Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Genet Metab ; 141(1): 108111, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38103461

RESUMO

Methionine dependence, the inability to grow in culture when methionine in the medium is replaced by its metabolic precursor homocysteine, occurs in many tumor cell lines. In most affected lines, the cause of methionine dependence is not known. An exception is the melanoma-derived cell line MeWo-LC1, in which hypermethylation of the MMACHC gene is associated with decreased MMACHC expression. Decreased expression results in decreased provision of the methylcobalamin cofactor required for activity of methionine synthase and thus decreased conversion of homocysteine to methionine. Analysis of data in the Cancer Cell Line Encyclopedia Archive demonstrated that MMACHC hypermethylation and decreased MMACHC expression occurred more frequently in melanoma cell lines when compared to other tumor cell lines. We further investigated methionine dependence and aspects of MMACHC function in a panel of six melanoma lines, including both melanoma lines with known methionine dependence status (MeWo, which is methionine independent, and A375, which is methionine dependent). We found that the previously unclassified melanoma lines HMCB, Colo829 and SH-4 were methionine dependent, while SK-Mel-28 was methionine independent. However, despite varying levels of MMACHC methylation and expression, none of the tested lines had decreased methylcobalamin and adenosylcobalamin synthesis as seen in MeWo-LC1, and the functions of both cobalamin-dependent enzymes methionine synthase and methylmalonyl-CoA mutase were intact. Thus, while melanoma lines were characterized by relatively high levels of MMACHC methylation and low expression, the defect in metabolism observed in MeWo-LC1 was unique, and decreased MMACHC expression was not a cause of methionine dependence in the other melanoma lines.


Assuntos
Melanoma , Metionina , Humanos , Metionina/metabolismo , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Racemetionina/metabolismo , Linhagem Celular Tumoral , Metilação de DNA , Homocisteína/metabolismo , Vitamina B 12/metabolismo , Oxirredutases/metabolismo
2.
Mol Genet Metab ; 142(1): 108345, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38387306

RESUMO

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.


Assuntos
Proteínas de Transporte , Hidroxocobalamina , Fenótipo , Deficiência de Vitamina B 12 , Vitamina B 12 , Humanos , Hidroxocobalamina/administração & dosagem , Hidroxocobalamina/uso terapêutico , Masculino , Feminino , Deficiência de Vitamina B 12/genética , Deficiência de Vitamina B 12/tratamento farmacológico , Deficiência de Vitamina B 12/sangue , Vitamina B 12/sangue , Pré-Escolar , Proteínas de Transporte/genética , Estudos Retrospectivos , Oxirredutases/genética , Criança , Ácido Metilmalônico/sangue , Homocistinúria/tratamento farmacológico , Homocistinúria/genética , Lactente , Mutação de Sentido Incorreto , Homozigoto , Heterozigoto , Homocisteína/sangue , Adolescente , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Erros Inatos do Metabolismo dos Aminoácidos/sangue , Adulto
3.
Hum Mol Genet ; 29(12): 1969-1985, 2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32068834

RESUMO

Vitamin B12 or cobalamin (Cbl) metabolism can be affected by genetic defects leading to defective activity of either methylmalonyl-CoA mutase or methionine synthase or both enzymes. Patients usually present with a wide spectrum of pathologies suggesting that various cellular processes could be affected by modifications in gene expression. We have previously demonstrated that these genetic defects are associated with subcellular mislocalization of RNA-binding proteins (RBP) and subsequent altered nucleo-cytoplasmic shuttling of mRNAs. In order to characterize the possible changes of gene expression in these diseases, we have investigated global gene expression in fibroblasts from patients with cblC and cblG inherited disorders by RNA-seq. The most differentially expressed genes are strongly associated with developmental processes, neurological, ophthalmologic and cardiovascular diseases. These associations are consistent with the clinical presentation of cblC and cblG disorders. Multivariate analysis of transcript processing revaled splicing alterations that led to dramatic changes in cytoskeleton organization, response to stress, methylation of macromolecules and RNA binding. The RNA motifs associated with this differential splicing reflected a potential role of RBP such as HuR and HNRNPL. Proteomic analysis confirmed that mRNA processing was significantly disturbed. This study reports a dramatic alteration of gene expression in fibroblasts of patients with cblC and cblG disorders, which resulted partly from disturbed function of RBP. These data suggest to evaluate the rescue of the mislocalization of RBP as a potential strategy in the treatment of severe cases who are resistant to classical treatments with co-enzyme supplements.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , Oxirredutases/genética , Deficiência de Vitamina B 12/genética , Vitamina B 12/genética , Processamento Alternativo/genética , Linhagem Celular , Proteína Semelhante a ELAV 1/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica/genética , Humanos , Proteômica , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas/genética , Vitamina B 12/metabolismo , Deficiência de Vitamina B 12/patologia
4.
Hum Genet ; 141(7): 1309-1325, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35190856

RESUMO

Epigenetic diseases can be produced by a stable alteration, called an epimutation, in DNA methylation, in which epigenome alterations are directly involved in the underlying molecular mechanisms of the disease. This review focuses on the epigenetics of two inherited metabolic diseases, epi-cblC, an inherited metabolic disorder of cobalamin (vitamin B12) metabolism, and alpha-thalassemia type α-ZF, an inherited disorder of α2-globin synthesis, with a particular interest in the role of aberrant antisense transcription of flanking genes in the generation of epimutations in CpG islands of gene promoters. In both disorders, the epimutation is triggered by an aberrant antisense transcription through the promoter, which produces an H3K36me3 histone mark involved in the recruitment of DNA methyltransferases. It results from diverse genetic alterations. In alpha-thalassemia type α-ZF, a deletion removes HBA1 and HBQ1 genes and juxtaposes the antisense LUC7L gene to the HBA2 gene. In epi-cblC, the epimutation in the MMACHC promoter is produced by mutations in the antisense flanking gene PRDX1, which induces a prolonged antisense transcription through the MMACHC promoter. The presence of the epimutation in sperm, its transgenerational inheritance via the mutated PRDX1, and the high expression of PRDX1 in spermatogonia but its nearly undetectable transcription in spermatids and spermatocytes, suggest that the epimutation could be maintained during germline reprogramming and despite removal of aberrant transcription. The epivariation seen in the MMACHC promoter (0.95 × 10-3) is highly frequent compared to epivariations affecting other genes of the Online Catalog of Human Genes and Genetic Disorders in an epigenome-wide dataset of 23,116 individuals. This and the comparison of epigrams of two monozygotic twins suggest that the aberrant transcription could also be influenced by post-zygotic environmental exposures.


Assuntos
Doenças Metabólicas , Talassemia alfa , Metilação de DNA , Epigênese Genética , Humanos , Masculino , Doenças Metabólicas/genética , Oxirredutases/genética , Sêmen , Talassemia alfa/genética
5.
Am J Med Genet A ; 188(4): 1124-1141, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35107211

RESUMO

The biological and clinical significance of the p.E88del variant in the transcobalamin receptor, CD320, is unknown. This allele is annotated in ClinVar as likely benign, pathogenic, and of uncertain significance. To determine functional consequence and clinical relevance of this allele, we employed cell culture and genetic association studies. Fibroblasts from 16 CD320 p.E88del homozygotes exhibited reduced binding and uptake of cobalamin. Complete ascertainment of newborns with transiently elevated C3 (propionylcarnitine) in New York State demonstrated that homozygosity for CD320 p.E88del was over-represented (7/348, p < 6 × 10-5 ). Using population data, we estimate that ~85% of the p.E88del homozygotes born in the same period did not have elevated C3, suggesting that cobalamin metabolism in the majority of these infants with this genotype is unaffected. Clinical follow-up of 4/9 homozygous individuals uncovered neuropsychological findings, mostly in speech and language development. None of these nine individuals exhibited perturbation of cobalamin metabolism beyond the newborn stage even during periods of acute illness. Newborns homozygous for this allele in the absence of other factors are at low risk of requiring clinical intervention, although more studies are required to clarify the natural history of various CD320 variants across patient populations.


Assuntos
Receptores de Superfície Celular , Transcobalaminas , Antígenos CD , Estudos de Associação Genética , Humanos , Lactente , Recém-Nascido , Receptores de Superfície Celular/genética , Transcobalaminas/genética , Transcobalaminas/metabolismo , Vitamina B 12/metabolismo
6.
J Inherit Metab Dis ; 45(2): 157-168, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34625984

RESUMO

Methionine synthase deficiency (cblG complementation group) is a rare inborn error of metabolism affecting the homocysteine re-methylation pathway. It leads to a biochemical phenotype of hyperhomocysteinemia and hypomethioninemia. The clinical presentation of cblG is variable, ranging from seizures, encephalopathy, macrocytic anemia, hypotonia, and feeding difficulties in the neonatal period to onset of psychiatric symptoms or acute neurologic changes in adolescence or adulthood. Given the variable and nonspecific symptoms seen in cblG, the diagnosis of affected patients is often delayed. Medical management of cblG includes the use of hydroxocobalamin, betaine, folinic acid, and in some cases methionine supplementation. Treatment has been shown to lead to improvement in the biochemical profile of affected patients, with lowering of total homocysteine levels and increasing methionine levels. However, the published literature contains differing conclusions on whether treatment is effective in changing the natural history of the disease. Herein, we present five patients with cblG who have shown substantial clinical benefit from treatment with objective improvement in their neurologic outcomes. We demonstrate more favorable outcomes in our patients who were treated early in life, especially those who were treated before neurologic symptoms manifested. Given improved outcomes from treatment of presymptomatic patients, cblG warrants inclusion in newborn screening.


Assuntos
Metionina , Vitamina B 12 , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/deficiência , Adulto , Erros Inatos do Metabolismo dos Aminoácidos , Diagnóstico Precoce , Homocisteína , Humanos , Erros Inatos do Metabolismo , Vitamina B 12/metabolismo
7.
Mol Genet Metab ; 132(3): 155-161, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33487542

RESUMO

Methionine dependence of tumor cell lines, the inability to grow in tissue culture media lacking methionine but supplemented with homocysteine, has been known for decades, but an understanding of the mechanism underlying this phenomenon remains incomplete. Methionine dependence of certain glioma and melanoma cell lines has been linked to alterations in the metabolism of cobalamin (vitamin B12). In the MeWo LC1 melanoma line, complementation analysis demonstrated that the genetic defect affected the same locus mutated in the cblC inborn error of cobalamin metabolism; hypermethylation of the MMACHC promoter was subsequently demonstrated. Analysis of data in the Cancer Cell Line Encyclopedia showed increased MMACHC methylation levels in melanoma lines compared to other types of cancer. RNA sequencing data from isolated tumors, tabulated at the cBioPortal for Cancer Genomics website, showed decreased MMACHC expression compared to other tumors; and methylation data tabulated at the TGGA Wanderer website demonstrated increased MMACHC methylation. These data suggest that disruptions in cobalamin metabolism might play a more general role in methionine dependence, and potentially in the pathogenesis of melanoma cell lines and primary tumors.


Assuntos
Genômica , Neoplasias/genética , Oxirredutases/genética , Vitamina B 12/uso terapêutico , Metilação de DNA/efeitos dos fármacos , Humanos , Metionina/metabolismo , Neoplasias/metabolismo , Análise de Sequência de RNA
8.
Genet Med ; 22(7): 1280, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32483295

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Genet Med ; 22(2): 432-436, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31462756

