Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 251
Filter
1.
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
2.
Cell Host Microbe ; 32(3): 382-395.e10, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38309259

ABSTRACT

Methionine is an essential proteinogenic amino acid, but its excess can lead to deleterious effects. Inborn errors of methionine metabolism resulting from loss of function in cystathionine ß-synthase (CBS) cause classic homocystinuria (HCU), which is managed by a methionine-restricted diet. Synthetic biotics are gastrointestinal tract-targeted live biotherapeutics that can be engineered to replicate the benefits of dietary restriction. In this study, we assess whether SYNB1353, an E. coli Nissle 1917 derivative, impacts circulating methionine and homocysteine levels in animals and healthy volunteers. In both mice and nonhuman primates (NHPs), SYNB1353 blunts the appearance of plasma methionine and plasma homocysteine in response to an oral methionine load. A phase 1 clinical study conducted in healthy volunteers subjected to an oral methionine challenge demonstrates that SYNB1353 is well tolerated and blunts plasma methionine by 26%. Overall, SYNB1353 represents a promising approach for methionine reduction with potential utility for the treatment of HCU.


Subject(s)
Homocystinuria , Methionine , Humans , Mice , Animals , Methionine/metabolism , Methionine/therapeutic use , Healthy Volunteers , Escherichia coli/genetics , Escherichia coli/metabolism , Disease Models, Animal , Homocystinuria/drug therapy , Homocystinuria/metabolism , Racemethionine , Homocysteine/therapeutic use
3.
Mol Cell Biol ; 43(12): 664-674, 2023.
Article in English | MEDLINE | ID: mdl-38051092

ABSTRACT

Homocystinuria (HCU), an inherited metabolic disorder caused by lack of cystathionine beta-synthase (CBS) activity, is chiefly caused by misfolding of single amino acid residue missense pathogenic variants. Previous studies showed that chemical, pharmacological chaperones or proteasome inhibitors could rescue function of multiple pathogenic CBS variants; however, the underlying mechanisms remain poorly understood. Using Chinese hamster DON fibroblasts devoid of CBS and stably overexpressing human WT or mutant CBS, we showed that expression of pathogenic CBS variant mostly dysregulates gene expression of small heat shock proteins HSPB3 and HSPB8 and members of HSP40 family. Endoplasmic reticulum stress sensor BiP was found upregulated with CBS I278T variant associated with proteasomes suggesting proteotoxic stress and degradation of misfolded CBS. Co-expression of the main effector HSP70 or master regulator HSF1 rescued steady-state levels of CBS I278T and R125Q variants with partial functional rescue of the latter. Pharmacological proteostasis modulators partially rescued expression and activity of CBS R125Q likely due to reduced proteotoxic stress as indicated by decreased BiP levels and promotion of refolding as indicated by induction of HSP70. In conclusion, targeted manipulation of cellular proteostasis may represent a viable therapeutic approach for the permissive pathogenic CBS variants causing HCU.


Subject(s)
Cystathionine beta-Synthase , Homocystinuria , Humans , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/chemistry , Cystathionine beta-Synthase/metabolism , Homocystinuria/drug therapy , Homocystinuria/genetics , Homocystinuria/metabolism , Cystathionine/metabolism , Cystathionine/therapeutic use , Proteostasis , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism
4.
PLoS One ; 18(6): e0286550, 2023.
Article in English | MEDLINE | ID: mdl-37319242

ABSTRACT

Classic homocystinuria is an inborn error of metabolism caused mainly by missense mutations leading to misfolded and/or unstable human cystathionine ß-synthase (CBS) protein, causing the accumulation of excess total homocysteine (tHcy) in tissues. Previously, it has been shown that certain missense containing human CBS proteins can be functionally rescued in mouse models of CBS deficiency by treatment with proteasome inhibitors. The rescue by proteasome inhibitors is thought to work both by inhibiting the degradation of misfolded CBS protein and by inducing the levels of heat-shock chaperone proteins in the liver. Here we examine the effectiveness of two FDA approved protease inhibitors, carfilzomib and bortezomib, on various transgenic mouse models of human CBS deficiency. Our results show that although both drugs are effective in inducing the liver chaperone proteins Hsp70 and Hsp27, and are effective in inhibiting proteasome function, bortezomib was somewhat more robust in restoring the mutant CBS function. Moreover, there was no significant correlation between proteasome inhibition and CBS activity, suggesting that some of bortezomib's effects are via other mechanisms. We also test the use of low-doses of bortezomib and carfilzomib on various mouse models for extended periods of time and find that while low-doses are less toxic, they are also less effective at restoring CBS function. Overall, these results show that while restoration of mutant CBS function is possible with proteasome inhibitors, the exact mechanism is complicated and it will likely be too toxic for long-term patient treatment.


Subject(s)
Cystathionine beta-Synthase , Homocystinuria , Humans , Mice , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Homocystinuria/drug therapy , Homocystinuria/genetics , Homocystinuria/metabolism , Bortezomib/pharmacology , Bortezomib/therapeutic use , Proteasome Endopeptidase Complex , Mice, Transgenic
5.
Br J Pharmacol ; 180(3): 264-278, 2023 02.
Article in English | MEDLINE | ID: mdl-36417581

ABSTRACT

Cystathionine beta-synthase (CBS)-deficient homocystinuria (HCU) is the most common inborn error of sulfur amino acid metabolism. The pyridoxine non-responsive form of the disease manifests itself by massively increasing plasma and tissue concentrations of homocysteine, a toxic intermediate of methionine metabolism that is thought to be the major cause of clinical complications including skeletal deformities, connective tissue defects, thromboembolism and cognitive impairment. The current standard of care involves significant dietary interventions that, despite being effective, often adversely affect quality of life of HCU patients, leading to poor adherence to therapy and inadequate biochemical control with clinical complications. In recent years, the unmet need for better therapeutic options has resulted in development of novel enzyme and gene therapies and exploration of pharmacological approaches to rescue CBS folding defects caused by missense pathogenic mutations. Here, we review scientific evidence and current state of affairs in development of recent approaches to treat HCU.


Subject(s)
Homocystinuria , Thromboembolism , Humans , Homocystinuria/drug therapy , Homocystinuria/genetics , Homocystinuria/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Quality of Life , Mutation, Missense
6.
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
7.
Cells ; 11(11)2022 05 25.
Article in English | MEDLINE | ID: mdl-35681432

ABSTRACT

Cystathionine beta synthase (CBS) catalyzes the first step of the transsulfuration pathway from homocysteine to cystathionine, and its deficiency leads to hyperhomocysteinemia (HHcy) in humans and rodents. To date, scarce information is available about the HHcy effect on insulin secretion, and the link between CBS activity and the setting of type 2 diabetes is still unknown. We aimed to decipher the consequences of an inborn defect in CBS on glucose homeostasis in mice. We used a mouse model heterozygous for CBS (CBS+/-) that presented a mild HHcy. Other groups were supplemented with methionine in drinking water to increase the mild to intermediate HHcy, and were submitted to a high-fat diet (HFD). We measured the food intake, body weight gain, body composition, glucose homeostasis, plasma homocysteine level, and CBS activity. We evidenced a defect in the stimulated insulin secretion in CBS+/- mice with mild and intermediate HHcy, while mice with intermediate HHcy under HFD presented an improvement in insulin sensitivity that compensated for the decreased insulin secretion and permitted them to maintain a glucose tolerance similar to the CBS+/+ mice. Islets isolated from CBS+/- mice maintained their ability to respond to the elevated glucose levels, and we showed that a lower parasympathetic tone could, at least in part, be responsible for the insulin secretion defect. Our results emphasize the important role of Hcy metabolic enzymes in insulin secretion and overall glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 2 , Homocystinuria , Hyperhomocysteinemia , Animals , Cystathionine beta-Synthase/metabolism , Glucose , Homeostasis , Homocysteine , Homocystinuria/metabolism , Hyperhomocysteinemia/metabolism , Mice
8.
Clin Epigenetics ; 14(1): 52, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440018

ABSTRACT

BACKGROUND: epi-cblC is a recently discovered inherited disorder of intracellular vitamin B12 metabolism associating hematological, neurological, and cardiometabolic outcomes. It is produced by an epimutation at the promoter common to CCDC163P and MMACHC, which results from an aberrant antisense transcription due to splicing mutations in the antisense PRDX1 gene neighboring MMACHC. We studied whether the aberrant transcription produced a second epimutation by encompassing the CpG island of the TESK2 gene neighboring CCDC163P. METHODS: We unraveled the methylome architecture of the CCDC163P-MMACHC CpG island (CpG:33) and the TESK2 CpG island (CpG:51) of 17 epi-cblC cases. We performed an integrative analysis of the DNA methylome profiling, transcriptome reconstruction of RNA-sequencing (RNA-seq), chromatin immunoprecipitation sequencing (ChIP-Seq) of histone H3, and transcription expression of MMACHC and TESK2. RESULTS: The PRDX1 splice mutations and activation of numerous cryptic splice sites produced antisense readthrough transcripts encompassing the bidirectional MMACHC/CCDC163P promoter and the TESK2 promoter, resulting in the silencing of both the MMACHC and TESK2 genes through the deposition of SETD2-dependent H3K36me3 marks and the generation of epimutations in the CpG islands of the two promoters. CONCLUSIONS: The antisense readthrough transcription of the mutated PRDX1 produces an epigenetic silencing of MMACHC and TESK2. We propose using the term 'epi-digenism' to define this epigenetic disorder that affects two genes. Epi-cblC is an entity that differs from cblC. Indeed, the PRDX1 and TESK2 altered expressions are observed in epi-cblC but not in cblC, suggesting further evaluating the potential consequences on cancer risk and spermatogenesis.


Subject(s)
Homocystinuria , Vitamin B 12 , DNA Methylation , Homocystinuria/genetics , Homocystinuria/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mutation , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Serine-Threonine Kinases , Vitamins
9.
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
10.
Nat Commun ; 13(1): 134, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013307

ABSTRACT

Combined methylmalonic acidemia and homocystinuria (cblC) is the most common inborn error of intracellular cobalamin metabolism and due to mutations in Methylmalonic Aciduria type C and Homocystinuria (MMACHC). Recently, mutations in the transcriptional regulators HCFC1 and RONIN (THAP11) were shown to result in cellular phenocopies of cblC. Since HCFC1/RONIN jointly regulate MMACHC, patients with mutations in these factors suffer from reduced MMACHC expression and exhibit a cblC-like disease. However, additional de-regulated genes and the resulting pathophysiology is unknown. Therefore, we have generated mouse models of this disease. In addition to exhibiting loss of Mmachc, metabolic perturbations, and developmental defects previously observed in cblC, we uncovered reduced expression of target genes that encode ribosome protein subunits. We also identified specific phenotypes that we ascribe to deregulation of ribosome biogenesis impacting normal translation during development. These findings identify HCFC1/RONIN as transcriptional regulators of ribosome biogenesis during development and their mutation results in complex syndromes exhibiting aspects of both cblC and ribosomopathies.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Homocystinuria/genetics , Host Cell Factor C1/genetics , Oxidoreductases/genetics , Repressor Proteins/genetics , Ribosomes/genetics , Vitamin B 12 Deficiency/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Disease Models, Animal , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Homocystinuria/metabolism , Homocystinuria/pathology , Host Cell Factor C1/deficiency , Humans , Male , Mice , Mice, Knockout , Mutation , Organelle Biogenesis , Oxidoreductases/deficiency , Protein Biosynthesis , Protein Subunits/genetics , Protein Subunits/metabolism , Repressor Proteins/deficiency , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Ribosomes/pathology , Vitamin B 12/metabolism , Vitamin B 12 Deficiency/metabolism , Vitamin B 12 Deficiency/pathology
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
Eur J Med Genet ; 62(10): 103713, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31279840

ABSTRACT

Cobalamin (cbl) C disease is a rare autosomal recessive inheritance disease, which is the most common cobalamin metabolic disorder. Its clinical phenotype involves multiple systems with varying degrees of severity, where in mild cases can be asymptomatic for many years, whereas severe cases may cause death during the neonatal period. The disease is caused by mutations in the MMACHC gene located on chromosome 1p34.1 that contains 5 exons; among which, exons 1-4 have an 849 bp coding sequence that encodes a protein containing 282 amino acids. Through clinical physical examination and laboratory tests, especially blood and urine screening, we found 28 cblC pediatric patients with clinical manifestations, such as mental retardation, motor development delay, epilepsy, metabolic acidosis, vomiting and diarrhea. By Sanger sequencing, we found homozygous or compound heterozygous mutations of MMACHC in 27 of the patients, and single heterozygous mutation of MMACHC in one of them. The c.609G > A, c.658-660delAAG, c.80A > G and c.482G > A mutations accounted for 43.64% (24/55), 10.91% (6/55), 9.09% (5/55) and 7.27% (4/55) of all the mutations, respectively. This spectrum finding is basically consistent with the previously reported data in Chinese patients. The most common c.609G > A mutation may likely lead to early-onset cblC disease. In previous literature involving a large sample of Caucasian cblC cases, the mutation spectrum of MMACHC gene is almost completely different from that of the Chinese population. The most common mutations in the Caucasian population were c.271dupA, c.394C > T and c.331C > T, which account for 48.05% (542/1128), 13.65% (154/1128) and 7.36% (83/1128) of all the mutant alleles, respectively. The c.271dupA mutation and c.331C > T mutation were mainly associated with early-onset cblC in children less than 1 year old, whilst the c.394C > T mutation was mainly associated with late-onset cblC patients characterised by isolated acute nervous system abnormalities. We also analysed the cause behind the different mutation spectrum of MMACHC gene between the Chinese and Caucasian populations.


Subject(s)
Genetic Predisposition to Disease , Homocystinuria/genetics , Mutation , Oxidoreductases/genetics , Vitamin B 12 Deficiency/congenital , Age of Onset , Alleles , Child , Child, Preschool , Female , Genetic Association Studies , Genotype , Homocystinuria/diagnosis , Homocystinuria/metabolism , Humans , Infant , Infant, Newborn , Male , Oxidoreductases/metabolism , Phenotype , Vitamin B 12 Deficiency/diagnosis , Vitamin B 12 Deficiency/genetics , Vitamin B 12 Deficiency/metabolism
18.
Hum Gene Ther ; 30(9): 1093-1100, 2019 09.
Article in English | MEDLINE | ID: mdl-31084364

ABSTRACT

Cystathionine ß-synthase (CBS) deficiency is a recessive inborn error of metabolism characterized by extremely elevated total homocysteine (tHcy) in the blood. Patients diagnosed with CBS deficiency have a variety of clinical problems, including dislocated lenses, osteoporosis, cognitive and behavioral issues, and a significantly increased risk of thrombosis. Current treatment strategies involve a combination of vitamin supplementation and restriction of foods containing the homocysteine precursor methionine. Here, a mouse model for CBS deficiency (Tg-I278T Cbs-/-) was used to evaluate the potential of minicircle-based naked DNA gene therapy to treat CBS deficiency. A 2.3 kb DNA-minicircle containing the liver-specific P3 promoter driving the human CBS cDNA (MC.P3-hCBS) was delivered into Tg-I278T Cbs-/- mice via a single hydrodynamic tail vein injection. Mean serum tHcy decreased from 351 µM before injection to 176 µM 7 days after injection (p = 0.0005), and remained decreased for at least 42 days. Western blot analysis reveals significant minicircle-directed CBS expression in the liver tissue. Liver CBS activity increased 34-fold (12.8 vs. 432 units; p = 0.0004) in MC.P3-hCBS-injected animals. Injection of MC.P3-hCBS in young mice, subsequently followed for 202 days, showed that the vector can ameliorate the mouse homocystinuria alopecia phenotype. The present findings show that minicircle-based gene therapy can lower tHcy in a mouse model of CBS deficiency.


Subject(s)
Cystathionine beta-Synthase/genetics , DNA, Circular/genetics , Genetic Therapy , Genetic Vectors/genetics , Homocystinuria/genetics , Homocystinuria/therapy , Animals , Biomarkers , Cystathionine beta-Synthase/blood , Cystathionine beta-Synthase/deficiency , DNA, Circular/administration & dosage , Disease Models, Animal , Female , Gene Expression , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Homocystinuria/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Phenotype , Transfection/methods , Treatment Outcome
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...