Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 250
Filter
1.
FASEB J ; 38(13): e23795, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38984928

ABSTRACT

Cystathionine beta-synthase-deficient homocystinuria (HCU) is a life-threatening disorder of sulfur metabolism. HCU can be treated by using betaine to lower tissue and plasma levels of homocysteine (Hcy). Here, we show that mice with severely elevated Hcy and potentially deficient in the folate species tetrahydrofolate (THF) exhibit a very limited response to betaine indicating that THF plays a critical role in treatment efficacy. Analysis of a mouse model of HCU revealed a 10-fold increase in hepatic levels of 5-methyl -THF and a 30-fold accumulation of formiminoglutamic acid, consistent with a paucity of THF. Neither of these metabolite accumulations were reversed or ameliorated by betaine treatment. Hepatic expression of the THF-generating enzyme dihydrofolate reductase (DHFR) was significantly repressed in HCU mice and expression was not increased by betaine treatment but appears to be sensitive to cellular redox status. Expression of the DHFR reaction partner thymidylate synthase was also repressed and metabolomic analysis detected widespread alteration of hepatic histidine and glutamine metabolism. Many individuals with HCU exhibit endothelial dysfunction. DHFR plays a key role in nitric oxide (NO) generation due to its role in regenerating oxidized tetrahydrobiopterin, and we observed a significant decrease in plasma NOx (NO2 + NO3) levels in HCU mice. Additional impairment of NO generation may also come from the HCU-mediated induction of the 20-hydroxyeicosatetraenoic acid generating cytochrome CYP4A. Collectively, our data shows that HCU induces dysfunctional one-carbon metabolism with the potential to both impair betaine treatment and contribute to multiple aspects of pathogenesis in this disease.


Subject(s)
Homocystinuria , Liver , Oxidation-Reduction , Tetrahydrofolate Dehydrogenase , Tetrahydrofolates , Animals , Homocystinuria/metabolism , Homocystinuria/drug therapy , Homocystinuria/genetics , Mice , Tetrahydrofolates/metabolism , Liver/metabolism , Tetrahydrofolate Dehydrogenase/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Betaine/metabolism , Betaine/pharmacology , Homocysteine/metabolism , Mice, Inbred C57BL , Cystathionine beta-Synthase/metabolism , Cystathionine beta-Synthase/genetics , Carbon/metabolism , Male , Folic Acid/metabolism , Female
2.
Biochem J ; 481(8): 569-585, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38563463

ABSTRACT

Homocystinuria is a rare disease caused by mutations in the CBS gene that results in a deficiency of cystathionine ß-synthase (CBS). CBS is an essential pyridoxal 5'-phosphate (PLP)-dependent enzyme in the transsulfuration pathway, responsible for combining serine with homocysteine to produce cystathionine, whose activity is enhanced by the allosteric regulator S-adenosylmethionine (SAM). CBS also plays a role in generating hydrogen sulfide (H2S), a gaseous signaling molecule with diverse regulatory functions within the vascular, nervous, and immune systems. In this study, we present the clinical and biochemical characterization of two novel CBS missense mutations that do not respond to pyridoxine treatment, namely c.689T > A (L230Q) and 215A > T (K72I), identified in a Chinese patient. We observed that the disease-associated K72I genetic variant had no apparent effects on the spectroscopic and catalytic properties of the full-length enzyme. In contrast, the L230Q variant expressed in Escherichia coli did not fully retain heme and when compared with the wild-type enzyme, it exhibited more significant impairments in both the canonical cystathionine-synthesis and the alternative H2S-producing reactions. This reduced activity is consistent with both in vitro and in silico evidence, which indicates that the L230Q mutation significantly decreases the overall protein's stability, which in turn, may represent the underlying cause of its pathogenicity.


Subject(s)
Cystathionine beta-Synthase , Homocystinuria , Mutation, Missense , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/metabolism , Homocystinuria/genetics , Homocystinuria/metabolism , Homocystinuria/enzymology , Humans , Male , Female
3.
FASEB J ; 36(4): e22260, 2022 04.
Article in English | MEDLINE | ID: mdl-35315960

ABSTRACT

Deficiencies in Cystathionine-ß-synthase (CBS) lead to hyperhomocysteinemia (HHCy), which is considered a risk factor for cardiovascular, bone and neurological disease. Moreover, CBS is important for the production of cysteine, hydrogen sulfide (H2 S) and glutathione. Studying the biological role of CBS in adult mice has been severely hampered by embryological disturbances and perinatal mortality. To overcome these issues and assess the effects of whole-body CBS deficiency in adult mice, we engineered and characterized a Cre-inducible Cbs knockout model during ageing. No perinatal mortality occurred before Cbs-/- induction at 10 weeks of age. Mice were followed until 90 weeks of age and ablation of Cbs was confirmed in liver and kidney but not in brain. Severe HHCy was observed in Cbs-/- (289 ± 58 µM) but not in Cbs+/- or control mice (<10 µM). Cbs-/- showed impaired growth, facial alopecia, endothelial dysfunction in absence of increased mortality, and signs of liver or kidney damage. CBS expression in skin localized to sebaceous glands and epidermis, suggesting local effects of Cbs-/- on alopecia. Cbs-/- showed increased markers of oxidative stress and senescence but expression of other H2 S producing enzymes (CSE and 3-MST) was not affected. CBS deficiency severely impaired H2 S production capacity in liver, but not in brain or kidney. In summary, Cbs-/- mice presented a mild phenotype without mortality despite severe HHCy. The findings demonstrate that HHCy is not directly linked to development of end organ damage.


