Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Dis Model Mech ; 17(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38235578

ABSTRACT

Skeletal muscular diseases predominantly affect skeletal and cardiac muscle, resulting in muscle weakness, impaired respiratory function and decreased lifespan. These harmful outcomes lead to poor health-related quality of life and carry a high healthcare economic burden. The absence of promising treatments and new therapies for muscular disorders requires new methods for candidate drug identification and advancement in animal models. Consequently, the rapid screening of drug compounds in an animal model that mimics features of human muscle disease is warranted. Zebrafish are a versatile model in preclinical studies that support developmental biology and drug discovery programs for novel chemical entities and repurposing of established drugs. Due to several advantages, there is an increasing number of applications of the zebrafish model for high-throughput drug screening for human disorders and developmental studies. Consequently, standardization of key drug screening parameters, such as animal husbandry protocols, drug compound administration and outcome measures, is paramount for the continued advancement of the model and field. Here, we seek to summarize and explore critical drug treatment and drug screening parameters in the zebrafish-based modeling of human muscle diseases. Through improved standardization and harmonization of drug screening parameters and protocols, we aim to promote more effective drug discovery programs.


Subject(s)
Muscular Diseases , Zebrafish , Animals , Humans , Zebrafish/physiology , Quality of Life , Disease Models, Animal , Muscular Diseases/drug therapy , Drug Evaluation, Preclinical/methods , Muscles
2.
Dis Model Mech ; 16(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36457161

ABSTRACT

Although renal macrophages have been shown to contribute to cyst development in polycystic kidney disease (PKD) animal models, it remains unclear whether there is a specific macrophage subpopulation involved. Here, we analyzed changes in macrophage populations during renal maturation in association with cystogenesis rates in conditional Pkd2 mutant mice. We observed that CD206+ resident macrophages were minimal in a normal adult kidney but accumulated in cystic areas in adult-induced Pkd2 mutants. Using Cx3cr1 null mice, we reduced macrophage number, including CD206+ macrophages, and showed that this significantly reduced cyst severity in adult-induced Pkd2 mutant kidneys. We also found that the number of CD206+ resident macrophage-like cells increased in kidneys and in the urine from autosomal-dominant PKD (ADPKD) patients relative to the rate of renal functional decline. These data indicate a direct correlation between CD206+ resident macrophages and cyst formation, and reveal that the CD206+ resident macrophages in urine could serve as a biomarker for renal cystic disease activity in preclinical models and ADPKD patients. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cysts , Polycystic Kidney, Autosomal Dominant , Mice , Animals , Kidney , Macrophages , Mice, Knockout , Biomarkers , Disease Models, Animal
3.
Dis Model Mech ; 15(12)2022 12 01.
Article in English | MEDLINE | ID: mdl-36533556

ABSTRACT

Meckel syndrome, nephronophthisis, Joubert syndrome and Bardet-Biedl syndrome are caused by mutations in proteins that localize to the ciliary transition zone (TZ). The phenotypically distinct syndromes suggest that these TZ proteins have differing functions. However, mutations in a single TZ gene can result in multiple syndromes, suggesting that the phenotype is influenced by modifier genes. We performed a comprehensive analysis of ten zebrafish TZ mutants, including mks1, tmem216, tmem67, rpgrip1l, cc2d2a, b9d2, cep290, tctn1, nphp1 and nphp4, as well as mutants in ift88 and ift172. Our data indicate that variations in phenotypes exist between different TZ mutants, supporting different tissue-specific functions of these TZ genes. Further, we observed phenotypic variations within progeny of a single TZ mutant, reminiscent of multiple disease syndromes being associated with mutations in one gene. In some mutants, the dynamics of the phenotype became complex with transitory phenotypes that are corrected over time. We also demonstrated that multiple-guide-derived CRISPR/Cas9 F0 'crispant' embryos recapitulate zygotic null phenotypes, and rapidly identified ciliary phenotypes in 11 cilia-associated gene candidates (ankfn1, ccdc65, cfap57, fhad1, nme7, pacrg, saxo2, c1orf194, ttc26, zmynd12 and cfap52).


Subject(s)
Cilia , Polycystic Kidney Diseases , Animals , Cilia/metabolism , Zebrafish/genetics , Penetrance , Syndrome , Polycystic Kidney Diseases/metabolism , Biological Variation, Population , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Vesicular Transport Proteins/genetics
4.
PLoS Genet ; 18(8): e1010341, 2022 08.
Article in English | MEDLINE | ID: mdl-35994499

ABSTRACT

Sister chromatid cohesion (SCC) is an important process in chromosome segregation. ESCO2 is essential for establishment of SCC and is often deleted/altered in human cancers. We demonstrate that esco2 haploinsufficiency results in reduced SCC and accelerates the timing of tumor onset in both zebrafish and mouse p53 heterozygous null models, but not in p53 homozygous mutant or wild-type animals. These data indicate that esco2 haploinsufficiency accelerates tumor onset in a loss of heterozygosity (LOH) sensitive background. Analysis of The Cancer Genome Atlas (TCGA) confirmed ESCO2 deficient tumors have elevated number of LOH events throughout the genome. Further, we demonstrated heterozygous loss of sgo1, important in maintaining SCC, also results in reduced SCC and accelerated tumor formation in a p53 heterozygous background. Surprisingly, while we did observe elevated levels of chromosome missegregation and micronuclei formation in esco2 heterozygous mutant animals, this chromosomal instability did not contribute to the accelerated tumor onset in a p53 heterozygous background. Interestingly, SCC also plays a role in homologous recombination, and we did observe elevated levels of mitotic recombination derived p53 LOH in tumors from esco2 haploinsufficient animals; as well as elevated levels of mitotic recombination throughout the genome of human ESCO2 deficient tumors. Together these data suggest that reduced SCC contributes to accelerated tumor penetrance through elevated mitotic recombination.


Subject(s)
Chromosome Segregation , Neoplasms , Acetyltransferases/genetics , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatids/genetics , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation/genetics , Humans , Mice , Neoplasms/genetics , Penetrance , Tumor Suppressor Protein p53/genetics , Zebrafish/genetics
5.
Genetics ; 220(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34850872

ABSTRACT

Primary cilia are sensory and signaling hubs with a protein composition that is distinct from the rest of the cell due to the barrier function of the transition zone (TZ) at the base of the cilium. Protein transport across the TZ is mediated in part by the BBSome, and mutations disrupting TZ and BBSome proteins cause human ciliopathy syndromes. Ciliopathies have phenotypic variability even among patients with identical genetic variants, suggesting a role for modifier loci. To identify potential ciliopathy modifiers, we performed a mutagenesis screen on nphp-4 mutant Caenorhabditis elegans and uncovered a novel allele of bbs-5. Nphp-4;bbs-5 double mutant worms have phenotypes not observed in either individual mutant strain. To test whether this genetic interaction is conserved, we also analyzed zebrafish and mouse mutants. While Nphp4 mutant zebrafish appeared overtly normal, Bbs5 mutants exhibited scoliosis. When combined, Nphp4;Bbs5 double mutant zebrafish did not exhibit synergistic effects, but the lack of a phenotype in Nphp4 mutants makes interpreting these data difficult. In contrast, Nphp4;Bbs5 double mutant mice were not viable and there were fewer mice than expected carrying three mutant alleles. In addition, postnatal loss of Bbs5 in mice using a conditional allele compromised survival when combined with an Nphp4 allele. As cilia are still formed in the double mutant mice, the exacerbated phenotype is likely a consequence of disrupted ciliary signaling. Collectively, these data support an evolutionarily conserved genetic interaction between Bbs5 and Nphp4 alleles that may contribute to the variability in ciliopathy phenotypes.


Subject(s)
Caenorhabditis elegans , Animals , Zebrafish
6.
Cell Death Dis ; 12(7): 659, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34193827

ABSTRACT

Cellular stress can lead to several human disease pathologies due to aberrant cell death. The p53 family (tp53, tp63, and tp73) and downstream transcriptional apoptotic target genes (PUMA/BBC3 and NOXA/PMAIP1) have been implicated as mediators of stress signals. To evaluate the importance of key stress response components in vivo, we have generated zebrafish null alleles in puma, noxa, p53, p63, and p73. Utilizing these genetic mutants, we have deciphered that the apoptotic response to genotoxic stress requires p53 and puma, but not p63, p73, or noxa. We also identified a delayed secondary wave of genotoxic stress-induced apoptosis that is p53/puma independent. Contrary to genotoxic stress, ER stress-induced apoptosis requires p63 and puma, but not p53, p73, or noxa. Lastly, the oxidative stress-induced apoptotic response requires p63, and both noxa and puma. Our data also indicate that while the neural tube is poised for apoptosis due to genotoxic stress, the epidermis is poised for apoptosis due to ER and oxidative stress. These data indicate there are convergent as well as unique molecular pathways involved in the different stress responses. The commonality of puma in these stress pathways, and the lack of gross or tumorigenic phenotypes with puma loss suggest that a inhibitor of Puma may have therapeutic application. In addition, we have also generated a knockout of the negative regulator of p53, mdm2 to further evaluate the p53-induced apoptosis. Our data indicate that the p53 null allele completely rescues the mdm2 null lethality, while the puma null completely rescues the mdm2 null apoptosis but only partially rescues the phenotype. Indicating Puma is the key mediator of p53-dependent apoptosis. Interestingly the p53 homozygous null zebrafish develop tumors faster than the previously described p53 homozygous missense mutant zebrafish, suggesting the missense allele may be hypomorphic allele.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , DNA Damage , Endoplasmic Reticulum Stress , Oxidative Stress , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Apoptosis/drug effects , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/genetics , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation , Macrolides/pharmacology , Oxidative Stress/drug effects , Phosphoproteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology , Time Factors , Trans-Activators/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , X-Rays , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Genome Med ; 13(1): 90, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34020708

ABSTRACT

BACKGROUND: We aimed to define the clinical and variant spectrum and to provide novel molecular insights into the DHX30-associated neurodevelopmental disorder. METHODS: Clinical and genetic data from affected individuals were collected through Facebook-based family support group, GeneMatcher, and our network of collaborators. We investigated the impact of novel missense variants with respect to ATPase and helicase activity, stress granule (SG) formation, global translation, and their effect on embryonic development in zebrafish. SG formation was additionally analyzed in CRISPR/Cas9-mediated DHX30-deficient HEK293T and zebrafish models, along with in vivo behavioral assays. RESULTS: We identified 25 previously unreported individuals, ten of whom carry novel variants, two of which are recurrent, and provide evidence of gonadal mosaicism in one family. All 19 individuals harboring heterozygous missense variants within helicase core motifs (HCMs) have global developmental delay, intellectual disability, severe speech impairment, and gait abnormalities. These variants impair the ATPase and helicase activity of DHX30, trigger SG formation, interfere with global translation, and cause developmental defects in a zebrafish model. Notably, 4 individuals harboring heterozygous variants resulting either in haploinsufficiency or truncated proteins presented with a milder clinical course, similar to an individual harboring a de novo mosaic HCM missense variant. Functionally, we established DHX30 as an ATP-dependent RNA helicase and as an evolutionary conserved factor in SG assembly. Based on the clinical course, the variant location, and type we establish two distinct clinical subtypes. DHX30 loss-of-function variants cause a milder phenotype whereas a severe phenotype is caused by HCM missense variants that, in addition to the loss of ATPase and helicase activity, lead to a detrimental gain-of-function with respect to SG formation. Behavioral characterization of dhx30-deficient zebrafish revealed altered sleep-wake activity and social interaction, partially resembling the human phenotype. CONCLUSIONS: Our study highlights the usefulness of social media to define novel Mendelian disorders and exemplifies how functional analyses accompanied by clinical and genetic findings can define clinically distinct subtypes for ultra-rare disorders. Such approaches require close interdisciplinary collaboration between families/legal representatives of the affected individuals, clinicians, molecular genetics diagnostic laboratories, and research laboratories.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , RNA Helicases/genetics , Animals , Biomarkers , Gene Expression , Gene Knockdown Techniques , Genetic Association Studies/methods , Germ-Line Mutation , HEK293 Cells , Humans , Immunohistochemistry , Mutation , Phenotype , RNA Helicases/chemistry , RNA Helicases/metabolism , Zebrafish
8.
Sci Rep ; 10(1): 19710, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33184372

ABSTRACT

The four transcription factors OCT4, SOX2, KLF4, and MYC (OSKM) together can convert human fibroblasts to induced pluripotent stem cells (iPSCs). It is, however, perplexing that they can do so only for a rare population of the starting cells with a long latency. Transcription factors (TFs) define identities of both the starting fibroblasts and the end product, iPSCs, and are also of paramount importance for the reprogramming process. It is critical to upregulate or activate the iPSC-enriched TFs while downregulate or silence the fibroblast-enriched TFs. This report explores the initial TF responses to OSKM as the molecular underpinnings for both the potency aspects and the limitation sides of the OSKM reprogramming. The authors first defined the TF reprogramome, i.e., the full complement of TFs to be reprogrammed. Most TFs were resistant to OSKM reprogramming at the initial stages, an observation consistent with the inefficiency and long latency of iPSC reprogramming. Surprisingly, the current analyses also revealed that most of the TFs (at least 83 genes) that did respond to OSKM induction underwent legitimate reprogramming. The initial legitimate transcriptional responses of TFs to OSKM reprogramming were also observed in the reprogramming fibroblasts from a different individual. Such early biased legitimate reprogramming of the responsive TFs aligns well with the robustness aspect of the otherwise inefficient and stochastic OSKM reprogramming.


Subject(s)
Cellular Reprogramming Techniques/methods , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Transcription Factors/genetics , Cells, Cultured , Cellular Reprogramming/drug effects , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Lentivirus/genetics , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic
9.
Am J Physiol Renal Physiol ; 316(3): F414-F425, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30566001

ABSTRACT

Deficiency in polycystin 1 triggers specific changes in energy metabolism. To determine whether defects in other human cystoproteins have similar effects, we studied extracellular acidification and glucose metabolism in human embryonic kidney (HEK-293) cell lines with polycystic kidney and hepatic disease 1 ( PKHD1) and polycystic kidney disease (PKD) 2 ( PKD2) truncating defects along multiple sites of truncating mutations found in patients with autosomal recessive and dominant PKDs. While neither the PKHD1 or PKD2 gene mutations nor their position enhanced cell proliferation rate in our cell line models, truncating mutations in these genes progressively increased overall extracellular acidification over time ( P < 0.001 for PKHD1 and PKD2 mutations). PKHD1 mutations increased nonglycolytic acidification rate (1.19 vs. 1.03, P = 0.002), consistent with an increase in tricarboxylic acid cycle activity or breakdown of intracellular glycogen. In addition, they increased basal and ATP-linked oxygen consumption rates [7.59 vs. 5.42 ( P = 0.015) and 4.55 vs. 2.98 ( P = 0.004)]. The PKHD1 and PKD2 mutations also altered mitochondrial morphology, resembling the effects of polycystin 1 deficiency. Together, these data suggest that defects in major PKD genes trigger changes in mitochondrial energy metabolism. After validation in in vivo models, these initial observations would indicate potential benefits of targeting energy metabolism in the treatment of PKDs.


Subject(s)
Energy Metabolism/genetics , Glucose/metabolism , Protein Kinases/genetics , Receptors, Cell Surface/genetics , Cell Proliferation/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , HEK293 Cells , Humans , Mutation , Protein Kinase D2 , Protein Kinases/metabolism , Receptors, Cell Surface/metabolism
10.
J Vis Exp ; (113)2016 07 15.
Article in English | MEDLINE | ID: mdl-27501381

ABSTRACT

Mitosis is critical for organismal growth and differentiation. The process is highly dynamic and requires ordered events to accomplish proper chromatin condensation, microtubule-kinetochore attachment, chromosome segregation, and cytokinesis in a small time frame. Errors in the delicate process can result in human disease, including birth defects and cancer. Traditional approaches investigating human mitotic disease states often rely on cell culture systems, which lack the natural physiology and developmental/tissue-specific context advantageous when studying human disease. This protocol overcomes many obstacles by providing a way to visualize, with high resolution, chromosome dynamics in a vertebrate system, the zebrafish. This protocol will detail an approach that can be used to obtain dynamic images of dividing cells, which include: in vitro transcription, zebrafish breeding/collecting, embryo embedding, and time-lapse imaging. Optimization and modifications of this protocol are also explored. Using H2A.F/Z-EGFP (labels chromatin) and mCherry-CAAX (labels cell membrane) mRNA-injected embryos, mitosis in AB wild-type, auroraB(hi1045) (,) and esco2(hi2865) mutant zebrafish is visualized. High resolution live imaging in zebrafish allows one to observe multiple mitoses to statistically quantify mitotic defects and timing of mitotic progression. In addition, observation of qualitative aspects that define improper mitotic processes (i.e., congression defects, missegregation of chromosomes, etc.) and improper chromosomal outcomes (i.e., aneuploidy, polyploidy, micronuclei, etc.) are observed. This assay can be applied to the observation of tissue differentiation/development and is amenable to the use of mutant zebrafish and pharmacological agents. Visualization of how defects in mitosis lead to cancer and developmental disorders will greatly enhance understanding of the pathogenesis of disease.


Subject(s)
Embryo, Nonmammalian/cytology , Mitosis , Time-Lapse Imaging , Zebrafish/embryology , Animals , Cell Cycle Proteins , Chromosome Segregation , Cytokinesis , Humans , Kinetochores , Microtubules
11.
PLoS Genet ; 12(7): e1006220, 2016 07.
Article in English | MEDLINE | ID: mdl-27472056

ABSTRACT

Ciliopathies are genetic disorders arising from dysfunction of microtubule-based cellular appendages called cilia. Different cilia types possess distinct stereotypic microtubule doublet arrangements with non-motile or 'primary' cilia having a 9+0 and motile cilia have a 9+2 array of microtubule doublets. Primary cilia are critical sensory and signaling centers needed for normal mammalian development. Defects in their structure/function result in a spectrum of clinical and developmental pathologies including abnormal neural tube and limb patterning. Altered patterning phenotypes in the limb and neural tube are due to perturbations in the hedgehog (Hh) signaling pathway. Motile cilia are important in fluid movement and defects in motility result in chronic respiratory infections, altered left-right asymmetry, and infertility. These features are the hallmarks of Primary Ciliary Dyskinesia (PCD, OMIM 244400). While mutations in several genes are associated with PCD in patients and animal models, the genetic lesion in many cases is unknown. We assessed the in vivo functions of Growth Arrest Specific 8 (GAS8). GAS8 shares strong sequence similarity with the Chlamydomonas Nexin-Dynein Regulatory Complex (NDRC) protein 4 (DRC4) where it is needed for proper flagella motility. In mammalian cells, the GAS8 protein localizes not only to the microtubule axoneme of motile cilia, but also to the base of non-motile cilia. Gas8 was recently implicated in the Hh signaling pathway as a regulator of Smoothened trafficking into the cilium. Here, we generate the first mouse with a Gas8 mutation and show that it causes severe PCD phenotypes; however, there were no overt Hh pathway phenotypes. In addition, we identified two human patients with missense variants in Gas8. Rescue experiments in Chlamydomonas revealed a subtle defect in swim velocity compared to controls. Further experiments using CRISPR/Cas9 homology driven repair (HDR) to generate one of these human missense variants in mice demonstrated that this allele is likely pathogenic.


Subject(s)
Body Patterning/genetics , Cilia/genetics , Kartagener Syndrome/genetics , Proteins/genetics , Animals , Cell Movement/genetics , Chlamydomonas/genetics , Cilia/pathology , Cytoskeletal Proteins , Cytoskeleton/genetics , Disease Models, Animal , Extremities/growth & development , Extremities/pathology , Genetic Predisposition to Disease , Humans , Kartagener Syndrome/pathology , Mice , Microtubules/genetics , Mutation , Neural Tube/growth & development , Neural Tube/pathology , Signal Transduction/genetics
12.
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
13.
Dis Model Mech ; 8(8): 941-55, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26044958

ABSTRACT

Mutations in ESCO2, one of two establishment of cohesion factors necessary for proper sister chromatid cohesion (SCC), cause a spectrum of developmental defects in the autosomal-recessive disorder Roberts syndrome (RBS), warranting in vivo analysis of the consequence of cohesion dysfunction. Through a genetic screen in zebrafish targeting embryonic-lethal mutants that have increased genomic instability, we have identified an esco2 mutant zebrafish. Utilizing the natural transparency of zebrafish embryos, we have developed a novel technique to observe chromosome dynamics within a single cell during mitosis in a live vertebrate embryo. Within esco2 mutant embryos, we observed premature chromatid separation, a unique chromosome scattering, prolonged mitotic delay, and genomic instability in the form of anaphase bridges and micronuclei formation. Cytogenetic studies indicated complete chromatid separation and high levels of aneuploidy within mutant embryos. Amongst aneuploid spreads, we predominantly observed decreases in chromosome number, suggesting that either cells with micronuclei or micronuclei themselves are eliminated. We also demonstrated that the genomic instability leads to p53-dependent neural tube apoptosis. Surprisingly, although many cells required Esco2 to establish cohesion, 10-20% of cells had only weakened cohesion in the absence of Esco2, suggesting that compensatory cohesion mechanisms exist in these cells that undergo a normal mitotic division. These studies provide a unique in vivo vertebrate view of the mitotic defects and consequences of cohesion establishment loss, and they provide a compensation-based model to explain the RBS phenotypes.


Subject(s)
Acetyltransferases/genetics , Chromatids/metabolism , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Ectromelia/genetics , Ectromelia/pathology , Hypertelorism/genetics , Hypertelorism/pathology , Mutation/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Acetyltransferases/deficiency , Acetyltransferases/metabolism , Animals , Apoptosis , Chromosome Segregation , Chromosomes/metabolism , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Genomic Instability , Mitotic Index , Models, Biological , Mutagenesis, Insertional/genetics , Neural Tube/metabolism , Neural Tube/pathology , Phenotype , Retroviridae/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/embryology , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism
14.
Drug Metab Dispos ; 43(7): 1037-44, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25934576

ABSTRACT

Since its identification in 2000, sulfotransferase (SULT) 4A1 has presented an enigma to the field of cytosolic SULT biology. SULT4A1 is exclusively expressed in neural tissue, is highly conserved, and has been identified in every vertebrate studied to date. Despite this singular level of conservation, no substrate or function for SULT4A1 has been identified. Previous studies demonstrated that SULT4A1 does not bind the obligate sulfate donor, 3'-phosphoadenosine-5'-phosphosulfate, yet SULT4A1 is classified as a SULT superfamily member based on sequence and structural similarities to the other SULTs. In this study, transcription activator-like effector nucleases were used to generate heritable mutations in the SULT4A1 gene of zebrafish. The mutation (SULT4A1(Δ8)) consists of an 8-nucleotide deletion within the second exon of the gene, resulting in a frameshift mutation and premature stop codon after 132 AA. During early adulthood, casual observations were made that mutant zebrafish were exhibiting excessively sedentary behavior during the day. These observations were inconsistent with published reports on activity in zebrafish that are largely diurnal organisms and are highly active during the day. Thus, a decrease in activity during the day represents an abnormal behavior and warranted further systematic analysis. EthoVision video tracking software was used to monitor activity levels in wild-type (WT) and SULT4A1(Δ8/Δ8) fish over 48 hours of a normal light/dark cycle. SULT4A1(Δ8/Δ8) fish were shown to exhibit increased inactivity bout length and frequency as well as a general decrease in daytime activity levels when compared with their WT counterparts.


Subject(s)
Frameshift Mutation/genetics , Motor Activity/genetics , Sulfotransferases/genetics , Sulfotransferases/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Anxiety/genetics , Anxiety/psychology , Base Sequence , Deoxyribonucleases/administration & dosage , Deoxyribonucleases/pharmacology , Embryo, Nonmammalian , Exons , Microinjections , Molecular Sequence Data , Mutation , Social Behavior
15.
PLoS One ; 9(12): e114632, 2014.
Article in English | MEDLINE | ID: mdl-25503746

ABSTRACT

With the goal to generate and characterize the phenotypes of null alleles in all genes within an organism and the recent advances in custom nucleases, genome editing limitations have moved from mutation generation to mutation detection. We previously demonstrated that High Resolution Melting (HRM) analysis is a rapid and efficient means of genotyping known zebrafish mutants. Here we establish optimized conditions for HRM based detection of novel mutant alleles. Using these conditions, we demonstrate that HRM is highly efficient at mutation detection across multiple genome editing platforms (ZFNs, TALENs, and CRISPRs); we observed nuclease generated HRM positive targeting in 1 of 6 (16%) open pool derived ZFNs, 14 of 23 (60%) TALENs, and 58 of 77 (75%) CRISPR nucleases. Successful targeting, based on HRM of G0 embryos correlates well with successful germline transmission (46 of 47 nucleases); yet, surprisingly mutations in the somatic tail DNA weakly correlate with mutations in the germline F1 progeny DNA. This suggests that analysis of G0 tail DNA is a good indicator of the efficiency of the nuclease, but not necessarily a good indicator of germline alleles that will be present in the F1s. However, we demonstrate that small amplicon HRM curve profiles of F1 progeny DNA can be used to differentiate between specific mutant alleles, facilitating rare allele identification and isolation; and that HRM is a powerful technique for screening possible off-target mutations that may be generated by the nucleases. Our data suggest that micro-homology based alternative NHEJ repair is primarily utilized in the generation of CRISPR mutant alleles and allows us to predict likelihood of generating a null allele. Lastly, we demonstrate that HRM can be used to quickly distinguish genotype-phenotype correlations within F1 embryos derived from G0 intercrosses. Together these data indicate that custom nucleases, in conjunction with the ease and speed of HRM, will facilitate future high-throughput mutation generation and analysis needed to establish mutants in all genes of an organism.


Subject(s)
Genetic Engineering/methods , Genomics , Genotyping Techniques/methods , Phenotype , Transition Temperature , Zebrafish/genetics , Animals , Base Composition , Base Sequence , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA Damage , DNA Mutational Analysis , Deoxyribonucleases/chemistry , Deoxyribonucleases/metabolism , Hybridization, Genetic , Mutation , Nucleic Acid Denaturation , Zinc Fingers
16.
Drug Metab Dispos ; 42(5): 947-53, 2014 May.
Article in English | MEDLINE | ID: mdl-24553382

ABSTRACT

Sulfotransferase (SULT) 4A1 is an orphan enzyme that shares distinct structure and sequence similarities with other cytosolic SULTs. SULT4A1 is primarily expressed in neuronal tissue and is also the most conserved SULT, having been identified in every vertebrate investigated to date. Certain haplotypes of the SULT4A1 gene are correlated with higher baseline psychopathology in schizophrenic patients, but no substrate or function for SULT4A1 has yet been identified despite its high level of sequence conservation. In this study, deep RNA sequencing was used to search for alterations in gene expression in 72-hour postfertilization zebrafish larvae following transient SULT4A1 knockdown (KD) utilizing splice blocking morpholino oligonucleotides. This study demonstrates that transient inhibition of SULT4A1 expression in developing zebrafish larvae results in the up-regulation of several genes involved in phototransduction. SULT4A1 KD was verified by immunoblot analysis and quantitative real-time polymerase chain reaction (qPCR). Gene regulation changes identified by deep RNA sequencing were validated by qPCR. This study is the first identification of a cellular process whose regulation appears to be associated with SULT4A1 expression.


Subject(s)
Gene Expression Regulation, Developmental , Light Signal Transduction/genetics , Sulfotransferases/physiology , Transcriptome , Zebrafish Proteins/physiology , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/embryology , Brain/metabolism , Eye/embryology , Eye/metabolism , Fertilization , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Larva , Molecular Sequence Data , Morpholinos/pharmacology , RNA/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Sequence Homology, Amino Acid , Sulfotransferases/genetics , Up-Regulation , Zebrafish/embryology , Zebrafish Proteins/genetics
17.
Dis Model Mech ; 3(1-2): 45-56, 2010.
Article in English | MEDLINE | ID: mdl-20075382

ABSTRACT

Li-Fraumeni syndrome (LFS) is a highly penetrant, autosomal dominant, human familial cancer predisposition. Although a key role for the tumor suppressor p53 has been implicated in LFS, the genetic and cellular mechanisms underpinning this disease remain unknown. Therefore, modeling LFS in a vertebrate system that is accessible to both large-scale genetic screens and in vivo cell biological studies will facilitate the in vivo dissection of disease mechanisms, help identify candidate genes, and spur the discovery of therapeutic compounds. Here, we describe a forward genetic screen in zebrafish embryos that was used to identify LFS candidate genes, which yielded a p53 mutant (p53(I166T)) that as an adult develops tumors, predominantly sarcomas, with 100% penetrance. As in humans with LFS, tumors arise in heterozygotes and display loss of heterozygosity (LOH). This report of LOH indicates that Knudson's two-hit hypothesis, a hallmark of human autosomal dominant cancer syndromes, can be modeled in zebrafish. Furthermore, as with some LFS mutations, the zebrafish p53(I166T) allele is a loss-of-function allele with dominant-negative activity in vivo. Additionally, we demonstrate that the p53 regulatory pathway, including Mdm2 regulation, is evolutionarily conserved in zebrafish, providing a bona fide biological context in which to systematically uncover novel modifier genes and therapeutic agents for human LFS.


Subject(s)
Li-Fraumeni Syndrome/genetics , Models, Genetic , Zebrafish/genetics , Alleles , Animals , Apoptosis/radiation effects , DNA Damage , Disease Models, Animal , Gene Knockdown Techniques , Genes, Dominant/genetics , Genetic Testing , Heterozygote , Loss of Heterozygosity/genetics , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Stability/radiation effects , Proto-Oncogene Proteins c-mdm2/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects , Transcriptional Activation/genetics , Transcriptional Activation/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
Dev Dyn ; 238(12): 3168-74, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19890916

ABSTRACT

In order to facilitate high throughput genotyping of zebrafish, we have developed a novel technique that uses High Resolution Melting Analysis (HRMA) to distinguish wild-type, heterozygous mutants and homogyzous mutants. This one hour technique removes the need for restriction enzymes and agarose gels. The generated melting curve profiles are sensitive enough to detect non-specific PCR products. We have been able to reliably genotype three classes of mutations in zebrafish, including point mutants, apc(hu745) (apc(mcr)), and p53(zy7) (p53(I166T)), a small deletion mutant (bap28(y75)) and a retroviral insertion mutant (wdr43(hi821a)). This technique can genotype individual zebrafish embryos and adults (by tail-clip) and is applicable to other model organisms.


Subject(s)
Cytogenetic Analysis/methods , DNA Mutational Analysis/methods , Zebrafish/genetics , Animals , Animals, Genetically Modified , Base Sequence , Efficiency , Embryo, Nonmammalian , Gene Deletion , Genotype , Molecular Sequence Data , Mutagenesis, Insertional , Mutant Proteins/analysis , Mutant Proteins/genetics , Polymorphism, Single Nucleotide , Retroviridae/genetics , Retroviridae/physiology , Time Factors , Zebrafish/embryology , Zebrafish/growth & development , Zebrafish/metabolism
19.
Cancer Res ; 68(21): 8968-75, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18974141

ABSTRACT

Ewing's sarcoma is characterized by the t(11;22)(q24:q12) reciprocal translocation. To study the effects of the fusion gene EWS-FLI1 on development and tumor formation, a transgenic mouse model was created. A strategy of conditional expression was used to limit the potentially deleterious effects of EWS-FLI1 to certain tissues. In the absence of Cre recombinase, EWS-FLI1 was not expressed in the EWS-FLI1 transgenic mice, and they had a normal phenotype. When crossed to the Prx1-Cre transgenic mouse, which expresses Cre recombinase in the primitive mesenchymal cells of the embryonic limb bud, the EF mice were noted to have a number of developmental defects of the limbs. These included shortening of the limbs, muscle atrophy, cartilage dysplasia, and immature bone. By itself, EWS-FLI1 did not induce the formation of tumors in the EF transgenic mice. However, in the setting of p53 deletion, EWS-FLI1 accelerated the formation of sarcomas from a median time of 50 to 21 weeks. Furthermore, EWS-FLI1 altered the type of tumor that formed. Conditional deletion of p53 in mesenchymal cells (Prx1-Cre p53(lox/lox)) produced osteosarcomas as the predominant tumor. The presence of EWS-FLI1 shifted the tumor phenotype to a poorly differentiated sarcoma. The results taken together suggest that EWS-FLI1 inhibits normal limb development and accelerates the formation of poorly differentiated sarcomas.


Subject(s)
Limb Deformities, Congenital/genetics , Oncogene Proteins, Fusion/physiology , Sarcoma, Experimental/pathology , Transcription Factors/physiology , Animals , Base Sequence , Blotting, Western , Cell Division , DNA Primers , Immunohistochemistry , Mice , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Experimental/genetics , Transcription Factors/genetics
20.
Dev Dyn ; 236(11): 3088-99, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17937395

ABSTRACT

Transgenesis is an important tool for assessing gene function. In zebrafish, transgenesis has suffered from three problems: the labor of building complex expression constructs using conventional subcloning; low transgenesis efficiency, leading to mosaicism in transient transgenics and infrequent germline incorporation; and difficulty in identifying germline integrations unless using a fluorescent marker transgene. The Tol2kit system uses site-specific recombination-based cloning (multisite Gateway technology) to allow quick, modular assembly of [promoter]-[coding sequence]-[3' tag] constructs in a Tol2 transposon backbone. It includes a destination vector with a cmlc2:EGFP (enhanced green fluorescent protein) transgenesis marker and a variety of widely useful entry clones, including hsp70 and beta-actin promoters; cytoplasmic, nuclear, and membrane-localized fluorescent proteins; and internal ribosome entry sequence-driven EGFP cassettes for bicistronic expression. The Tol2kit greatly facilitates zebrafish transgenesis, simplifies the sharing of clones, and enables large-scale projects testing the functions of libraries of regulatory or coding sequences.


Subject(s)
Animals, Genetically Modified , Cloning, Molecular/methods , DNA Transposable Elements , DNA, Recombinant/genetics , Gene Transfer Techniques , Zebrafish/genetics , Animals , Genetic Techniques , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Plasmids/genetics , Recombination, Genetic , Transposases/metabolism , Zebrafish/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...