Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Redox Biol ; 73: 103192, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38776754

ABSTRACT

BACKGROUND: In animals, dietary sulfur amino acid restriction (SAAR) improves metabolic health, possibly mediated by altering sulfur amino acid metabolism and enhanced anti-obesogenic processes in adipose tissue. AIM: To assess the effects of SAAR over time on the plasma and urine SAA-related metabolites (sulfurome) in humans with overweight and obesity, and explore whether such changes were associated with body weight, body fat and adipose tissue gene expression. METHODS: Fifty-nine subjects were randomly allocated to SAAR (∼2 g SAA, n = 31) or a control diet (∼5.6 g SAA, n = 28) consisting of plant-based whole-foods and supplemented with capsules to titrate contents of SAA. Sulfurome metabolites in plasma and urine at baseline, 4 and 8 weeks were measured using HPLC and LC-MS/MS. mRNA-sequencing of subcutaneous white adipose tissue (scWAT) was performed to assess changes in gene expression. Data were analyzed with mixed model regression. Principal component analyses (PCA) were performed on the sulfurome data to identify potential signatures characterizing the response to SAAR. RESULTS: SAAR led to marked decrease of the main urinary excretion product sulfate (p < 0.001) and plasma and/or 24-h urine concentrations of cystathionine, sulfite, thiosulfate, H2S, hypotaurine and taurine. PCA revealed a distinct metabolic signature related to decreased transsulfuration and H2S catabolism that predicted greater weight loss and android fat mass loss in SAAR vs. controls (all pinteraction < 0.05). This signature correlated positively with scWAT expression of genes in the tricarboxylic acid cycle, electron transport and ß-oxidation (FDR = 0.02). CONCLUSION: SAAR leads to distinct alterations of the plasma and urine sulfurome in humans, and predicted increased loss of weight and android fat mass, and adipose tissue lipolytic gene expression in scWAT. Our data suggest that SAA are linked to obesogenic processes and that SAAR may be useful for obesity and related disorders. TRIAL IDENTIFIER: https://clinicaltrials.gov/study/NCT04701346.

2.
Redox Biol ; 58: 102517, 2022 12.
Article in English | MEDLINE | ID: mdl-36306676

ABSTRACT

Regulation of H2S homeostasis in humans is poorly understood. Therefore, we assessed the importance of individual enzymes in synthesis and catabolism of H2S by studying patients with respective genetic defects. We analyzed sulfur compounds (including bioavailable sulfide) in 37 untreated or insufficiently treated patients with seven ultrarare enzyme deficiencies and compared them to 63 controls. Surprisingly, we observed that patients with severe deficiency in cystathionine ß-synthase (CBS) or cystathionine γ-lyase (CSE) - the enzymes primarily responsible for H2S synthesis - exhibited increased and normal levels of bioavailable sulfide, respectively. However, an approximately 21-fold increase of urinary homolanthionine in CBS deficiency strongly suggests that lacking CBS activity is compensated for by an increase in CSE-dependent H2S synthesis from accumulating homocysteine, which suggests a control of H2S homeostasis in vivo. In deficiency of sulfide:quinone oxidoreductase - the first enzyme in mitochondrial H2S oxidation - we found normal H2S concentrations in a symptomatic patient and his asymptomatic sibling, and elevated levels in an asymptomatic sibling, challenging the requirement for this enzyme in catabolizing H2S under physiological conditions. Patients with ethylmalonic encephalopathy and sulfite oxidase/molybdenum cofactor deficiencies exhibited massive accumulation of thiosulfate and sulfite with formation of large amounts of S-sulfocysteine and S-sulfohomocysteine, increased renal losses of sulfur compounds and concomitant strong reduction in plasma total cysteine. Our results demonstrate the value of a comprehensive assessment of sulfur compounds in severe disorders of homocysteine/cysteine metabolism and provide evidence for redundancy and compensatory mechanisms in the maintenance of H2S homeostasis.


Subject(s)
Hydrogen Sulfide , Humans , Hydrogen Sulfide/metabolism , Cysteine , Sulfides/metabolism , Homeostasis , Sulfur , Homocysteine
3.
Viruses ; 13(12)2021 12 14.
Article in English | MEDLINE | ID: mdl-34960774

ABSTRACT

The chicken Tva cell surface protein, a member of the low-density lipoprotein receptor family, has been identified as an entry receptor for avian leukosis virus of classic subgroup A and newly emerging subgroup K. Because both viruses represent an important concern for the poultry industry, we introduced a frame-shifting deletion into the chicken tva locus with the aim of knocking-out Tva expression and creating a virus-resistant chicken line. The tva knock-out was prepared by CRISPR/Cas9 gene editing in chicken primordial germ cells and orthotopic transplantation of edited cells into the testes of sterilized recipient roosters. The resulting tva -/- chickens tested fully resistant to avian leukosis virus subgroups A and K, both in in vitro and in vivo assays, in contrast to their susceptible tva +/+ and tva +/- siblings. We also found a specific disorder of the cobalamin/vitamin B12 metabolism in the tva knock-out chickens, which is in accordance with the recently recognized physiological function of Tva as a receptor for cobalamin in complex with transcobalamin transporter. Last but not least, we bring a new example of the de novo resistance created by CRISPR/Cas9 editing of pathogen dependence genes in farm animals and, furthermore, a new example of gene editing in chicken.


Subject(s)
Avian Leukosis Virus/physiology , Avian Proteins/physiology , Chickens/virology , Receptors, Virus/physiology , Vitamin B 12/metabolism , Animals , Avian Leukosis Virus/classification , Avian Proteins/genetics , Chick Embryo , Female , Frameshift Mutation , Gene Editing , Gene Knockout Techniques , Male , Methylmalonic Acid/blood , Receptors, Virus/genetics
4.
Biochim Biophys Acta Mol Basis Dis ; 1867(10): 166201, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34147638

ABSTRACT

Combined methylmalonic aciduria with homocystinuria (cblC type) is a rare disease caused by mutations in the MMACHC gene. MMACHC encodes an enzyme crucial for intracellular vitamin B12 metabolism, leading to the accumulation of toxic metabolites e.g. methylmalonic acid (MMA) and homocysteine (Hcy), and secondary disturbances in folate and one-carbon metabolism when not fully functional. Patients with cblC deficiency often present in the neonatal or early childhood period with a severe multisystem pathology, which comprises a broad spectrum of treatment-resistant ophthalmological phenotypes, including retinal degeneration, impaired vision, and vascular changes. To examine the potential function of MMACHC in the retina and how its loss may impact disease, we performed gene expression studies in human and mouse, which showed that local expression of MMACHC in the retina and retinal pigment epithelium is relatively stable over time. To study whether functional MMACHC is required for retinal function and tissue integrity, we generated a transgenic mouse lacking Mmachc expression in cells of the peripheral retina. Characterization of this mouse revealed accumulation of cblC disease related metabolites, including MMA and the folate-dependent purine synthesis intermediates AICA-riboside and SAICA-riboside in the retina. Nevertheless, fundus appearance, morphology, vasculature, and cellular composition of the retina, as well as ocular function, remained normal in mice up to 6 or 12 months of age. Our data indicates that peripheral retinal neurons do not require intrinsic expression of Mmachc for survival and function and questions whether a local MMACHC deficiency is responsible for the retinal phenotypes in patients.


Subject(s)
Oxidoreductases/metabolism , Retina/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Female , Homocysteine/metabolism , Homocystinuria/metabolism , Humans , Male , Methylmalonic Acid/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Mutation/genetics , Oxidoreductases/genetics , Phenotype , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Vitamin B 12/metabolism , Young Adult
5.
J Inherit Metab Dis ; 44(3): 677-692, 2021 05.
Article in English | MEDLINE | ID: mdl-33295057

ABSTRACT

Cystathionine ß-synthase (CBS) deficiency has a wide clinical spectrum, ranging from neurodevelopmental problems, lens dislocation and marfanoid features in early childhood to adult onset disease with predominantly thromboembolic complications. We have analysed clinical and laboratory data at the time of diagnosis in 328 patients with CBS deficiency from the E-HOD (European network and registry for Homocystinurias and methylation Defects) registry. We developed comprehensive criteria to classify patients into four groups of pyridoxine responsivity: non-responders (NR), partial, full and extreme responders (PR, FR and ER, respectively). All groups showed overlapping concentrations of plasma total homocysteine while pyridoxine responsiveness inversely correlated with plasma/serum methionine concentrations. The FR and ER groups had a later age of onset and diagnosis and a longer diagnostic delay than NR and PR patients. Lens dislocation was common in all groups except ER but the age of dislocation increased with increasing responsiveness. Developmental delay was commonest in the NR group while no ER patient had cognitive impairment. Thromboembolism was the commonest presenting feature in ER patients, whereas it was least likely at presentation in the NR group. This probably is due to the differences in ages at presentation: all groups had a similar number of thromboembolic events per 1000 patient-years. Clinical severity of CBS deficiency depends on the degree of pyridoxine responsiveness. Therefore, a standardised pyridoxine-responsiveness test in newly diagnosed patients and a critical review of previous assessments is indispensable to ensure adequate therapy and to prevent or reduce long-term complications.


Subject(s)
Cystathionine beta-Synthase/deficiency , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Pyridoxine/therapeutic use , Adolescent , Adult , Aged , Child , Child, Preschool , Delayed Diagnosis , Europe , Female , Homocystinuria/enzymology , Humans , Infant , Linear Models , Male , Methionine/blood , Middle Aged , Phenotype , Registries , Severity of Illness Index , Young Adult
6.
Hum Mutat ; 41(9): 1662-1670, 2020 09.
Article in English | MEDLINE | ID: mdl-32623804

ABSTRACT

Classical homocystinuria (HCU) is an inborn error of metabolism caused by loss of cystathionine ß-synthase (CBS) activity with the concomitant buildup of homocysteine. In knockout (KO) mice, a mouse model of HCU, complete lack of CBS is neonatally lethal. Administration of OT-58, an enzyme therapy for HCU, during the first 5 weeks of life rescued KO mice survival by preventing liver disease. Here, we studied the impact of a long-term uninterrupted OT-58 treatment or its absence beyond the neonatal period on liver pathology and metabolism. Plasma and liver metabolites of KO mice on OT-58 treatment were substantially improved or normalized compared with those receiving vehicle. Increased plasma activities of alanine aminotransferase and aspartate aminotransferase of vehicle-injected KO mice suggested the progression of liver damage with age and lack of treatment. At 3 months of age, liver histology showed no signs of hepatopathy in both vehicle- and OT-58-treated KO mice. However, moderate to severe liver disease, characterized by steatosis, hepatocellular necroses, disorganized endoplasmic reticulum, and swollen mitochondria, developed in 6-month-old vehicle-injected KO mice. KO mice on OT-58 treatment remained asymptomatic and were indistinguishable from age-matched healthy controls. Long-term uninterrupted OT-58 treatment was essential to prevent severe liver disease in the KO mouse model of HCU.


Subject(s)
Enzyme Replacement Therapy , Homocystinuria/drug therapy , Liver Diseases/prevention & control , Animals , Disease Models, Animal , Female , Liver/pathology , Male , Mice , Mice, Knockout
8.
J Inherit Metab Dis ; 42(6): 1064-1076, 2019 11.
Article in English | MEDLINE | ID: mdl-30714172

ABSTRACT

The most common ureagenesis defect is X-linked ornithine transcarbamylase (OTC) deficiency which is a main target for novel therapeutic interventions. The spf ash mouse model carries a variant (c.386G>A, p.Arg129His) that is also found in patients. Male spf ash mice have a mild biochemical phenotype with low OTC activity (5%-10% of wild-type), resulting in elevated urinary orotic acid but no hyperammonemia. We recently established a dried blood spot method for in vivo quantification of ureagenesis by Gas chromatography-mass spectrometry (GC-MS) using stable isotopes. Here, we applied this assay to wild-type and spf ash mice to assess ureagenesis at different ages. Unexpectedly, we found an age-dependency with a higher capacity for ammonia detoxification in young mice after weaning. A parallel pattern was observed for carbamoylphosphate synthetase 1 and OTC enzyme expression and activities, which may act as pacemaker of this ammonia detoxification pathway. Moreover, high ureagenesis in younger mice was accompanied by elevated periportal expression of hepatic glutamine synthetase, another main enzyme required for ammonia detoxification. These observations led us to perform a more extensive analysis of the spf ash mouse in comparison to the wild-type, including characterization of the corresponding metabolites, enzyme activities in the liver and plasma and the gut microbiota. In conclusion, the comprehensive enzymatic and metabolic analysis of ureagenesis performed in the presented depth was only possible in animals. Our findings suggest such analyses being essential when using the mouse as a model and revealed age-dependent activity of ammonia detoxification.


Subject(s)
Aging/physiology , Ammonia/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/pathology , Ornithine Carbamoyltransferase/genetics , Urea/metabolism , Age Factors , Animals , Disease Models, Animal , Humans , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/pathology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Transgenic , Ornithine Carbamoyltransferase Deficiency Disease/genetics
9.
Br J Pharmacol ; 176(4): 594-606, 2019 02.
Article in English | MEDLINE | ID: mdl-30341787

ABSTRACT

BACKGROUND AND PURPOSE: Homocystinurias are rare genetic defects characterized by altered fluxes of sulfur compounds including homocysteine and cysteine. We explored whether the severely perturbed sulfur amino acid metabolism in patients with homocystinurias affects the metabolism of hydrogen sulfide. EXPERIMENTAL APPROACH: We studied 10 treated patients with a block in the conversion of homocysteine to cysteine due to cystathionine ß-synthase deficiency (CBSD) and six treated patients with remethylation defects (RMD) and an enhanced flux of sulfur metabolites via transsulfuration. Control groups for CBSD and RMD patients consisted of 22 patients with phenylketonuria on a low-protein diet and of 12 healthy controls respectively. Plasma and urine concentrations of selected sulfur compounds were analysed by HPLC and LC-MS/MS. KEY RESULTS: Patients with CBSD exhibited plasma concentrations of monobromobimane-detected sulfide similar to appropriate controls. Urinary homolanthionine and thiosulfate in CBSD were increased significantly 1.9 and 3 times suggesting higher hydrogen sulfide synthesis by γ-cystathionase and detoxification respectively. Surprisingly, patients with RMD had significantly lower plasma sulfide levels (53 and 64% of controls) with lower sulfite concentrations, and higher taurine and thiosulfate levels suggesting enhanced cysteine oxidation and hydrogen sulfide catabolism respectively. CONCLUSION AND IMPLICATIONS: The results from this study suggest that severe inherited defects in sulfur amino acid metabolism may be accompanied by only moderately perturbed hydrogen sulfide metabolism and lends support to the hypothesis that enzymes in the transsulfuration pathway may not be the major contributors to the endogenous hydrogen sulfide pool. LINKED ARTICLES: This article is part of a themed section on Chemical Biology of Reactive Sulfur Species. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.4/issuetoc.


Subject(s)
Homocystinuria/metabolism , Sulfur Compounds/metabolism , Adolescent , Adult , Child , Child, Preschool , Female , Homocystinuria/blood , Homocystinuria/urine , Humans , Infant , Male , Sulfur Compounds/blood , Sulfur Compounds/urine , Young Adult
10.
Toxicol Appl Pharmacol ; 353: 102-108, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29935280

ABSTRACT

BACKGROUND: The aim of our study was to identify the genetic background of thiopurine-induced toxicity in a patient with a wild-type thiopurine methyltransferase genotype and activity. A 38-year-old Caucasian woman presented with cutaneous necrotizing vasculitis pancytopenia one month after starting azathioprine therapy. METHODS: During a routine biochemical follow-up of the patient, undetectable serum uric acid (<10 µl) was observed. A high performance liquid chromatography analysis of urinary purines revealed increased levels of xanthine (137 mmol/mol creatinine). The suspected diagnosis of hereditary xanthinuria, a rare autosomal recessive disorder of the last two steps of purine metabolism, was confirmed by sequence analysis. RESULTS: An analysis of XDH/XO and AOX1 revealed common polymorphisms, while analysis of the MOCOS gene identified a rare homozygous variant c.362C > T. Dysfunction of this variant was confirmed by significantly decreased xanthine dehydrogenase/oxidase activity in the patient's plasma (<2% of control mean activity). CONCLUSIONS: We present a biochemical, enzymatic, and molecular genetic case study suggesting an important association between a hitherto undescribed dysfunction variant in the MOCOS gene and thiopurine-induced toxicity. The identified variant c.362C > T results in slower thiopurine metabolism caused by inhibition of 6-mercaptopurine oxidation (catabolism) to 6-thioxanthine and 6-thiouric acid, which increases the formation of the nucleotide 6-thioguanine, which is toxic. This is the first clinical case to identify the crucial role of the MOCOS gene in thiopurine intolerance and confirm the impact of genetic variability of purine enzymes on different therapeutic outcomes in patients undergoing thiopurine treatment.


Subject(s)
Aldehyde Oxidase/deficiency , Mercaptopurine/analogs & derivatives , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Sulfurtransferases/genetics , Xanthine Dehydrogenase/deficiency , Adult , Aldehyde Oxidase/genetics , Female , Humans , Mercaptopurine/adverse effects , Mercaptopurine/metabolism , Methyltransferases/genetics , Polymorphism, Genetic/genetics , Uric Acid/blood , Xanthine/urine , Xanthine Dehydrogenase/genetics , Xanthine Oxidase/genetics
11.
Virchows Arch ; 472(6): 1029-1039, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29623395

ABSTRACT

Ornithine transcarbamylase (OTC) deficiency is an X-linked disorder that causes recurrent and life-threatening episodes of hyperammonemia. The clinical picture in heterozygous females is highly diverse and derives from the genotype and the degree of inactivation of the mutated X chromosome in hepatocytes. Here, we describe molecular genetic, biochemical, and histopathological findings in the livers explanted from two female patients with late-onset OTC deficiency. Analysis of X-inactivation ratios by DNA methylation-based assays showed remarkable intra-organ variation ranging from 46:54 to 82:18 (average 70:30, n = 37), in favor of the active X chromosome carrying the mutation c.583G>C (p.G195R), in the first patient and from 75:25 to 90:10 (average 82:18, n = 20) in favor of the active X chromosome carrying the splicing mutation c.663+1G>A in the second patient. The X-inactivation ratios in liver samples correlated highly with the proportions of OTC-positive hepatocytes calculated from high-resolution image analyses of the immunohistochemically detected OTC in frozen sections that was performed on total area > 5 cm2. X-inactivation ratios in blood in both female patients corresponded to the lower limit of the liver values. Our data indicate that the proportion of about 20-30% of hepatocytes expressing the functional OTC protein is not sufficient to maintain metabolic stability. X-inactivation ratios assessed in liver biopsies taken from heterozygous females with X-linked disorders should not be considered representative of the whole liver.


Subject(s)
Chromosomes, Human, X/genetics , Glutamate-Ammonia Ligase/metabolism , Liver/enzymology , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase/metabolism , X Chromosome Inactivation , Biopsy , Female , Genotype , Glutamate-Ammonia Ligase/genetics , Heterozygote , Humans , Male , Ornithine Carbamoyltransferase/genetics , Sex Characteristics
12.
Mol Ther ; 26(3): 834-844, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29398487

ABSTRACT

Classical homocystinuria (HCU) is the most common inherited disorder of sulfur amino acid metabolism caused by deficiency in cystathionine beta-synthase (CBS) activity and characterized by severe elevation of homocysteine in blood and tissues. Treatment with dietary methionine restriction is not optimal, and poor compliance leads to serious complications. We developed an enzyme replacement therapy (ERT) and studied its efficacy in a severe form of HCU in mouse (the I278T model). Treatment was initiated before or after the onset of clinical symptoms in an effort to prevent or reverse the phenotype. ERT substantially reduced and sustained plasma homocysteine concentration at around 100 µM and normalized plasma cysteine for up to 9 months of treatment. Biochemical balance was also restored in the liver, kidney, and brain. Furthermore, ERT corrected liver glucose and lipid metabolism. The treatment prevented or reversed facial alopecia, fragile and lean phenotype, and low bone mass. In addition, structurally defective ciliary zonules in the eyes of I278T mice contained low density and/or broken fibers, while administration of ERT from birth partially rescued the ocular phenotype. In conclusion, ERT maintained an improved metabolic pattern and ameliorated many of the clinical complications in the I278T mouse model of HCU.


Subject(s)
Cystathionine beta-Synthase/administration & dosage , Enzyme Replacement Therapy , Homocystinuria/diagnosis , Homocystinuria/therapy , Phenotype , Amino Acids, Sulfur/blood , Amino Acids, Sulfur/metabolism , Animals , Cystathionine beta-Synthase/chemistry , Disease Models, Animal , Drug Evaluation, Preclinical , Glucose/metabolism , Homocystinuria/metabolism , Lipid Metabolism , Liver/drug effects , Liver/metabolism , Mice , Oxidative Stress , Polyethylene Glycols/chemistry
13.
Antioxid Redox Signal ; 28(4): 311-323, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28874062

ABSTRACT

AIMS: The transsulfuration pathway enzymes cystathionine beta-synthase (CBS) and cystathionine gamma-lyase are thought to be the major source of hydrogen sulfide (H2S). In this study, we assessed the role of CBS in H2S biogenesis. RESULTS: We show that despite discouraging enzyme kinetics of alternative H2S-producing reactions utilizing cysteine compared with the canonical condensation of serine and homocysteine, our simulations of substrate competitions at biologically relevant conditions suggest that cysteine is able to partially compete with serine on CBS, thus leading to generation of appreciable amounts of H2S. The leading H2S-producing reaction is condensation of cysteine with homocysteine, while cysteine desulfuration plays a dominant role when cysteine is more abundant than serine and homocysteine is limited. We found that the serine-to-cysteine ratio is the main determinant of CBS H2S productivity. Abundance of cysteine over serine, for example, in plasma, allowed for up to 43% of CBS activity being responsible for H2S production, while excess of serine typical for intracellular levels effectively limited such activity to less than 1.5%. CBS also produced lanthionine from serine and cysteine and a third of lanthionine coming from condensation of two cysteines contributed to the H2S pool. INNOVATION: Our study characterizes the H2S-producing potential of CBS under biologically relevant conditions and highlights the serine-to-cysteine ratio as the main determinant of H2S production by CBS in vivo. CONCLUSION: Our data clarify the function of CBS in H2S biogenesis and the role of thioethers as surrogate H2S markers. Antioxid. Redox Signal. 28, 311-323.


Subject(s)
Biomarkers/metabolism , Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/metabolism , Sulfides/metabolism , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/metabolism , Animals , Biomarkers/chemistry , Catalysis , Cystathionine beta-Synthase/chemistry , Cysteine/chemistry , Haplorhini , Homocysteine/chemistry , Hydrogen Sulfide/chemistry , Kinetics , Mice , Mice, Knockout , Serine/chemistry , Sulfides/chemistry , Sulfur/metabolism
14.
FASEB J ; 31(12): 5495-5506, 2017 12.
Article in English | MEDLINE | ID: mdl-28821635

ABSTRACT

Classical homocystinuria (HCU) is an inborn error of sulfur amino acid metabolism caused by deficient activity of cystathionine ß-synthase (CBS), resulting in an accumulation of homocysteine and a concomitant decrease of cystathionine and cysteine in blood and tissues. In mice, the complete lack of CBS is neonatally lethal. In this study, newborn CBS-knockout (KO) mice were treated with recombinant polyethyleneglycolylated human truncated CBS (PEG-CBS). Full survival of the treated KO mice, along with a positive impact on metabolite levels in plasma, liver, brain, and kidneys, was observed. The PEG-CBS treatment prevented an otherwise fatal liver disease characterized by steatosis, death of hepatocytes, and ultrastructural abnormalities of endoplasmic reticulum and mitochondria. Furthermore, treatment of the KO mice for 5 mo maintained the plasma metabolite balance and completely prevented osteoporosis and changes in body composition that characterize both the KO model and human patients. These findings argue that early treatment of patients with HCU with PEG-CBS may prevent clinical symptoms of the disease possibly without the need of dietary protein restriction.-Majtan, T., Hulková, H., Park, I., Krijt, J., Kozich, V., Bublil, E. M., Kraus, J. P. Enzyme replacement prevents neonatal death, liver damage, and osteoporosis in murine homocystinuria.


Subject(s)
Cystathionine beta-Synthase/metabolism , Cystathionine beta-Synthase/therapeutic use , Fatty Liver/prevention & control , Homocystinuria/drug therapy , Homocystinuria/enzymology , Liver Diseases/prevention & control , Osteoporosis/prevention & control , Animals , Body Composition/drug effects , Cystathionine beta-Synthase/genetics , Disease Models, Animal , Fatty Liver/enzymology , Female , Homocystinuria/metabolism , Homocystinuria/pathology , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver/pathology , Liver Diseases/enzymology , Male , Mice , Mice, Knockout , Recombinant Proteins/therapeutic use
15.
Clin Chem Lab Med ; 55(8): 1168-1177, 2017 Jul 26.
Article in English | MEDLINE | ID: mdl-28107167

ABSTRACT

BACKGROUND: Liver enzymes are released from hepatocytes into circulation and their activity can be measured in the blood. We examined whether the plasma activity of the liver enzyme ornithine carbamoyltransferase, determined by a novel liquid chromatography-mass spectrometry (LC-MS/MS) assay, could be utilized for the detection of OTC deficiency (OTCD), an X-linked inborn error of the urea cycle. METHODS: The plasma ornithine carbamoyltransferase (OTC) activity was assayed in the reverse reaction using isotopically labeled citrulline-d4 as a substrate and by determination of the product, ornithine-d4, by LC-MS/MS analysis. RESULTS: The plasma OTC activity in the controls was in the range of 111-658 pkat/L (n=49, median 272 pkat/L), and the activity increased linearly with serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities in patients with hepatopathy. The OTC activity was subsequently determined in 32 individuals carrying mutations in the OTC gene, and OTC/ALT and OTC/AST ratios were calculated to account for the degree of hepatopathy, which is a common finding in OTCD. The OTC/ALT ratio enabled clear differentiation of OTCD hemizygotes (n=11, range 0-69×10-6) from controls (504-3440×10-6). This ratio also enabled the detection of 11 of 12 symptomatic heterozygotes (range 38-794×10-6), while this marker did not allow for reliable differentiation of asymptomatic heterozygotes (n=9) from controls. CONCLUSIONS: LC-MS/MS assay of plasma OTC activity enabled the detection of all hemizygous and the majority of symptomatic heterozygous OTCD patients in the tested cohort. This study demonstrates that non-invasive assay of enzymes expressed predominantly in the liver could be used as an alternative approach for diagnosing inborn errors of metabolism.


Subject(s)
Enzyme Assays/methods , Liver/enzymology , Ornithine Carbamoyltransferase Deficiency Disease/blood , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Ornithine Carbamoyltransferase/blood , Calibration , Chromatography, Liquid , Chromosomes, Human, X/genetics , Cohort Studies , Enzyme Stability , Female , Heterozygote , Humans , Linear Models , Male , Mutation , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/enzymology , Tandem Mass Spectrometry
16.
J Clin Invest ; 126(6): 2372-84, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183385

ABSTRACT

Homocystinuria, which typically results from cystathionine ß-synthase (CBS) deficiency, is the most common defect of sulfur amino acid metabolism. CBS condenses homocysteine and serine to cystathionine that is then converted to cysteine. Individuals with homocystinuria have markedly elevated plasma levels of homocysteine and methionine and reduced concentrations of cystathionine and cysteine. Clinical disease manifestations include thromboembolism and neuropsychiatric, ocular, and skeletal complications. Here, we have shown that administration of PEGylated CBS into the circulation of homocystinuria model mice alters the extra- and intracellular equilibrium of sulfur amino acids, resulting in a decrease of approximately 75% in plasma total homocysteine (tHcy) and normalization of cysteine concentrations. Moreover, the decrease in homocysteine and the normalization of cysteine in PEGylated CBS-treated model mice were accompanied by improvement of histopathological liver symptoms and increased survival. Together, these data suggest that CBS enzyme replacement therapy (ERT) is a promising approach for the treatment of homocystinuria and that ERT for metabolic diseases may not necessitate introduction of the deficient enzyme into its natural intracellular compartment.


Subject(s)
Cystathionine beta-Synthase/deficiency , Cystathionine beta-Synthase/therapeutic use , Homocystinuria/drug therapy , Homocystinuria/metabolism , Animals , Cystathionine beta-Synthase/genetics , Disease Models, Animal , Homocystinuria/pathology , Humans , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyethylene Glycols , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use
17.
Biochimie ; 126: 14-20, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26791043

ABSTRACT

Two enzymes in the transsulfuration pathway of homocysteine -cystathionine beta-synthase (CBS) and gamma-cystathionase (CTH)-use cysteine and/or homocysteine to produce the important signaling molecule hydrogen sulfide (H2S) and simultaneously the thioethers lanthionine, cystathionine or homolanthionine. In this study we explored whether impaired flux of substrates for H2S synthesis and/or deficient enzyme activities alter production of hydrogen sulfide in patients with homocystinurias. As an indirect measure of H2S synthesis we determined by LC-MS/MS concentrations of thioethers in plasma samples from 33 patients with different types of homocystinurias, in 8 patient derived fibroblast cell lines, and as reaction products of seven purified mutant CBS enzymes. Since chaperoned recombinant mutant CBS enzymes retained capacity of H2S synthesis in vitro it can be stipulated that deficient CBS activity in vivo may impair H2S production. Indeed, in patients with classical homocystinuria we observed significantly decreased cystathionine and lanthionine concentrations in plasma (46% and 74% of median control levels, respectively) and significantly lower cystathionine in fibroblasts (8% of median control concentrations) indicating that H2S production from cysteine and homocysteine may be also impaired. In contrast, the grossly elevated plasma levels of homolanthionine in CBS deficient patients (32-times elevation compared to median of controls) clearly demonstrates a simultaneous overproduction of H2S from homocysteine by CTH. In the remethylation defects the accumulation of homocysteine and the increased flux of metabolites through the transsulfuration pathway resulted in elevation of cystathionine and homolanthionine (857% and 400% of median control values, respectively) indicating a possibility of an increased biosynthesis of H2S by both CBS and CTH. This study shows clearly disturbed thioether concentrations in homocystinurias, and modeling using these data indicates that H2S synthesis may be increased in these conditions. Further studies are needed to confirm our findings and to explore the possible implications for pathophysiology of these disorders.


Subject(s)
Alanine/analogs & derivatives , Cystathionine/metabolism , Fibroblasts/metabolism , Homocystinuria/metabolism , Hydrogen Sulfide/metabolism , Sulfides/metabolism , Alanine/metabolism , Cells, Cultured , Cystathionine beta-Synthase/metabolism , Female , Fibroblasts/pathology , Homocystinuria/pathology , Humans , Male
18.
Hypertension ; 67(2): 335-41, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26667416

ABSTRACT

Metabolism of homocysteine and other sulfur amino acids is closely associated with metabolism of folates. In this study, we analyzed the possible role of folates and sulfur amino acids in the development of features of the metabolic syndrome in the BXH/HXB recombinant inbred strains derived from the spontaneously hypertensive rat (SHR) and Brown Norway progenitors. We mapped a quantitative trait locus for cysteine concentrations to a region of chromosome 1 that contains a cis-acting expression quantitative trait locus regulating mRNA levels of folate receptor 1 (Folr1) in the kidney. Sequence analysis revealed a deletion variant in the Folr1 promoter region of the SHR. Transfection studies demonstrated that the SHR-promoter region of Folr1 is less effective in driving luciferase reporter gene expression than the Brown Norway promoter region of Folr1. Results in the SHR.BN-chr.1 congenic strain confirmed that the SHR variant in Folr1 cosegregates with markedly reduced renal expression of Folr1 and renal folate reabsorption, decreased serum levels of folate, increased serum levels of cysteine and homocysteine, increased adiposity, ectopic fat accumulation in liver and muscle, reduced muscle insulin sensitivity, and increased blood pressure. Transgenic rescue experiments performed by expressing a Folr1 transgene in the SHR ameliorated most of the metabolic disturbances. These findings are consistent with the hypothesis that inherited variation in the expression of Folr1 in the kidney influences the development of the metabolic syndrome and constitutes a previously unrecognized genetic mechanism that may contribute to increased risk for diabetes mellitus and cardiovascular disease.


Subject(s)
Folate Receptor 1/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Hypertension/complications , Kidney/metabolism , Metabolic Syndrome/genetics , RNA/genetics , Animals , Blood Pressure/physiology , Folate Receptor 1/biosynthesis , Genetic Variation , Hypertension/genetics , Hypertension/metabolism , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Rats , Rats, Inbred BN , Rats, Inbred SHR , Real-Time Polymerase Chain Reaction
19.
Clin Biochem ; 48(1-2): 2-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25445730

ABSTRACT

OBJECTIVES: Stable isotope dilution coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS) is the sensitive method for screening for various inherited metabolic disorders using dried blood spots (DBSs). We present a method for LC-MS/MS determination of succinyladenosine (SAdo) and succinylaminoimidazole carboxamide riboside (SAICAr), biomarkers for adenylosuccinate lyase deficiency (dADSL), in DBS. DESIGN AND METHODS: SAICAr and SAdo were separated on a Symmetry-C18 column and detected using positive electrospray ionisation in selected reaction monitoring mode. The quantification was performed using the isotopically labelled internal standards SAdo-(13)C4 and SAICAr-(13)C4, which were prepared via ADSL-catalysed reactions of fumarate-(13)C4 with adenosine monophosphate and aminoimidazole carboxamide ribotide, respectively, and subsequent alkaline phosphatase-catalysed dephosphorylation of the resulting products. RESULTS: The detection of SAICAr and SAdo in DBS was linear over the range of 0-25µmol/L. The respective intra-assay and inter-assay imprecision values were less than 10.7% and 15.2% for SAICAr and 4.7% and 5.7% for SAdo. The recoveries from DBS spiked with different concentrations of SAICAr and SAdo were between 94% and 117%. The concentrations of SAICAr and SAdo were higher in the archived DBS from dADSL patients (SAICAr, 0.03-4.7µmol/L; SAdo, 1.5-21.3µmol/L; n=5) compared to those of the control subjects (SAICAr, 0-0.026µmol/L; SAdo, 0.06-0.14µmol/L; n=31), even after DBSs from dADSL patients were stored for 2-23years. CONCLUSIONS: We developed and validated a method of succinylpurine analysis in DBS that improves selective screening for dADSL in the paediatric population and may be used for retrospective diagnosis to aid the genetic counselling of affected families.


Subject(s)
Adenosine/analogs & derivatives , Adenylosuccinate Lyase/deficiency , Aminoimidazole Carboxamide/analogs & derivatives , Dried Blood Spot Testing/methods , Purine-Pyrimidine Metabolism, Inborn Errors/blood , Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , Ribonucleosides/blood , Tandem Mass Spectrometry/methods , Adenosine/blood , Adenylosuccinate Lyase/blood , Aminoimidazole Carboxamide/blood , Autistic Disorder , Carbon Isotopes , Chromatography, Liquid , Humans , Infant, Newborn , Limit of Detection , Reference Standards
20.
J Inherit Metab Dis ; 38(2): 287-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25331909

ABSTRACT

Classical homocystinuria is caused by mutations in the cystathionine ß-synthase (CBS) gene. Previous experiments in bacterial and yeast cells showed that many mutant CBS enzymes misfold and that chemical chaperones enable proper folding of a number of mutations. In the present study, we tested the extent of misfolding of 27 CBS mutations previously tested in E. coli under the more folding-permissive conditions of mammalian CHO-K1 cells and the ability of chaperones to rescue the conformation of these mutations. Expression of mutations in mammalian cells increased the median activity 16-fold and the amount of tetramers 3.2-fold compared with expression in bacteria. Subsequently, we tested the responses of seven selected mutations to three compounds with chaperone-like activity. Aminooxyacetic acid and 4-phenylbutyric acid exhibited only a weak effect. In contrast, heme arginate substantially increased the formation of mutant CBS protein tetramers (up to sixfold) and rescued catalytic activity (up to ninefold) of five out of seven mutations (p.A114V, p.K102N, p.R125Q, p.R266K, and p.R369C). The greatest effect of heme arginate was observed for the mutation p.R125Q, which is non-responsive to in vivo treatment with vitamin B(6). Moreover, the heme responsiveness of the p.R125Q mutation was confirmed in fibroblasts derived from a patient homozygous for this genetic variant. Based on these data, we propose that a distinct group of heme-responsive CBS mutations may exist and that the heme pocket of CBS may become an important target for designing novel therapies for homocystinuria.


Subject(s)
Arginine/pharmacology , Cystathionine beta-Synthase/genetics , Fibroblasts/drug effects , Heme/pharmacology , Homocystinuria/drug therapy , Molecular Chaperones/pharmacology , Mutation , Proteostasis Deficiencies/drug therapy , Animals , CHO Cells , Catalytic Domain , Cricetulus , Cystathionine beta-Synthase/metabolism , Female , Fibroblasts/enzymology , Genetic Predisposition to Disease , Homocystinuria/diagnosis , Homocystinuria/enzymology , Homocystinuria/genetics , Homozygote , Humans , Models, Molecular , Phenotype , Protein Conformation , Protein Folding , Proteostasis Deficiencies/diagnosis , Proteostasis Deficiencies/enzymology , Proteostasis Deficiencies/genetics , Structure-Activity Relationship , Substrate Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...