Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 546(7656): 168-172, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28538732

ABSTRACT

Metabolic reprogramming by oncogenic signals promotes cancer initiation and progression. The oncogene KRAS and tumour suppressor STK11, which encodes the kinase LKB1, regulate metabolism and are frequently mutated in non-small-cell lung cancer (NSCLC). Concurrent occurrence of oncogenic KRAS and loss of LKB1 (KL) in cells specifies aggressive oncological behaviour. Here we show that human KL cells and tumours share metabolomic signatures of perturbed nitrogen handling. KL cells express the urea cycle enzyme carbamoyl phosphate synthetase-1 (CPS1), which produces carbamoyl phosphate in the mitochondria from ammonia and bicarbonate, initiating nitrogen disposal. Transcription of CPS1 is suppressed by LKB1 through AMPK, and CPS1 expression correlates inversely with LKB1 in human NSCLC. Silencing CPS1 in KL cells induces cell death and reduces tumour growth. Notably, cell death results from pyrimidine depletion rather than ammonia toxicity, as CPS1 enables an unconventional pathway of nitrogen flow from ammonia into pyrimidines. CPS1 loss reduces the pyrimidine to purine ratio, compromises S-phase progression and induces DNA-polymerase stalling and DNA damage. Exogenous pyrimidines reverse DNA damage and rescue growth. The data indicate that the KL oncological genotype imposes a metabolic vulnerability related to a dependence on a cross-compartmental pathway of pyrimidine metabolism in an aggressive subset of NSCLC.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , DNA/biosynthesis , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Ammonia/metabolism , Animals , Bicarbonates/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamyl Phosphate/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Death , Cell Proliferation , DNA Damage/drug effects , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Female , Gene Silencing , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Metabolomics , Mice , Mitochondria/metabolism , Nitrogen/metabolism , Protein Serine-Threonine Kinases/metabolism , Purines/metabolism , Pyrimidines/pharmacology , S Phase , Transcription, Genetic , Xenograft Model Antitumor Assays
2.
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
3.
J Inherit Metab Dis ; 42(6): 1044-1053, 2019 11.
Article in English | MEDLINE | ID: mdl-30835861

ABSTRACT

The enzyme carbamoyl phosphate synthetase 1 (CPS1; EC 6.3.4.16) forms carbamoyl phosphate from bicarbonate, ammonia, and adenosine triphosphate (ATP) and is activated allosterically by N-acetylglutamate. The neonatal presentation of bi-allelic mutations of CPS1 results in hyperammonemia with reduced citrulline and is reported as the most challenging nitrogen metabolism disorder to treat. As therapeutic interventions are limited, patients often develop neurological injury or die from hyperammonemia. Survivors remain vulnerable to nitrogen overload, being at risk for repetitive neurological injury. With transgenic technology, our lab developed a constitutive Cps1 mutant mouse and reports its characterization herein. Within 24 hours of birth, all Cps1 -/- mice developed hyperammonemia and expired. No CPS1 protein by Western blot or immunostaining was detected in livers nor was Cps1 mRNA present. CPS1 enzymatic activity was markedly decreased in knockout livers and reduced in Cps1+/- mice. Plasma analysis found markedly reduced citrulline and arginine and markedly increased glutamine and alanine, both intermolecular carriers of nitrogen, along with elevated ammonia, taurine, and lysine. Derangements in multiple other amino acids were also detected. While hepatic amino acids also demonstrated markedly reduced citrulline, arginine, while decreased, was not statistically significant; alanine and lysine were markedly increased while glutamine was trending towards significance. In conclusion we have determined that this constitutive neonatal mouse model of CPS1 deficiency replicates the neonatal human phenotype and demonstrates the key biochemical features of the disorder. These mice will be integral for addressing the challenges of developing new therapeutic approaches for this, at present, poorly treated disorder.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/complications , Carbamoyl-Phosphate Synthase I Deficiency Disease/mortality , Glutamine/blood , Hyperammonemia , Animals , Animals, Newborn , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase I Deficiency Disease/blood , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Hyperammonemia/blood , Hyperammonemia/complications , Hyperammonemia/genetics , Hyperammonemia/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation
4.
Mol Genet Metab ; 119(4): 307-310, 2016 12.
Article in English | MEDLINE | ID: mdl-27771289

ABSTRACT

BACKGROUND: N-acetylglutamate synthase (NAGS) plays a key role in the removal of ammonia via the urea cycle by catalyzing the synthesis of N-acetylglutamate (NAG), the obligatory cofactor in the carbamyl phosphate synthetase 1 reaction. Enzymatic analysis of NAGS in liver homogenates has remained insensitive and inaccurate, which prompted the development of a novel method. METHODS: UPLC-MS/MS was used in conjunction with stable isotope (N-acetylglutamic-2,3,3,4,4-d5 acid) dilution for the quantitative detection of NAG produced by the NAGS enzyme. The assay conditions were optimized using purified human NAGS and the optimized enzyme conditions were used to measure the activity in mouse liver homogenates. RESULTS: A low signal-to-noise ratio in liver tissue samples was observed due to non-enzymatic formation of N-acetylglutamate and low specific activity, which interfered with quantitative analysis. Quenching of acetyl-CoA immediately after the incubation circumvented this analytical difficulty and allowed accurate and sensitive determination of mammalian NAGS activity. The specificity of the assay was validated by demonstrating a complete deficiency of NAGS in liver homogenates from Nags -/- mice. CONCLUSION: The novel NAGS enzyme assay reported herein can be used for the diagnosis of inherited NAGS deficiency and may also be of value in the study of secondary hyperammonemia present in various inborn errors of metabolism as well as drug treatment.


Subject(s)
Amino-Acid N-Acetyltransferase/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Hyperammonemia/diagnosis , Urea Cycle Disorders, Inborn/diagnosis , Acetyl Coenzyme A/metabolism , Amino-Acid N-Acetyltransferase/metabolism , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Humans , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/physiopathology , Liver/enzymology , Mice , Mice, Knockout , Tandem Mass Spectrometry , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/physiopathology
5.
Mol Genet Metab ; 114(3): 438-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25639153

ABSTRACT

Fatal hyperammonemia secondary to chemotherapy for hematological malignancies or following bone marrow transplantation has been described in few patients so far. In these, the pathogenesis of hyperammonemia remained unclear and was suggested to be multifactorial. We observed severe hyperammonemia (maximum 475 Āµmol/L) in a 2-year-old male patient, who underwent high-dose chemotherapy with carboplatin, etoposide and melphalan, and autologous hematopoietic stem cell transplantation for a neuroblastoma stage IV. Despite intensive care treatment, hyperammonemia persisted and the patient died due to cerebral edema. The biochemical profile with elevations of ammonia and glutamine (maximum 1757 Āµmol/L) suggested urea cycle dysfunction. In liver homogenates, enzymatic activity and protein expression of the urea cycle enzyme carbamoyl phosphate synthetase 1 (CPS1) were virtually absent. However, no mutation was found in CPS1 cDNA from liver and CPS1 mRNA expression was only slightly decreased. We therefore hypothesized that the acute onset of hyperammonemia was due to an acquired, chemotherapy-induced (posttranscriptional) CPS1 deficiency. This was further supported by in vitro experiments in HepG2 cells treated with carboplatin and etoposide showing a dose-dependent decrease in CPS1 protein expression. Due to severe hyperlactatemia, we analysed oxidative phosphorylation complexes in liver tissue and found reduced activities of complexes I and V, which suggested a more general mitochondrial dysfunction. This study adds to the understanding of chemotherapy-induced hyperammonemia as drug-induced CPS1 deficiency is suggested. Moreover, we highlight the need for urgent diagnostic and therapeutic strategies addressing a possible secondary urea cycle failure in future patients with hyperammonemia during chemotherapy and stem cell transplantation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Hematopoietic Stem Cell Transplantation/adverse effects , Hyperammonemia/etiology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Edema/etiology , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamyl Phosphate/metabolism , Carboplatin/adverse effects , Carboplatin/pharmacology , Child, Preschool , Combined Modality Therapy , Etoposide/adverse effects , Etoposide/pharmacology , Fatal Outcome , Glutamine/blood , Hep G2 Cells , Humans , Hyperammonemia/chemically induced , Liver/enzymology , Liver/metabolism , Male , Middle Aged , Neuroblastoma/drug therapy , Ornithine Carbamoyltransferase/genetics , Oxidative Phosphorylation
6.
Hum Mutat ; 34(8): 1149-59, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23649895

ABSTRACT

The urea cycle disease carbamoyl-phosphate synthetase deficiency (CPS1D) has been associated with many mutations in the CPS1 gene [HƤberle et al., 2011. Hum Mutat 32:579-589]. The disease-causing potential of most of these mutations is unclear. To test the mutations effects, we have developed a system for recombinant expression, mutagenesis, and purification of human carbamoyl-phosphate synthetase 1 (CPS1), a very large, complex, and fastidious enzyme. The kinetic and molecular properties of recombinant CPS1 are essentially the same as for natural human CPS1. Glycerol partially replaces the essential activator N-acetyl-l-glutamate (NAG), opening possibilities for treating CPS1D due to NAG site defects. The value of our expression system for elucidating the effects of mutations is demonstrated with eight clinical CPS1 mutations. Five of these mutations decreased enzyme stability, two mutations drastically hampered catalysis, and one vastly impaired NAG activation. In contrast, the polymorphisms p.Thr344Ala and p.Gly1376Ser had no detectable effects. Site-limited proteolysis proved the correctness of the working model for the human CPS1 domain architecture generally used for rationalizing the mutations effects. NAG and its analogue and orphan drug N-carbamoyl-l-glutamate, protected human CPS1 against proteolytic and thermal inactivation in the presence of MgATP, raising hopes of treating CPS1D by chemical chaperoning with N-carbamoyl-l-glutamate.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/chemistry , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Mutation, Missense , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/etiology , Carbamoyl-Phosphate Synthase I Deficiency Disease/pathology , Humans , Mutagenesis , Polymorphism, Genetic , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
7.
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
8.
Clin Chim Acta ; 526: 55-61, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34973183

ABSTRACT

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.


Subject(s)
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
9.
Liver Transpl ; 17(12): 1481-4, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21837743

ABSTRACT

Urea cycle disorders (UCDs) are rare causes of hyperammonemic encephalopathy in adults. Most UCDs present in childhood and, if unrecognized, are rapidly fatal. Affected individuals who survive to adulthood may remain undiagnosed because of clinicians' unawareness of the condition or atypical presentations. We describe the case of a 49-year-old man who initially presented with a stroke and developed hyperammonemic encephalopathy over a period of 8 months. A diagnosis of carbamoyl phosphate synthetase type 1 deficiency was made, and the patient was referred for liver transplantation. One year after liver transplantation, the patient had normal plasma ammonia concentrations and had returned to work.


Subject(s)
Liver Transplantation , Urea Cycle Disorders, Inborn/surgery , Age of Onset , Brain Diseases, Metabolic/etiology , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase I Deficiency Disease , Humans , Hyperammonemia/etiology , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Stroke/etiology , Treatment Outcome , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/enzymology
10.
Anesth Analg ; 113(4): 858-61, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21821508

ABSTRACT

We present a case of significant deterioration of chronic hyperammonemia after general anesthesia for neurosurgery despite aggressive treatment. Preoperative evaluation demonstrated that hyperammonemia was most likely related to valproic acid treatment. Genomic analysis revealed that the patient was heterozygotic for a missense polymorphism in the carbamoyl phosphate synthase 1 gene (4217C>A, rs1047891). This mutation was previously suggested to be associated with chronic hyperammonemia. Replacement of threonine with asparagine decreases the activity of carbamoyl phosphate synthase in the urea cycle. Genetic screening can potentially identify a population at risk before initiation of antiepileptic therapy.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Hyperammonemia/etiology , Neurosurgical Procedures/adverse effects , Urea Cycle Disorders, Inborn/genetics , Valproic Acid/adverse effects , Adult , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase I Deficiency Disease , Chronic Disease , DNA Mutational Analysis , Device Removal , Electric Power Supplies , Genetic Predisposition to Disease , Heterozygote , Humans , Hyperammonemia/chemically induced , Hyperammonemia/enzymology , Hyperammonemia/genetics , Male , Mutation, Missense , Polymorphism, Single Nucleotide , Risk Factors , Urea Cycle Disorders, Inborn/complications , Vagus Nerve Stimulation/instrumentation
11.
Mol Genet Metab ; 101(4): 311-23, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20800523

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) plays a paramount role in liver ureagenesis since it catalyzes the first and rate-limiting step of the urea cycle, the major pathway for nitrogen disposal in humans. CPS1 deficiency (CPS1D) is an autosomal recessive inborn error which leads to hyperammonemia due to mutations in the CPS1 gene, or is caused secondarily by lack of its allosteric activator NAG. Proteolytic, immunological and structural data indicate that human CPS1 resembles Escherichia coli CPS in structure, and a 3D model of CPS1 has been presented for elucidating the pathogenic role of missense mutations. Recent availability of CPS1 expression systems also can provide valuable tools for structure-function analysis and pathogenicity-testing of mutations in CPS1. In this paper, we provide a comprehensive compilation of clinical CPS1 mutations, and discuss how structural knowledge of CPS enzymes in combination with in vitro analyses can be a useful tool for diagnosis of CPS1D.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/chemistry , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease , Humans , Protein Structure, Tertiary , Urea Cycle Disorders, Inborn/enzymology , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/pathology
12.
Am J Obstet Gynecol ; 203(1): e10-1, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20471629

ABSTRACT

Carbamyl phosphate synthetase (CPS) is an enzyme that converts ammonia to carbamyl phosphate in the urea cycle. CPS deficiency is a genetic disorder that causes hyperammonemia because of enzyme activity deficiency. It is primarily diagnosed in neonates and infants and has a poor prognosis. We report an adult woman with CPS deficiency who developed hyperammonemia postpartum.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Hyperammonemia/enzymology , Postpartum Period/blood , Adult , Ammonia/blood , Female , Humans , Hyperammonemia/blood , Hyperammonemia/diagnosis , Hyperammonemia/therapy , Pregnancy , Renal Dialysis
13.
Hum Mutat ; 30(1): 56-60, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18666241

ABSTRACT

The urea cycle is the primary means of nitrogen metabolism in humans and other ureotelic organisms. There are five key enzymes in the urea cycle: carbamoyl-phosphate synthetase 1 (CPS1), ornithine transcarbamylase (OTC), argininosuccinate synthetase (ASS1), argininosuccinate lyase (ASL), and arginase 1 (ARG1). Additionally, a sixth enzyme, N-acetylglutamate synthase (NAGS), is critical for urea cycle function, providing CPS1 with its necessary cofactor. Deficiencies in any of these enzymes result in elevated blood ammonia concentrations, which can have detrimental effects, including central nervous system dysfunction, brain damage, coma, and death. Functional variants, which confer susceptibility for disease or dysfunction, have been described for enzymes within the cycle; however, a comprehensive screen of all the urea cycle enzymes has not been performed. We examined the exons and intron/exon boundaries of the five key urea cycle enzymes, NAGS, and two solute carrier transporter genes (SLC25A13 and SLC25A15) for sequence alterations using single-stranded conformational polymorphism (SSCP) analysis and high-resolution melt profiling. SSCP was performed on a set of DNA from 47 unrelated North American individuals with a mixture of ethnic backgrounds. High-resolution melt profiling was performed on a nonoverlapping DNA set of either 47 or 100 unrelated individuals with a mixture of backgrounds. We identified 33 unarchived polymorphisms in this screen that potentially play a role in the variation observed in urea cycle function. Screening all the genes in the pathway provides a catalog of variants that can be used in investigating candidate diseases.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Urea/metabolism , Ammonia/blood , Arginase/genetics , Argininosuccinate Lyase/genetics , Argininosuccinate Synthase/deficiency , Argininosuccinate Synthase/genetics , Argininosuccinic Aciduria , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Humans , Hyperargininemia , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase Deficiency Disease , Polymorphism, Single-Stranded Conformational
14.
Science ; 207(4431): 659-61, 1980 Feb 08.
Article in English | MEDLINE | ID: mdl-6243418

ABSTRACT

Treatment of a patient deficient in carbamyl phosphate synthetase with benzoate or phenylacetic acid resulted in an increase in urinary nitrogen, which could be accounted for by the respective amino acid acylation product, hippurate or phenylacetylgultamine. Benzoate treatment of four hyperammonemic comatose patients led to clinical improvement and a return of plasma ammonium levels toward normal.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Benzoates/therapeutic use , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Ornithine Carbamoyltransferase Deficiency Disease , Phenylacetates/therapeutic use , Phosphotransferases/deficiency , Adolescent , Amino Acids/blood , Ammonia/blood , Child , Female , Glutamine/metabolism , Glycine/metabolism , Hippurates/urine , Humans , Infant , Male , Nitrogen/blood
15.
J Clin Invest ; 74(6): 2144-8, 1984 Dec.
Article in English | MEDLINE | ID: mdl-6511918

ABSTRACT

The role of arginine as an essential amino was evaluated in four children with one of the deficiencies of carbamyl phosphate synthetase, ornithine transcarbamylase, argininosuccinate synthetase, and argininosuccinase. Within 15-68 h after arginine deprivation nitrogen accumulated as ammonium or glutamine or both, but glutamine was quantitatively the largest nitrogen accumulation product. Concomitantly plasma and urinary urea levels decreased. Resumption of arginine intake (or citrulline in the case of ornithine transcarbamylase deficiency) promptly led to correction of the hyperammonemia, hyperglutaminemia and hypoargininemia. Ornithine was an unsatisfactory substitute for arginine. Arginine deprivation did not interfere with carbamyl phosphate synthesis as manifested by orotic aciduria. It is concluded that arginine is an indispensable amino acid for children with inborn errors of ureagenesis and its absence results in the rapid onset of symptomatic hyperammonemia.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Arginine/physiology , Urea/biosynthesis , Ammonia/blood , Arginine/therapeutic use , Argininosuccinate Synthase/deficiency , Argininosuccinic Aciduria , Blood Urea Nitrogen , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Child, Preschool , Female , Glutamine/blood , Humans , Infant , Infant, Newborn , Male , Ornithine/blood , Ornithine Carbamoyltransferase Deficiency Disease
16.
J Clin Invest ; 91(5): 1884-7, 1993 May.
Article in English | MEDLINE | ID: mdl-8486760

ABSTRACT

Carbamyl phosphate synthetase I (CPS I; EC6,3,4,16) is an autosomal recessive disorder characterized by hyperammonemia. We studied the molecular bases of CPS I deficiency in a newborn Japanese girl with consanguineous parents. Northern and Western blots revealed a marked decrease in CPS I mRNA and enzyme protein but with a size similar to that of the control, respectively. Sequencing of the patient's cDNA revealed a nine-nucleotide deletion at position 832-840. Sequencing analysis of the genomic DNA revealed a G to C transversion at position 840, the last nucleotide of an exon in the splice donor site. This substitution altered the consensus sequence of the splice donor site and the newly cryptical donor site in the exon caused the 9-bp in-frame deletion. This report seems to be the first complete definition of CPS I deficiency, at the molecular level.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Liver/enzymology , RNA Splicing , RNA, Messenger/genetics , Sequence Deletion , Amino Acid Sequence , Base Composition , Base Sequence , Blotting, Northern , Blotting, Western , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , DNA/genetics , DNA/isolation & purification , Exons , Female , Humans , Infant, Newborn , Male , Molecular Sequence Data , Oligodeoxyribonucleotides , Pedigree , Polymerase Chain Reaction/methods , RNA, Messenger/metabolism
17.
Gene ; 107(2): 335-40, 1991 Nov 15.
Article in English | MEDLINE | ID: mdl-1840546

ABSTRACT

Carbamyl phosphate synthetase I (CPSI) is the first enzyme involved in urea synthesis. CPSI deficiency is an autosomal recessive disorder characterized by hyperammonemic coma in the neonatal period. To analyze the enzyme and gene structures, and to elucidate the nature of mutations in CPSI deficiency, we isolated cDNA clones encoding human liver CPSI. Oligo(dT)-primed and random primer human liver cDNA libraries in lambda gt11 were screened using 5', middle, and 3' fragments of the rat CPSI cDNA as probes. Seven positive clones covered the full-length cDNA sequence with an open reading frame encoding a precursor polypeptide of 1500 amino acids (aa) (deduced Mr, 164,828) with a putative N-terminal presequence of 38 or 39 aa, a 5'-untranslated sequence of 118 bp and a 3'-untranslated sequence of 597 bp. Comparison with the rat CPSI cDNA showed that the deduced aa sequence of the human liver CPSI precursor is 94.4% identical to the rat enzyme precursor. A molecular analysis was made of the genomic DNA from three patients with CPSI deficiency. Heterogeneity of hybridized fragments that may or may not be the cause of the deficiency was apparent on the DNA blots from tissues from one patient.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Ammonia/blood , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Mitochondria/enzymology , Amino Acid Sequence , Base Sequence , Blotting, Southern , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Cloning, Molecular , Female , Genes, Recessive/genetics , Humans , Male , Molecular Sequence Data , Mutation/genetics
18.
Arch Neurol ; 41(7): 758-60, 1984 Jul.
Article in English | MEDLINE | ID: mdl-6743067

ABSTRACT

Inborn errors of ureagenesis must be considered in the differential diagnosis of recurrent vomiting and lethargy in childhood. Elevations of liver enzyme levels are often present during these episodes and may lead to an erroneous diagnosis of hepatic encephalopathy. We studied two cases of urea cycle defects.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Urea/metabolism , Amino Acid Metabolism, Inborn Errors/enzymology , Ammonia/blood , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Child, Preschool , Female , Humans , Infant, Newborn , Male , Ornithine Carbamoyltransferase Deficiency Disease
20.
Pediatrics ; 58(2): 227-35, 1976 Aug.
Article in English | MEDLINE | ID: mdl-181726

ABSTRACT

A 13-year-old girl with congenital deficiency of carbamyl phosphate synthetase has been treated intermittently for one year with a restricted protein diet supplemented by various mixtures of the alpha-ketoanalogues of valine, leucine, isoleucine, and phenylalanine, the D,L-alpha-hydroxyanalogue of methionine, and five amino acids (lysine, arginine, histidine, threonine and tryptophan). Numerous adjustments in the composition of this mixture were made. Eventually normal levels of plasma ammonia and most amino acids were achieved, with three exceptions: slightly increased glutamine, pronounced alloisoleucinemia, and persistently low phenylalanine. Alloisoleucine was shown not to be incorporated into plasma protein and not to be excreted in the urine; hence this abnormality was viewed as being clinically insiginificant. Hypophenylalaninemia was unexplained, and failed to respond to increased phenylpyruvate dosage or phenylalanine itself; renal clearance of phenylalanine was high but could not account for the low plasma level. Compared to the pretreatment period her clinical status has improved markedly. Physical and mental development has continued at the same rate. Temporary withdrawal of the supplements led to prompt increases in plasma ammonia, glutamine, and alanine. We conclude that this therapy provides safe and effective long-term management for this patient's disorder and may be useful in other cases of congenital hyperammonemia.


Subject(s)
Amino Acids/therapeutic use , Carbamoyl-Phosphate Synthase (Ammonia)/deficiency , Phosphotransferases/deficiency , Adolescent , Arginine/analogs & derivatives , Dietary Proteins , Female , Histidine/analogs & derivatives , Humans , Isoleucine/therapeutic use , Long-Term Care , Lysine/analogs & derivatives , Methionine/therapeutic use , Phenylalanine/therapeutic use , Threonine/analogs & derivatives , Tryptophan/analogs & derivatives , Valine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL