Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Elife ; 122024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847802

ABSTRACT

CRISPR prime editing (PE) requires a Cas9 nickase-reverse transcriptase fusion protein (known as PE2) and a prime editing guide RNA (pegRNA), an extended version of a standard guide RNA (gRNA) that both specifies the intended target genomic sequence and encodes the desired genetic edit. Here, we show that sequence complementarity between the 5' and the 3' regions of a pegRNA can negatively impact its ability to complex with Cas9, thereby potentially reducing PE efficiency. We demonstrate this limitation can be overcome by a simple pegRNA refolding procedure, which improved ribonucleoprotein-mediated PE efficiencies in zebrafish embryos by up to nearly 25-fold. Further gains in PE efficiencies of as much as sixfold could also be achieved by introducing point mutations designed to disrupt internal interactions within the pegRNA. Our work defines simple strategies that can be implemented to improve the efficiency of PE.


Subject(s)
CRISPR-Cas Systems , Gene Editing , RNA, Guide, CRISPR-Cas Systems , Zebrafish , Zebrafish/genetics , Animals , Gene Editing/methods , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , Embryo, Nonmammalian/metabolism , RNA Folding
2.
bioRxiv ; 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37645936

ABSTRACT

CRISPR prime editing (PE) requires a Cas9 nickase-reverse transcriptase fusion protein (known as PE2) and a prime editing guide RNA (pegRNA), an extended version of a standard guide RNA (gRNA) that both specifies the intended target genomic sequence and encodes the desired genetic edit. Here we show that sequence complementarity between the 5' and the 3' regions of a pegRNA can negatively impact its ability to complex with Cas9, thereby potentially reducing PE efficiency. We demonstrate this limitation can be overcome by a simple pegRNA refolding procedure, which improved ribonucleoprotein-mediated PE efficiencies in zebrafish embryos by up to nearly 25-fold. Further gains in PE efficiencies of as much as 6-fold could also be achieved by introducing point mutations designed to disrupt internal interactions within the pegRNA. Our work defines simple strategies that can be implemented to improve the efficiency of PE.

3.
Sci Adv ; 8(47): eabm7069, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36417527

ABSTRACT

Little is understood about the embryonic development of sociality. We screened 1120 known drugs and found that embryonic inhibition of topoisomerase IIα (Top2a) resulted in lasting social deficits in zebrafish. In mice, prenatal Top2 inhibition caused defects in social interaction and communication, which are behaviors that relate to core symptoms of autism. Mutation of Top2a in zebrafish caused down-regulation of a set of genes highly enriched for genes associated with autism in humans. Both the Top2a-regulated and autism-associated gene sets have binding sites for polycomb repressive complex 2 (PRC2), a regulatory complex responsible for H3K27 trimethylation (H3K27me3). Moreover, both gene sets are highly enriched for H3K27me3. Inhibition of the PRC2 component Ezh2 rescued social deficits caused by Top2 inhibition. Therefore, Top2a is a key component of an evolutionarily conserved pathway that promotes the development of social behavior through PRC2 and H3K27me3.

4.
Nat Biotechnol ; 40(2): 189-193, 2022 02.
Article in English | MEDLINE | ID: mdl-33927418

ABSTRACT

Prime editors have been delivered using DNA or RNA vectors. Here we demonstrate prime editing with purified ribonucleoprotein complexes. We introduced somatic mutations in zebrafish embryos with frequencies as high as 30% and demonstrate germline transmission. We also observed unintended insertions, deletions and prime editing guide RNA (pegRNA) scaffold incorporations. In HEK293T and primary human T cells, prime editing with purified ribonucleoprotein complexes introduced desired edits with frequencies of up to 21 and 7.5%, respectively.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Zebrafish , Animals , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing , HEK293 Cells , Humans , RNA, Guide, Kinetoplastida/genetics , Ribonucleoproteins/genetics , Zebrafish/genetics
6.
Biomolecules ; 11(11)2021 11 10.
Article in English | MEDLINE | ID: mdl-34827664

ABSTRACT

A better understanding of the metabolic constraints of a tumor may lead to more effective anticancer treatments. Evidence has emerged in recent years shedding light on a crucial aspartate dependency of many tumor types. As a precursor for nucleotide synthesis, aspartate is indispensable for cell proliferation. Moreover, the malate-aspartate shuttle plays a key role in redox balance, and a deficit in aspartate can lead to oxidative stress. It is now recognized that aspartate biosynthesis is largely governed by mitochondrial metabolism, including respiration and glutaminolysis in cancer cells. Therefore, under conditions that suppress mitochondrial metabolism, including mutations, hypoxia, or chemical inhibitors, aspartate can become a limiting factor for tumor growth and cancer cell survival. Notably, aspartate availability has been associated with sensitivity or resistance to various therapeutics that are presently in the clinic or in clinical trials, arguing for a critical need for more effective aspartate-targeting approaches. In this review, we present current knowledge of the metabolic roles of aspartate in cancer cells and describe how cancer cells maintain aspartate levels under different metabolic states. We also highlight several promising aspartate level-modulating agents that are currently under investigation.


Subject(s)
Aspartic Acid , Mitochondria , Cell Line, Tumor , Cell Proliferation , Cell Respiration , Glutamine , Humans
7.
Science ; 373(6559): 1146-1151, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34413171

ABSTRACT

CRISPR-Cas9 can be scaled up for large-scale screens in cultured cells, but CRISPR screens in animals have been challenging because generating, validating, and keeping track of large numbers of mutant animals is prohibitive. Here, we introduce Multiplexed Intermixed CRISPR Droplets (MIC-Drop), a platform combining droplet microfluidics, single-needle en masse CRISPR ribonucleoprotein injections, and DNA barcoding to enable large-scale functional genetic screens in zebrafish. The platform can efficiently identify genes responsible for morphological or behavioral phenotypes. In one application, we showed that MIC-Drop could identify small-molecule targets. Furthermore, in a MIC-Drop screen of 188 poorly characterized genes, we discovered several genes important for cardiac development and function. With the potential to scale to thousands of genes, MIC-Drop enables genome-scale reverse genetic screens in model organisms.


Subject(s)
CRISPR-Cas Systems , Genetic Testing , Microfluidic Analytical Techniques , Zebrafish/genetics , Animals , Cardiovascular System/growth & development , Cell Culture Techniques , High-Throughput Nucleotide Sequencing , Zebrafish/growth & development
8.
J Exp Clin Cancer Res ; 40(1): 251, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34376225

ABSTRACT

BACKGROUND: Pancreatic cancer is a complex disease with a desmoplastic stroma, extreme hypoxia, and inherent resistance to therapy. Understanding the signaling and adaptive response of such an aggressive cancer is key to making advances in therapeutic efficacy. Redox factor-1 (Ref-1), a redox signaling protein, regulates the conversion of several transcription factors (TFs), including HIF-1α, STAT3 and NFκB from an oxidized to reduced state leading to enhancement of their DNA binding. In our previously published work, knockdown of Ref-1 under normoxia resulted in altered gene expression patterns on pathways including EIF2, protein kinase A, and mTOR. In this study, single cell RNA sequencing (scRNA-seq) and proteomics were used to explore the effects of Ref-1 on metabolic pathways under hypoxia. METHODS: scRNA-seq comparing pancreatic cancer cells expressing less than 20% of the Ref-1 protein was analyzed using left truncated mixture Gaussian model and validated using proteomics and qRT-PCR. The identified Ref-1's role in mitochondrial function was confirmed using mitochondrial function assays, qRT-PCR, western blotting and NADP assay. Further, the effect of Ref-1 redox function inhibition against pancreatic cancer metabolism was assayed using 3D co-culture in vitro and xenograft studies in vivo. RESULTS: Distinct transcriptional variation in central metabolism, cell cycle, apoptosis, immune response, and genes downstream of a series of signaling pathways and transcriptional regulatory factors were identified in Ref-1 knockdown vs Scrambled control from the scRNA-seq data. Mitochondrial DEG subsets downregulated with Ref-1 knockdown were significantly reduced following Ref-1 redox inhibition and more dramatically in combination with Devimistat in vitro. Mitochondrial function assays demonstrated that Ref-1 knockdown and Ref-1 redox signaling inhibition decreased utilization of TCA cycle substrates and slowed the growth of pancreatic cancer co-culture spheroids. In Ref-1 knockdown cells, a higher flux rate of NADP + consuming reactions was observed suggesting the less availability of NADP + and a higher level of oxidative stress in these cells. In vivo xenograft studies demonstrated that tumor reduction was potent with Ref-1 redox inhibitor similar to Devimistat. CONCLUSION: Ref-1 redox signaling inhibition conclusively alters cancer cell metabolism by causing TCA cycle dysfunction while also reducing the pancreatic tumor growth in vitro as well as in vivo.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Pancreatic Neoplasms/genetics , Animals , Humans , Mice , Pancreatic Neoplasms/pathology , Transfection
9.
Cancer Res ; 80(17): 3492-3506, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32651261

ABSTRACT

Under conditions of inherent or induced mitochondrial dysfunction, cancer cells manifest overlapping metabolic phenotypes, suggesting that they may be targeted via a common approach. Here, we use multiple oxidative phosphorylation (OXPHOS)-competent and incompetent cancer cell pairs to demonstrate that treatment with α-ketoglutarate (aKG) esters elicits rapid death of OXPHOS-deficient cancer cells by elevating intracellular aKG concentrations, thereby sequestering nitrogen from aspartate through glutamic-oxaloacetic transaminase 1 (GOT1). Exhaustion of aspartate in these cells resulted in immediate depletion of adenylates, which plays a central role in mediating mTOR inactivation and inhibition of glycolysis. aKG esters also conferred cytotoxicity in a variety of cancer types if their cell respiration was obstructed by hypoxia or by chemical inhibition of the electron transport chain (ETC), both of which are known to increase aspartate and GOT1 dependencies. Furthermore, preclinical mouse studies suggested that cell-permeable aKG displays a good biosafety profile, eliminates aspartate only in OXPHOS-incompetent tumors, and prevents their growth and metastasis. This study reveals a novel cytotoxic mechanism for the metabolite aKG and identifies cell-permeable aKG, either by itself or in combination with ETC inhibitors, as a potential anticancer approach. SIGNIFICANCE: These findings demonstrate that OXPHOS deficiency caused by either hypoxia or mutations, which can significantly increase cancer virulence, renders tumors sensitive to aKG esters by targeting their dependence upon GOT1 for aspartate synthesis. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/17/3492/F1.large.jpg.


Subject(s)
Ketoglutaric Acids/pharmacology , Mitochondrial Diseases/metabolism , Neoplasms/metabolism , Nitrogen/metabolism , Oxidative Phosphorylation/drug effects , Animals , Cell Line, Tumor , Humans , Mice, Nude , Xenograft Model Antitumor Assays
10.
FASEB J ; 34(1): 1546-1557, 2020 01.
Article in English | MEDLINE | ID: mdl-31914600

ABSTRACT

G protein-coupled receptors (GPCRs) comprise the largest group of membrane receptors in eukaryotic genomes and collectively they regulate nearly all cellular processes. Despite the widely recognized importance of this class of proteins, many GPCRs remain understudied. G protein-coupled receptor 27 (Gpr27) is an orphan GPCR that displays high conservation during vertebrate evolution. Although, GPR27 is known to be expressed in tissues that regulate metabolism including the pancreas, skeletal muscle, and adipose tissue, its functions are poorly characterized. Therefore, to investigate the potential roles of Gpr27 in energy metabolism, we generated a whole body gpr27 knockout zebrafish line. Loss of gpr27 potentiated the elevation in glucose levels induced by pharmacological or nutritional perturbations. We next leveraged a mass spectrometry metabolite profiling platform to identify other potential metabolic functions of Gpr27. Notably, genetic deletion of gpr27 elevated medium-chain acylcarnitines, in particular C6-hexanoylcarnitine, C8-octanoylcarnitine, C9-nonanoylcarnitine, and C10-decanoylcarnitine, lipid species known to be associated with insulin resistance in humans. Concordantly, gpr27 deletion in zebrafish abrogated insulin-dependent Akt phosphorylation and glucose utilization. Finally, loss of gpr27 increased the expression of key enzymes in carnitine shuttle complex, in particular the homolog to the brain-specific isoform of CPT1C which functions as a hypothalamic energy senor. In summary, our findings shed light on the biochemical functions of Gpr27 by illuminating its role in lipid metabolism, insulin signaling, and glucose homeostasis.


Subject(s)
Carnitine/analogs & derivatives , Glucose/metabolism , Homeostasis/genetics , Insulin Resistance/genetics , Receptors, G-Protein-Coupled/genetics , Zebrafish/genetics , Animals , Carnitine/genetics , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Gene Deletion , Glucose/genetics , Insulin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Zebrafish/metabolism
11.
Nat Chem Biol ; 14(9): 844-852, 2018 09.
Article in English | MEDLINE | ID: mdl-29988067

ABSTRACT

Primordial germ cells (PGCs) form during early embryogenesis with a supply of maternal mRNAs that contain shorter poly(A) tails. How translation of maternal mRNAs is regulated during PGC development remains elusive. Here we describe a small-molecule screen with zebrafish embryos that identified primordazine, a compound that selectively ablates PGCs. Primordazine's effect on PGCs arises from translation repression through primordazine-response elements in the 3' UTRs. Systematic dissection of primordazine's mechanism of action revealed that translation of mRNAs during early embryogenesis occurs by two distinct pathways, depending on the length of their poly(A) tails. In addition to poly(A)-tail-dependent translation (PAT), early embryos perform poly(A)-tail-independent noncanonical translation (PAINT) via deadenylated 3' UTRs. Primordazine inhibits PAINT without inhibiting PAT, an effect that was also observed in quiescent, but not proliferating, mammalian cells. These studies reveal that PAINT is an alternative form of translation in the early embryo and is indispensable for PGC maintenance.


Subject(s)
3' Untranslated Regions/genetics , Germ Cells/metabolism , Peptide Chain Initiation, Translational/genetics , Animals , Cell Line, Tumor , Hydrazines/pharmacology , Mice , Peptide Chain Initiation, Translational/drug effects , Zebrafish
12.
ACS Chem Biol ; 13(8): 2143-2152, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29897731

ABSTRACT

Hereditary hemochromatosis (HH) is one of the most common genetic disorders in Caucasian populations, with no viable therapeutic options except phlebotomy. We describe a zebrafish model of human HH (HH) created by targeted mutagenesis of the gene encoding transferrin receptor 2 ( tfr2). TFR2 mutations in humans lead to HH Type 3, a rare but severe form of the disease. The tfr2 mutant model in zebrafish recapitulates the defining features of HH3: iron overload and suppression of hepcidin, the iron regulatory hormone. Using in vivo chemical screens in zebrafish embryos, we identify a new small molecule inducer of hepcidin: SC-514, a specific chemical inhibitor of NFkB signaling. Using independent small molecule inhibitors of the NFkB pathway, we demonstrate that inhibition of NFkB signaling causes induction of hepcidin transcription and reduction of iron overload in the HH3 model. This first successful chemical intervention for hereditary hemochromatosis may also have relevance in treatment of other very prevalent iron regulatory iron overload disorders such as thalassemia.


Subject(s)
Hemochromatosis/drug therapy , NF-kappa B/antagonists & inhibitors , Receptors, Transferrin/deficiency , Thiophenes/therapeutic use , Animals , Disease Models, Animal , Gene Knockdown Techniques , Hemochromatosis/metabolism , Hepcidins/genetics , Hepcidins/metabolism , Iron/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , Up-Regulation/drug effects , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Pharmacol Biochem Behav ; 157: 1-8, 2017 06.
Article in English | MEDLINE | ID: mdl-28408289

ABSTRACT

Changes in the expression of the dopamine transporter (DAT), or the sensitivity of dopamine receptors, are associated with aging and substance abuse and may underlie some of the symptoms common to both conditions. In this study, we explored the role of the dopaminergic system in the anxiogenic effects of aging and acute cocaine exposure by comparing the behavioral phenotypes of wild type (WT) and DAT knockout zebrafish (DAT-KO) of different ages. To determine the involvement of specific dopamine receptors in anxiety states, antagonists to D1 (SCH23390) and D2/D3 (sulpiride) were employed. We established that DAT-KO results in a chronic anxiety-like state, seen as an increase in bottom-dwelling and thigmotaxis. Similar effects were produced by aging and acute cocaine administration, both leading to reduction in DAT mRNA abundance (qPCR). Inhibition of D1 activity counteracted the anxiety-like effects associated with DAT deficit, independent of its origin. Inhibition of D2/D3 receptors reduced anxiety in young DAT-KO, and enhanced the anxiogenic effects of cocaine in WT, but did not affect aged WT or DAT-KO fish. These findings provide new evidence that the dopaminergic system plays a critical role in anxiety-like states, and suggest that adult zebrafish provide a sensitive diurnal vertebrate model for elucidating the molecular mechanisms of anxiety and a platform for anxiolytic drug screens.


Subject(s)
Aging/metabolism , Anxiety/metabolism , Dopamine Plasma Membrane Transport Proteins/deficiency , Dopamine/deficiency , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Aging/drug effects , Aging/genetics , Animals , Animals, Genetically Modified , Anxiety/genetics , Base Sequence , Dopamine/genetics , Dopamine Antagonists/pharmacology , Dopamine Plasma Membrane Transport Proteins/genetics , Male , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Zebrafish
14.
Adv Exp Med Biol ; 916: 61-86, 2016.
Article in English | MEDLINE | ID: mdl-27165349

ABSTRACT

Animal models of tumor initiation and tumor progression are essential components toward understanding cancer and designing/validating future therapies. Zebrafish is a powerful model for studying tumorigenesis and has been successfully exploited in drug discovery. According to the zebrafish reference genome, 82 % of disease-associated genes in the Online Mendelian Inheritance in Man (OMIM) database have clear zebrafish orthologues. Using a variety of large-scale random mutagenesis methods developed to date, zebrafish can provide a unique opportunity to identify gene mutations that may be associated with cancer predisposition. On the other hand, newer technologies enabling targeted mutagenesis can facilitate reverse cancer genetic studies and open the door for complex genetic analysis of tumorigenesis. In this chapter, we will describe the various technologies for conducting genome editing in zebrafish with special emphasis on the approaches to inactivate genes.


Subject(s)
Disease Models, Animal , Neoplasms/genetics , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Mutagenesis , Retroviridae/genetics , Transgenes , Zebrafish
15.
Nat Chem Biol ; 12(7): 552-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27239788

ABSTRACT

Humans and many animals show 'freezing' behavior in response to threatening stimuli. In humans, inappropriate threat responses are fundamental characteristics of several mental illnesses. To identify small molecules that modulate threat responses, we developed a high-throughput behavioral assay in zebrafish (Danio rerio) and evaluated 10,000 compounds for their effects on freezing behavior. We found three classes of compounds that switch the threat response from freezing to escape-like behavior. We then screened these for binding activity across 45 candidate targets. Using target profile clustering, we identified the sigma-1 (σ1) receptor as having a role in the mechanism of behavioral switching and confirmed that known σ1 ligands also disrupt freezing behavior. Furthermore, mutation of the gene encoding σ1 prevented the behavioral effect of escape-inducing compounds. One compound, which we call finazine, potently bound mammalian σ1 and altered threat-response behavior in mice. Thus, pharmacological and genetic interrogation of the freezing response revealed σ1 as a mediator of threat responses in vertebrates.


Subject(s)
Escape Reaction/drug effects , Freezing Reaction, Cataleptic/drug effects , Larva/drug effects , Receptors, sigma/metabolism , Small Molecule Libraries/pharmacology , Zebrafish , Anilides/chemistry , Anilides/metabolism , Anilides/pharmacology , Animals , Escape Reaction/radiation effects , Freezing Reaction, Cataleptic/radiation effects , High-Throughput Screening Assays , Larva/radiation effects , Ligands , Light , Mice , Molecular Structure , Piperazines/chemistry , Piperazines/metabolism , Piperazines/pharmacology , Receptors, sigma/genetics , Small Molecule Libraries/chemistry , Zebrafish/growth & development , Sigma-1 Receptor
16.
Cancer Cell ; 28(6): 773-784, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26678339

ABSTRACT

Heterozygous mutation of IDH1 in cancers modifies IDH1 enzymatic activity, reprogramming metabolite flux and markedly elevating 2-hydroxyglutarate (2-HG). Here, we found that 2-HG depletion did not inhibit growth of several IDH1 mutant solid cancer types. To identify other metabolic therapeutic targets, we systematically profiled metabolites in endogenous IDH1 mutant cancer cells after mutant IDH1 inhibition and discovered a profound vulnerability to depletion of the coenzyme NAD+. Mutant IDH1 lowered NAD+ levels by downregulating the NAD+ salvage pathway enzyme nicotinate phosphoribosyltransferase (Naprt1), sensitizing to NAD+ depletion via concomitant nicotinamide phosphoribosyltransferase (NAMPT) inhibition. NAD+ depletion activated the intracellular energy sensor AMPK, triggered autophagy, and resulted in cytotoxicity. Thus, we identify NAD+ depletion as a metabolic susceptibility of IDH1 mutant cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cytokines/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Isocitrate Dehydrogenase/genetics , Mutation , NAD/deficiency , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/drug effects , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cytokines/metabolism , Energy Metabolism/drug effects , Enzyme Activation , Female , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Glutarates/metabolism , HEK293 Cells , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/metabolism , Metabolomics/methods , Mice, SCID , Molecular Targeted Therapy , Nicotinamide Phosphoribosyltransferase/metabolism , Pentosyltransferases/metabolism , Signal Transduction/drug effects , Spheroids, Cellular , Time Factors , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Nature ; 523(7561): 481-5, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26098369

ABSTRACT

Although CRISPR-Cas9 nucleases are widely used for genome editing, the range of sequences that Cas9 can recognize is constrained by the need for a specific protospacer adjacent motif (PAM). As a result, it can often be difficult to target double-stranded breaks (DSBs) with the precision that is necessary for various genome-editing applications. The ability to engineer Cas9 derivatives with purposefully altered PAM specificities would address this limitation. Here we show that the commonly used Streptococcus pyogenes Cas9 (SpCas9) can be modified to recognize alternative PAM sequences using structural information, bacterial selection-based directed evolution, and combinatorial design. These altered PAM specificity variants enable robust editing of endogenous gene sites in zebrafish and human cells not currently targetable by wild-type SpCas9, and their genome-wide specificities are comparable to wild-type SpCas9 as judged by GUIDE-seq analysis. In addition, we identify and characterize another SpCas9 variant that exhibits improved specificity in human cells, possessing better discrimination against off-target sites with non-canonical NAG and NGA PAMs and/or mismatched spacers. We also find that two smaller-size Cas9 orthologues, Streptococcus thermophilus Cas9 (St1Cas9) and Staphylococcus aureus Cas9 (SaCas9), function efficiently in the bacterial selection systems and in human cells, suggesting that our engineering strategies could be extended to Cas9s from other species. Our findings provide broadly useful SpCas9 variants and, more importantly, establish the feasibility of engineering a wide range of Cas9s with altered and improved PAM specificities.


Subject(s)
CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Nucleotide Motifs , Protein Engineering/methods , Streptococcus pyogenes/enzymology , Amino Acid Substitution/genetics , Animals , CRISPR-Cas Systems , Cell Line , Directed Molecular Evolution , Genome/genetics , Humans , Mutation/genetics , Staphylococcus aureus/enzymology , Streptococcus thermophilus/enzymology , Substrate Specificity/genetics , Zebrafish/embryology , Zebrafish/genetics
18.
Methods Mol Biol ; 1311: 317-34, 2015.
Article in English | MEDLINE | ID: mdl-25981483

ABSTRACT

In recent years, the zebrafish has become a critical contributor to various areas of biomedical research, advancing our fundamental understanding of biomedicine and helping discover candidate therapeutics for human diseases. Nevertheless, to further extend the power of this important model organism requires a robust and simple-to-use genome editing platform that will enable targeted gene knockouts and introduction of specific mutations identified in human diseases into the zebrafish genome. We describe here protocols for creating insertion or deletion (indel) mutations or precise sequence modifications in zebrafish genes using customizable CRISPR-Cas9 RNA-guided nucleases (RGNs). These methods can be easily implemented in any lab and may also potentially be extended for use in other organisms.


Subject(s)
CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , Deoxyribonucleases/metabolism , Genetic Engineering/methods , Mutagenesis , RNA/genetics , Zebrafish/genetics , Animals , Embryo, Nonmammalian/metabolism , Genetic Vectors/genetics , Mutation , Zebrafish/embryology
19.
J Clin Invest ; 125(5): 1987-97, 2015 May.
Article in English | MEDLINE | ID: mdl-25866969

ABSTRACT

Patients with a germline mutation in von Hippel-Lindau (VHL) develop renal cell cancers and hypervascular tumors of the brain, adrenal glands, and pancreas as well as erythrocytosis. These phenotypes are driven by aberrant expression of HIF2α, which induces expression of genes involved in cell proliferation, angiogenesis, and red blood cell production. Currently, there are no effective treatments available for VHL disease. Here, using an animal model of VHL, we report a marked improvement of VHL-associated phenotypes following treatment with HIF2α inhibitors. Inactivation of vhl in zebrafish led to constitutive activation of HIF2α orthologs and modeled several aspects of the human disease, including erythrocytosis, pathologic angiogenesis in the brain and retina, and aberrant kidney and liver proliferation. Treatment of vhl(-/-) mutant embryos with HIF2α-specific inhibitors downregulated Hif target gene expression in a dose-dependent manner, improved abnormal hematopoiesis, and substantially suppressed erythrocytosis and angiogenic sprouting. Moreover, pharmacologic inhibition of HIF2α reversed the compromised cardiac contractility of vhl(-/-) embryos and partially rescued early lethality. This study demonstrates that small-molecule targeting of HIF2α improves VHL-related phenotypes in a vertebrate animal model and supports further exploration of this strategy for treating VHL disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Hydrazones/therapeutic use , Sulfones/therapeutic use , von Hippel-Lindau Disease/drug therapy , 5' Untranslated Regions , Amino Acids, Dicarboxylic/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/blood supply , Disease Models, Animal , Drug Evaluation, Preclinical , Embryo, Nonmammalian , Gene Expression Regulation/drug effects , Humans , Hydrazones/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney/pathology , Liver/pathology , Myocardial Contraction/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Phenotype , Polycythemia/drug therapy , Polycythemia/genetics , Retinal Vessels/pathology , Sulfones/pharmacology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology , von Hippel-Lindau Disease/physiopathology
20.
Methods Enzymol ; 546: 377-413, 2014.
Article in English | MEDLINE | ID: mdl-25398350

ABSTRACT

Genome editing using the Cas9 endonuclease of Streptococcus pyogenes has demonstrated unprecedented efficacy and facility in a wide variety of biological systems. In zebrafish, specifically, studies have shown that Cas9 can be directed to user-defined genomic target sites via synthetic guide RNAs, enabling random or homology-directed sequence alterations, long-range chromosomal deletions, simultaneous disruption of multiple genes, and targeted integration of several kilobases of DNA. Altogether, these methods are opening new doors for the engineering of knock-outs, conditional alleles, tagged proteins, reporter lines, and disease models. In addition, the ease and high efficiency of generating Cas9-mediated gene knock-outs provides great promise for high-throughput functional genomics studies in zebrafish. In this chapter, we briefly review the origin of CRISPR/Cas technology and discuss current Cas9-based genome-editing applications in zebrafish with particular emphasis on their designs and implementations.


Subject(s)
CRISPR-Cas Systems , Gene Targeting/methods , Zebrafish/genetics , Animals , Genetic Engineering/methods , Genome , INDEL Mutation , RNA, Guide, Kinetoplastida/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...