RESUMO

PURPOSE: Our laboratory has classified patients with methylmalonic aciduria using somatic cell studies for over four decades. We have accumulated 127 fibroblast lines from patients with persistent elevated methylmalonic acid (MMA) levels in which no genetic cause could be identified. Cultured fibroblasts from 26 of these patients had low [14C]propionate incorporation into macromolecules, possibly reflecting decreased methylmalonyl-CoA mutase function. METHODS: Genome sequencing (GS), copy-number variation (CNV) analysis, and RNA sequencing were performed on genomic DNA and complementary DNA (cDNA) from these 26 patients. RESULTS: No patient had two pathogenic variants in any gene associated with cobalamin metabolism. Nine patients had heterozygous variants of unknown significance previously identified by a next-generation sequencing (NGS) panel targeting cobalamin metabolic genes. Three patients had pathogenic changes in genes not associated with cobalamin metabolism (PCCA, EPCAM, and a 17q12 duplication) that explain parts of their phenotypes other than elevated MMA. CONCLUSION: Genome and RNA sequencing did not detect any additional putative causal genetic defects in known cobalamin genes following somatic cell studies and the use of a targeted NGS panel. They did detect pathogenic variants in other genes in three patients that explained some aspects of their clinical presentation.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Ácido Metilmalônico/metabolismo , Vitamina B 12/genética , Sequência de Bases/genética , Linhagem Celular , Feminino , Fibroblastos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Lactente , Recém-Nascido , Masculino , Metilmalonil-CoA Mutase/genética , Metilmalonil-CoA Mutase/metabolismo , Mutação/genética , Análise de Sequência de RNA/métodos , Vitamina B 12/metabolismo , Sequenciamento do Exoma/métodos
10.
Mol Genet Metab ; 130(3): 179-182, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32414565

RESUMO

MTHFD1 is a trifunctional protein containing 10-formyltetrahydrofolate synthetase, 5,10-methenyltetrahydrofolate cyclohydrolase and 5,10-methylenetetrahydrofolate dehydrogenase activities. It is encoded by MTHFD1 and functions in the cytoplasmic folate cycle where it is involved in de novo purine synthesis, synthesis of thymidylate and remethylation of homocysteine to methionine. Since the first reported case of severe combined immunodeficiency resulting from MTHFD1 mutations, seven additional patients ascertained through molecular analysis have been reported with variable phenotypes, including megaloblastic anemia, atypical hemolytic uremic syndrome, hyperhomocysteinemia, microangiopathy, infections and autoimmune diseases. We determined the level of MTHFD1 expression and dehydrogenase specific activity in cell extracts from cultured fibroblasts of three previously reported patients, as well as a patient with megaloblastic anemia and recurrent infections with compound heterozygous MTHFD1 variants that were predicted to be deleterious. MTHFD1 protein expression determined by Western blotting in fibroblast extracts from three of the patients was markedly decreased compared to expression in wild type cells (between 4.8 and 14.3% of mean control values). MTHFD1 expression in the fourth patient was approximately 44% of mean control values. There was no detectable methylenetetrahydrofolate dehydrogenase specific activity in extracts from any of the four patients. This is the first measurement of MTHFD1 function in MTHFD1 deficient patients and confirms the previous molecular diagnoses.


Assuntos
Fibroblastos/patologia , Deficiência de Ácido Fólico/diagnóstico , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Metilenotetra-Hidrofolato Desidrogenase (NADP)/metabolismo , Antígenos de Histocompatibilidade Menor/genética , Antígenos de Histocompatibilidade Menor/metabolismo , Mutação , Imunodeficiência Combinada Severa/diagnóstico , Estudos de Casos e Controles , Células Cultivadas , Fibroblastos/metabolismo , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Humanos , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/metabolismo
11.
Curr Opin Clin Nutr Metab Care ; 23(4): 241-246, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32412981

RESUMO

PURPOSE OF REVIEW: Immune dysfunction, including severe combined immunodeficiency, has been described in genetic disorders affecting the metabolism of the vitamins cobalamin (vitamin B12) and folate. We have reviewed reports of clinical findings in patients with a number of inborn errors of cobalamin or folate metabolism, specifically looking for immune problems. RECENT FINDINGS: There is little evidence that immune function is affected in most of the disorders. Exceptions are Imerslund-Gräsbeck syndrome and hereditary folate malabsorption (affecting intestinal absorption of cobalamin and folate, respectively), transcobalamin deficiency (affecting transport of cobalamin in blood and cellular cobalamin uptake), and methylenetetrahydrofolate dehydrogenase 1 deficiency (catalyzing cytoplasmic interconversion of reduced folate coenzyme derivatives). SUMMARY: Although some inborn errors of cobalamin or folate can be associated with immune dysfunction, the degree and type of immune dysfunction vary with no obvious pattern.


Assuntos
Deficiência de Ácido Fólico/imunologia , Síndromes de Malabsorção/imunologia , Fenômenos Fisiológicos da Nutrição/imunologia , Doenças da Imunodeficiência Primária/imunologia , Deficiência de Vitamina B 12/imunologia , Anemia Megaloblástica/congênito , Anemia Megaloblástica/imunologia , Ácido Fólico/genética , Ácido Fólico/imunologia , Deficiência de Ácido Fólico/congênito , Humanos , Síndromes de Malabsorção/congênito , Metilenotetra-Hidrofolato Desidrogenase (NADP)/deficiência , Metilenotetra-Hidrofolato Desidrogenase (NADP)/imunologia , Antígenos de Histocompatibilidade Menor/imunologia , Proteinúria/congênito , Proteinúria/imunologia , Transcobalaminas/deficiência , Transcobalaminas/imunologia , Vitamina B 12/genética , Vitamina B 12/imunologia , Deficiência de Vitamina B 12/congênito
12.
Hum Mol Genet ; 26(15): 2838-2849, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28449119

RESUMO

CblX (MIM309541) is an X-linked recessive disorder characterized by defects in cobalamin (vitamin B12) metabolism and other developmental defects. Mutations in HCFC1, a transcriptional co-regulator which interacts with multiple transcription factors, have been associated with cblX. HCFC1 regulates cobalamin metabolism via the regulation of MMACHC expression through its interaction with THAP11, a THAP domain-containing transcription factor. The HCFC1/THAP11 complex potentially regulates genes involved in diverse cellular functions including cell cycle, proliferation, and transcription. Thus, it is likely that mutation of THAP11 also results in biochemical and other phenotypes similar to those observed in patients with cblX. We report a patient who presented with clinical and biochemical phenotypic features that overlap cblX, but who does not have any mutations in either MMACHC or HCFC1. We sequenced THAP11 by Sanger sequencing and discovered a potentially pathogenic, homozygous variant, c.240C > G (p.Phe80Leu). Functional analysis in the developing zebrafish embryo demonstrated that both THAP11 and HCFC1 regulate the proliferation and differentiation of neural precursors, suggesting important roles in normal brain development. The loss of THAP11 in zebrafish embryos results in craniofacial abnormalities including the complete loss of Meckel's cartilage, the ceratohyal, and all of the ceratobranchial cartilages. These data are consistent with our previous work that demonstrated a role for HCFC1 in vertebrate craniofacial development. High throughput RNA-sequencing analysis reveals several overlapping gene targets of HCFC1 and THAP11. Thus, both HCFC1 and THAP11 play important roles in the regulation of cobalamin metabolism as well as other pathways involved in early vertebrate development.


Assuntos
Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Vitamina B 12/metabolismo , Animais , Sequência de Bases , Região Branquial/metabolismo , Diferenciação Celular , Criança , Anormalidades Craniofaciais/genética , Fibroblastos , Regulação da Expressão Gênica/genética , Fator C1 de Célula Hospedeira/química , Fator C1 de Célula Hospedeira/genética , Fator C1 de Célula Hospedeira/metabolismo , Humanos , Mutação , Cultura Primária de Células , Transcrição Gênica , Vitamina B 12/genética , Peixe-Zebra/genética
13.
J Am Chem Soc ; 140(41): 13205-13208, 2018 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-30282455

RESUMO

A sophisticated intracellular trafficking pathway in humans is used to tailor vitamin B12 into its active cofactor forms, and to deliver it to two known B12-dependent enzymes. Herein, we report an unexpected strategy for cellular retention of B12, an essential and reactive cofactor. If methylmalonyl-CoA mutase is unavailable to accept the coenzyme B12 product of adenosyltransferase, the latter catalyzes homolytic scission of the cobalt-carbon bond in an unconventional reversal of the nucleophilic displacement reaction that was used to make it. The resulting homolysis product binds more tightly to adenosyltransferase than does coenzyme B12, facilitating cofactor retention. We have trapped, and characterized spectroscopically, an intermediate in which the cobalt-carbon bond is weakened prior to being broken. The physiological relevance of this sacrificial catalytic activity for cofactor retention is supported by the significantly lower coenzyme B12 concentration in patients with dysfunctional methylmalonyl-CoA mutase but normal adenosyltransferase activity.


Assuntos
Cobamidas/metabolismo , Alquil e Aril Transferases/química , Alquil e Aril Transferases/metabolismo , Carbono/química , Domínio Catalítico , Cobalto/química , Cobamidas/química , Fibroblastos/metabolismo , Humanos , Metilmalonil-CoA Mutase/metabolismo , Estrutura Molecular
15.
Proc Natl Acad Sci U S A ; 112(2): 400-5, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25548164

RESUMO

An inborn error of metabolism associated with mutations in the human methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) gene has been identified. The proband presented with SCID, megaloblastic anemia, and neurologic abnormalities, but the causal metabolic impairment is unknown. SCID has been associated with impaired purine nucleotide metabolism, whereas megaloblastic anemia has been associated with impaired de novo thymidylate (dTMP) biosynthesis. MTHFD1 functions to condense formate with tetrahydrofolate and serves as the primary entry point of single carbons into folate-dependent one-carbon metabolism in the cytosol. In this study, we examined the impact of MTHFD1 loss of function on folate-dependent purine, dTMP, and methionine biosynthesis in fibroblasts from the proband with MTHFD1 deficiency. The flux of formate incorporation into methionine and dTMP was decreased by 90% and 50%, respectively, whereas formate flux through de novo purine biosynthesis was unaffected. Patient fibroblasts exhibited enriched MTHFD1 in the nucleus, elevated uracil in DNA, lower rates of de novo dTMP synthesis, and increased salvage pathway dTMP biosynthesis relative to control fibroblasts. These results provide evidence that impaired nuclear de novo dTMP biosynthesis can lead to both megaloblastic anemia and SCID in MTHFD1 deficiency.


Assuntos
Metilenotetra-Hidrofolato Desidrogenase (NADP)/deficiência , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Timidina Monofosfato/biossíntese , Substituição de Aminoácidos , Anemia Megaloblástica/genética , Anemia Megaloblástica/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Códon sem Sentido , Dano ao DNA , Fibroblastos/metabolismo , Humanos , Redes e Vias Metabólicas , Metilenotetra-Hidrofolato Desidrogenase (NADP)/química , Antígenos de Histocompatibilidade Menor , Proteínas Mutantes/química , Fenótipo , Mutação Puntual , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/metabolismo
16.
Hum Mutat ; 38(5): 511-516, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28185376

RESUMO

Deleterious variants in the same gene present in two or more families with overlapping clinical features provide convincing evidence of a disease-gene association; this can be a challenge in the study of ultrarare diseases. To facilitate the identification of additional families, several groups have created "matching" platforms. We describe four individuals from three unrelated families "matched" by GeneMatcher and MatchMakerExchange. Individuals had microcephaly, developmental delay, epilepsy, and recessive mutations in TRIT1. A single homozygous mutation in TRIT1 associated with similar features had previously been reported in one family. The identification of these individuals provides additional evidence to support TRIT1 as the disease-causing gene and interprets the variants as "pathogenic." TRIT1 functions to modify mitochondrial tRNAs and is necessary for protein translation. We show that dysfunctional TRIT1 results in decreased levels of select mitochondrial proteins. Our findings confirm the TRIT1 disease association and advance the phenotypic and molecular understanding of this disorder.


Assuntos
Alquil e Aril Transferases/genética , Alelos , Genes Recessivos , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Mutação , Adolescente , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Criança , Pré-Escolar , Fácies , Feminino , Testes Genéticos , Homozigoto , Humanos , Imageamento por Ressonância Magnética , Masculino , Fenótipo
19.
J Med Genet ; 53(10): 705-9, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27330106

RESUMO

BACKGROUND: Osteonecrosis of the femoral head is a debilitating disease that involves impaired blood supply to the femoral head and leads to femoral head collapse. METHODS: We use whole-exome sequencing and Sanger sequencing to analyse a family with inherited osteonecrosis of the femoral head and fluorescent Ca(2+) imaging to functionally characterise the variant protein. RESULTS: We report a family with four siblings affected with inherited osteonecrosis of the femoral head and the identification of a c.2480_2483delCCCG frameshift deletion followed by a c.2486T>A substitution in one allele of the transient receptor potential vanilloid 4 (TRPV4) gene. TRPV4 encodes a Ca(2+)-permeable cation channel known to play a role in vasoregulation and osteoclast differentiation. While pathogenic TRPV4 mutations affect the skeletal or nervous systems, association with osteonecrosis of the femoral head is novel. Functional measurements of Ca(2+) influx through mutant TRPV4 channels in HEK293 cells and patient-derived dermal fibroblasts identified a TRPV4 gain of function. Analysis of channel open times, determined indirectly from measurement of TRPV4 activity within a cluster of TRPV4 channels, revealed that the TRPV4 gain of function was caused by longer channel openings. CONCLUSIONS: These findings identify a novel TRPV4 mutation implicating TRPV4 and altered calcium homeostasis in the pathogenesis of osteonecrosis while reinforcing the importance of TRPV4 in bone diseases and vascular endothelium.


Assuntos
Necrose da Cabeça do Fêmur/metabolismo , Mutação , Canais de Cátion TRPV/genética , Sequência de Aminoácidos , Cálcio/metabolismo , Feminino , Necrose da Cabeça do Fêmur/genética , Humanos , Masculino , Linhagem , Alinhamento de Sequência , Canais de Cátion TRPV/química , Canais de Cátion TRPV/metabolismo , Adulto Jovem
20.
Hum Mutat ; 37(9): 976-82, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27349184

RESUMO

Vitamin B12 (cobalamin, Cbl) cofactors adenosylcobalamin (AdoCbl) and methylcobalamin (MeCbl) are required for the activity of the enzymes methylmalonyl-CoA mutase (MCM) and methionine synthase (MS). Inborn errors of Cbl metabolism are rare Mendelian disorders associated with hematological and neurological manifestations, and elevations of methylmalonic acid and/or homocysteine in the blood and urine. We describe a patient whose fibroblasts had decreased functional activity of MCM and MS and decreased synthesis of AdoCbl and MeCbl (3.4% and 1.0% of cellular Cbl, respectively). The defect in cultured patient fibroblasts complemented those from all known complementation groups. Patient cells accumulated transcobalamin-bound-Cbl, a complex which usually dissociates in the lysosome to release free Cbl. Whole-exome sequencing identified putative disease-causing variants c.851T>G (p.L284*) and c.1019C>T (p.T340I) in transcription factor ZNF143. Proximity biotinylation analysis confirmed the interaction between ZNF143 and HCFC1, a protein that regulates expression of the Cbl trafficking enzyme MMACHC. qRT-PCR analysis revealed low MMACHC expression levels both in patient fibroblasts, and in control fibroblasts incubated with ZNF143 siRNA.


Assuntos
Citoplasma/metabolismo , Erros Inatos do Metabolismo/genética , Transativadores/genética , Transcobalaminas/metabolismo , Vitamina B 12/metabolismo , Proteínas de Transporte/metabolismo , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Lactente , Masculino , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Mutação , Oxirredutases , Linhagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA