Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 99
1.
J Inherit Metab Dis ; 47(1): 50-62, 2024 Jan.
Article En | MEDLINE | ID: mdl-37026568

Urea cycle defects (UCDs) are severe inherited metabolic diseases with high unmet needs which present a permanent risk of hyperammonaemic decompensation and subsequent acute death or neurological sequelae, when treated with conventional dietetic and medical therapies. Liver transplantation is currently the only curative option, but has the potential to be supplanted by highly effective gene therapy interventions without the attendant need for life-long immunosuppression or limitations imposed by donor liver supply. Over the last three decades, pioneering genetic technologies have been explored to circumvent the consequences of UCDs, improve quality of life and long-term outcomes: adenoviral vectors, adeno-associated viral vectors, gene editing, genome integration and non-viral technology with messenger RNA. In this review, we present a summarised view of this historical path, which includes some seminal milestones of the gene therapy's epic. We provide an update about the state of the art of gene therapy technologies for UCDs and the current advantages and pitfalls driving future directions for research and development.


Liver Transplantation , Ornithine Carbamoyltransferase Deficiency Disease , Urea Cycle Disorders, Inborn , Humans , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Quality of Life , Urea/metabolism , Living Donors , Genetic Therapy , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/therapy , Urea Cycle Disorders, Inborn/complications
2.
Genet Med ; 26(4): 101039, 2024 Apr.
Article En | MEDLINE | ID: mdl-38054409

PURPOSE: Liver transplantation (LTx) is performed in individuals with urea cycle disorders when medical management (MM) insufficiently prevents the occurrence of hyperammonemic events. However, there is a paucity of systematic analyses on the effects of LTx on health-related outcome parameters compared to individuals with comparable severity who are medically managed. METHODS: We investigated the effects of LTx and MM on validated health-related outcome parameters, including the metabolic disease course, linear growth, and neurocognitive outcomes. Individuals were stratified into "severe" and "attenuated" categories based on the genotype-specific and validated in vitro enzyme activity. RESULTS: LTx enabled metabolic stability by prevention of further hyperammonemic events after transplantation and was associated with a more favorable growth outcome compared with individuals remaining under MM. However, neurocognitive outcome in individuals with LTx did not differ from the medically managed counterparts as reflected by the frequency of motor abnormality and cognitive standard deviation score at last observation. CONCLUSION: Whereas LTx enabled metabolic stability without further need of protein restriction or nitrogen-scavenging therapy and was associated with a more favorable growth outcome, LTx-as currently performed-was not associated with improved neurocognitive outcomes compared with long-term MM in the investigated urea cycle disorders.


Liver Transplantation , Urea Cycle Disorders, Inborn , Humans , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/surgery , Proteins , Outcome Assessment, Health Care
3.
Int J Dev Neurosci ; 82(8): 772-788, 2022 Dec.
Article En | MEDLINE | ID: mdl-36129623

Urea cycle disorders (UCD) are a group of genetic diseases caused by deficiencies in the enzymes and transporters involved in the urea cycle. The impairment of the cycle results in ammonia accumulation, leading to neurological dysfunctions and poor outcomes to affected patients. The aim of this study is to investigate and describe UCD patients' principal clinical and biochemical presentations to support professionals on urgent diagnosis and quick management, aiming better outcomes for patients. We explored medical records of 30 patients diagnosed in a referral center from Brazil to delineate UCD clinical and biochemical profile. Patients demonstrated a range of signs and symptoms, such as altered levels of consciousness, acute encephalopathy, seizures, progressive loss of appetite, vomiting, coma, and respiratory distress, in most cases combined with high levels of ammonia, which is an immediate biomarker, leading to a UCD suspicion. The most prevalent UCD detected were ornithine transcarbamylase deficiency, followed by citrullinemia type 1, hyperargininemia, carbamoyl phosphate synthase 1 deficiency, and argininosuccinic aciduria. Clinical symptoms were highly severe, being the majority developmental and neurological disabilities, with 20% of death rate. Laboratory analysis revealed high levels of ammonia (mean ± SD: 860 ± 470 µmol/L; reference value: ≤80 µmol/L), hypoglycemia, metabolic acidosis, and high excretion of orotic acid in the urine (except in carbamoyl phosphate synthetase 1 [CPS1] deficiency). We emphasize the need of urgent identification of UCD clinical and biochemical conditions, and immediate measurement of ammonia, to enable the correct diagnosis and increase the chances of patients' survival, minimizing neurological and psychomotor damage caused by hepatic encephalopathy.


Hepatic Encephalopathy , Hyperammonemia , Ornithine Carbamoyltransferase Deficiency Disease , Urea Cycle Disorders, Inborn , Humans , Hyperammonemia/complications , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Hepatic Encephalopathy/complications , Hepatic Encephalopathy/diagnosis , Ammonia , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics , Ornithine Carbamoyltransferase Deficiency Disease/complications , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Ornithine Carbamoyltransferase Deficiency Disease/genetics
4.
Front Immunol ; 13: 861516, 2022.
Article En | MEDLINE | ID: mdl-35711415

The hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome is a rare autosomal recessive inborn error of the urea cycle caused by mutations in the SLC25A15 gene. Besides the well-known metabolic complications, patients often present intercurrent infections associated with acute hyperammonemia and metabolic decompensation. However, it is currently unknown whether intercurrent infections are associated with immunological alterations besides the known metabolic imbalances. Herein, we describe the case of a 3-years-old girl affected by the HHH syndrome caused by two novel SLC25A15 gene mutations associated with immune phenotypic and functional alterations. She was admitted to the hospital with an episode of recurrent otitis, somnolence, confusion, and lethargy. Laboratory tests revealed severe hyperammonemia, elevated serum levels of liver transaminases, hemostasis alterations, hyperglutaminemia and strikingly increased orotic aciduria. Noteworthy, serum protein electrophoresis showed a reduction in the gamma globulin fraction. Direct sequencing of the SLC25A15 gene revealed two heterozygous non-conservative substitutions in the exon 5: c.649G>A (p.Gly217Arg) and c.706A>G (p.Arg236Gly). In silico analysis indicated that both mutations significantly impair protein structure and function and are consistent with the patient clinical status confirming the diagnosis of HHH syndrome. In addition, the immune analysis revealed reduced levels of serum IgG and striking phenotypic and functional alterations in the T and B cell immune compartments. Our study has identified two non-previously described mutations in the SLC25A15 gene underlying the HHH syndrome. Moreover, we are reporting for the first time functional and phenotypic immunologic alterations in this rare inborn error of metabolism that would render the patient immunocompromised and might be related to the high frequency of intercurrent infections observed in patients bearing urea cycle disorders. Our results point out the importance of a comprehensive analysis to gain further insights into the underlying pathophysiology of the disease that would allow better patient care and quality of life.


Hyperammonemia , Urea Cycle Disorders, Inborn , Amino Acid Transport Systems, Basic/genetics , Child, Preschool , Female , Humans , Hyperammonemia/complications , Hyperammonemia/diagnosis , Mitochondrial Membrane Transport Proteins , Ornithine/deficiency , Quality of Life , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics
5.
Mol Genet Metab ; 135(4): 327-332, 2022 04.
Article En | MEDLINE | ID: mdl-35279366

Citrulline is a target analyte measured at expanded newborn screening (NBS) and its elevation represents a biomarker for distal urea cycle disorders and citrin deficiency. Altered ratios of citrulline with other urea cycle-related amino acids are helpful for the differential diagnosis. However, the use of cut-off values in screening programmes has raised the issue about the interpretation of mild elevation of citrulline levels detected at NBS, below the usual range observed in the "classical/severe" forms of distal urea cycle disorders and in citrin deficiency. Herein, we report ten subjects with positive NBS for a mild elevation of citrulline (<100 µmol/L), in whom molecular investigations revealed carriers status for argininosuccinate synthase deficiency, a milder form of argininosuccinate lyase deficiency and two other diseases, lysinuric protein intolerance and dihydrolipoamide dehydrogenase deficiency, not primarily affecting the urea cycle. To guide the diagnostic process, we have designed an algorithm for mild citrulline elevation (<100 µmol/L) at NBS, which expands the list of disorders to be included in the differential diagnosis.


Citrulline , Urea Cycle Disorders, Inborn , Citrullinemia , Humans , Infant, Newborn , Neonatal Screening , Urea , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics
6.
J Inherit Metab Dis ; 45(3): 470-480, 2022 05.
Article En | MEDLINE | ID: mdl-34988999

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.


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
7.
Clin Chim Acta ; 526: 55-61, 2022 Feb 01.
Article En | MEDLINE | ID: mdl-34973183

PURPOSE: Carbamoyl phosphate synthetase 1 (CPS1) deficiency affects the first step of urea cycle and is a severe form of urea cycle disorder (UCD). The severity of hyperammonemic encephalopathy determines the clinical course of UCDs. Here, we describe the genetic and clinical characteristics of CPS1 deficiency in Korea. PATIENT AND METHODS: This study included seven patients with CPS1 deficiency genetically confirmed from January 1992 to September 2020. The peak ammonia level during the first crisis, the half time of peak ammonia level, the initial plasma amino acid levels, and neurological outcomes were compared between CPS1 deficiency and two common UCDs (i.e., 17 patients with argininosuccinate synthetase 1 deficiency and 24 patients with ornithine transcarbamylase deficiency). RESULT: Eleven CPS1 mutations were identified, including 10 novel mutations. Eight mutations were missense. Six patients with CPS1 deficiency had neonatal type. The peak ammonia level, initial glutamate level, and accompanying rate of irreversible neurological damages were highest in patients with CPS1 deficiency. The patient with late-onset CPS1 deficiency responded dramatically to N-carbamylglutamate treatment. CONCLUSION: The clinical manifestations of CPS1 deficiency were the most severe among UCDs. Considering the high proportion of missense mutations, responsiveness to N-carbamylglutamate would be evaluated in a future study.


Carbamoyl-Phosphate Synthase (Ammonia) , Carbamoyl-Phosphate Synthase I Deficiency Disease , Urea Cycle Disorders, Inborn , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/diagnosis , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Carbamyl Phosphate , Humans , Infant, Newborn , Mutation , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics
8.
Intern Med ; 61(4): 553-557, 2022 Feb 15.
Article En | MEDLINE | ID: mdl-34433721

A 49-year-old Japanese man had shown developmental delay, learning difficulties, epilepsy, and slowly progressive gait disturbance in elementary school. At 46 years old, he experienced repeated drowsiness with or without generalized convulsions, and hyperammonemia was detected. Brain magnetic resonance imaging detected multiple cerebral white matter lesions. An electroencephalogram showed diffuse slow basic activities with 2- to 3-Hz δ waves. Genetic tests confirmed a diagnosis of hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome. Leukoencephalopathy was resolved following the administration of L-arginine and lactulose with a decrease in plasma ammonia levels and glutamine-glutamate peak on magnetic resonance spectroscopy. Leukoencephalopathy in HHH syndrome may be reversible with the resolution of hyperammonemia-induced glutamine toxicity.


Hyperammonemia , Leukoencephalopathies , Urea Cycle Disorders, Inborn , Ammonia , Child , Humans , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Male , Middle Aged , Ornithine/deficiency , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics
9.
Hum Mutat ; 42(12): 1624-1636, 2021 12.
Article En | MEDLINE | ID: mdl-34510628

N-acetylglutamate synthase deficiency is an autosomal recessive urea cycle disorder caused either by decreased expression of the NAGS gene or defective NAGS enzyme resulting in decreased production of N-acetylglutamate (NAG), an allosteric activator of carbamylphosphate synthetase 1 (CPS1). NAGSD is the only urea cycle disorder that can be effectively treated with a single drug, N-carbamylglutamate (NCG), a stable NAG analog, which activates CPS1 to restore ureagenesis. We describe three patients with NAGSD due to four novel noncoding sequence variants in the NAGS regulatory regions. All three patients had hyperammonemia that resolved upon treatment with NCG. Sequence variants NM_153006.2:c.427-222G>A and NM_153006.2:c.427-218A>C reside in the 547 bp-long first intron of NAGS and define a novel NAGS regulatory element that binds retinoic X receptor α. Sequence variants NC_000017.10:g.42078967A>T (NM_153006.2:c.-3065A>T) and NC_000017.10:g.42078934C>T (NM_153006.2:c.-3098C>T) reside in the NAGS enhancer, within known HNF1 and predicted glucocorticoid receptor binding sites, respectively. Reporter gene assays in HepG2 and HuH-7 cells demonstrated that all four substitutions could result in reduced expression of NAGS. These findings show that analyzing noncoding regions of NAGS and other urea cycle genes can reveal molecular causes of disease and identify novel regulators of ureagenesis.


Amino-Acid N-Acetyltransferase , Hyperammonemia , Urea Cycle Disorders, Inborn , Amino-Acid N-Acetyltransferase/chemistry , Amino-Acid N-Acetyltransferase/genetics , Humans , Hyperammonemia/genetics , Introns , Regulatory Sequences, Nucleic Acid , Urea Cycle Disorders, Inborn/genetics
10.
Hepatology ; 74(6): 3249-3268, 2021 12.
Article En | MEDLINE | ID: mdl-34343359

BACKGROUND AND AIMS: Metabolic reprogramming plays an important role in tumorigenesis. However, the metabolic types of different tumors are diverse and lack in-depth study. Here, through analysis of big databases and clinical samples, we identified a carbamoyl phosphate synthetase 1 (CPS1)-deficient hepatocellular carcinoma (HCC) subtype, explored tumorigenesis mechanism of this HCC subtype, and aimed to investigate metabolic reprogramming as a target for HCC prevention. APPROACH AND RESULTS: A pan-cancer study involving differentially expressed metabolic genes of 7,764 tumor samples in 16 cancer types provided by The Cancer Genome Atlas (TCGA) demonstrated that urea cycle (UC) was liver-specific and was down-regulated in HCC. A large-scale gene expression data analysis including 2,596 HCC cases in 7 HCC cohorts from Database of HCC Expression Atlas and 17,444 HCC cases from in-house hepatectomy cohort identified a specific CPS1-deficent HCC subtype with poor clinical prognosis. In vitro and in vivo validation confirmed the crucial role of CPS1 in HCC. Liquid chromatography-mass spectrometry assay and Seahorse analysis revealed that UC disorder (UCD) led to the deceleration of the tricarboxylic acid cycle, whereas excess ammonia caused by CPS1 deficiency activated fatty acid oxidation (FAO) through phosphorylated adenosine monophosphate-activated protein kinase. Mechanistically, FAO provided sufficient ATP for cell proliferation and enhanced chemoresistance of HCC cells by activating forkhead box protein M1. Subcutaneous xenograft tumor models and patient-derived organoids were employed to identify that blocking FAO by etomoxir may provide therapeutic benefit to HCC patients with CPS1 deficiency. CONCLUSIONS: In conclusion, our results prove a direct link between UCD and cancer stemness in HCC, define a CPS1-deficient HCC subtype through big-data mining, and provide insights for therapeutics for this type of HCC through targeting FAO.


Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Case-Control Studies , Cell Line, Tumor , DNA Methylation , Gas Chromatography-Mass Spectrometry , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Transcriptome , Urea Cycle Disorders, Inborn/enzymology , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology
11.
BMJ Case Rep ; 14(7)2021 Jul 01.
Article En | MEDLINE | ID: mdl-34210698

Hyperornithinaemia-hyperammonaemia-homocitrullinuria (HHH) syndrome is a rare inherited metabolic disorder of the urea cycle. Few reports exist to guide practices during pregnancy and fetal delivery. Yet, with affected patients often surviving into reproductive age, appropriate management of the peripartum phase is essential to ensure positive maternal and fetal outcomes.Reassuringly, the vast majority of offspring of parturients with HHH syndrome have normal developmental outcomes; yet as seen here, fetal growth restriction does appear more frequently. Furthermore, in addition to the absent fetal corpus callosum observed in this case, other fetal cerebral abnormalities, including speech delay and intellectual impairment, have been recognised.Unregulated dietary intake is one proposed factor for the observed disruption in fetal growth and early cerebral development. These stipulations not only reinforce the importance of extensive planning and teamwork, but also demonstrate the importance of timely intervention by a metabolic dietician and dietary compliance in the early organogenesis stage of pregnancy.


Hyperammonemia , Urea Cycle Disorders, Inborn , Citrulline , Female , Humans , Hyperammonemia/diagnosis , Hyperammonemia/therapy , Ornithine/deficiency , Pregnancy , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics
12.
Am J Med Genet A ; 185(7): 2026-2036, 2021 07.
Article En | MEDLINE | ID: mdl-33851512

Urea cycle disorders (UCDs) are inherited metabolic diseases that lead to hyperammonemia with variable clinical manifestations. Using data from a nationwide study, we investigated the onset time, gene variants, clinical manifestations, and treatment of patients with UCDs in Japan. Of the 229 patients with UCDs diagnosed and/or treated between January 2000 and March 2018, identified gene variants and clinical information were available for 102 patients, including 62 patients with ornithine transcarbamylase (OTC) deficiency, 18 patients with carbamoyl phosphate synthetase 1 (CPS1) deficiency, 16 patients with argininosuccinate synthetase (ASS) deficiency, and 6 patients with argininosuccinate lyase (ASL) deficiency. A total of 13, 10, 4, and 5 variants in the OTC, CPS1, ASS, and ASL genes were respectively identified as novel variants, which were neither registered in ClinVar databases nor previously reported. The onset time and severity in patients with UCD could be predicted based on the identified gene variants in each patient from this nationwide study and previous studies. This genetic information may help in predicting the long-term outcome and determining specific treatment strategies such as liver transplantation in patients with UCDs.


Argininosuccinate Lyase/genetics , Argininosuccinate Synthase/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Ornithine Carbamoyltransferase/genetics , Urea Cycle Disorders, Inborn/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Genetic Variation/genetics , Humans , Hyperammonemia/enzymology , Hyperammonemia/genetics , Hyperammonemia/pathology , Infant , Male , Metabolic Diseases/enzymology , Metabolic Diseases/genetics , Metabolic Diseases/pathology , Urea Cycle Disorders, Inborn/enzymology , Urea Cycle Disorders, Inborn/pathology , Young Adult
13.
Mol Genet Metab ; 133(2): 148-156, 2021 06.
Article En | MEDLINE | ID: mdl-33846069

BACKGROUND: Urea cycle disorders (UCDs) are among the most common inborn errors of liver metabolism. As therapies for hyperammonemia associated with urea cycle dysfunction have improved, chronic complications, such as liver disease, have become increasingly apparent in individuals with UCDs. Liver disease in UCDs may be associated with hepatic inflammation, hepatic fibrosis, portal hypertension, liver cancer and even liver failure. However, except for monitoring serum aminotransferases, there are no clear guidelines for screening and/or monitoring individuals with UCDs for liver disease. Thus, we systematically evaluated the potential utility of several non-invasive biomarkers for liver fibrosis in UCDs. METHODS: We evaluated grey-scale ultrasonography, liver stiffness obtained from shear wave elastography (SWE), and various serum biomarkers for hepatic fibrosis and necroinflammation, in a cohort of 28 children and adults with various UCDs. RESULTS: Overall, we demonstrate a high burden of liver disease in our participants with 46% of participants having abnormal grey-scale ultrasound pattern of the liver parenchyma, and 52% of individuals having increased liver stiffness. The analysis of serum biomarkers revealed that 32% of participants had elevated FibroTest™ score, a marker for hepatic fibrosis, and 25% of participants had increased ActiTest™ score, a marker for necroinflammation. Interestingly, liver stiffness did not correlate with ultrasound appearance or FibroTest™. CONCLUSION: Overall, our results demonstrate the high overall burden of liver disease in UCDs and highlights the need for further studies exploring new tools for identifying and monitoring individuals with UCDs who are at risk for this complication. TRIAL REGISTRATION: This study has been registered in ClinicalTrials.gov (NCT03721367).


Argininosuccinate Lyase/blood , Genetic Diseases, Inborn/blood , Liver Cirrhosis/blood , Liver Diseases/blood , Urea Cycle Disorders, Inborn/blood , Adolescent , Adult , Biomarkers/blood , Child , Child, Preschool , Elasticity Imaging Techniques , Female , Genetic Diseases, Inborn/diagnostic imaging , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Humans , Hyperammonemia/blood , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/pathology , Liver/diagnostic imaging , Liver/pathology , Liver Cirrhosis/diagnostic imaging , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Diseases/genetics , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Metabolism, Inborn Errors/genetics , Middle Aged , Ultrasonography , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology , Young Adult
14.
Mol Genet Metab ; 132(4): 220-226, 2021 04.
Article En | MEDLINE | ID: mdl-33648834

Urea cycle disorders (UCDs), inborn errors of hepatocyte metabolism, result in the systemic accumulation of ammonia to toxic levels. Sodium 4-phenylbutyrate (NaPB), a standard therapy for UCDs for over 20 years, generates an alternative pathway of nitrogen deposition through glutamine consumption. Administration during or immediately after a meal is the accepted use of NaPB. However, this regimen is not based on clinical evidence. Here, an open-label, single-dose, five-period crossover study was conducted in healthy adults to investigate the effect of food on the pharmacokinetics of NaPB and determine any subsequent change in amino acid availability. Twenty subjects were randomized to one of four treatment groups. Following an overnight fast, NaPB was administered orally at 4.3 g/m2 (high dose, HD) or 1.4 g/m2 (low dose, LD) either 30 min before or just after breakfast. At both doses, compared with post-breakfast administration, pre-breakfast administration significantly increased systemic exposure of PB and decreased plasma glutamine availability. Pre-breakfast LD administration attenuated plasma glutamine availability to the same extent as post-breakfast HD administration. Regardless of the regimen, plasma levels of branched-chain amino acids (BCAA) were decreased below baseline in a dose-dependent manner. In conclusion, preprandial oral administration of NaPB maximized systemic exposure of the drug and thereby its potency to consume plasma glutamine. This finding may improve poor medication compliance because of the issues with odor, taste, and pill burden of NaPB and reduce the risk of BCAA deficiency in NaPB therapy.


Eating/genetics , Pharmacokinetics , Phenylbutyrates/administration & dosage , Urea Cycle Disorders, Inborn/drug therapy , Administration, Oral , Adult , Amino Acids/genetics , Amino Acids, Branched-Chain/genetics , Biological Availability , Female , Glutamine/genetics , Healthy Volunteers , Humans , Male , Middle Aged , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/pathology , Young Adult
15.
Sci Rep ; 11(1): 3580, 2021 02 11.
Article En | MEDLINE | ID: mdl-33574402

The urea cycle protects the central nervous system from ammonia toxicity by converting ammonia to urea. N-acetylglutamate synthase (NAGS) catalyzes formation of N-acetylglutamate, an essential allosteric activator of carbamylphosphate synthetase 1. Enzymatic activity of mammalian NAGS doubles in the presence of L-arginine, but the physiological significance of NAGS activation by L-arginine has been unknown. The NAGS knockout (Nags-/-) mouse is an animal model of inducible hyperammonemia, which develops hyperammonemia without N-carbamylglutamate and L-citrulline supplementation (NCG + Cit). We used adeno associated virus (AAV) based gene transfer to correct NAGS deficiency in the Nags-/- mice, established the dose of the vector needed to rescue Nags-/- mice from hyperammonemia and measured expression levels of Nags mRNA and NAGS protein in the livers of rescued animals. This methodology was used to investigate the effect of L-arginine on ureagenesis in vivo by treating Nags-/- mice with AAV vectors encoding either wild-type or E354A mutant mouse NAGS (mNAGS), which is not activated by L-arginine. The Nags-/- mice expressing E354A mNAGS were viable but had elevated plasma ammonia concentration despite similar levels of the E354A and wild-type mNAGS proteins. The corresponding mutation in human NAGS (NP_694551.1:p.E360D) that abolishes binding and activation by L-arginine was identified in a patient with NAGS deficiency. Our results show that NAGS deficiency can be rescued by gene therapy, and suggest that L-arginine binding to the NAGS enzyme is essential for normal ureagenesis.


Amino-Acid N-Acetyltransferase/genetics , Gene Transfer Techniques , Hyperammonemia/genetics , Urea Cycle Disorders, Inborn/genetics , Amino-Acid N-Acetyltransferase/metabolism , Animals , Arginine/metabolism , Arginine/pharmacology , Citrulline/metabolism , Citrulline/pharmacology , Dependovirus/genetics , Disease Models, Animal , Glutamates/metabolism , Glutamates/pharmacology , Humans , Hyperammonemia/metabolism , Hyperammonemia/pathology , Hyperammonemia/therapy , Mice , Mice, Knockout , Mutant Proteins/genetics , Urea/metabolism , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology , Urea Cycle Disorders, Inborn/therapy
16.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 38(1): 74-77, 2021 Jan 10.
Article Zh | MEDLINE | ID: mdl-33423264

OBJECTIVE: To explore the genetic basis for a child with clinically suspected 3-methylcrotonyl-coenzyme A carboxylase deficiency (MCCD). METHODS: Genomic DNA was extracted from peripheral blood samples of the proband and her parents. Whole exome sequencing was used to screen pathogenic variant in the proband. Suspected variant was verified by Sanger sequencing. Impact of the variant on the structure and function of protein product was analyzed by using bioinformatic software. RESULTS: Sanger sequencing showed that the proband has carried homozygous missense c.1342G>A (p.Gly448Ala) variant of the MCCC2 gene, for which her mother was a heterozygous carrier. The same variant was not detected in her father. The variant was predicted to be pathogenic by PolyPhen-2 and Mutation Taster software, and the site was highly conserved among various species. Based on the American College of Medical Genetics and Genomics standards and guidelines, the c.1342G>A (p.Gly448Ala) variant of MCCC2 gene was predicted to be likely pathogenic(PM2+PP2-PP5). CONCLUSION: The homozygous missense variant of the MCCC2 gene c.1342G>A (p.Gly448Ala) probably underlay the molecular pathogenesis of the proband. Genetic testing has confirmed the clinical diagnosis.


Carbon-Carbon Ligases/deficiency , Mutation, Missense , Urea Cycle Disorders, Inborn , Carbon-Carbon Ligases/genetics , Child , Female , Humans , Male , Mutation, Missense/genetics , Pedigree , Urea Cycle Disorders, Inborn/genetics
17.
Pharmacopsychiatry ; 54(1): 5-17, 2021 Jan.
Article En | MEDLINE | ID: mdl-33147643

The implementation of pharmacogenomic (PGx) testing in psychiatry remains modest, in part due to divergent perceptions of the quality and completeness of the evidence base and diverse perspectives on the clinical utility of PGx testing among psychiatrists and other healthcare providers. Recognizing the current lack of consensus within the field, the International Society of Psychiatric Genetics assembled a group of experts to conduct a narrative synthesis of the PGx literature, prescribing guidelines, and product labels related to psychotropic medications as well as the key considerations and limitations related to the use of PGx testing in psychiatry. The group concluded that to inform medication selection and dosing of several commonly-used antidepressant and antipsychotic medications, current published evidence, prescribing guidelines, and product labels support the use of PGx testing for 2 cytochrome P450 genes (CYP2D6, CYP2C19). In addition, the evidence supports testing for human leukocyte antigen genes when using the mood stabilizers carbamazepine (HLA-A and HLA-B), oxcarbazepine (HLA-B), and phenytoin (CYP2C9, HLA-B). For valproate, screening for variants in certain genes (POLG, OTC, CSP1) is recommended when a mitochondrial disorder or a urea cycle disorder is suspected. Although barriers to implementing PGx testing remain to be fully resolved, the current trajectory of discovery and innovation in the field suggests these barriers will be overcome and testing will become an important tool in psychiatry.


Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Pharmacogenomic Testing/methods , Psychiatry/methods , Anticonvulsants/therapeutic use , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2D6/genetics , Dose-Response Relationship, Drug , HLA Antigens/genetics , Humans , Pharmacogenomic Testing/standards , Practice Guidelines as Topic , Psychiatry/standards , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/genetics
18.
Biochimie ; 183: 89-99, 2021 Apr.
Article En | MEDLINE | ID: mdl-33309754

Despite biochemical and genetic testing being the golden standards for identification of proximal urea cycle disorders (UCDs), genotype-phenotype correlations are often unclear. Co-occurring partial defects affecting more than one gene have not been demonstrated so far in proximal UCDs. Here, we analyzed the mutational spectrum of 557 suspected proximal UCD individuals. We probed oligomerizing forms of NAGS, CPS1 and OTC, and evaluated the surface exposure of residues mutated in heterozygously affected individuals. BN-PAGE and gel-filtration chromatography were employed to discover protein-protein interactions within recombinant enzymes. From a total of 281 confirmed patients, only 15 were identified as "heterozygous-only" candidates (i.e. single defective allele). Within these cases, the only missense variants to potentially qualify as dominant negative triggers were CPS1 p.Gly401Arg and NAGS p.Thr181Ala and p.Tyr512Cys, as assessed by residue oligomerization capacity and surface exposure. However, all three candidates seem to participate in critical intramolecular functions, thus, unlikely to facilitate protein-protein interactions. This interpretation is further supported by BN-PAGE and gel-filtration analyses revealing no multiprotein proximal urea cycle complex formation. Collectively, genetic analysis, structural considerations and in vitro experiments point against a prominent role of dominant negative effects in human proximal UCDs.


Amino-Acid N-Acetyltransferase , Carbamoyl-Phosphate Synthase (Ammonia) , Genes, Dominant , Mutation, Missense , Ornithine Carbamoyltransferase , Urea Cycle Disorders, Inborn , Amino Acid Substitution , Amino-Acid N-Acetyltransferase/chemistry , Amino-Acid N-Acetyltransferase/genetics , Amino-Acid N-Acetyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/chemistry , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Female , Heterozygote , Homozygote , Humans , Male , Ornithine Carbamoyltransferase/chemistry , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Protein Domains , Urea Cycle Disorders, Inborn/enzymology , Urea Cycle Disorders, Inborn/genetics
19.
Article Zh | WPRIM | ID: wpr-879527

OBJECTIVE@#To explore the genetic basis for a child with clinically suspected 3-methylcrotonyl-coenzyme A carboxylase deficiency (MCCD).@*METHODS@#Genomic DNA was extracted from peripheral blood samples of the proband and her parents. Whole exome sequencing was used to screen pathogenic variant in the proband. Suspected variant was verified by Sanger sequencing. Impact of the variant on the structure and function of protein product was analyzed by using bioinformatic software.@*RESULTS@#Sanger sequencing showed that the proband has carried homozygous missense c.1342G>A (p.Gly448Ala) variant of the MCCC2 gene, for which her mother was a heterozygous carrier. The same variant was not detected in her father. The variant was predicted to be pathogenic by PolyPhen-2 and Mutation Taster software, and the site was highly conserved among various species. Based on the American College of Medical Genetics and Genomics standards and guidelines, the c.1342G>A (p.Gly448Ala) variant of MCCC2 gene was predicted to be likely pathogenic(PM2+PP2-PP5).@*CONCLUSION@#The homozygous missense variant of the MCCC2 gene c.1342G>A (p.Gly448Ala) probably underlay the molecular pathogenesis of the proband. Genetic testing has confirmed the clinical diagnosis.


Child , Female , Humans , Male , Carbon-Carbon Ligases/genetics , Mutation, Missense/genetics , Pedigree , Urea Cycle Disorders, Inborn/genetics
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 37(12): 1360-1363, 2020 Dec 10.
Article Zh | MEDLINE | ID: mdl-33306822

OBJECTIVE: To explore the genetic basis for a Chinese pedigree affected with N-acetylglutamate synthase deficiency. METHODS: Trio whole exome sequencing (WES) was carried out for the pedigree. Pathogenicity of the identified variant was predicted based on the latest recommendation of the American College of Medical Genetics and Genomics (ACMG). Prenatal diagnosis was provided for subsequent pregnancy through Sanger sequencing. RESULTS: Trio WES showed that the proband has carried compound heterozygous c.68delG and c.796G>C variants of NAGS gene, for which the mother and father were respectively heterozygous carriers. Neither variant was reported previously. Based on the ACMG guidelines, the c.68delG variant was classified as "likely pathogenic" (PVS1+PM2), while the c.796G>C variant was classified as with "uncertain significance" (PM2+BP4). Sanger sequencing validated the above findings, and only detected the heterozygous c.796G>C variant in the amniotic fluid sample. The fetus was followed up till 6 month after birth with no obvious abnormality. CONCLUSION: The compound heterozygous c.68delG and c.796G>C variants of the NAGS gene probably underlay the disorder in this pedigree, and the resulth asenabled genetic counseling and prenatal diagnosis for this pedigree.


Genetic Testing , Prenatal Diagnosis , Urea Cycle Disorders, Inborn , Amino-Acid N-Acetyltransferase/genetics , China , Female , Humans , Male , Mutation/genetics , Pedigree , Pregnancy , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/genetics , Exome Sequencing
...