Results 201-225 of about 1,000
  1. Abstract Background The PELO–HBS1L complex is a critical mediator of ribosome-associated quality control, responsible for resolving stalled ribosomes and maintaining translational fidelity. Recent CRISPR-based synthetic lethality screens have identified PELO and HBS1L as selective vulnerabilities in cancers with 9p21.3 deletion or microsatellite instability-high (MSI-H) phenotypes. These cancers exhibit dependency on the PELO–HBS1L complex due to coexisting destabilization of the superkiller complex (SKIc), presenting a unique opportunity for targeted therapeutic intervention. However, no small-molecule inhibitors of this complex have been reported. Methods We employed AlphaFold2-multimer modeling to generate high-confidence structural models of the PELO–HBS1L heterodimer. Structural evaluation included predicted aligned error (PAE), per-residue confidence (pLDDT), and MSA coverage. Fragment-based pocket mapping identified a candidate binding pocket, selected for docking based on accessibility and proximity to inter-chain contacts. A fragment-guided design strategy was implemented using apogossypol scaffolds bearing hydroxyamide linkers and terminal polar groups. Ligands were prepared via Open Babel and docked into the interface pocket using AutoDock Vina. Binding energy, interaction geometry, and contact distances were used to rank binding poses. Results AlphaFold2 models revealed a rigid PELO core and a flexible HBS1L tail, with PAE and pLDDT scores indicating moderate interface confidence and ligand-accessible surface features. Indole-2-carboxylic acid with guanidine substitution formed key polar contacts in the candidate binding pocket. Among apogossypol analogs, methylamine and guanidine-tailed derivatives displayed strong polar interactions at the pocket interface, including engagement with GLU215, ASN, and THR. A methylated guanidine analog further enhanced interaction density, while piperidine substitutions were poorly tolerated. Conclusion A therapeutic small molecule docked in a binding pocket at the interface of two interacting proteins can disrupt a protein-protein complex. This approach, known as protein-protein interaction (PPI) inhibition, is a well-established strategy in drug discovery. The PELO–HBS1L interface contains a druggable surface pocket that can be targeted by small molecules. Apogossypol-derived ligands with flexible, cationic extensions demonstrate promising interaction profiles and provide a foundation for further hit-to-lead optimization. These findings support development of interface-directed inhibitors to exploit synthetic lethality in PELO-dependent cancers.
    Date: 2025-04-11
    Authors: Lehrer S, Rheinstein P.
    Ref: Research Square
  2. Abstract The oriental fruit fly Bactrocera dorsalis (Hendel) is considered as a quarantine pest in many countries and regions. Challenges remain in distinguishing this species with morphological similarities, especially in relevant development stages. In recent years, CRISPR-Cas12b genetic diagnostics has seen rapid advancements and offers an efficient tool for the identification of pathogens, viruses, and other genetic targets. Here we developed a new and rapid visual detection assay of B. dorsalis using recombinase polymerase amplification (RPA) and the CRISPR/Cas12b system. The system can detect different developmental stages of B. dorsalis within 30–35 minutes at 43 ℃ and the results are easily observed by the naked eye based on the color change in the tube during the reaction. The specificity and high sensitivity of this method was demonstrated, allowing for detection from 3.2 pg µL − 1 of DNA. With crude DNA, this diagnostic system successfully identified B. dorsalis by holding the reaction tubes in the hand. Our study demonstrates that RPA-CRISPR/Cas12b visualization system is effective to detect B. dorsalis rapidly and accurately. This approach can be applied for monitoring and identification of other pests in border and relevant locations, preventing biological invasions and ensuring pest control.
    Date: 2025-04-11
    Authors: Lv C, Zhang F, Ren L, Chen C, Zhu P, Cheng X, Yang X, Liu B.
    Ref: Research Square
  3. ABSTRACT Recently, de novo variants in an 18 nucleotide region in the centre of RNU4-2 were shown to cause ReNU syndrome, a syndromic neurodevelopmental disorder (NDD) that is predicted to affect tens of thousands of individuals worldwide 1,2 . RNU4-2 is a non-protein-coding gene that is transcribed into the U4 small nuclear RNA (snRNA) component of the major spliceosome 3 . ReNU syndrome variants disrupt spliceosome function and alter 5’ splice site selection 1,4 . Here, we performed saturation genome editing (SGE) of RNU4-2 to identify the functional and clinical impact of variants across the entire gene. The resulting SGE function scores, derived from variants’ effects on cell fitness, discriminate ReNU syndrome variants from those observed in the population and dramatically outperform in silico variant effect prediction. Using these data, we redefine the ReNU syndrome critical region at single nucleotide resolution, resolve variant pathogenicity for variants of uncertain significance, and show that SGE function scores delineate variants by phenotypic severity. Further, we identify variants impacting function in regions of RNU4-2 that are critical for interactions with other spliceosome components. We show that these variants cause a novel recessive NDD that is clinically distinct from ReNU syndrome. Together, this work defines the landscape of variant function across RNU4-2 , providing critical insights for both diagnosis and therapeutic development.
    Date: 2025-04-11
    Authors: De Jonghe J, Kim HC, Adedeji A, Leitão E, Dawes R, Chen Y, Blakes AJ, Simons C, Rius R, Alvi JR, Amblard F, Austin-Tse C, Baer S, Balton EV, Blanc P, Calame DG, Coutton C, Cunningham CA, Dargie N, Dipple KM, Du H, El Chehadeh S, Glass I, Gleeson JG, Grunewald O, Gueguen P, Harbuz R, Jacquemont M, Leventer RJ, Marijon P, Messaoud O, Sultan T, Thauvin C, Vincent-Delorme C, Yilmaz Gulec E, Thevenon J, Mendez R, MacArthur DG, Depienne C, Nava C, Whiffin N, Findlay GM.
    Ref: medRxiv
  4. The ribosome biases the conformations sampled by nascent polypeptide chains along folding pathways towards biologically active states. A hallmark of the co-translational folding (coTF) of many proteins are highly stable folding intermediates that are absent or only transiently populated off the ribosome, yet persist during translation well-beyond complete emergence of the domain from the ribosome exit tunnel. Intermediates are important for folding fidelity; however, their structures have remained elusive. Here, we have structurally characterised two coTF intermediates of an immunoglobulin-like domain by developing comprehensive 19 F NMR analyses using chemical shifts, paramagnetic relaxation enhancement (PRE), and protein engineering. We integrated these experimental data with extensive molecular dynamics (MD) simulations to obtain atomistic structures of the folding intermediates on the ribosome. The resulting intermediate structures are distinguished by native-like folds initiated from either their N-or C-termini, and reveal parallel folding pathways, which are structurally conserved within the protein domain family, in contrast to their in vitro refolding mechanisms. By redirecting proteins to fold along hierarchical, parallel routes, the ribosome may promote efficient folding by avoiding kinetic traps, and regulate nascent chain assembly and targeting by auxiliary factors to maintain cellular proteostasis.
    Date: 2025-04-11
    Authors: Chan SH, Streit JO, Włodarski T, Mitropoulou A, Cabrita LD, Christodoulou J.
    Ref: bioRxiv
  5. ABSTRACT Large scale loss-of-function screens utilising CRISPR or siRNA can provide profound insights into the importance of individual genes for the survival of a cancer cell and can drive the identification of therapeutic targets and biomarkers, and the development of targeted drugs. However, the analysis of these data and the substantial bodies of metadata that relate to them, is technically challenging and typically requires substantial expertise in data science and computer coding. To facilitate the analysis of cancer gene dependency data by cancer biologists and clinical scientists, we have developed DepMine – a computational toolkit providing a powerful system for framing complex queries relating cancer gene dependency to the underlying genetic changes that occur in cancer cells. DepMine identifies synthetic lethal relationships between putative target genes and complex ‘cancer profiles’ built from user-specified combinations of mutations, copy-number variation, and expression levels, and can refine these to optimal biomarker definitions for target dependency.
    Date: 2025-04-11
    Authors: Pearl LH, Pearl FM.
    Ref: bioRxiv
  6. SUMMARY While high and stable transgene expression can be achieved in undifferentiated pluripotent stem cells, conventional transgene expression systems are often silenced upon differentiation. Silencing occurs with both randomly integrated transgenes, introduced via transposase or lentiviral methods, and with transgenes targeted to specific genomic sites, including at commonly used safe harbor loci. The challenge to robustly express experimental transgenes in differentiated pluripotent stem cells is a major bottleneck in the field for applications such as CRISPR screening. Here, we conducted a comparative analysis to systematically evaluate the impact of various promoters, transcriptional regulatory elements, insulators, and genomic integration sites on transgene silencing during neuronal differentiation. Our findings reveal that specific combinations of promoters and transcriptional stability elements are able to prevent transgene silencing during differentiation, whereas chromatin insulators had less impact on silencing and three novel safe harbor integration sites performed similarly to the CLYBL locus. Guided by these insights we developed the PiggyBac vector TK4, which showed complete resistance to transgene silencing across various neuronal and microglial differentiation protocols from six different pluripotent stem cell lines, as independently confirmed by seven different laboratories. This construct will be highly useful for assays requiring stable transgene expression during differentiation, and holds the potential for broad applications in various research fields.
    Date: 2025-04-11
    Authors: Uenaka T, Napole AB, Saha AD, Sun D, Singavarapu A, Calzada L, Chen J, Erlebach L, McQuade A, Ramos D, Rigamonti A, Salazar L, Samelson AJ, Sedov K, Welsh NJ, Wild K, Wu Q, Arenas E, Bassett AR, Kampmann M, Kronenberg-Versteeg D, Merkle FT, Schüle B, Thompson LM, Skarnes WC, Ward ME, Wernig M.
    Ref: bioRxiv
  7. We present Alpha-BET, a structure-guided strategy leveraging AlphaFold to identify optimal ALFA-tag insertion sites for minimally disruptive labeling of viral glycoproteins with high-affinity nanobodies. Applied to HIV-1 Env, SARS-CoV-2 S, and NiV G, Alpha-BET preserves structural integrity and function. For HIV-1 Env, we demonstrate super-resolution DNA-PAINT MINFLUX 3D imaging enabled by tag insertion, showcasing its power for visualizing native trimers in single virions and potential for broader applications in virus research.
    Date: 2025-04-11
    Authors: Willliamson DJ, Zaza C, Carlon-Andres I, Gentili A, Daly JL, Holmes H, Thrush JW, Starling T, Neil S, Owens R, Malim MH, Tynan C, Simoncelli S, Padilla-Parra S.
    Ref: bioRxiv
  8. Abstract Genome editing by CRISPR-Cas9 is promising for genetic disease and cancer gene therapy. However, safety concerns are still present, particularly the ON-target genotoxicity for protocols using nucleases. Quality control of edited cells before and after graft is mandatory, especially to assay megabase-scale genomic rearrangements induced at the targeted locus. These unintended events are fortunately rare but potentially deleterious. Classical PCR-based bulk approaches do not detect them or underestimate their frequency. Single-cell approaches are promising but RNA sequencing only estimates copy number variation of the genome. Here, we propose single-cell DNA sequencing to accurately evaluate and monitor CRISPR-mediated genotoxicity in primary cells (human fibroblasts and hematopoietic stem/progenitor cells). We designed a homemade panel using single nucleotide polymorphisms to detect, map and characterize induced-losses of heterozygosity (terminal, interstitial, copy-loss and copy-neutral). This innovative approach revealed intense genotoxicity linked to the double strand break. The risk was associated with DNA repair modulators in particular DNA-PKcs inhibitor AZD7648. Importantly, the CDK4/6 inhibitor palbociclib prevented these rearrangements in hematopoietic stem/progenitor cells. This work strongly suggests that single-cell DNA sequencing should be routinely implemented in clinical applications before CRISPR-edited cell infusions.
    Date: 2025-04-11
    Authors: Moreau-Gaudry F, Boutin J, Fayet S, Marin V, BERGES C, Riandière M, Toutain J, Lamrissi-Garcia I, Thibault C, Cappellen D, Dabernat S, Poulet A, Francillette M, Droin N, Bedel A.
    Ref: Research Square
  9. Hemophilia B gene therapy treatments currently have not addressed the need for predictable, durable, active, and redosable factor IX (FIX). Unlike conventional gene therapy, engineered B Cell Medicines (BCMs) are durable, redosable, and titratable, and thus have the potential to address significant unmet needs in the Hemophilia B treatment paradigm. BE-101 is an autologous BCM comprised of expanded and differentiated B lymphocyte lineage cells genetically engineered ex vivo to secrete FIX-Padua. CRISPR/Cas9 mediated gene editing at the C-C chemokine receptor type 5 locus was used to facilitate transgene insertion of an AAV6-encoded DNA template via homology-directed repair. Transgene insertion did not alter B cell biology, viability, or differentiation into plasma cells. Appreciable levels of BE-101-derived FIX-Padua were detected within 1 day after IV administration in mouse and steady state was reached within 2 weeks and persisted for over 184 days. Redosing produced an increase in FIX-Padua production close to linear dose proportionality. Comprehensive genotoxicity analysis found no off-target issues of concern. No safety signals were observed in animal tolerability and GLP toxicology studies. In conclusion, BE-101 produces sustained levels of active FIX-Padua with the ability to engraft without host preconditioning and with the potential for redosing and titratability.
    Date: 2025-04-10
    Authors: Liu H, Singh S, Mullen TJ, Bullock C, Keegan S, Patterson T, Thakur S, Lundberg A, Shenker S, Couto R, Yadav C, Dastagir S, Li L, Bainter W, Liberzon E, Malloy CR, Lazzarotto CR, Ohsumi TK, Chilakala S, Chen H, Kshirsagar R, Hohmann AF, Arlauckas S, Lazorchak A, Scull C, Morgan RA.
    Ref: bioRxiv
  10. Simple Summary Diffuse midline gliomas (DMGs) are aggressive childhood brain tumours with no effective treatments. Over 80% of cases carry the histone H3K27M mutation, which alters chromatin structure and gene regulation and induce tumours growth. Immunotherapy, which uses the body’s immune system to fight cancer, relies on tumour- specific antigen molecules that immune cells can recognise. However, because DMGs have few mutations, finding suitable antigens for therapeutic purposes has been challenging. In this study, we investigated how the H3K27M mutation affects tumour antigen presentation in DMG. Using patient-derived DMG models, we found that H3K27M alters the landscape of antigens displayed on tumour cell surface, creating unique immune targets. We identified six immunogenic peptides, that triggered strong T cell responses. These antigens were absent when H3K27M was removed, confirming their link to the mutation. Our findings provide a blueprint for developing T cell-based immunotherapies for DMG, offering new hope for targeted treatments against this devastating disease. Background: Diffuse midline gliomas (DMGs) are among the most aggressive paediatric brain tumours, with the pathognomonic H3K27M mutation present in over 80% of cases. This mutation drives epigenetic dysregulation and transcriptional reprogramming, yet its impact on the tumour antigenic landscape remains poorly understood. Given the low mutational burden of DMG, an expanded search beyond neoantigens to include epigenetically dysregulated tumour-associated antigens (TAAs) is critical for advancing antigen-specific immunotherapies. Methods: To assess how H3K27M influences antigenic landscape of DMG, we performed a comprehensive immunopeptidomic analysis using patient-derived DMG cell line models (SU-DIPG13 and BT245) that harbour the H3K27M mutation and their CRISPR-edited H3K27M-knockout (KO) counterparts. High-resolution mass spectrometry and bioinformatics were employed to define H3K27M-driven changes in the immunopeptidome. Functional T cell assays using HLA-matched healthy donor PBMCs were conducted to evaluate the immunogenicity of H3K27M-associated peptides. Results: Our findings reveal that the H3K27M mutation reshapes the tumour antigenic landscape in a model-specific manner. While H3K27M knockout increased HLA-I expression in SU-DIPG13 but not BT245, immunopeptidomic profiling uncovered distinct shifts in the presentation of tumour-associated peptides, independent of direct effects on antigen processing machinery. Among these, we identified six immunogenic peptides, derived from SLITRK2, PRAME, XKR5, and CBX2, that elicited CD8⁺ T cell responses in in vitro functional assays. Notably, PRAME, a well-characterised cancer-testis antigen was confirmed as an H3K27M-associated immunogenic target, reinforcing its therapeutic relevance. Peptides identified exclusively in H3K27M+ cells were absent in KO models, demonstrating a direct link between H3K27M-driven transcriptional dysregulation and tumour antigenicity. Conclusions: This study provides the first systematic assessment of how H3K27M reshapes the antigenic landscape in DMG, uncovering novel, immunogenic tumour- associated peptides that could serve as targets for precision immunotherapy. By demonstrating that H3K27M mutation drives context-dependent antigen presentation, our findings establish a foundation for T cell-based therapies targeting H3K27M-associated antigens. These insights pave the way for next-generation personalised immunotherapies for this otherwise treatment-refractory disease. Key Points H3K27M mutation induces expression of tumour-associated antigens in DMG H3K27M alters the DMG immunopeptidome without uniformly changing HLA-I levels PRAME- and CBX2-derived peptides are immunogenic and targetable by CD8⁺ T cells Importance of Study Diffuse midline gliomas (DMGs) are universally fatal paediatric brain tumours with limited treatment options and poor immune visibility. While the H3K27M mutation is a defining hallmark, its impact on tumour immunogenicity remains unclear. This study presents the first comprehensive to explore the effect of H3K27M-mution on the DMG immunopeptidome, revealing six immunogenic peptides derived from epigenetically dysregulated tumour- associated antigens, including SLITRK2, PRAME, XKR5, and CBX2. These antigens elicited CD8⁺ T cell responses, establishing a direct link between H3K27M-driven transcriptional dysregulation and tumour antigenicity. By leveraging these altered antigens, we highlight actionable vulnerabilities for T cell-based immunotherapy.
    Date: 2025-04-10
    Authors: Shamekhi T, Zeng B, Sun CX, Daniel P, Kam Sian TCL, Goncalves G, Huang G, Fahimi F, Selvakumar N, Tanuwidjaya E, Woodhouse I, Kritzer B, Schittenhelm RB, Mazzieri R, Cain JE, Nazarin J, Hansford JR, Firestein R, Dolcetti R, Faridi P.
    Ref: bioRxiv
  11. Microgravity presents unique challenges to human physiology, particularly the immune system, during spaceflight. T lymphocytes, key components of adaptive immunity, play a vital role in immune regulation, yet the effects of microgravity on T-cell function and gene expression remain incompletely understood. This study, conducted as part of the MESSAGE (Microgravity Associated Genetics) Science Mission in the Axiom-3 mission ( https://www.nasa.gov/mission/station/research-explorer/investigation/?#id=9100 ). We aimed to investigate how microgravity influences immune cell responses by analysing blood samples from three astronauts between pre-flight and during their space mission on the ISS (post-launch Day 4, Day 7, and Day 10 in ISS). Hemogram analysis revealed no statistically significant differences in leukocyte, erythrocyte, haemoglobin, and haematocrit levels between pre-flight and in-flight samples, suggesting a stable haematological profile under microgravity conditions. T cell subpopulation analysis indicated fluctuations in effector memory T cells (TemEARLY and TemLATE), but these changes did not reach statistical significance. These findings suggest that, within the short duration of spaceflight, haematological and immune parameters remain largely stable, underscoring the need for further research into long-term immune adaptations in microgravity. As a next step, transcriptomic analysis will be performed to identify microgravity-associated gene candidates, and CRISPR-based knock-out of the genes in T cells will be generated to explore their functional roles in microgravity-induced immune modulation.
    Date: 2025-04-10
    Authors: Tastan C, Gulden G, Demir O, Cam E, Erkek F, Sert B, Gozler T, Yuksel I, Tekirdagli B.
    Ref: bioRxiv
  12. During their vectorial biosynthesis on the ribosome, elongating nascent polypeptide chains explore a range of conformational states towards their biologically functional structure. However, this high structural heterogeneity has limited their observation at high-resolution. Here, we have used an integrated structural biology approach to explore the structures of the multi-domain immunoglobulin-like FLN5-6 during its biosynthesis, capturing early folding through to native folding. We developed an in-silico purification approach for cryo-EM of ribosome-nascent chain complexes (RNCs), and integrated the resulting cryo-EM maps with NMR spectroscopy and atomistic molecular dynamics (MD) simulations to produce experimentally reweighted structural ensembles of RNC. The resulting atomistic structures reveal insights into the orientational heterogeneity of the nascent chain and its dynamic interactions with the ribosome. In particular, we find that two distinct pathways exist for nascent polypeptides in the exit tunnel vestibule, influenced by their stage of biosynthesis, folding conformational state and ribosomal RNA helices lining the tunnel. Our systematic analysis of the structures of nascent proteins translation-stalled at multiple time-points provides insights into how the ribosome dynamically modulates its pathway out of the exit tunnel to regulate its folding and accessibility for auxiliary factors of other co-translational events.
    Date: 2025-04-10
    Authors: Mitropoulou AN, Tomasz Włodarski T, Streit JO, Chan SH, Plessa E, Woodburn LF, Cabrita LD, Christodoulou J.
    Ref: bioRxiv
  13. ABSTRACT Human pluripotent stem cell (hPSC)-derived cardiac therapies hold great promise for heart regeneration but face major translational barriers due to allogeneic immune rejection. Here, we engineered hypoimmunogenic hPSCs using a two-step CRISPR-Cas9 strategy: (1) B2M knockout, eliminating HLA class I surface expression, and (2) knock-in of HLA-E or HLA-G trimer constructs in the AAVS1 safe harbor locus to confer robust immune evasion. Hypoimmunogenic hPSCs maintained pluripotency, efficiently differentiated into cardiac cell types that resisted both T and NK cell-mediated cytotoxicity in vitro , and self-assembled into engineered cardiac organoids. Comprehensive analyses of the hypoimmunogenic cells and organoids revealed preservation of transcriptomic, structural, and functional properties with minimal off-target effects from gene editing. In vivo , hypoimmunogenic cardiac organoids restored contractile function in infarcted rat hearts and demonstrated superior graft retention and immune evasion in humanized mice compared to wild-type counterparts. These findings establish the therapeutic potential of hypoimmunogenic hPSC-CMs in the cardiac organoid platform, laying the foundation for off-the-shelf cardiac cell therapies to treat cardiovascular disease, the leading cause of death worldwide.
    Date: 2025-04-09
    Authors: Silver SE, Howells AR, Arhontoulis DC, Randolph LN, Hyams NA, Barrs RW, Li M, Kerr CM, Robino RA, Morningstar JE, Bain JD, Floy ME, Norris RA, Bao X, Ruddy JM, Palecek SP, Ferreira LMR, Lian XL, Mei Y.
    Ref: bioRxiv
  14. Zebrafish (Danio rerio) has become a pivotal vertebrate model in biomedical research, renowned for its genetic similarity to humans, optical transparency, rapid embryonic development, and amenability to experimental manipulation. In recent years, the derivation of cell lines from zebrafish embryos has unlocked new possibilities for in vitro studies across developmental biology, toxicology, disease modeling, and genetic engineering. These embryo-derived cultures offer scalable, reproducible, and ethically favorable alternatives to in vivo approaches, enabling high-throughput screening and mechanistic exploration under defined conditions. This review provides a comprehensive overview of protocols for establishing and maintaining zebrafish embryonic cell lines, emphasizing culture conditions, pluripotency features, transfection strategies, and recent innovations such as genotype-defined mutant lines generated via CRISPR/Cas9 and feeder-free systems. We also highlight emerging applications in oncology, regenerative medicine, and functional genomics, positioning zebrafish cell lines as versatile platforms bridging animal models and next-generation in vitro systems. Their ongoing optimization holds promise for improved reproducibility, reduced animal use, and expanded translational impact in biomedical research.
    Date: 2025-04-09
    Authors: Arana ÁJ, González-Llera L, Barreiro-Iglesias A, Sánchez L.
    Ref: Preprints.org
  15. Summary Replication stress is a key driver of DNA damage and genome instability. Replication stress-induced fork remodelling generates a new DNA end that is vulnerable to the action of nucleases, and which is protected by a range of factors including the canonical tumour suppressors BRCA1 and BRCA2. Here we report that replication stress drives elevated production of cytokines and chemokines in the absence of DNA damage. The DNA sensor IFI16 binds nascent DNA at stalled replication forks and signals via the DNA sensing adaptor STING, to induce the activation of NF-κB and the production of pro-inflammatory cytokines in response to replication stress. IFI16 also acts directly at stalled replication forks to protect nascent DNA from degradation by the nucleases MRE11 and DNA2. Furthermore, IFI16 is required for the interferon-mediated rescue of fork protection in BRCA-deficient cells, highlighting the critical role of IFI16 in the cross-talk between innate immunity and fork protection during replication stress. Highlights Replication stress induces an early innate immune response, which is dependent on the DNA sensing factors IFI16 and STING, but not cGAS IFI16 binds directly to nascent DNA at stalled replication forks IFI16 prevents nucleolytic degradation of reversed forks IFI16 is required for interferon-mediated fork protection in BRCA-deficient cells
    Date: 2025-04-09
    Authors: Gamble A, Ward TA, Wheeler OP, Jones CM, Bennett LG, Vernon EG, Thanendran V, Morris JP, Ceppi I, Halder S, Borello D, Walker TDJ, Rajan J, Dunphy G, Cejka P, Unterholzner L, Staples CJ.
    Ref: bioRxiv
  16. Cell transfer experiments complement the rigorous investigation of antiviral and antitumor functions of natural killer (NK) cells. Success in these endeavors is enhanced by expansion of small numbers of input NK cells driven by viral antigens or homeostatic proliferation in immunodeficient hosts. In contrast, analysis of other NK-cell functions, including immunoregulation, are non-proliferative and require an intact immune system in recipient mice. We reveal poor persistence of conventional congenic (CD45.1) BoyJ NK cells following adoptive transfer in comparison to CRISPR-generated CD45.1+ (JAXBoy) NK cells. Reciprocal transfers between C57BL/6 and JAXBoy mice substantially improve seeding and maintenance of donor NK cells. Using this system, we confirm that CXCR3 re-positions NK cells in the white pulp of the spleen after infection, which is vital for immunoregulation. Moreover, we discovered that the transcription factor ASCL2 is required for recruitment of NK cells into the spleen and white pulp. These results provide improved tools and novel insights into NK cell biology. Key points JAXBoy are superior to conventional BoyJ mice for NK cell persistence after transfer. CXCR3 repositions donor NK cells in T/B-zones of the spleen after infection. The transcription factor ASCL2 is required for NK-cell recruitment to spleen white pulp.
    Date: 2025-04-09
    Authors: Canaday LM, Cox A, Feldman HA, Seelamneni H, Ali A, Tuazon JA, Calderon LB, Bennett SN, Yan A, Wilson M, Velu V, Waggoner SN.
    Ref: bioRxiv
  17. Guide RNA (gRNA) arrays can enable targeting multiple genomic loci simultaneously using CRISPR-Cas9. In this study, we present a streamlined and efficient method to rapidly construct gRNA arrays with up to 10 gRNA units in a single day. We demonstrate that gRNA arrays maintain robust functional activity across all positions, and can incorporate libraries of gRNAs, combining scalability and multiplexing. Our approach will streamline combinatorial perturbation research by enabling the economical and rapid construction, testing, and iteration of gRNA arrays.
    Date: 2025-04-09
    Authors: Salaudeen AL, Mateyko N, de Boer CG.
    Ref: bioRxiv
  18. Several members of the epidermal growth factor (EGF) family have been implicated in the biology of schizophrenia (Ketharanathan et al., 2024). The EGF-related ligand, Betacellulin (BTC), plays an important role in the proliferation and differentiation of neural stem cells and our group found markedly reduced BTC levels in patients with schizophrenia. Nevertheless, the interplay of affected BTC and its participation in neural specification and neurodevelopment remains elusive. We generated Knockout (KO) - BTC clones from an existing hiPSC line through CRISPR/Cas9-mediated modification. Furthermore, we validated BTC-KO through genotyping/sequencing, FACS and Western Blot. Finally, we demonstrated trilineage differentiation potential in vitro .
    Date: 2025-04-09
    Authors: Cota-Coronado A, Manning M, Kim D, Lee J, Gibbons A, Rosenbluh J, Hill RA, Sundram S.
    Ref: bioRxiv
  19. The CRISPR/Cas9 system is a powerful gene-editing tool. Its specificity and stability rely on complex allosteric regulation. Understanding these allosteric regulations is essential for developing high-fidelity Cas9 variants with reduced off-target effects. Here, we introduce a novel structure-based machine learning (ML) approach to systematically identify long-range allosteric networks in Cas9. Our ML model was trained using all available Cas9 structures, ensuring a comprehensive representation of Cas9 structural landscape. We then applied this model to Streptococcus pyogenes Cas9 (SpCas9) to demonstrate the feature selection process. Using the Cα-Cα inter-residue distances, we mapped key allosteric networks and refined them through a two-stage SHAP feature selection (FS) strategy, reducing a vast feature space to 28 critical Lysine-Arginine (Lys-Arg) residue pairs that mediate SpCas9 interdomain communication, stability, and specificity. These Lys-Arg pairs initially shared a 46.5Å inter-residue distance, but molecular dynamics simulations revealed distinct stabilization behaviors, indicating a hierarchical allosteric network. Further mutational analysis of R78A-K855A (M1) and R765A-K1246A (M2) identified an electrostatic valley, a stabilizing network where positively charged residues interact with negatively charged DNA to maintain SpCas9 structural integrity. Disrupting this valley through direct (M2) or allosteric (M1) mutations destabilized SpCas9 DNA-bound conformation, leading to distinct pathways for improving SpCas9 specificity. This study provides a new framework for understanding allostery in Cas9, integrating ML-driven structural analysis with MD simulations. By identifying key allosteric residues and introducing the electrostatic valley as a central concept, we offer a rational strategy for engineering high-fidelity Cas9 variants. Beyond Cas9, our approach can be applied to uncover allosteric hotspots in other enzyme regulation and rational protein design.
    Date: 2025-04-09
    Authors: Madugula SS, Jayasinghe-Arachchige VM, Norgan Radler C, Wang S, Liu J.
    Ref: bioRxiv
  20. Identification of new therapeutic targets in hepatocellular carcinoma (HCC) remains critical. Chromatin regulating complexes are frequently mutated or aberrantly expressed in HCC, suggesting dysregulation of chromatin environments is a key feature driving liver cancer. To investigate whether the altered chromatin state in HCC cells could be targeted, we designed and utilized an epigenome-focused CRISPR library that targets genes involved in chromatin regulation. This focused approach allowed us to test multiple HCC cell lines in both 2D and 3D growth conditions, which revealed striking differences in the essentiality of genes involved in ubiquitination and multiple chromatin regulators vital for HCC cell survival in 2D but whose loss promoted growth in 3D. We found the core subunits of the menin-MLL1 complex among the strongest essential genes for HCC survival in all screens and thoroughly characterized the mechanism through which the menin-MLL1 complex promotes HCC cell growth. Inhibition of the menin-MLL1 interaction led to global changes in occupancy of the complex with concomitant decreases in H3K4me3 and expression of genes involved in PI3K/AKT/mTOR signaling pathway. Menin inhibition affected chromatin accessibility in HCC cells, revealing that increased chromatin accessibility at sites not bound by menin-MLL1 was associated with the recruitment of the pioneer transcription factor complex NF-Y. A CRISPR/Cas9 screen of chromatin regulators in the presence of menin inhibitor SNDX-5613 revealed a significantly increased cell death when combined with NFYB knockout. Together these data show that menin-MLL1 is necessary for HCC cell survival and cooperates with NF-Y to regulate oncogenic gene transcription.
    Date: 2025-04-08
    Authors: Dzama-Karels M, Kuhlers P, Sokolowski M, Brinkman JA, Morris JP, Raab JR.
    Ref: bioRxiv
  21. DNA methyltransferase 1 inhibitor (DNMT1i) therapy is a promising option for increasing immune response as part of combination cancer therapy. High–grade serous ovarian carcinoma (HGSOC) is a highly aggressive cancer with poor survival outcomes, where DNMT1i therapy is being increasingly explored. HGSOC with epigenetically silenced BRCA1 has been shown to respond to PARP inhibitor (PARPi) treatment – a core targeted therapy for HGSOC. However, loss of silencing of even a single BRCA1 allele causes PARPi and platinum chemotherapy resistance. We tested whether BRCA1 silencing was robust to DNMT1i therapy, or would be reversed, thus driving PARPi resistance. We previously generated two homozygously silenced BRCA1 HGSOC cell lines: WEHI–CS62 and an OVCAR8 derivative. DNMT1i treatment caused sustained BRCA1 promoter methylation loss, gene re–expression and PARPi resistance in both of these silenced BRCA1 lines, but not in mutated BRCA1/2 or RAD51C lines. Methylation arrays confirmed transient global CpG methylation losses following DNMT1i. CRISPR deletion of the re–expressed BRCA1 copy in WEHI-CS62 restored silencing and PARPi sensitivity. Furthermore, DNMT1i treatment of a silenced BRCA1 PDX caused heterogeneous BRCA1 promoter methylation loss. In summary, DNMT1 inhibitors caused sustained reduction of BRCA1 promoter methylation in HGSOC cells. This resulted in BRCA1 re-expression and PARP inhibitor resistance, presenting a significant risk for up to 17% of HGSOC patients with BRCA1 gene silencing who could benefit from PARP inhibitor therapy. We conclude that DNA demethylation therapy should be avoided for HGSOC patients with epigenetically silenced BRCA1.
    Date: 2025-04-08
    Authors: Nesic K, Geissler F, Xu L, Kyran E, Beard S, Vandenberg CJ, Liddell B, Olechnowicz SW, Topp M, McNally O, Ratnayake G, Traficante N, Australian Ovarian Cancer Study, DeFazio A, Bowtell DD, Pappenfuss T, Zhang F, Dobrovic A, Waddell N, Scott CL, Kondrashova O, Wakefield MJ.
    Ref: bioRxiv
  22. ABSTRACT Spatial RNA organization plays a pivotal role in diverse cellular processes and diseases, but the functional implications of spatial RNA localization remain underexplored. We present CRISPR-mediated transcriptome o rganization (CRISPR-TO) that harnesses RNA-guided, nuclease-dead dCas13 for programmable control of RNA localization in live cells. CRISPR-TO enables targeted localization of RNAs to diverse subcellular compartments, including p-bodies, stress granules, telomeres, and nuclear stress bodies, across cell types. In primary cortical neurons, we demonstrate that repositioned mRNAs undergo local translation along neurites and at neurite tips and co-transport with ribosomes, with β -actin mRNA localization enhancing the formation of dynamic filopodial protrusions and inhibiting axonal regeneration. Furthermore, CRISPR-TO-enabled parallel screening in primary neurons identifies Stmn2 mRNA localization as a driver of neurite outgrowth. By enabling large-scale perturbation of the spatial transcriptome, CRISPR-TO bridges a critical gap left by current sequencing and imaging technologies, offering a versatile platform for high-throughput functional interrogation of RNA localization in living cells and organisms.
    Date: 2025-04-08
    Authors: Han M, Fu ML, Zhu Y, Choi AA, Li E, Bezney J, Cai S, Miles L, Ma Y, Qi S.
    Ref: bioRxiv
  23. Abstract Background: Dissecting the functional impact of genetic mutations is essential to advancing our understanding of genotype-phenotype relationships and identifying new therapeutic targets. Despite the progress in sequencing and CRISPR technologies, proteome-wide mutation effect prediction remains challenging. Here, we introduce ProteoCast, a scalable and interpretable computational method for proteome-wide classification of genetic variants and functional protein site identification. It relies solely on evolutionary information, leveraging protein sequence data across organisms. Results: Using ProteoCast, we generated mutational landscapes for 22,169 Drosophila melanogaster protein isoforms, categorising over 293 million amino acid substitutions as functionally neutral, uncertain, or impactful. We validated our predictions with over 380 thousand natural polymorphisms observed in the Drosophila Genetic Reference Panel (DGRP) and Drosophila Evolution over Space and Time (DEST) datasets and with FlyBase's developmentally lethal mutations. About 86% of known lethal mutations were classified as impactful or uncertain, versus only 13% and 18% of DGRP and DEST mutations. Moreover, we performed ProteoCastguided genome editing experiments, providing a proof-of-concept of the validity of this strategy. Beyond variant effect prediction, ProteoCast detected evolutionary conservation signals in about one-third of 40.5K annotated post-translational modification sites and 83% of ~90 known short linear motifs. These results support its usefulness for uncovering interaction and regulatory sites in unstructured protein regions. Conclusions: Our results demonstrate ProteoCast applicability for model organisms, contributing to basic genetic research and translational studies. This work provides a publicly available dataset, userfriendly interactive web services, and a locally deployable pipeline tool for further research into gene function and mutation effects in any organism.
    Date: 2025-04-08
    Authors: Rera M, Abakarova M, Freiberger M, Lierhmann A, Laine E.
    Ref: Research Square
  24. Enzymatic browning and cold-induced sweetening (CIS) affect the post-harvest quality of potato tubers. Browning is caused by Polyphenol Oxidase 2 (PPO2), which is activated by mechanical damage during harvest and storage. CIS occurs when vacuolar invertase converts sucrose into reducing sugars, which react with amino acids during frying, forming brown pigments and acrylamide. While cold storage prevents sprouting and disease, it also increases vacuolar invertase expression, leading to quality loss. Using CRISPR/Cas9, we developed gene-edited potato lines with improved resistance to browning and CIS. Line 6A (cv. Atlantic) and E03-3 (cv. Spunta) exhibited complete vacuolar invertase (InvVac) knockout, maintaining chip quality for at least 60 days at 4°C. Line 6A, renamed PIRU INTA, was tested in field trials and preserved frying quality for up to 90 days under cold storage. PIRU INTA is currently undergoing registration as a new variety. Additionally, lines E04-5B and E03-3 (cv. Spunta) showed partial PPO2 gene edits, reducing enzymatic browning by 80% and 40%, respectively. This study demonstrates the potential of CRISPR/Cas9 to develop non-transgenic, gene-edited potatoes with enhanced storage quality, benefiting both growers and the food industry.
    Date: 2025-04-07
    Authors: Massa GA, Décima Oneto CA, González MN, Poulsen Hornum A, Arizmendi A, Sucar S, Divito SB, Feingold SE.
    Ref: Preprints.org
  25. Hepatic stellate cells (HSCs) are critical for normal liver development and regeneration. Podocalyxin-like (podxl) is highly expressed in zebrafish HSCs, but its role in liver development is not known. Here we report that podxl knockdown using CRISPR/Cas9 (“CRISPants”) significantly decreased HSC number in zebrafish larvae at different time points and in two independent HSC reporter lines, supporting a role for podxl in HSC development. We generated five podxl mutants, including two mutants lacking the predicted podxl promoter region, and found that none of the mutants recapitulated the knockdown phenotype. Podxl CRISPR/Cas9 injection in mutants lacking the podxl guide RNA cut site did not affect HSC number, supporting the hypothesis that the CRISPant phenotype was specific, requiring intact podxl. Podxl mRNA levels in three podxl mutants were similar to those of wildtype controls. RNA sequencing of podxl mutants and controls showed no significant change in transcript levels of genes with sequence similarity to podxl , but it revealed upregulation of a network of extracellular matrix genes in podxl mutants. These results support a role for podxl in zebrafish liver development and suggest that upregulation of a group of functionally related genes represents the main mechanism of compensation for podxl genomic loss.
    Date: 2025-04-07
    Authors: Ross AN, Miscik NM, Kalasekar SM, Harris JD, Tran M, Saxena A, Baker SA, Evason KJ.
    Ref: bioRxiv
first · previous · 4 · 5 · 6 · 7 · 8 · 9 · 10 · 11 · 12 · 13 · next · last