens of thousands of images, detecting small amplicon colonies across sequencing cycles, and calling reads. To address these challenges, we introduce STARCall: STitching, Alignment and Read Calling for in situ sequencing. STARCall uses a novel stitching algorithm that minimizes both inter-cycle and intra-cycle alignment error, and improved filters and normalization for base calling. When applied to a set of 9 in situ sequencing image sets, STARCall yielded an 8-40% increase in genotyped cells. STARCall is open-source and applicable to a variety of experiments, providing a robust pipeline for in situ sequencing data.
    DOI:  https://doi.org/10.1101/2025.10.31.685785
  17. Int J Mol Sci. 2025 Nov 12. pii: 10932. [Epub ahead of print]26(22):
      Cancer cells can sustain survival independently of exogenous growth factors. To investigate their adaptation to serum deprivation, we analyzed transcriptomic responses in two cancer cell lines. Transcriptome analysis revealed upregulation of mRNAs encoding cholesterol biosynthesis enzymes. This was a critical adaptive response, as a pharmacological inhibition of the pathway with statin triggered a robust apoptotic cell death accompanied by generation of a mitochondrial reactive oxygen species. The mechanistic target of rapamycin complex 1 (mTORC1), a master regulator of cell growth, is known to be engaged in controlling lipid biosynthesis. We detected the high polysomal and preribosomal peaks not only in serum-containing medium but also under serum deprivation, indicating a high rate of protein synthesis and ribosomal biogenesis independent of serum. In addition, the inhibition of mTOR kinase activity substantially reduced polysome abundance, with a more pronounced effect in serum-deprived cancer cells. Notably, the mTOR kinase inhibition also prevented the upregulation of the cholesterol synthesis enzyme that established a direct link between mTOR activity, protein synthesis, and cholesterol biosynthesis. Together, our results show that cancer cells adapt to serum withdrawal by activating the cholesterol synthesis pathway through mTOR-dependent regulation of gene expression and protein synthesis, underscoring a critical mechanism of survival under serum withdrawal.
    Keywords:  apoptosis; cholesterol synthesis; mechanistic target of rapamycin complex 1 (mTORC1); polysomes; protein synthesis; reactive oxygen species; serum deprivation
    DOI:  https://doi.org/10.3390/ijms262210932
  18. bioRxiv. 2025 Oct 13. pii: 2025.10.12.681903. [Epub ahead of print]
      Growth factor induced receptor dimerization and activation of downstream pathways can modulate cell fate decisions. Here, we investigate the potential of de novo designed synthetic ligands, termed Novokines, to reprogram cell identity by inducing proximity of novel pairs of receptor subunits. We find that a design, H2F, that brings together HER2 (which has no known natural ligand) and the FGF receptor has potent signaling activity. H2F induces robust signaling and reprograms fibroblasts into myogenic cells. Unlike native FGF ligands, H2F selectively activates the MAPK pathway without engaging PLCγ-mediated Ca²⁺ signaling. FRET assays confirm H2F-mediated HER2-FGFR proximity, and phosphoproteomic analysis reveals activation of MAPK effectors. H2F-induced ERK phosphorylation is abolished in cells expressing a kinase-dead FGFR1 (K514M) mutant, confirming the requirement for FGFR catalytic activity. H2F treatment significantly increases myofiber formation from adult patient-derived primary myoblasts, demonstrating its capacity to promote myogenic regeneration. Our findings demonstrate that synthetic receptor pairings can rewire signaling outputs to drive regeneration, providing a programmable platform for cell fate engineering.
    DOI:  https://doi.org/10.1101/2025.10.12.681903
  19. Nature. 2025 Nov 26.
      Targeted protein degradation is a pharmacological strategy that relies on small molecules such as proteolysis-targeting chimeras (PROTACs) or molecular glues, which induce proximity between a target protein and an E3 ubiquitin ligase to prompt target ubiquitination and proteasomal degradation1. Sporadic reports indicated that ligands designed to inhibit a target can also induce its destabilization2-4. Among others, this has repeatedly been observed for kinase inhibitors5-7. However, we lack an understanding of the frequency, generalizability and mechanistic underpinnings of these phenomena. Here, to address this knowledge gap, we generated dynamic abundance profiles of 98 kinases after cellular perturbations with 1,570 kinase inhibitors, revealing 160 selective instances of inhibitor-induced kinase destabilization. Kinases prone to degradation are frequently annotated as HSP90 clients, therefore affirming chaperone deprivation as an important route of destabilization. However, detailed investigation of inhibitor-induced degradation of LYN, BLK and RIPK2 revealed a differentiated, common mechanistic logic whereby inhibitors function by inducing a kinase state that is more efficiently cleared by endogenous degradation mechanisms. Mechanistically, effects can manifest by ligand-induced changes in cellular activity, localization or higher-order assemblies, which may be triggered by direct target engagement or network effects. Collectively, our data suggest that inhibitor-induced kinase degradation is a common event and positions supercharging of endogenous degradation circuits as an alternative to classical proximity-inducing degraders.
    DOI:  https://doi.org/10.1038/s41586-025-09763-9
  20. medRxiv. 2025 Nov 14. pii: 2025.11.12.25340127. [Epub ahead of print]
      Multiplexed assays of variant effect (MAVEs) systematically measure variant function but have been limited to cancer cell lines rather than disease-relevant cell types. We developed saturation genome editing in human iPSCs (iPSC-SGE) to introduce variant libraries into a single allele of a target gene while programming the genetic background of the second allele, enabling variant assessment across differentiated cell types and genetic contexts at scale. We edited 1,137 variants into MYBPC3 and measured protein abundance in cardiomyocytes and cardiac organoids, accurately identifying pathogenic variants, and resolving variants of uncertain significance. Highlighting the importance of genetic context, we edited 437 POLG variants in two genetic backgrounds and identified loss-of-function and dominant-negative variants. Finally, we illuminate a path for scaling iPSC-SGE by identifying 443 disease genes essential for iPSC or iPSC-derived neuron growth. iPSC-SGE enables systematic assessment of variants in specialized human cell types, advancing MAVEs to empower genomic medicine.
    DOI:  https://doi.org/10.1101/2025.11.12.25340127
  21. bioRxiv. 2025 Nov 08. pii: 2025.11.06.687062. [Epub ahead of print]
      Waddington's epigenetic landscape has served as biology's central metaphor for cellular differentiation for over half a century, depicting mature cell types as balls resting in stable valley floors. Boolean networks - introduced by Kauffman in 1969 to model gene regulatory dynamics - provide a mathematical formalization of this landscape, where attractors represent phenotypes and basins of attraction correspond to developmental valleys. Traditional stability measures quantify robustness by perturbing arbitrary states, yet biological systems typically reside at attractors rather than in transient states. Here we formalize and systematically analyze attractor coherence - a stability measure Kauffman originally envisioned but never rigorously developed - which quantifies how likely a perturbation of an attractor state causes phenotype switching. Analyzing 122 expertcurated biological Boolean models, we reveal a striking paradox: attractors representing mature cell types are consistently less stable than the developmental trajectories approaching them. Largescale simulations of random networks demonstrate that this coherence gap arises from canalization - a hallmark of biological regulation where individual genes can override others. While canalization increases overall network stability, it disproportionately stabilizes transient states, positioning attractors near basin boundaries. The gap's magnitude is almost perfectly predicted by network bias (Spearman's ρ = -0.997), itself modulated by canalization. These findings revise Waddington's landscape: canalization carves deep protective valleys ensuring developmental robustness, yet simultaneously flattens ridges near valley floors, facilitating phenotypic plasticity when multiple fates coexist. This explains how biological systems achieve both reliable development and plasticity, with implications for understanding development, disease-related transitions, and designing robust yet controllable synthetic gene circuits.
    DOI:  https://doi.org/10.1101/2025.11.06.687062
  22. bioRxiv. 2025 Nov 09. pii: 2025.11.08.687374. [Epub ahead of print]
      Understanding how individual genetic backgrounds shape the effects of disease-associated mutations is central to elucidating the biology of complex psychiatric disorders. We developed a scalable 'village editing' strategy that enables simultaneous genome editing across multiple induced pluripotent stem cell (iPSC) lines, allowing systematic assessment of how polygenic context modulates the impact of specific mutations. Using pooled CRISPR editing in 15 iPSC lines spanning a range of schizophrenia (SCZ) polygenic risk scores, we generated homozygous and heterozygous knockouts in two known SCZ-associated genes: LRP1 , involved in cholesterol import, and NRXN1 , a presynaptic adhesion molecule. By mixing all lines prior to editing and de-multiplexing them afterward, we efficiently produced multi-donor knockout neurons at scale. Transcriptomic profiling revealed that LRP1 and NRXN1 loss produce both shared and donor-specific effects on neuronal gene expression, with variable perturbation of neurotransmitter transport and cholesterol biosynthesis pathways across genetic backgrounds. These results demonstrate that village editing enables systematic dissection of gene-background interactions in human neurons, offering a powerful framework for studying the polygenic architecture of psychiatric disease.
