Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Hum Mol Genet ; 32(11): 1922-1931, 2023 05 18.
Article in English | MEDLINE | ID: mdl-36881658

ABSTRACT

Citrin deficiency (CD) is an inborn error of metabolism caused by loss-of-function of the mitochondrial aspartate/glutamate transporter, CITRIN, which is involved in both the urea cycle and malate-aspartate shuttle. Patients with CD develop hepatosteatosis and hyperammonemia but there is no effective therapy for CD. Currently, there are no animal models that faithfully recapitulate the human CD phenotype. Accordingly, we generated a CITRIN knockout HepG2 cell line using Clustered Regularly Interspaced Short Palindromic Repeats/Cas 9 genome editing technology to study metabolic and cell signaling defects in CD. CITRIN KO cells showed increased ammonia accumulation, higher cytosolic ratio of reduced versus oxidized form of nicotinamide adenine dinucleotide (NAD) and reduced glycolysis. Surprisingly, these cells showed impaired fatty acid metabolism and mitochondrial activity. CITRIN KO cells also displayed increased cholesterol and bile acid metabolism resembling those observed in CD patients. Remarkably, normalizing cytosolic NADH:NAD+ ratio by nicotinamide riboside increased glycolysis and fatty acid oxidation but had no effect on the hyperammonemia suggesting the urea cycle defect was independent of the aspartate/malate shuttle defect of CD. The correction of glycolysis and fatty acid metabolism defects in CITRIN KO cells by reducing cytoplasmic NADH:NAD+ levels suggests this may be a novel strategy to treat some of the metabolic defects of CD and other mitochondrial diseases.


Subject(s)
Citrullinemia , Hyperammonemia , Humans , Citrullinemia/genetics , Citrullinemia/metabolism , NAD/metabolism , Malates , Aspartic Acid/metabolism , Hyperammonemia/genetics , Mitochondrial Membrane Transport Proteins/genetics , Hepatocytes/metabolism , Glycolysis , Urea/metabolism , Fatty Acids
2.
Trends Endocrinol Metab ; 33(8): 539-553, 2022 08.
Article in English | MEDLINE | ID: mdl-35725541

ABSTRACT

Citrin deficiency is a pan-ethnic and highly prevalent mitochondrial disease with three different stages: neonatal intrahepatic cholestasis (NICCD), a relatively mild adaptation stage, and type II citrullinemia in adulthood (CTLN2). The cause is the absence or dysfunction of the calcium-regulated mitochondrial aspartate/glutamate carrier 2 (AGC2/SLC25A13), also called citrin, which imports glutamate into the mitochondrial matrix and exports aspartate to the cytosol. In citrin deficiency, these missing transport steps lead to impairment of the malate-aspartate shuttle, gluconeogenesis, amino acid homeostasis, and the urea cycle. In this review, we describe the geological spread and occurrence of citrin deficiency, the metabolic consequences and use our current knowledge of the structure to predict the impact of the known pathogenic mutations on the calcium-regulatory and transport mechanism of citrin.


Subject(s)
Citrullinemia , Adult , Aspartic Acid/genetics , Calcium , Citrullinemia/genetics , Citrullinemia/metabolism , Glutamates/genetics , Humans , Infant, Newborn , Mitochondrial Membrane Transport Proteins/genetics , Mutation
3.
Mol Genet Metab ; 133(1): 63-70, 2021 05.
Article in English | MEDLINE | ID: mdl-33741270

ABSTRACT

Patients with citrin deficiency during the adaptation/compensation period exhibit diverse clinical features and have characteristic diet of high protein, high fat, and low carbohydrate. Japanese cuisine typically contains high carbohydrate but evaluation of diet of citrin-deficient patients in 2008 showed a low energy intake and a protein:fat:carbohydrate (PFC) ratio of 19:44:37, which indicates low carbohydrate consumption rate. These findings prompted the need for diet intervention to prevent the adult onset of type II citrullinemia (CTLN2). Since the publication of the report about 10 years ago, patients are generally advised to eat what they wish under active dietary consultation and intervention. In this study, citrin-deficient patients and control subjects living in the same household provided answers to a questionnaire, filled-up a maximum 6-day food diary, and supplied physical data and information on medications if any. To study the effects of the current diet, the survey collected data from 62 patients and 45 controls comparing daily intakes of energy, protein, fat, and carbohydrate. Food analysis showed that patient's energy intake was 115% compared to the Japanese standard. The confidence interval of the PFC ratio of patients was 20-22:47-51:28-32, indicating higher protein, higher fat and lower carbohydrate relative to previous reports. The mean PFC ratio of female patients (22:53:25) was significantly different from that of male patients (20:46:34), which may explain the lower frequency of CTLN2 in females. Comparison of the present data to those published 10 years ago, energy, protein, and fat intakes were significantly higher but the amount of carbohydrate consumption remained the same. Regardless of age, most patients (except for adolescents) consumed 100-200 g/day of carbohydrates, which met the estimated average requirement of 100 g/day for healthy individuals. Finally, patients were generally not overweight and some CTLN2 patients were underweight although their energy intake was higher compared with the control subjects. We speculate that high-energy of a low carbohydrate diet under dietary intervention may help citrin-deficient patients attain normal growth and prevent the onset of CTLN2.