Subject(s)
Homocystinuria , Hydrogen Sulfide , Hyperhomocysteinemia , Aging , Alopecia , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Disease Models, Animal , Female , Homocystinuria/metabolism , Hydrogen Sulfide/metabolism , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , Mice , Mice, Knockout , Pregnancy
4.
Cell Mol Life Sci ; 79(8): 410, 2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35821533

ABSTRACT

Beyond well-assessed risk factors, cardiovascular events could be also associated with the presence of epigenetic and genetic alterations, such as the methylenetetrahydrofolate-reductase (MTHFR) C677T polymorphism. This gene variant is related to increased circulating levels of homocysteine (Hcy) and cardiovascular risk. However, heterozygous carriers have an augmented risk of cardiovascular accidents independently from normal Hcy levels, suggesting the presence of additional deregulated processes in MTHFR C677T carriers. Here, we hypothesize that targeting Sirtuin 1 (SIRT1) could be an alternative mechanism to control the cardiovascular risk associated to MTHFR deficiency condition. Flow Mediated Dilatation (FMD) and light transmission aggregometry assay were performed in subjects carrying MTHFR C677T allele after administration of resveratrol, the most powerful natural clinical usable compound that owns SIRT1 activating properties. MTHFR C677T carriers with normal Hcy levels revealed endothelial dysfunction and enhanced platelet aggregation associated with SIRT1 downregulation. SIRT1 activity stimulation by resveratrol intake was able to override these abnormalities without affecting Hcy levels. Impaired endothelial function, bleeding time, and wire-induced thrombus formation were rescued in a heterozygous Mthfr-deficient (Mthfr+/-) mouse model after resveratrol treatment. Using a cell-based high-throughput multiplexed screening (HTS) assay, a novel selective synthetic SIRT1 activator, namely ISIDE11, was identified. Ex vivo and in vivo treatment of Mthfr+/- mice with ISIDE11 rescues endothelial vasorelaxation and reduces wire-induced thrombus formation, effects that were abolished by SIRT1 inhibitor. Moreover, platelets from MTHFR C677T allele carriers treated with ISIDE11 showed normalization of their typical hyper-reactivity. These results candidate SIRT1 activation as a new therapeutic strategy to contain cardio and cerebrovascular events in MTHFR carriers.


Subject(s)
Homocystinuria , Methylenetetrahydrofolate Reductase (NADPH2) , Sirtuin 1 , Thrombosis , Animals , Genotype , Homocystinuria/drug therapy , Homocystinuria/metabolism , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Muscle Spasticity , Psychotic Disorders/metabolism , Resveratrol/pharmacology , Sirtuin 1/genetics , Sirtuin 1/metabolism , Thrombosis/drug therapy , Thrombosis/genetics , Thrombosis/metabolism , Thrombosis/prevention & control
5.
Dev Biol ; 468(1-2): 1-13, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32941884

ABSTRACT

Combined methylmalonic acidemia and homocystinuria, cblC type, is the most common inherited disorder of cobalamin metabolism and is characterized by severe fetal developmental defects primarily impacting the central nervous system, hematopoietic system, and heart. CblC was previously shown to be due to mutations in the MMACHC gene, which encodes a protein thought to function in intracellular cobalamin trafficking and biosynthesis of adenosylcobalamin (AdoCbl) and methylcobalamin (MeCbl). These coenzymes are required for the production of succinyl-CoA and methionine, respectively. However, it is currently unclear whether additional roles for MMACHC exist outside of cobalamin metabolism. Furthermore, due to a lack of sufficient animal models, the exact pathophysiology of cblC remains unknown. Here, we report the generation and characterization of two new mouse models to study the role of MMACHC in vivo. CRISPR/Cas9 genome editing was used to develop a Mmachc floxed allele (Mmachcflox/flox), which we validated as a conditional null. For a gain-of-function approach, we generated a transgenic mouse line that over-expresses functional Mmachc (Mmachc-OE+/tg) capable of rescuing Mmachc homozygous mutant lethality. Surprisingly, our data also suggest that these mice may exhibit a partially penetrant maternal-effect rescue, which might have implications for in utero therapeutic interventions to treat cblC. Both the Mmachcflox/flox and Mmachc-OE+/tg mouse models will be valuable resources for understanding the biological roles of MMACHC in a variety of tissue contexts and allow for deeper understanding of the pathophysiology of cblC.


Subject(s)
Homocystinuria , Oxidoreductases , Vitamin B 12 Deficiency/congenital , Animals , Disease Models, Animal , Homocystinuria/genetics , Homocystinuria/metabolism , Homocystinuria/pathology , Homocystinuria/physiopathology , Mice , Mice, Transgenic , Oxidoreductases/genetics , Oxidoreductases/metabolism , Vitamin B 12 Deficiency/genetics , Vitamin B 12 Deficiency/metabolism , Vitamin B 12 Deficiency/pathology , Vitamin B 12 Deficiency/physiopathology
6.
Mol Genet Metab ; 132(2): 128-138, 2021 02.
Article in English | MEDLINE | ID: mdl-33483253

ABSTRACT

Cystathionine beta-synthase deficient homocystinuria (HCU) is a life-threatening disorder of sulfur metabolism. Our knowledge of the metabolic changes induced in HCU are based almost exclusively on data derived from plasma. In the present study, we present a comprehensive analysis on the effects of HCU upon the hepatic metabolites and enzyme expression levels of the methionine-folate cycles in a mouse model of HCU. HCU induced a 10-fold increase in hepatic total homocysteine and in contrast to plasma, this metabolite was only lowered by approximately 20% by betaine treatment indicating that this toxic metabolite remains unacceptably elevated. Hepatic methionine, S-adenosylmethionine, S-adenosylhomocysteine, N-acetlymethionine, N-formylmethionine, methionine sulfoxide, S-methylcysteine, serine, N-acetylserine, taurocyamine and N-acetyltaurine levels were also significantly increased by HCU while cysteine, N-acetylcysteine and hypotaurine were all significantly decreased. In terms of polyamine metabolism, HCU significantly decreased spermine and spermidine levels while increasing 5'-methylthioadenosine. Betaine treatment restored normal spermine and spermidine levels but further increased 5'-methylthioadenosine. HCU induced a 2-fold induction in expression of both S-adenosylhomocysteine hydrolase and methylenetetrahydrofolate reductase. Induction of this latter enzyme was accompanied by a 10-fold accumulation of its product, 5-methyl-tetrahydrofolate, with the potential to significantly perturb one­carbon metabolism. Expression of the cytoplasmic isoform of serine hydroxymethyltransferase was unaffected by HCU but the mitochondrial isoform was repressed indicating differential regulation of one­carbon metabolism in different sub-cellular compartments. All HCU-induced changes in enzyme expression were completely reversed by either betaine or taurine treatment. Collectively, our data show significant alterations of polyamine, folate and methionine cycle metabolism in HCU hepatic tissues that in some cases, differ significantly from those observed in plasma, and have the potential to contribute to multiple aspects of pathogenesis.


Subject(s)
Cystathionine beta-Synthase/genetics , Homocystinuria/metabolism , Liver/metabolism , Methionine/metabolism , Adenosylhomocysteinase/genetics , Animals , Betaine/pharmacology , Cystathionine beta-Synthase/metabolism , Disease Models, Animal , Folic Acid/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Glycine Hydroxymethyltransferase/genetics , Homocysteine/blood , Homocysteine/metabolism , Homocystinuria/drug therapy , Homocystinuria/genetics , Homocystinuria/pathology , Humans , Liver/enzymology , Methionine/analogs & derivatives , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Polyamines/metabolism
7.
J Inherit Metab Dis ; 44(6): 1382-1392, 2021 11.
Article in English | MEDLINE | ID: mdl-34528713

ABSTRACT

Cystathionine ß-synthase (CBS) deficiency is a recessive inborn error of sulfur metabolism characterized by elevated blood levels of total homocysteine (tHcy). Patients diagnosed with CBS deficiency are currently treated by a combination of vitamin supplementation and restriction of foods containing the homocysteine precursor methionine, but the effectiveness of this therapy is limited due to poor compliance. A mouse model for CBS deficiency (Tg-I278T Cbs-/- ) was used to evaluate a potential gene therapy approach to treat CBS deficiency utilizing an AAVrh.10-based vector containing the human CBS cDNA downstream of the constitutive, strong CAG promoter (AAVrh.10hCBS). Mice were administered a single dose of virus and followed for up to 1 year. The data demonstrated a dose-dependent increase in liver CBS activity and a dose-dependent decrease in serum tHcy. Liver CBS enzyme activity at 1 year was similar to Cbs+/- control mice. Mice given the highest dose (5.6 × 1011 genomes/mouse) had mean serum tHcy decrease of 97% 1 week after injection and an 81% reduction 1 year after injection. Treated mice had either full- or substantial correction of alopecia, bone loss, and fat mass phenotypes associated with Cbs deficiency in mice. Our findings show that AAVrh.10-based gene therapy is highly effective in treating CBS deficiency in mice and supports additional pre-clinical testing for eventual use human trials.


Subject(s)
Cystathionine beta-Synthase/genetics , Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Homocystinuria/genetics , Homocystinuria/therapy , Animals , Cystathionine beta-Synthase/blood , Cystathionine beta-Synthase/deficiency , Disease Models, Animal , Female , Gene Expression , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Homocystinuria/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Phenotype
8.
J Nutr ; 150(Suppl 1): 2506S-2517S, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000152

ABSTRACT

The metabolism of sulfur-containing amino acids (SAAs) requires an orchestrated interplay among several dozen enzymes and transporters, and an adequate dietary intake of methionine (Met), cysteine (Cys), and B vitamins. Known human genetic disorders are due to defects in Met demethylation, homocysteine (Hcy) remethylation, or cobalamin and folate metabolism, in Hcy transsulfuration, and Cys and hydrogen sulfide (H2S) catabolism. These disorders may manifest between the newborn period and late adulthood by a combination of neuropsychiatric abnormalities, thromboembolism, megaloblastic anemia, hepatopathy, myopathy, and bone and connective tissue abnormalities. Biochemical features include metabolite deficiencies (e.g. Met, S-adenosylmethionine (AdoMet), intermediates in 1-carbon metabolism, Cys, or glutathione) and/or their accumulation (e.g. S-adenosylhomocysteine, Hcy, H2S, or sulfite). Treatment should be started as early as possible and may include a low-protein/low-Met diet with Cys-enriched amino acid supplements, pharmacological doses of B vitamins, betaine to stimulate Hcy remethylation, the provision of N-acetylcysteine or AdoMet, or experimental approaches such as liver transplantation or enzyme replacement therapy. In several disorders, patients are exposed to long-term markedly elevated Met concentrations. Although these conditions may inform on Met toxicity, interpretation is difficult due to the presence of additional metabolic changes. Two disorders seem to exhibit Met-associated toxicity in the brain. An increased risk of demyelination in patients with Met adenosyltransferase I/III (MATI/III) deficiency due to biallelic mutations in the MATIA gene has been attributed to very high blood Met concentrations (typically >800 µmol/L) and possibly also to decreased liver AdoMet synthesis. An excessively high Met concentration in some patients with cystathionine ß-synthase deficiency has been associated with encephalopathy and brain edema, and direct toxicity of Met has been postulated. In summary, studies in patients with various disorders of SAA metabolism showed complex metabolic changes with distant cellular consequences, most of which are not attributable to direct Met toxicity.


Subject(s)
Amino Acids, Sulfur/metabolism , Cysteine/metabolism , Homocysteine/metabolism , Metabolic Diseases/genetics , Methionine/metabolism , Sulfur Compounds/metabolism , Sulfur/metabolism , Animals , Brain Diseases/etiology , Brain Diseases/metabolism , Glutathione/metabolism , Homocystinuria/etiology , Homocystinuria/metabolism , Humans , Hydrogen Sulfide/metabolism , Liver/metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Metabolic Diseases/therapy , Metabolism, Inborn Errors/pathology , Metabolism, Inborn Errors/therapy , Methionine Adenosyltransferase/metabolism , Methylation , S-Adenosylmethionine/metabolism , Sulfites/metabolism
9.
FASEB J ; 33(11): 12477-12486, 2019 11.
Article in English | MEDLINE | ID: mdl-31450979

ABSTRACT

Classic homocystinuria (HCU) is an inherited disorder characterized by elevated homocysteine (Hcy) in plasma and tissues resulting from cystathionine ß-synthase (CBS) deficiency. There is no cure, and patients are predominantly managed by methionine-restricted diet (MRD) to limit the production of Hcy. In this study, we used the I278T mouse model of HCU to evaluate the long-term impact of a novel enzyme replacement therapy [truncated human CBS C15S mutant modified with linear 20-kDa N-hydroxysuccinimide ester polyethylene glycol (OT-58)] on clinical end points relevant to human patients with HCU. In addition, we compared its efficacy on a background of either MRD or normal methionine intake [regular diet (REG)] to that of MRD alone. We found that, compared with untreated I278T mice, OT-58 treatment of I278T mice fed with the REG diet resulted in a 90% decrease in plasma Hcy concentrations and correction of learning/cognition, endothelial dysfunction, hemostasis, bone mineralization, and body composition. On background of the MRD, OT-58 performed equally well with plasma Hcy entirely normalized. The MRD alone decreased plasma Hcy by 67% and corrected the HCU phenotype in I278T mice. However, the MRD increased anxiety and reduced bone mineral content in both I278T mice and wild-type controls. This study shows that OT-58 is a highly efficacious novel treatment for HCU on the background of either normal or restricted methionine intake.-Majtan, T., Park, I., Cox, A., Branchford, B. R., di Paola, J., Bublil, E. M., Kraus, J. P. Behavior, body composition, and vascular phenotype of homocystinuric mice on methionine-restricted diet or enzyme replacement therapy.


Subject(s)
Behavior, Animal , Body Composition , Cystathionine beta-Synthase/therapeutic use , Enzyme Replacement Therapy , Homocystinuria/drug therapy , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Disease Models, Animal , Homocystinuria/genetics , Homocystinuria/metabolism , Homocystinuria/pathology , Humans , Methionine/pharmacology , Mice , Mice, Transgenic
10.
FASEB J ; 33(5): 6339-6353, 2019 05.
Article in English | MEDLINE | ID: mdl-30768359

ABSTRACT

Classical cystathionine ß-synthase-deficient homocystinuria (HCU) is a life-threatening inborn error of sulfur metabolism. Treatment for pyridoxine-nonresponsive HCU involves lowering homocysteine (Hcy) with a methionine (Met)-restricted diet and betaine supplementation. Betaine treatment efficacy diminishes significantly over time due to impairment of betaine-Hcy S-methyltransferase (BHMT) function. Little is known regarding the regulation of BHMT in HCU. Using a betaine-responsive preclinical mouse model of HCU, we observed that this condition induces a 75% repression of BHMT mRNA, protein and enzyme activity, and significant depletion of hepatic betaine levels. BHMT repression was proportional to plasma Hcy levels but was not observed in mouse models of homocystinuria due to either methylenetetrahydrofolate reductase or Met synthase deficiency. Both Met supplementation and chemically induced glutathione depletion exacerbated hepatic BHMT repression in HCU mice but not wild-type (WT) controls. Conversely, cysteine treatment normalized hepatic BHMT expression in HCU mice but had no effect in WT control animals. Taurine treatment induced BHMT expression in HCU mice by 5-fold and restored maximal lowering of Hcy levels during long-term betaine treatment with a concomitant normalization of inflammatory cytokine expression and a significantly improved coagulative phenotype. Collectively, our findings indicate that adjuvantial taurine treatment has the potential to significantly improve clinical outcomes in HCU.-Maclean, K. N., Jiang, H, Phinney, W. N., Keating, A. K., Hurt, K. J., Stabler, S. P. Taurine alleviates repression of betaine-homocysteine S-methyltransferase and significantly improves the efficacy of long-term betaine treatment in a mouse model of cystathionine ß-synthase-deficient homocystinuria.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/metabolism , Betaine/pharmacology , Homocystinuria , Liver/enzymology , Taurine/pharmacology , Animals , Betaine-Homocysteine S-Methyltransferase/genetics , Disease Models, Animal , Homocystinuria/drug therapy , Homocystinuria/genetics , Homocystinuria/metabolism , Homocystinuria/pathology , Humans , Liver/pathology , Mice , Mice, Knockout
11.
Hum Mutat ; 40(2): 230-240, 2019 02.
Article in English | MEDLINE | ID: mdl-30408270

ABSTRACT

Homocystinuria is a rare inborn error of methionine metabolism caused by cystathionine ß-synthase (CBS) deficiency. The prevalence of homocystinuria in Qatar is 1:1,800 births, mainly due to a founder Qatari missense mutation, c.1006C>T; p.R336C (p.Arg336Cys). We characterized the structure-function relationship of the p.R336C-mutant protein and investigated the effect of different chemical chaperones to restore p.R336C-CBS activity using three models: in silico, ΔCBS yeast, and CRISPR/Cas9 p.R336C knock-in HEK293T and HepG2 cell lines. Protein modeling suggested that the p.R336C induces severe conformational and structural changes, perhaps influencing CBS activity. Wild-type CBS, but not the p.R336C mutant, was able to restore the yeast growth in ΔCBS-deficient yeast in a complementation assay. The p.R336C knock-in HEK293T and HepG2 cells decreased the level of CBS expression and reduced its structural stability; however, treatment of the p.R336C knock-in HEK293T cells with betaine, a chemical chaperone, restored the stability and tetrameric conformation of CBS, but not its activity. Collectively, these results indicate that the p.R336C mutation has a deleterious effect on CBS structure, stability, and activity, and using the chemical chaperones approach for treatment could be ineffective in restoring p.R336C CBS activity.


Subject(s)
Cystathionine beta-Synthase/genetics , Homocystinuria/genetics , Molecular Chaperones/genetics , Mutant Proteins/genetics , Computer Simulation , Cystathionine beta-Synthase/chemistry , Enzyme Stability , Gene Expression Regulation, Enzymologic/genetics , HEK293 Cells , Hep G2 Cells , Homocystinuria/metabolism , Homocystinuria/pathology , Humans , Methionine/metabolism , Molecular Chaperones/chemistry , Mutant Proteins/chemistry , Mutation, Missense/genetics , Protein Folding , Protein Structure, Tertiary , Qatar , Structure-Activity Relationship
12.
FASEB J ; 32(3): 1265-1280, 2018 03.
Article in English | MEDLINE | ID: mdl-29101223

ABSTRACT

Cystathionine ß-synthase-deficient homocystinuria (HCU) is a poorly understood, life-threatening inborn error of sulfur metabolism. Analysis of hepatic glutathione (GSH) metabolism in a mouse model of HCU demonstrated significant depletion of cysteine, GSH, and GSH disulfide independent of the block in trans-sulfuration compared with wild-type controls. HCU induced the expression of the catalytic and regulatory subunits of γ-glutamyl ligase, GSH synthase (GS), γ-glutamyl transpeptidase 1, 5-oxoprolinase (OPLAH), and the GSH-dependent methylglyoxal detoxification enzyme, glyoxalase-1. Multiple components of the transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated antioxidant-response regulatory axis were induced without any detectable activation of Nrf2. Metabolomic analysis revealed the accumulation of multiple γ-glutamyl amino acids and that plasma ophthalmate levels could serve as a noninvasive marker for hepatic redox stress. Neither cysteine, nor betaine treatment was able to reverse the observed enzyme inductions. Taurine treatment normalized the expression levels of γ-glutamyl ligase C/M, GS, OPLAH, and glyoxalase-1, and reversed HCU-induced deficits in protein glutathionylation by acting to double GSH levels relative to controls. Collectively, our data indicate that the perturbation of the γ-glutamyl cycle could contribute to multiple sequelae in HCU and that taurine has significant therapeutic potential for both HCU and other diseases for which GSH depletion is a critical pathogenic factor.-Maclean, K. N., Jiang, H., Aivazidis, S., Kim, E., Shearn, C. T., Harris, P. S., Petersen, D. R., Allen, R. H., Stabler, S. P., Roede, J. R. Taurine treatment prevents derangement of the hepatic γ-glutamyl cycle and methylglyoxal metabolism in a mouse model of classical homocystinuria: regulatory crosstalk between thiol and sulfinic acid metabolism.


Subject(s)
Aminobutyrates/metabolism , Homocystinuria/metabolism , Liver/metabolism , Pyruvaldehyde/metabolism , Sulfhydryl Compounds/metabolism , Sulfinic Acids/metabolism , Taurine/pharmacology , Amino Acids/metabolism , Animals , Cystathionine beta-Synthase/metabolism , Disease Models, Animal , Female , Homocystinuria/drug therapy , Homocystinuria/pathology , Liver/drug effects , Male , Metabolome , Mice , Mice, Inbred C57BL , Oxidation-Reduction , gamma-Glutamyltransferase/metabolism
13.
J Inherit Metab Dis ; 42(2): 333-352, 2019 03.
Article in English | MEDLINE | ID: mdl-30773687

ABSTRACT

AIM: To explore the clinical presentation, course, treatment and impact of early treatment in patients with remethylation disorders from the European Network and Registry for Homocystinurias and Methylation Defects (E-HOD) international web-based registry. RESULTS: This review comprises 238 patients (cobalamin C defect n = 161; methylenetetrahydrofolate reductase deficiency n = 50; cobalamin G defect n = 11; cobalamin E defect n = 10; cobalamin D defect n = 5; and cobalamin J defect n = 1) from 47 centres for whom the E-HOD registry includes, as a minimum, data on medical history and enrolment visit. The duration of observation was 127 patient years. In 181 clinically diagnosed patients, the median age at presentation was 30 days (range 1 day to 42 years) and the median age at diagnosis was 3.7 months (range 3 days to 56 years). Seventy-five percent of pre-clinically diagnosed patients with cobalamin C disease became symptomatic within the first 15 days of life. Total homocysteine (tHcy), amino acids and urinary methylmalonic acid (MMA) were the most frequently assessed disease markers; confirmatory diagnostics were mainly molecular genetic studies. Remethylation disorders are multisystem diseases dominated by neurological and eye disease and failure to thrive. In this cohort, mortality, thromboembolic, psychiatric and renal disease were rarer than reported elsewhere. Early treatment correlates with lower overall morbidity but is less effective in preventing eye disease and cognitive impairment. The wide variation in treatment hampers the evaluation of particular therapeutic modalities. CONCLUSION: Treatment improves the clinical course of remethylation disorders and reduces morbidity, especially if started early, but neurocognitive and eye symptoms are less responsive. Current treatment is highly variable. This study has the inevitable limitations of a retrospective, registry-based design.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/therapy , Homocystinuria/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity/metabolism , Vitamin B 12/metabolism , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Cross-Sectional Studies , Disease Progression , Europe , Female , Humans , Infant , Infant, Newborn , Male , Methylation , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Methylmalonic Acid/urine , Phenotype , Pregnancy , Psychotic Disorders/metabolism , Registries , Retrospective Studies , Young Adult
14.
J Inherit Metab Dis ; 42(1): 128-139, 2019 01.
Article in English | MEDLINE | ID: mdl-30740731

ABSTRACT

PURPOSE: To assess how the current practice of newborn screening (NBS) for homocystinurias compares with published recommendations. METHODS: Twenty-two of 32 NBS programmes from 18 countries screened for at least one form of homocystinuria. Centres provided pseudonymised NBS data from patients with cystathionine beta-synthase deficiency (CBSD, n = 19), methionine adenosyltransferase I/III deficiency (MATI/IIID, n = 28), combined remethylation disorder (cRMD, n = 56) and isolated remethylation disorder (iRMD), including methylenetetrahydrofolate reductase deficiency (MTHFRD) (n = 8). Markers and decision limits were converted to multiples of the median (MoM) to allow comparison between centres. RESULTS: NBS programmes, algorithms and decision limits varied considerably. Only nine centres used the recommended second-tier marker total homocysteine (tHcy). The median decision limits of all centres were ≥ 2.35 for high and ≤ 0.44 MoM for low methionine, ≥ 1.95 for high and ≤ 0.47 MoM for low methionine/phenylalanine, ≥ 2.54 for high propionylcarnitine and ≥ 2.78 MoM for propionylcarnitine/acetylcarnitine. These decision limits alone had a 100%, 100%, 86% and 84% sensitivity for the detection of CBSD, MATI/IIID, iRMD and cRMD, respectively, but failed to detect six individuals with cRMD. To enhance sensitivity and decrease second-tier testing costs, we further adapted these decision limits using the data of 15 000 healthy newborns. CONCLUSIONS: Due to the favorable outcome of early treated patients, NBS for homocystinurias is recommended. To improve NBS, decision limits should be revised considering the population median. Relevant markers should be combined; use of the postanalytical tools offered by the CLIR project (Collaborative Laboratory Integrated Reports, which considers, for example, birth weight and gestational age) is recommended. tHcy and methylmalonic acid should be implemented as second-tier markers.


Subject(s)
Homocystinuria/diagnosis , Acetylcarnitine/metabolism , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/metabolism , Carnitine/analogs & derivatives , Carnitine/metabolism , Female , Glycine N-Methyltransferase/deficiency , Glycine N-Methyltransferase/metabolism , Homocysteine/metabolism , Homocystinuria/metabolism , Humans , Infant, Newborn , Male , Methionine/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Methylmalonic Acid/metabolism , Muscle Spasticity/diagnosis , Muscle Spasticity/metabolism , Neonatal Screening/methods , Phenylalanine/metabolism , Psychotic Disorders/diagnosis , Psychotic Disorders/metabolism
15.
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
16.
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
17.
Curr Osteoporos Rep ; 16(5): 554-560, 2018 10.
Article in English | MEDLINE | ID: mdl-30116976

ABSTRACT

PURPOSE OF REVIEW: Homocystinuria is a congenital metabolic disorder in which cystathionine ß-synthase deficiency results in a prominent increase in homocysteine (serum levels > 100 µM), causing mental retardation, atherosclerotic cerebral infarction, and osteoporosis accompanied by fragility fractures. Encountering a case with excessive homocysteinemia such as that seen in hereditary homocystinuria is unlikely during usual medical examinations. However, in individuals who have vitamin B or folate deficiency, serum homocysteine concentrations are known to increase. These individuals may also have a polymorphism in methylenetetrahydrofolate reductase, MTHFR (C677T: TT type), which regulates homocysteine metabolism. These changes in homocysteine levels may elicit symptoms resembling those of homocystinuria (e.g., Alzheimer's disease, atherosclerosis, osteoporosis). RECENT FINDINGS: High serum homocysteine has been shown to have detrimental effects on neural cells, vascular endothelial cells, osteoblasts, and osteoclasts. Homocysteine is also known to increase oxidative stress, disrupt cross-linking of collagen molecules, and increase levels of advanced glycation end products, which results in reduced bone strength through a mechanism that goes beyond low bone density and increased bone resorption. Therefore, high serum homocysteine may be regarded as a factor that can reduce both bone mass and impair bone quality. In this review, we outline the epidemiology and pathophysiology of osteoporosis associated with hyperhomocysteinemia.


Subject(s)
Bone and Bones/metabolism , Homocysteine/metabolism , Hyperhomocysteinemia/metabolism , Osteoporosis/metabolism , Collagen/metabolism , Folic Acid Deficiency/metabolism , Glycation End Products, Advanced/metabolism , Homocystinuria/complications , Homocystinuria/genetics , Homocystinuria/metabolism , Humans , Hyperhomocysteinemia/complications , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Muscle Spasticity/complications , Muscle Spasticity/genetics , Muscle Spasticity/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/etiology , Polymorphism, Genetic , Psychotic Disorders/complications , Psychotic Disorders/genetics , Psychotic Disorders/metabolism , Vitamin B Deficiency/metabolism
18.
Cell Mol Neurobiol ; 37(8): 1477-1485, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28258516

ABSTRACT

Homocystinuria is an inborn error of amino acid metabolism caused by deficiency of cystathionine ß-synthase (CBS) activity, biochemically characterized by homocysteine (Hcy) and methionine (Met) accumulation in biological fluids and high urinary excretion of homocystine. Clinical manifestations include thinning and lengthening of long bones, osteoporosis, dislocation of the ocular lens, thromboembolism, and mental retardation. Although the pathophysiology of this disease is poorly known, the present review summarizes the available experimental findings obtained from patients and animal models indicating that oxidative stress may contribute to the pathogenesis of homocystinuria. In this scenario, several studies have shown that enzymatic and non-enzymatic antioxidant defenses are decreased in individuals affected by this disease. Furthermore, markers of lipid, protein, and DNA oxidative damage have been reported to be increased in blood, brain, liver, and skeletal muscle in animal models studied and in homocystinuric patients, probably as a result of increased free radical generation. On the other hand, in vitro and in vivo studies have shown that Hcy induces reactive species formation in brain, so that this major accumulating metabolite may underlie the oxidative damage observed in the animal model and human condition. Taken together, it may be presumed that the disruption of redox homeostasis may contribute to the tissue damage found in homocystinuria. Therefore, it is proposed that the use of appropriate antioxidants may represent a novel adjuvant therapy for patients affected by this disease.


Subject(s)
Cystathionine beta-Synthase/deficiency , Disease Models, Animal , Homocystinuria/metabolism , Oxidative Stress/physiology , Animals , Antioxidants/metabolism , Brain/metabolism , Homocystinuria/pathology , Humans
19.
J Inherit Metab Dis ; 40(1): 113-120, 2017 01.
Article in English | MEDLINE | ID: mdl-27444757

ABSTRACT

Cystathionine ß-synthase (CBS) deficiency is a recessive inborn error of metabolism in which patients have extremely elevated plasma total homocysteine and have clinical manifestations in the vascular, visual, skeletal, and nervous systems. Homocysteine is an intermediary metabolite produced from the hydrolysis of S-adenosylhomocysteine (SAH), which is a by-product of methylation reactions involving the methyl-donor S-adenosylmethionine (SAM). Here, we have measured SAM, SAH, DNA and histone methylation status in an inducible mouse model of CBS deficiency to test the hypothesis that homocysteine-related phenotypes are caused by inhibition of methylation due to elevated SAH and reduced SAM/SAH ratio. We found that mice lacking CBS have elevated cellular SAH and reduced SAM/SAH ratios in both liver and kidney, but this was not associated with alterations in the level of 5-methylcytosine or various histone modifications. Using methylated DNA immunoprecipitation in combination with microarray, we found that of the 241 most differentially methylated promoter probes, 89 % were actually hypermethylated in CBS deficient mice. In addition, we did not find that changes in DNA methylation correlated well with changes in RNA expression in the livers of induced and uninduced CBS mice. Our data indicates that reduction in the SAM/SAH ratio, due to loss of CBS activity, does not result in overall hypomethylation of either DNA or histones.


Subject(s)
Cystathionine beta-Synthase/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Homocystinuria/genetics , Animals , Cystathionine beta-Synthase/metabolism , DNA/genetics , Disease Models, Animal , Epigenomics/methods , Homocysteine/genetics , Homocysteine/metabolism , Homocystinuria/metabolism , Kidney/metabolism , Liver/metabolism , Mice , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism
20.
Natl Med J India ; 30(3): 139-141, 2017.
Article in English | MEDLINE | ID: mdl-28936998

ABSTRACT

An 18-year-old male presented to our hospital with complaints of episodic abdominal pain, dry cough and right pleuritic chest pain. He was diagnosed as a case of right tuberculous pleural effusion on the basis of the pleural fluid Genexpert report of Mycobacterium tuberculosis detected sensitive to rifampicin and was started on antituberculous therapy. Forty-five days later, he presented with acute onset breathlessness, swelling of the right leg, streaky haemoptysis and a fresh left-sided pleural effusion. Evaluation revealed venous thromboembolism (right lower lobar segment pulmonary embolism with right leg deep vein thrombosis). Workup for malignancy was negative. However, he had vitamin B12 deficiency with increased homocysteine levels and heterozygous mutation of the MTHFR gene at A1298C. He was treated with optimal anticoagulation, vitamin B12 supplementation and antitubercular treatment. This is a rare combination of events perhaps related to the MTHFR gene mutation.


Subject(s)
Antitubercular Agents/therapeutic use , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Pulmonary Embolism/complications , Rifampin/therapeutic use , Tuberculosis/drug therapy , Venous Thromboembolism/complications , Homocystinuria/metabolism , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/chemistry , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Muscle Spasticity/metabolism , Pleural Effusion , Psychotic Disorders/metabolism , Rifampin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL