Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
ACS Chem Neurosci ; 15(5): 916-931, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38369717

ABSTRACT

Novel insights into the etiology of metabolic disorders have recently been uncovered through the study of metabolite amyloids. In particular, inborn errors of metabolism (IEMs), including gout, Lesch-Nyhan syndrome (LNS), xanthinuria, citrullinemia, and hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome, are attributed to the dysfunction of the urea cycle and uric acid pathway. In this study, we endeavored to understand and mechanistically characterize the aggregative property exhibited by the principal metabolites of the urea cycle and uric acid pathway, specifically hypoxanthine, xanthine, citrulline, and ornithine. Employing scanning electron microscopy (SEM), transmission electron microscopy (TEM), and atomic force microscopy (AFM), we studied the aggregation profiles of the metabolites. Insights obtained through molecular dynamics (MD) simulation underscore the vital roles of π-π stacking and hydrogen bonding interactions in the self-assembly process, and thioflavin T (ThT) assays further corroborate the amyloid nature of these metabolites. The in vitro MTT assay revealed the cytotoxic trait of these assemblies, a finding that was substantiated by in vivo assays employing the Caenorhabditis elegans (C. elegans) model, which revealed that the toxic effects were more pronounced and dose-specific in the case of metabolites that had aged via longer preincubation. We hence report a compelling phenomenon wherein these metabolites not only aggregate but transform into a soft, ordered assembly over time, eventually crystallizing upon extended incubation, leading to pathological implications. Our study suggests that the amyloidogenic nature of the involved metabolites could be a common etiological link in IEMs, potentially providing a unified perspective to study their pathophysiology, thus offering exciting insights into the development of targeted interventions for these metabolic disorders.


Subject(s)
Hyperammonemia , Ornithine/deficiency , Urea Cycle Disorders, Inborn , Uric Acid , Animals , Caenorhabditis elegans , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology , Amyloid/metabolism , Ornithine/metabolism , Urea
2.
Eur Rev Med Pharmacol Sci ; 27(22): 11131-11142, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38039045

ABSTRACT

BACKGROUND: Urea cycle disorders (UCDs) are a group of rare inborn diseases caused by a deficiency in one of the six enzymes or one of the two transporters involved in the urea cycle. The most common biochemical feature is elevated blood ammonia levels, which can be toxic at high levels, especially to the brain and may manifest as encephalopathy if left untreated. Glycerol phenylbutyrate (GPB) is currently approved for use in the USA and Europe for patients of all ages with UCD who cannot be managed with protein restriction and/or amino acid supplementation alone. This article presents the author's experience in different exemplary settings and depicts the most efficient management of UCDs with GPB. CASE PRESENTATION: Six patient histories are described. 4 had OCT, one citrullinemia, and one argininosuccinic aciduria. Treatment with GPB was started between 2 days and 14 years of age. Before GPB, one patient had not been treated, 4 had received sodium phenylbutyrate (NaPB), and one Na benzoate. CONCLUSIONS: Overall, treatment with GPB was followed by a relevant metabolic improvement, resulting in better therapeutic compliance, reduced hospitalization, and improved quality of life.


Subject(s)
Quality of Life , Urea Cycle Disorders, Inborn , Humans , Glutamine/metabolism , Ammonia/metabolism , Ammonia/therapeutic use , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/metabolism , Urea/therapeutic use , Urea/metabolism
3.
Arch Biochem Biophys ; 736: 109526, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36702451

ABSTRACT

Urea cycle disorders (UCD) are inborn errors of metabolism that occur due to a loss of function in enzymes and transporters involved in the urea cycle, causing an intoxication by hyperammonemia and accumulation of metabolites. Patients can develop hepatic encephalopathy (HE), severe neurological and motor disabilities, and often death. The mechanisms involved in the pathophysiology of UCD are many and complex, but there are strong indications that oxidative stress and inflammation are present, being responsible for at least part of the cellular damage that occurs in these diseases. The aim of this study was to evaluate oxidative and nitrosative damage and inflammation in UCD, to better understand the pathophysiology mechanisms of these diseases. We evaluated the nitrite and nitrate content, thiobarbituric acid-reactive substances (TBARS), carbonyl protein content and a panel of cytokines in plasma sample of 14 patients. The UCD patients group consisted of individuals affected with ornithine transcarbamylase deficiency (n = 8), carbamoyl phosphate synthetase deficiency (n = 2), argininosuccinate synthetase deficiency (n = 2); arginase 1 deficiency (n = 1) and argininosuccinate lyase deficiency (n = 1). Patients mean age at diagnosis was 5.25 ± 9.86 years-old and mean concentrations were compared with healthy individuals of matched age and gender. We found a significant reduction in nitrogen reactive species in patients when compared to controls. TBARS was increased in patients, indicating lipid peroxidation. To evaluate protein oxidative damage in UCD, the carbonyl content was measured, and the results also demonstrated an increase in this biomarker. Finally, we found that UCD patients have enhanced concentrations of cytokines, with pro-inflammatory interleukins IL-6, IL-8, interferon-γ and TNF-α, and anti-inflammatory IL-10 being increased when compared to the control group. In conclusion, our results demonstrate that oxidative stress and inflammation occurs in UCD and probably contribute to the severe brain damage present in patients.


Subject(s)
Urea Cycle Disorders, Inborn , Adolescent , Child , Child, Preschool , Humans , Cytokines/metabolism , Inflammation , Oxidative Stress , Thiobarbituric Acid Reactive Substances , Urea , Urea Cycle Disorders, Inborn/metabolism , Infant, Newborn , Infant
4.
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
5.
Anal Biochem ; 636: 114343, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34637785

ABSTRACT

Urea cycle disorders (UCD) are inherited diseases resulting from deficiency in one of six enzymes or two carriers that are required to remove ammonia from the body. UCD may be associated with neurological damage encompassing a spectrum from asymptomatic/mild to severe encephalopathy, which results in most cases from Hyperammonemia (HA) and elevation of other neurotoxic intermediates of metabolism. Electroencephalography (EEG), Magnetic resonance imaging (MRI) and Proton Magnetic resonance spectroscopy (MRS) are noninvasive measures of brain function and structure that can be used during HA to guide management and provide prognostic information, in addition to being research tools to understand the pathophysiology of UCD associated brain injury. The Urea Cycle Rare disorders Consortium (UCDC) has been invested in research to understand the immediate and downstream effects of hyperammonemia (HA) on brain using electroencephalogram (EEG) and multimodal brain MRI to establish early patterns of brain injury and to track recovery and prognosis. This review highlights the evolving knowledge about the impact of UCD and HA in particular on neurological injury and recovery and use of EEG and MRI to study and evaluate prognostic factors for risk and recovery. It recognizes the work of others and discusses the UCDC's prior work and future research priorities.


Subject(s)
Brain , Electroencephalography , Hyperammonemia , Magnetic Resonance Imaging , Proton Magnetic Resonance Spectroscopy , Urea Cycle Disorders, Inborn , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain/physiopathology , History, 21st Century , Hyperammonemia/diagnostic imaging , Hyperammonemia/history , Hyperammonemia/metabolism , Hyperammonemia/physiopathology , Urea Cycle Disorders, Inborn/diagnostic imaging , Urea Cycle Disorders, Inborn/history , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/physiopathology
6.
Mol Cell ; 81(18): 3749-3759, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34469752

ABSTRACT

The expression of the urea cycle (UC) proteins is dysregulated in multiple cancers, providing metabolic benefits to tumor survival, proliferation, and growth. Here, we review the main changes described in the expression of UC enzymes and metabolites in different cancers at various stages and suggest that these changes are dynamic and should hence be viewed in a context-specific manner. Understanding the evolvability in the activity of the UC pathway in cancer has implications for cancer-immune cell interactions and for cancer diagnosis and therapy.


Subject(s)
Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Urea/metabolism , Ammonia/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/physiopathology
7.
Hepatology ; 74(6): 3249-3268, 2021 12.
Article in English | MEDLINE | ID: mdl-34343359

ABSTRACT

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.


Subject(s)
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
8.
J Inherit Metab Dis ; 44(6): 1311-1322, 2021 11.
Article in English | MEDLINE | ID: mdl-34232532

ABSTRACT

Urea cycle disorders (UCDs) are inherited metabolic diseases causing hyperammonemia by defects in urea cycle enzymes or transporters. Liver transplantation (LT) currently is the only curative treatment option until novel therapies become available. We performed a nationwide questionnaire-based study between January 2000 and March 2018 to investigate the effect of LT in patients with UCDs in Japan. A total of 231 patients with UCDs were enrolled in this study. Of them, a total of 78 patients with UCDs (30 male and 16 female ornithine transcarbamylase deficiency (OTCD), 21 carbamoyl phosphate synthetase 1 deficiency (CPSD), 10 argininosuccinate synthetase deficiency (ASSD) and 1 arginase 1 deficiency (ARGD)) had undergone LT. Concerning the maximum blood ammonia levels at the onset time in the transplanted male OTCD (N = 28), female OTCD (N = 15), CPSD (N = 21) and ASSD (N = 10), those were median 634 (IQR: 277-1172), 268 (211-352), 806 (535-1382), and 628 (425-957) µmol/L, respectively. The maximum blood ammonia levels in female OTCD were thus significantly lower than in the other UCDs (all P < .01). LT was effective for long-term survival, prevented recurrent hyperammonemia attack, and lowered baseline blood ammonia levels in patients with UCDs. LT had limited effect for ameliorating neurodevelopmental outcome in patients with severe disease because hyperammonemia at the onset time already had a significant impact on the brain. Patients with ASSD may be more likely to survive without cognitive impairment by receiving early LT despite severe neonatal hyperammonemia ≥ 360 µmol/L. In patients with neonatal onset OTCD or CPSD, there may be additional factors with adverse effects on the brain that are not improved by LT.


Subject(s)
Liver Transplantation , Urea Cycle Disorders, Inborn/surgery , Adolescent , Brain/metabolism , Child , Child Development/physiology , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Japan , Male , Survival Rate , Treatment Outcome , Urea Cycle Disorders, Inborn/metabolism
9.
Mol Genet Metab ; 133(2): 148-156, 2021 06.
Article in English | MEDLINE | ID: mdl-33846069

ABSTRACT

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).


Subject(s)
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
10.
J Cell Mol Med ; 25(8): 4099-4109, 2021 04.
Article in English | MEDLINE | ID: mdl-33611823

ABSTRACT

Urea cycle disorders (UCDs) are a group of rare metabolic conditions characterized by hyperammonemia and a broad spectrum of phenotypic severity. They are caused by the congenital deficiency in the eight biomolecules involved in urea cycle. In the present study, five cases of UCD were recruited and submitted to a series of clinical, biochemical, and genetic analysis with a combination of high throughput techniques. Moreover, in silico analysis was conducted on the identified missense genetic variants. Various clinical and biochemical indications (including profiles of amino acids and urinary orotic acids) of UCD were manifested by the five probands. Sequence analysis revealed nine diagnostic variants, including three novel ones, which caused Argininosuccinic aciduria (ASA) in one case, Carbamoyl phosphate synthetase 1deficiency (CPS1D) in two cases, Ornithine transcarbamylase deficiency (OTCD) in one case, and Citrin deficiency in 1case. Results of in silico biophysical analysis strongly suggested the pathogenicity of each the five missense variants and provided insight into their intramolecular impacts. In conclusion, this study expanded the genetic variation spectrum of UCD, gave solid evidence for counselling to the affected families, and should facilitate the functional study on the proteins in urea cycle.


Subject(s)
Computer Simulation , Mutation, Missense , Ornithine Carbamoyltransferase/genetics , Urea Cycle Disorders, Inborn/pathology , DNA Mutational Analysis , Female , Humans , Infant , Infant, Newborn , Male , Pedigree , Prognosis , Urea Cycle Disorders, Inborn/etiology , Urea Cycle Disorders, Inborn/metabolism
11.
Sci Rep ; 11(1): 3580, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574402

ABSTRACT

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.


Subject(s)
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
12.
Mol Genet Metab ; 132(1): 19-26, 2021 01.
Article in English | MEDLINE | ID: mdl-33388234

ABSTRACT

BACKGROUND/AIMS: Neonatal onset Urea cycle disorders (UCDs) can be life threatening with severe hyperammonemia and poor neurological outcomes. Glycerol phenylbutyrate (GPB) is safe and effective in reducing ammonia levels in patients with UCD above 2 months of age. This study assesses safety, ammonia control and pharmacokinetics (PK) of GPB in UCD patients below 2 months of age. METHODS: This was an open-label study in UCD patients aged 0 - 2 months, consisting of an initiation/transition period (1 - 4 days) to GPB, followed by a safety extension period (6 months to 2 years). Patients presenting with a hyperammonemic crisis (HAC) did not initiate GPB until blood ammonia levels decreased to below 100 µmol/L while receiving sodium phenylacetate/sodium benzoate and/or hemodialysis. Ammonia levels, PK analytes and safety were evaluated during transition and monthly during the safety extension for 6 months and every 3 months thereafter. RESULTS: All 16 patients with UCD (median age 0.48 months, range 0.1 to 2.0 months) successfully transitioned to GPB within 3 days. Average plasma ammonia level excluding HAC was 94.3 µmol/L at baseline and 50.4 µmol/L at the end of the transition period (p = 0.21). No patient had a HAC during the transition period. During the safety extension, the majority of patients had controlled ammonia levels, with mean plasma ammonia levels lower during GPB treatment than baseline. Mean glutamine levels remained within normal limits throughout the study. PK analyses indicate that UCD patients <2 months are able to hydrolyze GPB with subsequent absorption of phenylbutyric acid (PBA), metabolism to phenylacetic acid (PAA) and conjugation with glutamine. Plasma concentrations of PBA, PAA, and phenylacetylglutamine (PAGN) were stable during the safety extension phase and mean plasma phenylacetic acid: phenylacetylglutamine ratio remained below 2.5 suggesting no accumulation of GPB. All patients reported at least 1 treatment emergent adverse event with gastroesophageal reflux disease, vomiting, hyperammonemia, diaper dermatitis (37.5% each), diarrhea, upper respiratory tract infection and rash (31.3% each) being the most frequently reported. CONCLUSIONS: This study supports safety and efficacy of GPB in UCD patients aged 0 -2 months who cannot be managed by dietary protein restriction and/or amino acid supplementation alone. GPB undergoes intestinal hydrolysis with no accumulation in this population.


Subject(s)
Glycerol/analogs & derivatives , Hyperammonemia/drug therapy , Phenylbutyrates/administration & dosage , Urea Cycle Disorders, Inborn/drug therapy , Age of Onset , Ammonia/blood , Child, Preschool , Female , Glycerol/administration & dosage , Humans , Hyperammonemia/blood , Hyperammonemia/pathology , Infant , Infant, Newborn , Male , Pediatrics , Phenylacetates/administration & dosage , Renal Dialysis , Urea Cycle Disorders, Inborn/blood , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology
13.
Mol Genet Metab ; 131(3): 289-298, 2020 11.
Article in English | MEDLINE | ID: mdl-33317798

ABSTRACT

The mammalian urea cycle (UC) is responsible for siphoning catabolic waste nitrogen into urea for excretion. Disruptions of the functions of any of the enzymes or transporters lead to elevated ammonia and neurological injury. Carbamoyl phosphate synthetase 1 (CPS1) is the first and rate-limiting UC enzyme responsible for the direct incorporation of ammonia into UC intermediates. Symptoms in CPS1 deficiency are typically the most severe of all UC disorders, and current clinical management is insufficient to prevent the associated morbidities and high mortality. With recent advances in basic and translational studies of CPS1, appreciation for this enzyme's essential role in the UC has been broadened to include systemic metabolic regulation during homeostasis and disease. Here, we review recent advances in CPS1 biology and contextualize them around the role of CPS1 in health and disease.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Urea Cycle Disorders, Inborn/genetics , Urea/metabolism , Ammonia/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/pathology , Homeostasis/genetics , Humans , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology
14.
Mol Genet Metab ; 131(4): 390-397, 2020 12.
Article in English | MEDLINE | ID: mdl-33288448

ABSTRACT

OBJECTIVE: The implementation of newborn screening (NBS) programs for citrullinemia type 1 (CTLN1) and argininosuccinic aciduria (ASA) is subject to controversial debate. The aim of this study was to assess the impact of NBS on the metabolic disease course and clinical outcome of affected individuals. METHODS: In 115 individuals with CTLN1 and ASA, we compared the severity of the initial hyperammonemic episode (HAE) and the frequency of (subsequent) HAEs with the mode of diagnosis. Based on a recently established functional disease prediction model, individuals were stratified according to their predicted severe or attenuated phenotype. RESULTS: Individuals with predicted attenuated forms of CTLN1 and ASA were overrepresented in the NBS group, while those with a predicted severe phenotype were underrepresented compared to individuals identified after the manifestation of symptoms (SX). Identification by NBS was associated with reduced severity of the initial HAE both in individuals with predicted severe and attenuated phenotypes, while it was not associated with lower frequency of (subsequent) HAEs. Similar results were obtained when including some patients diagnosed presymptomatically (i.e. prenatal testing, and high-risk family screening) in this analysis. CONCLUSION: Since one of the major challenges of NBS outcome studies is the potential overrepresentation of individuals with predicted attenuated phenotypes in NBS cohorts, severity-adjusted evaluation of screened and unscreened individuals is important to avoid overestimation of the NBS effect. NBS enables the attenuation of the initial HAE but does not affect the frequency of subsequent metabolic decompensations in individuals with CTLN1 and ASA. Future long-term studies will need to evaluate the clinical impact of this finding, especially with regard to mortality, as well as cognitive outcome and quality of life of survivors.


Subject(s)
Argininosuccinic Aciduria/diagnosis , Citrullinemia/diagnosis , Metabolic Diseases/genetics , Urea Cycle Disorders, Inborn/diagnosis , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/metabolism , Argininosuccinic Aciduria/pathology , Citrullinemia/genetics , Citrullinemia/metabolism , Citrullinemia/pathology , Female , Humans , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/pathology , Infant, Newborn , Male , Metabolic Diseases/diagnosis , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Neonatal Screening , Quality of Life , Severity of Illness Index , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology
15.
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
16.
Nat Metab ; 2(5): 432-446, 2020 05.
Article in English | MEDLINE | ID: mdl-32694660

ABSTRACT

Chronic inflammation is linked to diverse disease processes, but the intrinsic mechanisms that determine cellular sensitivity to inflammation are incompletely understood. Here, we show the contribution of glucose metabolism to inflammation-induced changes in the survival of pancreatic islet ß-cells. Using metabolomic, biochemical and functional analyses, we investigate the protective versus non-protective effects of glucose in the presence of pro-inflammatory cytokines. When protective, glucose metabolism augments anaplerotic input into the TCA cycle via pyruvate carboxylase (PC) activity, leading to increased aspartate levels. This metabolic mechanism supports the argininosuccinate shunt, which fuels ureagenesis from arginine and conversely diminishes arginine utilization for production of nitric oxide (NO), a chief mediator of inflammatory cytotoxicity. Activation of the PC-urea cycle axis is sufficient to suppress NO synthesis and shield cells from death in the context of inflammation and other stress paradigms. Overall, these studies uncover a previously unappreciated link between glucose metabolism and arginine-utilizing pathways via PC-directed ureagenesis as a protective mechanism.


Subject(s)
Arginine/metabolism , Glucose/metabolism , Glucose/pharmacology , Inflammation/prevention & control , Insulin-Secreting Cells/drug effects , Urea Cycle Disorders, Inborn/pathology , Urea/metabolism , Adolescent , Adult , Aged , Aspartic Acid/metabolism , Cell Survival , Citric Acid Cycle/drug effects , Female , Humans , Inflammation/pathology , Insulin-Secreting Cells/pathology , Male , Metabolomics , Middle Aged , Nitric Oxide/metabolism , Pyruvate Carboxylase/metabolism , Urea Cycle Disorders, Inborn/metabolism , Young Adult
17.
Nat Rev Nephrol ; 16(8): 471-482, 2020 08.
Article in English | MEDLINE | ID: mdl-32269302

ABSTRACT

Hyperammonaemia in children can lead to grave consequences in the form of cerebral oedema, severe neurological impairment and even death. In infants and children, common causes of hyperammonaemia include urea cycle disorders or organic acidaemias. Few studies have assessed the role of extracorporeal therapies in the management of hyperammonaemia in neonates and children. Moreover, consensus guidelines are lacking for the use of non-kidney replacement therapy (NKRT) and kidney replacement therapies (KRTs, including peritoneal dialysis, continuous KRT, haemodialysis and hybrid therapy) to manage hyperammonaemia in neonates and children. Prompt treatment with KRT and/or NKRT, the choice of which depends on the ammonia concentrations and presenting symptoms of the patient, is crucial. This expert Consensus Statement presents recommendations for the management of hyperammonaemia requiring KRT in paediatric populations. Additional studies are required to strengthen these recommendations.


Subject(s)
Continuous Renal Replacement Therapy/methods , Hyperammonemia/therapy , Peritoneal Dialysis/methods , Urea Cycle Disorders, Inborn/therapy , Arginine/therapeutic use , Carnitine/therapeutic use , Child , Child, Preschool , Delphi Technique , Diet, Protein-Restricted , Humans , Hybrid Renal Replacement Therapy , Hyperammonemia/metabolism , Infant , Infant, Newborn , Parenteral Nutrition/methods , Phenylacetates/therapeutic use , Phenylbutyrates/therapeutic use , Practice Guidelines as Topic , Renal Dialysis/methods , Sodium Benzoate/therapeutic use , Urea Cycle Disorders, Inborn/metabolism , Vitamin B Complex/therapeutic use
18.
Soft Matter ; 16(11): 2725-2735, 2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32115597

ABSTRACT

Transmembrane pH gradient poly(isoprene)-block-poly(ethylene glycol) (PI-b-PEG) polymersomes were investigated for their potential use in the detoxification of ammonia, a metabolite that is excessively present in patients suffering from urea cycle disorders and advanced liver diseases, and which causes neurotoxic effects (e.g., hepatic encephalopathy). Polymers varying in PI and PEG block length were synthesized via nitroxide-mediated polymerization and screened for their ability to self-assemble into polymersomes in aqueous media. Ammonia sequestration by the polymersomes was investigated in vitro. While most vesicular systems were able to capture ammonia in simulated intestinal fluids, uptake was lost in partially dehydrated medium mimicking conditions in the colon. Polymeric crosslinking of residual olefinic bonds in the PI block increased polymersome stability, partially preserving the ammonia capture capacity in the simulated colon environment. These more stable vesicular systems hold promise for the chronic oral treatment of hyperammonemia.


Subject(s)
Ammonia/chemistry , Drug Carriers/chemistry , Hepatic Encephalopathy/drug therapy , Inactivation, Metabolic/genetics , Ammonia/metabolism , Butadienes/chemistry , Butadienes/pharmacology , Drug Carriers/pharmacology , Fluorescein-5-isothiocyanate/chemistry , Hemiterpenes/chemistry , Hemiterpenes/pharmacology , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/metabolism , Humans , Hydrogen-Ion Concentration , Liver Diseases/complications , Liver Diseases/drug therapy , Liver Diseases/metabolism , Methacrylates/chemistry , Particle Size , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polymerization , Polymers/chemistry , Polymers/pharmacology , Proton-Motive Force/drug effects , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/metabolism , Water/metabolism
19.
Int J Mol Sci ; 22(1)2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33396517

ABSTRACT

Osteopetrosis is a hereditary disorder characterized by sclerotic, thick, weak, and brittle bone. The biological behavior of mesenchymal cells obtained from osteopetrosis patients has not been well-studied. Isolated mesenchymal stem/stromal cells from dental pulp (DP-MSSCs) of recently extracted deciduous teeth from osteopetrosis (OP) patients and healthy controls (HCs) were compared. We evaluated whether the dental pulp of OP patients has a population of MSSCs with similar multilineage differentiation capability to DP-MSSCs of healthy subjects. Stem/progenitor cells were characterized using immunohistochemistry, flow cytometry, and proteomics. Our DP-MSSCs were strongly positive for CD44, CD73, CD105, and CD90. DP-MSSCs obtained from HC subjects and OP patients showed similar patterns of proliferation and differentiation as well as gene expression. Proteomic analysis identified 1499 unique proteins with 94.3% similarity in global protein fingerprints of HCs and OP patients. Interestingly, we observed subtle differences in expressed proteins of osteopetrosis disease-related in pathways, including MAPK, ERK 1/2, PI3K, and integrin, rather than in the stem cell signaling network. Our findings of similar protein expression signatures in DP-MSSCs of HC and OP patients are of paramount interest, and further in vivo validation study is needed. There is the possibility that OP patients could have their exfoliating deciduous teeth banked for future use in regenerative dentistry.


Subject(s)
Acidosis, Renal Tubular/metabolism , Acidosis, Renal Tubular/pathology , Biomarkers/metabolism , Carbonic Anhydrases/deficiency , Dental Pulp/metabolism , Mesenchymal Stem Cells/metabolism , Osteopetrosis/metabolism , Osteopetrosis/pathology , Proteome/analysis , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/pathology , Adolescent , Biomarkers/analysis , Carbonic Anhydrases/metabolism , Case-Control Studies , Cell Differentiation , Cell Proliferation , Child , Dental Pulp/cytology , Female , Humans , Male , Mesenchymal Stem Cells/cytology
20.
Cell Rep ; 29(8): 2144-2153.e7, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31747589

ABSTRACT

Patients with germline mutations in the urea-cycle enzyme argininosuccinate lyase (ASL) are at risk for developing neurobehavioral and cognitive deficits. We find that ASL is prominently expressed in the nucleus locus coeruleus (LC), the central source of norepinephrine. Using natural history data, we show that individuals with ASL deficiency are at risk for developing attention deficits. By generating LC-ASL-conditional knockout (cKO) mice, we further demonstrate altered response to stressful stimuli with increased seizure reactivity in LC-ASL-cKO mice. Depletion of ASL in LC neurons leads to reduced amount and activity of tyrosine hydroxylase (TH) and to decreased catecholamines synthesis, due to decreased nitric oxide (NO) signaling. NO donors normalize catecholamine levels in the LC, seizure sensitivity, and the stress response in LC-ASL-cKO mice. Our data emphasize ASL importance for the metabolic regulation of LC function with translational relevance for ASL deficiency (ASLD) patients as well as for LC-related pathologies.


Subject(s)
Argininosuccinate Lyase/metabolism , Locus Coeruleus/metabolism , Tyrosine 3-Monooxygenase/metabolism , Urea Cycle Disorders, Inborn/metabolism , Animals , Catecholamines/metabolism , Cell Nucleus/metabolism , Mice , Mice, Knockout , Nitric Oxide/metabolism , Seizures/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...