    DOI:  https://doi.org/10.1101/2025.11.08.687374
  23. Nat Protoc. 2025 Nov 28.
      Understanding how cells sense and respond to mechanical forces is crucial for many biological processes, including adhesion, migration, differentiation and immune activation. In this protocol, we describe two advanced DNA-based tension probes, the reversible shearing DNA-based tension probe (RSDTP) and ForceChrono probe, which provide powerful tools for studying mechanotransduction in living cells. RSDTPs enable dynamic quantification of forces ranging from 4 to 60 pN, offering the advantage of reversibility without ligand depletion, making them ideal for ensemble force measurements across populations of cells. ForceChrono probes not only measure the magnitude of force but also capture its duration and loading rate, providing essential insights into the temporal dynamics of single-molecule force transmission. We detail the fundamental principles, design strategies and step-by-step procedures for synthesizing, purifying and applying these probes, including surface preparation, cell experiments, image acquisition and data analysis. In addition, we describe the computational tools for image analysis. Together, these probes enable a detailed analysis of cellular mechanobiology, with applications in integrin mechanobiology and cell adhesion biology. This protocol is suitable for researchers with a background in cell biology, molecular biology, surface chemistry, optical imaging and data analysis and can be completed by a graduate student in 3-4 days.
    DOI:  https://doi.org/10.1038/s41596-025-01277-y
  24. Oncogene. 2025 Nov 25.
      EGFR family receptor tyrosine kinase signaling is commonly dysregulated in cancer by amplification or activating mutations. Although studies have investigated dual EGFR/PI3K inhibition in breast cancer, they have not determined biomarkers which predict success. We present evidence of a patient subset with EGFR amplification and PI3Kinase pathway mutations in breast cancer which can be synergistically targeted by dual EGFR/PI3K inhibition. This study identified that EGFR amplification occurs in ~1-5% of breast cancer patients with shorter overall survival compared to unamplified patients. Up to 71% of EGFR amplified tumors have activating mutations in the PI3K pathway. Dual EGFR/PI3K inhibition more dramatically reduced mTOR and AKT signaling in BT20 and MDA-MB-468 cells which both have EGFR amplification and PI3K pathway activating mutations, compared to control cells. Dual inhibition synergistically reduced cell viability and increased apoptosis in MDA-MB-468 and BT20 compared to control. Single agent therapy in a BT20 xenograft model reduced tumor volume, however only the combination statistically significantly reduced tumor volume compared to control. We conclude that EGFR amplification with co-incident PI3K pathway mutations are driver mutations in a subset of breast cancers and present a subgroup of breast cancers that are more likely to respond to dual targeted therapy.
    DOI:  https://doi.org/10.1038/s41388-025-03634-3
  25. bioRxiv. 2025 Nov 06. pii: 2025.09.17.676780. [Epub ahead of print]
      Cell state transitions underlie the emergence of diverse cell types and are traditionally defined by changes in gene expression. Yet these transitions also involve coordinated shifts in cell morphology and behaviour, which remain poorly characterized in densely packed epithelia. We developed a quantitative live-imaging and computational framework to track thousands of individual cells over time in the rapidly differentiating Xenopus mucociliary epithelium (MCE). From segmentations and trajectories, we extracted dynamic features-cell and nuclear shape, movement, position-to create a time-resolved morphodynamic dataset spanning the full course of differentiation. While single features showed high noise and low separability of ground-truth cell types, supervised machine learning revealed that integrating time-resolved features robustly predicts final cell fate. Gradient-boosted trees and multinomial logistic regression achieved moderate but consistent accuracy, especially for abundant epithelial lineages. Key discriminants included normalized Z-position, membrane-nucleus offset, and absolute experimental time, whereas movement contributed minimally to the results. Our results show that morphodynamic signatures encode predictive information about cell identity and provide a framework linking physical dynamics with molecular state.
    DOI:  https://doi.org/10.1101/2025.09.17.676780
  26. Nature. 2025 Nov 24.
      The colorectal epithelium is rapidly renewing, with remarkable capacity to regenerate following injury. In colorectal cancer (CRC), this regenerative capacity can be co-opted to drive epithelial plasticity. While oncogenic MAPK signalling in CRC is common, with frequent mutations of both KRAS (40-50%) and BRAF (10%)1, inhibition of this pathway typically drives resistance clinically. Given the development of KRAS inhibitors, and licensing of BRAF inhibitor combinations2-4, we have interrogated key mechanisms of resistance to these agents in advanced preclinical CRC models. We show that oncogenic MAPK signalling induces epithelial state changes in vivo, driving adoption of a regenerative/revival stem like population, while inhibition leads to rapid transcriptional remodeling of both Kras- and Braf-mutant tumours, favoring a Wnt-associated, canonical stem phenotype. This drives acute therapeutic resistance in Kras- and delayed resistance in Braf-driven models. Importantly, where plasticity is restrained, such as in early metastatic disease, or through targeting ligand-dependent Wnt-pathway Rnf43 mutations, marked therapeutic responses are observed. This explains the super response to BRAF+EGFR targeted therapies previously observed in a BRAF/RNF43 co-mutant patient population, highlighting the criticality of cellular plasticity in therapeutic response. Together, our data provides clear insight into the mechanisms underpinning resistance to MAPK targeted therapies in CRC. Moreover, strategies that aim to corral stem cell fate, restrict epithelial plasticity or intervene when tumours lack heterogeneity may improve therapeutic efficacy of these agents.
    DOI:  https://doi.org/10.1038/s41586-025-09916-w
  27. bioRxiv. 2025 Nov 14. pii: 2025.11.13.688264. [Epub ahead of print]
      The advent of spatial omics has revolutionized our understanding of tissue biology; however, these technologies remain largely descriptive and do not capture how changes in gene regulation propagate across spatial neighborhoods. While in-silico perturbation methods and foundation models aim to model the impact of genetic perturbations, these methods are limited to single-cell approaches that lack spatial resolution. Other studies can delineate morphological domains based on transcriptional similarity, but not spatial functional microniches. We address this major unmet need by developing SpaceTravLR (Spatially perturbing Transcription factors, Ligands and Receptors), a novel interpretable machine learning approach that generalizes across tissues and species, uncovering spatial features linked to functional outcomes, thereby capturing functional microniches with spatial resolution. SpaceTravLR infers how single or combinatorial genetic perturbations rewire signals across the tissue neighborhood, by propagating effects through underlying spatially resolved molecular networks, thereby modeling how perturbations can reshape both the targeted cell and its surrounding neighborhood. SpaceTravLR defines novel spatial microniches across a range of tissues at different scales of organization (niches, neighborhoods and tissues), disease and developmental contexts. SpaceTravLR's perturbation predictions are made solely from spatial omics data and closely align with experimental validation or known outcomes based on mechanistic studies. Critically, our approach enables the generation of mechanistic hypotheses underlying identified niches. We show SpaceTravLR discovered a novel mechanism for Ccr4 that drives the spatial location of a pathogenic population of allergen-specific T helper 2 (Th2) cells as they develop in the lymph node, which was experimentally validated in a murine model. Overall, SpaceTravLR provides a novel interpretable and experimentally validated framework for uncovering how genes act individually and combinatorially through cell-intrinsic and cell-extrinsic circuits to shape spatial tissue organization and function.
    DOI:  https://doi.org/10.1101/2025.11.13.688264
  28. Nucleic Acids Res. 2025 Nov 28. pii: gkaf1230. [Epub ahead of print]
      CellMiner Cross-Database (CellMinerCDB) (https://discover.nci.nih.gov/cellminercdb/) is an established interactive application providing direct access and enabling exploration of cancer cell line pharmacogenomics without extensive programming experience. Data are compiled from many sources, including the National Cancer Institute(NCI), Broad Institute Dependency Map (DepMap), Sanger/MGH Genomics of DrugSensitivity in Cancer (GDSC), MD Anderson Cell Lines Project (MCLP), andNational Center for Advancing Translational Sciences (NCATS). In the version 2.2 update, our collection has expanded to pharmacogenomics data for 1916 cancer cell lines and over 25 000 drugs. Drug screening data include many additional compounds for potential drug repurposing from the Broad PRISM, NCATS, and NCI. The user interface facilitates uncovering specific samples of interest and identifying drug and cell lines across databases. We also expanded the annotations for cross-referencing other databases and downloading our data for further cancer biology and drug discovery studies. Herein, we provide use cases for CellMinerCDB, including (i) data reproducibility given overlaps of cell lines, genes, and drugs across databases; (ii) candidate biomarker discovery; and (iii) cross-dataset analyses.
    DOI:  https://doi.org/10.1093/nar/gkaf1230
  29. bioRxiv. 2025 Oct 13. pii: 2025.10.13.681693. [Epub ahead of print]
      Although we now have a rich toolset for genome editing, an equivalent framework for manipulating the proteome with a comparable flexibility and specificity remains elusive. A promising strategy for "proteome editing" is to use bifunctional molecules (e.g. PROteolysis-Targeting Chimeras or PROTACs1) that bring a target protein into proximity with a degradation or stabilization effector, but their broader application is constrained by a limited repertoire of well-characterized target or effector "handles". We asked whether coupling de novo protein design to a multiplex screening framework could address this gap by accelerating the discovery of effector handles for intracellular protein degradation, stabilization, or relocalization. Using LABEL-seq2, a sequencing-based assay that enables multiplex, quantitative measurement of protein abundance, we screened 9,715 de novo designed candidate effector handles for their ability to recruit a target protein to components of the ubiquitin-proteasome system3 (UPS) (FBXL12, TRAF2, UCHL1, USP38) or the autophagy pathway4 (GABARAP, GABARAPL2, MAP1LC3A). In a single experiment, we discovered hundreds of de novo designed effector handles that reproducibly drove either intracellular degradation (n = 277) or stabilization (n = 204) of a reporter protein. Validation of a subset of these hits in an orthogonal assay confirmed that sequencing-based measurements from the primary screen reliably reflected changes in intracellular abundance of the target protein. Successful effector handles were discovered for both the UPS (n = 194) and autophagy (n = 287) pathways, which provide complementary routes for programmable proteome editing. Autophagy-recruiting effector handles generalized to endogenous targets, as substituting the reporter-specific target handle with a high-affinity MCL1 binder5 reduced endogenous levels of this intracellular oncoprotein6. Moreover, directing autophagy-recruiting effector handles to the outer mitochondrial membrane dramatically perturbed mitochondrial networks in a manner consistent with synthetic tethering and sequestration7,8. Beyond generating a diverse repertoire of protein abundance or localization effector handles, our results establish a scalable, low-cost platform that links deep learning-guided protein design to functional cellular readouts, and chart a course toward a general framework for programmable proteome editing.
    DOI:  https://doi.org/10.1101/2025.10.13.681693
  30. Cancer Gene Ther. 2025 Nov 24.
      Acral melanoma (AM), a rare and aggressive subtype with 5-year survival rates below 16%, exhibits limited response to CDK4/6 inhibitors (CDK4i/6i) despite frequent pathway alterations. Here, we identify AKT-mTOR signaling as a critical escape mechanism triggered by CDK4/6 inhibition. Using a genetically diverse panel of AM cell lines, we demonstrate that CDK4i/6i induces rapid hyperactivation of AKT (pS473) and mTORC1 (pS6 S240/244) alongside Rb dephosphorylation. Interestingly, CDK4i/6i disrupts the cytoplasmic interaction of Rb with the mTORC2 subunit Sin1, suggesting the loss of the Rb-Sin1 protein-protein interaction may lead to mTORC2-mediated AKT hyperactivation following CDK4i/6i. Pharmacological inhibition of the AKT-mTOR axis significantly increases CDK4i/6i efficacy, as seen in the ability to reduce clonogenic survival and the ability to increase annexin+ cytotoxicity relative to single-agent CDK4i/6i, AKTi, or mTORi activity alone. These findings provide preclinical rationale for co-targeting CDK4/6 and mTORC1/2 to improve AM outcomes.