Subject(s)
Calcium-Binding Proteins/genetics , Citrullinemia/diet therapy , Energy Metabolism/physiology , Organic Anion Transporters/genetics , Adolescent , Adult , Calcium-Binding Proteins/deficiency , Carbohydrate Metabolism/physiology , Carbohydrates/administration & dosage , Citrullinemia/epidemiology , Citrullinemia/metabolism , Citrullinemia/pathology , Dietary Fats/administration & dosage , Dietary Fats/metabolism , Eating/physiology , Female , Humans , Japan/epidemiology , Male , Mitochondrial Membrane Transport Proteins/genetics , Organic Anion Transporters/deficiency , Proteins/administration & dosage , Proteins/metabolism
4.
Metab Brain Dis ; 36(4): 685-699, 2021 04.
Article in English | MEDLINE | ID: mdl-33555496

ABSTRACT

Citrullinemia Type I is an inborn error, which leads to accumulation of citrulline and ammonia in blood and body tissues. We evaluated the in vitro effects of citrulline, ammonia and the influence of resveratrol on oxidative stress parameters in the cerebrum of 30- and 60-day-old male Wistar rats. Citrulline (0.1, 2.5, 5.0 mM), ammonia (0.01, 0.1, 1.0 mM) and resveratrol (0.01, 0.1, 0.5 mM) were added to the assays to measure thiobarbituric acid reactive substances (TBA-RS), total sulfhydryl content and the activity of antioxidant enzymes catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Citrulline (2.5 and 5.0 mM) increased TBA-RS in the cerebellum of 30-day-old and in the cerebral cortex and cerebellum of 60-day-old. Citrulline (5.0 mM) increased SOD and reduced GSH-Px in the hippocampus of 30-day-old, whereas in the cerebellum it increased GSH-Px. In the cerebral cortex, 2.5 and 5.0 mM citrulline reduced GSH-Px. In 60-day-old, 2.5 and 5.0 mM citrulline increased SOD in the cerebellum, increased GSH-Px in the cerebral cortex and 5.0 mM citrulline reduced CAT and increased SOD in the cerebral cortex. Ammonia (0.1 and 1.0 mM) reduced the sulfhydryl content in the cerebral cortex of 30- and 60-day-old, 1.0 mM ammonia increased SOD and reduced GSH-Px in the cerebellum of 30-day-old and increased SOD in the hippocampus and cerebellum of 60-day-old. Resveratrol was able to prevent the majority of these alterations. Thus, citrulline and ammonia induce oxidative stress in the cerebrum of rats; however, resveratrol was able to exert antioxidant effects against these substances.


Subject(s)
Antioxidants/pharmacology , Brain/metabolism , Citrullinemia/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/physiology , Resveratrol/pharmacology , Ammonia/toxicity , Animals , Antioxidants/therapeutic use , Brain/drug effects , Citrulline/toxicity , Citrullinemia/chemically induced , Citrullinemia/prevention & control , Dose-Response Relationship, Drug , Male , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Rats , Rats, Wistar , Resveratrol/therapeutic use
5.
J Inherit Metab Dis ; 44(1): 110-117, 2021 01.
Article in English | MEDLINE | ID: mdl-32740958

ABSTRACT

Citrin deficiency is a hereditary disorder caused by SLC25A13 mutations and manifests as neonatal intrahepatic cholestasis (NICCD), failure to thrive and dyslipidemia (FTTDCD), and adult-onset type II citrullinemia (CTLN2). Citrin is a component of the malate-aspartate nicotinamide adenine dinucleotide hydrogen (NADH) shuttle, an essential shuttle for hepatic glycolysis. Hepatic glycolysis and the coupled lipogenesis are impaired in citrin deficiency. Hepatic lipogenesis plays a significant role in fat supply during growth spurt periods: the fetal period, infancy, and puberty. Growth impairment in these periods is characteristic of citrin deficiency. Hepatocytes with citrin deficiency cannot use glucose and fatty acids as energy sources due to defects in the NADH shuttle and downregulation of peroxisome proliferator-activated receptor α (PPARα), respectively. An energy deficit in hepatocytes is considered a fundamental pathogenesis of citrin deficiency. Medium-chain triglyceride (MCT) supplementation with a lactose-restricted formula and MCT supplementation under a low-carbohydrate diet are recommended for NICCD and CTLN2, respectively. MCT supplementation therapy can provide energy to hepatocytes, promote lipogenesis, correct the cytosolic NAD+ /NADH ratio via the malate-citrate shuttle and improve ammonia detoxification, and it is a reasonable therapy for citrin deficiency. It is very important to administer MCT at a dose equivalent to the liver's energy requirements in divided doses with meals. MCT supplementation therapy is certainly promising for promoting growth spurts during infancy and adolescence and for preventing CTLN2 onset. Intravenous administration of solutions containing fructose is contraindicated, and persistent hyperglycemia should be avoided due to glucose intoxication for patients receiving hyperalimentation or with complicating diabetes.


Subject(s)
Citrullinemia/drug therapy , Citrullinemia/prevention & control , Triglycerides/therapeutic use , Adolescent , Citrullinemia/metabolism , Energy Metabolism , Hepatocytes/metabolism , Humans , Infant
6.
Naunyn Schmiedebergs Arch Pharmacol ; 394(5): 873-884, 2021 05.
Article in English | MEDLINE | ID: mdl-33205249

ABSTRACT

We investigated the in vitro effects of citrulline (0.1, 2.5 and 5.0 mM) and ammonia (0.01, 0.1 and 1.0 mM), and the influence of resveratrol (0.01 mM, 0.1 mM and 0.5 mM) on pyruvate kinase, citrate synthase, succinate dehydrogenase (SDH), complex II, and cytochrome c oxidase activities in cerebral cortex, cerebellum and hippocampus homogenates of 60-day-old male Wistar rats. Results showed that 2.5 and 5.0 mM citrulline decreased pyruvate kinase activity in cerebral cortex and, at a concentration of 5.0 mM, increased its activity in hippocampus. Additionally, 5.0 mM citrulline increased citrate synthase activity in the cerebellum of rats. Citrulline (5.0 mM) reduced complex II and cytochrome c oxidase activities in cerebral cortex and hippocampus. With regard to ammonia, at 0.1 and 1.0 mM, decreased complex II activity in cerebral cortex and at 1.0 mM decreased its activity in cerebellum and hippocampus. Ammonia (1.0 mM) also decreased cytochrome c oxidase activity in cerebral cortex and cerebellum of rats. Resveratrol was able to prevent most of the alterations caused by these metabolites in the biomarkers of energy metabolism measured in the cerebrum of rats. Data suggest that these alterations in energy metabolism, caused by citrulline and ammonia, are probably mediated by the generation of free radicals, which can in turn be scavenged by resveratrol.


Subject(s)
Citrullinemia/drug therapy , Energy Metabolism/drug effects , Free Radical Scavengers/pharmacology , Resveratrol/pharmacology , Ammonia/administration & dosage , Ammonia/toxicity , Animals , Cerebellum/drug effects , Cerebellum/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Citrulline/administration & dosage , Citrulline/toxicity , Citrullinemia/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Free Radical Scavengers/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Male , Rats , Rats, Wistar , Resveratrol/administration & dosage
7.
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
8.
Biomolecules ; 10(8)2020 07 24.
Article in English | MEDLINE | ID: mdl-32722104

ABSTRACT

Can you imagine a disease in which intake of an excess amount of sugars or carbohydrates causes hyperammonemia? It is hard to imagine the intake causing hyperammonemia. AGC2 or citrin deficiency shows their symptoms following sugar/carbohydrates intake excess and this disease is now known as a pan-ethnic disease. AGC2 (aspartate glutamate carrier 2) or citrin is a mitochondrial transporter which transports aspartate (Asp) from mitochondria to cytosol in exchange with glutamate (Glu) and H+. Asp is originally supplied from mitochondria to cytosol where it is necessary for synthesis of proteins, nucleotides, and urea. In cytosol, Asp can be synthesized from oxaloacetate and Glu by cytosolic Asp aminotransferase, but oxaloacetate formation is limited by the amount of NAD+. This means an increase in NADH causes suppression of Asp formation in the cytosol. Metabolism of carbohydrates and other substances which produce cytosolic NADH such as alcohol and glycerol suppress oxaloacetate formation. It is forced under citrin deficiency since citrin is a member of malate/Asp shuttle. In this review, we will describe history of identification of the SLC25A13 gene as the causative gene for adult-onset type II citrullinemia (CTLN2), a type of citrin deficiency, pathophysiology of citrin deficiency together with animal models and possible treatments for citrin deficiency newly developing.


Subject(s)
Aspartic Acid/metabolism , Calcium-Binding Proteins/metabolism , Citrullinemia/metabolism , Glutamic Acid/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Organic Anion Transporters/metabolism , Animals , Biological Transport , Calcium-Binding Proteins/genetics , Citrullinemia/genetics , Citrullinemia/therapy , Genetic Predisposition to Disease/genetics , Humans , Mitochondrial Membrane Transport Proteins/genetics , Organic Anion Transporters/genetics
9.
Oncogene ; 39(1): 164-175, 2020 01.
Article in English | MEDLINE | ID: mdl-31462712

ABSTRACT

Citrin, encoded by SLC25A13 gene, is an inner mitochondrial transporter that is part of the malate-aspartate shuttle, which regulates the NAD+/NADH ratio between the cytosol and mitochondria. Citrullinemia type II (CTLN-II) is an inherited disorder caused by germline mutations in SLC25A13, manifesting clinically in growth failure that can be alleviated by dietary restriction of carbohydrates. The association of citrin with glycolysis and NAD+/NADH ratio led us to hypothesize that it may play a role in carcinogenesis. Indeed, we find that citrin is upregulated in multiple cancer types and is essential for supplementing NAD+ for glycolysis and NADH for oxidative phosphorylation. Consequently, citrin deficiency associates with autophagy, whereas its overexpression in cancer cells increases energy production and cancer invasion. Furthermore, based on the human deleterious mutations in citrin, we found a potential inhibitor of citrin that restricts cancerous phenotypes in cells. Collectively, our findings suggest that targeting citrin may be of benefit for cancer therapy.


Subject(s)
Carcinogenesis/genetics , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Neoplasms/genetics , Carbohydrates/genetics , Citrullinemia/genetics , Citrullinemia/metabolism , Cytosol/metabolism , Cytosol/pathology , Gene Expression Regulation, Neoplastic/genetics , Germ-Line Mutation/genetics , Glutamates/pharmacology , Glutamic Acid/analogs & derivatives , Glutamic Acid/pharmacology , Glycolysis/genetics , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Oxidative Phosphorylation/drug effects
10.
Mol Ther ; 27(7): 1242-1251, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31056400

ABSTRACT

Citrin deficiency is an autosomal recessive disorder caused by loss-of-function mutations in SLC25A13, encoding the liver-specific mitochondrial aspartate/glutamate transporter. It has a broad spectrum of clinical phenotypes, including life-threatening neurological complications. Conventional protein replacement therapy is not an option for these patients because of drug delivery hurdles, and current gene therapy approaches (e.g., AAV) have been hampered by immunogenicity and genotoxicity. Although dietary approaches have shown some benefits in managing citrin deficiency, the only curative treatment option for these patients is liver transplantation, which is high-risk and associated with long-term complications because of chronic immunosuppression. To develop a new class of therapy for citrin deficiency, codon-optimized mRNA encoding human citrin (hCitrin) was encapsulated in lipid nanoparticles (LNPs). We demonstrate the efficacy of hCitrin-mRNA-LNP therapy in cultured human cells and in a murine model of citrin deficiency that resembles the human condition. Of note, intravenous (i.v.) administration of the hCitrin-mRNA resulted in a significant reduction in (1) hepatic citrulline and blood ammonia levels following oral sucrose challenge and (2) sucrose aversion, hallmarks of hCitrin deficiency. In conclusion, mRNA-LNP therapy could have a significant therapeutic effect on the treatment of citrin deficiency and other mitochondrial enzymopathies with limited treatment options.


Subject(s)
Citrullinemia/drug therapy , Citrullinemia/metabolism , Drug Delivery Systems/methods , Genetic Therapy/methods , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , RNA, Messenger/therapeutic use , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Gene Knockout Techniques , Glucosephosphate Dehydrogenase/genetics , HeLa Cells , Hep G2 Cells , Humans , Lipids/chemistry , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Nanoparticles/chemistry , Open Reading Frames/genetics , RNA, Messenger/chemical synthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Transfection , Treatment Outcome
11.
Sci Rep ; 9(1): 4179, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862943

ABSTRACT

Previous studies using citrin/mitochondrial glycerol-3-phosphate (G3P) dehydrogenase (mGPD) double-knockout mice have demonstrated that increased dietary protein reduces the extent of carbohydrate-induced hyperammonemia observed in these mice. This study aimed to further elucidate the mechanisms of this effect. Specific amino acids were initially found to decrease hepatic G3P, or increase aspartate or citrulline levels, in mGPD-knockout mice administered ethanol. Unexpectedly, oral glycine increased ammonia in addition to lowering G3P and increasing citrulline. Subsequently, simultaneous glycine-plus-sucrose (Gly + Suc) administration led to a more severe hyperammonemic state in double-KO mice compared to sucrose alone. Oral arginine, ornithine, aspartate, alanine, glutamate and medium-chain triglycerides all lowered blood ammonia following Gly + Suc administration, with combinations of ornithine-plus-aspartate (Orn + Asp) or ornithine-plus-alanine (Orn + Ala) suppressing levels similar to wild-type. Liver perfusion and portal vein-arterial amino acid differences suggest that oral aspartate, similar to alanine, likely activated ureagenesis from ammonia and lowered the cytosolic NADH/NAD+ ratio through conversion to alanine in the small intestine. In conclusion, Gly + Suc administration induces a more severe hyperammonemic state in double-KO mice that Orn + Asp or Orn + Ala both effectively suppress. Aspartate-to-alanine conversion in the small intestine allows for effective oral administration of either, demonstrating a pivotal role of inter-organ aspartate metabolism for the treatment of citrin deficiency.


Subject(s)
Aspartic Acid/metabolism , Citrullinemia/metabolism , Mitochondrial Membrane Transport Proteins/deficiency , Organ Specificity , Amino Acids/blood , Amino Acids/pharmacology , Ammonia/blood , Ammonium Chloride/metabolism , Animals , Citrulline/pharmacology , Disease Models, Animal , Glycerolphosphate Dehydrogenase/metabolism , Hyperammonemia/blood , Intestine, Small/metabolism , Lactates/metabolism , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Ornithine/pharmacology , Perfusion , Portal Vein/metabolism , Pyruvic Acid/metabolism , Urea/metabolism
12.
J Clin Endocrinol Metab ; 103(7): 2488-2497, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29659898

ABSTRACT

Context: Citrin-deficient infants present neonatal intrahepatic cholestasis caused by citrin deficiency (NICCD), which resolves at 12 months. Thereafter, they have normal liver function associated with hypercholesterolemia, and a preference for lipid-rich carbohydrate-restricted diets. However, some develop adult-onset type II citrullinemia, which is associated with metabolic abnormalities. Objectives: To identify the causes of hypercholesterolemia in citrin-deficient children post-NICCD. Design and Setting: We determined the concentrations of sterol markers of cholesterol synthesis, absorption, and catabolism by liquid chromatography-electrospray ionization-tandem mass spectrometry and evaluated serum lipoprotein profiles. Subjects: Twenty citrin-deficient children aged 5 to 13 years and 37 age-matched healthy children. Intervention: None. Main Outcome Measures: Relationship between serum lipoproteins and sterol markers of cholesterol metabolism. Results: The citrin-deficient group had a significantly higher high-density lipoprotein cholesterol (HDL-C) concentration than did the control group (78 ± 11 mg/dL vs 62 ± 14 mg/dL, P < 0.001), whereas the two groups had similar low-density lipoprotein cholesterol and triglyceride concentrations. The concentrations of markers of cholesterol synthesis (lathosterol and 7-dehydrocholesterol) and bile acids synthesis (7α-hydroxycholesterol and 27-hydroxycholesterol) were 1.5- to 2.8-fold and 1.5- to 3.9-fold, respectively, higher in the citrin-deficient group than in the control group. The concentration of 24S-hydroxycholesterol, a marker of cholesterol catabolism in the brain, was 2.5-fold higher in the citrin-deficient group. In both groups, the HDL-C concentration was significantly positively correlated with that of 27-hydroxycholesterol, the first product of the alternative bile acid synthesis pathway. Conclusions: HDL-C and sterol marker concentrations are elevated in citrin-deficient children post-NICCD. Moreover, cholesterol synthesis and elimination are markedly enhanced in the liver and brain of citrin-deficient children.


Subject(s)
Brain/metabolism , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Citrullinemia/metabolism , Hypercholesterolemia/etiology , Liver/metabolism , Adolescent , Bile Acids and Salts/biosynthesis , Biomarkers/blood , Case-Control Studies , Child , Child, Preschool , Cholesterol/blood , Citrullinemia/complications , Dehydrocholesterols/blood , Female , Humans , Hydroxycholesterols/blood , Male , Triglycerides/blood
13.
World J Gastroenterol ; 23(44): 7930-7938, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29209134

ABSTRACT

The underlying pathophysiology of liver dysfunction in urea cycle disorders (UCDs) is still largely elusive. There is some evidence that the accumulation of urea cycle (UC) intermediates are toxic for hepatocyte mitochondria. It is possible that liver injury is directly caused by the toxicity of ammonia. The rarity of UCDs, the lack of checking of iron level in these patients, superficial knowledge of UC and an underestimation of the metabolic role of fumaric acid, are the main reasons that are responsible for the incomprehension of the mechanism of liver injury in patients suffering from UCDs. Owing to our routine clinical practice to screen for iron overload in severely ill neonates, with the focus on the newborns suffering from acute liver failure, we report a case of citrullinemia with neonatal liver failure and high blood parameters of iron overload. We hypothesize that the key is in the decreased-deficient fumaric acid production in the course of UC in UCDs that causes several sequentially intertwined metabolic disturbances with final result of liver iron overload. The presented hypothesis could be easily tested by examining the patients suffering from UCDs, for liver iron overload. This could be easily performed in countries with a high population and comprehensive national register for inborn errors of metabolism. CONCLUSION: Providing the hypothesis is correct, neonatal liver damage in patients having UCD can be prevented by the supplementation of pregnant women with fumaric or succinic acid, prepared in the form of iron supplementation pills. After birth, liver damage in patients having UCDs can be prevented by supplementation of these patients with zinc fumarate or zinc succinylate, as well.


Subject(s)
Citrullinemia/metabolism , Hepatocytes/metabolism , Hyperammonemia/metabolism , Liver Diseases/etiology , Liver/physiopathology , Ammonia/blood , Ammonia/toxicity , Central Nervous System/physiopathology , Citrullinemia/blood , Citrullinemia/diagnosis , Citrullinemia/therapy , Enterobacter/isolation & purification , Fatal Outcome , Humans , Hyperammonemia/blood , Hyperammonemia/diagnosis , Hyperammonemia/therapy , Infant, Newborn , Lethargy/etiology , Lethargy/metabolism , Lethargy/physiopathology , Lethargy/therapy , Liver/cytology , Liver/metabolism , Liver Diseases/metabolism , Liver Diseases/physiopathology , Liver Diseases/therapy , Male , Sepsis/microbiology , Sepsis/therapy , Urea/blood , Urea/metabolism , Urea/toxicity
14.
Stem Cell Res Ther ; 8(1): 176, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28750687

ABSTRACT

BACKGROUND: Argininosuccinate synthase (ASS)1 is a urea cycle enzyme that catalyzes the conversion of citrulline and aspartate to argininosuccinate. Mutations in the ASS1 gene cause citrullinemia type I, a rare autosomal recessive disorder characterized by neonatal hyperammonemia, elevated citrulline levels, and early neonatal death. Treatment for this disease is currently restricted to liver transplantation; however, due to limited organ availability, substitute therapies are required. Recently, extracellular vesicles (EVs) have been reported to act as intercellular transporters carrying genetic information responsible for cell reprogramming. In previous studies, we isolated a population of stem cell-like cells known as human liver stem cells (HLSCs) from healthy liver tissue. Moreover, EVs derived from HLSCs were reported to exhibit regenerative effects on the liver parenchyma in models of acute liver injury. The aim of this study was to evaluate whether EVs derived from normal HLSCs restored ASS1 enzymatic activity and urea production in hepatocytes differentiated from HLSCs derived from a patient with type I citrullinemia. METHODS: HLSCs were isolated from the liver of a patient with type I citrullinemia (ASS1-HLSCs) and characterized by fluorescence-activated cell sorting (FACS), immunofluorescence, and DNA sequencing analysis. Furthermore, their differentiation capabilities in vitro were also assessed. Hepatocytes differentiated from ASS1-HLSCs were evaluated by the production of urea and ASS enzymatic activity. EVs derived from normal HLSCs were purified by differential ultracentrifugation followed by floating density gradient. The EV content was analyzed to identify the presence of ASS1 protein, mRNA, and ASS1 gene. In order to obtain ASS1-depleted EVs, a knockdown of the ASS1 gene in HLSCs was performed followed by EV isolation from these cells. RESULTS: Treating ASS1-HLSCs with EVs from HLSCs restored both ASS1 activity and urea production mainly through the transfer of ASS1 enzyme and mRNA. In fact, EVs from ASS1-knockdown HLSCs contained low amounts of ASS1 mRNA and protein, and were unable to restore urea production in hepatocytes differentiated from ASS1-HLSCs. CONCLUSIONS: Collectively, these results suggest that EVs derived from normal HLSCs may compensate the loss of ASS1 enzyme activity in hepatocytes differentiated from ASS1-HLSCs.


Subject(s)
Argininosuccinate Synthase , Citrullinemia , Extracellular Vesicles/metabolism , Liver/metabolism , Stem Cells/metabolism , Argininosuccinate Synthase/biosynthesis , Argininosuccinate Synthase/genetics , Citrullinemia/genetics , Citrullinemia/metabolism , Citrullinemia/therapy , Hepatocytes/metabolism , Humans , Urea/metabolism
15.
Mol Genet Metab ; 120(4): 306-316, 2017 04.
Article in English | MEDLINE | ID: mdl-28259708

ABSTRACT

Mice carrying simultaneous homozygous mutations in the genes encoding citrin, the mitochondrial aspartate-glutamate carrier 2 (AGC2) protein, and mitochondrial glycerol-3-phosphate dehydrogenase (mGPD), are a phenotypically representative model of human citrin (a.k.a., AGC2) deficiency. In this study, we investigated the voluntary oral intake and preference for sucrose, glycerol or ethanol solutions by wild-type, citrin (Ctrn)-knockout (KO), mGPD-KO, and Ctrn/mGPD double-KO mice; all substances that are known or suspected precipitating factors in the pathogenesis of human citrin deficiency. The double-KO mice showed clear suppressed intake of sucrose, consuming less with progressively higher concentrations compared to the other mice. Similar observations were made when glycerol or ethanol were given. The preference of Ctrn-KO and mGPD-KO mice varied with the different treatments; essentially no differences were observed for sucrose, while an intermediate intake or similar to that of the double-KO mice was observed for glycerol and ethanol. We next examined the hepatic glycerol 3-phosphate, citrate, citrulline, lysine, glutamate and adenine nucleotide levels following forced enteral administration of these solutions. A strong correlation between the simultaneous increased hepatic glycerol 3-phosphate and decreased ATP or total adenine nucleotide content and observed aversion of the mice during evaluation of their voluntary preferences was found. Overall, our results suggest that the aversion observed in the double-KO mice to these solutions is initiated and/or mediated by hepatic metabolic perturbations, resulting in a behavioral response to increased hepatic cytosolic NADH and a decreased cellular adenine nucleotide pool. These findings may underlie the dietary predilections observed in human citrin deficient patients.


Subject(s)
Citrullinemia/metabolism , Dietary Sucrose/administration & dosage , Ethanol/administration & dosage , Glycerol/administration & dosage , Liver/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Transport Systems, Acidic/genetics , Animals , Antiporters/genetics , Disease Models, Animal , Glycerolphosphate Dehydrogenase/genetics , Glycerophosphates/metabolism , Humans , Mice , Mice, Knockout
16.
Mol Genet Metab ; 120(3): 207-212, 2017 03.
Article in English | MEDLINE | ID: mdl-28041819

ABSTRACT

Citrin deficiency causes adult-onset type II citrullinemia (CTLN-2), which later manifests as severe liver steatosis and life-threatening encephalopathy. Long-standing energy deficit of the liver and brain may predispose ones to CTLN-2. Here, we compared the energy-driving tricarboxylic acid (TCA) cycle and fatty acid ß-oxidation cycle between 22 citrin-deficient children (age, 3-13years) with normal liver functions and 37 healthy controls (age, 5-13years). TCA cycle analysis showed that basal plasma citrate and α-ketoglutarate levels were significantly higher in the affected than the control group (p<0.01). Conversely, basal plasma fumarate and malate levels were significantly lower than those for the control (p<0.001). The plasma level of 3-OH-butyrate derived from fatty acid ß-oxidation was significantly higher in the affected group (p<0.01). Ten patients underwent sodium pyruvate therapy. However, this therapy did not correct or attenuate such deviations in both cycles. Sodium pyruvate therapy significantly increased fasting insulin secretion (p<0.01); the fasting sugar level remained unchanged. Our results suggest that citrin-deficient children show considerable deviations of TCA cycle metabolite profiles that are resistant to sodium pyruvate treatment. Thus, long-standing and considerable TCA cycle dysfunction might be a pivotal metabolic background of CTLN-2 development.


Subject(s)
Citric Acid Cycle , Citrullinemia/drug therapy , Citrullinemia/metabolism , Fatty Acids/metabolism , Pyruvates/administration & dosage , Adolescent , Child , Child, Preschool , Citric Acid/blood , Citric Acid Cycle/drug effects , Female , Fumarates/blood , Humans , Ketoglutaric Acids/blood , Malates/blood , Male , Oxidative Stress/drug effects , Pyruvates/pharmacology , Treatment Outcome
17.
J Hum Genet ; 62(2): 305-307, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27829683

ABSTRACT

Mutations in SLC25A13 cause citrin deficiency, which has three phenotypes: neonatal intrahepatic cholestasis caused by citrin deficiency (NICCD), failure to thrive and dyslipidemia caused by citrin deficiency (FTTDCD) and adult-onset type 2 citrullinemia (CTLN2). The purpose of this study was to determine the mutation spectrum and the clinical and biochemical characteristics of citrin deficiency in Korean patients. Thirty-four patients were diagnosed with citrin deficiency based on mutations in SLC25A13, as verified by direct sequencing and long PCR screening of a large transposon insertion. A total of 66 alleles from 33 unrelated families of 34 patients with citrin deficiency (27 NICCD, 2 FTTDCD and 5 CTLN2) were retrospectively identified. The common pathogenic alleles were IVS16ins3kb (33%), c.851_854del (30%) and c.1177+1G>A (12%), and three novel variants were identified. Levels of citrulline, threonine, methionine, tyrosine and arginine and the threonine-to-serine ratio were higher in children with neonatal intrahepatic cholestasis caused by NICCD compared with that in patients with idiopathic neonatal hepatitis (INH). We concluded that Korean patients with citrin deficiency showed the highest frequency of the IVS16ins3kb mutation and that plasma amino-acid profiles can be used to differentiate between NICCD and INH.


Subject(s)
Citrullinemia/genetics , Failure to Thrive/genetics , Mitochondrial Membrane Transport Proteins/genetics , Adolescent , Adult , Alleles , Amino Acids/metabolism , Asian People/genetics , Base Sequence , Citrullinemia/metabolism , Failure to Thrive/metabolism , Female , Gene Frequency/genetics , Humans , Infant , Infant, Newborn , Male , Mitochondrial Membrane Transport Proteins/metabolism , Mutation , Polymerase Chain Reaction , Republic of Korea , Retrospective Studies , Sequence Analysis, DNA , Young Adult
18.
J Inherit Metab Dis ; 39(5): 661-672, 2016 09.
Article in English | MEDLINE | ID: mdl-27106216

ABSTRACT

BACKGROUND: Patients with urea cycle disorders (UCDs) have an increased risk of neurological disease manifestation. AIMS: Determining the effect of diagnostic and therapeutic interventions on the neurological outcome. METHODS: Evaluation of baseline, regular follow-up and emergency visits of 456 UCD patients prospectively followed between 2011 and 2015 by the E-IMD patient registry. RESULTS: About two-thirds of UCD patients remained asymptomatic until age 12 days [i.e. the median age at diagnosis of patients identified by newborn screening (NBS)] suggesting a potential benefit of NBS. In fact, NBS lowered the age at diagnosis in patients with late onset of symptoms (>28 days), and a trend towards improved long-term neurological outcome was found for patients with argininosuccinate synthetase and lyase deficiency as well as argininemia identified by NBS. Three to 17 different drug combinations were used for maintenance therapy, but superiority of any single drug or specific drug combination above other combinations was not demonstrated. Importantly, non-interventional variables of disease severity, such as age at disease onset and peak ammonium level of the initial hyperammonemic crisis (cut-off level: 500 µmol/L) best predicted the neurological outcome. CONCLUSIONS: Promising results of NBS for late onset UCD patients are reported and should be re-evaluated in a larger and more advanced age group. However, non-interventional variables affect the neurological outcome of UCD patients. Available evidence-based guideline recommendations are currently heterogeneously implemented into practice, leading to a high variability of drug combinations that hamper our understanding of optimised long-term and emergency treatment.


Subject(s)
Ammonium Compounds/metabolism , Nervous System Diseases/diagnosis , Nervous System Diseases/metabolism , Urea Cycle Disorders, Inborn/diagnosis , Urea Cycle Disorders, Inborn/metabolism , Adolescent , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/metabolism , Argininosuccinate Synthase/metabolism , Child , Citrullinemia/diagnosis , Citrullinemia/metabolism , Female , Humans , Hyperammonemia/diagnosis , Hyperammonemia/metabolism , Infant, Newborn , Late Onset Disorders/diagnosis , Late Onset Disorders/metabolism , Male , Neonatal Screening/methods , Prospective Studies , Urea/metabolism
19.
Nature ; 527(7578): 379-383, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26560030

ABSTRACT

Cancer cells hijack and remodel existing metabolic pathways for their benefit. Argininosuccinate synthase (ASS1) is a urea cycle enzyme that is essential in the conversion of nitrogen from ammonia and aspartate to urea. A decrease in nitrogen flux through ASS1 in the liver causes the urea cycle disorder citrullinaemia. In contrast to the well-studied consequences of loss of ASS1 activity on ureagenesis, the purpose of its somatic silencing in multiple cancers is largely unknown. Here we show that decreased activity of ASS1 in cancers supports proliferation by facilitating pyrimidine synthesis via CAD (carbamoyl-phosphate synthase 2, aspartate transcarbamylase, and dihydroorotase complex) activation. Our studies were initiated by delineating the consequences of loss of ASS1 activity in humans with two types of citrullinaemia. We find that in citrullinaemia type I (CTLN I), which is caused by deficiency of ASS1, there is increased pyrimidine synthesis and proliferation compared with citrullinaemia type II (CTLN II), in which there is decreased substrate availability for ASS1 caused by deficiency of the aspartate transporter citrin. Building on these results, we demonstrate that ASS1 deficiency in cancer increases cytosolic aspartate levels, which increases CAD activation by upregulating its substrate availability and by increasing its phosphorylation by S6K1 through the mammalian target of rapamycin (mTOR) pathway. Decreasing CAD activity by blocking citrin, the mTOR signalling, or pyrimidine synthesis decreases proliferation and thus may serve as a therapeutic strategy in multiple cancers where ASS1 is downregulated. Our results demonstrate that ASS1 downregulation is a novel mechanism supporting cancerous proliferation, and they provide a metabolic link between the urea cycle enzymes and pyrimidine synthesis.


Subject(s)
Argininosuccinate Synthase/deficiency , Aspartic Acid/metabolism , Neoplasms/metabolism , Pyrimidines/biosynthesis , Animals , Argininosuccinate Synthase/metabolism , Aspartate Carbamoyltransferase/metabolism , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Cell Line, Tumor , Cell Proliferation , Citrullinemia/metabolism , Cytosol/metabolism , Dihydroorotase/metabolism , Down-Regulation , Enzyme Activation , Humans , Male , Mice , Mice, SCID , Neoplasms/enzymology , Neoplasms/pathology , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/metabolism , Phosphorylation , TOR Serine-Threonine Kinases/metabolism
20.
Brain Nerve ; 67(6): 739-47, 2015 Jun.
Article in Japanese | MEDLINE | ID: mdl-26062589

ABSTRACT

Citrin, encoded by SLC25A13, is a component of the malate-aspartate shuttle, which is the main NADH-transporting system in the liver. Citrin deficiency causes neonatal intrahepatic cholestasis (NICCD), which usually resolves within the first year of life. However, a small number of adults with citrin deficiency develop adult-onset type II citrullinemia (CTLN2), which causes hyperammonemic encephalopathy leading to death due to cerebral edema. Liver transplantation is the only definitive therapy for patients with CTLN2. Hepatic glycolysis is coupled with hepatic lipogenesis via the NADH shuttles composed of the malate-aspartate shuttle and malate-citrate shuttle. Citrin deficiency is expected to impair glycolysis and lipogenesis in hepatocytes. We noticed that a lactose (galactose)-restricted and medium-chain triglyceride (MCT)-supplemented formula is notably effective for patients with NICCD. We extended this therapy for CTLN2 and found that an MCT supplementation therapy under a low-carbohydrate formula prevented the relapse of hyperammonemic encephalopathy, normalized the liver dysfunction (including the Fisher ratio), and gradually improved the level of plasma citrulline and fatty liver. An MCT supplement can provide energy to hepatocytes and promote hepatic lipogenesis, leading to improvement of the cytosolic NAD+/NADH ratio via the malate-citrate shuttle. MCT supplementation could be a promising therapy for citrin deficiency.


Subject(s)
Citrullinemia/drug therapy , Triglycerides/therapeutic use , Adult , Citrullinemia/metabolism , Humans , Infant
SELECTION OF CITATIONS
SEARCH DETAIL
...