Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 301
Filter
1.
Mol Genet Metab ; 142(1): 108345, 2024 May.
Article in English | MEDLINE | ID: mdl-38387306

ABSTRACT

Mutations in MMACHC cause cobalamin C disease (cblC, OMIM 277400), the commonest inborn error of vitamin B12 metabolism. In cblC, deficient activation of cobalamin results in methylcobalamin and adenosylcobalamin deficiency, elevating methylmalonic acid (MMA) and total plasma homocysteine (tHcy). We retrospectively reviewed the medical files of seven cblC patients: three compound heterozygotes for the MMACHC (NM_015506.3) missense variant c.158T>C p.(Leu53Pro) in trans with the common pathogenic mutation c.271dupA (p.(Arg91Lysfs*14), "compounds"), and four c.271dupA homozygotes ("homozygotes"). Compounds receiving hydroxocobalamin intramuscular injection monotherapy had age-appropriate psychomotor performance and normal ophthalmological examinations. In contrast, c.271dupA homozygotes showed marked psychomotor retardation, retinopathy and feeding problems despite penta-therapy (hydroxocobalamin, betaine, folinic acid, l-carnitine and acetylsalicylic acid). Pretreatment levels of plasma and urine MMA and tHcy were higher in c.271dupA homozygotes than in compounds. Under treatment, levels of the compounds approached or entered the reference range but not those of c.271dupA homozygotes (tHcy: compounds 9.8-32.9 µM, homozygotes 41.6-106.8 (normal (N) < 14); plasma MMA: compounds 0.14-0.81 µM, homozygotes, 10.4-61 (N < 0.4); urine MMA: compounds 1.75-48 mmol/mol creatinine, homozygotes 143-493 (N < 10)). Patient skin fibroblasts all had low cobalamin uptake, but this was milder in compound cells. Also, the distribution pattern of cobalamin species was qualitatively different between cells from compounds and from homozygotes. Compared to the classic cblC phenotype presented by c.271dupA homozygous patients, c.[158T>C];[271dupA] compounds had mild clinical and biochemical phenotypes and responded strikingly to hydroxocobalamin monotherapy.


Subject(s)
Carrier Proteins , Hydroxocobalamin , Phenotype , Vitamin B 12 Deficiency , Vitamin B 12 , Humans , Hydroxocobalamin/administration & dosage , Hydroxocobalamin/therapeutic use , Male , Female , Vitamin B 12 Deficiency/genetics , Vitamin B 12 Deficiency/drug therapy , Vitamin B 12 Deficiency/blood , Vitamin B 12/blood , Child, Preschool , Carrier Proteins/genetics , Retrospective Studies , Oxidoreductases/genetics , Child , Methylmalonic Acid/blood , Homocystinuria/drug therapy , Homocystinuria/genetics , Infant , Mutation, Missense , Homozygote , Heterozygote , Homocysteine/blood , Adolescent , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/blood , Adult
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.
Dis Model Mech ; 16(11)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37994477

ABSTRACT

Amino acids are organic molecules that serve as basic substrates for protein synthesis and have additional key roles in a diverse array of cellular functions, including cell signaling, gene expression, energy production and molecular biosynthesis. Genetic defects in the synthesis, catabolism or transport of amino acids underlie a diverse class of diseases known as inborn errors of amino acid metabolism. Individually, these disorders are rare, but collectively, they represent an important group of potentially treatable disorders. In this Clinical Puzzle, we discuss the pathophysiology, clinical features and management of three disorders that showcase the diverse clinical presentations of disorders of amino acid metabolism: phenylketonuria, lysinuric protein intolerance and homocystinuria due to cystathionine ß-synthase (CBS) deficiency. Understanding the biochemical perturbations caused by defects in amino acid metabolism will contribute to ongoing development of diagnostic and management strategies aimed at improving the morbidity and mortality associated with this diverse group of disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Homocystinuria , Humans , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/diagnosis , Homocystinuria/drug therapy , Amino Acids
5.
Mol Genet Metab ; 140(3): 107681, 2023 11.
Article in English | MEDLINE | ID: mdl-37604084

ABSTRACT

In early-onset (EO) cblC deficiency (MMACHC), hydroxocobalamin dose-intensification (OHCBL-DI) improved biochemical and clinical outcome. In mammals, Cobalamin is reduced, in a reaction mediated by MMACHC. Pathogenic variants in MMACHC disrupt the synthesis pathway of methyl-cobalamin (MetCbl) and 5'-deoxy-adenosyl-cobalamin (AdoCbl), cofactors for both methionine synthase (MS) and methyl-malonyl-CoA mutase (MCM) enzymes. In 5 patients (pts.), with EO cblC deficiency, biochemical and clinical responses were studied following OHCbl-DI (mean ± SD 6,5 ± 3,3 mg/kg/day), given early, before age 5 months (pts. 1, 2, 3 and 4) or lately, at age 5 years (pt. 5). In all pts., total homocysteine (tHcy), methyl-malonic acid (MMA) and Cob(III)alamin levels were measured. Follow-up was performed during 74/12 years (pts. 1, 2, 3), 33/12 years (pt. 4) and 34/12 years (pt. 5). OHCbl was delivered intravenously or subcutaneously. Mean ± SD serum Cob(III)alamin levels were 42,2 × 106 ± 28, 0 × 106 pg/ml (normal: 200-900 pg/ml). In all pts., biomarkers were well controlled. All pts., except pt. 5, who had poor vision, had central vision, mild to moderate nystagmus, and with peri-foveolar irregularity in pts. 1, 2 and 4, yet none had the classic bulls' eye maculopathy and retinal degeneration characteristic of pts. with EO cblC deficiency. Only pt. 5, had severe cognitive deficiency. Both visual and cognitive functions were better preserved with early than with late OHCBL-DI. OHCBL-DI is suggested to bypass MMACHC, subsequently to be rescued by methionine synthase reductase (MSR) and adenosyl-transferase (ATR) to obtain Cob(I)alamin resulting in improved cognitive and retinal function in pts. with EO cblC deficiency.


Subject(s)
Cognitive Dysfunction , Homocystinuria , Macular Degeneration , Vitamin B 12 Deficiency , Child, Preschool , Humans , Infant , Male , Cognitive Dysfunction/drug therapy , Homocystinuria/drug therapy , Homocystinuria/genetics , Hydroxocobalamin/therapeutic use , Macular Degeneration/drug therapy , Mammals , Oxidoreductases , Vitamin B 12/metabolism , Vitamin B 12 Deficiency/drug therapy
6.
J Pediatr Endocrinol Metab ; 36(8): 761-771, 2023 Aug 28.
Article in English | MEDLINE | ID: mdl-37440674

ABSTRACT

OBJECTIVES: To study the biochemical, clinical and molecular characteristics of 5,10- methylenetetrahydrofolate reductase (MTHFR) deficiency in Pakistani patients from a single center. METHODS: Medical charts, urine organic acid chromatograms, plasma methionine and Hcys levels, and molecular testing results of MTHFR gene of patients presenting at the Biochemical Genetics Clinic, AKUH from 2016 to 2022 were reviewed. RESULTS: Neonatal MTHFR deficiency was found in five patients. The median (IQR) age of symptom onset and diagnosis were 18 (8.5-22) and 26 (16.5-31) days. The median lag between symptom onset and diagnosis was 8 (4.5-12.5) days. The median age of treatment initiation and duration of treatment were 26 (16.5-49) and 32 (25.5-54) days. The most common clinical features were lethargy, poor feeding, and seizures. The MTHFR gene sequencing revealed homozygous variants p.K510K, p.R567*, and p.R157W. Renal insufficiency manifesting as elevated serum creatinine and responding to betaine therapy was noted in one patient. This has not been previously reported in neonatal MTHFR deficiency and may reflect engagement of alternate pathways of remethylation. Adult onset MTHFR deficiency was found in six patients, with a heterogeneous neurological presentation. The median lag between symptoms onset and diagnosis was 7 (3-11) years. MTHFR gene sequencing revealed homozygous variant p.A195V in five patients from one family and p.G261V in the other. Two of the five reported variants are novel that include p.R157W and p.G261V. CONCLUSIONS: Eleven patients of this rare disorder from a single center indicate the need for clinical awareness and appropriate biochemical evaluation to ensure optimal outcomes.


Subject(s)
Homocystinuria , Psychotic Disorders , Adult , Humans , Homocystinuria/diagnosis , Homocystinuria/genetics , Homocystinuria/drug therapy , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Pakistan , Psychotic Disorders/diagnosis , Psychotic Disorders/drug therapy , Psychotic Disorders/genetics
7.
Mol Genet Metab ; 139(4): 107653, 2023 08.
Article in English | MEDLINE | ID: mdl-37463544

ABSTRACT

Classical homocystinuria (HCU) is a rare inborn error of amino acid metabolism characterized by accumulation of homocysteine, an intermediate product of methionine metabolism, leading to significant systemic toxicities, particularly within the vascular, skeletal, and ocular systems. Most patients require lifelong dietary therapy with severe restriction of natural protein to minimize methionine intake, and many patients still struggle to maintain healthy homocysteine levels. Since eliminating methionine from the diet reduces homocysteine levels, we hypothesized that an enzyme that can degrade methionine within the gastrointestinal (GI) tract could help HCU patients maintain healthy levels while easing natural protein restrictions. We describe the preclinical development of CDX-6512, a methionine gamma lyase (MGL) enzyme that was engineered for stability and activity within the GI tract for oral administration to locally degrade methionine. CDX-6512 is stable to low pH and intestinal proteases, enabling it to survive the harsh GI environment without enteric coating and to degrade methionine freed from dietary protein within the small intestine. Administering CDX-6512 to healthy non-human primates following a high protein meal led to a dose-dependent suppression of plasma methionine. In Tg-I278T Cbs-/- mice, an animal model that recapitulates aspects of HCU disease including highly elevated serum homocysteine levels, oral dosing of CDX-6512 after a high protein meal led to suppression in serum levels of both methionine and homocysteine. When animals received a daily dose of CDX-6512 with a high protein meal for two weeks, the Tg-I278T Cbs-/- mice maintained baseline homocysteine levels, whereas homocysteine levels in untreated animals increased by 39%. These preclinical data demonstrate the potential of CDX-6512 as an oral enzyme therapy for HCU.


Subject(s)
Homocystinuria , Humans , Mice , Animals , Homocystinuria/drug therapy , Homocystinuria/genetics , Methionine/metabolism , Homocysteine , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Racemethionine , Gastrointestinal Tract/metabolism
8.
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
9.
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
10.
Orphanet J Rare Dis ; 17(1): 417, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36376887

ABSTRACT

BACKGROUND: Betaine is an "alternate" methyl donor for homocysteine remethylation catalyzed by betaine homocysteine methyltransferase (BHMT), an enzyme mainly expressed in the liver and kidney. Betaine has been used for more than 30 years in pyridoxine non-responsive cystathionine beta-synthase (pnrCBS) and cobalamin C (cblC) deficiencies to lower the hyperhomocysteinemia, although little is known about the optimal therapeutic dosage and its pharmacokinetic in these patients. AIMS: We compared 2 betaine doses (100 mg/kg/day vs. 250 mg/kg/day) in children affected by pnrCBS or cblC deficiencies. We also measured the pharmacokinetics parameters after a single dose of betaine (100 or 250 mg/kg) in these patients. METHODS: We conducted a prospective, randomized, crossover clinical trial with blinded evaluation. The primary outcome was the equivalence of total plasma homocysteine (tHcy) concentrations upon one-month oral treatment with betaine at 100 versus 250 mg/kg/day. RESULTS: Eleven patients completed the study (5 pnrCBS and 6 cblC). tHcy concentrations were equivalent after a one-month treatment period for the two betaine dosages. Multivariate analysis showed a significant effect of betaine dose on methionine (Met) (p = 0.01) and S-adenosylmethionine (SAM) concentrations (p = 0.006). CONCLUSIONS: Our analysis shows that there is no overt benefit to increasing betaine dosage higher than 100 mg/kg/day to lower tHcy concentrations in pnrCBS and cblC deficiencies. However, increasing betaine up to 250 mg/kg/d could benefit cblC patients through the increase of methionine and SAM concentrations, as low Met and SAM concentrations are involved in the pathophysiology of this disease. In contrast, in pnrCBS deficiency, betaine doses higher than 100 mg/kg/day could be harmful to these patients with pre-existing hypermethioninemia. TRIAL REGISTRATION: Clinical Trials, NCT02404337. Registered 23 May 2015-prospectively registered, https://clinicaltrials.gov .


Subject(s)
Homocystinuria , Vitamin B 12 Deficiency , Humans , Child , Betaine/therapeutic use , Prospective Studies , Homocystinuria/drug therapy , Cystathionine beta-Synthase/therapeutic use , Methionine , S-Adenosylmethionine/therapeutic use , Homocysteine
11.
Brasília; CONITEC; nov. 2022.
Non-conventional in Portuguese | BRISA/RedTESA | ID: biblio-1434441

ABSTRACT

INTRODUÇÃO: A homocistinúria ocorre por conta de mutações em enzimas envolvidas no metabolismo da metionina, sendo elas: deficiência da cistationina-beta-sintetase (CBS), deficiência da 5,10- metilenotetrahidrofolato redutase (MTHFR) ou defeito no cofator do metabolismo da cobalamina (cbl). É uma condição clínica que acomete um pequeno número de pessoas e designa um grupo de doenças metabólicas hereditárias, e em função de decorrer de mutações em enzimas do metabolismo da metionina, a homocistinúria, é uma doença progressiva, sendo mais grave a evolução quanto mais precocemente se manifestam os sintomas. Por ser uma doença progressiva e sistêmica, o quadro clínico da homocistinúria é caracterizado por complicações que podem atingir diversos sistemas biológicos. Algumas das principais manifestações clínicas são o deslocamento das lentes dos olhos para baixo (ectopia lentis), osteoporose, retardo mental, convulsões e, principalmente, fenômenos tromboembólicos. Os fenômenos tromboembólicos revestem-se da maior importância, por determinarem a ocorrência de eventos cardiovasculares sérios, como infarto do miocárdio e acidentes vasculares encefálicos muito precocemente na vida, com impacto significativo na morbimortalidade. PERGUNTA DE PESQUISA: O uso de betaína anidra como terapia complementar no tratamento de pacientes com homocistinúria com deficiências ou defeitos da cistationina-betasintetase, não responsivos a piridoxina (B6) é eficaz, seguro e custo-efetivo em relação às terapias disponíveis no SUS? EVIDÊNCIAS CIENTÍFICAS: Em revisão sistemática da literatura, o demandante selecionou 1 ECR e 04 séries de casos e 02 estudos observacionais com subgrupo de interesse. No relatório de avaliação crítica foi realizada nova busca nas bases de dados e foram excluídas da análise as quatro séries de casos pela limitação metodológica do tipo de estudo e o número muito baixo de pacientes. Observou-se que na maioria dos estudos, o uso de betaína levou a redução pelo menos numérica dos níveis de homocisteína. Destaca-se que em ECR, essa redução foi significativa para o grupo betaína frente ao placebo.) Ainda, foi observado aumento também pelo menos numérico da cisteína e da metionina em pacientes em uso de betaína.Entretanto, no ECR, o aumento da metionina no grupo betaína apesar de numericamente maior não apresentou significância estatística na comparação frente ao placebo. Os valores de densidade mineral óssea avaliados não foram observadas diferenças significativas entre betaína e placebo, apesar de pacientes em uso de betaína apresentarem resultados numericamente melhores. Em termos de segurança, a betaína foi considerada tolerável, sem relatos de disfunção hepática, renal ou de medula óssea e nenhum relato de efeito indesejado. Ainda, não foram reportados eventos adversos ou intolerância ao tratamento. AVALIAÇÃO ECONÔMICA: A análise econômica realizada pelo demandante baseou-se em modelo de custoutilidade e os resultados são apresentados na forma de custo incremental por ano de vida ganho e por QALY.O curso da doença foi simulado utilizando o modelo de Markov. O modelo incluiu três estados de saúde:Pacientes sem evento cardiovascular, pacientes com evento cardiovascular e morte. O horizonte temporal foi de 20 anos. Para um paciente viver um ano livre da ocorrência de eventos cardiovasculares (1 AVL de ECV) a RCEI foi de R$ 492.124,91. Para um paciente viver um ano com a qualidade de saúde plena (1 QALY) a RCEI foi de R$ 1.525.349,65 milhões. ANÁLISE DE IMPACTO ORÇAMENTÁRIO: Foi realizada pelo demandante, análise de impacto orçamentário (AIO) tendo como população alvo pacientes com homocistinúria clássica não responsivos à piridoxina (vitamina B6) O demandante criou dois cenários, um baseado em dados epidemiológicos e outro baseado em demanda aferida.A demanda epidemiológica , resulta em um investimento incremental, acumulados em 5 anos de R$ R$ 11.025.966 e de R$ 3.570.275 para a demanda aferida considerando um Market-share de 50% a 90% no quinto ano. MONITORAMENTO DO HORIZONTE TECNOLÓGICO: No horizonte considerado nesta análise, não se detectou nenhuma tecnologia potencial para o tratamento de pacientes com homocistinúria clássica, não responsivos a piridoxina (B6). Porém, cabe informar que existem duas tecnologias com estudos de fase 1/2 em andamento para homocistinúria clássica, independentemente da responsividade à piridoxina, a pegtarviliase e a pegtibatinase. CONSIDERAÇÕES FINAIS: A análise da evidência clínica apresentada pelo demandante sugere que a tecnologia proposta apresenta efetividade superior quando usada em combinação às alternativas disponíveis atualmente no SUS. Os estudos avaliando betaína são estudos com baixo número amostral,suscetíveis a vieses importantes e há ausência de um ensaio clínico randomizado que avalie a betaína anidra em um número de pacientes maior.O fato de ser uma doença rara corrobora para não termos evidências robustas dessa tecnologia. O modelo de custo-efetividade/utilidade construído pelo demandante utilizou uma abordagem considerando apenas três estados de transições e um horizonte temporal de 20 anos, porém não justificou essa escolha que difere do recomendado nas diretrizes brasileiras. Não foi referida a utilização de taxa de desconto. O modelo adotado considerou manifestações clínicas relacionadas apenas a eventos cardiovasculares, porém a homocistinúria têm outras manifestações clínicas importantes que podem impactar diretamente na qualidade de vida dos pacientes que não estão avaliadas no modelo. Os dados de custos dos medicamentos consideraram PMVG DE 17% sendo mais adequado a utilização de valores considerando 18% de imposto. Como a homocistinúria acomete pacientes pediátricos seria importante apresentar análises de sensibilidade ou justificar o valor do tratamento para não superestimar os custos já que o medicamento avaliado é calculado por peso do paciente. O demandante apresentou uma AIO adequada em relação ao market share e considerou a fragilidade dos dados da população ­alvo apresentando uma análise por demanda aferida e por demanda epidemiológica. A demanda aferida calculada apresenta dados mais perto da realidade dos pacientes diagnosticados no Brasil com homocistinúria. RECOMENDAÇÃO PRELIMINAR DA CONITEC: Os membros do Plenário, presentes na 113ª Reunião Ordinária da Conitec, no dia 05 de outubro de 2022, deliberaram que deliberaram que a matéria fosse disponibilizada em consulta pública com recomendação preliminar desfavorável à incorporação ao SUS da betaína anidra para o tratamento de pacientes com homocistinúria com deficiências ou defeitos da cistationina-beta- sintetase, não responsivos a piridoxina (B6). CONSULTA PÚBLICA: Foram recebidas 121 contribuições, sendo 07 pelo formulário técnico-científico e 114 pelo formulário sobre experiência ou opinião. Todas as contribuições de cunho técnico-científico recebidas foram contra a recomendação inicial da Conitec. A maioria dos respondentes do formulário de experiência e opinião manifestou-se favorável à incorporação da betaína anidra. Entre as opiniões favoráveis, destacaram-se os temas sobre direito à saúde e o papel essencial do medicamento avaliado para o tratamento dos pacientes não responsivos à piridoxina. Os participantes também mencionaram o aumento da qualidade de vida, o fato de o medicamento se destinar ao tratamento de uma doença rara, ser um medicamento recomendado por profissionais de saúde, o tratamento da doença demandar a associação do medicamento com outras tecnologias, a segurança em relação à padronização na produção do medicamento e se tratar de uma questão de sobrevivência. Em relação à experiência com a betaína anidra, como efeitos positivos ou facilidades, os respondentes relataram o controle da doença, boa resposta terapêutica, prevenção de manifestações clínicas da doença e ação anticoagulante. Em contraponto, reportaram a dificuldade de acesso ao medicamento. RECOMENDAÇÃO FINAL DA CONITEC: Pelo exposto, o Plenário da Conitec, em sua 12ª Reunião Extraordinária, no dia 29 de novembro de 2022, deliberou, por unanimidade, recomendar a não incorporação da betaína anidra para o tratamento de pacientes com homocistinúria com deficiências ou defeitos da cistationina-beta-sintetase, não responsivos a piridoxina (B6). Por fim, foi assinado o Registro de Deliberação nº 788/2022. DECISÃO: Não incorporar, no âmbito do Sistema Único de Saúde - SUS, a betaína anidra para o tratamento de pacientes com homocistinúria com deficiências ou defeitos da cistationina-betasintetase, não responsivos a piridoxina (B6), conforme protocolo estabelecido pelo Ministério da Saúde, conforme a Portaria nº 176, publicada no Diário Oficial da União nº 238, seção 1, página 231, em 20 de dezembro de 2022.


Subject(s)
Humans , Pyridoxine/adverse effects , Betaine/therapeutic use , Cystathionine beta-Synthase/deficiency , Homocystinuria/drug therapy , Unified Health System , Brazil , Cost-Benefit Analysis/economics
12.
Neurocase ; 28(4): 388-392, 2022 08.
Article in English | MEDLINE | ID: mdl-36219783

ABSTRACT

Combined methylmalonic acidemia and homocystinuria, is a rare autosomal recessive disorder due to defective intracellular cobalamin metabolism. We report an 18-year-old Chinese male who presented with hypermyotonia, seizures, and congenital heart diseases. Mutation analysis revealed c.365A>T and c.482 G>A mutations in the MMACHC gene, diagnosed with methylmalonic aciduria and homocystinuria (CblC type). After treatment with vitamin B12, L-carnitine, betaine, and folate, which resulted in an improvement in his clinical symptoms and laboratory values. This case emphasizes that inborn errors of metabolism should be considered for a teenager presenting with challenging or neurologic symptoms, especially when combined with unexplained heart diseases.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Heart Defects, Congenital , Homocystinuria , Male , Adolescent , Humans , Homocystinuria/complications , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/therapeutic use , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/genetics , Vitamin B 12 , Mutation , Heart Defects, Congenital/complications , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/drug therapy , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases/therapeutic use
13.
Indian J Ophthalmol ; 70(7): 2272-2278, 2022 07.
Article in English | MEDLINE | ID: mdl-35791106

ABSTRACT

Homocystinuria is a rare metabolic inborn disorder caused due to dysfunctional cystathionine ß-synthase (CBS) enzyme activity, thus resulting in elevated levels of methionine and homocysteine in the blood and urine. The timely recognition of this rare metabolic disorder and prompt methionine-restricted diet are crucial in lessening the systemic consequences. The recalcitrant cases have a higher risk for cardiovascular diseases, neurodegenerative diseases, neural tube defects, and other severe clinical complications. This review aims to present the ophthalmic spectrum of homocystinuria and its molecular basis, the disease management, as well as the current and potential treatment approaches with a greater emphasis on preventive strategies.


Subject(s)
Homocystinuria , Cystathionine beta-Synthase/therapeutic use , Homocystinuria/complications , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Humans , Methionine/therapeutic use
14.
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
15.
Zhonghua Er Ke Za Zhi ; 60(6): 533-538, 2022 Jun 02.
Article in Chinese | MEDLINE | ID: mdl-35658358

ABSTRACT

Objective: To analyze the clinical features and CBS gene variants of 13 patients with classic homocystinuria, and the strategies of individual treatment and prevention were explored. Methods: The general information, clinical manifestations, laboratory tests, cranial images, CBS gene variants, diagnosis and therapeutic strategies of 13 patients with classic homocystinuria admitted to the Department of Pediatrics of Children's Hospital Affiliated to Zhengzhou University and Peking University First Hospital from November 2013 to June 2021 were analyzed retrospectively. Results: There were 13 patients diagnosed at the age of 10 days to 14 years, 6 were male and 7 were female. There were 3 patients detected by newborn screening and received treatment at the asymptomatic stage. There were 10 patients clinically diagnosed at the age of 5 to 14 years. Their symptoms appeared at age of 1 to 6 years. The major clinical manifestations were marfanoid features, lens dislocation and (or) myopia, developmental delay, osteoporosis, and cardiovascular diseases. Brain magnetic resonance imaging showed asymmetric infarcts in 4 patients and hypomyelination in 1 case. Increased blood methionine, plasma total homocysteine and urinary total homocysteine with normal urinary methylmalonic acid were found in 13 patients. The biochemical features were consistent with classic homocystinuria. Totally 18 variants were identified in CBS gene of 13 patients, 10 variants were novel and 8 were reported. only 1 patient was partially responsive to vitamin B6 treatment, while 12 cases were non-responsive. They were mainly treated with low methionine diet and betaine supplement. Three vitamin B6 non-responsive cases received liver transplantation at age of 3, 8 and 8 years, respectively. Their blood methionine and total homocysteine returned to normal within a week after liver transplantation. One patient died. Prenatal diagnosis was performed for a fetus when the mother was pregnant again. Two pathogenic CBS gene variants were identified from the amniocytes as same as the proband. Conclusions: The clinical manifestations of classic homocystinuria are complex and variable. Blood amino acid analysis, serum or urine total homocysteine assay and gene analysis are critical for its diagnosis. There were 10 novel CBS gene varients were identified expanding the CBS gene varient spectrum. Liver transplantation is an effective treatment. Prenatal diagnosis is important to prevent classic homocysteinuria.


Subject(s)
Homocystinuria , Adolescent , Child , Child, Preschool , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/therapeutic use , Female , Homocysteine/therapeutic use , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Homocystinuria/genetics , Humans , Infant , Infant, Newborn , Male , Methionine/therapeutic use , Pyridoxine/therapeutic use , Retrospective Studies , Vitamins/therapeutic use
16.
Biochim Biophys Acta Proteins Proteom ; 1870(6): 140793, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35618206

ABSTRACT

The cblC disease is an inborn disorder of the vitamin B12 (cobalamin, Cbl) metabolism characterized by methylmalonic aciduria and homocystinuria. The clinical consequences of this disease are devastating and, even when early treated with current therapies, the affected children manifest symptoms involving vision, growth, and learning. The illness is caused by mutations in the gene codifying for MMACHC, a 282aa protein that transports and transforms the different Cbl forms. Here we present data on the structural properties of the truncated protein p.R132X resulting from the c.394C > T mutation that, along with c.271dupA and c.331C > T, is among the most common mutations in cblC. Although missing part of the Cbl binding domain, p.R132X is associated to late-onset symptoms and, therefore, it is supposed to retain residual function. However, to our knowledge structural-functional studies on c.394C > T mutant aimed at verifying this hypothesis are still lacking. By using a biophysical approach including Circular Dichroism, fluorescence, Small Angle X-ray Scattering, and Molecular Dynamics, we show that the mutant protein MMACHC-R132X retains secondary structure elements and remains compact in solution, partly preserving its binding affinity for Cbl. Insights on the fragile stability of MMACHC-R132X-Cbl are provided.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Homocystinuria , Amino Acid Metabolism, Inborn Errors/genetics , Carrier Proteins , Child , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Homocystinuria/genetics , Humans , Mutation , Oxidoreductases/metabolism , Vitamin B 12/metabolism
17.
J Inherit Metab Dis ; 45(4): 848-861, 2022 07.
Article in English | MEDLINE | ID: mdl-35460084

ABSTRACT

MTHFR deficiency is a severe inborn error of metabolism leading to impairment of the remethylation of homocysteine to methionine. Neonatal and early-onset patients mostly exhibit a life-threatening acute neurologic deterioration. Furthermore, data on early-onset patients' long-term outcomes are scarce. The aims of this study were (1) to study and describe the clinical and laboratory parameters of early-onset MTHFR-deficient patients (i.e., ≤3 months of age) and (2) to identify predictive factors for severe neurodevelopmental outcomes in a cohort with early and late onset MTHFR-deficient patients. To this end, we conducted a retrospective, multicentric, international cohort study on 72 patients with MTHFR deficiency from 32 international metabolic centres. Characteristics of the 32 patients with early-onset MTHFR deficiency were described at time of diagnosis and at the last follow-up visit. Logistic regression analysis was used to identify predictive factors of severe neurodevelopmental outcome in a broader set of patients with early and non-early-onset MTHFR deficiency. The majority of early-onset MTHFR-deficient patients (n = 32) exhibited neurologic symptoms (76%) and feeding difficulties (70%) at time of diagnosis. At the last follow-up visit (median follow-up time of 8.1 years), 76% of treated early-onset patients (n = 29) exhibited a severe neurodevelopmental outcome. Among the whole study population of 64 patients, pre-symptomatic diagnosis was independently associated with a significantly better neurodevelopmental outcome (adjusted OR 0.004, [0.002-0.232]; p = 0.003). This study provides evidence for benefits of pre-symptomatic diagnosis and appropriate therapeutic management, highlighting the need for systematic newborn screening for MTHFR deficiency and pre-symptomatic treatment that may improve outcome.


Subject(s)
Homocystinuria , Cohort Studies , Homocysteine , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Humans , Infant, Newborn , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Muscle Spasticity/diagnosis , Psychotic Disorders , Retrospective Studies
19.
J Inherit Metab Dis ; 45(4): 719-733, 2022 07.
Article in English | MEDLINE | ID: mdl-35358327

ABSTRACT

Patient registries for rare diseases enable systematic data collection and can also be used to facilitate postauthorization safety studies (PASS) for orphan drugs. This study evaluates the PASS for betaine anhydrous (Cystadane), conducted as public private partnership (PPP) between the European network and registry for homocystinurias and methylation defects and the marketing authorization holder (MAH). Data were prospectively collected, 2013-2016, in a noninterventional, international, multicenter, registry study. Putative adverse and severe adverse events were reported to the MAH's pharmacovigilance. In total, 130 individuals with vitamin B6 nonresponsive (N = 54) and partially responsive (N = 7) cystathionine beta-synthase (CBS) deficiency, as well as 5,10-methylenetetrahydrofolate reductase (MTHFR; N = 21) deficiency and cobalamin C (N = 48) disease were included. Median (range) duration of treatment with betaine anhydrous was 6.8 (0-9.8) years. The prescribed betaine dose exceeded the recommended maximum (6 g/day) in 49% of individuals older than 10 years because of continued dose adaptation to weight; however, with disease-specific differences (minimum: 31% in B6 nonresponsive CBS deficiency, maximum: 67% in MTHFR deficiency). Despite dose escalation no new or potential risk was identified. Combined disease-specific treatment decreased mean ± SD total plasma homocysteine concentrations from 203 ± 116 to 81 ± 51 µmol/L (p < 0.0001), except in MTHFR deficiency. Recommendations for betaine anhydrous dosage were revised for individuals ≥ 10 years. PPPs between MAH and international scientific consortia can be considered a reliable model for implementing a PASS, reutilizing well-established structures and avoiding data duplication and fragmentation.


Subject(s)
Homocystinuria , Psychotic Disorders , Betaine/adverse effects , Cystathionine beta-Synthase , Homocysteine , Homocystinuria/drug therapy , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Muscle Spasticity
20.
Orphanet J Rare Dis ; 17(1): 33, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35109910

ABSTRACT

BACKGROUND: Methylmalonic aciduria and homocystinuria, CblC type (OMIM #277400) is the most common disorder of cobalamin intracellular metabolism, an autosomal recessive disease, whose biochemical hallmarks are hyperhomocysteinemia, methylmalonic aciduria and low plasma methionine. Despite being a well-recognized disease for pediatricians, there is scarce awareness of its adult presentation. A thorough analysis and discussion of cobalamin C defect presentation in adult patients has never been extensively performed. This article reviews the published data and adds a new case of the latest onset of symptoms ever described for the disease. RESULTS: We present the emblematic case of a 45-year-old male, describing the diagnostic odyssey he ventured through to get to the appropriate treatment and molecular diagnosis. Furthermore, available clinical, biochemical and molecular data from 22 reports on cases and case series were collected, resulting in 45 adult-onset CblC cases, including our own. We describe the onset of the disease in adulthood, encompassing neurological, psychiatric, renal, ophthalmic and thromboembolic symptoms. In all cases treatment with intramuscular hydroxycobalamin was effective in reversing symptoms. From a molecular point of view adult patients are usually compound heterozygous carriers of a truncating and a non-truncating variant in the MMACHC gene. CONCLUSION: Adult onset CblC disease is a rare disorder whose diagnosis can be delayed due to poor awareness regarding its presenting insidious symptoms and biochemical hallmarks. To avoid misdiagnosis, we suggest that adult onset CblC deficiency is acknowledged as a separate entity from pediatric late onset cases, and that the disease is considered in the differential diagnosis in adult patients with atypical hemolytic uremic syndromes and/or slow unexplained decline in renal function and/or idiopathic neuropathies, spinal cord degenerations, ataxias and/or recurrent thrombosis and/or visual field defects, maculopathy and optic disc atrophy. Plasma homocysteine measurement should be the first line for differential diagnosis when the disease is suspected. To further aid diagnosis, it is important that genes belonging to the intracellular cobalamin pathway are included within gene panels routinely tested for atypical hemolytic uremic syndrome and chronic kidney disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Homocystinuria , Vitamin B 12 Deficiency , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/therapy , Heterozygote , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Homocystinuria/genetics , Humans , Male , Middle Aged , Oxidoreductases/genetics , Oxidoreductases/therapeutic use , Vitamin B 12/therapeutic use , Vitamin B 12 Deficiency/diagnosis , Vitamin B 12 Deficiency/drug therapy , Vitamin B 12 Deficiency/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...