    DOI:  https://doi.org/10.1038/s41417-025-00987-5
  31. Nat Commun. 2025 Nov 27. 16(1): 10656
      Silencing remains a significant challenge for exogenous gene expression, limiting both the penetrance and expressivity of transgenes. In particular, silencing of Cas9 expression is a major technical limitation for many gene editing and CRISPR screening applications. Here, we demonstrate that including introns in Cas9 expression cassettes significantly reduces silencing across multiple cell lines. Notably, the incorporation of an intron into a CRISPRa construct results in reduced silencing, increased expression levels, and markedly enhanced activation of target genes. We investigate diverse intron sequences and discover that T-rich introns over 2 kb confer the greatest protection against silencing. In addition, we find that introns can work synergistically with chromatin opening elements to further mitigate silencing, suggesting regulatory mechanisms are acting at both the DNA and RNA level to silence exogenous genes. Our work highlights the potential of introns to optimize genetic constructs for enhanced expression and improved cellular engineering requiring constitutive expression of large transgenes.
    DOI:  https://doi.org/10.1038/s41467-025-65669-0
  32. Trends Cancer. 2025 Nov 25. pii: S2405-8033(25)00260-2. [Epub ahead of print]
      While the initial transformation of cancer cells is driven by genetic alterations, tumor cell behaviors and functional states are dynamically regulated by cell-intrinsic factors including proteins, metabolites and lipids, and extrinsic microenvironmental factors. Emerging multi-omics technologies highlighted that cancer cells exhibit distinct lipidome compositions and employ specific lipid metabolic circuits for chemical conversions - collectively defined as 'lipotypes'. We review the interplay between cancer lipotypes and cellular states, focusing on interpreting how being at different positions along the spectra of representative lipid metabolic axes influences cancerous traits. We aim to instill a system biology perspective to integrate 'lipotypes' into the established 'genotype-phenotype' framework in cancer.
    Keywords:  cancer progression and metastasis; lipid biosynthesis; lipid metabolism; lipidomics; lipotypes; storage and degradation; tumor microenvironment; uptake
    DOI:  https://doi.org/10.1016/j.trecan.2025.10.009
  33. PNAS Nexus. 2025 Nov;4(11): pgaf355
      Scratch wound healing assays are widely used to study collective cell migration, essential for understanding tissue regeneration, drug effects, and wound healing mechanisms. However, conventional analyses often rely on wound edge dynamics or individual cell tracks, limiting spatial insight into migration behavior. We present a sector-based analytical framework that reinterprets time-lapse microscopy data by dividing each image into defined spatial regions across the field of view. This enables spatially resolved characterization of how cell populations migrate over time. To address challenges of low contrast and uneven illumination in bright-field microscopy, we apply a level-set segmentation algorithm that robustly detects the wound edge. Using this approach, we show that both cell velocity and trajectory vary with distance from the wound boundary. We introduce a novel metric, the sector-boundary distance, to identify regions where cells migrate faster along nonradial paths. To assess chemotactic activation, cells were treated with the chemokine CXCL10 to stimulate motility via CXCR3-mediated signaling. Statistical testing showed that, in treated cells, the proportion of highly motile cells was significantly associated with wound closure, even in regions distant from the scratch, whereas directionality played a limited role. By contrast, untreated cells exhibited weaker and less organized migration patterns. These findings highlight how local cellular activity contributes to healing in a treatment-dependent manner. Our method bridges global wound-level analysis and local cell-scale behavior by combining single-cell tracking with precise boundary detection. The complete framework is available as open-source software, including a user-friendly web application that enables interactive analysis of microscopy data.
    Keywords:  collective cell migration; image processing; level-sets; scratch wound healing assay; time-lapse microscopy
    DOI:  https://doi.org/10.1093/pnasnexus/pgaf355
  34. Nat Commun. 2025 Nov 27. 16(1): 10652
      Multiplexed imaging has transformed our ability to study tissue organization by capturing thousands of cells and molecules in their native context. However, these datasets are enormous, often comprising tens of gigabytes per image, and require complex workflows that limit their broader use. Current tools are often fragmented, inefficient, and difficult to adopt across disciplines. Here we show that SPACEc, a scalable Python platform, streamlines spatial imaging analysis from start to finish. The platform integrates image processing, cell segmentation, and data preprocessing into a single workflow, while improving computational performance through parallelization and GPU acceleration. We introduce innovative methods, including patch proximity analysis, to more accurately map local cellular neighborhoods and interactions. SPACEc also simplifies advanced approaches such as deep-learning annotation, making them accessible through an intuitive interface. By combining efficiency, accuracy, and usability, this platform enables researchers from diverse backgrounds to gain deeper insights into tissue architecture and cellular microenvironments.
    DOI:  https://doi.org/10.1038/s41467-025-65658-3