Genome-wide CRISPR-Cas9 screening identifies CLK1 inhibition as a strategy to restore PARP inhibitor sensitivity via ERCC1 isoform switching
Epithelial ovarian cancer (EOC) is an aggressive malignancy with limited therapeutic options. Poly(ADP-ribose) polymerase inhibitors (PARPi) have shown remarkable efficacy, especially in BRCA-mutant patients, and are approved as maintenance therapy to prevent recurrence after initial response to chemotherapy. However, the development of PARPi resistance poses a major clinical challenge. This study utilized a whole-genome CRISPR-Cas9 genetic screening to identify genes associated with PARPi sensitivity upon knockout. Based on the screening and validated through further experiments, we confirmed that CLK1 knockdown is synthetically lethal with PARPi in ovarian cancer. The combination of the PARPi Olaparib and CLK1 inhibitor TG003 exhibited potent anti-proliferative effects both in vitro and in vivo. Mechanistically, CLK1 inhibition downregulated the functional ERCC1-202 isoform, resulting in enhanced DNA damage and apoptosis. Our findings reveal a novel mechanism underlying PARPi sensitivity and suggest that targeting CLK1 in combination with PARPi may represent a promising therapeutic strategy for PARPi-resistant ovarian cancer.
Lipid deposition promotes YTHDF3-mediated m6A modification of PPARα to facilitate liver metastasis of colorectal cancer
The liver is a common site for cancer metastasis and a key metabolic organ. Lipid metabolism irregularities are linked to liver metastasis risk, but the mechanisms are not fully understood. Herein, in colorectal liver metastases (CRLM) clinical samples, lipid metabolism was broadly dysregulated, and lipid metabolites accumulated, as shown by integrated transcriptome and lipidomics analyses. Functionally, lipids deposition promotes liver metastasis in vitro and in vivo. Mechanistically, lipid deposition significantly enhances YTHDF3-mediated m6A modification and degradation of PPARα, which is crucial for liver metastasis. This process reduces the β-hydroxybutyrylation of YTHDF3, thereby promoting LLPS and increasing the stability of YTHDF3, which in turn facilitates the progression of CRC and liver metastasis. Furthermore, lipid deposition induces the interaction between STAT3 and YAP, activating YTHDF3 transcription. These two regulatory mechanisms synergize to drive YTHDF3 accumulation in lipid-rich metastatic lesions. In summary, our findings reveal that lipid deposition promotes LLPS-mediated m6A modification and decreases β-hydroxybutyrylation in liver metastasis, offering new strategies for the treatment of CRLM.
GlycoRNA research: from unknown unknowns to known unknowns
Recent discoveries have revealed the existence of glycosylated RNAs (glycoRNA), in which glycans are covalently linked to small non‑coding RNAs and are predominantly localized to the cell surface. Since the initial discovery in 2021, glycoRNA have become an emerging field: four years in glycoRNA research have produced advances in labeling, imaging, and mass spectrometry that now highlight the role of glycoRNA in cell communication, immune regulation, and disease progression. In this review, we summarize current knowledge of glycoRNA biogenesis, detection techniques, and biological functions, and discuss how these findings reshape the future interface between glycobiology and RNA biology.
Human meibomian gland organoids to study epithelial homeostasis and dysfunction
Meibomian glands (MGs) are holocrine glands that secrete lipids to maintain the homeostasis of ocular surface, and their dysfunction leads to dry eye disease. Herein, we established long-term 3D organoid culture for murine and human MGs, which retained the cell lineages and lipid-producing ability. The organoids mimicked the drug treatment responses and generated functional MGs after orthotopic transplantation. Inspired by organoid cultures, we found FGF10 eye drops could rescue all-trans retinoic acid-induced MG dysfunction in mice. Besides, nicotinamide uniquely hampered the human MG organoid expansion by inhibiting FGF10 signaling. Single-cell atlas and lipidome not only aligned the delineated cell types and featured lipids between human MGs and organoids, but highlighting MAPK signaling inhibition enhanced acinar cell differentiation and functional maturation of MG organoids. In summary, this study established an organoid platform to explore epithelial homeostasis and dysfunction of MGs, facilitating drug development and regenerative medicine for dry eye disease.
DNA - histone cross-link locks the nucleosome structure and disrupts its recognition and processing
DNA-histone cross-links (DHCs) frequently arise within nucleosomes during DNA damage and repair processes. However, the functional consequences of DHC within nucleosomes remain largely unexplored. In this study, we prepared structurally homogeneous nucleosomes containing a single, site-specific DHC using click chemistry and systematically evaluated the impact of DHC on nucleosome structure and function. Our results show that DHC markedly enhances nucleosome thermal stability and completely blocks both thermally induced passive sliding and chromatin remodeler-mediated active sliding. Moreover, DHC obstructs SP6 RNA polymerase-driven transcription elongation through nucleosomes, leading to premature termination approximately 15 bp upstream of the cross-linking site. DHC also increases histone resistance to proteolytic digestion within nucleosomes. These findings suggest that even a single DHC can substantially lock and rigidify the nucleosome structure and broadly interfere with the recognition and processing of nucleosomes by various cellular machineries, thereby rendering DHC a highly toxic and persistent form of DNA damage. This in vitro study highlights the unique impact of DHC on nucleosome architecture and is expected to motivate further exploration of its biological roles in vivo.
Capturing trophectoderm-like stem cells enables step-wisely remodeling of placental development
The trophectoderm produced from totipotent blastomeres initiates trophoblast development, while placental deficiencies can cause pregnancy disorders. Yet, a culture system that fully recapitulates the entire placenta development is still lacking, greatly limiting related studies. Here, we captured mouse trophectoderm-like stem cells (TELSCs), which can give rise to all trophoblast lineages and be applied to generate trophoblast organoids. We achieved the induction and maintenance of TELSCs from totipotent blastomere-like stem cells or early embryos through a Hippo-YAP/Notch-to-TGFβ1 signaling switch. At the molecular level, TELSCs resemble E4.5 trophectoderm and are distinct from all previously known trophoblast-like stem cells. Functionally, TELSCs can generate all trophoblast lineages in both teratoma and chimera assays. We further applied TELSCs to generate trophoblast organoids containing various mature trophoblasts and a self-renewing extraembryonic ectoderm (ExE)-like progenitor population. Interestingly, we observed transiently formed rosette-like structures that rely on Itgb1, which are essential to induce ExE-like progenitors and to generate organoids eventually. Thus, the capture of TELSCs enables comprehensive insights into placental development.
Peptide Hormones and Bile Acids Shaping Immune Tolerance of the Liver: Implications and Applications
Postprandially reabsorbed bile acids, along with various peptide hormones released following a meal, orchestrate complex events associated with digestion and prepare the body for the disposal of incoming nutrients by regulating metabolism. Interestingly, these factors have also been shown to modulate immune function. For example, recent interest in weight-loss agents such as semaglutide has demonstrated their ability to attenuate inflammation and provide benefits in diverse clinical contexts characterized by inflammatory responses. This raises an important question: why do hormones with well-established roles in digestion and metabolism also influence immunity? Here, we propose that the immune-regulatory activity of peptide hormones, together with postprandially reabsorbed bile acids, contributes to another remarkable phenomenon: the exceptional immune tolerance of the liver. While it is well established that the liver is an immunologically tolerant organ, the precise mechanisms underlying this skewed immunological tone remain poorly understood. Hepatic immune tolerance has generally been considered an intrinsic property of the liver, arising from autonomous mechanisms. Here, we highlight that various entero-pancreatic endocrine factors delivered to the liver via the portal vein activate cAMP signalling, thereby promoting immune tolerance and attenuating inflammatory tone within the liver. Critically, because these endocrine factors reach the liver at elevated concentrations through the portal vein before dilution in the systemic circulation, they profoundly shape the hepatic immune environment. Physiologically, this system ensures that the liver tolerates diet- and gut-derived inflammogens. Finally, we discuss several implications of this mechanism.
Primate ASPM knockout causes severe microcephaly and oligodendrocyte loss in the brain
AhR-Siglec-15 axis regulates lysosomal Ca2+ release for sonic hedgehog medulloblastoma growth via TRPML1
Sonic hedgehog subgroup medulloblastoma (SHH-MB), an aggressive pediatric brain tumor that originates from granule neuron precursors, faces the challenge of poor treatment owing to its unclear molecular mechanisms. Here, we show that sialic acid-binding immunoglobulin-like receptor 15 (Siglec-15), an immunosuppressive membrane protein, is upregulated and mediates SHH-MB growth through its translocation to the lysosomal membrane. We found that SHH-MB cells use the cation-independent mannose 6-phosphate receptor (CI-MPR) to transport Siglec-15 from the trans-Golgi network (TGN) to lysosomes, where Siglec-15 induces lysosomal Ca2+ release by interacting with mucolipin TRP cation channel 1 (TRPML1), leading to the nuclear translocation of the transcription factor EB (TFEB). Blockade of Siglec-15, TRPML1 or TFEB hinders SHH-MB growth in vitro and in vivo. Importantly, aryl hydrocarbon receptor (AhR), a cytoplasmic transcription factor, upregulates Siglec-15 expression. AhR inhibition by CH-223191 or StemRegenin 1 (SR1) achieved therapeutic efficacy against orthotopic SHH-MB xenografts in mice. These findings reveal an essential role for the AhR-siglec-15 axis in SHH-MB development, providing a potential strategy for SHH-MB treatment.
Tumor cell-specific loss of GPX4 reprograms triacylglycerol metabolism to escape ferroptosis and impair antitumor immunity in NSCLC
Glutathione peroxidase 4 (GPX4) is a master regulator of ferroptosis, a process that has been proposed as a potential therapeutic strategy for cancer. Here we have unexpectedly found that inducible knockout of GPX4 in tumor cells significantly promotes non-small cell lung cancer (NSCLC) progression in the autochthonous Kras LSL-G12D/+ Lkb1 fl/fl (KL) and Kras LSL-G12D/+ Tp53 fl/fl (KP) mouse models, whereas inducible overexpression of GPX4 in tumor cells exerts the opposite effect. GPX4-deficient tumor cells evade ferroptosis by upregulating the expression of DGAT1/2 to promote the synthesis of triacylglycerol (TAG) and oxidized TAG (oxTAG) and the formation of lipid droplets in cells. In addition, GPX4-deficient tumor cells secrete TAG and oxTAG into the extracellular space to induce dysfunction of antitumor CD8+ T cells, thereby coordinating an immunoinhibitory tumor microenvironment (TME). Consistently, treatment with DGAT1/2 inhibitors or inducible overexpression of GPX4 in tumor cells significantly resensitizes tumor cells to ferroptosis and ignites the activation of T cells in the TME to inhibit NSCLC progression. These findings highlight a previously uncharacterized role of tumor cell-specific GPX4 in NSCLC progression and provide potential therapeutic strategies for NSCLC.
CTCF's loop-independent functions prevail over chromatin looping in the acute degradation system
Molecular Recognition at the Opioid-modulating Neuropeptide FF Receptor 1
Multidimensional single-cell analysis reveals immune dysfunction and inflammatory response in Lymphatic malformations
Lymphatic malformations (LMs) are debilitating and potentially life-threatening diseases. However, the immune phenotype of circulating cells and underlying molecular mechanisms in LMs remain poorly understood. Here, we performed integrated single-cell RNA, T-cell receptor, and B-cell receptor sequencing (scRNA-seq, scTCR-seq, and scBCR-seq) of peripheral blood and pleural effusion from patients with LMs to delineate their immune landscape. We identified an expansion of pro-inflammatory CD14+CD16+ monocytes and atypical memory B cells, accompanied by reduced cytotoxic CD8+ T and NK cells. Functional analysis revealed impaired antigen processing and presentation in CD14+ monocytes, and dysregulated transcription factor activity, potentially driving immune dysfunction. Additionally, LMs exhibited substantial remodeling of TCR and BCR repertoires, with shifts in clonality and diversity. Moreover, the CXCL16-CXCR6 interaction was associated with inflammatory responses, while upregulation of the inhibitory checkpoint HLA-E: CD94-NKG2A potentially contributed to impaired NK cell activity. Finally, we constructed a shared pro-inflammatory monocyte program and revealed S100A8 as a potential therapeutic target for LMs. We further demonstrated that S100A8 pharmacological inhibition could ameliorate the pathological lymphatic malformation phenotype. Collectively, our findings delineate cell type-specific immune dysregulation in LMs, offering insights for therapeutic development.
Pleural cavity macrophages promote lung tumor establishment through tissue invasion
GABPA ensures proper blastocyst development by suppressing SMAD3-mediated TGF-β signaling
Fibulin-7 in progenitor cells promotes adipose tissue fibrosis and disrupts metabolic homeostasis in obesity
Fibrosis, resulting from excess extracellular matrix (ECM) deposition, is a feature of adipose tissue (AT) dysfunction and obesity-related insulin resistance. Emerging evidence indicates that adipogenic stem and precursor cells (ASPCs) are a crucial origin of ECM proteins and possess the potential to induce AT fibrosis. Here, we employed single-cell RNA-seq and identified a unique subset of ASPCs that closely associated with ECM function. Within this subset, we discerned a notable upregulation in the expression of Fibulin-7 (FBLN7), a secreted glycoprotein, in obese mice. Similarly, in humans, FBLN7 levels exhibited an increase in visceral fat among obese individuals and demonstrated a correlation with clinical metabolic traits. Functional studies further revealed that, in response to caloric excess, ASPCs-specific FBLN7 knockout mice display a diminished state of AT fibrosis-inflammation, along with improved systemic metabolic health. Notably, the depletion of FBLN7 in ASPCs suppressed TGF-β-induced fibrogenic responses, whereas its overexpression amplified such responses. Mechanistically, FBLN7 interacted with thrombospondin-1 (TSP1) via its EGF-like calcium-binding domain, thereby enhancing the stability of the TSP1 protein. This, in turn, facilitated the conversion of latent TGF-β to its bio-active form, subsequently promoting TGFBR1/Smad signaling pathways. Furthermore, we developed an anti-FBLN7 neutralizing antibody, which could dramatically alleviate diet-induced AT fibrosis. These results suggest that FBLN7, produced by ASPCs, exerts a major influence in the development of AT fibrosis and may represent a potential target for therapeutic intervention.
Engineering oncolytic bacteria as precision cancer therapeutics: design principles, therapeutic strategies, and translational perspectives
Engineered oncolytic bacteria are emerging as a promising platform for precision cancer therapy, combining inherent tumor tropism, immunogenicity, and programmable gene control. Advances in synthetic biology now enable inducible and autonomous circuits that sense exogenous inputs (chemical signals or physical signals), bacterial self-cues (quorum sensing, bacterial invasion switches, or nitric oxide-responsive promoters), and tumor-specific pathophysiology (hypoxia, low pH, or lactate). These designs regulate colonization, lysis, and the spatiotemporally confined release of therapeutic cargos-including prodrug-converting enzymes, cytokines, and antibody/nanobody fragments-thereby enhancing antitumor efficacy while limiting off-target toxicity. Beyond monotherapy, oncolytic bacteria integrate with complementary modalities-including immune-checkpoint blockade, adoptive cell therapies (CAR-T/NK), radiotherapy/chemotherapy, nanomedicine, and oncolytic viruses-to amplify immune activation and to enable multimodal, synergistic regimens. Concurrently, biosensor modules transform bacterial chassis into programmable "microbial factories" that couple therapy with real-time imaging and adaptive responses within the tumor microenvironment. This review synthesizes design principles for bacterial gene regulation, surveys recent preclinical advances, and highlights emerging combination strategies, while outlining translational considerations for safety, manufacturability, and dosing, and patient selection. Together, these developments position engineered oncolytic bacteria as a promising route toward safe, effective, and ultimately personalized bacteria-based cancer therapeutics.
Tuning the Hsp70 chaperone cycle: emerging roles of GrpE-like NEFs in proteostasis and organelle function
The heat shock protein 70 (Hsp70) family of molecular chaperones is essential for nearly every cell to support protein homeostasis through folding, signaling, and quality control. Hsp70 functionality critically depends on co-chaperones, including the GrpE-like family of nucleotide exchange factors (NEFs), first identified in Escherichia coli as GrpE. These factors have long been recognized for their ability to catalyze the release of Hsp70 nucleotide and protein substrates, but recent structural and functional studies have revealed that GrpE-like NEFs are more than passive exchange catalysts, instead acting as dynamic regulators that coordinate chaperone activity with cellular stress responses, organelle-specific demands, and allosteric control of substrate binding and release. In this review, we synthesize decades of research on GrpE-like proteins across bacteria and eukaryotes, culminating in high-resolution structures of the human mitochondrial NEF, GrpEL1, in complex with mitochondrial Hsp70. We examine how architectural features of GrpE-like NEFs have evolved to meet specialized demands, such as thermosensing in bacteria, redox-responsive regulation in vertebrates, and coordination of protein import in mitochondria. We further describe how discrete structural domains dynamically control chaperone cycling, including nucleotide and substrate release, and how gene duplication and domain specialization have driven functional diversification in higher eukaryotes. Finally, we highlight emerging evidence linking NEF activity to mitochondrial homeostasis, stress adaptation, and disease, reframing GrpE-like NEFs as tunable regulators rather than static cofactors. This perspective positions them as stress-adaptive control points in proteostasis and offers a conceptual framework for understanding how ancient chaperone systems have evolved to meet the regulatory needs of modern and complex eukaryotic cells.
Deciphering the RNA Landscapes on Mammalian Cell Surfaces
Cell surface RNAs, notably glycoRNAs, have been reported, yet the precise compositions of surface RNAs across different primary cell types remain unclear. Here, we introduce a comprehensive suite of methodologies for profiling, imaging, and quantifying specific surface RNAs. We present AMOUR, a method leveraging T7-based linear amplification, to accurately profile surface RNAs while preserving plasma membrane integrity. By integrating fluorescently labeled DNA probes with live primary cells, and employing imaging along with flow cytometry analysis, we can effectively image and quantify representative surface RNAs. Utilizing these techniques, we have identified diverse non-coding RNAs present on mammalian cell surfaces, expanding beyond the known glycoRNAs. We confirm the membrane anchorage and quantify the abundance of several representative surface RNA molecules in cultured HeLa cells and human umbilical cord blood mononuclear cells (hUCB-MNCs). Our imaging and flow cytometry analyses unequivocally confirm the membrane localization of Y family RNAs, spliceosomal snRNA U5, mitochondrial rRNA MTRNR2, mitochondrial tRNA MT-TA, VTRNA1-1, and the long non-coding RNA XIST. Our study not only introduces effective approaches for investigating surface RNAs but also provides a detailed portrayal of the surface RNA landscapes of hUCB-MNCs and murine blood cells, paving the way for future research in the field of surface RNAs.
Circadian rhythm disruption declines oocyte quality for fertility via PTEN/AKT pathway
Structural and mechanistic insights into symmetry conversion in plant GORK K+ channel regulation
GORK is a shaker-like potassium channel in plants that contains ankyrin (ANK) repeats. In guard cells, activation of GORK causes K+ efflux, reducing turgor pressure and closing stomata. However, how GORK is regulated remains largely elusive. Here, we solved the cryo-EM structure of Arabidopsis GORK, revealing an unusual symmetry reduction (from C4 to C2) feature within its tetrameric assembly. This symmetry reduction in GORK channel is driven by ANK dimerization, which disrupts the coupling between transmembrane helices and cytoplasmic domains, thus maintaining GORK in an autoinhibited state. Electrophysiological and structural analyses further confirmed that ANK dimerization inhibits GORK, and its removal restores C4 symmetry, converting GORK to an activatable state. This dynamic switching between C2 and C4 symmetry, mediated by ANK dimerization, presents a GORK target site that guard cells regulate to switch the plant K+ channel between inhibited and activatable states, thus controlling stomatal movement in response to environmental stimuli.
