Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Circ Res ; 130(10): 1565-1582, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35410483

RESUMO

BACKGROUND: S-adenosylhomocysteine (SAH) is a risk factor of cardiovascular disease; inhibition of SAH hydrolase (SAHH) results in SAH accumulation and induces endothelial dysfunction and atherosclerosis. However, the effect and mechanism of SAHH in atherosclerotic calcification is still unclear. We aimed to explore the role and mechanism of SAHH in atherosclerotic calcification. METHODS: The relationship between SAHH and atherosclerotic calcification was investigated in patients with coronary atherosclerotic calcification. Different in vivo genetic models were used to examine the effect of SAHH deficiency on atherosclerotic calcification. Human aortic and murine vascular smooth muscle cells (VSMCs) were cultured to explore the underlying mechanism of SAHH on osteoblastic differentiation of VSMCs. RESULTS: The expression and activity of SAHH were decreased in calcified human coronary arteries and inversely associated with coronary atherosclerotic calcification severity, whereas plasma SAH and total homocysteine levels were positively associated with coronary atherosclerotic calcification severity. Heterozygote knockout of SAHH promoted atherosclerotic calcification. Specifically, VSMC-deficient but not endothelial cell-deficient or macrophage-deficient SAHH promoted atherosclerotic calcification. Mechanistically, SAHH deficiency accumulated SAH levels and induced H19-mediated Runx2 (runt-related transcription factor 2)-dependent osteoblastic differentiation of VSMCs by inhibiting DNMT3b (DNA methyltransferase 3b) and leading to hypomethylation of the H19 promoter. On the contrary, SAHH deficiency resulted in lower intracellular levels of adenosine and reduced AMPK (AMP-activated protein kinase) activation. Adenosine supplementation activated AMPK and abolished SAHH deficiency-induced expression of H19 and Runx2 and osteoblastic differentiation of VSMCs. Finally, AMPK activation by adenosine inhibited H19 expression by inducing Sirt1 (sirtuin-1)-mediated histone H3 hypoacetylation and DNMT3b-mediated hypermethylation of the H19 promoter in SAHH deficiency VSMCs. CONCLUSIONS: We have confirmed a novel correlation between SAHH deficiency and atherosclerotic calcification and clarified a new mechanism that epigenetic upregulation of H19 and AMPK inhibition concurrently contribute to SAHH deficiency-promoted Runx2-dependent atherosclerotic calcification.


Assuntos
Aterosclerose , Calcinose , Calcificação Vascular , Proteínas Quinases Ativadas por AMP/metabolismo , Adenosina/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos , Animais , Aterosclerose/metabolismo , Calcinose/genética , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Epigênese Genética , Glicina N-Metiltransferase/deficiência , Humanos , Camundongos , Miócitos de Músculo Liso/metabolismo , RNA Longo não Codificante , S-Adenosil-Homocisteína/metabolismo , Regulação para Cima , Calcificação Vascular/genética , Calcificação Vascular/metabolismo
2.
Langmuir ; 38(27): 8252-8265, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35758025

RESUMO

The incorrect metabolic breakdown of the nonaromatic amino acid methionine (Met) leads to the disorder called hypermethioninemia via an unknown mechanism. To understand the molecular level pathogenesis of this disorder, we prepared a DMPC lipid membrane, the mimicking setup of the cell membrane, and explored the effect of the millimolar level of Met on it. We found that Met forms toxic fibrillar aggregates that disrupt the rigidity of the membrane bilayer, and increases the dynamic response of water molecules surrounding the membrane as well as the heterogeneity of the membrane. Such aggregates strongly deform red blood cells. This opens the requirement to consider therapeutic antagonists either to resist or to inhibit the toxic amyloid aggregates against hypermethioninemia. Moreover, such disrupting effect on membrane bilayer and cytotoxicity along with deformation effect on RBC by the cross amyloids of Met and Phenylalanine (Phe) was found to be most virulent. This exclusive observation of the enhanced virulent effect of the cross amyloids is expected to be an informative asset to explain the coexistence of two amyloid disorders.


Assuntos
Aminoácidos , Metionina , Erros Inatos do Metabolismo dos Aminoácidos , Amiloide/química , Glicina N-Metiltransferase/deficiência , Metionina/química , Fenilalanina , Fosfolipídeos
3.
Mol Genet Metab ; 132(1): 38-43, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33309011

RESUMO

Adenosine kinase (ADK) deficiency is characterized by liver disease, dysmorphic features, epilepsy and developmental delay. This defect disrupts the adenosine/AMP futile cycle and interferes with the upstream methionine cycle. We report the clinical, histological and biochemical courses of three ADK children carrying two new mutations and presenting with neonatal cholestasis and neurological disorders. One of them died of liver failure whereas the other two recovered from their liver damage. As the phenotype was consistent with a mitochondrial disorder, we studied liver mitochondrial respiratory chain activities in two patients and revealed a combined defect of several complexes. In addition, we retrospectively analyzed methionine plasma concentration, a hallmark of ADK deficiency, in a cohort of children and showed that methionine level in patients with ADK deficiency was strongly increased compared with patients with other liver diseases. ADK deficiency is a cause of neonatal or early infantile liver disease that may mimic primary mitochondrial disorders. In this context, an elevation of methionine plasma levels over twice the upper limit should not be considered as a nonspecific finding. ADK deficiency induced-liver dysfunction is most often transient, but could be life-threatening.


Assuntos
Adenosina Quinase/genética , Erros Inatos do Metabolismo dos Aminoácidos/genética , Deficiências do Desenvolvimento/genética , Epilepsia/genética , Glicina N-Metiltransferase/deficiência , Adenosina/genética , Adenosina/metabolismo , Adenosina Quinase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Criança , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/patologia , Epilepsia/complicações , Epilepsia/patologia , Feminino , Predisposição Genética para Doença , Glicina N-Metiltransferase/genética , Humanos , Lactente , Recém-Nascido , Hepatopatias/complicações , Hepatopatias/genética , Hepatopatias/patologia , Masculino , Estudos Retrospectivos
4.
J Nutr ; 150(Suppl 1): 2494S-2505S, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33000151

RESUMO

Metabolism of excess methionine (Met) to homocysteine (Hcy) by transmethylation is facilitated by the expression of methionine adenosyltransferase (MAT) I/III and glycine N-methyltransferase (GNMT) in liver, and a lack of either enzyme results in hypermethioninemia despite normal concentrations of MATII and methyltransferases other than GNMT. The further metabolism of Hcy by the transsulfuration pathway is facilitated by activation of cystathionine ß-synthase (CBS) by S-adenosylmethionine (SAM) as well as the relatively high KM of CBS for Hcy. Transmethylation plus transsulfuration effects catabolism of the Met molecule along with transfer of the sulfur atom of Met to serine to synthesize cysteine (Cys). Oxidation and excretion of Met sulfur depend upon Cys catabolism and sulfur oxidation pathways. Excess Cys is oxidized by cysteine dioxygenase 1 (CDO1) and further metabolized to taurine or sulfate. Some Cys is normally metabolized by desulfhydration pathways, and the hydrogen sulfide (H2S) produced is further oxidized to sulfate. If Cys or Hcy concentrations are elevated, Cys or Hcy desulfhydration can result in excess H2S and thiosulfate production. Excess Cys or Met may also promote their limited metabolism by transamination pathways.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Cisteína/metabolismo , Glicina N-Metiltransferase/deficiência , Homocisteína/metabolismo , Fígado/metabolismo , Metionina/metabolismo , Sulfetos/metabolismo , Enxofre/metabolismo , Aminoácidos/metabolismo , Animais , Cistationina beta-Sintase/metabolismo , Glicina N-Metiltransferase/metabolismo , Humanos , Sulfeto de Hidrogênio/metabolismo , S-Adenosilmetionina/metabolismo , Serina/metabolismo , Tiossulfatos/metabolismo
5.
Am J Med Genet A ; 182(4): 740-745, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31957987

RESUMO

S-adenosylhomocysteine hydrolase deficiency is an autosomal recessive neurometabolic disorder affecting the muscles, liver, and nervous system. The disease occurs by pathogenic variants of AHCY gene encoding S-adenosylhomocysteine hydrolase (AHCY) enzyme. This article reports a patient with presumed AHCY deficiency who was diagnosed by whole exome sequencing due to compound heterozygosity of novel p.T57I (c.170C>T) and p.V217M (c.649G>A) variants of AHCY gene. The patient had diffuse edema, coagulopathy, central nervous system abnormalities, and hypotonia. She died in 3 months due to cardiovascular collapse. Clinical findings of the present case were compatible with previously reported AHCY deficiency patients and the novel variants we found are considered to be the cause of the symptoms. This article also compiles the previous reports and expands clinical spectrum of AHCY deficiency by adding new features.


Assuntos
Adenosil-Homocisteinase/genética , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Glicina N-Metiltransferase/deficiência , Mutação , Erros Inatos do Metabolismo dos Aminoácidos/genética , Feminino , Glicina N-Metiltransferase/genética , Humanos , Recém-Nascido , Prognóstico
6.
Amino Acids ; 52(4): 629-638, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32246211

RESUMO

Hypermethioninemia is a disorder characterized by high plasma levels of methionine (Met) and its metabolites such as methionine sulfoxide (MetO). Studies have reported associated inflammatory complications, but the mechanisms involved in the pathophysiology of hypermethioninemia are still uncertain. The present study aims to evaluate the effect of chronic administration of Met and/or MetO on phenotypic characteristics of macrophages, in addition to oxidative stress, purinergic system, and inflammatory mediators in macrophages. In this study, Swiss male mice were subcutaneously injected with Met and MetO at concentrations of 0.35-1.2 g/kg body weight and 0.09-0.3 g/kg body weight, respectively, from the 10th-38th day post-birth, while the control group was treated with saline solution. The results revealed that Met and/or MetO induce an M1/classical activation phenotype associated with increased levels of tumor necrosis factor alpha and nitrite, and reduced arginase activity. It was also found that Met and/or MetO alter the activity of antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase, as well as the levels of thiol and reactive oxygen species in macrophages. The chronic administration of Met and/or MetO also promotes alteration in the hydrolysis of ATP and ADP, as indicated by the increased activity of ectonucleotidases. These results demonstrate that chronic administration of Met and/or MetO promotes activated pro-inflammatory profile by inducing M1/classical macrophage polarization. Thus, the changes in redox status and purinergic system upon chronic Met and/or MetO exposure may contribute towards better understanding of the alterations consistent with hypermethioninemic patients.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/imunologia , Glicina N-Metiltransferase/deficiência , Macrófagos/imunologia , Metionina/análogos & derivados , Animais , Catalase/metabolismo , Polaridade Celular , Glutationa Peroxidase/metabolismo , Glicina N-Metiltransferase/imunologia , Macrófagos/efeitos dos fármacos , Masculino , Metionina/administração & dosagem , Metionina/metabolismo , Metionina/farmacologia , Camundongos , Oxirredução , Estresse Oxidativo , Fenótipo , Superóxido Dismutase/metabolismo
7.
Amino Acids ; 52(3): 371-385, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31902007

RESUMO

The aim of this study was to investigate the effect of the chronic administration of methionine (Met) and/or its metabolite, methionine sulfoxide (MetO), on the behavior and neurochemical parameters of young rats. Rats were treated with saline (control), Met (0.2-0.4 g/kg), MetO (0.05-0.1 g/kg), and/or a combination of Met + MetO, subcutaneously twice a day from postnatal day 6 (P6) to P28. The results showed that Met, MetO, and Met + MetO impaired short-term and spatial memories (P < 0.05), reduced rearing and grooming (P < 0.05), but did not alter locomotor activity (P > 0.05). Acetylcholinesterase activity was increased in the cerebral cortex, hippocampus, and striatum following Met and/or MetO (P < 0.05) treatment, while Na+, K+-ATPase activity was reduced in the hippocampus (P < 0.05). There was an increase in the level of thiobarbituric acid reactive substances (TBARS) in the cerebral cortex in Met-, MetO-, and Met + MetO-treated rats (P < 0.05). Met and/or MetO treatment reduced superoxide dismutase, catalase, and glutathione peroxidase activity, total thiol content, and nitrite levels, and increased reactive oxygen species and TBARS levels in the hippocampus and striatum (P < 0.05). Hippocampal brain-derived neurotrophic factor was reduced by MetO and Met + MetO compared with the control group. The number of NeuN-positive cells was decreased in the CA3 in Met + MetO group and in the dentate gyrus in the Met, MetO, and Met + MetO groups compared to control group (P < 0.05). Taken together, these findings further increase our understanding of changes in the brain in hypermethioninemia by elucidating behavioral alterations, biological mechanisms, and the vulnerability of brain function to high concentrations of Met and MetO.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/complicações , Glicina N-Metiltransferase/deficiência , Hipocampo/patologia , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Metionina/análogos & derivados , Espécies Reativas de Oxigênio/metabolismo , Acetilcolinesterase/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/induzido quimicamente , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Animais , Catalase/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Feminino , Glutationa Peroxidase/deficiência , Glicina N-Metiltransferase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Transtornos da Memória/metabolismo , Memória de Curto Prazo/efeitos dos fármacos , Metionina/metabolismo , Metionina/toxicidade , Ratos , Ratos Wistar , Memória Espacial/efeitos dos fármacos , Superóxido Dismutase/deficiência , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
8.
Amino Acids ; 52(11-12): 1545-1558, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33184691

RESUMO

We investigated the ability of tannic acid (TA) to prevent oxidative and nitrosative damage in the brain, liver, kidney, and serum of a rat model of acute hypermethioninemia. Young Wistar rats were divided into four groups: I (control), II (TA 30 mg/kg), III (methionine (Met) 0.4 g/kg + methionine sulfoxide (MetO) 0.1 g/kg), and IV (TA/Met + MetO). Rats in groups II and IV received TA orally for seven days, and rats of groups I and III received an equal volume of water. After pretreatment with TA, rats from groups II and IV received a single subcutaneous injection of Met + MetO, and were euthanized 3 h afterwards. In specific brain structures and the kidneys, we observed that Met + MetO led to increased reactive oxygen species (ROS), nitrite, and lipid peroxidation levels, followed by a reduction in thiol content and antioxidant enzyme activity. On the other hand, pretreatment with TA prevented both oxidative and nitrosative damage. In the serum, Met + MetO caused a decrease in the activity of antioxidant enzymes, which was again prevented by TA pretreatment. In contrast, in the liver, there was a reduction in ROS levels and an increase in total thiol content, which was accompanied by a reduction in catalase and superoxide dismutase activities in the Met + MetO group, and pretreatment with TA was able to prevent only the reduction in catalase activity. Conclusively, pretreatment with TA has proven effective in preventing oxidative and nitrosative changes caused by the administration of Met + MetO, and may thus represent an adjunctive therapeutic approach for treatment of hypermethioninemia.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Glicina N-Metiltransferase/deficiência , Estresse Nitrosativo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Taninos/farmacologia , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Glutationa Peroxidase/genética , Glicina N-Metiltransferase/metabolismo , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Estresse Nitrosativo/genética , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo , Soro/efeitos dos fármacos , Soro/metabolismo , Superóxido Dismutase/genética
9.
Biosci Biotechnol Biochem ; 84(5): 927-935, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31959065

RESUMO

For many years, clinical studies have suggested that blood levels of l-methionine and L-homocysteine correlate with health status or homocystinuria/hypermethioninemia. l-Methionine in a solution containing 0%, 10%, or 20% human serum was detected in 10-200 µM using l-methionine decarboxylase (MetDC). Spike and recovery tests showed that the enzymatic assay could accurately and reproducibly determine the increases in l-methionine in serum samples. These results suggest that our enzymatic method using MetDC is useful for primary screening of hypermethioninemia or homocystinuria based on serum l-methionine concentration. Additionally, we confirmed that l-methionine (100 nmol) in solution was degraded to less than the detection limit by incubation at 37ºC for 10 min using 2 U of MetDC. Therefore, l-homocysteine in serum samples can be detected with equivalent sensitivity using l-methionine γ-lyase (MGL), in solutions that either did not contain l-methionine or contained l-methionine preincubated with MetDC.Abbreviations: DTT: dithiothreitol; IPTG: isopropyl-ß-d-thiogalactopyranoside; KPB: potassium phosphate buffer; MBTH: 3-methyl-2-benzothiazolinonehydrazone; mdc: the gene coding l-methionine decarboxylase; MetDC: l-methionine decarboxylase; mgl: the gene coding l-methionine γ-lyase; MGL: l-methionine γ-lyase; PLP: pyridoxal 5'-phosphate.


Assuntos
Liases de Carbono-Enxofre/metabolismo , Carboxiliases/metabolismo , Ensaios Enzimáticos/métodos , Homocisteína/sangue , Metionina/sangue , Pseudomonas putida/enzimologia , Streptomyces/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/sangue , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Escherichia coli/genética , Escherichia coli/metabolismo , Glicina N-Metiltransferase/sangue , Glicina N-Metiltransferase/deficiência , Homocistinúria/sangue , Homocistinúria/diagnóstico , Humanos , Plasmídeos/genética , Pseudomonas putida/genética , Espectrofotometria/métodos , Streptomyces/genética
10.
BMC Med Genet ; 20(1): 3, 2019 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-30612563

RESUMO

BACKGROUND: Tandem mass spectrometry (MS MS) and simple fluorometric assays are currently used in newborn screening programs to detect inborn errors of metabolism (IEM). The aim of the study was to evaluate the clinical utility of exome sequencing as a second tier screening method to assist clinical diagnosis of the newborn. METHODS: A novel PCR-exome amplification and re-sequencing (PEARS) assay was designed and used to detect mutations in 122 genes associated with 101 IEM. Newborn bloodspots positive by biochemical testing were analysed by PEARS assay to detect pathogenic mutations relevant to the IEM. RESULTS: In initial validation studies of genomic DNA samples, PEARS assay correctly detected 25 known mutations associated with 17 different IEM. Retrospective gene analysis of newborns with clinical phenylketonuria (PKU), identified compound heterozygote phenylalanine hydroxylase (PAH) gene mutations in eight of nine samples (89%). Prospective analysis of 211 bloodspots correctly identified the two true PKU samples, yielding positive and negative predictive values of 100%. Testing of 8 true positive MS MS samples correctly identified potentially pathogenic compound heterozygote genotypes in 2 cases of citrullinemia type 1 and one case each of methylmalonic acidemia, isobutyryl-CoA dehydrogenase deficiency, short chain acyl-CoA dehydrogenase deficiency and glutaric acid type II and heterozygous genotypes in 2 cases of autosomal dominant methioninemia. Analysis of 11 of 12 false positive MS MS samples for other IEM identified heterozygous carriers in 8 cases for the relevant genes associated with the suspected IEM. In the remaining 3 cases, the test revealed compound heterozygote mutations in other metabolic genes not associated with the suspected IEM, indicating a misinterpretation of the original MS MS data. CONCLUSIONS: The PEARS assay has clinical utility as a rapid and cost effective second-tier test to assist the clinician to accurately diagnose newborns with a suspected IEM.


Assuntos
Sequenciamento do Exoma/métodos , Exoma/genética , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/genética , Reação em Cadeia da Polimerase Multiplex/métodos , Triagem Neonatal/métodos , Acil-CoA Desidrogenase/deficiência , Acil-CoA Desidrogenase/genética , Erros Inatos do Metabolismo dos Aminoácidos/genética , Citrulinemia/genética , Aconselhamento Genético , Genótipo , Glutaratos , Glicina N-Metiltransferase/deficiência , Glicina N-Metiltransferase/genética , Heterozigoto , Humanos , Recém-Nascido , Erros Inatos do Metabolismo Lipídico/genética , Masculino , Técnicas de Diagnóstico Molecular/métodos , Mutação , Fenilalanina Hidroxilase/genética , Fenilcetonúrias/diagnóstico , Fenilcetonúrias/genética , Estudos Prospectivos , Estudos Retrospectivos , Espectrometria de Massas em Tandem/métodos
11.
J Vasc Res ; 56(5): 230-240, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31307051

RESUMO

OBJECTIVE: The relationship between methionine (Met) and abdominal aortic aneurysm (AAA) has been previously demonstrated, but the mechanisms controlling this association remain unclear. This study investigated the potential contribution of hypermethioninemia (HMet) to the development of AAA. METHODS: A model of AAA was induced by intraluminal porcine pancreatic elastase (PPE) infusion in 60 male Sprague-Dawley rats divided into 4 groups (n = 15 per group). Met was supplied by intragastric administration (1 g/kg body weight/day) from 1 week before surgery until 4 weeks after surgery. The aortic diameter was measured by ultrasound. Aortas were collected 4 weeks after surgery and subjected to biochemical analysis, histological assays, and transmission electron microscopy. RESULTS: After 5 weeks of Met supplementation, HMet increased the dilation ratio of the HMet + PPE group, and hyperhomocysteinemia was also induced in HMet and HMet + PPE rats. Increased matrix metalloproteinase-2 (MMP-2), osteopontin, and interleukin-6 expression was detected in HMet + PPE rats. Furthermore, increased autophagy was detected in the HMet + PPE group. CONCLUSION: This study demonstrates that HMet may exacerbate the formation of AAA due to the increased dilation ratio partially via enhancing MMP-2 and inflammatory responses.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/induzido quimicamente , Aneurisma da Aorta Abdominal/induzido quimicamente , Glicina N-Metiltransferase/deficiência , Metionina , Erros Inatos do Metabolismo dos Aminoácidos/sangue , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/ultraestrutura , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Dilatação Patológica , Modelos Animais de Doenças , Progressão da Doença , Glicina N-Metiltransferase/sangue , Interleucina-6/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Osteopontina/metabolismo , Elastase Pancreática , Ratos Sprague-Dawley , Fatores de Risco , Fatores de Tempo
12.
J Inherit Metab Dis ; 42(1): 128-139, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30740731

RESUMO

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.


Assuntos
Homocistinúria/diagnóstico , Acetilcarnitina/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Feminino , Glicina N-Metiltransferase/deficiência , Glicina N-Metiltransferase/metabolismo , Homocisteína/metabolismo , Homocistinúria/metabolismo , Humanos , Recém-Nascido , Masculino , Metionina/metabolismo , Metilenotetra-Hidrofolato Redutase (NADPH2)/deficiência , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Ácido Metilmalônico/metabolismo , Espasticidade Muscular/diagnóstico , Espasticidade Muscular/metabolismo , Triagem Neonatal/métodos , Fenilalanina/metabolismo , Transtornos Psicóticos/diagnóstico , Transtornos Psicóticos/metabolismo
13.
Amino Acids ; 49(1): 129-138, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27718024

RESUMO

High levels of methionine (Met) and methionine sulfoxide (MetO) are found in several genetic abnormalities. Oxidative stress is involved in the pathophysiology of many inborn errors of metabolism. However, little is known about the role of oxidative damage in hepatic and renal changes in hypermethioninemia. We investigated the effect of chronic treatment with Met and/or MetO on oxidative stress parameters in liver and kidney, as lipid peroxidation (TBARS), total sulfhydryl content (SH), reactive oxygen species (ROS) and enzymes activities superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and delta aminolevulinic dehydratase (ALA-D). Serum biochemical parameters were evaluated. Wistar rats were treated daily with two subcutaneous injections of saline (control), Met (0.2-0.4 g/kg), MetO (0.05-0.1 g/kg) and the association between these (Met plus MetO) from the 6th to the 28th day of life. Our data demonstrated an increase of glucose and urea levels in all experimental groups. Cholesterol (MetO and Met plus MetO) were decreased and triglycerides (MetO) were increased. SOD (MetO and Met plus MetO) and CAT (Met, MetO and Met plus MetO) activities were decreased, while GPx was enhanced by MetO and Met plus MetO treatment in liver. In kidney, we observed a reduction of SH levels, SOD and CAT activities and an increase of TBARS levels in all experimental groups. ROS levels in kidney were increased in MetO and Met plus MetO groups. ALA-D activity was enhanced in liver (MetO and Met plus MetO) and kidney (Met plus MetO). These findings help to understand the pathophysiology of hepatic and renal alterations present in hypermethioninemia.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Glicina N-Metiltransferase/deficiência , Metionina/análogos & derivados , Metionina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Sintase do Porfobilinogênio/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/induzido quimicamente , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Animais , Catalase/metabolismo , Colesterol/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Glucose/metabolismo , Glutationa Peroxidase/metabolismo , Glicina N-Metiltransferase/metabolismo , Injeções Subcutâneas , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Peroxidação de Lipídeos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Metionina/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Compostos de Sulfidrila/metabolismo , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Triglicerídeos/metabolismo , Ureia/metabolismo
14.
J Inherit Metab Dis ; 40(4): 491-495, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28168361

RESUMO

The indications and outcomes of liver transplantation for metabolic disease have been reviewed recently and this short review concentrates on recent developments and advances. Recently recognized metabolic causes of acute liver failure are reviewed and their implications for transplantation discussed. Newly described indications for liver transplantation in systemic metabolic diseases are described and an update is given on the role of auxiliary and domino liver transplantation.


Assuntos
Falência Hepática Aguda/cirurgia , Transplante de Fígado/tendências , Doenças Metabólicas/cirurgia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/cirurgia , Animais , Encefalopatias Metabólicas Congênitas/genética , Encefalopatias Metabólicas Congênitas/cirurgia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/cirurgia , Terapia de Reposição de Enzimas , Epífises/anormalidades , Epífises/cirurgia , Glicina N-Metiltransferase/deficiência , Glicina N-Metiltransferase/genética , Humanos , Pseudo-Obstrução Intestinal/genética , Pseudo-Obstrução Intestinal/cirurgia , Neoplasias Hepáticas/cirurgia , Encefalomiopatias Mitocondriais/genética , Encefalomiopatias Mitocondriais/cirurgia , Distrofia Muscular Oculofaríngea , Proteínas de Neoplasias/genética , Oftalmoplegia/congênito , Osteocondrodisplasias/genética , Osteocondrodisplasias/cirurgia , Púrpura/genética , Púrpura/cirurgia , Doença de Refsum Infantil/genética , Doença de Refsum Infantil/cirurgia
15.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 34(1): 98-101, 2017 Feb 10.
Artigo em Zh | MEDLINE | ID: mdl-28186605

RESUMO

OBJECTIVE: To detect potential mutations of MAT1A gene in a child suspected with simple hypermethioninemia by MS/MS neonatal screening. METHODS: Clinical data of the child was collected. Genomic DNA was extracted by a standard method and subjected to targeted sequencing using an Ion AmpliseqTM Inherited Disease Panel. Detected mutations were verified by Sanger sequencing. RESULTS: The child showed no clinical features except evaluated methionine. A novel compound mutation of the MAT1A gene, i.e., c.345delA and c.529C>T, was identified in the child. His father and mother were found to be heterozygous for the c.345delA mutation and c.529C>T mutation, respectively. CONCLUSION: The compound mutation c.345delA and c.529C>T of the MAT1A gene probably underlie the disease in the child. The semi-conductor sequencing has provided an important means for the diagnosis of hereditary diseases.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Predisposição Genética para Doença/genética , Glicina N-Metiltransferase/deficiência , Doenças do Recém-Nascido/genética , Metionina Adenosiltransferase/genética , Mutação , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Sequência de Bases , Análise Mutacional de DNA/métodos , Saúde da Família , Pai , Feminino , Glicina N-Metiltransferase/genética , Heterozigoto , Humanos , Recém-Nascido , Doenças do Recém-Nascido/patologia , Masculino , Mães
16.
Mol Genet Metab ; 118(3): 143-144, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27177696

RESUMO

Harvey Mudd was the father of methionine metabolic disorders. Beginning with his identification of the enzyme defect in homocystinuria, he co-discovered cobalamin C disorder as the first known human disorder of vitamin B12 metabolism, thereby extending our concept of homocystinuria as a key feature of related disorders rather than a single disease, and identified new disorders that produce hypermethioninemia. He had no equal in our understanding of how critical methionine metabolism is to human homeostasis.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Glicina N-Metiltransferase/deficiência , Homocistinúria/diagnóstico , Metionina/metabolismo , Deficiência de Vitamina B 12/congênito , Pesquisa Biomédica , História do Século XX , História do Século XXI , Humanos , Masculino , Deficiência de Vitamina B 12/diagnóstico
17.
J Biomed Sci ; 23(1): 69, 2016 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-27716281

RESUMO

BACKGROUND: Glycine N-methyltransferase (GNMT) is abundantly expressed in the normal liver but is down-regulated in liver cancer tissues. GNMT knockout (Gnmt-/-) mice can spontaneously develop chronic hepatitis, fatty liver, and liver cancer. We previously demonstrated that hepatic GNMT is decreased in high-fat-diet-induced type 2 diabetes mellitus, but its contribution to metabolic syndrome is unclear. Here we show that GNMT modulates key aspects of metabolic syndrome in mice. METHODS: Eleven-week-old Gnmt-/- and wild-type (WT) mice with a C57BL/6 genetic background were used in this study. The metabolic defects of GNMT deficiency were measured by glucose and insulin tolerance tests, lipid homeostasis, gluconeogenesis, and insulin signaling. RESULTS: Gnmt-/- mice, especially females, exhibited glucose intolerance and insulin resistance. However, their body fat and lean mass, food and water intakes, and energy expenditure did not differ from those of WT mice. In addition, glucose-stimulated insulin secretion and insulin-stimulated glucagon secretion were normal in the serum and pancreatic islets of Gnmt-/- mice. Importantly, we found that GNMT deficiency increased lipogenesis and triglycerides in the liver. The elevated triglycerides disrupted the ability of insulin to induce Akt and S6 ribosomal protein phosphorylation, and then triggered insulin resistance and gluconeogenesis in female Gnmt-/- mice. CONCLUSIONS: Our data indicate that hepatic GNMT regulates lipid and glucose homeostasis, and provide insight into the development of insulin resistance through modulating the PI3K/Akt pathway.


Assuntos
Gluconeogênese , Glicina N-Metiltransferase/deficiência , Glicina N-Metiltransferase/genética , Insulina/metabolismo , Fígado/enzimologia , Síndrome Metabólica/genética , Transdução de Sinais , Animais , Feminino , Síndrome Metabólica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
18.
Amino Acids ; 48(11): 2479-2489, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27465642

RESUMO

Hypermethioninemia is a condition defined as elevated plasma methionine levels and may be a consequence of different conditions that include non-genetic and genetic causes. In severe cases, hypermethioninemia may lead to development of neurological and hepatic impairments, but mechanisms are still not well elucidated. Therefore, this review aims to reunite the knowledge acquired about the methionine-induced brain and liver toxicity focusing on the results obtained by studies from patients, in vitro experiments, and in vivo animal models. In general, some studies have shown that methionine decreases Na+,K+-ATPase activity, induces oxidative stress, increases acetylcholinesterase activity, and leads to dendritic spine downregulation in brain. Concerning to liver, hypermethioninemia seems to provoke changes in cell morphology, lipid accumulation, oxidative stress, inflammation, and ATP depletion. It is possible to infer that oxidative damage is one of the most important mechanisms responsible for methionine toxicity, since different studies showed that this amino acid induces oxidative stress in brain and liver tissues. Besides, reactive oxygen species may mediate other alterations induced by methionine, such as the reduction in brain Na+,K+-ATPase activity, and liver inflammation.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encéfalo/metabolismo , Glicina N-Metiltransferase/deficiência , Hepatopatias/metabolismo , Fígado/metabolismo , Doenças do Sistema Nervoso/metabolismo , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Animais , Encéfalo/patologia , Glicina N-Metiltransferase/genética , Glicina N-Metiltransferase/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Metabolismo dos Lipídeos/genética , Fígado/patologia , Hepatopatias/etiologia , Hepatopatias/genética , Hepatopatias/patologia , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/patologia , Estresse Oxidativo/genética , Espécies Reativas de Oxigênio/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo
19.
Lab Invest ; 95(2): 223-36, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25531568

RESUMO

Glycine-N-methyltransferase (GNMT) is essential to preserve liver homeostasis. Cirrhotic patients show low expression of GNMT that is absent in hepatocellular carcinoma (HCC) samples. Accordingly, GNMT deficiency in mice leads to steatohepatitis, fibrosis, cirrhosis, and HCC. Lack of GNMT triggers NK cell activation in GNMT(-/-) mice and depletion of TRAIL significantly attenuates acute liver injury and inflammation in these animals. Chronic inflammation leads to fibrogenesis, further contributing to the progression of chronic liver injury regardless of the etiology. The aim of our study is to elucidate the implication of TRAIL-producing NK cells in the progression of chronic liver injury and fibrogenesis. For this we generated double TRAIL(-/-)/GNMT(-/-) mice in which we found that TRAIL deficiency efficiently protected the liver against chronic liver injury and fibrogenesis in the context of GNMT deficiency. Next, to better delineate the implication of TRAIL-producing NK cells during fibrogenesis we performed bile duct ligation (BDL) to GNMT(-/-) and TRAIL(-/-)/GNMT(-/-) mice. In GNMT(-/-) mice, exacerbated fibrogenic response after BDL concurred with NK1.1(+) cell activation. Importantly, specific inhibition of TRAIL-producing NK cells efficiently protected GNMT(-/-) mice from BDL-induced liver injury and fibrogenesis. Finally, TRAIL(-/-)/GNMT(-/-) mice showed significantly less fibrosis after BDL than GNMT(-/-) mice further underlining the relevance of the TRAIL/DR5 axis in mediating liver injury and fibrogenesis in GNMT(-/-) mice. Finally, in vivo silencing of DR5 efficiently protected GNMT(-/-) mice from BDL-liver injury and fibrogenesis, overall underscoring the key role of the TRAIL/DR5 axis in promoting fibrogenesis in the context of absence of GNMT. Overall, our work demonstrates that TRAIL-producing NK cells actively contribute to liver injury and further fibrogenesis in the pathological context of GNMT deficiency, a molecular scenario characteristic of chronic human liver disease.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/imunologia , Doença Hepática Terminal/etiologia , Doença Hepática Terminal/patologia , Glicina N-Metiltransferase/deficiência , Células Matadoras Naturais/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Ductos Biliares/cirurgia , Western Blotting , Citometria de Fluxo , Glicina N-Metiltransferase/imunologia , Humanos , Imuno-Histoquímica , Ligadura , Camundongos , Camundongos Knockout , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
20.
J Hepatol ; 62(3): 673-81, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25457203

RESUMO

BACKGROUND & AIMS: Very-low-density lipoproteins (VLDLs) export lipids from the liver to peripheral tissues and are the precursors of low-density-lipoproteins. Low levels of hepatic S-adenosylmethionine (SAMe) decrease triglyceride (TG) secretion in VLDLs, contributing to hepatosteatosis in methionine adenosyltransferase 1A knockout mice but nothing is known about the effect of SAMe on the circulating VLDL metabolism. We wanted to investigate whether excess SAMe could disrupt VLDL plasma metabolism and unravel the mechanisms involved. METHODS: Glycine N-methyltransferase (GNMT) knockout (KO) mice, GNMT and perilipin-2 (PLIN2) double KO (GNMT-PLIN2-KO) and their respective wild type (WT) controls were used. A high fat diet (HFD) or a methionine deficient diet (MDD) was administrated to exacerbate or recover VLDL metabolism, respectively. Finally, 33 patients with non-alcoholic fatty-liver disease (NAFLD); 11 with hypertriglyceridemia and 22 with normal lipidemia were used in this study. RESULTS: We found that excess SAMe increases the turnover of hepatic TG stores for secretion in VLDL in GNMT-KO mice, a model of NAFLD with high SAMe levels. The disrupted VLDL assembly resulted in the secretion of enlarged, phosphatidylethanolamine-poor, TG- and apoE-enriched VLDL-particles; special features that lead to increased VLDL clearance and decreased serum TG levels. Re-establishing normal SAMe levels restored VLDL secretion, features and metabolism. In NAFLD patients, serum TG levels were lower when hepatic GNMT-protein expression was decreased. CONCLUSIONS: Excess hepatic SAMe levels disrupt VLDL assembly and features and increase circulating VLDL clearance, which will cause increased VLDL-lipid supply to tissues and might contribute to the extrahepatic complications of NAFLD.


Assuntos
Lipoproteínas VLDL/sangue , Hepatopatia Gordurosa não Alcoólica/metabolismo , S-Adenosilmetionina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Glicina N-Metiltransferase/deficiência , Glicina N-Metiltransferase/genética , Glicina N-Metiltransferase/metabolismo , Humanos , Lipoproteínas VLDL/metabolismo , Fígado/metabolismo , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Modelos Biológicos , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Perilipina-2 , S-Adenosilmetionina/deficiência , Triglicerídeos/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA