학술논문

Conditional disruption of hepatic carbamoyl phosphate synthetase 1 in mice results in hyperammonemia without orotic aciduria and can be corrected by liver-directed gene therapy
Document Type
article
Source
Molecular Genetics and Metabolism. 124(4)
Subject
Medical Biotechnology
Biomedical and Clinical Sciences
Nutrition
Gene Therapy
Liver Disease
Biotechnology
Digestive Diseases
Genetics
Neurosciences
Development of treatments and therapeutic interventions
5.2 Cellular and gene therapies
Ammonia
Animals
Carbamoyl-Phosphate Synthase (Ammonia)
Carbamoyl-Phosphate Synthase I Deficiency Disease
Carbamyl Phosphate
Female
Gene Expression Regulation
Enzymologic
Genetic Therapy
Glutamine
Humans
Hyperammonemia
Liver
Male
Mice
Mice
Knockout
Mutation
Orotate Phosphoribosyltransferase
Orotidine-5'-Phosphate Decarboxylase
Purine-Pyrimidine Metabolism
Inborn Errors
Carbamoyl phosphate
synthetase deficiency
Gene therapy
Clinical Sciences
Genetics & Heredity
Clinical sciences
Language
Abstract
Carbamoyl phosphate synthetase 1 (CPS1) is a urea cycle enzyme that forms carbamoyl phosphate from bicarbonate, ammonia and ATP. Bi-allelic mutations of the CPS1 gene result in a urea cycle disorder presenting with hyperammonemia, often with reduced citrulline, and without orotic aciduria. CPS1 deficiency is particularly challenging to treat and lack of early recognition typically results in early neonatal death. Therapeutic interventions have limited efficacy and most patients develop long-term neurologic sequelae. Using transgenic techniques, we generated a conditional Cps1 knockout mouse. By loxP/Cre recombinase technology, deletion of the Cps1 locus was achieved in adult transgenic animals using a Cre recombinase-expressing adeno-associated viral vector. Within four weeks from vector injection, all animals developed hyperammonemia without orotic aciduria and died. Minimal CPS1 protein was detectable in livers. To investigate the efficacy of gene therapy for CPS deficiency following knock-down of hepatic endogenous CPS1 expression, we injected these mice with a helper-dependent adenoviral vector (HDAd) expressing the large murine CPS1 cDNA under control of the phosphoenolpyruvate carboxykinase promoter. Liver-directed HDAd-mediated gene therapy resulted in survival, normalization of plasma ammonia and glutamine, and 13% of normal Cps1 expression. A gender difference in survival suggests that female mice may require higher hepatic CPS1 expression. We conclude that this conditional murine model recapitulates the clinical and biochemical phenotype detected in human patients with CPS1 deficiency and will be useful to investigate ammonia-mediated neurotoxicity and for the development of cell- and gene-based therapeutic approaches.