Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.803
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 93(1): 139-161, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38598855

ABSTRACT

CRISPR-Cas (clustered regularly interspaced short palindromic repeats-CRISPR-associated nuclease) defense systems have been naturally coopted for guide RNA-directed transposition on multiple occasions. In all cases, cooption occurred with diverse elements related to the bacterial transposon Tn7. Tn7 tightly controls transposition; the transposase is activated only when special targets are recognized by dedicated target-site selection proteins. Tn7 and the Tn7-like elements that coopted CRISPR-Cas systems evolved complementary targeting pathways: one that recognizes a highly conserved site in the chromosome and a second pathway that targets mobile plasmids capable of cell-to-cell transfer. Tn7 and Tn7-like elements deliver a single integration into the site they recognize and also control the orientation of the integration event, providing future potential for use as programmable gene-integration tools. Early work has shown that guide RNA-directed transposition systems can be adapted to diverse hosts, even within microbial communities, suggesting great potential for engineering these systems as powerful gene-editing tools.


Subject(s)
CRISPR-Cas Systems , DNA Transposable Elements , RNA, Guide, CRISPR-Cas Systems , Transposases , DNA Transposable Elements/genetics , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Transposases/metabolism , Transposases/genetics , Gene Editing/methods , Bacteria/genetics , Plasmids/metabolism , Plasmids/genetics , Clustered Regularly Interspaced Short Palindromic Repeats
2.
Mol Cell ; 84(8): 1585-1600.e7, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38479385

ABSTRACT

Myriad physiological and pathogenic processes are governed by protein levels and modifications. Controlled protein activity perturbation is essential to studying protein function in cells and animals. Based on Trim-Away technology, we screened for truncation variants of E3 ubiquitinase Trim21 with elevated efficiency (ΔTrim21) and developed multiple ΔTrim21-based targeted protein-degradation systems (ΔTrim-TPD) that can be transfected into host cells. Three ΔTrim-TPD variants are developed to enable chemical and light-triggered programmable activation of TPD in cells and animals. Specifically, we used ΔTrim-TPD for (1) red-light-triggered inhibition of HSV-1 virus proliferation by degrading the packaging protein gD, (2) for chemical-triggered control of the activity of Cas9/dCas9 protein for gene editing, and (3) for blue-light-triggered degradation of two tumor-associated proteins for spatiotemporal inhibition of melanoma tumor growth in mice. Our study demonstrates that multiple ΔTrim21-based controllable TPD systems provide powerful tools for basic biology research and highlight their potential biomedical applications.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Mice , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Proteins/metabolism , Proteolysis , Mammals/metabolism
3.
Mol Cell ; 84(10): 1842-1854.e7, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759624

ABSTRACT

Genomic context critically modulates regulatory function but is difficult to manipulate systematically. The murine insulin-like growth factor 2 (Igf2)/H19 locus is a paradigmatic model of enhancer selectivity, whereby CTCF occupancy at an imprinting control region directs downstream enhancers to activate either H19 or Igf2. We used synthetic regulatory genomics to repeatedly replace the native locus with 157-kb payloads, and we systematically dissected its architecture. Enhancer deletion and ectopic delivery revealed previously uncharacterized long-range regulatory dependencies at the native locus. Exchanging the H19 enhancer cluster with the Sox2 locus control region (LCR) showed that the H19 enhancers relied on their native surroundings while the Sox2 LCR functioned autonomously. Analysis of regulatory DNA actuation across cell types revealed that these enhancer clusters typify broader classes of context sensitivity genome wide. These results show that unexpected dependencies influence even well-studied loci, and our approach permits large-scale manipulation of complete loci to investigate the relationship between regulatory architecture and function.


Subject(s)
CCCTC-Binding Factor , Enhancer Elements, Genetic , Insulin-Like Growth Factor II , RNA, Long Noncoding , SOXB1 Transcription Factors , Animals , Mice , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Locus Control Region/genetics , Genomic Imprinting , Genomics/methods
4.
Mol Cell ; 83(7): 1140-1152.e7, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36931273

ABSTRACT

Sox2 expression in mouse embryonic stem cells (mESCs) depends on a distal cluster of DNase I hypersensitive sites (DHSs), but their individual contributions and degree of interdependence remain a mystery. We analyzed the endogenous Sox2 locus using Big-IN to scarlessly integrate large DNA payloads incorporating deletions, rearrangements, and inversions affecting single or multiple DHSs, as well as surgical alterations to transcription factor (TF) recognition sequences. Multiple mESC clones were derived for each payload, sequence-verified, and analyzed for Sox2 expression. We found that two DHSs comprising a handful of key TF recognition sequences were each sufficient for long-range activation of Sox2 expression. By contrast, three nearby DHSs were entirely context dependent, showing no activity alone but dramatically augmenting the activity of the autonomous DHSs. Our results highlight the role of context in modulating genomic regulatory element function, and our synthetic regulatory genomics approach provides a roadmap for the dissection of other genomic loci.


Subject(s)
Gene Expression Regulation , Regulatory Sequences, Nucleic Acid , Animals , Mice , Enhancer Elements, Genetic , Genomics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , SOXB1 Transcription Factors/metabolism
5.
Mol Cell ; 73(3): 589-600.e4, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30639240

ABSTRACT

CRISPR-Cas12a (Cpf1) is an RNA-guided DNA-cutting nuclease that has been repurposed for genome editing. Upon target DNA binding, Cas12a cleaves both the target DNA in cis and non-target single-stranded DNAs (ssDNAs) in trans. To elucidate the molecular basis for both DNase cleavage modes, we performed structural and biochemical studies on Francisella novicida Cas12a. We show that guide RNA-target strand DNA hybridization conformationally activates Cas12a, triggering its trans-acting, non-specific, single-stranded DNase activity. In turn, cis cleavage of double-stranded DNA targets is a result of protospacer adjacent motif (PAM)-dependent DNA duplex unwinding, electrostatic stabilization of the displaced non-target DNA strand, and ordered sequential cleavage of the non-target and target DNA strands. Cas12a releases the PAM-distal DNA cleavage product and remains bound to the PAM-proximal DNA cleavage product in a catalytically competent, trans-active state. Together, these results provide a revised model for the molecular mechanisms of both the cis- and the trans-acting DNase activities of Cas12a enzymes, enabling their further exploitation as genome editing tools.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , DNA, Single-Stranded/metabolism , Francisella/enzymology , Gene Editing/methods , RNA, Guide, Kinetoplastida/metabolism , Bacterial Proteins/genetics , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/genetics , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , Enzyme Activation , Francisella/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , Structure-Activity Relationship , Substrate Specificity
6.
Trends Genet ; 39(1): 34-45, 2023 01.
Article in English | MEDLINE | ID: mdl-36055901

ABSTRACT

Chromoanagenesis is a single catastrophic event that involves, in most cases, localized chromosomal shattering and reorganization, resulting in a dramatically restructured chromosome. First discovered in cancer cells, it has since been observed in various other systems, including plants. In this review, we discuss the origin, characteristics, and potential mechanisms underlying chromoanagenesis in plants. We report that multiple processes, including mutagenesis and genetic engineering, can trigger chromoanagenesis via a variety of mechanisms such as micronucleation, breakage-fusion-bridge (BFB) cycles, or chain-like translocations. The resulting rearranged chromosomes can be preserved during subsequent plant growth, and sometimes inherited to the next generation. Because of their high tolerance to genome restructuring, plants offer a unique system for investigating the evolutionary consequences and potential practical applications of chromoanagenesis.


Subject(s)
Chromosomes , Chromothripsis , Humans , Genome , Plants/genetics
7.
Trends Genet ; 39(5): 347-357, 2023 05.
Article in English | MEDLINE | ID: mdl-36997427

ABSTRACT

Genetic drive represents a fundamental evolutionary force that can exact profound change to the genetic composition of populations by biasing allele transmission. Herein I propose that the use of synthetic homing gene drives, the human-mediated analog of endogenous genetic drives, warrants the designation of 'genetic welding' as an anthropogenic evolutionary force. Conceptually, this distinction parallels that of artificial and natural selection. Genetic welding is capable of imposing complex and rapid heritable phenotypic change on entire populations, whether motivated by biodiversity conservation or public health. Unanticipated possible long-term evolutionary outcomes, however, demand further investigation and bioethical consideration. The emerging importance of genetic welding also compels our explicit recognition of genetic drive as an addition to the other four fundamental forces of evolution.


Subject(s)
Gene Drive Technology , Genes, Synthetic , Humans , Selection, Genetic , Alleles
8.
Mol Cell Proteomics ; 23(7): 100796, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38851451

ABSTRACT

Protein O-linked mannose (O-Man) glycosylation is an evolutionary conserved posttranslational modification that fulfills important biological roles during embryonic development. Three nonredundant enzyme families, POMT1/POMT2, TMTC1-4, and TMEM260, selectively coordinate the initiation of protein O-Man glycosylation on distinct classes of transmembrane proteins, including α-dystroglycan, cadherins, and plexin receptors. However, a systematic investigation of their substrate specificities is lacking, in part due to the ubiquitous expression of O-Man glycosyltransferases in cells, which precludes analysis of pathway-specific O-Man glycosylation on a proteome-wide scale. Here, we apply a targeted workflow for membrane glycoproteomics across five human cell lines to extensively map O-Man substrates and genetically deconstruct O-Man initiation by individual and combinatorial knockout of O-Man glycosyltransferase genes. We established a human cell library for the analysis of substrate specificities of individual O-Man initiation pathways by quantitative glycoproteomics. Our results identify 180 O-Man glycoproteins, demonstrate new protein targets for the POMT1/POMT2 pathway, and show that TMTC1-4 and TMEM260 pathways widely target distinct Ig-like protein domains of plasma membrane proteins involved in cell-cell and cell-extracellular matrix interactions. The identification of O-Man on Ig-like folds adds further knowledge on the emerging concept of domain-specific O-Man glycosylation which opens for functional studies of O-Man-glycosylated adhesion molecules and receptors.


Subject(s)
Mannose , Humans , Glycosylation , Mannose/metabolism , Substrate Specificity , Glycoproteins/metabolism , Proteomics/methods , Cell Line , Glycosyltransferases/metabolism , Glycosyltransferases/genetics , Protein Processing, Post-Translational , Cell Engineering/methods
9.
Proc Natl Acad Sci U S A ; 120(51): e2221680120, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38096407

ABSTRACT

Animals integrate sensory information from the environment and display various behaviors in response to external stimuli. In Caenorhabditis elegans hermaphrodites, 33 types of sensory neurons are responsible for chemosensation, olfaction, and mechanosensation. However, the functional roles of all sensory neurons have not been systematically studied due to the lack of facile genetic accessibility. A bipartite cGAL-UAS system has been previously developed to study tissue- or cell-specific functions in C. elegans. Here, we report a toolkit of new cGAL drivers that can facilitate the analysis of a vast majority of the 60 sensory neurons in C. elegans hermaphrodites. We generated 37 sensory neuronal cGAL drivers that drive cGAL expression by cell-specific regulatory sequences or intersection of two distinct regulatory regions with overlapping expression (split cGAL). Most cGAL-drivers exhibit expression in single types of cells. We also constructed 28 UAS effectors that allow expression of proteins to perturb or interrogate sensory neurons of choice. This cGAL-UAS sensory neuron toolkit provides a genetic platform to systematically study the functions of C. elegans sensory neurons.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Sensory Receptor Cells/metabolism
10.
Proc Natl Acad Sci U S A ; 120(10): e2200626120, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36853939

ABSTRACT

Engagement of the inhibitory T cell receptor programmed cell death protein 1 (PD-1) associates with dysfunctional states of pathogen- or tumor-specific T cells. Accordingly, systemic antibody-mediated blockade of PD-1 has become a central target for immunotherapies but is also associated with severe toxicities due to loss of peripheral tolerance. Therefore, selective ablation of PD-1 expression on adoptively transferred T cells through direct genetic knockout (KO) is currently being explored as an alternative therapeutic approach. However, since PD-1 might also be required for the regulation of physiological T cell function and differentiation, the suitability of PD-1 as an engineering target is controversial. In this study, we systematically investigated the maintenance of T cell functionality after CRISPR/Cas9-mediated PD-1 KO in vivo during and after acute and chronic antigen encounter. Under all tested conditions, PD-1 ablation preserved the persistence, differentiation, and memory formation of adoptively transferred receptor transgenic T cells. Functional PD-1 KO T cells expressing chimeric antigen receptors (CARs) targeting CD19 could be robustly detected for over 390 d in a syngeneic immunocompetent mouse model, in which constant antigen exposure was provided by continuous B cell renewal, representing the longest in vivo follow-up of CAR-T cells described to date. PD-1 KO CAR-T cells showed no evidence for malignant transformation during the entire observation period. Our data demonstrate that genetic ablation of PD-1 does not impair functionality and longevity of adoptively transferred T cells per se and therefore may be pursued more generally in engineered T cell-based immunotherapy to overcome a central immunosuppressive axis.


Subject(s)
Programmed Cell Death 1 Receptor , T-Lymphocytes , Animals , Mice , Programmed Cell Death 1 Receptor/genetics , Adaptor Proteins, Signal Transducing , Animals, Genetically Modified , Antibodies, Blocking
11.
J Biol Chem ; 300(5): 107248, 2024 May.
Article in English | MEDLINE | ID: mdl-38556082

ABSTRACT

P2X receptors are a family of ligand gated ion channels found in a range of eukaryotic species including humans but are not naturally present in the yeast Saccharomyces cerevisiae. We demonstrate the first recombinant expression and functional gating of the P2X2 receptor in baker's yeast. We leverage the yeast host for facile genetic screens of mutant P2X2 by performing site saturation mutagenesis at residues of interest, including SNPs implicated in deafness and at residues involved in native binding. Deep mutational analysis and rounds of genetic engineering yield mutant P2X2 F303Y A304W, which has altered ligand selectivity toward the ATP analog AMP-PNP. The F303Y A304W variant shows over 100-fold increased intracellular calcium amplitudes with AMP-PNP compared to the WT receptor and has a much lower desensitization rate. Since AMP-PNP does not naturally activate P2X receptors, the F303Y A304W P2X2 may be a starting point for downstream applications in chemogenetic cellular control. Interestingly, the A304W mutation selectively destabilizes the desensitized state, which may provide a mechanistic basis for receptor opening with suboptimal agonists. The yeast system represents an inexpensive, scalable platform for ion channel characterization and engineering by circumventing the more expensive and time-consuming methodologies involving mammalian hosts.


Subject(s)
Receptors, Purinergic P2X2 , Saccharomyces cerevisiae , Humans , Amino Acid Substitution , Ligands , Protein Engineering/methods , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X2/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Models, Molecular , Protein Structure, Tertiary , Protein Structure, Quaternary , Structural Homology, Protein , Mutation
12.
Mol Biol Evol ; 41(1)2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38128148

ABSTRACT

The mosquito family Culicidae is divided into 2 subfamilies named the Culicinae and Anophelinae. Nix, the dominant male-determining factor, has only been found in the culicines Aedes aegypti and Aedes albopictus, 2 important arboviral vectors that belong to the subgenus Stegomyia. Here we performed sex-specific whole-genome sequencing and RNAseq of divergent mosquito species and explored additional male-inclusive datasets to investigate the distribution of Nix. Except for the Culex genus, Nix homologs were found in all species surveyed from the Culicinae subfamily, including 12 additional species from 3 highly divergent tribes comprising 4 genera, suggesting Nix originated at least 133 to 165 million years ago (MYA). Heterologous expression of 1 of 3 divergent Nix open reading frames (ORFs) in Ae. aegypti resulted in partial masculinization of genetic females as evidenced by morphology and doublesex splicing. Phylogenetic analysis suggests Nix is related to femaleless (fle), a recently described intermediate sex-determining factor found exclusively in anopheline mosquitoes. Nix from all species has a conserved structure, including 3 RNA-recognition motifs (RRMs), as does fle. However, Nix has evolved at a much faster rate than fle. The RRM3 of both Nix and fle are distantly related to the single RRM of a widely distributed and conserved splicing factor transformer-2 (tra2). The RRM3-based phylogenetic analysis suggests this domain in Nix and fle may have evolved from tra2 or a tra2-related gene in a common ancestor of mosquitoes. Our results provide insights into the evolution of sex determination in mosquitoes and will inform broad applications of mosquito-control strategies based on manipulating sex ratios toward nonbiting males.


Subject(s)
Aedes , Mosquito Vectors , Animals , Female , Male , Phylogeny , Mosquito Vectors/genetics , Aedes/genetics , Aedes/metabolism , RNA Splicing
13.
Trends Genet ; 38(8): 869-880, 2022 08.
Article in English | MEDLINE | ID: mdl-35599021

ABSTRACT

The mitochondrial genome has been difficult to manipulate because it is shielded by the organelle double membranes, preventing efficient nucleic acid entry. Moreover, mitochondrial DNA (mtDNA) recombination is not a robust system in most species. This limitation has forced investigators to rely on naturally occurring alterations to study both mitochondrial function and pathobiology. Because most pathogenic mtDNA mutations are heteroplasmic, the development of specific nucleases has allowed us to selectively eliminate mutant species. Several 'protein only' gene-editing platforms have been successfully used for this purpose. More recently, a DNA double-strand cytidine deaminase has been identified and adapted to edit mtDNA. This enzyme was also used as a component to adapt a DNA single-strand deoxyadenosine deaminase to mtDNA editing. These are major advances in our ability to precisely alter the mtDNA in animal cells.


Subject(s)
Genome, Mitochondrial , Animals , DNA, Mitochondrial/genetics , Gene Editing , Genome, Mitochondrial/genetics , Mitochondria/genetics
14.
Eur J Immunol ; 54(2): e2350778, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38105295

ABSTRACT

As the understanding of the tumor microenvironment has deepened, immunotherapy has become a promising strategy for cancer treatment. In contrast to traditional therapies, immunotherapy is more precise and induces fewer adverse effects. In this field, some bacteria have attracted increased attention because of their natural ability to preferentially colonize and proliferate inside tumor sites and exert antitumor effects. Moreover, bacterial components may activate innate and adaptive immunity to resist tumor progression. However, the application of bacteria-based cancer immunotherapy is hampered by potential infection-associated toxicity and unpredictable behavior in vivo. Owing to modern developments in genetic engineering, bacteria can be modified to weaken their toxicity and enhance their ability to eliminate tumor cells or activate the antitumor immune response. This review summarizes the roles of bacteria in the tumor microenvironment, current strategies for bacterial engineering, and the synergistic efficiency of bacteria with other immunotherapies. In addition, the prospects and challenges of the clinical translation of engineered bacteria are summarized.


Subject(s)
Neoplasms , Humans , Neoplasms/therapy , Immunotherapy , Bacteria , Tumor Microenvironment , Genetic Engineering
15.
Mol Ther ; 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38273655

ABSTRACT

The last decade has seen rapid development in the field of cellular immunotherapy, particularly in regard to chimeric antigen receptor (CAR)-modified T cells. However, challenges, such as severe treatment-related toxicities and inconsistent quality of autologous products, have hindered the broader use of CAR-T cell therapy, highlighting the need to explore alternative immune cells for cancer targeting. In this regard, natural killer (NK) cells have been extensively studied in cellular immunotherapy and were found to exert cytotoxic effects without being restricted by human leukocyte antigen and have a lower risk of causing graft-versus-host disease; making them favorable for the development of readily available "off-the-shelf" products. Clinical trials utilizing unedited NK cells or reprogrammed NK cells have shown early signs of their effectiveness against tumors. However, limitations, including limited in vivo persistence and expansion potential, remained. To enhance the antitumor function of NK cells, advanced gene-editing technologies and combination approaches have been explored. In this review, we summarize current clinical trials of antitumor NK cell therapy, provide an overview of innovative strategies for reprogramming NK cells, which include improvements in persistence, cytotoxicity, trafficking and the ability to counteract the immunosuppressive tumor microenvironment, and also discuss some potential combination therapies.

16.
Mol Ther ; 32(2): 440-456, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38213031

ABSTRACT

Here we introduce a first-in-class microRNA-sensitive oncolytic Zika virus (ZIKV) for virotherapy application against central nervous system (CNS) tumors. The described methodology produced two synthetic modified ZIKV strains that are safe in normal cells, including neural stem cells, while preserving brain tropism and oncolytic effects in tumor cells. The microRNA-sensitive ZIKV introduces genetic modifications in two different virus sites: first, in the established 3'UTR region, and secondly, in the ZIKV protein coding sequence, demonstrating for the first time that the miRNA inhibition systems can be functional outside the UTR RNA sites. The total tumor remission in mice bearing human CNS tumors, including metastatic tumor growth, after intraventricular and systemic modified ZIKV administration, confirms the promise of this virotherapy as a novel agent against brain tumors-highly deadly diseases in urgent need of effective advanced therapies.


Subject(s)
Central Nervous System Neoplasms , MicroRNAs , Oncolytic Virotherapy , Oncolytic Viruses , Zika Virus Infection , Zika Virus , Humans , Mice , Animals , Oncolytic Viruses/genetics , Zika Virus/genetics , MicroRNAs/genetics , Zika Virus Infection/therapy , Oncolytic Virotherapy/methods
17.
Proc Natl Acad Sci U S A ; 119(45): e2105458119, 2022 Nov 08.
Article in English | MEDLINE | ID: mdl-36322728

ABSTRACT

Despite dramatic advances in genomics, connecting genotypes to phenotypes is still challenging. Sexual genetics combined with linkage analysis is a powerful solution to this problem but generally unavailable in bacteria. We build upon a strong negative selection system to invent mass allelic exchange (MAE), which enables hybridization of arbitrary (including pathogenic) strains of Escherichia coli. MAE reimplements the natural phenomenon of random cross-overs, enabling classical linkage analysis. We demonstrate the utility of MAE with virulence-related gain-of-function screens, discovering that transfer of a single operon from a uropathogenic strain is sufficient for enabling a commensal E. coli to form large intracellular bacterial collections within bladder epithelial cells. MAE thus enables assaying natural allelic variation in E. coli (and potentially other bacteria), complementing existing loss-of-function genomic techniques.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Urinary Tract Infections , Uropathogenic Escherichia coli , Humans , Uropathogenic Escherichia coli/genetics , Urinary Tract Infections/microbiology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Virulence/genetics , Virulence Factors/genetics
18.
Plant Mol Biol ; 114(2): 21, 2024 Feb 18.
Article in English | MEDLINE | ID: mdl-38368585

ABSTRACT

Arbuscular mycorrhizal symbiosis (AM) is a beneficial trait originating with the first land plants, which has subsequently been lost by species scattered throughout the radiation of plant diversity to the present day, including the model Arabidopsis thaliana. To explore if elements of this apparently beneficial trait are still present and could be reactivated we generated Arabidopsis plants expressing a constitutively active form of Interacting Protein of DMI3, a key transcription factor that enables AM within the Common Symbiosis Pathway, which was lost from Arabidopsis along with the AM host trait. We characterize the transcriptomic effect of expressing IPD3 in Arabidopsis with and without exposure to the AM fungus (AMF) Rhizophagus irregularis, and compare these results to the AM model Lotus japonicus and its ipd3 knockout mutant cyclops-4. Despite its long history as a non-AM species, restoring IPD3 in the form of its constitutively active DNA-binding domain to Arabidopsis altered expression of specific gene networks. Surprisingly, the effect of expressing IPD3 in Arabidopsis and knocking it out in Lotus was strongest in plants not exposed to AMF, which is revealed to be due to changes in IPD3 genotype causing a transcriptional state, which partially mimics AMF exposure in non-inoculated plants. Our results indicate that molecular connections to symbiosis machinery remain in place in this nonAM species, with implications for both basic science and the prospect of engineering this trait for agriculture.


Subject(s)
Arabidopsis , Lotus , Arabidopsis/genetics , Symbiosis/genetics , Genotype , Agriculture , Biological Evolution , Lotus/genetics
19.
Kidney Int ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38901605

ABSTRACT

Vascularization plays a critical role in organ maturation and cell-type development. Drug discovery, organ mimicry, and ultimately transplantation hinge on achieving robust vascularization of in vitro engineered organs. Here, focusing on human kidney organoids, we overcame this hurdle by combining a human induced pluripotent stem cell (iPSC) line containing an inducible ETS translocation variant 2 (ETV2) (a transcription factor playing a role in endothelial cell development) that directs endothelial differentiation in vitro, with a non-transgenic iPSC line in suspension organoid culture. The resulting human kidney organoids show extensive endothelialization with a cellular identity most closely related to human kidney endothelia. Endothelialized kidney organoids also show increased maturation of nephron structures, an associated fenestrated endothelium with de novo formation of glomerular and venous subtypes, and the emergence of drug-responsive renin expressing cells. The creation of an engineered vascular niche capable of improving kidney organoid maturation and cell type complexity is a significant step forward in the path to clinical translation. Thus, incorporation of an engineered endothelial niche into a previously published kidney organoid protocol allowed the orthogonal differentiation of endothelial and parenchymal cell types, demonstrating the potential for applicability to other basic and translational organoid studies.

20.
Trends Genet ; 37(8): 685-687, 2021 08.
Article in English | MEDLINE | ID: mdl-33975753

ABSTRACT

The characteristics of RNA editing, including the lower risk compared with genome editing, may loosen the ethical barriers that are currently imposed on genetic engineering, thus opening new possibilities for research, therapy, and human enhancement. We should start considering the future ethical and social implications of this new and promising technology.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Engineering/ethics , RNA Editing/ethics , Gene Editing/ethics , Genome, Human/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL