Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Hum Mol Genet ; 30(3-4): 182-197, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33517446

ABSTRACT

Lipotoxicity was recently reported in several forms of kidney disease, including focal segmental glomerulosclerosis (FSGS). Susceptibility to FSGS in African Americans is associated with the presence of genetic variants of the Apolipoprotein L1 gene (APOL1) named G1 and G2. If and how endogenous APOL1 may alter mitochondrial function by the modifying cellular lipid metabolism is unknown. Using transgenic mice expressing the APOL1 variants (G0, G1 or G2) under endogenous promoter, we show that APOL1 risk variant expression in transgenic mice does not impair kidney function at baseline. However, APOL1 G1 expression worsens proteinuria and kidney function in mice characterized by the podocyte inducible expression of nuclear factor of activated T-cells (NFAT), which we have found to cause FSGS. APOL1 G1 expression in this FSGS-model also results in increased triglyceride and cholesterol ester contents in kidney cortices, where lipid accumulation correlated with loss of renal function. In vitro, we show that the expression of endogenous APOL1 G1/G2 in human urinary podocytes is associated with increased cellular triglyceride content and is accompanied by mitochondrial dysfunction in the presence of compensatory oxidative phosphorylation (OXPHOS) complexes elevation. Our findings indicate that APOL1 risk variant expression increases the susceptibility to lipid-dependent podocyte injury, ultimately leading to mitochondrial dysfunction.


Subject(s)
Apolipoprotein L1/genetics , Genetic Variation , Glomerulosclerosis, Focal Segmental/metabolism , Lipid Metabolism , Mitochondria/metabolism , Podocytes/metabolism , Black or African American/genetics , Animals , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/physiopathology , Homeostasis , Humans , Mice , Mice, Transgenic , Mitochondria/physiology , Podocytes/physiology , Proteinuria , Triglycerides/metabolism
2.
Dev Biol ; 363(2): 373-87, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22266424

ABSTRACT

The enteric nervous system (ENS) arises from the coordinated migration, expansion and differentiation of vagal and sacral neural crest progenitor cells. During development, vagal neural crest cells enter the foregut and migrate in a rostro-to-caudal direction, colonizing the entire gastrointestinal tract and generating the majority of the ENS. Sacral neural crest contributes to a subset of enteric ganglia in the hindgut, colonizing the colon in a caudal-to-rostral wave. During this process, enteric neural crest-derived progenitors (ENPs) self-renew and begin expressing markers of neural and glial lineages as they populate the intestine. Our earlier work demonstrated that the transcription factor Foxd3 is required early in neural crest-derived progenitors for self-renewal, multipotency and establishment of multiple neural crest-derived cells and structures including the ENS. Here, we describe Foxd3 expression within the fetal and postnatal intestine: Foxd3 was strongly expressed in ENPs as they colonize the gastrointestinal tract and was progressively restricted to enteric glial cells. Using a novel Ednrb-iCre transgene to delete Foxd3 after vagal neural crest cells migrate into the midgut, we demonstrated a late temporal requirement for Foxd3 during ENS development. Lineage labeling of Ednrb-iCre expressing cells in Foxd3 mutant embryos revealed a reduction of ENPs throughout the gut and loss of Ednrb-iCre lineage cells in the distal colon. Although mutant mice were viable, defects in patterning and distribution of ENPs were associated with reduced proliferation and severe reduction of glial cells derived from the Ednrb-iCre lineage. Analyses of ENS-lineage and differentiation in mutant embryos suggested activation of a compensatory population of Foxd3-positive ENPs that did not express the Ednrb-iCre transgene. Our findings highlight the crucial roles played by Foxd3 during ENS development including progenitor proliferation, neural patterning, and glial differentiation and may help delineate distinct molecular programs controlling vagal versus sacral neural crest development.


Subject(s)
Enteric Nervous System/growth & development , Forkhead Transcription Factors/metabolism , Gene Deletion , Intestines/innervation , Neurogenesis , Neuroglia/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism , Animals , Cell Movement , Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Female , Forkhead Transcription Factors/genetics , Intestines/embryology , Intestines/growth & development , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neural Crest/embryology , Repressor Proteins/genetics
3.
J Cell Sci ; 124(Pt 16): 2837-50, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21807948

ABSTRACT

RAD18 is an ubiquitin ligase that is involved in replication damage bypass and DNA double-strand break (DSB) repair processes in mitotic cells. Here, we investigated the testicular phenotype of Rad18-knockdown mice to determine the function of RAD18 in meiosis, and in particular, in the repair of meiotic DSBs induced by the meiosis-specific topoisomerase-like enzyme SPO11. We found that RAD18 is recruited to a specific subfraction of persistent meiotic DSBs. In addition, RAD18 is recruited to the chromatin of the XY chromosome pair, which forms the transcriptionally silent XY body. At the XY body, RAD18 mediates the chromatin association of its interaction partners, the ubiquitin-conjugating enzymes HR6A and HR6B. Moreover, RAD18 was found to regulate the level of dimethylation of histone H3 at Lys4 and maintain meiotic sex chromosome inactivation, in a manner similar to that previously observed for HR6B. Finally, we show that RAD18 and HR6B have a role in the efficient repair of a small subset of meiotic DSBs.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Meiosis , Testis/metabolism , Animals , Chromatin Assembly and Disassembly/genetics , DNA Methylation , DNA Repair/genetics , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Histones/genetics , Histones/metabolism , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Small Interfering/genetics , Testis/pathology , Ubiquitin-Conjugating Enzymes/metabolism , X Chromosome Inactivation/genetics
4.
Nat Genet ; 36(7): 732-7, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15170213

ABSTRACT

Hirschsprung disease (HSCR) is a multigenic, congenital disorder that affects 1 in 5,000 newborns and is characterized by the absence of neural crest-derived enteric ganglia in the colon. One of the primary genes affected in HSCR encodes the G protein-coupled endothelin receptor-B (EDNRB). The expression of Ednrb is required at a defined time period during the migration of the precursors of the enteric nervous system (ENS) into the colon. In this study, we describe a conserved spatiotemporal ENS enhancer of Ednrb. This 1-kb enhancer is activated as the ENS precursors approach the colon, and partial deletion of this enhancer at the endogenous Ednrb locus results in pigmented mice that die postnatally from megacolon. We identified binding sites for SOX10, an SRY-related transcription factor associated with HSCR, in the Ednrb ENS enhancer, and mutational analyses of these sites suggested that SOX10 may have multiple roles in regulating Ednrb in the ENS.


Subject(s)
Gene Expression Regulation/physiology , High Mobility Group Proteins/physiology , Neoplasm Proteins/physiology , Receptors, Endothelin/genetics , Animals , Base Sequence , Enhancer Elements, Genetic , Enteric Nervous System/physiology , Mice , Mice, Transgenic , Molecular Sequence Data , SOXE Transcription Factors , Sequence Homology, Nucleic Acid , Transcription Factors
5.
Mol Pharmacol ; 81(2): 220-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22049154

ABSTRACT

The development of zinc finger nuclease (ZFN) technology has enabled the genetic engineering of the rat genome. The ability to manipulate the rat genome has great promise to augment the utility of rats for biological and pharmacological studies. A Wistar Hannover rat model lacking the multidrug resistance protein Mdr1a P-glycoprotein (P-gp) was generated using a rat Mdr1a-specific ZFN. Mdr1a was completely absent in tissues, including brain and small intestine, of the knockout rat. Pharmacokinetic studies with the Mdr1a P-gp substrates loperamide, indinavir, and talinolol indicated that Mdr1a was functionally inactive in the blood-brain barrier and intestine in Mdr1a(-/-) rats. To identify possible compensatory mechanisms in Mdr1a(-/-) rats, the expression levels of drug-metabolizing enzyme and transporter-related genes were compared in brain, liver, kidney, and intestine of male and female Mdr1a(-/-) and control rats. In general, alterations in gene expression of these genes in Mdr1a(-/-) rats seemed to be modest, with more changes in female than in male rats. Taken together, our studies demonstrate that the ZFN-generated Mdr1a(-/-) rat will be a valuable tool for central nervous system drug target validation and determining the role of P-gp in drug absorption and disposition.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Genetic Engineering/methods , Zinc Fingers/genetics , ATP Binding Cassette Transporter, Subfamily B/deficiency , Animals , Endonucleases , Female , Gene Expression , Genome , Male , Rats , Rats, Transgenic , Rats, Wistar , Tissue Distribution
6.
Am J Physiol Endocrinol Metab ; 302(2): E209-17, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22045313

ABSTRACT

The liver is a crossroad for metabolism of lipid and carbohydrates, with acetyl-CoA serving as an important metabolic intermediate and a precursor for fatty acid and cholesterol biosynthesis pathways. A better understanding of the regulation of these pathways requires an experimental approach that provides both quantitative metabolic flux measurements and mechanistic insight. Under conditions of high carbohydrate availability, excess carbon is converted into free fatty acids and triglyceride for storage, but it is not clear how excessive carbohydrate availability affects cholesterol biosynthesis. To address this, C57BL/6J mice were fed either a low-fat, high-carbohydrate diet or a high-fat, carbohydrate-free diet. At the end of the dietary intervention, the two groups received (2)H(2)O to trace de novo fatty acid and cholesterol synthesis, and livers were collected for gene expression analysis. Expression of lipid and glucose metabolism genes was determined using a custom-designed pathway focused PCR-based gene expression array. The expression analysis showed downregulation of cholesterol biosynthesis genes and upregulation of fatty acid synthesis genes in mice receiving the high-carbohydrate diet compared with the carbohydrate-free diet. In support of these findings, (2)H(2)O tracer data showed that fatty acid synthesis was increased 10-fold and cholesterol synthesis was reduced by 1.6-fold in mice fed the respective diets. In conclusion, by applying gene expression analysis and tracer methodology, we show that fatty acid and cholesterol synthesis are differentially regulated when the carbohydrate intake in mice is altered.


Subject(s)
Cholesterol/biosynthesis , Diet, Carbohydrate-Restricted , Diet, High-Fat , Fatty Acids/biosynthesis , Liver/metabolism , Animals , Cholesterol/genetics , Dietary Carbohydrates/metabolism , Dietary Fats/metabolism , Fatty Acids/genetics , Gene Expression , Gene Expression Profiling , Male , Mice
7.
PLoS One ; 16(6): e0253197, 2021.
Article in English | MEDLINE | ID: mdl-34138902

ABSTRACT

The mechanism of pathogenesis associated with APOL1 polymorphisms and risk for non-diabetic chronic kidney disease (CKD) is not fully understood. Prior studies have minimized a causal role for the circulating APOL1 protein, thus efforts to understand kidney pathogenesis have focused on APOL1 expressed in renal cells. Of the kidney cells reported to express APOL1, the proximal tubule expression patterns are inconsistent in published reports, and whether APOL1 is synthesized by the proximal tubule or possibly APOL1 protein in the blood is filtered and reabsorbed by the proximal tubule remains unclear. Using both protein and mRNA in situ methods, the kidney expression pattern of APOL1 was examined in normal human and APOL1 bacterial artificial chromosome transgenic mice with and without proteinuria. APOL1 protein and mRNA was detected in podocytes and endothelial cells, but not in tubular epithelia. In the setting of proteinuria, plasma APOL1 protein did not appear to be filtered or reabsorbed by the proximal tubule. A side-by-side examination of commercial antibodies used in prior studies suggest the original reports of APOL1 in proximal tubules likely reflects antibody non-specificity. As such, APOL1 expression in podocytes and endothelia should remain the focus for mechanistic studies in the APOL1-mediated kidney diseases.


Subject(s)
Apolipoprotein L1/metabolism , Kidney Tubules, Proximal/metabolism , Proteinuria/metabolism , Alleles , Animals , Apolipoprotein L1/genetics , Endothelial Cells/metabolism , Humans , Kidney , Liver/metabolism , Mice , Mice, Transgenic , Podocytes/metabolism , Proteinuria/genetics
8.
SLAS Discov ; 26(9): 1225-1237, 2021 10.
Article in English | MEDLINE | ID: mdl-34218698

ABSTRACT

High-throughput phenotypic screening is a key driver for the identification of novel chemical matter in drug discovery for challenging targets, especially for those with an unclear mechanism of pathology. For toxic or gain-of-function proteins, small-molecule suppressors are a targeting/therapeutic strategy that has been successfully applied. As with other high-throughput screens, the screening strategy and proper assays are critical for successfully identifying selective suppressors of the target of interest. We executed a small-molecule suppressor screen to identify compounds that specifically reduce apolipoprotein L1 (APOL1) protein levels, a genetically validated target associated with increased risk of chronic kidney disease. To enable this study, we developed homogeneous time-resolved fluorescence (HTRF) assays to measure intracellular APOL1 and apolipoprotein L2 (APOL2) protein levels and miniaturized them to 1536-well format. The APOL1 HTRF assay served as the primary assay, and the APOL2 and a commercially available p53 HTRF assay were applied as counterscreens. Cell viability was also measured with CellTiter-Glo to assess the cytotoxicity of compounds. From a 310,000-compound screening library, we identified 1490 confirmed primary hits with 12 different profiles. One hundred fifty-three hits selectively reduced APOL1 in 786-O, a renal cell adenocarcinoma cell line. Thirty-one of these selective suppressors also reduced APOL1 levels in conditionally immortalized human podocytes. The activity and specificity of seven resynthesized compounds were validated in both 786-O and podocytes.


Subject(s)
Apolipoprotein L1/antagonists & inhibitors , Drug Discovery/methods , High-Throughput Screening Assays , Podocytes/drug effects , Podocytes/metabolism , Humans , Small Molecule Libraries
9.
Nat Genet ; 53(9): 1322-1333, 2021 09.
Article in English | MEDLINE | ID: mdl-34385711

ABSTRACT

The functional interpretation of genome-wide association studies (GWAS) is challenging due to the cell-type-dependent influences of genetic variants. Here, we generated comprehensive maps of expression quantitative trait loci (eQTLs) for 659 microdissected human kidney samples and identified cell-type-eQTLs by mapping interactions between cell type abundances and genotypes. By partitioning heritability using stratified linkage disequilibrium score regression to integrate GWAS with single-cell RNA sequencing and single-nucleus assay for transposase-accessible chromatin with high-throughput sequencing data, we prioritized proximal tubules for kidney function and endothelial cells and distal tubule segments for blood pressure pathogenesis. Bayesian colocalization analysis nominated more than 200 genes for kidney function and hypertension. Our study clarifies the mechanism of commonly used antihypertensive and renal-protective drugs and identifies drug repurposing opportunities for kidney disease.


Subject(s)
Hypertension/genetics , Kidney Tubules, Distal/pathology , Kidney Tubules, Proximal/pathology , Quantitative Trait Loci/genetics , Renal Insufficiency, Chronic/genetics , Base Sequence , Chromosome Mapping , Endothelial Cells/pathology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Genotype , High-Throughput Nucleotide Sequencing , Humans , Polymorphism, Single Nucleotide/genetics , Quantitative Trait, Heritable , Renal Insufficiency, Chronic/pathology , Sequence Analysis, RNA , Single-Cell Analysis
10.
PLoS One ; 14(4): e0211559, 2019.
Article in English | MEDLINE | ID: mdl-30998685

ABSTRACT

Apolipoprotein L1 (APOL1) genetic variants G1 and G2, compared to the common allele G0, are major risk factors for non-diabetic kidney disease in African descent populations. APOL1 is a minor protein component of HDL, as well as being expressed in podocytes and vascular cells. Reverse cholesterol transport involves the transport of cholesterol to HDL by cellular ATP-binding cassette; ABCA1 and ABCG1 with subsequent delivery from peripheral tissues to the liver. With impaired reverse cholesterol transport, lipid accumulation occurs and macrophages morphologically transform into foam cells, releasing inflammatory factors. We asked whether the APOL1 risk variants alter peripheral cholesterol metabolism and specifically affect macrophage cholesterol efflux. Tissues and bone marrow (BM)-derived monocytes were isolated from wild-type mice (WT) and from BAC/APOL1 transgenic (APOL1-G0, APOL1-G1, and APOL1-G2) mice, which carry a bacterial artificial chromosome that contains the human APOL1 genomic region. Monocytes were differentiated into macrophages using M-CSF, and then polarized into M1 and M2 macrophages. Cholesterol content, cholesterol efflux, and ABCA1 and ABCG1 mRNA expression were measured. Kidney, spleen, and bone marrow-derived macrophages from APOL1-G1 and -G2 mice showed increased cholesterol accumulation and decreased ABCA1 and ABCG1 mRNA levels. BM-derived macrophages from APOL1-G1 and -G2 mice showed significantly reduced cholesterol efflux compared to WT or APOL1-G0 macrophages. Taken together, the evidence suggests that APOL1-G1 and -G2 risk variants impaired reverse cholesterol transport through decreased expression of cholesterol efflux transporters suggesting a possible mechanism to promote macrophage foam cell formation, driving inflammation in the glomerulus and renal interstitium.


Subject(s)
Apolipoprotein L1/metabolism , Cholesterol/metabolism , Kidney/metabolism , Macrophages/metabolism , Animals , Apolipoprotein L1/genetics , Biological Transport , Cells, Cultured , Genetic Variation , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Male , Mice , Mice, Transgenic , Spleen/metabolism
11.
Commun Biol ; 1: 188, 2018.
Article in English | MEDLINE | ID: mdl-30417125

ABSTRACT

APOL1 risk alleles associate with chronic kidney disease in African Americans, but the mechanisms remain to be fully understood. We show that APOL1 risk alleles activate protein kinase R (PKR) in cultured cells and transgenic mice. This effect is preserved when a premature stop codon is introduced to APOL1 risk alleles, suggesting that APOL1 RNA but not protein is required for the effect. Podocyte expression of APOL1 risk allele RNA, but not protein, in transgenic mice induces glomerular injury and proteinuria. Structural analysis of the APOL1 RNA shows that the risk variants possess secondary structure serving as a scaffold for tandem PKR binding and activation. These findings provide a mechanism by which APOL1 variants damage podocytes and suggest novel therapeutic strategies.

12.
Cell Metab ; 27(6): 1236-1248.e6, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29706567

ABSTRACT

Diacylglycerol acyltransferase 2 (DGAT2) catalyzes the final step in triglyceride (TG) synthesis and has been shown to play a role in regulating hepatic very-low-density lipoprotein (VLDL) production in rodents. To explore the potential of DGAT2 as a therapeutic target for the treatment of dyslipidemia, we tested the effects of small-molecule inhibitors and gene silencing both in vitro and in vivo. Consistent with prior reports, chronic inhibition of DGAT2 in a murine model of obesity led to correction of multiple lipid parameters. In contrast, experiments in primary human, rhesus, and cynomolgus hepatocytes demonstrated that selective inhibition of DGAT2 has only a modest effect. Acute and chronic inhibition of DGAT2 in rhesus primates recapitulated the in vitro data yielding no significant effects on production of plasma TG or VLDL apolipoprotein B. These results call into question whether selective inhibition of DGAT2 is sufficient for remediation of dyslipidemia.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Dyslipidemias/metabolism , Hepatocytes/metabolism , Obesity/metabolism , Triglycerides/metabolism , Animals , Apolipoproteins B/metabolism , Cells, Cultured , Diacylglycerol O-Acyltransferase/genetics , Disease Models, Animal , Gene Silencing , Humans , Lipoproteins, VLDL/metabolism , Macaca fascicularis , Macaca mulatta , Mice , Mice, Inbred C57BL
13.
Sci Rep ; 7(1): 7272, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28779103

ABSTRACT

In response to various types of injury, melanocyte stem cells (McSCs) located in the bulge of hair follicles can regenerate mature melanocytes for hair and skin pigmentation. How McSCs respond to injury, however, remains largely unknown. Here we show that after epilation of mice, McSCs regenerate follicular and epidermal melanocytes, resulting in skin and hair hyperpigmentation. We further show that epilation leads to endogenous EDN3 upregulation in the dermal papilla, the secondary hair germ cells, and the epidermis. Genetic and pharmacological disruption of the EDN3 receptor EDNRB in vivo significantly blocks the effect of epilation on follicular and epidermal melanocyte regeneration as well as skin and hair hyperpigmentation. Taken together, these results indicate that epilation induces McSCs activation through EDN3/EDNRB signaling and in turn leads to skin and hair hyperpigmentation. The findings suggest that EDN/EDNRB signaling may serve as a potential therapeutic target to promote repigmentation in hypopigmentation disorders.


Subject(s)
Cell Differentiation/genetics , Hair , Melanocytes/cytology , Receptor, Endothelin B/genetics , Regeneration/genetics , Skin Pigmentation , Stem Cells/cytology , Animals , Cell Proliferation , Epidermal Cells/metabolism , Gene Expression , Hair Follicle/cytology , Hair Follicle/metabolism , Hyperpigmentation/genetics , Melanins/metabolism , Mice , Mice, Transgenic
14.
Blood Coagul Fibrinolysis ; 26(8): 893-902, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26192114

ABSTRACT

This report aims at exploring quantitatively the relationship between FXII inhibition and thromboprotection. FXII full and partial null in rats were established via zinc finger nuclease-mediated knockout and siRNA-mediated knockdown, respectively. The rats were subsequently characterized in thrombosis and hemostasis models. Knockout rats exhibited complete thromboprotection in both the arteriovenous shunt model (∼100% clot weight reduction) and the FeCl3-induced arterial thrombosis model (no reduction in blood flow), without any increase in cuticle bleeding time compared with wild-type control rats. Ex-vivo aPTT and the ellagic acid-triggered thrombin generation assay (TGA) exhibited anticoagulant changes. In contrast, ex-vivo PT or high tissue factor-triggered TGA was indistinguishable from control. Rats receiving single doses (0, 0.01, 0.03, 0.1, 0.3, 1 mg/kg) of FXII siRNA exhibited dose-dependent knockdown in liver FXII mRNA and plasma FXII protein (95 and 99%, respectively, at 1 mg/kg) at day 7 post dosing. FXII knockdown was associated with dose-dependent thromboprotection (maximal efficacy achieved with 1 mg/kg in both models) and negligible change in cuticle bleeding times. Ex-vivo TGA triggered with low-level (0.5 µmol/l) ellagic acid tracked best with the knockdown levels and efficacy. Our findings confirm and extend literature reports of an attractive benefit-to-risk profile of targeting FXII for antithrombotic therapies. Titrating of FXII is instructive for its pharmacological inhibition. The knockout rat is valuable for evaluating both mechanism-based safety concerns and off-target effects of FXII(a) inhibitors. Detailed TGA analyses will inform on optimal trigger conditions in studying pharmacodynamic effects of FXII(a) inhibition.


Subject(s)
Endodeoxyribonucleases/genetics , Factor XII/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Thrombolytic Therapy/methods , Thrombosis/therapy , Animals , Arteriovenous Shunt, Surgical , Chlorides/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Ellagic Acid/pharmacology , Endodeoxyribonucleases/metabolism , Factor XII/genetics , Factor XII/metabolism , Ferric Compounds/pharmacology , Gene Knockout Techniques , Hemorrhage/prevention & control , Humans , Liver/metabolism , Male , Partial Thromboplastin Time , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Thrombin/metabolism , Thrombosis/chemically induced , Thrombosis/genetics , Thrombosis/pathology , Zinc Fingers/genetics
15.
Thromb Haemost ; 113(6): 1300-11, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25790442

ABSTRACT

Haemophilia A and B are characterised by a life-long bleeding predisposition, and several lines of evidence suggest that risks of atherothrombotic events may also be reduced. Establishing a direct correlation between coagulation factor levels, thrombotic risks and bleeding propensity has long been hampered by an inability to selectively and specifically inhibit coagulation factor levels. Here, the exquisite selectivity of gene silencing combined with a gene knockout (KO) approach was used to define the relative contribution of factor IX (fIX) to thrombosis and primary haemostasis in the rat. Using a lipid nanoparticle (LNP) formulation, we successfully delivered fIX siRNAs to the liver by intravenous administration. The knockdown (KD) of target gene mRNA was achieved rapidly (within 24 hour post-siRNA dosing), sustained (maintained for at least 7 days post dosing) and not associated with changes in mRNA expression levels of other coagulation factors. We found that intermediate levels of liver fIX mRNA silencing (60-95 %) translating into a 50-99 % reduction of plasma fIX activity provided protection from thrombosis without prolonging the cuticle bleeding time. Over 99 % inhibition of fIX activity was required to observe increase in bleeding, a phenotype confirmed in fIX KO rats. These data provide substantial evidence of a participation of fIX in the mechanisms regulating thrombosis prior to those regulating primary haemostasis, therefore highlighting the potential of fIX as a therapeutic target. In addition, hepatic mRNA silencing using LNP-encapsulated siRNAs may represent a promising novel approach for the chronic treatment and prevention of coagulation-dependent thrombotic disorders in humans.


Subject(s)
Factor IX/genetics , Hemophilia B/genetics , Hemorrhage/genetics , Liver/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNAi Therapeutics , Thrombosis/prevention & control , Animals , Cell Line , Chlorides , Disease Models, Animal , Factor IX/metabolism , Ferric Compounds , Gene Expression Regulation , Genotype , Hemophilia B/blood , Hemorrhage/blood , Hemostasis/genetics , Male , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/genetics , Time Factors , Transfection
16.
PLoS One ; 8(1): e54480, 2013.
Article in English | MEDLINE | ID: mdl-23336002

ABSTRACT

Diacylglycerol acyltransferase-1 (DGAT1) is a potential therapeutic target for treatment of obesity and related metabolic diseases. However, the degree of DGAT1 inhibition required for metabolic benefits is unclear. Here we show that partial DGAT1 deficiency in mice suppressed postprandial triglyceridemia, led to elevations in glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) only following meals with very high lipid content, and did not protect from diet-induced obesity. Maximal DGAT1 inhibition led to enhanced GLP-1 and PYY secretion following meals with physiologically relevant lipid content. Finally, combination of DGAT1 inhibition with dipeptidyl-peptidase-4 (DPP-4) inhibition led to further enhancements in active GLP-1 in mice and dogs. The current study suggests that targeting DGAT1 to enhance postprandial gut hormone secretion requires maximal inhibition, and suggests combination with DPP-4i as a potential strategy to develop DGAT1 inhibitors for treatment of metabolic diseases.


Subject(s)
Diacylglycerol O-Acyltransferase/genetics , Gastrointestinal Hormones/metabolism , Gastrointestinal Tract/metabolism , Postprandial Period , Animals , Base Sequence , Diacylglycerol O-Acyltransferase/deficiency , Diacylglycerol O-Acyltransferase/metabolism , Diet , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Dogs , Enzyme Activation , Female , Gastric Emptying/genetics , Gene Dosage , Gene Expression Regulation , Gene Order , Genotype , Glucagon-Like Peptide 1/metabolism , Lipid Metabolism , Male , Mice , Mice, Knockout , Molecular Sequence Data , Triglycerides/blood
17.
Cancer Res ; 71(8): 3052-65, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21493594

ABSTRACT

PDK1 activates AKT suggesting that PDK1 inhibition might suppress tumor development. However, while PDK1 has been investigated intensively as an oncology target, selective inhibitors suitable for in vivo studies have remained elusive. In this study we present the results of in vivo PDK1 inhibition through a universally applicable RNAi approach for functional drug target validation in oncogenic pathway contexts. This approach, which relies on doxycycline-inducible shRNA expression from the Rosa26 locus, is ideal for functional studies of genes like PDK1 where constitutive mouse models lead to strong developmental phenotypes or embryonic lethality. We achieved more than 90% PDK1 knockdown in vivo, a level sufficient to impact physiological functions resulting in hyperinsulinemia and hyperglycemia. This phenotype was reversible on PDK1 reexpression. Unexpectedly, long-term PDK1 knockdown revealed a lack of potent antitumor efficacy in 3 different mouse models of PTEN-deficient cancer. Thus, despite efficient PDK1 knockdown, inhibition of the PI3K pathway was marginal suggesting that PDK1 was not a rate limiting factor. Ex vivo analysis of pharmacological inhibitors revealed that AKT and mTOR inhibitors undergoing clinical development are more effective than PDK1 inhibitors at blocking activated PI3K pathway signaling. Taken together our findings weaken the widely held expectation that PDK1 represents an appealing oncology target.


Subject(s)
Neoplasms, Experimental/enzymology , PTEN Phosphohydrolase/deficiency , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Gene Knockdown Techniques , Gene Silencing , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Oncogene Protein v-akt/antagonists & inhibitors , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/genetics , Phosphorylation , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference
18.
Methods Enzymol ; 477: 415-27, 2010.
Article in English | MEDLINE | ID: mdl-20699153

ABSTRACT

Expression of small hairpin RNA (shRNA) in mammalian cells can trigger potent RNAi-mediated gene silencing. The dominant-acting RNAi can often result in phenotypes similar to that of a null allele. Moreover, the generation of shRNA knockdown mice and subsequent phenotypic analysis can be achieved in a condensed timeline compared to that of conventional gene-targeting knockout strategies. Here, we discuss methods for the in vivo analysis of gene function in adult mouse tissues following tetracycline-induced RNA knockdown in a single-copy inducible polymerase III promoter-driven shRNA system.


Subject(s)
Gene Silencing/physiology , RNA Interference/physiology , Tetracycline/pharmacology , Animals , Doxycycline/pharmacology , Gene Expression/drug effects , Mice , RNA Interference/drug effects , RNA, Small Interfering/genetics
19.
Dev Biol ; 259(1): 162-75, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12812796

ABSTRACT

Mutations in the genes encoding endothelin receptor-B (Ednrb) and its ligand endothelin-3 (Edn3) affect the development of two neural crest-derived cell types, melanocytes and enteric neurons. EDNRB signaling is exclusively required between E10.5 and E12.5 during the migratory phase of melanoblast and enteric neuroblast development. To determine the fate of Ednrb-expressing cells during this critical period, we generated a strain of mice with the bacterial beta-galactosidase (lacZ) gene inserted downstream of the endogenous Ednrb promoter. The expression of the lacZ gene was detected in melanoblasts and precursors of the enteric neuron system (ENS), as well as other neural crest cells and nonneural crest-derived lineages. By comparing Ednrb(lacZ)/+ and Ednrb(lacZ)/Ednrb(lacZ) embryos, we determined that the Ednrb pathway is not required for the initial specification and dispersal of melanoblasts and ENS precursors from the neural crest progenitors. Rather, the EDNRB-mediated signaling is required for the terminal migration of melanoblasts and ENS precursors, and this pathway is not required for the survival of the migratory cells.


Subject(s)
Enteric Nervous System/growth & development , Melanocytes/physiology , Neural Crest/cytology , Neurons/physiology , Receptors, Endothelin/physiology , Stem Cells/physiology , Animals , Cell Differentiation , Cell Movement , Lac Operon , Mice , Receptor, Endothelin B
20.
Development ; 131(14): 3239-47, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15201217

ABSTRACT

The endothelin receptor B gene (Ednrb) encodes a G-protein-coupled receptor that is expressed in a variety of cell types and is specifically required for the development of neural crest-derived melanocytes and enteric ganglia. In humans, mutations in this gene are associated with Waardenburg-Shah syndrome, a disorder characterized by pigmentation defects, deafness and megacolon. To address the question of whether melanocyte development depends entirely on a cell-autonomous action of Ednrb, we performed a series of tissue recombination experiments in vitro, using neural crest cell cultures from mouse embryos carrying a novel Ednrb-null allele characterized by the insertion of a lacZ marker gene. The results show that Ednrb is not required for the generation of early neural crest-derived melanoblasts but is required for the expression of the differentiation marker tyrosinase. Tyrosinase expression can be rescued, however, by the addition of Ednrb wild-type neural tubes. These Ednrb wild-type neural tubes need not be capable of generating melanocytes themselves, but must be capable of providing KIT ligand, the cognate ligand for the tyrosine kinase receptor KIT. In fact, soluble KIT ligand is sufficient to induce tyrosinase expression in Ednrb-deficient cultures. Nevertheless, these tyrosinase-expressing, Ednrb-deficient cells do not develop to terminally differentiated, pigmented melanocytes. Pigmentation can be induced, however, by treatment with tetradecanoyl phorbol acetate, which mimics EDNRB signaling, but not by treatment with endothelin 1, which stimulates the paralogous receptor EDNRA. The results suggest that Ednrb plays a significant role during melanocyte differentiation and effects melanocyte development by both cell non-autonomous and cell-autonomous signaling mechanisms.


Subject(s)
Melanocytes/cytology , Receptor, Endothelin B/metabolism , Signal Transduction , Alleles , Animals , Cell Differentiation , Cell Division , Cell Lineage , DNA-Binding Proteins/metabolism , Genetic Markers , Genotype , Lac Operon , Melanocytes/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Microphthalmia-Associated Transcription Factor , Models, Biological , Monophenol Monooxygenase/metabolism , Mutation , Neural Crest/cytology , Phorbol Esters/metabolism , Pigmentation , Receptors, G-Protein-Coupled/metabolism , Stem Cell Factor/metabolism , Time Factors , Transcription Factors/metabolism , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL