Results 251-275 of about 1,000
  1. Epigenetic modifications play a crucial role in gene regulation and have been implicated in various physiological processes and disease conditions. DNA methylation (DNAm) has been implicated in the etiology and progression of many stress-related psychiatric behaviors such as depression. The ability to manipulate DNAm may provide a means to reverse and treat such disorders. Although CRISPR-based technologies have enabled locus-specific DNAm editing, their clinical applicability may be limited due to immunogenicity concerns and off-target effects. In this study, we introduce a novel approach for targeted DNAm manipulation using single-stranded, methylated DNA probes. We designed probes targeting the glucocorticoid response element (GRE) within the FKBP5 (FK506 binding protein 5) gene, a key regulator of stress response and depressive symptoms, and the promoter region of the MAOA (monoamine oxidase A) gene. In both HEK293 human embryonic kidney and mouse pituitary AtT-20 cells, transfection with their respective methylated probes significantly increased DNAm at targeted CpG sites in a persistent and dose-dependent manner. Importantly, the induced methylation effectively attenuated glucocorticoid-induced upregulation of FKBP5 gene expression. Alteration of methylation was specific to single-stranded probes, as double-stranded methylated probes and unmethylated probes showed no significant effects. Our findings suggest that methylated DNA probes have the potential to function as a simple tool for targeted epigenetic manipulation and serve as a safer alternative to CRISPR-based epigenome editing tools for the treatment of stress-related disorders such as depression.
    Date: 2025-04-01
    Authors: Modi N, Guo J, Lee R, Greenstein A, Lee R.
    Ref: Preprints.org
  2. Abstract Background Propionic acidemia (PA) is a rare autosomal recessive metabolic disorder caused by defects in propionyl-CoA carboxylase (PCC), a mitochondrial enzyme composed of six alpha (PCCA) and six beta (PCCB) subunits. Mutations in PCCA/PCCB genes disrupt PCC function, leading to toxic metabolite accumulation and clinical manifestations. Current research is limited by inadequate patient-derived cellular models and ethical constraints in sample acquisition. Method Using CRISPR/Cas9-mediated gene editing, we established an isogenic human induced pluripotent stem cell (iPSC) line carrying the PCCA c.2002G > A mutation. Results The mutant iPSCs showed sustained expression of pluripotency markers (OCT4, NANOG, SOX-2), maintained normal karyotype (46, XY), and retained trilineage differentiation capacity. Functional characterization demonstrated significantly reduced PCC enzyme activity, accurately modeling PA metabolic pathology. Conclusions This isogenic iPSC line provides an ethically unconstrained platform to investigate PA molecular mechanisms and genotype-phenotype relationships. The model enables systematic drug screening and therapeutic development while overcoming patient sample limitations.
    Date: 2025-04-01
    Authors: Tao T, Lin L, Tang Y, Liu Z, Liu Y, Xie Y, Hu X, Wang J, Wang T, Zhang G, Wang Y, Zhu S.
    Ref: Research Square
  3. The CRISPR/Cas9 system, derived from the adaptive immune defence of bacteria and archaea, has emerged as a powerful tool for genome engineering by enabling precise, site-specific modifications. Type II bacterial CRISPR systems recognize and cleave DNA in various microorganisms using the RNA-guided Cas9 endonuclease. We present a streamlined and efficient approach for genome editing in two strains of Saccharomyces cerevisiae , focused on targeted mutagenesis of the ADE 2 gene of the BY4741 strain and homology-directed repair of the ade2 locus of the W303-1A strain, involving a single-step transformation strategy utilizing the pML104 plasmid, which encodes the Cas9 endonuclease and a user-defined single-guide RNA (gRNA) complementary to the target site. In the BY4741 (wildtype) strain, pML104-gRNA components achieved successful ADE2 disruption rates of 40% (95% CI: 0.10–0.70), confirmed by adenine auxotrophy screening and Sanger sequencing. Additionally, co-transformation of gRNA plasmid and donor repair templates in the W303-1A ( ade2 mutant) strain also resulted in 40% recombination efficiency, restoring the function of the mutant gene to allow activation of de novo purine biosynthesis activity when cultured on SC-Ade medium. This optimized, cost-effective CRISPR/Cas9 method enhances editing efficiency in S. cerevisiae , providing a robust and practical platform for high-precision genetic modifications with broad applications in molecular biology and biotechnology.
    Date: 2025-04-01
    Authors: Jarra E.
    Ref: bioRxiv
  4. Protein modification by interferon-stimulated gene 15 (ISG15), termed ISGylation, exhibits antiviral properties and influences tumorigenesis, genome stability and metabolic processes. ISGylation is counteracted by the specific protease USP18. Likewise, viral proteases such as the papain-like protease (PLpro) from SARS-CoV-2 cleave ISG15 to undermine the host immune response. Beyond its role as a deISGylating enzyme, USP18 acts as a major negative regulator of the IFN signaling pathway in a STAT2-dependent manner. In humans, unconjugated ISG15 secures USP18 stability and the absence of USP18 or impaired STAT2/USP18 binding cause fatal interferonopathies. Thus, the USP18 signaling hub represents a critical checkpoint for type I IFN signaling and ISGylation, qualifying it as a promising immune and cancer drug target. However, suitable assays to monitor protein-protein interactions (PPIs) within the USP18/ISG15/STAT2 signaling hub and to screen for PPI modulators are missing and no specific inhibitors targeting USP18 interactions are available. To address this gap, we developed a method based on the NanoLuc luciferase (NLuc) Bioluminescence Energy Transfer (NanoBRET) assay system to study PPIs. Firstly, we generated stable cell lines suitable to monitor USP18/ISG15 and USP18/STAT2 interactions, providing a semi high-throughput screening (HTS)-compatible platform. In combination with a virtual pre-screen of 60,000 compounds against USP18 in silico , this assay allowed us to identify a first small molecule (ZHAWOC8655) that compromises cellular USP18/ISG15 binding and inhibits USP18 protease activity in vitro . To further explore the potential of using the NanoBRET system for testing PPI modulators, we evaluated the effect of GRL0617, a compound which was shown to disrupt the interaction between SARS-CoV-2 PLpro/ISG15 as well as SARS-CoV-2 PLpro/ubiquitin. NanoBRET based stable cell lines as presented here will be suitable for monitoring PPIs in other multiprotein complexes after various stimuli, mutations or small molecule administration and can be challenged with siRNA or CRISPR/Cas9 libraries to identify previously unrecognized regulators.
    Date: 2025-04-01
    Authors: Hess S, Alonso MC, Brand M, Lauw S, Lindenmann U, Göbel K, Geurink PP, Fritz G, Riedl R, Knobeloch K.
    Ref: bioRxiv
  5. In the evolutionary battle between bacteria and mobile genetic elements, such as bacteriophage viruses and plasmids, bacteria have developed intricate defense systems. Among these, the CRISPR-Cas system has been extensively studied and harnessed as a revolutionary gene editing tool. However, while the biochemical process by which this microbial immune system acquires genetic CRISPR memory and immunity against invaders has been comprehensively examined, fundamental questions about the bacterial physiological state underlying how and when CRISPR memory is formed have only been partially explored. Naïve CRISPR adaptation is generally rare, but occurs more frequently when bacteria are challenged with replication-deficient phages. In such scenarios, bacteria are not under immediate threat and have ample time to adapt to the phage DNA, without risking cell death. Accordingly, slow growth caused by low temperatures, low aeration, or bacteriostatic antibiotics promotes CRISPR adaptation, possibly by allowing the Cas complexes more time to adapt before being outpaced. Persister cells are dormant antibiotic-tolerant subpopulations of cells with limited metabolic activity. When a mobile genetic element invades a persister cell, its replication is halted until the host cell resumes growth, providing an ideal opportunity for CRISPR adaptation. Here, we show that transiently dormant Escherichia coli persister cells acquire CRISPR immunity 10-fold more frequently than the general bacterial population. Thus, persister cells, in addition to being notoriously antibiotic tolerant, are primed for CRISPR-Cas adaptation and may be in a state of heightened immune capacity and evolution, securing the survival of the population.
    Date: 2025-04-01
    Authors: Mauritzen JJ, Wajn AS, Hitz IF, Høyland-Kroghsbo NM.
    Ref: bioRxiv
  6. ABSTRACT While GWAS have been successful in providing variant-to-trait associations for human complex diseases, functional dissection of the discovered loci has lagged behind. Here, we describe a variant-to-gene (V2G) mapping effort for Alzheimer’s disease (AD) to implicate causal variants and effector genes from the most recent AD GWAS meta-analyses (101 loci). We leveraged our genomics datasets comprising high-resolution promoter Capture C, ATAC-seq, and RNA-seq from brain-relevant cell types to fine-map AD GWAS variants, identifying 89 candidate causal SNPs and 69 effector genes. We then designed a single-cell CRISPRi screen to perturb candidate regulatory regions (n=74) and assess the transcriptional response in the human microglial cell line, HMC3. Our screen across ∼97,000 cells identified 19 regulatory regions and 19 effector genes. We then elected to functionally dissect our top hit, the TSPAN14 locus, and we show that an intronic region containing AD-associated SNPs rs7080009, rs1870138, and rs1870137 is a microglia-specific enhancer, with the AD risk haplotype increasing its activity. CRISPR precise genomic deletion of this region decreases TSPAN14 expression, alters specific cellular pathways including cell adhesion, and decreases secreted levels of pro-inflammatory cytokines IL-6 and IL-8, which are known biomarkers of aging and AD. Our work provides a systematic framework to map GWAS signals to their effector genes for AD and other brain-related disorders, and provides robust leads to follow up with in-depth functional investigations.
    Date: 2025-04-01
    Authors: Laub S, Tulina N, Hoffman M, Faryean J, Ramachandran S, Trang K, Lewkiewicz S, Chesi A.
    Ref: bioRxiv
  7. ABSTRACT Synthetic lethality (SL) is an extreme form of negative genetic interaction, where simultaneous disruption of two non-essential genes causes cell death. SL can be exploited to develop cancer therapies that target tumour cells with specific mutations, potentially limiting toxicity. Pooled combinatorial CRISPR screens, where two genes are simultaneously perturbed and the resulting impacts on fitness estimated, are now widely used for the identification of SL targets in cancer. Various scoring methods have been developed to infer SL genetic interactions from these screens, but there has been no systematic comparison of these approaches. Here, we performed a comprehensive analysis of 5 scoring methods for SL detection using 5 combinatorial CRISPR datasets. We assessed the performance of each algorithm on each screen dataset using two different benchmarks of paralog synthetic lethality. We find that no single method performs best across all screens but identify two methods that perform well across most datasets. GRAPHICAL ABSTRACT Figure 1. Graphical Abstract. Benchmarking Scoring Methods for Synthetic Lethality Detection from CRISPR screen data. Experimental setup for benchmarking experiments. Five different CRISPR double knockout (DKO) screens are scored for genetic interaction using 5 different scoring methods. The calculated scores are analysed using two different benchmarks (De Kegel Hits and Köferle Hits). Area under the receiver operating characteristic curve (AUROC) and Area under the precision recall curve (AUPR) for each scoring method on each dataset are calculated and compared.
    Date: 2025-04-01
    Authors: Ajmal H, Nandi S, Kebabci N, Ryan CJ.
    Ref: bioRxiv
  8. The mouse model is by far the most widely used animal model in preclinical neuroscience, but translating findings to humans suffers from the lack of a formal framework establishing the correspondence between the mouse and the human brain. In this study, we build on the concept of common brain space, and on previous work embedding gene expression profiles, to bring the two species into alignment for comparative analysis. Using a variational autoencoder (VAE) combined with a latent classifier, we create a latent space that strikes a balance between abstract features related to reconstruction and features pertaining to regional segregation. This approach demonstrates improved cross-species homology and within-species locality compared to existing comparative models. In addition, we show that brain alterations in mouse disease models can be translated to humans, predicting patterns of brain changes in Alzheimer's and Parkinson's diseases. The flexibility and scalability of this approach offer a promising framework to bridge between more animal models, comparing quantitative imaging modalities, and disease phenotypes. This in turn will help advance our understanding of species similarities and differences, enhancing both fundamental translational neuroscience and disease modelling.
    Date: 2025-04-01
    Authors: Jaroszynski CYG, Amer M, Beauchamp A, Lerch JP, Sotiropoulos S, Mars RB.
    Ref: bioRxiv
  9. ABSTRACT RNA sequencing studies on human dorsal root ganglion (hDRG) from patients suffering from neuropathic pain show upregulation of OSM, linking this IL-6 family cytokine to pain disorders. In mice, however, OSM signaling causes itch behaviors through a direct effect on its cognate receptor expressed uniquely by pruriceptive sensory neurons. We hypothesized that an expansion in function of OSM-OSM receptor (OSMR) in sensory disorders in humans could be explained by species differences in receptor expression and signaling. Our in situ hybridization and immunohistochemical findings demonstrate broad expression of OSMR in DRG nociceptors and afferent fibers innervating the superficial and deep skin of humans. In patch-clamp electrophysiology, OSM directly activates human sensory neurons engaging MAPK signaling to promote action potential firing. Using CRISPR editing we show that OSM activation of MAPK signaling is dependent on OSMR and not LIFR in hDRG. Bulk, single-nuclei, and single-cell RNA-seq of OSM-treated hDRG cultures reveal expansive similarities in the transcriptomic signature observed in pain DRGs from neuropathic patients, indicating that OSM alone can orchestrate transcriptomic signatures associated with pain. We conclude that OSM-OSMR signaling via MAPKs is a critical signaling factor for DRG plasticity that may underlie neuropathic pain in patients.
    Date: 2025-04-01
    Authors: Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Gabriel KA, Palomino S, Li Y, Uhelski ML, Shiers S, Franco-Enzástiga Ú, Wangzhou A, Lesnak JB, Bandaru S, Shrivastava A, Inturi N, Albrecht PJ, Dockum M, Cervantes AM, Horton P, Funk G, North RY, Tatsui CE, Corrales G, Yousuf MS, Curatolo M, Gereau RW, Patwardhan A, Dussor G, Dougherty PM, Rice FL, Price TJ.
    Ref: bioRxiv
  10. Exonic enhancers (EEs) occupy an under-appreciated niche in gene regulation. By integrating transcription factor binding, chromatin accessibility, and high-throughput enhancer-reporter assays, we demonstrate that many protein-coding exons possess enhancer activity across species. These EEs exhibit characteristic epigenomic signatures, form long-range interactions with gene promoters, and can be altered by both nonsynonymous and synonymous variants. CRISPR–mediated inactivation demonstrated the involvement of EEs in the cis-regulation of host and distal gene expression. Through large-scale cancer genome analyses, we reveal that EE mutations correlate with dysregulated target-gene expression and clinical outcomes, highlighting their potential relevance in disease. Evolutionary comparisons show that EEs exhibit both strong sequence constraint and lineage-specific plasticity, suggesting that they serve ancient regulatory functions while also contributing to species divergence. Our findings redefine the landscape of functional elements by establishing EEs as a component of gene regulation, while revealing how coding regions can simultaneously fulfil both protein-coding and cis-regulatory roles.
    Date: 2025-04-01
    Authors: Mouren J, Torres M, van Ouwerkerk A, Manosalva I, Gallardo F, Spicuglia S, Ballester B.
    Ref: bioRxiv
  11. Cells generate purine nucleotides through both de novo purine biosynthesis (DNPB) and purine salvage. Purine accumulation represses energetically costly DNPB through feedback inhibition of the enzymatic steps that produce the precursor phosphoribosylamine. Excessive DNPB is associated with human diseases including neurological dysfunction and hyperuricemia. However, the mechanisms explaining how cells balance DNPB and purine salvage are incompletely understood. Data from a genome-wide CRISPR loss-of-function screen and extensive stable isotope tracing identified Nudix hydrolase 5 (NUDT5) as a suppressor of DNPB during purine salvage. NUDT5 ablation allows DNPB to persist in the presence of either native purines or thiopurine drugs; this renders NUDT5-deficient cells insensitive to thiopurine treatment. Surprisingly, this regulation occurs independently of NUDT5’s known function in hydrolyzing ADP-ribose to AMP and ribose-5-phosphate. Rather, NUDT5 interacts with phosphoribosyl pyrophosphate amidotransferase (PPAT), the rate-limiting enzyme in DNPB that generates phosphoribosylamine. Upon induction of purine salvage, the PPAT-NUDT5 interaction is required to trigger disassembly of the purinosome, a cytosolic metabolon involved in efficient DNPB. Mutations that disrupt NUDT5’s interaction with PPAT but leave its catalytic activity intact permit excessive DNPB during purine salvage, inducing thiopurine resistance. Collectively, our findings identify NUDT5 as a regulator governing the balance between DNPB and purine salvage, underscoring its impact on nucleotide metabolism and efficacy of thiopurine treatment.
    Date: 2025-04-01
    Authors: Wu Z, Nguyen PT, Sondhi V, Yao R, Dai T, Chiang J, Shang Z, Cai F, Cai L, Zhang J, Moore MD, Alshamleh I, Li X, Ogu T, Zacharias LG, Winston R, Patricio JS, Johnson X, Chen W, Cong Q, Mathews TP, Zhang Y, DeBerardinis RJ.
    Ref: bioRxiv
  12. Objectives Candidozyma ( Candida ) auris is an emerging fungal pathogen of global concern that often exhibits multi-drug resistance. Over 90% of isolates are resistant to fluconazole. Of the six described clades of C. auris , Clade III has been found to be nearly universally fluconazole resistant and almost every Clade III isolate described carries a mutation in the gene encoding the fluconazole target sterol demethylase ( ERG11 ) leading to a VF125AL substitution and a mutation leading to a N647T substitution in the gene encoding Mrr1a, a transcriptional regulator of the Mdr1 transporter. Both mutations have been shown to contribute to fluconazole resistance in C. auris . Methods In the present study we introduced the Clade III MRR1A mutation into a Clade I background using CRISPR-Cas9 gene editing. In two Clade III clinical isolates we corrected the native MRR1A and ERG11 mutations to their wild-type sequences as well as disrupted MDR1 . Triazole susceptibilities and MDR1 gene expression were measured in all strains. Results Introduction: of the N647T substitution in a Clade I background confers a modest reduction in fluconazole and voriconazole susceptibility. Similarly, correction of MRR1A or disruption of MDR1 in each Clade III background resulted in a one-dilution decrease in fluconazole and voriconazole MIC while the ERG11 correction resulted in a three-dilution decrease in fluconazole and voriconazole MIC. Conclusions Our findings show that while the MRR1A mutation makes a modest contribution, the ERG11 mutation is responsible for most of the fluconazole resistance observed in Clade III isolates. We also show that while these mutations likewise affect voriconazole susceptibility, they have no effect on susceptibility to itraconazole, isavuconazole, or posaconazole suggesting the potential therapeutic utility of these antifungals for infections due to Clade III isolates of C. auris .
    Date: 2025-03-31
    Authors: Barker KS, Zhang Q, Peters TL, Rybak JM, Morschhäuser J, Cuomo CA, Rogers PD.
    Ref: bioRxiv
  13. Interplay among gene-editing technologies, artificial intelligence (AI), and nanotechnology is revolutionizing personalized drug delivery with greater accuracy, efficiency, and individualized treatment regimens. This paper summarizes the development of lipid nanoparticles, vesicular drug carriers, and intelligent drug delivery systems and how these could improve drug bioavailability and targeted therapies. AI-based predictive models are revolutionizing drug discovery, formulation, and personalized treatment development to enable more efficient and personalized therapy.In addition, nanorobotics and magnetically triggered drug delivery are facilitating site-specific therapy with the specific significance in neurology and oncology. Further, CRISPR-based gene editing and artificial intelligence are facilitating precision of therapy with highly targeted nature's gene therapy. Advanced academe and industry technology are foreseeing the future for autonomous delivery systems and real-time monitoring facilitated by artificial intelligence to redefine precision medicine's era.This article highlights the revolutionizing capability of such inter-disciplinary advances and their ability to redefine contemporary therapeutics. Medicine in the future, supported by AI, nanotechnology, and gene editing, will be more effective, more specific, and patient-centric. Increased research and technological development will be the driving force for these advances to reach the clinic, and the outcome of therapy will become safer and more efficient.
    Date: 2025-03-31
    Authors: Sengar A.
    Ref: Preprints.org
  14. The growing threat of antimicrobial resistance has driven the search for new bioactive compounds in extreme environments such as Antarctica. Streptomyces fildesensis So13.3, isolated from Antarctic soil, has been shown to contain a biosynthetic gene cluster associated with producing actinomycin D, an antibiotic with therapeutic potential. In this study, we analysed the regulatory role of TetR/AcrR family transcription factors present within this BGC, focusing on their activation under different nutritional conditions and their structural scharacterisation using bioinformatics tools and molecular dynamics simulations. The results showed that TetR/AcrR expression increased significantly in ISP4 and IMA media, suggesting their involvement in nutrient-dependent regulation of the cluster. At the structural level, two TetR proteins (TetR-206 and TetR-279) were modelled, with the latter standing out due to a C-terminal tetracycline repressor-like domain. A 200 ns molecular dynamics simulation was performed in GROMACS to evaluate the stability and flexibility of TetR-279, including analysis of point mutations (S166P, V167A, V167I). The S166P mutation significantly impacted structural flexibility, while V167A and V167I caused only minor alterations. This work demonstrates the value of integrating omics approaches, structural modelling, and gene editing with CRISPR to study and potentially activate silent BGCs in non-model bacteria such as Antarctic Streptomyces. In particular, the targeted inhibition of TetR-279 may trigger metabolic rewiring and facilitate the expression of novel antibiotics encoded in cryptic biosynthetic gene clusters.
    Date: 2025-03-31
    Authors: Leal K, Gajardo HA, Contreras MJ, Palma M, Machuca Aguirre J, Nuñez Montero K, Gutierrez A, Barrientos L.
    Ref: Preprints.org
  15. Neuron cell culture stands at the forefront of neuroscience innovation, offering unparalleled insights into neuronal development, pathology, and regeneration. This review critically examines advances and persistent obstacles in culturing neurons, with a focus on axon and dendrite growth—a pivotal yet underexplored frontier for regenerative medicine. We synthesize breakthroughs in extracellular matrix (ECM) engineering, 3D biomimetic microenvironments, and molecular interventions while highlighting intrinsic challenges such as limited neuronal longevity, tumorigenicity risks in stem cell approaches, and reproducibility gaps. Introducing a biomimetic engineering framework, we liken neuronal regeneration to a multidimensional optimization problem, where balancing mechanical, biochemical, and epigenetic variables dictates functional outcomes. Key findings include: 1. 3D hydrogels mimicking brain ECM enhance neurite outgrowth by 40–60% compared to 2D systems. 2. Secretory pathway disparities between axons and dendrites reveal evolutionarily conserved growth mechanisms. 3. Tumorigenicity remains a critical barrier, with CRISPR-Lin28-edited iPSCs reducing teratoma formation by 65% in preclinical models. We advocate for standardized, scalable protocols and CRISPR-epigenetic tools to silence inhibitory pathways (e.g., Nogo-A). By bridging in vitro models with clinical translation, this work charts a roadmap for overcoming regenerative bottlenecks in neurodegenerative diseases and CNS injuries.
    Date: 2025-03-31
    Authors: Zehtabian K.
    Ref: Preprints.org
  16. Prokaryotes carry clusters of phage defense systems in “defense islands” that have been extensively exploited bioinformatically and experimentally for discovery of immune functions. However, little effort has been dedicated to determining which specific system(s) within defense islands limit lytic phage reproduction in clinical bacterial strains. Here, we employed the CRISPR-based Cascade-Cas3 system to delete defense islands in a Pseudomonas aeruginosa clinical isolate to identify mechanisms of lytic phage antagonism. Deletion of one island in a cystic fibrosis-derived clinical isolate sensitized the strain to phages from the Pbunavirus family, which are commonly used as therapeutics. The causal defense system is a Type IIS restriction endonuclease-like protein (END PaCF1 ), common in Pseudomonads, however it lacks an associated methyltransferase typical Type IIS R-M systems. END PaCF1 protects bacteria against phages with hypermodified DNA and is surprisingly agnostic to the specific structure of the modification, which is unlike typical type IV restriction endonucleases. In END PaCF1 , the endonuclease domain is fused to a catalytically inactive Endonuclease III (iEndoIII), a domain that recognizes non-canonical bases to repair DNA in prokaryotes and eukaryotes. We therefore propose that nucleases containing an i En doIII d omain ( END nucleases) can sense diverse DNA hypermodifications. Our findings reveal modularity of the sensing and cleavage domains, as expected of a modification-dependent endonucleases. We further show that some hypermodified phages, including Pbunavirus family members and Wrowclawvirus family (Pa5oct-like) of jumbo phages, encode END nuclease inhibitors that directly bind to the nuclease, likely via the iEndoIII domain. These inhibitors are necessary for Pbunavirus to plaque on clinical isolates and sufficient to enable other hypermodified phages to plaque in the presence of this defense system.
    Date: 2025-03-31
    Authors: Yee W, Lee Y, Klein TA, Wirganowicz A, Gabagat AE, Csörgő B, Makarova KS, Koonin EV, Weigele PR, Bondy-Denomy J.
    Ref: bioRxiv
  17. Prairie voles ( Microtus ochrogaster ) are a powerful model for studying the neurobiology of social bonding, yet tools for region- and cell type-specific gene regulation remain underdeveloped in this species. Here, we present a lentivirus-mediated CRISPR activation and interference (CRISPRa/i) platform for somatic gene modulation in the prairie vole brain. This system enables non-mutagenic, titratable regulation of gene expression in the adult brain without germline modification. Our dual-vector system includes one construct expressing dCas9-VPR (CRISPRa) or dCas9-KRAB-MeCP2 (CRISPRi) under a neuron-specific promoter, and a second construct delivering a U6-driven sgRNA alongside an EF1α-driven mCherry reporter. We detail the design, production, and stereotaxic delivery of these tools and demonstrate their application by targeting four genes implicated in social behavior ( Oxtr, Avpr1a, Drd1, Drd2 ) across two mesolimbic brain regions: the nucleus accumbens and ventral pallidum. Gene expression analyses confirmed robust, bidirectional transcriptional modulation for select targets, establishing proof of concept for CRISPRa/i in this non-traditional model. The dual-vector design is readily adaptable to other gene targets, cell types, and brain regions, and can be multiplexed to provide a flexible and scalable framework for investigating gene function in behaviorally relevant circuits. These advances represent the first successful implementation of somatic CRISPRa/i in prairie voles and expand the genetic toolkit available for this species.
    Date: 2025-03-31
    Authors: Loth MK, Mesch KT, Herrera-Garcia C, Brusman LE, Donaldson ZR.
    Ref: bioRxiv
  18. Effective and scalable sex separation remains a critical challenge for mosquito genetic control strategies. Genetic sexing strains (GSS) address this by genetically linking maleness with selectable traits, enabling efficient removal of females before release. Here, we describe a robust platform for the development of GSSs in the invasive Aedes albopictus mosquito by integrating a CRISPR-engineered selectable phenotype with sex conversion via nix , the male-determining factor. As a proof-of-concept, we disrupt the yellow gene to generate a vivid pigmentation marker, then rescue its function in males using nix -containing transgenes, creating a stable strain where all females are yellow and all engineered males are dark. The resulting GSS males are fertile, robust, and despite lacking the ancestral M locus, exhibit gene expression profiles closely resembling wild-type males. We benchmark sex separation based on pigmentation and discover that yellow mutant females exhibit slower larval development, enhancing protandry-based sorting. The GSS strain is compatible with existing size-based sex sorting systems, allowing for improved separation accuracy through the integration of natural and engineered sexually dimorphic traits. Additionally, we find that GSS females lay desiccation-sensitive eggs, reducing the risk of accidental female releases. Our approach is the first to engineer a sex-linked selectable trait by precisely targeting an endogenous gene and restoring its function in males, establishing a versatile platform for GSS development in Aedes mosquitoes.
    Date: 2025-03-31
    Authors: Zaada DS, Toren O, Krsticevic F, Haber DA, Gildman D, Galpaz N, Häcker I, Schetelig MF, Marois E, Arien Y, Papathanos PA.
    Ref: bioRxiv
  19. A major challenge in human evolutionary biology is to pinpoint genetic differences that underlie human-specific traits, such as increased neuron number and differences in cognitive behaviors. We used human-chimpanzee tetraploid cells to distinguish gene expression changes due to cis -acting sequence variants that change local gene regulation, from trans expression changes due to species differences in the cellular environment. In neural progenitor cells, examination of both cis and trans changes — combined with CRISPR inhibition and transcription factor motif analyses — identified cis -acting, species-specific gene regulatory changes, including to TNIK, FOSL2 , and MAZ , with widespread trans effects on neurogenesis-related gene programs. In excitatory neurons, we identified POU3F2 as a key cis -regulated gene with trans effects on synaptic gene expression and neuronal firing. This study identifies cis -acting genomic changes that cause cascading trans gene regulatory effects to contribute to human neural specializations, and provides a general framework for discovering genetic differences underlying human traits.
    Date: 2025-03-31
    Authors: Song JH, Carter AC, Bushinsky EM, Beck SG, Petrocelli JE, Koreman GT, Babu J, Kingsley DM, Greenberg ME, Walsh CA.
    Ref: bioRxiv
  20. Abstract LIMD1 is a tumour suppressor gene frequently lost in non-small cell lung cancer (NSCLC), but its role in cancer-immune cell interactions remains unexplored. Here, we demonstrate that LIMD1 loss results in upregulation of the key immune checkpoint protein PD-L1. Using multi-region sequencing from the TRACERx dataset, we identify that LIMD1 loss is clonal in over 80% of squamous cell carcinoma (LUSC) and 40% of lung adenocarcinoma (LUAD) cases, correlating with increased PD-L1 expression. LIMD1 deficiency results in upregulation of basal and IFNγ-induced PD-L1 expression in NSCLC cells and, consistent with its early loss during oncogenesis, in primary human small airway epithelial cells. Mechanistically, we demonstrate that LIMD1 interacts with the E3 ubiquitin ligase ARIH1 to mediate efficient PD-L1 ubiquitination and degradation, a process that is significantly impaired in LIMD1-deficient cells, resulting in increased PD-L1 stability. As a consequence, LIMD1-deficient tumour cells suppressed CD8+ T cell activation in vitro, and blockade of PD-L1 reversed this suppression. Clinically, we show that LIMD1 loss is associated with enhanced response to immune checkpoint inhibitors (ICIs) in NSCLC patient cohorts, revealing a novel cancer cell-intrinsic correlation of ICI efficacy. Our results uncover a tumour suppressor-mediated mechanism of PD-L1 expression and pave the way for stratified immunotherapy approaches in LIMD1 -/- NSCLC.
    Date: 2025-03-31
    Authors: Sharp T, Shah K, Kennedy P, Black J, Litchfield K, Thakkar K, Maria Contreras-Gerenas1, KBrooksbank K, Yuan O, Grevitt P, Dahal L, Lagos D, McGranahan N, Charrot S, Davies J.
    Ref: Research Square
  21. Genetic defects in glycine decarboxylase (GLDC) cause non-ketotic hyperglycinemia (NKH), a rare and frequently fatal neurometabolic disease, which lacks FDA-approved therapies. We characterized CRISPR Cas9-edited humanized mice expressing a prevalent clinical mutation after administration with a single intraperitoneal dose of a novel recombinant of adeno-associated viral vector 9 expressing GLDC (rAAV9-GLDC). Long term biological activity of rAAV9-GLDC was first validated by assessment of its systemic efficacy over five and ten months in mice. Access of rAAV9 to the brain was confirmed by tracking green fluorescent protein (GFP) after a single intraperitoneal dose of rAAV9-GFP. Over five months, control ‘mock’ treated GFP-mice showed reduction in astrocytes but not microglia, oligodendrocytes or neurons in the brain. 37% of these animals suffered long term neurological disease and/or death. rAAV9-GLDC boosted novel astrocyte proliferation linked to neurogenesis, in absence of inflammation to confer 100% protection against sickness and fatality due to NKH.
    Date: 2025-03-31
    Authors: Lopez-Ramirez A, Bali A, Alam MS, Padmanabhan P, Calhoun S, Bickerton C, Flores-Mireles AL, Haldar K.
    Ref: bioRxiv
  22. Microbial and viral co-evolution has created immunity mechanisms involving oligonucleotide signaling that share mechanistic features with human anti-viral systems 1 . In these pathways, including CBASS and type III CRISPR systems in bacteria and cGAS-STING in humans, oligonucleotide synthesis occurs upon detection of virus or foreign genetic material in the cell, triggering the antiviral response 2–4 . In a surprising inversion of this process, we show here that the CRISPR-related enzyme mCpol synthesizes cyclic oligonucleotides constitutively as part of an active mechanism that maintains cell health. Cell-based experiments demonstrated that the absence or loss of mCpol-produced cyclic oligonucleotides triggers cell death, preventing spread of viruses that attempt immune evasion by depleting host cyclic nucleotides. Structural and mechanistic investigation revealed mCpol to be a di-adenylate cyclase whose product, c-di-AMP, prevents toxic oligomerization of the effector protein 2TMβ. Analysis of cells by fluorescence microscopy showed that lack of mCpol allows 2TMβ-mediated cell death due to inner membrane collapse. These findings unveil a powerful new defense strategy against virus-mediated immune suppression, expanding our understanding of oligonucleotides in cell health and disease. These results raise the possibility of similar protective roles for cyclic oligonucleotides in other organisms including humans.
    Date: 2025-03-30
    Authors: Doherty EE, Adler BA, Yoon PH, Hsieh K, Loi K, Armbuster EG, Lahiri A, Bolling CS, Wilcox XE, Akkati A, Iavarone AT, Pogliano J, Doudna JA.
    Ref: bioRxiv
  23. The novel duck reovirus (NDRV) disease presents a significant threat to the poultry industry due to the absence of effective therapeutic measures. As a result, there is an urgent need to develop innovative rapid diagnostic methods for early virus detection. In this study, we developed a Rapid Visual CRISPR Assay to detect the NDRV S3 gene using novel Cas12a orthologs. Specifically, we compared the performance of two candidates, Gs12-16 and Gs12-18, in detecting the NDRV S3 gene to identify a highly sensitive and efficient CRISPR-based diagnostic method. Our results demonstrated that both Gs12-16 and Gs12-18 exhibited strong cis - and trans -cleavage activities for classical “TTTV” protospacer adjacent motif (PAM)-containing targets in vitro , although they required different reaction temperatures. Notably, Gs12-18 showed relatively higher activity for dsDNA targets compared to Gs12-16, indicating that Gs12-18 is more suitable for CRISPR-based nucleic acid detection applications. To leverage these properties, we integrated Gs12-18 with loop-mediated isothermal amplification (LAMP) technology to establish a LAMP-CRISPR/Gs12-18-mediated method for detecting the NDRV S3 gene. This approach enables highly sensitive and visually detectable on-site identification of the NDRV S3 gene, achieving a sensitivity of 38 copies per reaction. Our LAMP-CRISPR/Gs12-18-based method can be utilized for highly sensitive detection of NDRV nucleic acids.
    Date: 2025-03-30
    Authors: Wang Y, Fu L, Li S, Tao D, Gong P, Yang Y, Ruan J, Xie S, Wang C, He D.
    Ref: bioRxiv
  24. Bacteria use antiphage systems to combat phages, their ubiquitous competitors, and evolve new defenses through repeated reshuffling of basic functional units into novel reformulations. A common theme is generating a nucleotide-derived second messenger in response to phage that activates an effector protein to halt virion production. Phages respond with counter-defenses that deplete these second messengers, leading to an escalating arms race with the host. Here we discover a novel antiphage system we call Panoptes that detects phage infection by surveying the cytosol for phage proteins that antagonize the nucleotide-derived second messenger pool. Panoptes is a two-gene operon, optSE . OptS is predicted to synthesize a second messenger using a minimal CRISPR polymerase (mCpol) domain, a version of the polymerase domain found in Type III CRISPR systems (Cas10) that is distantly related to GGDEF and Thg1 tRNA repair polymerase domains. OptE is predicted to be a transmembrane effector protein that binds cyclic nucleotides. optSE potently restricted phage replication but mutant phages that had loss-of-function mutations in anti-CBASS protein 2 (Acb2) escaped defense. These findings were unexpected because Acb2 is a nucleotide “sponge” that antagonizes second messenger signaling. Using genetic and biochemical assays, we found that Acb2 bound the OptS-synthesized nucleotide, 2′,3′-cyclic adenosine monophosphate (2′,3′-c-di-AMP); however, 2′,3′-c-di-AMP was synthesized constitutively by OptS and inhibited OptE. Nucleotide depletion by Acb2 released OptE toxicity thereby initiating abortive infection to halt phage replication. These data demonstrate a sophisticated immune strategy that hosts use to guard their second messenger pool and turn immune evasion against the virus.
    Date: 2025-03-30
    Authors: Sullivan AE, Nabhani A, Schinkel K, Dinh DM, Duncan ML, Ednacot EMQ, Hoffman CR, Izrailevsky DS, Kibby EM, Nagy TA, Nguyen CM, Tak U, Burroughs AM, Aravind L, Whiteley AT, Morehouse BR.
    Ref: bioRxiv
  25. ABSTRACT Animal models with a clinically relevant phenotype remain important for robust evaluation of novel therapeutics for the fatal, X-linked genetic disorder, Duchenne Muscular Dystrophy (DMD). Demonstration of functional improvement is crucial for both patients and regulatory authorities. DMD is associated with a decline in musculoskeletal function with progressive paresis, muscle atrophy and fibrosis: phenotypic features that are also seen in the DE50-MD canine model of DMD. Here we investigate non-invasive methods to quantify changes in activity and behaviour in DE50-MD dogs, using collar-based, tri-axial accelerometers. We measured activity in affected DE50-MD male dogs (3-8 per age point) and littermate wild-type (WT) male controls (3-13 per age point) at monthly intervals from 3 to 18 months of age using Axivity-AX3 accelerometers attached ventrally on each dog’s collar. Data were recorded for 48 hours while dogs remained in their kennels with outside runs following their normal routine. Acceleration vector magnitudes were used to derive various activity indicators over a 24-hour period. Mixed model analyses were used to examine differences between affected and WT groups at different ages. DE50-MD dogs’ activity indicators were significantly higher for % time spent at rest (p<0.001) and significantly lower for all other activity indicators (all p<0.05), when compared to age-matched WT dogs. Sample size calculations reveal that these non-invasive and objective biomarkers offer significant promise for preclinical testing of therapeutics in this model of DMD. Our approach reveals opportunities for cross-model standardisation of activity monitoring methods, applicable to both research and companion animal settings. Summary statement The DE50-MD dog model of Duchenne muscular dystrophy shows significant age-associated reduction in activity quantified through non-invasive, wearable accelerometers. Activity metrics tested show promise for objective assessment of activity patterns for preclinical trials.
    Date: 2025-03-29
    Authors: Karimjee K, River RC, Olsen E, Chang Y, Wells DJ, Daley MA, Piercy RJ.
    Ref: bioRxiv
first · previous · 6 · 7 · 8 · 9 · 10 · 11 · 12 · 13 · 14 · 15 · next · last