45 research outputs found

    Combined congenic mapping and nuclease-based gene targeting for studying allele-specific effects of Tnfrsf9 within the Idd9.3 autoimmune diabetes locus.

    Get PDF
    Rodent complex trait genetic studies involving a cross between two inbred strains are usually followed by congenic mapping to refine the loci responsible for the phenotype. However, progressing from a chromosomal region to the actual causal gene remains challenging because multiple polymorphic genes are often closely linked. The goal of this study was to develop a strategy that allows candidate gene testing by allele-specific expression without prior knowledge of the credible causal variant. Tnfrsf9 (encoding CD137) is a candidate gene for the Idd9.3 type 1 diabetes (T1D) susceptibility locus in the nonobese diabetic (NOD) mouse model. A C57BL/10Sn (B10)-derived diabetes resistance Idd9.3 congenic region has been shown to enhance accumulation of CD137+ regulatory T cells and serum soluble CD137 in NOD mice. By combining the power of congenic mapping and nuclease-based gene targeting, we established a system where a pair of F1 hybrids expressed either the B10 or NOD Tnfrsf9 allele mimicking coisogenic strains. Using this approach, we demonstrated that the allelic difference in B10 and NOD Tnfrsf9 alone was sufficient to cause differential accumulation of CD137+ regulatory T cells and serum soluble CD137 levels. This strategy can be broadly applied to other rodent genetic mapping studies

    Virus-Free CRISPR CAR T cells induce solid tumor regression

    Get PDF
    Chimeric antigen receptor (CAR) T cell therapy has shown promising efficacy in treating hematologic malignancies and has led to the FDA-approval of three CAR T cell products. However, there has been little success in treating solid tumors, as clinical trials to date have yielded little to no responses and no improvement in survival. Current methods of CAR T cell production typically involve the use of viral vectors which can give rise to complications such as insertional mutagenesis, leading to gene silencing or oncogene activation. In addition, GMP-grade viral vector manufacturing can be expensive with lengthy wait times for new batches. Here we have developed a virus-free strategy in primary T cells that has eliminated the use of viral vectors through the use of CRISPR-Cas9 to precisely edit the chimeric antigen receptor into the TRAC gene1. Our method of virus free production begins through the generation of a double stranded DNA (dsDNA) template produced by polymerase chain reaction (PCR). This template is then combined with a SpCas9-single guide RNA to create a ribonucleoprotein (RNP) complex. Isolated human primary T cells from adult healthy donors are then nucleofected with the RNP and dsDNA template on day 2 of ex vivo expansion. Flow cytometry is then utilized to immunophenotype the cell product and analyze the percent of efficiency of CAR gene transfer. Within the cell product, the editing efficiencies are \u3e95% TCR knockout and 35% CAR+. Transcriptional profiling indicates that the virus-free CART cells have a favorable memory-like phenotype. In addition to our in vitro work, in vivo mice studies with anti-GD2 CART products demonstrate regression of GD2+ solid tumors upon virus-free CART treatment, showing similar potency and survival to viral-produced CAR T cells. The production of virus-free CAR T cells has high potential to enable the rapid and flexible manufacturing of highly defined and highly potent CAR T cell products for the treatment of solid tumors. 1 Mueller, K. et al. CRISPR-mediated insertion of a chimeric antigen receptor produces nonviral T cell products capable of inducing solid tumor regression. bioRxiv preprint doi: https://doi.org/10.1101/2021.08.06.455489 (2021)

    Virus-Free CRISPR CAR T cells induce solid tumor regression

    Get PDF
    Chimeric antigen receptor (CAR) T cell therapy has shown promising efficacy in treating hematologic malignancies and has led to the FDA-approval of three CAR T cell products. However, there has been little success in treating solid tumors, as clinical trials to date have yielded little to no responses and no improvement in survival. Current methods of CAR T cell production typically involve the use of viral vectors which can give rise to complications such as insertional mutagenesis, leading to gene silencing or oncogene activation. In addition, GMP-grade viral vector manufacturing can be expensive with lengthy wait times for new batches. Here we have developed a virus-free strategy in primary T cells that has eliminated the use of viral vectors through the use of CRISPR-Cas9 to precisely edit the chimeric antigen receptor into the TRAC gene1. Our method of virus free production begins through the generation of a double stranded DNA (dsDNA) template produced by polymerase chain reaction (PCR). This template is then combined with a SpCas9-single guide RNA to create a ribonucleoprotein (RNP) complex. Isolated human primary T cells from adult healthy donors are then nucleofected with the RNP and dsDNA template on day 2 of ex vivo expansion. Flow cytometry is then utilized to immunophenotype the cell product and analyze the percent of efficiency of CAR gene transfer. Within the cell product, the editing efficiencies are \u3e95% TCR knockout and 35% CAR+. Transcriptional profiling indicates that the virus-free CART cells have a favorable memory-like phenotype. In addition to our in vitro work, in vivo mice studies with anti-GD2 CART products demonstrate regression of GD2+ solid tumors upon virus-free CART treatment, showing similar potency and survival to viral-produced CAR T cells. The production of virus-free CAR T cells has high potential to enable the rapid and flexible manufacturing of highly defined and highly potent CAR T cell products for the treatment of solid tumors. 1 Mueller, K. et al. CRISPR-mediated insertion of a chimeric antigen receptor produces nonviral T cell products capable of inducing solid tumor regression. bioRxiv preprint doi: https://doi.org/10.1101/2021.08.06.455489 (2021)

    Comparative Genomic Characterization of Francisella tularensis Strains Belonging to Low and High Virulence Subspecies

    Get PDF
    Tularemia is a geographically widespread, severely debilitating, and occasionally lethal disease in humans. It is caused by infection by a gram-negative bacterium, Francisella tularensis. In order to better understand its potency as an etiological agent as well as its potential as a biological weapon, we have completed draft assemblies and report the first complete genomic characterization of five strains belonging to the following different Francisella subspecies (subsp.): the F. tularensis subsp. tularensis FSC033, F. tularensis subsp. holarctica FSC257 and FSC022, and F. tularensis subsp. novicida GA99-3548 and GA99-3549 strains. Here, we report the sequencing of these strains and comparative genomic analysis with recently available public Francisella sequences, including the rare F. tularensis subsp. mediasiatica FSC147 strain isolate from the Central Asian Region. We report evidence for the occurrence of large-scale rearrangement events in strains of the holarctica subspecies, supporting previous proposals that further phylogenetic subdivisions of the Type B clade are likely. We also find a significant enrichment of disrupted or absent ORFs proximal to predicted breakpoints in the FSC022 strain, including a genetic component of the Type I restriction-modification defense system. Many of the pseudogenes identified are also disrupted in the closely related rarely human pathogenic F. tularensis subsp. mediasiatica FSC147 strain, including modulator of drug activity B (mdaB) (FTT0961), which encodes a known NADPH quinone reductase involved in oxidative stress resistance. We have also identified genes exhibiting sequence similarity to effectors of the Type III (T3SS) and components of the Type IV secretion systems (T4SS). One of the genes, msrA2 (FTT1797c), is disrupted in F. tularensis subsp. mediasiatica and has recently been shown to mediate bacterial pathogen survival in host organisms. Our findings suggest that in addition to the duplication of the Francisella Pathogenicity Island, and acquisition of individual loci, adaptation by gene loss in the more recently emerged tularensis, holarctica, and mediasiatica subspecies occurred and was distinct from evolutionary events that differentiated these subspecies, and the novicida subspecies, from a common ancestor. Our findings are applicable to future studies focused on variations in Francisella subspecies pathogenesis, and of broader interest to studies of genomic pathoadaptation in bacteria

    Diving into the vertical dimension of elasmobranch movement ecology

    Get PDF
    Knowledge of the three-dimensional movement patterns of elasmobranchs is vital to understand their ecological roles and exposure to anthropogenic pressures. To date, comparative studies among species at global scales have mostly focused on horizontal movements. Our study addresses the knowledge gap of vertical movements by compiling the first global synthesis of vertical habitat use by elasmobranchs from data obtained by deployment of 989 biotelemetry tags on 38 elasmobranch species. Elasmobranchs displayed high intra- and interspecific variability in vertical movement patterns. Substantial vertical overlap was observed for many epipelagic elasmobranchs, indicating an increased likelihood to display spatial overlap, biologically interact, and share similar risk to anthropogenic threats that vary on a vertical gradient. We highlight the critical next steps toward incorporating vertical movement into global management and monitoring strategies for elasmobranchs, emphasizing the need to address geographic and taxonomic biases in deployments and to concurrently consider both horizontal and vertical movements

    Diving into the vertical dimension of elasmobranch movement ecology

    Get PDF
    Knowledge of the three-dimensional movement patterns of elasmobranchs is vital to understand their ecological roles and exposure to anthropogenic pressures. To date, comparative studies among species at global scales have mostly focused on horizontal movements. Our study addresses the knowledge gap of vertical movements by compiling the first global synthesis of vertical habitat use by elasmobranchs from data obtained by deployment of 989 biotelemetry tags on 38 elasmobranch species. Elasmobranchs displayed high intra- and interspecific variability in vertical movement patterns. Substantial vertical overlap was observed for many epipelagic elasmobranchs, indicating an increased likelihood to display spatial overlap, biologically interact, and share similar risk to anthropogenic threats that vary on a vertical gradient. We highlight the critical next steps toward incorporating vertical movement into global management and monitoring strategies for elasmobranchs, emphasizing the need to address geographic and taxonomic biases in deployments and to concurrently consider both horizontal and vertical movements

    Diving into the vertical dimension of elasmobranch movement ecology

    Get PDF
    Knowledge of the three-dimensional movement patterns of elasmobranchs is vital to understand their ecological roles and exposure to anthropogenic pressures. To date, comparative studies among species at global scales have mostly focused on horizontal movements. Our study addresses the knowledge gap of vertical movements by compiling the first global synthesis of vertical habitat use by elasmobranchs from data obtained by deployment of 989 biotelemetry tags on 38 elasmobranch species. Elasmobranchs displayed high intra- and interspecific variability in vertical movement patterns. Substantial vertical overlap was observed for many epipelagic elasmobranchs, indicating an increased likelihood to display spatial overlap, biologically interact, and share similar risk to anthropogenic threats that vary on a vertical gradient. We highlight the critical next steps toward incorporating vertical movement into global management and monitoring strategies for elasmobranchs, emphasizing the need to address geographic and taxonomic biases in deployments and to concurrently consider both horizontal and vertical movements

    Effects of Anacetrapib in Patients with Atherosclerotic Vascular Disease

    Get PDF
    BACKGROUND: Patients with atherosclerotic vascular disease remain at high risk for cardiovascular events despite effective statin-based treatment of low-density lipoprotein (LDL) cholesterol levels. The inhibition of cholesteryl ester transfer protein (CETP) by anacetrapib reduces LDL cholesterol levels and increases high-density lipoprotein (HDL) cholesterol levels. However, trials of other CETP inhibitors have shown neutral or adverse effects on cardiovascular outcomes. METHODS: We conducted a randomized, double-blind, placebo-controlled trial involving 30,449 adults with atherosclerotic vascular disease who were receiving intensive atorvastatin therapy and who had a mean LDL cholesterol level of 61 mg per deciliter (1.58 mmol per liter), a mean non-HDL cholesterol level of 92 mg per deciliter (2.38 mmol per liter), and a mean HDL cholesterol level of 40 mg per deciliter (1.03 mmol per liter). The patients were assigned to receive either 100 mg of anacetrapib once daily (15,225 patients) or matching placebo (15,224 patients). The primary outcome was the first major coronary event, a composite of coronary death, myocardial infarction, or coronary revascularization. RESULTS: During the median follow-up period of 4.1 years, the primary outcome occurred in significantly fewer patients in the anacetrapib group than in the placebo group (1640 of 15,225 patients [10.8%] vs. 1803 of 15,224 patients [11.8%]; rate ratio, 0.91; 95% confidence interval, 0.85 to 0.97; P=0.004). The relative difference in risk was similar across multiple prespecified subgroups. At the trial midpoint, the mean level of HDL cholesterol was higher by 43 mg per deciliter (1.12 mmol per liter) in the anacetrapib group than in the placebo group (a relative difference of 104%), and the mean level of non-HDL cholesterol was lower by 17 mg per deciliter (0.44 mmol per liter), a relative difference of -18%. There were no significant between-group differences in the risk of death, cancer, or other serious adverse events. CONCLUSIONS: Among patients with atherosclerotic vascular disease who were receiving intensive statin therapy, the use of anacetrapib resulted in a lower incidence of major coronary events than the use of placebo. (Funded by Merck and others; Current Controlled Trials number, ISRCTN48678192 ; ClinicalTrials.gov number, NCT01252953 ; and EudraCT number, 2010-023467-18 .)
    corecore