bims-pideca Biomed News
on Class IA PI3K signalling in development and cancer
Issue of 2025–09–21
eighteen papers selected by
Ralitsa Radostinova Madsen, MRC-PPU



  1. Nature. 2025 Sep 17.
      The mechanistic target of rapamycin complex 1 (mTORC1) integrates growth factor (GF) and nutrient signals to stimulate anabolic processes connected to cell growth and inhibit catabolic processes such as autophagy1,2. GF signalling through the tuberous sclerosis complex regulates the lysosomally localized small GTPase RAS homologue enriched in brain (RHEB)3. Direct binding of RHEB-GTP to the mTOR kinase subunit of mTORC1 allosterically activates the kinase by inducing a large-scale conformational change4. Here we reconstituted mTORC1 activation on membranes by RHEB, RAGs and Ragulator. Cryo-electron microscopy showed that RAPTOR and mTOR interact directly with the membrane. Full engagement of the membrane anchors is required for optimal alignment of the catalytic residues in the mTOR kinase active site. Converging signals from GFs and nutrients drive mTORC1 recruitment to and activation on lysosomal membrane in a four-step process, consisting of (1) RAG-Ragulator-driven recruitment to within ~100 Å of the lysosomal membrane; (2) RHEB-driven recruitment to within ~40 Å; (3) RAPTOR-membrane engagement and intermediate enzyme activation; and (4) mTOR-membrane engagement and full enzyme activation. RHEB and membrane engagement combined leads to full catalytic activation and structurally explains GF and nutrient signal integration at the lysosome.
    DOI:  https://doi.org/10.1038/s41586-025-09545-3
  2. Nat Methods. 2025 Sep 15.
      Artificial intelligence (AI) methods are powerful tools for biological image analysis and processing. High-quality annotated images are key to training and developing new algorithms, but access to such data is often hindered by the lack of standards for sharing datasets. We discuss the barriers to sharing annotated image datasets and suggest specific guidelines to improve the reuse of bioimages and annotations for AI applications. These include standards on data formats, metadata, data presentation and sharing, and incentives to generate new datasets. We are sure that the Metadata, Incentives, Formats and Accessibility (MIFA) recommendations will accelerate the development of AI tools for bioimage analysis by facilitating access to high-quality training and benchmarking data.
    DOI:  https://doi.org/10.1038/s41592-025-02835-8
  3. EMBO Rep. 2025 Sep 16.
      mTOR (mechanistic target of rapamycin) kinase is a pivotal regulator of cellular growth and metabolism, integrating signals from nutrients and growth factors. It functions through the assembly of two distinct complexes, mTORC1 and mTORC2, which differ in their substrate specificity and regulation. While the regulation of mTORC1 is well-characterized, less is known about the modulators of mTORC2 signaling. In this study, we identify tyrosine phosphatase PTPN22 as an mTORC2-associated protein. We provide evidence that PTPN22 is essential for the activation of the mTORC2/AKT axis, independent of cell lineage. Loss of PTPN22 results in impaired AKT phosphorylation in response to both basal and growth factor signals. Mechanistically, PTPN22 functions as a scaffolding protein that promotes the mSIN-RICTOR interaction, thereby maintaining mTORC2 complex integrity. Notably, this adaptor function of PTPN22 is independent of its tyrosine phosphatase activity. Functionally, we demonstrate that PTPN22 is required for cell growth and survival in both cellular models and nude mouse xenografts. Together, these findings reveal a non-catalytic role for phosphatase PTPN22 in mTORC2 assembly and function.
    Keywords:  AKT; PTPN22; Rictor; mSIN; mTOR
    DOI:  https://doi.org/10.1038/s44319-025-00576-5
  4. bioRxiv. 2025 Sep 05. pii: 2025.09.05.674553. [Epub ahead of print]
      Genetically encoded DNA recorders convert transient biological events into stable genomic mutations, offering a means to reconstruct past cellular states. However, current approaches to log historical events by modifying genomic DNA have limited capacity to record the magnitude of biological signals within individual cells. Here, we introduce MitoScribe, a mitochondrial DNA (mtDNA)-based recording platform that uses mtDNA base editors (DdCBEs) to write graded biological signals into mtDNA as neutral, single-nucleotide substitutions at a defined site. Taking advantage of the hundreds to thousands of mitochondrial genome copies per cell, we demonstrate MitoScribe enables reproducible, highly sensitive, non-destructive, durable, and high-throughput measurements of molecular signals, including hypoxia, NF-κB activity, BMP and Wnt signaling. We show multiple modes of operation, including multiplexed recordings of two independent signals, and coincidence detection of temporally overlapping signals. Coupling MitoScribe with single-cell RNA sequencing and mitochondrial transcript enrichment, we further reconstruct signaling dynamics at the single-cell transcriptome level. Applying this approach during the directed differentiation of human induced pluripotent stem cells (iPSCs) toward mesoderm, we show that early heterogeneity in response to a differentiation cue predicts the later cell state. Together, MitoScribe provides a scalable platform for high-resolution molecular recording in complex cellular contexts.
    DOI:  https://doi.org/10.1101/2025.09.05.674553
  5. Mol Cell. 2025 Sep 18. pii: S1097-2765(25)00706-3. [Epub ahead of print]85(18): 3486-3504.e7
      The mechanistic target of rapamycin (mTOR) is a key regulator of lipid homeostasis by controlling processes including lipid uptake and biosynthesis. mTOR dysregulation and consequent altered lipid metabolism are common in various diseases, including cancers, making mTOR a promising therapeutic target. Therefore, it is crucial to understand how mTOR activation and inhibition reprogram lipid homeostasis. In human cancer cell lines, mTOR inhibition induces alternative lipid uptake through translation eukaryotic initiation factor 3D (eIF3D)-mediated low-density lipoprotein receptor (LDLR)-related protein 6 (LRP6) increase and activates liver X receptor β (LXRβ), promoting cholesterol release from lysosomes and its transport to the plasma membrane via Niemann-Pick disease type C (NPC) intracellular cholesterol transporter 1 (NPC1). This signaling supports tumor cell survival and stress resistance. In mouse xenograft models, combining mTOR inhibition with LRP6 knockdown or NPC1 targeting significantly suppresses tumor growth. Our findings highlight mTOR feedback signaling in reprogramming lipid homeostasis and its therapeutic potential to treat diseases characterized by dysregulated mTOR.
    Keywords:  AKT; IGF1R; LRP6; NPC1; cholesterol; mTOR
    DOI:  https://doi.org/10.1016/j.molcel.2025.08.021
  6. Immunol Cell Biol. 2025 Sep 14.
      Activated PI3K delta syndrome 1 (APDS1) is caused by a heterozygous germline gain-of-function (GOF) variant in PIK3CD, which encodes the p110δ catalytic subunit of phosphoinositide 3-kinase (PI3K). APDS1 patients display a broad range of clinical manifestations and perturbations in cellular phenotype. One of the most striking features is the dysregulation of the T-cell compartment, characterized by an increase in memory T cells, including Tfh cells, and a concomitant decrease in naïve T cells. We have previously shown that many of these changes in T-cell populations were T-cell extrinsic and were also induced in WT T cells that developed in the presence of PI3K GOF cells. Here we dissected the drivers of dysregulated T-cell activation using a mouse model of APDS1. This revealed that PI3K GOF macrophages and DCs made little contribution to the aberrant T-cell activation. Instead, PI3K GOF T cells were able to drive the loss of WT naïve CD4+ T cells, while dysregulated PI3K GOF B cells mediated an increase in Tfh cells. Surprisingly, despite previous reports of increased PI3K signalling driving dysregulated inflammatory Tregs, we saw no evidence for Pik3cd GOF Tregs acquiring an inflammatory phenotype and driving T-cell activation. These studies provide new insights into the role of PI3K in immune cells and how increased PI3K can drive T- and B-cell dysregulation and contribute to the phenotype of APDS1 patients.
    Keywords:  CD4+ T cells; follicular T‐helper cells; primary immunodeficiency; regulatory T cells
    DOI:  https://doi.org/10.1111/imcb.70058
  7. Stem Cell Reports. 2025 Sep 18. pii: S2213-6711(25)00245-0. [Epub ahead of print] 102641
      Hematopoietic stem cells (HSCs) arise in embryogenesis from a specialized hemogenic endothelium (HE) via endothelial-to-hematopoietic transition (EHT). While induced pluripotent stem cells (iPSCs) give rise to HE with robust hemogenic potential, bona fide HSC generation from iPSCs remains challenging. We map single-cell dynamics of EHT from iPSCs and integrate it with human embryo datasets to identify ligand-receptor interactions that drive transcriptional divergence between iPSC-derived and embryonic cell states. The expression of endothelial genes predicted to be regulated by FGF signaling was incompletely repressed during iPSC-derived EHT. FGF activity declined at the onset of EHT to enable normal hematopoiesis in the zebrafish, and chemical inhibition of FGF signaling during EHT enhanced HSC and progenitor generation in the zebrafish and from iPSCs. In summary, we generate a single-cell map of iPSC-derived EHT, identify ligand-receptor interactions that can improve iPSC differentiation, and uncover elevated FGF signaling as a barrier to hematopoiesis.
    Keywords:  FGF; endothelial-to-hematopoietic transition; hematopoiesis; iPSC; single cell atlas
    DOI:  https://doi.org/10.1016/j.stemcr.2025.102641
  8. PLoS Comput Biol. 2025 Sep 16. 21(9): e1013300
      Biological networks provide a structured framework for analyzing the dynamic interplay and interactions between molecular entities, facilitating deeper insights into cellular functions and biological processes. Network construction often requires extensive manual curation based on scientific literature and public databases, a time-consuming and laborious task. To address this challenge, we introduce NeKo, a Python package to automate the construction of biological networks by integrating and prioritizing molecular interactions from various databases. NeKo allows users to provide their molecules of interest (e.g., genes, proteins or phosphosites), select interaction resources and apply flexible strategies to build networks based on prior knowledge. Users can filter interactions by various criteria, such as direct or indirect links and signed or unsigned interactions, to tailor the network to their needs and downstream analysis. We demonstrate some of NeKo's capabilities in two use cases: first we construct a network based on transcriptomics from medulloblastoma; in the second, we model drug synergies. NeKo streamlines the network-building process, making it more accessible and efficient for researchers.
    DOI:  https://doi.org/10.1371/journal.pcbi.1013300
  9. J Clin Invest. 2025 Sep 16. pii: e197764. [Epub ahead of print]135(18):
      Cutaneous melanoma (CM) is known for its aggressive behavior, high metastatic potential, and poor prognosis. Mutations in the BRAF gene are common in CM, and patients with BRAF-mutant melanoma often respond well to combined inhibition of BRAF and MEK (BRAFi + MEKi). Although BRAFi + MEKi therapy provides clinical efficacy, the response durability is limited by persistent drug-tolerant residual cells, culminating in relapse. In this issue of the JCI, Tiago et al. confirmed that NR2F1, a dormancy-associated transcription factor, is a key determinant of therapeutic resistance in melanoma. NR2F1 expression was elevated in transcriptomic datasets from patients with minimal residual disease, and in murine and human melanoma models, NR2F1 overexpression reduced therapeutic efficacy and suppressed tumor proliferation and invasion while sustaining mechanistic target of rapamycin complex 1 (mTORC1) transcriptional regulation of relevant genes. Combining BRAFi + MEKi with the mTORC1 inhibitor rapamycin effectively targeted these resistant melanoma cells, suggesting a potential path forward for targeting NR2F1 and mTORC1 signaling in patients with CM.
    DOI:  https://doi.org/10.1172/JCI197764
  10. Cell Rep Methods. 2025 Sep 17. pii: S2667-2375(25)00213-9. [Epub ahead of print] 101177
      In vitro human pluripotent stem cell (hPSC)-derived models have been crucial in advancing our understanding of the mechanisms underlying neurodevelopment, though knowledge of the earliest stages of brain formation is lacking. Micropatterning of cell populations as they transition from pluripotency through the process of neurulation can produce self-assembled neuroepithelial tissues (NETs) with precise spatiotemporal control, enhancing the fidelity of hPSC models to the early developing human brain and their use in phenotypic assessments. Here, we introduce an accessible, customizable, and scalable method to produce self-assembled NETs using bioprinting to rapidly deposit reproducibly sized extracellular matrix droplets. Matrix addition to the media provides a scaffold that promotes 3D tissue folding, reflecting neural tube development. We demonstrate that these scaffolded NETs (scNETs) exhibit key architectural and biological features of the human brain during normal and abnormal development-notably, hyperproliferation and structural malformations induced by TSC2 deficiency-and provide a robust drug screening tool.
    Keywords:  CP: Biotechnology; CP: Stem cell; bioprinting; cortical folding; cortical malformations; human pluripotent stem cells; micropatterning; neural tube; neurodevelopment; neuruloids; scaffolded neuroepithelial tissues; tuberous sclerosis complex
    DOI:  https://doi.org/10.1016/j.crmeth.2025.101177
  11. Stem Cell Reports. 2025 Sep 18. pii: S2213-6711(25)00247-4. [Epub ahead of print] 102643
      Human pluripotent stem cells (PSCs) are known to harbor mutations in tumor-associated genes, and here we aim to examine the status of adult stem cells (ASCs). We thus identify cancer-related mutations in 18% of about 600 mesenchymal stem cell samples, and in 41% of about 200 neural stem cell (NSC) samples. We show a lineage-specific profile of cancer-related genes, demonstrating that TP53 is a central mutated gene in human PSCs but not in mesenchymal or NSCs. We suggest that the lineage-specificity of tumor-associated genes correlates with their expression levels and with tumor-specific mutations in patients. We also show the consequences of mutations in oncogenes and tumor suppressor genes on the transcriptome of each specific stem cell lineage. We therefore propose a categorization of these mutated samples for further appreciation of their severity and emphasize the importance of genetic screening in pluripotent and ASC lines.
    Keywords:  adult stem cells; cancer-related mutations; embryonic stem cells; genomic integrity; lineage specificity; mesenchymal stem cells; neural stem cells; point mutations
    DOI:  https://doi.org/10.1016/j.stemcr.2025.102643
  12. Cell Rep. 2025 Sep 17. pii: S2211-1247(25)01067-8. [Epub ahead of print]44(10): 116296
      Mitochondrial metabolism determines bone marrow hematopoietic stem cell (HSC) heterogeneity and influences their repopulation potential, though its embryonic origins remain unclear. We show that during the endothelial-to-hematopoietic transition in the mouse embryo, dynamic changes in mitochondrial activity drive the production of hematopoietic stem and progenitor cells (HSPCs) with differing potencies. Lowering mitochondrial activity in the aorta-gonad-mesonephros (AGM) by pharmacological or genetic means activates Wnt signaling to promote HSPC expansion. Further, mitochondrial membrane potential (MMP) gives rise to functional heterogeneity in HSPCs. In-vitro and in-vivo functional assays and single-cell transcriptomics showed that MMPlow HSPCs in the AGM are myeloid biased, with enhanced differentiation potential, whereas MMPhigh HSPCs are lymphoid biased, with diminished differentiation potential. Mechanistically, low mitochondrial activity in HSPCs upregulates phosphoinositide 3-kinase signaling to promote differentiation. These insights into the initiation of metabolic heterogeneity could be leveraged to isolate the distinct HSPC subsets and to efficiently generate the desired lineages.
    Keywords:  CP: Developmental biology; PI3K signaling; Wnt signaling; cell fate; endothelial to hematopoietic transition; hematopoiesis; hematopoietic stem and progenitor cell; lymphoid; metabolism; mitochondria; myeloid
    DOI:  https://doi.org/10.1016/j.celrep.2025.116296
  13. Sci Signal. 2025 Sep 16. 18(904): eadx5186
      Mutated RAS proteins activate downstream effector pathways (RAF-MEK-ERK and PI3K-AKT) to drive oncogenic transformation and progression. Because RAS family members differentially engage these pathways, combined inhibition of both pathways is required to effectively treat RAS-mutated cancers. Here, we found that this was due to signaling contributed by wild-type RAS family members that activated an effector pathway that was poorly engaged by the mutant RAS family member. Wild-type KRAS and NRAS promoted RAF-MEK-ERK signaling in cells expressing mutant HRAS, whereas wild-type HRAS and NRAS promoted PI3K-AKT signaling in cells expressing mutant KRAS. Combining inhibitors targeting the poorly engaged RAS effector pathways with inhibitors targeting the mutant RAS resulted in synergistic cytotoxicity in a manner that depended on wild-type RAS expression. The farnesyltransferase inhibitor tipifarnib blocked mutant HRAS-PI3K signaling and synergized with MEK inhibitors in HRAS-mutated cells, whereas KRASG12C inhibitors blocked mutant KRAS-MEK signaling and synergized with PI3K inhibitors in KRASG12C-mutated cells. Synergy was abolished in MEFs lacking all RAS proteins and in cancer cell lines in which nonmutated RAS family members were deleted. Our data highlight the critical role of wild-type RAS family members in supporting mutant RAS signaling and its importance as a therapeutic cotarget in RAS-mutated cancers.
    DOI:  https://doi.org/10.1126/scisignal.adx5186
  14. Ann Biomed Eng. 2025 Sep 17.
      Cholesterol is a vital component of the cell membrane and plays an essential role in mediating integral membrane protein function. Altered cholesterol regulation has been implicated in neurological diseases that are associated with blood-brain barrier breakdown. However, the role of brain barrier function in inherited disorders of cholesterol metabolism, such as Niemann-Pick disease C1 (NP-C1), remains unclear. In this study, we determined how cholesterol depletion with U18666A, a chemical inhibitor of NPC1 protein, as well as with the cholesterol-depleting agent methyl-β cyclodextrin (MβCD), impacted brain endothelial cell barrier function. We hypothesized that cholesterol depletion would decrease barrier integrity by disrupting tight junction protein continuity. To test this hypothesis, we differentiated human-induced pluripotent stem cells into brain microvascular endothelial cells (hiBMECs). We then assessed barrier integrity by quantifying trans-endothelial electrical resistance (TEER), small fluorescent molecule permeability, and tight junction continuity and protein levels. We now show that U18666A-treated hiBMECs demonstrated a 75% decrease in TEER and 9-fold increase in sodium fluorescein permeability. Similar trends were observed for hiBMECs treated with MβCD, which showed significantly lowered TEER (93% decrease) and increased sodium fluorescein permeability (20-fold higher). We also observed decreased continuity of the tight junction proteins occludin (13% lower) and claudin-5 (8% lower) as well as a 53% decrease in claudin-5 protein with U18666A treatment. Co-treating U18666A-treated hiBMECs with hydroxypropyl-β cyclodextrin (HPβCD), which releases lysosomal cholesterol, prevented these changes. Together, our results demonstrate that cholesterol is vital for hiBMEC barrier function and tight junction continuity. This study highlights the potential of therapeutics targeted to brain endothelium in NP-C1 and other cholesterol metabolism disorders.
    Keywords:  Blood–brain barrier; Brain endothelial; Cholesterol; Niemann-Pick disease; Tight junction; iPSC-BMEC
    DOI:  https://doi.org/10.1007/s10439-025-03841-9
  15. FEBS Lett. 2025 Sep 19.
      The Target of Rapamycin (TOR) is a highly conserved protein kinase that regulates cell growth and metabolism through two distinct complexes, TORC1 and TORC2. Each complex regulates different downstream targets; however, both are activated and regulated on lipid membranes. In this Perspective, we will revise the structural biology of TOR complexes and the molecular mechanisms governing their regulation, highlighting the importance of lipid membranes for their function.
    Keywords:  (m)TORC1; (m)TORC2; cryoEM; lipid membrane; peripheral membrane protein complexes
    DOI:  https://doi.org/10.1002/1873-3468.70171
  16. bioRxiv. 2025 Sep 08. pii: 2025.09.03.673941. [Epub ahead of print]
      Many biological processes, like gene regulation or cell signalling, rely on molecules (inputs) that bind to targets leading to downstream responses. In the gene regulation field, recent data have shown that higher transcription factor (TF) concentrations may increase transcription levels of a gene without affecting the gene activation time. We call this behaviour output decoupling . Motivated by these observations, here we investigate mechanisms for output decoupling in Markov process models where a readout molecule is produced downstream of ligand binding. Our focus is on identifying regimes where the steady-state level of the readout changes with input concentration, while the activation time, quantified by mean first-passage times, remains unaffected. Through a combination of analytical and numerical investigations, we find two mechanisms through which output decoupling can arise: i) rate scale separation , where the system is comprised of slow and fast transitions that are differentially regulated by the input; and ii) incoherent regulation, where the input acts on two transitions with opposing effects on readout production, with all transitions operating on similar timescales in the absence of input. Such incoherent regulation has emerged as a plausible regulatory mode of TFs, and we suggest decoupling as a new characteristic feature of this regulatory mode. More broadly, our findings offer a mechanistic and conceptual framework for reasoning about output decoupling in input-output systems.
    Author summary: How biomolecular systems respond to signals often depends not only on the final level of activity they reach, but also on how quickly they reach it. These two outputs - steady-state level and activation time- are usually coupled: higher input concentration tends to produce both higher activity and faster responses. Yet recent experiments suggest that, in some cases, the strength of the response can change while the speed remains constant. In this work, we explore the conditions under which such "output decoupling" can occur. Using mathematical models of molecular systems, we identify two ways this can happen. In one case, the activation time is determined by slow steps in the system that the input does not control. In the other, the input exerts opposing effects on the system, simultaneously promoting and hindering the response, which balances the timing. By revealing how decoupling arises, our study provides a framework for interpreting puzzling experimental results. More broadly, it shows the value of considering both dynamics and steady-state behavior jointly when studying how molecular systems process information.
    DOI:  https://doi.org/10.1101/2025.09.03.673941
  17. bioRxiv. 2025 Sep 07. pii: 2025.09.02.673874. [Epub ahead of print]
       Background: Capillary malformation (CM) is a congenital vascular anomaly affecting the skin, mucosa, and brain, yet the understanding of its vascular pathogenesis remains limited.
    Methods: We applied spatial whole-transcriptome profiling (GeoMx) and gene set enrichment analysis within CM lesions at single vasculature level. Differentially expressed genes were validated by immunofluorescence staining. Phosphoproteomics was profiled to uncover lesion-wide phosphorylation sites on proteins. Single-cell RNA sequencing was performed on CM-derived induced pluripotent stem cells (iPSCs) to determine differentiation trajectories of lesional vascular lineages. In silico gene perturbation was used to predict candidate genes for modulating vascular pathological progression, followed by functional validation in CM iPSC-derived endothelial cells (ECs) using a Tet-on system.
    Results: A spatial transcriptomic atlas was constructed, and pathological landscape of individual CM vasculature was delineated. CM vessels exhibited hallmarks of endothelial-to-mesenchymal transition (EndMT), including disruption of adherens junctions (AJs), vascular identity transitions, and metabolic remodeling. Phosphoproteomics confirmed that differentially phosphorylated proteins were enriched in EndMT- and AJ-related pathways. Aberrant expression of venous transcriptional factor NR2F2 was observed in lesional ECs and correlated with progressive enlargement from capillaries to larger-caliber vessels containing multiple layers of smooth muscle cells (SMCs). In CM iPSCs, differentiation course yielded reduced ECs but increased SMCs. In silico knockout simulation predicted NR2F2 as a crucial regulator of facilitating SMC phenotype in CM. Consistently, enforced NR2F2 expression during iPSC differentiation suppressed endothelial markers while inducing SMC-associated genes.
    Conclusions: Single CM vasculature displays pathological hallmarks characterized by EndMT and AJ disruption, leading to progressive vascular remodeling. NR2F2 functions as a central regulatory factor orchestrating the acquisition of the SMC phenotype, thereby representing a potential therapeutic target in CM.
    Keywords:  Capillary malformation; NR2F2; adherens junctions; endothelial cells; endothelial-to-mesenchymal transition; induced pluripotent stem cells; whole transcriptome atlas
    DOI:  https://doi.org/10.1101/2025.09.02.673874