scholarly journals A mouse model of hereditary coproporphyria identified in an ENU mutagenesis screen

2017 ◽  
Vol 10 (8) ◽  
pp. 1005-1013 ◽  
Author(s):  
Ashlee J. Conway ◽  
Fiona C. Brown ◽  
Robert O. Fullinfaw ◽  
Benjamin T. Kile ◽  
Stephen M. Jane ◽  
...  
BMC Genetics ◽  
2009 ◽  
Vol 10 (1) ◽  
pp. 12 ◽  
Author(s):  
Melissa K Boles ◽  
Bonney M Wilkinson ◽  
Andrea Maxwell ◽  
Lihua Lai ◽  
Alea A Mills ◽  
...  

2007 ◽  
Vol 5 ◽  
pp. P-W-473-P-W-473
Author(s):  
R.J. Westrick ◽  
S.L. Manning ◽  
M.E. Winn ◽  
G.M. Stotz ◽  
E. Sanford ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2-2 ◽  
Author(s):  
Randal J. Westrick ◽  
Sara L. Manning ◽  
Sarah L. Dobies ◽  
Abigail L. Peterson ◽  
David R. Siemieniak ◽  
...  

Abstract Factor V Leiden, (FVL) is the most common known inherited thrombotic risk factor and is present in approximately 5% of most Western populations and 25–50% of patients presenting with venous thrombosis. However, FVL is incompletely penetrant, with only approximately 10% of FVL carriers developing thrombosis in their lifetimes. Though interactions between FVL and other known prothrombotic mutations have been documented in a few cases, the genetic factors responsible for the incomplete penetrance of FVL remain largely unknown. We previously reported a remarkable synthetic lethality in mice carrying the FVL mutation and partial deficiency of a key coagulation component, tissue factor pathway inhibitor (TFPI). Complete TFPI deficiency in mice is embryonic lethal, whereas heterozygosity is compatible with normal survival. However, homozygosity for FVL (FvQ/Q) in the context of heterozygosity for TFPI (Tfpi+/−) is uniformly lethal due to disseminated perinatal thrombosis. In order to identify potential modifier genes contributing to FVL penetrance, we have utilized this lethal genetic interaction as a phenotyping tool for a sensitized ENU mutagenesis screen in laboratory mice. We hypothesize that dominant mutations in key components of the coagulation system will improve hemostatic balance and allow survival in mice carrying the lethal FvQ/Q Tfpi+/− genotype combination. As an example, we propose that loss of one tissue factor allele might compensate for reduced TFPI and rescue FvQ/Q Tfpi+/− . To test this hypothesis, we bred tissue factor heterozygous mice (Tf+/−) with FvQ/Q Tfpi+/− mice and observed complete rescue, with normal survival and the expected number (8 of 57) of FvQ/Q Tfpi+/− Tf+/− mice from a FvQ/+ Tfpi+/− Tf+/−x FvQ/Q cross. In order to identify candidate modifier genes, we performed a whole genome mutagenesis screen. In this screen, male FvQ/Q mice were mutagenized with ENU and bred to FvQ/+ Tfpi+/− double heterozygous females. DNAs from surviving offspring were PCR assayed to identify rescued mice with the FvQ/Q Tfpi+/− genotype. Analysis of 2250 offspring, corresponding to approximately half genome coverage, has identified 15 mice that survived to weaning. Heritability was demonstrated for the 5 mutant lines subjected to progeny testing to date. Genetic crosses are in progress to map the mutant genes in 3 of the 5 progeny tested lines. These preliminary results demonstrate the feasibility of this sensitized screen for the identification of dominant suppressors of thrombosis. Based on our data, we estimate that there are likely 10–20 mammalian genes for which a <50% reduction in expression could result in a major shift in hemostatic balance sufficient to rescue the lethal thrombosis associated with the FvQ/Q Tfpi+/− lethal genotype. Each of these loci represent a candidate for a human modifier gene in patients with FVL and other thrombophilic mutations. Finally, the biologic pathways uncovered by these studies should provide new insights into the overall regulation of hemostatic balance and identify potential new targets for therapeutic intervention.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 493-493
Author(s):  
Randal Joseph Westrick ◽  
Guojing Zhu ◽  
Kart Tomberg ◽  
David R. Siemieniak ◽  
Sara A. Haynes ◽  
...  

Abstract Abstract 493 Only ∼10% of individuals carrying the common risk factor, Factor V Leiden (FVL), will develop venous thrombosis. In order to identify potential FVL modifier genes, we performed a sensitized dominant ENU mutagenesis screen based on the perinatal synthetic lethal thrombosis observed in mice homozygous for FVL (FVQ/Q) and hemizygous for tissue factor pathway inhibitor deficiency (Tfpi+/−). The screen was performed by crossing ENU-treated male FVQ/Q mice with FVQ/+ Tfpi+/− females. Surviving G1 offspring were analyzed to identify survivors with the lethal FVQ/Q Tfpi+/− genotype. Analysis of 7,128 G1 offspring (∼2X genome coverage) identified 98 FVQ/Q Tfpi+/− mice that survived to weaning. Fourteen FVQ/Q Tfpi+/− G1 mice exhibited successful transmission of a putative suppressor mutation to two or more FVQ/Q Tfpi+/− G2 offspring. Whole exome sequencing was performed on a progeny tested member of 8 of the 14 lines using the Agilent SureSelect mouse whole exome capture kit resulting in ∼100 fold coverage. Variant analysis revealed a small number of high confidence novel heterozygous (dominant) single nucleotide variants (SNVs) in each sample. Sanger re-sequencing of all 11 SNVs in a cohort of mice from line 1 confirmed that the G to C mutation (chromosome 11 base 19,977,300) in the Actr2 gene (present in 15/16 re-sequenced progeny p<0.0001) is the dominant FVL modifier in this line. This mutation resulted in an R286G substitution in a highly conserved amino acid in the Arp2 protein, which is essential for the function of the Arp2/3 complex. Arp2/3 is responsible for intracellular actin branching and polymerization, which is critical for the regulation of cell shape. Analysis of 31 progeny from an Arp2 R/G × Arp2 R/G cross revealed only one live Arp2 G/G mouse (p<0.05), suggesting that R286G is a loss of function mutation and that homozygous deficiency is lethal. Complete blood counts (Advia 2120) performed on 14 Actr2 heterozygous and 10 wildtype littermates revealed no significant differences in platelet count, red and white blood cell counts, hematocrit or hemoglobin. However, measurements of platelet size and size distribution including mean platelet dry mass, platelet volume distribution width and the platelet component distribution width were significantly altered in Actr2 heterozygous mutant mice (p<0.05 for each measure). Thus, partial deficiency of Arp2 appears to alter platelet structure/function resulting in a shift in hemostatic balance facilitating survival of the otherwise lethal FVQ/Q Tfpi+/− phenotype. These results suggest that variation in Arp2 or related genes could potentially modify thrombosis risk in humans, and might also identify novel therapeutic targets for the treatment of this class of disorders. Disclosures: Ginsburg: Shire Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Portola Pharmaceuticals: Consultancy; Catalyst Biosciences: Consultancy; Baxter Pharmaceuticals: benefit from payments to Children's Hosptial, Boston, and the University of Michigan Patents & Royalties; Merck Pharmaceuticals: Consultancy.


2007 ◽  
Vol 38 (2) ◽  
pp. 174-175
Author(s):  
Gerhard Rank ◽  
Rosemary Sutton ◽  
Ben Kile ◽  
Simon Foote ◽  
Jacinta Caddy ◽  
...  

2013 ◽  
Vol 14 (9) ◽  
pp. R96 ◽  
Author(s):  
Lucia Daxinger ◽  
Sarah K Harten ◽  
Harald Oey ◽  
Trevor Epp ◽  
Luke Isbel ◽  
...  

2018 ◽  
Vol 11 (5) ◽  
pp. dmm034678 ◽  
Author(s):  
Ashlee J. Conway ◽  
Fiona C. Brown ◽  
Elinor J. Hortle ◽  
Gaetan Burgio ◽  
Simon J. Foote ◽  
...  

2008 ◽  
Vol 19 (2) ◽  
pp. 77-84 ◽  
Author(s):  
Cornelius R. Pawlak ◽  
Carles Sanchis-Segura ◽  
Dian Soewarto ◽  
Sibylle Wagner ◽  
Martin Hrabé de Angelis ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document