Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
1.
Sci Rep ; 13(1): 20535, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37996510

ABSTRACT

A multi-class classification model for acute coronary syndrome (ACS) remains to be constructed based on multi-fluid metabolomics. Major confounders may exert spurious effects on the relationship between metabolism and ACS. The study aims to identify an independent biomarker panel for the multiclassification of HC, UA, and AMI by integrating serum and urinary metabolomics. We performed a liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based metabolomics study on 300 serum and urine samples from 44 patients with unstable angina (UA), 77 with acute myocardial infarction (AMI), and 29 healthy controls (HC). Multinomial machine learning approaches, including multinomial adaptive least absolute shrinkage and selection operator (LASSO) regression and random forest (RF), and assessment of the confounders were applied to integrate a multi-class classification biomarker panel for HC, UA and AMI. Different metabolic landscapes were portrayed during the transition from HC to UA and then to AMI. Glycerophospholipid metabolism and arginine biosynthesis were predominant during the progression from HC to UA and then to AMI. The multiclass metabolic diagnostic model (MDM) dependent on ACS, including 2-ketobutyric acid, LysoPC(18:2(9Z,12Z)), argininosuccinic acid, and cyclic GMP, demarcated HC, UA, and AMI, providing a C-index of 0.84 (HC vs. UA), 0.98 (HC vs. AMI), and 0.89 (UA vs. AMI). The diagnostic value of MDM largely derives from the contribution of 2-ketobutyric acid, and LysoPC(18:2(9Z,12Z)) in serum. Higher 2-ketobutyric acid and cyclic GMP levels were positively correlated with ACS risk and atherosclerosis plaque burden, while LysoPC(18:2(9Z,12Z)) and argininosuccinic acid showed the reverse relationship. An independent multiclass biomarker panel for HC, UA, and AMI was constructed using the multinomial machine learning methods based on serum and urinary metabolite signatures.


Subject(s)
Acute Coronary Syndrome , Myocardial Infarction , Humans , Acute Coronary Syndrome/diagnosis , Argininosuccinic Acid , Chromatography, Liquid , Tandem Mass Spectrometry , Biomarkers , Myocardial Infarction/diagnosis , Angina, Unstable , Cyclic GMP
2.
Nature ; 615(7952): 490-498, 2023 03.
Article in English | MEDLINE | ID: mdl-36890227

ABSTRACT

Metabolic rewiring underlies the effector functions of macrophages1-3, but the mechanisms involved remain incompletely defined. Here, using unbiased metabolomics and stable isotope-assisted tracing, we show that an inflammatory aspartate-argininosuccinate shunt is induced following lipopolysaccharide stimulation. The shunt, supported by increased argininosuccinate synthase (ASS1) expression, also leads to increased cytosolic fumarate levels and fumarate-mediated protein succination. Pharmacological inhibition and genetic ablation of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) further increases intracellular fumarate levels. Mitochondrial respiration is also suppressed and mitochondrial membrane potential increased. RNA sequencing and proteomics analyses demonstrate that there are strong inflammatory effects resulting from FH inhibition. Notably, acute FH inhibition suppresses interleukin-10 expression, which leads to increased tumour necrosis factor secretion, an effect recapitulated by fumarate esters. Moreover, FH inhibition, but not fumarate esters, increases interferon-ß production through mechanisms that are driven by mitochondrial RNA (mtRNA) release and activation of the RNA sensors TLR7, RIG-I and MDA5. This effect is recapitulated endogenously when FH is suppressed following prolonged lipopolysaccharide stimulation. Furthermore, cells from patients with systemic lupus erythematosus also exhibit FH suppression, which indicates a potential pathogenic role for this process in human disease. We therefore identify a protective role for FH in maintaining appropriate macrophage cytokine and interferon responses.


Subject(s)
Fumarate Hydratase , Interferon-beta , Macrophages , Mitochondria , RNA, Mitochondrial , Humans , Argininosuccinate Synthase/metabolism , Argininosuccinic Acid/metabolism , Aspartic Acid/metabolism , Cell Respiration , Cytosol/metabolism , Fumarate Hydratase/antagonists & inhibitors , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Fumarates/metabolism , Interferon-beta/biosynthesis , Interferon-beta/immunology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Lupus Erythematosus, Systemic/enzymology , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Membrane Potential, Mitochondrial , Metabolomics , Mitochondria/genetics , Mitochondria/metabolism , RNA, Mitochondrial/metabolism
3.
Appl Biochem Biotechnol ; 194(11): 5443-5455, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35789984

ABSTRACT

Acute ischemic stroke (AIS) is characterized by a sudden blockage of one of the main arteries supplying blood to the brain, leading to insufficient oxygen and nutrients for brain cells to function properly. Unfortunately, metabolic alterations in the biofluids with AIS are still not well understood. In this study, we performed high-throughput target metabolic analysis on 44 serum samples, including 22 from AIS patients and 22 from healthy controls. Multiple-reaction monitoring analysis of 180 common metabolites revealed a total of 29 metabolites that changed significantly (VIP > 1, p < 0.05). Multivariate statistical analysis unraveled a striking separation between AIS patients and healthy controls. Comparing the AIS group with the control group, the contents of argininosuccinic acid, beta-D-glucosamine, glycerophosphocholine, L-abrine, and L-pipecolic acid were remarkably downregulated in AIS patients. Twenty-nine out of 112 detected metabolites enriched in disturbed metabolic pathways, including aminoacyl-tRNA biosynthesis, glycerophospholipid metabolism, lysine degradation, phenylalanine, tyrosine, and tryptophan biosynthesis metabolic pathways. Collectively, these results will provide a sensitive, feasible diagnostic prospect for AIS patients.


Subject(s)
Ischemic Stroke , Stroke , Humans , Stroke/diagnosis , Tryptophan , Argininosuccinic Acid , Lysine , Biomarkers/metabolism , Metabolomics/methods , Tyrosine , Phenylalanine , Glucosamine , Oxygen , Glycerophospholipids , RNA, Transfer
4.
Anal Chem ; 92(19): 12891-12899, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32822159

ABSTRACT

Since l-argininosuccinic acid (ASA) is the characteristic biomarker for the diagnosis of certain diseases, its reliable detection in complex biological samples is necessary to obtain a complete evaluation with greater specificity and accuracy. ASA can undergo intramolecular cyclization, yielding an equilibrium with the resulting cyclic forms, which can predominate under different analytical conditions. In this work, the appearance and transformation of the different forms of ASA have been studied and a strategy for targeted screening analysis of ASA and its cyclic forms using capillary electrophoresis-electrospray ionization-time-of-flight mass spectrometry (CE-ESI-TOF-MS) has been developed. The data and spectra obtained allowed us to gain further insight into accurate identification, concluding that there is a dynamic equilibrium depending on the pH. Moreover, one- and two-dimensional NMR spectroscopy experiments have allowed us to determine the predominant tautomeric structure for the major cyclic ASA derivative, confirming the importance of intramolecular hydrogen bonds.


Subject(s)
Argininosuccinic Acid/chemical synthesis , Argininosuccinic Acid/urine , Argininosuccinic Acid/chemistry , Cyclization , Electrophoresis, Capillary , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Male , Molecular Conformation , Spectrometry, Mass, Electrospray Ionization
5.
PLoS One ; 15(4): e0230813, 2020.
Article in English | MEDLINE | ID: mdl-32267860

ABSTRACT

Lung macrophages (LM) are in the first line of defense against inhaled pathogens and can undergo phenotypic polarization to the proinflammatory M1 after stimulation with Toll-like receptor agonists. The objective of the present work was to characterize the metabolic alterations occurring during the experimental M1 LM polarization. Human LM were obtained from resected lungs and cultured for 24 hrs in medium alone or with 10 ng.mL-1 lipopolysaccharide. Cells and culture supernatants were subjected to extraction for metabolomic analysis with high-resolution LC-MS (HILIC and reverse phase -RP- chromatography in both negative and positive ionization modes) and GC-MS. The data were analyzed with R and the Worklow4Metabolomics and MetaboAnalyst online infrastructures. A total of 8,741 and 4,356 features were detected in the intracellular and extracellular content, respectively, after the filtering steps. Pathway analysis showed involvement of arachidonic acid metabolism, tryptophan metabolism and Krebs cycle in the response of LM to LPS, which was confirmed by the specific quantitation of selected compounds. This refined analysis highlighted a regulation of the kynurenin pathway as well as the serotonin biosynthesis pathway, and an involvement of aspartate-arginosuccinate shunt in the malate production. Macrophages M1 polarization is accompanied by changes in the cell metabolome, with the differential expression of metabolites involved in the promotion and regulation of inflammation and antimicrobial activity. The analysis of this macrophage immunometabolome may be of interest for the understanding of the pathophysiology of lung inflammatory disesases.


Subject(s)
Argininosuccinic Acid/metabolism , Aspartic Acid/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Tryptophan/metabolism , Aged , Cells, Cultured , Female , Humans , Inflammation/metabolism , Macrophage Activation/drug effects , Macrophage Activation/physiology , Male , Middle Aged , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Mol Neurobiol ; 57(2): 1233-1244, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31707633

ABSTRACT

Tissue accumulation and high urinary excretion of argininosuccinate (ASA) is the biochemical hallmark of argininosuccinate lyase deficiency (ASLD), a urea cycle disorder mainly characterized by neurologic abnormalities, whose pathogenesis is still unknown. Thus, in the present work, we evaluated the in vitro and in vivo effects of ASA on a large spectrum of oxidative stress parameters in brain of adolescent rats in order to test whether disruption of redox homeostasis could be involved in neurodegeneration of this disorder. ASA provoked in vitro lipid and protein oxidation, decreased reduced glutathione (GSH) concentrations, and increased reactive oxygen species generation in cerebral cortex and striatum. Furthermore, these effects were totally prevented or attenuated by the antioxidants melatonin and GSH. Similar results were obtained by intrastriatal administration of ASA, in addition to increased reactive nitrogen species generation and decreased activities of superoxide dismutase, glutathione peroxidase, and glutathione S-transferase. It was also observed that melatonin and N-acetylcysteine prevented most of ASA-induced in vivo pro-oxidant effects in striatum. Taken together, these data indicate that disturbance of redox homeostasis induced at least in part by high brain ASA concentrations per se may potentially represent an important pathomechanism of neurodegeneration in patients with ASLD and that therapeutic trials with appropriate antioxidants may be an adjuvant treatment for these patients.


Subject(s)
Argininosuccinic Acid/pharmacology , Brain/drug effects , Free Radical Scavengers/metabolism , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Brain/growth & development , Brain/metabolism , Glutathione Peroxidase/metabolism , Rats, Wistar , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
7.
Acta Medica Philippina ; : 366-372, 2020.
Article in English | WPRIM (Western Pacific) | ID: wpr-979856

ABSTRACT

Objective@#The study is a retrospective review which provides preliminary data on the correlation between biochemical profiles and initial clinical manifestation of patients diagnosed to have argininosuccinate synthetase deficiency (ASSD) and argininosuccinate lyase deficiency (ASLD) detected by expanded newborn screening (ENBS). @*Methods@#This is a study of five distal UCD patients initially detected by elevated citrulline on ENBS. Medical charts of the patients were reviewed. The initial clinical manifestations of the patients were correlated with results of biochemical tests. @*Results@#There were four cases of ASLD and one case of ASSD reviewed in this study. All cases of ASLD were confirmed by the presence of argininosuccinic acid (ASA) in the urine metabolic screen (UMS). The plasma citrulline level of the ASSD patient is significantly elevated as compared to the ASLD patients (2,690 µmol/L; NV: 10-45 µmol/L). The ASSD patient and one ASLD patient were symptomatic within the first six days of life. Both presented with significantly elevated plasma ammonia, citrulline and glutamine levels. Three ASLD patients were asymptomatic on initial screening. @*Conclusion@#ENBS has shown importance in the early detection and management of ASSD and ASLD. Early initiation of management may prevent hyperammonemic crises. Long term outcome studies are needed to look into the correlation of neurodevelopmental outcome with lifelong accumulation of citrulline and glutamine in ASSD and ASA in ASLD.


Subject(s)
Citrullinemia , Argininosuccinic Aciduria , Argininosuccinic Acid
8.
Article in English | MEDLINE | ID: mdl-31518899

ABSTRACT

Plasma elevations of the amino acids alloisoleucine and argininosuccinic acid (ASA) are pathognomonic for maple syrup urine disease and argininosuccinate lyase deficiency, respectively. Reliable detection of these biomarkers is typically achieved using methods with tedious sample preparations or long chromatographic separations, and many published amino acid assays report poor specificity and/or sensitivity for one or both of these compounds. This report describes a novel liquid chromatography tandem mass spectrometry (LC-MS/MS) method that provides rapid quantification of alloisoleucine and ASA in human plasma. The basis of this method is a mixed-mode solid phase separation that achieves baseline resolution of alloisoleucine from isobaric interferents without the use of derivatization or ion pairing agents. The inject-to-inject time is 6 min including elution, column washing and re-equilibration. Validation studies demonstrate excellent limits of quantification (1 µmol/L), linearity (r = 0.999 from 1 to 250 µmol/L), accuracy (bias = -3.8% and -10.1%), and inter-assay imprecision (CV < 8.06%) for plasma analyses. Data from long-term clinical application confirms chromatographic consistency equivalent to more traditional reversed-phase or HILIC based columns. Additional matrix studies indicate low suppression (<10%) for a wide range of amino acids and compatibility with other matrixes such as blood spot analyses. Finally, analysis of our first 257 clinical specimens demonstrates high analytic specificity and sensitivity, allowing the detection of subtle but clinically relevant elevations of alloisoleucine and ASA that may be missed by other less sensitive methods. In conclusion, the novel LC-MS/MS method reported here overcomes a number of the challenges associated with alloisoleucine and ASA quantification. Combining this approach with published incomplete amino acid quantification methods allows, for the first time, a rapid and comprehensive LC-MS/MS analysis of underivatized amino acids without the use of ion pairing agents.


Subject(s)
Argininosuccinic Acid/blood , Chromatography, Liquid/methods , Isoleucine/blood , Tandem Mass Spectrometry/methods , Argininosuccinic Acid/chemistry , Humans , Isoleucine/chemistry , Linear Models , Reproducibility of Results , Sensitivity and Specificity
9.
J Inherit Metab Dis ; 42(6): 1077-1087, 2019 11.
Article in English | MEDLINE | ID: mdl-30907007

ABSTRACT

The urea cycle disorder (UCD) argininosuccinate lyase (ASL) deficiency, caused by a defective ASL enzyme, exhibits a wide range of phenotypes, from life-threatening neonatal hyperammonemia to asymptomatic patients, with only the biochemical marker argininosuccinic acid (ASA) elevated in body fluids. Remarkably, even without ever suffering from hyperammonemia, patients often develop severe cognitive impairment and seizures. The goal of this study was to understand the effect on the known toxic metabolite ASA and the assumed toxic metabolite guanidinosuccinic acid (GSA) on developing brain cells, and to evaluate the potential role of creatine (Cr) supplementation, as it was described protective for brain cells exposed to ammonia. We used an in vitro model, in which we exposed three-dimensional (3D) organotypic rat brain cell cultures in aggregates to different combinations of the metabolites of interest at two time points (representing two different developmental stages). After harvest and cryopreservation of the cell cultures, the samples were analyzed mainly by metabolite analysis, immunohistochemistry, and western blotting. ASA and GSA were found toxic for astrocytes and neurons. This toxicity could be reverted in vitro by Cr. As well, an antiapoptotic effect of ASA was revealed, which could contribute to the neurotoxicity in ASL deficiency. Further studies in human ASL deficiency will be required to understand the biochemical situation in the brain of affected patients, and to investigate the impact of high or low arginine doses on brain Cr availability. In addition, clinical trials to evaluate the beneficial effect of Cr supplementation in ASL deficiency would be valuable.


Subject(s)
Argininosuccinic Acid/toxicity , Argininosuccinic Aciduria/pathology , Argininosuccinic Aciduria/prevention & control , Brain/pathology , Creatine/pharmacology , Neurotoxicity Syndromes/pathology , Animals , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/metabolism , Brain/drug effects , Brain/metabolism , Cells, Cultured , Humans , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/metabolism , Organ Culture Techniques/methods , Rats , Tissue Scaffolds/chemistry
10.
Br J Cancer ; 117(12): 1855-1864, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29073636

ABSTRACT

BACKGROUND: Uterine leiomyomas can be classified into molecularly distinct subtypes according to their genetic triggers: MED12 mutations, HMGA2 upregulation, or inactivation of FH. The aim of this study was to identify metabolites and metabolic pathways that are dysregulated in different subtypes of leiomyomas. METHODS: We performed global metabolomic profiling of 25 uterine leiomyomas and 17 corresponding myometrium specimens using liquid chromatography-tandem mass spectroscopy. RESULTS: A total of 641 metabolites were detected. All leiomyomas displayed reduced homocarnosine and haeme metabolite levels. We identified a clearly distinct metabolomic profile for leiomyomas of the FH subtype, characterised by metabolic alterations in the tricarboxylic acid cycle and pentose phosphate pathways, and increased levels of multiple lipids and amino acids. Several metabolites were uniquely elevated in leiomyomas of the FH subtype, including N6-succinyladenosine and argininosuccinate, serving as potential biomarkers for FH deficiency. In contrast, leiomyomas of the MED12 subtype displayed reduced levels of vitamin A, multiple membrane lipids and amino acids, and dysregulation of vitamin C metabolism, a finding which was also compatible with gene expression data. CONCLUSIONS: The study reveals the metabolomic heterogeneity of leiomyomas and provides the requisite framework for strategies designed to target metabolic alterations promoting the growth of these prevalent tumours.


Subject(s)
Leiomyoma/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Amino Acids/metabolism , Argininosuccinic Acid/metabolism , Ascorbic Acid/metabolism , Citric Acid Cycle , Female , Fumarate Hydratase/genetics , HMGA2 Protein/genetics , Humans , Leiomyoma/genetics , Lipid Metabolism , Mediator Complex/genetics , Metabolic Networks and Pathways , Metabolome , Pentose Phosphate Pathway , Vitamin A/metabolism
11.
IUBMB Life ; 69(11): 896-907, 2017 11.
Article in English | MEDLINE | ID: mdl-29044950

ABSTRACT

Arginine biosynthesis pathway is crucial to the survival and pathogenesis of Mycobacterium tuberculosis (Mtb). Arginine is a critical amino acid that contributes to the inflection of cellular immune responses during pathogenesis. Argininosuccinate lyase from Mtb (MtArgH), the last enzyme in the pathway, catalyzes the production of arginine from argininosuccinic acid. MtArgH is an essential enzyme for the growth and survival of M. tuberculosis. We biochemically characterized MtArgH and deciphered the role of a previously unexplored cysteine (Cys441 ) residue at the C-terminal region of the protein. Chemical modification of Cys441 completely abrogated the enzymatic activity suggesting its involvement in the catalytic mechanism. Replacement of Cys441 to alanine showed a striking decrease in the enzymatic activity, while retaining the overall secondary to quaternary structure of the protein, hence corroborating the involvement of Cys441 in the process of catalysis. Interestingly, replacement of Cys441 to serine, showed significant increase in activity, as compared to the wild-type MtArgH. Inactivity of C441 A and elevated activity of its conservative mutant (C441 S) confirmed the participation of Cys441 in the MtArgH activity. We also, observed that C441 S mutant has higher thermal stability and maintains significant activity at high temperatures. This is in concordance with our observation that Cys441 in Mtb is replaced by a serine in the ArgH from thermophilic microorganisms. Furthermore, we also propose a potential feedback mechanism, wherein the Cys441 is covalently modified to S-(2-succinyl) cysteine (succination) by one of the products, fumarate, thereby inactivating MtArgH. These insights into the mechanism of MtArgH activity unravel novel regulations of arginine biosynthetic pathway in Mtb. © 2017 IUBMB Life, 69(11):896-907, 2017.


Subject(s)
Argininosuccinate Lyase/chemistry , Bacterial Proteins/chemistry , Cysteine/chemistry , Mycobacterium tuberculosis/enzymology , Serine/chemistry , Amino Acid Sequence , Arginine/metabolism , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/metabolism , Argininosuccinic Acid/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biocatalysis , Cloning, Molecular , Cysteine/metabolism , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Kinetics , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/chemistry , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serine/metabolism , Structure-Activity Relationship , Substrate Specificity
12.
J Inherit Metab Dis ; 40(3): 357-368, 2017 05.
Article in English | MEDLINE | ID: mdl-28251416

ABSTRACT

OBJECTIVES: This UK-wide study defines the natural history of argininosuccinic aciduria and compares long-term neurological outcomes in patients presenting clinically or treated prospectively from birth with ammonia-lowering drugs. METHODS: Retrospective analysis of medical records prior to March 2013, then prospective analysis until December 2015. Blinded review of brain MRIs. ASL genotyping. RESULTS: Fifty-six patients were defined as early-onset (n = 23) if symptomatic < 28 days of age, late-onset (n = 23) if symptomatic later, or selectively screened perinatally due to a familial proband (n = 10). The median follow-up was 12.4 years (range 0-53). Long-term outcomes in all groups showed a similar neurological phenotype including developmental delay (48/52), epilepsy (24/52), ataxia (9/52), myopathy-like symptoms (6/52) and abnormal neuroimaging (12/21). Neuroimaging findings included parenchymal infarcts (4/21), focal white matter hyperintensity (4/21), cortical or cerebral atrophy (4/21), nodular heterotopia (2/21) and reduced creatine levels in white matter (4/4). 4/21 adult patients went to mainstream school without the need of additional educational support and 1/21 lives independently. Early-onset patients had more severe involvement of visceral organs including liver, kidney and gut. All early-onset and half of late-onset patients presented with hyperammonaemia. Screened patients had normal ammonia at birth and received treatment preventing severe hyperammonaemia. ASL was sequenced (n = 19) and 20 mutations were found. Plasma argininosuccinate was higher in early-onset compared to late-onset patients. CONCLUSIONS: Our study further defines the natural history of argininosuccinic aciduria and genotype-phenotype correlations. The neurological phenotype does not correlate with the severity of hyperammonaemia and plasma argininosuccinic acid levels. The disturbance in nitric oxide synthesis may be a contributor to the neurological disease. Clinical trials providing nitric oxide to the brain merit consideration.


Subject(s)
Argininosuccinic Aciduria/pathology , Argininosuccinic Aciduria/therapy , Adolescent , Adult , Ammonia/metabolism , Argininosuccinic Acid/blood , Argininosuccinic Aciduria/blood , Argininosuccinic Aciduria/genetics , Child , Child, Preschool , Female , Follow-Up Studies , Genotype , Humans , Hyperammonemia/metabolism , Hyperammonemia/pathology , Infant , Infant, Newborn , Male , Middle Aged , Mutation/genetics , Phenotype , Prospective Studies , Retrospective Studies , Young Adult
13.
Eksp Klin Farmakol ; 79(2): 34-9, 2016.
Article in Russian | MEDLINE | ID: mdl-27416681

ABSTRACT

The influence of angioprotector and endothelium-protector drugs pentoxifylline and unifuzol as components of supportive therapy on the efficacy of combined cytostatic treatment has been experimentally studied. It is established that pentoxifylline and unifuzol do not affect the antitumor and antimetastatic activity of doxorubicin and cyclophosphan with respect to Pliss lymphosarcoma and Walker 256 carcinosarcoma, and in some cases even potentiate the effect of cytostatic therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Argininosuccinic Acid/pharmacology , Carcinoma 256, Walker/drug therapy , Cytostatic Agents/pharmacology , Lymphoma, Non-Hodgkin/drug therapy , Pentoxifylline/pharmacology , Animals , Carcinoma 256, Walker/pathology , Cell Proliferation/drug effects , Cyclophosphamide/pharmacology , Doxorubicin/pharmacology , Drug Synergism , Lymphoma, Non-Hodgkin/pathology , Male , Rats , Rats, Wistar , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Immunity ; 42(3): 419-30, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25786174

ABSTRACT

Macrophage polarization involves a coordinated metabolic and transcriptional rewiring that is only partially understood. By using an integrated high-throughput transcriptional-metabolic profiling and analysis pipeline, we characterized systemic changes during murine macrophage M1 and M2 polarization. M2 polarization was found to activate glutamine catabolism and UDP-GlcNAc-associated modules. Correspondingly, glutamine deprivation or inhibition of N-glycosylation decreased M2 polarization and production of chemokine CCL22. In M1 macrophages, we identified a metabolic break at Idh, the enzyme that converts isocitrate to alpha-ketoglutarate, providing mechanistic explanation for TCA cycle fragmentation. (13)C-tracer studies suggested the presence of an active variant of the aspartate-arginosuccinate shunt that compensated for this break. Consistently, inhibition of aspartate-aminotransferase, a key enzyme of the shunt, inhibited nitric oxide and interleukin-6 production in M1 macrophages, while promoting mitochondrial respiration. This systems approach provides a highly integrated picture of the physiological modules supporting macrophage polarization, identifying potential pharmacologic control points for both macrophage phenotypes.


Subject(s)
Gene Regulatory Networks/immunology , Immunity, Innate , Macrophages/metabolism , Mitochondria/metabolism , Transcription, Genetic/immunology , Animals , Argininosuccinic Acid/immunology , Argininosuccinic Acid/metabolism , Aspartate Aminotransferase, Mitochondrial/genetics , Aspartate Aminotransferase, Mitochondrial/immunology , Aspartic Acid/immunology , Aspartic Acid/metabolism , Chemokine CCL22/genetics , Chemokine CCL22/immunology , Citric Acid Cycle , Gene Expression Regulation , Glutamine/deficiency , Glycosylation , Interleukin-6/genetics , Interleukin-6/immunology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Macrophages/classification , Macrophages/cytology , Macrophages/immunology , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/immunology , Mice , Mitochondria/genetics , Mitochondria/immunology , Nitric Oxide/immunology , Nitric Oxide/metabolism , Signal Transduction , Uridine Diphosphate N-Acetylglucosamine/immunology , Uridine Diphosphate N-Acetylglucosamine/metabolism
15.
Cell Rep ; 3(5): 1440-8, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23643539

ABSTRACT

The identification of mutated metabolic enzymes in hereditary cancer syndromes has established a direct link between metabolic dysregulation and cancer. Mutations in the Krebs cycle enzyme, fumarate hydratase (FH), predispose affected individuals to leiomyomas, renal cysts, and cancers, though the respective pathogenic roles of mitochondrial and cytosolic FH isoforms remain undefined. On the basis of comprehensive metabolomic analyses, we demonstrate that FH1-deficient cells and tissues exhibit defects in the urea cycle/arginine metabolism. Remarkably, transgenic re-expression of cytosolic FH ameliorated both renal cyst development and urea cycle defects associated with renal-specific FH1 deletion in mice. Furthermore, acute arginine depletion significantly reduced the viability of FH1-deficient cells in comparison to controls. Our findings highlight the importance of extramitochondrial metabolic pathways in FH-associated oncogenesis and the urea cycle/arginine metabolism as a potential therapeutic target.


Subject(s)
Fumarate Hydratase/metabolism , Kidney Neoplasms/enzymology , Animals , Arginine/metabolism , Argininosuccinic Acid/metabolism , Cell Line , Citric Acid Cycle , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Fumarates/metabolism , Kidney/enzymology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Metabolome , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Mutation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Urea/metabolism
16.
Mol Genet Metab ; 107(3): 315-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23040521

ABSTRACT

OBJECTIVE: To compare the effects of combinatorial therapy with low-dose arginine and a nitrogen scavenging agent (sodium phenylbutyrate) vs. monotherapy with high-dose arginine on liver function tests in patients with argininosuccinic aciduria (ASA). STUDY DESIGN: Twelve patients with ASA were enrolled in a double-blind, placebo-controlled, cross-over study design. Subjects were randomized to receive either a low-dose of arginine therapy (100 mg · kg(-1) · d(-1)) combined with sodium phenylbutyrate (500 mg · kg(-1) · d(-1)) (LDA arm) or a high-dose of arginine alone (500 mg · kg(-1) · d(-1)) (HDA arm) for one week. At the end of one week of therapy, liver function tests were assessed and metabolite fluxes were measured using a multi-tracer stable isotope protocol. RESULTS: Plasma aspartate aminotransferase (AST), alanine aminotransferase (ALT), and measures of synthetic functions of the liver were the primary outcomes. Subjects had significantly increased levels of argininosuccinate (P<0.03) and AST levels (P<0.01) after treatment with high-dose arginine. In the subset of subjects with elevated AST or ALT, treatment with high-dose of arginine was associated with further increases in plasma levels of both aminotransferases. Whereas subjects had increased arginine and citrulline flux with high-dose arginine therapy, the glutamine flux was not different between the two treatment arms. The synthetic liver functions as assessed by prothrombin time, INR, and coagulation factor levels were not different between the HDA and LDA arms. CONCLUSIONS: Administering higher doses of arginine in subjects with ASA results in increases in AST and ALT levels, especially in the subset of patients with elevated baseline aminotransferases. Hence, low-dose arginine sufficient to normalize arginine levels in plasma combined with nitrogen scavenging therapy should be considered as a therapeutic option for treatment of ASA in patients with elevations of hepatic aminotransferases.


Subject(s)
Arginine/therapeutic use , Argininosuccinic Aciduria/drug therapy , Phenylbutyrates/therapeutic use , Adolescent , Alanine Transaminase/blood , Arginine/blood , Argininosuccinic Acid/blood , Argininosuccinic Aciduria/blood , Aspartate Aminotransferases/blood , Child , Child, Preschool , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Liver Function Tests , Male , Phenylbutyrates/blood , Placebos , Young Adult
17.
Genet Med ; 14(5): 501-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22241104

ABSTRACT

The urea cycle consists of six consecutive enzymatic reactions that convert waste nitrogen into urea. Deficiencies of any of these enzymes of the cycle result in urea cycle disorders (UCDs), a group of inborn errors of hepatic metabolism that often result in life-threatening hyperammonemia. Argininosuccinate lyase (ASL) catalyzes the fourth reaction in this cycle, resulting in the breakdown of argininosuccinic acid to arginine and fumarate. ASL deficiency (ASLD) is the second most common UCD, with a prevalence of ~1 in 70,000 live births. ASLD can manifest as either a severe neonatal-onset form with hyperammonemia within the first few days after birth or as a late-onset form with episodic hyperammonemia and/or long-term complications that include liver dysfunction, neurocognitive deficits, and hypertension. These long-term complications can occur in the absence of hyperammonemic episodes, implying that ASL has functions outside of its role in ureagenesis and the tissue-specific lack of ASL may be responsible for these manifestations. The biochemical diagnosis of ASLD is typically established with elevation of plasma citrulline together with elevated argininosuccinic acid in the plasma or urine. Molecular genetic testing of ASL and assay of ASL enzyme activity are helpful when the biochemical findings are equivocal. However, there is no correlation between the genotype or enzyme activity and clinical outcome. Treatment of acute metabolic decompensations with hyperammonemia involves discontinuing oral protein intake, supplementing oral intake with intravenous lipids and/or glucose, and use of intravenous arginine and nitrogen-scavenging therapy. Dietary restriction of protein and dietary supplementation with arginine are the mainstays in long-term management. Orthotopic liver transplantation (OLT) is best considered only in patients with recurrent hyperammonemia or metabolic decompensations resistant to conventional medical therapy.


Subject(s)
Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Arginine/metabolism , Arginine/therapeutic use , Argininosuccinate Lyase/genetics , Argininosuccinic Acid/blood , Argininosuccinic Acid/metabolism , Argininosuccinic Acid/urine , Argininosuccinic Aciduria/therapy , Child, Preschool , Citrulline/blood , Cognition Disorders/enzymology , Cognition Disorders/genetics , Diet, Protein-Restricted , Fumarates/metabolism , Genetic Testing , Glucose/therapeutic use , Humans , Hyperammonemia/enzymology , Hyperammonemia/genetics , Hypertension/enzymology , Hypertension/genetics , Infant , Infant, Newborn , Lipids/therapeutic use , Liver Diseases/enzymology , Liver Diseases/genetics , Liver Transplantation , Neonatal Screening , Phenylbutyrates/therapeutic use , Sodium Benzoate/therapeutic use
18.
Am J Med Genet C Semin Med Genet ; 157C(1): 45-53, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21312326

ABSTRACT

The urea cycle consists of six consecutive enzymatic reactions that convert waste nitrogen into urea. Deficiencies of any of these enzymes of the cycle result in urea cycle disorders (UCD), a group of inborn errors of hepatic metabolism that often result in life threatening hyperammonemia. Argininosuccinate lyase (ASL) is a cytosolic enzyme which catalyzes the fourth reaction in the cycle and the first degradative step, that is, the breakdown of argininosuccinic acid to arginine and fumarate. Deficiency of ASL results in an accumulation of argininosuccinic acid in tissues, and excretion of argininosuccinic acid in urine leading to the condition argininosuccinic aciduria (ASA). ASA is an autosomal recessive disorder and is the second most common UCD. In addition to the accumulation of argininosuccinic acid, ASL deficiency results in decreased synthesis of arginine, a feature common to all UCDs except argininemia. Arginine is not only the precursor for the synthesis of urea and ornithine as part of the urea cycle but it is also the substrate for the synthesis of nitric oxide, polyamines, proline, glutamate, creatine, and agmatine. Hence, while ASL is the only enzyme in the body able to generate arginine, at least four enzymes use arginine as substrate: arginine decarboxylase, arginase, nitric oxide synthetase (NOS) and arginine/glycine aminotransferase. In the liver, the main function of ASL is ureagenesis, and hence, there is no net synthesis of arginine. In contrast, in most other tissues, its role is to generate arginine that is designated for the specific cell's needs. While patients with ASA share the acute clinical phenotype of hyperammonemia, encephalopathy, and respiratory alkalosis common to other UCD, they also present with unique chronic complications most probably caused by a combination of tissue specific deficiency of arginine and/or elevation of argininosuccinic acid. This review article summarizes the clinical characterization, biochemical, enzymatic, and molecular features of this disorder. Current treatment, prenatal diagnosis, diagnosis through the newborn screening as well as hypothesis driven future treatment modalities are discussed.


Subject(s)
Argininosuccinate Lyase/genetics , Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Argininosuccinic Aciduria/metabolism , Arginase/genetics , Arginase/metabolism , Arginine/genetics , Arginine/metabolism , Argininosuccinic Acid/metabolism , Argininosuccinic Aciduria/therapy , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Humans , Hyperammonemia/enzymology , Hyperammonemia/genetics , Hyperammonemia/metabolism , Infant, Newborn , Liver Diseases/enzymology , Liver Diseases/genetics , Liver Diseases/metabolism , Neonatal Screening , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Ornithine/genetics , Ornithine/metabolism , Urea Cycle Disorders, Inborn/enzymology , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/metabolism
19.
Rev. Fac. Med. (Bogotá) ; 58(4): 316-323, oct.-dic. 2010.
Article in Spanish | LILACS | ID: lil-613148

ABSTRACT

Los defectos del ciclo de la úrea se deben a deficiencias de diferentes enzimas; las manifestaciones clínicas son similares y están relacionadas con la hiperamonemia. Se presentan las historias clínicas de tres neonatos a término, sin evidencia de alteración al nacimiento. Se les detectó hiperamonemia y se sospechó enfermedad metabólica. La cromatografía de aminoácidos sugirió defectos del ciclo de la úrea. El manejo incluyó dieta con restricción de proteínas, administración de benzoato de sodio, exsanguinotransfusión y diálisis peritoneal pese a lo cual fallecieron. Se revisan las causas de hiperamonemia en el neonato y se propone una secuencia para su diagnóstico.


Subject(s)
Infant, Newborn , Brain Diseases , Citrullinemia , Hyperammonemia , Infant, Newborn , Metabolism, Inborn Errors , Argininosuccinic Acid
20.
Malays J Pathol ; 32(2): 87-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21329179

ABSTRACT

Argininosuccinic aciduria is an inborn error of the urea cycle caused by deficiency of argininosuccinate lyase (ASL). ASL-deficient patients present with progressive intoxication due to accumulation of ammonia in the body. Early diagnosis and treatment of hyperammonemia are necessary to improve survival and prevent long-term handicap. Two clinical phenotypes have been recognized--neonatal acute and milder late-onset form. We investigated patients with hyperammonemia by a stepwise approach in which quantitative amino acids analysis was the core diagnostic procedure. Here, we describe the clinical phenotypes and biochemical characteristics in diagnosing this group of patients. We have identified 13 patients with argininosuccinic aciduria from 2003 till 2009. Ten patients who presented with acute neonatal hyperammonemic encephalopathy had markedly elevated blood ammonia (> 430 micromol/L) within the first few days of life. Three patients with late-onset disease had more subtle clinical presentations and they developed hyperammonemia only during the acute catabolic state at two to twelve months of age. Their blood ammonia was mild to moderately elevated (> 75-265 micromol/L). The diagnosis was confirmed by detection of excessive levels of argininosuccinate in the urine and/or plasma. They also have moderately increased levels of citrulline and, low levels of arginine and ornithine in their plasma. Two patients succumbed to the disease. To date, eleven patients remained well on a dietary protein restriction, oral ammonia scavenging drugs and arginine supplementation. The majority of them have a reasonable good neurological outcome.


Subject(s)
Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/metabolism , Argininosuccinic Aciduria/physiopathology , Age of Onset , Amino Acids/analysis , Argininosuccinic Acid/blood , Argininosuccinic Acid/urine , Female , Humans , Infant , Infant, Newborn , Malaysia , Male , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...