Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Publication year range
1.
Hepatology ; 76(3): 646-659, 2022 09.
Article in English | MEDLINE | ID: mdl-34786702

ABSTRACT

BACKGROUND AND AIMS: Patient-derived human-induced pluripotent stem cells (hiPSCs) differentiated into hepatocytes (hiPSC-Heps) have facilitated the study of rare genetic liver diseases. Here, we aimed to establish an in vitro liver disease model of the urea cycle disorder ornithine transcarbamylase deficiency (OTCD) using patient-derived hiPSC-Heps. APPROACH AND RESULTS: Before modeling OTCD, we addressed the question of why hiPSC-Heps generally secrete less urea than adult primary human hepatocytes (PHHs). Because hiPSC-Heps are not completely differentiated and maintain some characteristics of fetal PHHs, we compared gene-expression levels in human fetal and adult liver tissue to identify genes responsible for reduced urea secretion in hiPSC-Heps. We found lack of aquaporin 9 (AQP9) expression in fetal liver tissue as well as in hiPSC-Heps, and showed that forced expression of AQP9 in hiPSC-Heps restores urea secretion and normalizes the response to ammonia challenge by increasing ureagenesis. Furthermore, we proved functional ureagenesis by challenging AQP9-expressing hiPSC-Heps with ammonium chloride labeled with the stable isotope [15 N] (15 NH4 Cl) and by assessing enrichment of [15 N]-labeled urea. Finally, using hiPSC-Heps derived from patients with OTCD, we generated a liver disease model that recapitulates the hepatic manifestation of the human disease. Restoring OTC expression-together with AQP9-was effective in fully correcting OTC activity and normalizing ureagenesis as assessed by 15 NH4 Cl stable-isotope challenge. CONCLUSION: Our results identify a critical role for AQP9 in functional urea metabolism and establish the feasibility of in vitro modeling of OTCD with hiPSC-Heps. By facilitating studies of OTCD genotype/phenotype correlation and drug screens, our model has potential for improving the therapy of OTCD.


Subject(s)
Aquaporins/metabolism , Induced Pluripotent Stem Cells , Liver Diseases , Ornithine Carbamoyltransferase Deficiency Disease , Adult , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Liver Diseases/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/therapy , Urea
2.
J Inherit Metab Dis ; 46(1): 55-65, 2023 01.
Article in English | MEDLINE | ID: mdl-36220785

ABSTRACT

Protein catabolism ultimately yields toxic ammonia, which must be converted to urea by the liver for renal excretion. In extrahepatic tissues, ammonia is temporarily converted primarily to glutamine for subsequent hepatic extraction. Urea cycle disorders (UCDs) are inborn errors of metabolism causing impaired ureagenesis, leading to neurotoxic accumulation of ammonia and brain glutamine. Treatment includes dietary protein restriction and oral "ammonia scavengers." These scavengers chemically combine with glutamine and glycine to yield excretable products, creating an alternate pathway of waste nitrogen disposal. The amino acid transporter SLC6A19 is responsible for >95% of absorption and reabsorption of free neutral amino acids in the small intestine and kidney, respectively. Genetic SLC6A19 deficiency causes massive neutral aminoaciduria but is typically benign. We hypothesized that inhibiting SLC6A19 would open a novel and effective alternate pathway of waste nitrogen disposal. To test this, we crossed SLC6A19 knockout (KO) mice with spfash mice, a model of ornithine transcarbamylase (OTC) deficiency. Loss of SLC6A19 in spfash mice normalized plasma ammonia and brain glutamine and increased median survival in response to a high protein diet from 7 to 97 days. While induced excretion of amino acid nitrogen is likely the primary therapeutic mechanism, reduced intestinal absorption of dietary free amino acids, and decreased muscle protein turnover due to loss of SLC6A19 may also play a role. In summary, the results suggest that SLC6A19 inhibition represents a promising approach to treating UCDs and related aminoacidopathies.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Amino Acid Transport Systems, Neutral , Ornithine Carbamoyltransferase Deficiency Disease , Mice , Animals , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Glutamine , Nitrogen/metabolism , Ammonia , Disease Models, Animal , Mice, Knockout , Urea/metabolism , Ornithine Carbamoyltransferase/genetics , Amino Acid Transport Systems, Neutral/genetics
3.
J Inherit Metab Dis ; 45(3): 470-480, 2022 05.
Article in English | MEDLINE | ID: mdl-34988999

ABSTRACT

Amino acids, the building blocks of proteins in the cells and tissues, are of fundamental importance for cell survival, maintenance, and proliferation. The liver plays a critical role in amino acid metabolism and detoxication of byproducts such as ammonia. Urea cycle disorders with hyperammonemia remain difficult to treat and eventually necessitate liver transplantation. In this study, ornithine transcarbamylase deficient (Otcspf-ash ) mouse model was used to test whether knockdown of a key glutamine metabolism enzyme glutaminase 2 (GLS2, gene name: Gls2) or glutamate dehydrogenase 1 (GLUD1, gene name: Glud1) could rescue the hyperammonemia and associated lethality induced by a high protein diet. We found that reduced hepatic expression of Gls2 but not Glud1 by AAV8-mediated delivery of a short hairpin RNA in Otcspf-ash mice diminished hyperammonemia and reduced lethality. Knockdown of Gls2 but not Glud1 in Otcspf-ash mice exhibited reduced body weight loss and increased plasma glutamine concentration. These data suggest that Gls2 hepatic knockdown could potentially help alleviate risk for hyperammonemia and other clinical manifestations of patients suffering from defects in the urea cycle.


Subject(s)
Glutaminase/metabolism , Hyperammonemia , Ornithine Carbamoyltransferase Deficiency Disease , Urea Cycle Disorders, Inborn , Ammonia , Animals , Disease Models, Animal , Glutaminase/genetics , Glutamine/metabolism , Humans , Hyperammonemia/metabolism , Liver/metabolism , Mice , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Urea/metabolism , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/metabolism
4.
BMC Gastroenterol ; 22(1): 144, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35346058

ABSTRACT

BACKGROUND: Ornithine transcarbamylase deficiency (OTCD) is most common among urea cycle disorders (UCDs), defined by defects in enzymes associated with ureagenesis. Corticosteroid administration to UCD patients, including OTCD patients, is suggested to be avoided, as it may induce life-threatening hyperammonemia. The mechanism has been considered nitrogen overload due to the catabolic effect of corticosteroids; however, the pathophysiological process is unclear. METHODS: To elucidate the mechanism of hyperammonemia induced by corticosteroid administration in OTCD patients, we analyzed a mouse model by administering corticosteroids to OTCspf-ash mice deficient in the OTC gene. Dexamethasone (DEX; 20 mg/kg) was administered to the OTCspf-ash and wild-type (WT) mice at 0 and 24 h, and the serum ammonia concentrations, the levels of the hepatic metabolites, and the gene expressions related with ammonia metabolism in the livers and muscles were analyzed. RESULTS: The ammonia levels in Otcspf-ash mice that were administered DEX tended to increase at 24 h and increased significantly at 48 h. The metabolomic analysis showed that the levels of citrulline, arginine, and ornithine did not differ significantly between Otcspf-ash mice that were administered DEX and normal saline; however, the level of aspartate was increased drastically in Otcspf-ash mice owing to DEX administration (P < 0.01). Among the enzymes associated with the urea cycle, mRNA expressions of carbamoyl-phosphate synthase 1, ornithine transcarbamylase, arginosuccinate synthase 1, and arginosuccinate lyase in the livers were significantly downregulated by DEX administration in both the Otcspf-ash and WT mice (P < 0.01). Among the enzymes associated with catabolism, mRNA expression of Muscle RING-finger protein-1 in the muscles was significantly upregulated in the muscles of WT mice by DEX administration (P < 0.05). CONCLUSIONS: We elucidated that corticosteroid administration induced hyperammonemia in Otcspf-ash mice by not only muscle catabolism but also suppressing urea-cycle-related gene expressions. Since the urea cycle intermediate amino acids, such as arginine, might not be effective because of the suppressed expression of urea-cycle-related genes by corticosteroid administration, we should consider an early intervention by renal replacement therapy in cases of UCD patients induced by corticosteroids to avoid brain injuries or fatal outcomes.


Subject(s)
Ornithine Carbamoyltransferase Deficiency Disease , Adrenal Cortex Hormones , Animals , Citrulline/genetics , Gene Expression , Humans , Mice , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Urea/metabolism
5.
Exp Cell Res ; 384(1): 111621, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31513782

ABSTRACT

A long-term hepatocyte culture maintaining liver-specific functions is very essential for both basic research and the development of bioartificial liver devices in clinical application. However, primary hepatocytes rapidly lose their proliferation and hepatic functions over a few days in culture. This work is to establish an ornithine transcarbamylase deficiency (OTCD) patient-derived primary human hepatocyte (OTCD-PHH) culture with hepatic functions for providing an in vitro cell model. Liver tissue from an infant with OTCD was dispersed into single cells. The cells were cultured using conditional reprogramming. To characterize the cells, we assessed activities and mRNA expression of CYP3A4, 1A1, 2C9, as well as albumin and urea secretion. We found that the OTCD-PHH can be subpassaged for more than 15 passages. The cells do not express mRNA of fibroblast-specific maker, whereas they highly express markers of epithelial cells and hepatocytes. In addition, the OTCD-PHH retain native CYP3A4, 1A1, 2C9 activities and albumin secretion function at early passages. The OTCD-PHH at passages 2, 6, 9 and 13 have identical DNA fingerprint as the original tissue. Furthermore, under 3D culture environment, low urea production and hepatocyte marker staining of the OTCD-PHH were detected. The established OTCD-PHH maintain liver-specific functions at early passages and can be long-term cultured in vitro. We believe the established long-term OTCD-PHH culture is highly relevant to study liver diseases, particularly in infants with OTCD.


Subject(s)
Hepatocytes/pathology , Liver Diseases/pathology , Liver/pathology , Ornithine Carbamoyltransferase Deficiency Disease/pathology , 3T3 Cells , Animals , Cell Line , Cell Line, Tumor , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP3A/metabolism , Epithelial Cells/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Infant , Liver/metabolism , Liver Diseases/metabolism , Male , Mice , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , RNA, Messenger/metabolism
6.
J Inherit Metab Dis ; 42(1): 93-106, 2019 01.
Article in English | MEDLINE | ID: mdl-30740724

ABSTRACT

BACKGROUND: To improve our understanding of urea cycle disorders (UCDs) prospectively followed by two North American (NA) and European (EU) patient cohorts. AIMS: Description of the NA and EU patient samples and investigation of the prospects of combined and comparative analyses for individuals with UCDs. METHODS: Retrieval and comparison of the data from 1095 individuals (NA: 620, EU: 475) from two electronic databases. RESULTS: The proportion of females with ornithine transcarbamylase deficiency (fOTC-D), particularly those being asymptomatic (asfOTC-D), was higher in the NA than in the EU sample. Exclusion of asfOTC-D resulted in similar distributions in both samples. The mean age at first symptoms was higher in NA than in EU patients with late onset (LO), but similar for those with early (≤ 28 days) onset (EO) of symptoms. Also, the mean age at diagnosis and diagnostic delay for EO and LO patients were similar in the NA and EU cohorts. In most patients (including fOTC-D), diagnosis was made after the onset of symptoms (59.9%) or by high-risk family screening (24.7%), and less often by newborn screening (8.9%) and prenatal testing (3.7%). Analysis of clinical phenotypes revealed that EO patients presented with more symptoms than LO individuals, but that numbers of symptoms correlated with plasma ammonium concentrations in EO patients only. Liver transplantation was reported for 90 NA and 25 EU patients. CONCLUSIONS: Combined analysis of databases drawn from distinct populations opens the possibility to increase sample sizes for natural history questions, while comparative analysis utilizing differences in approach to treatment can evaluate therapeutic options and enhance long-term outcome studies.


Subject(s)
Urea Cycle Disorders, Inborn/diagnosis , Cohort Studies , Data Analysis , Delayed Diagnosis , Europe , Female , Humans , Infant, Newborn , Male , Neonatal Screening/methods , North America , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Rare Diseases , Urea/metabolism , Urea Cycle Disorders, Inborn/metabolism
7.
J Inherit Metab Dis ; 42(6): 1064-1076, 2019 11.
Article in English | MEDLINE | ID: mdl-30714172

ABSTRACT

The most common ureagenesis defect is X-linked ornithine transcarbamylase (OTC) deficiency which is a main target for novel therapeutic interventions. The spf ash mouse model carries a variant (c.386G>A, p.Arg129His) that is also found in patients. Male spf ash mice have a mild biochemical phenotype with low OTC activity (5%-10% of wild-type), resulting in elevated urinary orotic acid but no hyperammonemia. We recently established a dried blood spot method for in vivo quantification of ureagenesis by Gas chromatography-mass spectrometry (GC-MS) using stable isotopes. Here, we applied this assay to wild-type and spf ash mice to assess ureagenesis at different ages. Unexpectedly, we found an age-dependency with a higher capacity for ammonia detoxification in young mice after weaning. A parallel pattern was observed for carbamoylphosphate synthetase 1 and OTC enzyme expression and activities, which may act as pacemaker of this ammonia detoxification pathway. Moreover, high ureagenesis in younger mice was accompanied by elevated periportal expression of hepatic glutamine synthetase, another main enzyme required for ammonia detoxification. These observations led us to perform a more extensive analysis of the spf ash mouse in comparison to the wild-type, including characterization of the corresponding metabolites, enzyme activities in the liver and plasma and the gut microbiota. In conclusion, the comprehensive enzymatic and metabolic analysis of ureagenesis performed in the presented depth was only possible in animals. Our findings suggest such analyses being essential when using the mouse as a model and revealed age-dependent activity of ammonia detoxification.


Subject(s)
Aging/physiology , Ammonia/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/pathology , Ornithine Carbamoyltransferase/genetics , Urea/metabolism , Age Factors , Animals , Disease Models, Animal , Humans , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/pathology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Transgenic , Ornithine Carbamoyltransferase Deficiency Disease/genetics
8.
Mol Ther ; 26(3): 801-813, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29433939

ABSTRACT

We describe a novel, two-nanoparticle mRNA delivery system and show that it is highly effective as a means of intracellular enzyme replacement therapy (i-ERT) using a murine model of ornithine transcarbamylase deficiency (OTCD). Our Hybrid mRNA Technology delivery system (HMT) comprises an inert lipid nanoparticle that protects the mRNA from nucleases in the blood as it distributes to the liver and a polymer micelle that targets hepatocytes and triggers endosomal release of mRNA. This results in high-level synthesis of the desired protein specifically in the liver. HMT delivery of human OTC mRNA normalizes plasma ammonia and urinary orotic acid levels, and leads to a prolonged survival benefit in the murine OTCD model. HMT represents a unique, non-viral mRNA delivery method that allows multi-dose, systemic administration for treatment of single-gene inherited metabolic diseases.


Subject(s)
Genetic Therapy , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase/genetics , RNA, Messenger/genetics , Animals , Disease Models, Animal , Genetic Therapy/methods , Male , Metabolic Networks and Pathways , Mice , Mice, Knockout , Micelles , Nanoparticles , Nanotechnology , Ornithine Carbamoyltransferase/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/therapy , Polymers , RNA, Messenger/administration & dosage , RNA, Small Interfering/genetics , Urea/metabolism
9.
BMC Med Genet ; 19(1): 210, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30541480

ABSTRACT

BACKGROUND: Ornithine transcarbamylase deficiency (OTCD) is an X-linked recessive disorder involving a defect in the urea cycle caused by OTC gene mutations. Although a total of 417 disease-causing mutations in OTC have been reported, structural abnormalities in this gene are rare. We here describe a female OTCD case caused by an exonic duplication of the OTC gene (exons 1-6). CASE PRESENTATION: A 23-year-old woman with late-onset OTCD diagnosed by biochemical testing was subjected to subsequent genetic testing. Sanger sequencing revealed no pathogenic mutation throughout the coding exons of the OTC gene, but multiplex ligation-dependent probe amplification (MLPA) revealed duplication of exons 1-6. Further genetic analyses revealed an inversion of duplicated exon 1 and a tandem duplication of exons 2-6. Each of the junctions of the inversion harbored a microhomology and non-templated microinsertion, respectively, suggesting a replication-based mechanism. The duplication was also of de novo origin but segregation analysis indicated that it took place in the paternal chromosome. CONCLUSION: We report the first OTCD case harboring an exonic duplication in the OTC gene. The functional defects caused by this anomaly were determined via structural analysis of its complex rearrangements.


Subject(s)
Chromosomes, Human, X/chemistry , Exons , Gene Duplication , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase/genetics , Age of Onset , Base Sequence , Female , Gene Expression , Genes, Recessive , Humans , Multiplex Polymerase Chain Reaction , Ornithine Carbamoyltransferase/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/physiopathology , Paternal Inheritance , Translocation, Genetic , Young Adult
10.
J Inherit Metab Dis ; 39(2): 231-41, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26310964

ABSTRACT

BACKGROUND: Organic acidurias (OADs) and urea cycle disorders (UCDs) are inborn metabolic disorders with a risk for acute and chronic metabolic decompensation resulting in impairments of the central nervous system and other organ systems. So far, there is no systematic study of intellectual functioning, behavioural/emotional problems and health-related quality of life (HRQoL), and how these domains are connected. METHODS: Data of 152 patients with OADs (n = 100) and UCDs (n = 52) from the European Registry and Network of intoxication type Metabolic Diseases (E-IMD) using standardized instruments were compared with normative data. RESULTS: Behavioural/emotional problems are increased in OADs or UCDs patients by a factor of 2.5 (3.0), in female asymptomatic carriers of X-linked inherited UCD ornithine transcarbamylase deficiency (fasOTCD) by a factor of 1.5. All groups show similar patterns of behavioural/emotional problems, not different from epidemiological data. Mental disability (IQ ≤ 70) was found in 31% of OAD, 43% of UCD, but not in fasOTCD subjects. HRQoL was decreased in the physical domain, but in the normal range. Behavioural/emotional problems were significantly associated with intellectual functioning (OR = 6.24, 95%CI: 1.39-27.99), but HRQoL was independent from both variables. CONCLUSIONS: Patients with OADs and UCDs show increased frequencies of mental disability and behavioural/emotional problems. Profiles of behavioural/emotional problems were similar to epidemiological data. Intellectual disability and behavioural/emotional problems were strongly associated. Patients' HRQoL was in the normal range, possibly compensated by coping strategies of their families. Diagnostics and clinical care of OAD/UCD patients should be improved regarding behavioural/emotional, intellectual and quality of life aspects.


Subject(s)
Intellectual Disability/etiology , Mental Disorders/etiology , Metabolic Diseases/complications , Metabolism, Inborn Errors/complications , Urea Cycle Disorders, Inborn/complications , Child, Preschool , Emotions , Female , Humans , Intellectual Disability/metabolism , Male , Mental Disorders/metabolism , Metabolic Diseases/metabolism , Metabolism, Inborn Errors/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/complications , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Quality of Life , Urea Cycle Disorders, Inborn/metabolism
11.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 31(2): 148-51, 2014 Apr.
Article in Zh | MEDLINE | ID: mdl-24711021

ABSTRACT

OBJECTIVE: To analyze the clinical features, metabolic profiling and gene mutations of patients with ornithine transcarbamylase deficiency (OTCD) and explore the molecular pathogenesis of OTCD in order to provide a solution for molecular diagnostics and genetic counseling. METHODS: Clinical data of 3 neonates were analyzed. The amino acids level in blood was analyzed with mass spectrum technology. PCR was used to amplify all the 10 exons of OTC gene. The PCR products were directly sequenced to detect the mutations. RESULTS: All of the 3 cases had neonatal onset and showed poor reaction, feeding difficulty, convulsion and neonatal infection. Citrulline levels were significantly decreased. Case 1 had a missense mutation of Y183C. Case 2 showed a missense mutation of V339G in exon 10. And a missense mutations of W332S in exon 9 was detected in case 3. CONCLUSION: Analysis of OTC gene sequences can be used for the diagnosis of OTCD and screening of asymptomatic carriers. Mutation analysis is important for prenatal diagnosis of individuals with a positive family history and genetic counseling. The V339G and W332S mutations have been discovered for the first time. Patients with such mutations may have onset of the disease during neonatal period.


Subject(s)
Mutation , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase/genetics , Humans , Male , Ornithine Carbamoyltransferase Deficiency Disease/metabolism
12.
Mol Genet Metab ; 109(3): 251-4, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23669167

ABSTRACT

Urea cycle deficient patients with prominent hyperammonemic often exhibit abnormal production of nitric oxide (NO), which reduces vascular tone, along with amino acid abnormalities. However, information related to the metabolic changes in heterozygotes of ornithine transcarbamylase deficiency (OTCD) lacking overt hyperammonemia is quite limited. We examined vascular mediators and amino acids in non-hyperammonemic heterozygotes. Twenty-four heterozygous OTCD adult females without hyperammonemic bouts, defined as non-hyperammonemic carriers, were enrolled. We measured blood amino acids constituting urea cycle and nitric oxide (NO) cycle. Blood concentrations of nitrate/nitrite (NOx) as stable NO-metabolites, asymmetric dimethylarginine (ADMA) inhibiting NO synthesis, and endothelin-1 (ET-1) raising vascular tone were also determined. NOx concentrations were significantly lower in non-hyperammonemic carriers (p < 0.01). However, ADMA and ET-1 levels in this group were comparable to those in the age-matched control group. Arginine and citrulline levels were also significantly lower in non-hyperammonemic carriers than in controls (p < 0.01). Of the 24 non-hyperammonemic carriers, 10 often developed headaches. Their daily NOx and arginine levels were significantly lower than those in headache-free carriers (p < 0.05). In three carriers receiving oral l-arginine, blood NOx concentrations were significantly higher. In two of those three, the occurrence of headaches was decreased. These results suggest that NO cycle coupling with the urea cycle is altered substantially even in non-hyperammonemic OTCD carriers, predisposing them to headaches.


Subject(s)
Heterozygote , Nitric Oxide/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Urea/metabolism , Amino Acids/blood , Ammonia/blood , Child, Preschool , Endothelin-1/blood , Female , Humans , Infant , Infant, Newborn , Male , Metabolic Networks and Pathways
13.
Mol Genet Metab ; 108(3): 161-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23403242

ABSTRACT

BACKGROUND: Urea cycle disorders (UCD) are caused by genetic defects in enzymes that constitute the hepatic ammonia detoxification pathway. Patients may present with variable clinical manifestations and with hyperammonaemia. Liver abnormalities have been associated with UCD, but only a few reports on the histopathological findings in the liver of UCD patients have been published. METHODS: We conducted a retrospective review of liver biopsies, ex-planted livers and livers at post-mortem of patients with UCD. A single pathologist reviewed all specimens. RESULTS: There were 18 liver samples from 13 patients with confirmed UCD: four ex-planted livers from patients with Ornithine Transcarbamylase (OTC) (n=3) and Carbamoyl Phosphate Synthetase 1 (CPS 1) (n=1) deficiencies, eight post-mortem samples from patients with CPS 1 (n=2), OTC (n=4), Argininosuccinate Synthetase (ASS) (n=1) and Argininosuccinate Lyase (ASL) (n=1) deficiencies, and six liver biopsies, three of which came from one patient with ASL deficiency. The other three liver biopsies were from patients who subsequently received liver transplantation. Histopathological findings in samples from neonates were non-specific. Samples from three late onset OTC deficient and one ASL deficient patients showed thin fibrous septa with portal to portal bridging fibrosis and focal marked enlargement and pallor of the hepatocytes due to accumulation of glycogen particles, resembling glycogenosis and resulting in a prominent nodular pattern. Serial liver biopsies in four UCD patients with interval between samples ranging from 1 year 2 months to 17 years showed progression in fibrosis in one OTC and one ASL deficient patients. Moderate fatty changes to no progression in liver disease were noted in the two patients (OTC=1 and CPS=1). A variety of non-specific features such as fatty change, mild inflammation, cholestasis and focal necrosis were seen in the other UCD patients. CONCLUSIONS: Histopathological changes in livers from neonates with UCD are non-specific. Older patients with UCD seem to show variable hepatic fibrosis compared to those who died early. Some of these patients also show focal and superficial resemblance to a glycogen storage disorder and cirrhosis. However, progression of these changes seems to be slow. To clarify the long term consequence of these changes, more extensive periods of follow up in a larger population series is needed.


Subject(s)
Argininosuccinate Synthase/deficiency , Argininosuccinic Aciduria/pathology , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Hepatocytes/pathology , Hyperammonemia/pathology , Liver/pathology , Ornithine Carbamoyltransferase Deficiency Disease/pathology , Argininosuccinic Aciduria/complications , Argininosuccinic Aciduria/metabolism , Autopsy , Biopsy , Child , Child, Preschool , Fatty Acids/metabolism , Female , Hepatocytes/metabolism , Histocytochemistry , Humans , Hyperammonemia/complications , Hyperammonemia/metabolism , Infant , Infant, Newborn , Liver/metabolism , Liver Transplantation , Male , Ornithine Carbamoyltransferase Deficiency Disease/complications , Ornithine Carbamoyltransferase Deficiency Disease/metabolism
14.
Am J Emerg Med ; 31(9): 1420.e5-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23790482

ABSTRACT

Ornithine transcarbamylase (OTC) deficiency is a genetic disorder involving a mutation of the ornithine transcarbamylase gene, located on the short arm of the X chromosome (Xp21.1). This makes the expression of the gene most common in homozygous males, but heterozygous females can also be affected and may be more likely to suffer from serious morbidity. Most males present early in the neonatal period with more devastating outcomes than their female counterparts. Up to 34% will present in the first 30 days of life (J Pediatr 2001;138:S30). Females often have partially functioning mitochondria due to uneven distribution of the mutant gene secondary to lyonization ("X-chromosome Inactivation". Genetics Home Reference, 2012). Occasionally, symptomatic females may not even present until they are placed under metabolic stress such as a severe illness, fasting, pregnancy, or new medication (Roth KS, Steiner RD. "Ornithine Transcarbamylase Deficiency". EMedicine, 2012). The urea cycle is the body's primary tool for the disposal of excess nitrogen, which is generated by the routine metabolism of proteins and amino acids. Mitochondrial dysfunction impairs urea production and result in hyperammonemia (Semin Neonatol 2002;7:27). The sine qua non among all degrees of OTC deficiency at presentation is hyperammonemia. As in adults, children will have similar symptoms of encephalopathy, but this may be expressed differently depending on the child's developmental level. We present an unusual case of OTC deficiency in an older child with undifferentiated symptoms of an anticholinergic toxidrome, liver failure, iron overdose, and mushroom poisoning.


Subject(s)
Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Child , Diagnosis, Differential , Emergency Service, Hospital , Female , Humans , Ornithine Carbamoyltransferase Deficiency Disease/complications , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Psychotic Disorders/etiology
15.
J Hepatol ; 56(5): 1136-1144, 2012 May.
Article in English | MEDLINE | ID: mdl-22245901

ABSTRACT

BACKGROUND & AIMS: Multidrug resistance-associated protein 2 (in humans, MRP2; in rodents, Mrp2) mediates biliary excretion of bilirubin glucuronides. Therefore, upregulation of MRP2/Mrp2 expression may improve hyperbilirubinemia. We investigated the effects of 4-phenylbutyrate (4PBA), a drug used to treat ornithine transcarbamylase deficiency (OTCD), on the cell surface expression and transport function of MRP2/Mrp2 and serum T-Bil concentration. METHODS: MRP2-expressing MDCKII (MRP2-MDCKII) cells and rats were studied to explore the change induced by 4PBA treatment in the cell surface expression and transport function of MRP2/Mrp2 and its underlying mechanism. Serum and liver specimens from OTCD patients were analyzed to examine the effect of 4PBA on hepatic MRP2 expression and serum T-Bil concentration in humans. RESULTS: In MRP2-MDCKII cells and the rat liver, 4PBA increased the cell surface expression and transport function of MRP2/Mrp2. In patients with OTCD, hepatic MRP2 expression increased and serum T-Bil concentration decreased significantly after 4PBA treatment. In vitro studies designed to explore the mechanism underlying this drug action suggested that cell surface-resident MRP2/Mrp2 is degraded via ubiquitination-mediated targeting to the endosomal/lysosomal degradation pathway and that 4PBA inhibits the degradation of cell surface-resident MRP2/Mrp2 by reducing its susceptibility to ubiquitination. CONCLUSIONS: 4PBA activates MRP2/Mrp2 function through increased expression of MRP2/Mrp2 at the hepatocanalicular membrane by modulating its ubiquitination, and thereby decreases serum T-Bil concentration. 4PBA has thus therapeutic potential for improving hyperbilirubinemia.


Subject(s)
Bilirubin/blood , Liver/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Phenylbutyrates/pharmacology , Ubiquitination/drug effects , Adult , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Child, Preschool , Dogs , Female , Humans , Infant , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Models, Animal , Multidrug Resistance-Associated Protein 2 , Ornithine Carbamoyltransferase Deficiency Disease/drug therapy , Phenylbutyrates/therapeutic use , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
16.
J Inherit Metab Dis ; 35(5): 777-85, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22167275

ABSTRACT

Urea cycle disorders (UCDs) are one of the most frequently inherited metabolic diseases in Japan, with an estimated prevalence of 1 per 50,000 live births. Here, we investigated the clinical manifestations, treatment, and prognosis of 177 patients with UCDs who were evaluated and treated from January 1999 to March 2009. These included 77 cases of neonatal-onset UCDs and 91 cases of late-onset UCDs. The most common UCD was ornithine transcarbamylase deficiency (OTCD), which accounted for 116 out of 177 patients. This result is similar to a previous study performed between 1978 and 1995 in Japan: OTCD accounted for about two-thirds of the total number of UCD cases. We studied the relationship between prognosis and the peak blood ammonia level at the onset in 151 UCD patients. Compared with a previous survey conducted in Japan, we found that a greater number of patients survived without any mental retardation despite their peak blood ammonia levels being greater than 360 µmol/l. The 5-year survival rate of patients with OTCD improved to 86% for those with the neonatal-onset type and to 92% for those with the late-onset type. We hypothesize that the increased survival rate is due to early diagnosis and better treatments that are now available in Japan. It is very important to diagnose and treat UCDs, especially OTCD, when the blood ammonia levels in patients are low. The outcome in patients with low blood ammonia levels was better than that in patients with high blood ammonia levels.


Subject(s)
Ammonia/blood , Ammonia/metabolism , Urea Cycle Disorders, Inborn/diagnosis , Urea/metabolism , Age of Onset , Female , Humans , Japan , Male , Ornithine Carbamoyltransferase Deficiency Disease/blood , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Ornithine Carbamoyltransferase Deficiency Disease/drug therapy , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Prognosis , Survival Rate , Treatment Outcome , Urea Cycle Disorders, Inborn/blood , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/metabolism
17.
Mol Ther ; 17(8): 1340-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19384294

ABSTRACT

Ornithine transcarbamylase (OTC) deficiency, the most common urea cycle disorder, is associated with severe hyperammonemia accompanied by a high risk of neurological damage and death in patients presenting with the neonatal-onset form. Contemporary therapies, including liver transplantation, remain inadequate with considerable morbidity, justifying vigorous investigation of alternate therapies. Clinical evidence suggests that as little as 3% normal enzyme activity is sufficient to ameliorate the severe neonatal phenotype, making OTC deficiency an ideal model for the development of liver-targeted gene therapy. In this study, we investigated metabolic correction in neonatal and adult male OTC-deficient Spf(ash) mice following adeno-associated virus (AAV)2/8-mediated delivery of the murine OTC complementary DNA under the transcriptional control of a liver-specific promoter. Substantially supraphysiological levels of OTC enzymatic activity were readily achieved in both adult and neonatal mice following a single intraperitoneal (i.p.) injection, with metabolic correction in adults being robust and life-long. In the neonates, however, full metabolic correction was transient, although modest levels of OTC expression persisted into adulthood. Although not directly testable in Spf(ash) mice, these levels were theoretically sufficient to prevent hyperammonemia in a null phenotype. This loss of expression in the neonatal liver is the consequence of hepatocellular proliferation and presents an added challenge to human therapy.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/therapy , Animals , Animals, Newborn , Blotting, Western , Cell Line , DNA, Complementary/genetics , Female , Humans , Immunohistochemistry , Liver/metabolism , Male , Mice , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Ornithine Carbamoyltransferase/physiology , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/urine , Orotic Acid/urine
18.
Clin Biochem ; 84: 63-72, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32569589

ABSTRACT

BACKGROUND: Ornithine transcarbamylase deficiency (OTCD) is pleomorphic congenital hyperammonemia, in which the prognosis of the patient is determined both by genotype and environmental factors. This study investigated the clinical and biochemical characteristics of OTCD patients with different prognosis. METHOD: Of 35 OTCD patients, six males deceased at the first disease-onset, 17 males survived and had controllable ammonia levels after treatment, and 12 females survived through the first disease-onset but had intractable hyperammonemia and high mortality. Fasting blood samples from patients collected at three disease stages were used for the analysis of amino acid (AA) profile, acylcarnitine profile, and micronutrients. Differences in profiles between patients and healthy controls and within patient groups were studied. RESULTS: All OTCD patients had accumulation of glutamine, homocitrulline, lysine, glutamate, cystathionine, and pipecolic acid, as well as deficiency of citrulline, tryptophan, threonine, and carnitine. For male non-survivors, most other AAs and long-chain acylcarnitines were elevated at disease onset, of which the levels of creatine, N-acetylaspartic acid, and homoarginine were remarkably high. Male survivors and female patients had most other AAs at low to normal levels. Compared with male survivors, female patients had much lower protein-intolerance, as indicated by significantly lower levels of protein consumption indicators, including essential AAs, 1-methylhistidine, acylcarnitines et al., but high levels of ammonia. Female patients still had significantly higher levels of citrulline, homocitrulline, and citrulline/arginine compared to male survivors. CONCLUSION: Unique profiles were observed in each group of OTCD patients, indicating specific physiological changes that happened to them.


Subject(s)
Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/physiopathology , Adolescent , Adult , Ammonia/blood , Arginine/blood , Child , Child, Preschool , China , Creatine/metabolism , Female , Humans , Hyperammonemia/physiopathology , Lysine/blood , Male , Ornithine/therapeutic use , Ornithine Carbamoyltransferase Deficiency Disease/blood , Urea/blood , Young Adult
19.
Biomed Res Int ; 2020: 5690915, 2020.
Article in English | MEDLINE | ID: mdl-32934962

ABSTRACT

During Jan. 2016-Dec. 2019, nine Chinese patients from eight unrelated families were diagnosed with neonatal-onset UCDs by targeted panel sequencing or whole-exome sequencing (WES). Their clinical manifestations, biochemical features, 180-day-age outcomes, and molecular genetic characteristics were reviewed retrospectively. NGS-based tests revealed 7 patients diagnosed with ornithine transcarbamylase deficiency (OTCD) and 2 with carbamoylphosphate synthetase I deficiency (CPS1D). The spectrum of the clinical presentation of nine affected individuals progressed from unspecific symptoms like poor feeding to somnolence, coma, and death. All patients presented with an acute hyperammonemia. The most robust metabolic pattern in OTCD was hyperglutaminemic hyperammonemia with high concentration of urine orotic acid, and it was reported in six patients. Of ten variants found on the OTC gene and CPS1 gene, 3 were novel: (c.176T>C (p.L59P)) in the OTC gene, c.2938G>A (p.G980S) and c.3734T>A (p.L1245H) in the CPS1 gene. There was a high mortality rate of 77.78% (7/9) for all the defects combined. An OTC-deficient male and a CPS1-deficient female survived from episodes of hyperammonemia. Although prompt recognition of UCD and the use of alternative pathway therapy in addition to provision of appropriate nutrition and dialysis improved survival, the overall outcomes for the neonatal-onset type are poor in China.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Hyperammonemia/genetics , Ornithine Carbamoyltransferase/genetics , Urea Cycle Disorders, Inborn/genetics , Age of Onset , China , Female , High-Throughput Nucleotide Sequencing , Humans , Hyperammonemia/complications , Hyperammonemia/metabolism , Hyperammonemia/pathology , Infant, Newborn , Male , Metabolomics/methods , Mutation/genetics , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/pathology , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology , Exome Sequencing
20.
Radiology ; 252(3): 833-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19567648

ABSTRACT

PURPOSE: To determine cerebral glutamate turnover rate in partial-ornithine transcarbamylase deficiency (OTCD) patients by using carbon 13 ((13)C) magnetic resonance (MR) spectroscopy. MATERIALS AND METHODS: The study was performed with approval of the institutional review board, in compliance with HIPAA regulations, and with written informed consent of the subjects. MR imaging, hydrogen 1 ((1)H) MR spectroscopy, and (13)C MR spectroscopy were performed at 1.5 T in 10 subjects, six patients with OTCD and four healthy control subjects, who were in stable condition. Each received intravenous (13)C-glucose (0.2 g/kg), C1 or C2 position, as a 15-minute bolus. Cerebral metabolites were determined with proton decoupling in a parieto-occipital region (n = 9) and without proton decoupling in a frontal region (n = 1) during 60-120 minutes. RESULTS: Uptake and removal of cerebral glucose ([1-(13)C]-glucose or [2-(13)C]-glucose) were comparable in healthy control subjects and subjects with OTCD (P = .1). Glucose C1 was metabolized to glutamate C4 and glucose C2 was metabolized to glutamate C5 at comparable rates, both of which were significantly reduced in OTCD (combined, P = .04). No significant differences in glutamine formation were found in subjects with OTCD (P = .1). [2-(13)C]-glucose and its metabolic products were observed in anterior cingulate gyrus without proton decoupling in one subject with OTCD. CONCLUSION: Treatments that improve cerebral glucose metabolism and glutamate neurotransmission may improve neurologic outcome in patients with OTCD, in whom prevention and treatment of hyperammonemic episodes appear to be insufficient.


Subject(s)
Brain/metabolism , Glutamic Acid/metabolism , Magnetic Resonance Spectroscopy/methods , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Adult , Carbon Isotopes , Case-Control Studies , Female , Glucose/metabolism , Humans , Male , Middle Aged , Ornithine Carbamoyltransferase Deficiency Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL