Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Yeast ; 36(10): 607-615, 2019 10.
Article in English | MEDLINE | ID: mdl-31301239

ABSTRACT

Saccharomyces cerevisiae is a genetically facile organism, yet multiple CRISPR/Cas9 techniques are widely used to edit its genome more efficiently and cost effectively than conventional methods. The absence of selective markers makes CRISPR/Cas9 editing particularly useful when making mutations within genes or regulatory sequences. Heterozygous mutations within genes frequently arise in the winners of evolution experiments. The genetic dissection of heterozygous alleles can be important to understanding gene structure and function. Unfortunately, the high efficiency of genome cutting and repair makes the introduction of heterozygous alleles by standard CRISPR/Cas9 technique impossible. To be able to quickly and reliably determine the individual phenotypes of the thousands of heterozygous mutations that can occur during directed evolutions is of particular interest to industrial strain improvement research. In this report, we describe a CRISPR/Cas9 method that introduces specific heterozygous mutations into the S. cerevisiae genome. This method relies upon creating silent point mutations in the protospacer adjacent motif site or removing the protospacer adjacent motif site entirely to stop the multiple rounds of genome editing that prevent heterozygous alleles from being generated. This technique should be able to create heterozygous alleles in other diploid yeasts and different allelic copy numbers in polyploid cells.


Subject(s)
Alleles , CRISPR-Cas Systems , Gene Editing/methods , Genome, Fungal , Saccharomyces cerevisiae/genetics , CRISPR-Associated Protein 9/genetics , Heterozygote , Mutation , Phenotype , RNA Editing
2.
PLoS One ; 11(3): e0151521, 2016.
Article in English | MEDLINE | ID: mdl-27010479

ABSTRACT

Biliary atresia (BA) is a progressive fibro-inflammatory disorder that is the leading indication for liver transplantation in children. Although there is evidence implicating genetic, infectious, environmental, and inflammatory causes, the etiology of BA remains unknown. We have recently reported that cholangiocytes from BA patients showed decreased DNA methylation relative to disease- and non-disease controls, supporting a potential role for DNA hypomethylation in BA etiopathogenesis. In the current study, we examined the methylation status of specific genes in human BA livers using methylation microarray technology. We found global DNA hypomethylation in BA samples as compared to disease- and non-disease controls at specific genetic loci. Hedgehog pathway members, SHH and GLI2, known to be upregulated in BA, were both hypomethylated, validating this approach as an investigative tool. Another region near the PDGFA locus was the most significantly hypomethylated in BA, suggesting potential aberrant expression. Validation assays confirmed increased transcriptional and protein expression of PDGFA in BA livers. We also show that PDGF-A protein is specifically localized to cholangiocytes in human liver samples. Injection of PDGF-AA protein dimer into zebrafish larvae caused biliary developmental and functional defects. In addition, activation of the Hedgehog pathway caused increased expression of PDGF-A in zebrafish larvae, providing a previously unrecognized link between PDGF and the Hedgehog pathway. Our findings implicate DNA hypomethylation as a specific factor in mediating overexpression of genes associated with BA and identify PDGF as a new candidate in BA pathogenesis.


Subject(s)
Biliary Atresia/genetics , DNA Methylation , Gene Expression Regulation , Platelet-Derived Growth Factor/genetics , Animals , Bile Ducts/pathology , Biliary Atresia/pathology , Genetic Loci , Hedgehog Proteins/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Liver/metabolism , Nuclear Proteins/genetics , Platelet-Derived Growth Factor/administration & dosage , Up-Regulation , Zebrafish/genetics , Zinc Finger Protein Gli2
3.
Hepatology ; 60(5): 1581-92, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25043405

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD), the accumulation of lipid within hepatocytes, is increasing in prevalence. Increasing fructose consumption correlates with this increased prevalence, and rodent studies directly support fructose leading to NAFLD. The mechanisms of NAFLD and in particular fructose-induced lipid accumulation remain unclear, although there is evidence for a role for endoplasmic reticulum (ER) stress and oxidative stress. We have evidence that NAFLD models demonstrate activation of the target of rapamycin complex 1 (Torc1) pathway. We set out to assess the contribution of ER stress, oxidative stress, and Torc1 up-regulation in the development of steatohepatitis in fructose-treated larval zebrafish. Zebrafish were treated with fructose or glucose as a calorie-matched control. We also treated larvae with rapamycin, tunicamycin (ER stress), or valinomycin (oxidative stress). Fish were stained with oil red O to assess hepatic lipid accumulation, and we also performed quantitative polymerase chain reaction (qPCR)and western blot analysis. We performed immunostaining on samples from patients with NAFLD and nonalcoholic steatohepatitis (NASH). Treatment with fructose induced hepatic lipid accumulation, mitochondrial abnormalities, and ER defects. In addition, fructose-treated fish showed activation of inflammatory and lipogenic genes. Treatment with tunicamycin or valinomycin also induced hepatic lipid accumulation. Expression microarray studies of zebrafish NAFLD models showed an elevation of genes downstream of Torc1 signaling. Rapamycin treatment of fructose-treated fish prevented development of hepatic steatosis, as did treatment of tunicamycin- or valinomycin-treated fish. Examination of liver samples from patients with hepatic steatosis demonstrated activation of Torc1 signaling. CONCLUSION: Fructose treatment of larval zebrafish induces hepatic lipid accumulation, inflammation, and oxidative stress. Our results indicate that Torc1 activation is required for hepatic lipid accumulation across models of NAFLD, and in patients.


Subject(s)
Disease Models, Animal , Fatty Liver/etiology , Fructose/adverse effects , TOR Serine-Threonine Kinases/metabolism , Zebrafish , Animals , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Endoplasmic Reticulum Stress , Fatty Liver/drug therapy , Fatty Liver/metabolism , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Up-Regulation
4.
Zebrafish ; 10(2): 177-83, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23448251

ABSTRACT

Biliary atresia (BA) is the most common identifiable hepatobiliary disease affecting infants, in which there are defects in intra- and extrahepatic bile ducts and progressive fibrosis. Activation of interferon-gamma (IFNγ) appears to be critical in both patients with BA and in rodent models of BA. We have recently reported a zebrafish model of biliary disease that shares features with BA, in which inhibition of DNA methylation leads to intrahepatic biliary defects and activation of IFNγ target genes. Here we report that ifng genes are hypomethylated and upregulated in zebrafish larvae treated with azacytidine (azaC), an inhibitor of DNA methylation. Injection of IFNγ protein into developing zebrafish larvae leads to biliary defects, suggesting that activation of the IFNγ pathway is sufficient to cause developmental biliary defects. These defects are associated with decreased cholangiocyte proliferation and with a decrease in the expression of vhnf1 (hnf1b, tcf2), which encodes a homeodomain protein with previously reported roles in biliary development in multiple models. These results support an importance of IFNγ in mediating biliary defects, and also demonstrate the feasibility of direct injection of intact protein into developing zebrafish larvae.


Subject(s)
Azacitidine/pharmacology , Biliary Atresia/genetics , Interferon-gamma/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Biliary Atresia/etiology , Biliary Tract/growth & development , Biliary Tract/metabolism , DNA Methylation/drug effects , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Interferon-gamma/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/metabolism
5.
Gastroenterology ; 144(5): 1107-1115.e3, 2013 May.
Article in English | MEDLINE | ID: mdl-23336978

ABSTRACT

BACKGROUND & AIMS: Biliary atresia (BA) is a progressive fibroinflammatory disorder of infants involving the extrahepatic and intrahepatic biliary tree. Its etiology is unclear but is believed to involve exposure of a genetically susceptible individual to certain environmental factors. BA occurs exclusively in the neonatal liver, so variants of genes expressed during hepatobiliary development could affect susceptibility. Genome-wide association studies previously identified a potential region of interest at 2q37. We continued these studies to narrow the region and identify BA susceptibility genes. METHODS: We searched for copy number variants that were increased among patients with BA (n = 61) compared with healthy individuals (controls; n = 5088). After identifying a candidate gene, we investigated expression patterns of orthologues in zebrafish liver and the effects of reducing expression, with morpholino antisense oligonucleotides, on biliary development, gene expression, and signal transduction. RESULTS: We observed a statistically significant increase in deletions at 2q37.3 in patients with BA that resulted in deletion of one copy of GPC1, which encodes glypican 1, a heparan sulfate proteoglycan that regulates Hedgehog signaling and inflammation. Knockdown of gpc1 in zebrafish led to developmental biliary defects. Exposure of the gpc1 morphants to cyclopamine, a Hedgehog antagonist, partially rescued the gpc1-knockdown phenotype. Injection of zebrafish with recombinant Sonic Hedgehog led to biliary defects similar to those of the gpc1 morphants. Liver samples from patients with BA had reduced levels of apical GPC1 in cholangiocytes compared with samples from controls. CONCLUSIONS: Based on genetic analysis of patients with BA and zebrafish, GPC1 appears to be a BA susceptibility gene. These findings also support a role for Hedgehog signaling in the pathogenesis of BA.


Subject(s)
Biliary Atresia/genetics , DNA/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Heparan Sulfate Proteoglycans/genetics , Zebrafish/genetics , Animals , Biliary Atresia/metabolism , Child , Disease Models, Animal , Genome-Wide Association Study , Heparan Sulfate Proteoglycans/biosynthesis , Humans , In Situ Hybridization , Polymerase Chain Reaction , Zebrafish/metabolism
6.
Dev Biol ; 365(2): 434-44, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22465374

ABSTRACT

We identified three zebrafish mutants with defects in biliary development. One of these mutants, pekin (pn), also demonstrated generalized hypopigmentation and other defects, including disruption of retinal cell layers, lack of zymogen granules in the pancreas, and dilated Golgi in intestinal epithelial cells. Bile duct cells in pn demonstrated an accumulation of electron dense bodies. We determined that the causative defect in pn was a splice site mutation in the atp6ap2 gene that leads to an inframe stop codon. atp6ap2 encodes a subunit of the vacuolar H(+)-ATPase (V-H(+)-ATPase), which modulates pH in intracellular compartments. The Atp6ap2 subunit has also been shown to function as an intracellular renin receptor that stimulates fibrogenesis. Here we show that mutants and morphants involving other V-H(+)-ATPase subunits also demonstrated developmental biliary defects, but did not demonstrate the inhibition of fibrogenic genes observed in pn. The defects in pn are reminiscent of those we and others have observed in class C VPS (vacuolar protein sorting) family mutants and morphants, and we report here that knockdown of atp6ap2 and vps33b had an additive negative effect on biliary development. Our findings suggest that pathways which are important in modulating intracompartmental pH lead to defects in digestive organ development, and support previous studies demonstrating the importance of intracellular sorting pathways in biliary development.


Subject(s)
Biliary Tract/abnormalities , Membrane Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/abnormalities , Animals , Biliary Tract/enzymology , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Membrane Proteins/genetics , Mutation , Protein Subunits/genetics , Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Hepatology ; 53(3): 905-14, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21319190

ABSTRACT

UNLABELLED: Infantile cholestatic disorders arise in the context of progressively developing intrahepatic bile ducts. Biliary atresia (BA), a progressive fibroinflammatory disorder of extra- and intrahepatic bile ducts, is the most common identifiable cause of infantile cholestasis and the leading indication for liver transplantation in children. The etiology of BA is unclear, and although there is some evidence for viral, toxic, and complex genetic causes, the exclusive occurrence of BA during a period of biliary growth and remodeling suggests an importance of developmental factors. Interestingly, interferon-γ (IFN-γ) signaling is activated in patients and in the frequently utilized rhesus rotavirus mouse model of BA, and is thought to play a key mechanistic role. Here we demonstrate intrahepatic biliary defects and up-regulated hepatic expression of IFN-γ pathway genes caused by genetic or pharmacological inhibition of DNA methylation in zebrafish larvae. Biliary defects elicited by inhibition of DNA methylation were reversed by treatment with glucocorticoid, suggesting that the activation of inflammatory pathways was critical. DNA methylation was significantly reduced in bile duct cells from BA patients compared to patients with other infantile cholestatic disorders, thereby establishing a possible etiologic link between decreased DNA methylation, activation of IFN-γ signaling, and biliary defects in patients. CONCLUSION: Inhibition of DNA methylation leads to biliary defects and activation of IFN-γ-responsive genes, thus sharing features with BA, which we determine to be associated with DNA hypomethylation. We propose epigenetic activation of IFN-γ signaling as a common etiologic mechanism of intrahepatic bile duct defects in BA.


Subject(s)
Bile Ducts/abnormalities , Bile Ducts/growth & development , DNA Methylation/drug effects , Adenosylhomocysteinase/genetics , Animals , Azacitidine/pharmacology , Bile Ducts/drug effects , Biliary Atresia/etiology , Biliary Atresia/physiopathology , Epigenomics , Humans , Inflammation/physiopathology , Interferon-gamma/physiology , Zebrafish
8.
Gastroenterology ; 136(3): 1081-90, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19185580

ABSTRACT

BACKGROUND & AIMS: The function of microRNA (miRNA) in liver development is unknown. To address this issue, we characterized miRNA expression in the embryonic mouse liver, performed functional miRNA analysis in zebrafish larvae, and identified novel hepatic miRNA targets. METHODS: Hepatic RNA isolated from mice at embryonic days 15.5, 18.5, and postnatal day 2 was hybridized to a mouse miRNA microarray. The microarray results were confirmed by Northern blot hybridization and quantitative reverse-transcription polymerase chain reaction. The spatial distribution of selected miRNAs was determined by in situ hybridization. Functional analysis of miR-30a was performed in zebrafish using antisense-mediated miRNA knockdown. Targets of miR-30a were identified by microarray analysis of gene expression following knockdown in cultured cells. RESULTS: A set of 38 differentially expressed fetal hepatic miRNAs was identified. Several of these miRNAs were found to exhibit distinct temporal and spatial patterns of expression in hepatocytes, cholangiocytes, and nonepithelial cells within the liver. Two (miR-30a and miR-30c) are the first examples of ductal plate and bile duct-specific hepatic miRNAs. Knockdown of miR-30a in the zebrafish larva results in defective biliary morphogenesis. Several newly identified targets of miR-30a are known regulators of liver development and function. CONCLUSIONS: We have identified miRNAs whose spatial and temporal patterns of expression are suggestive of functional roles in hepatic development and/or function. One of these, the biliary miRNA miR-30a, is required for biliary development in zebrafish. This is the first demonstration of a functional role for miRNA in hepatic organogenesis.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Liver/embryology , Liver/physiology , MicroRNAs/genetics , Adenylate Kinase/genetics , Animals , Autoantigens/genetics , Bile Ducts/embryology , Bile Ducts/physiology , Humans , Mice , Mice, Inbred Strains , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , RNA-Binding Proteins , Vertebrates , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...