Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 38(13): e23795, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38984928

RESUMO

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


Assuntos
Homocistinúria , Fígado , Oxirredução , Tetra-Hidrofolato Desidrogenase , Tetra-Hidrofolatos , Animais , Homocistinúria/metabolismo , Homocistinúria/tratamento farmacológico , Homocistinúria/genética , Camundongos , Tetra-Hidrofolatos/metabolismo , Fígado/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Betaína/metabolismo , Betaína/farmacologia , Homocisteína/metabolismo , Camundongos Endogâmicos C57BL , Cistationina beta-Sintase/metabolismo , Cistationina beta-Sintase/genética , Carbono/metabolismo , Masculino , Ácido Fólico/metabolismo , Feminino
2.
Mol Genet Metab ; 132(2): 128-138, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33483253

RESUMO

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


Assuntos
Cistationina beta-Sintase/genética , Homocistinúria/metabolismo , Fígado/metabolismo , Metionina/metabolismo , Adenosil-Homocisteinase/genética , Animais , Betaína/farmacologia , Cistationina beta-Sintase/metabolismo , Modelos Animais de Doenças , Ácido Fólico/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glicina Hidroximetiltransferase/genética , Homocisteína/sangue , Homocisteína/metabolismo , Homocistinúria/tratamento farmacológico , Homocistinúria/genética , Homocistinúria/patologia , Humanos , Fígado/enzimologia , Metionina/análogos & derivados , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Camundongos , Poliaminas/metabolismo
3.
J Nutr ; 151(10): 2882-2893, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34383924

RESUMO

BACKGROUND: Adequate cellular thymidylate (dTMP) pools are essential for preservation of nuclear and mitochondrial genome stability. Previous studies have indicated that disruption in nuclear dTMP synthesis leads to increased uracil misincorporation into DNA, affecting genome stability. To date, the effects of impaired mitochondrial dTMP synthesis in nontransformed tissues have been understudied. OBJECTIVES: This study aimed to determine the effects of decreased serine hydroxymethyltransferase 2 (Shmt2) expression and dietary folate deficiency on mitochondrial DNA (mtDNA) integrity and mitochondrial function in mouse tissues. METHODS: Liver mtDNA content, and uracil content in liver mtDNA, were measured in Shmt2+/- and Shmt2+/+ mice weaned onto either a folate-sufficient control diet (2 mg/kg folic acid; C) or a modified diet lacking folic acid (0 mg/kg folic acid) for 7 wk. Shmt2+/- and Shmt2+/+ mouse embryonic fibroblast (MEF) cells were cultured in defined culture medium containing either 0 or 25 nM folate (6S-5-formyl-tetrahydrofolate, folinate) to assess proliferative capacity and mitochondrial function. Chi-square tests, linear mixed models, and 2-factor ANOVA with Tukey post hoc analyses were used to analyze data. RESULTS: Shmt2 +/- mice exhibited a 48%-67% reduction in SHMT2 protein concentrations in tissues. Interestingly, Shmt2+/- mice consuming the folate-sufficient C diet exhibited a 25% reduction in total folate in liver mitochondria. There was also a >20-fold increase in uracil in liver mtDNA in Shmt2+/- mice consuming the C diet, and dietary folate deficiency also increased uracil content in mouse liver mtDNA from both Shmt2+/+ and Shmt2+/- mice. Furthermore, decreased Shmt2 expression in MEF cells reduced cell proliferation, mitochondrial membrane potential, and oxygen consumption rate. CONCLUSIONS: This study demonstrates that Shmt2 heterozygosity and dietary folate deficiency impair mitochondrial dTMP synthesis in mice, as evidenced by the increased uracil in mtDNA. In addition, Shmt2 heterozygosity impairs mitochondrial function in MEF cells. These findings suggest that elevated uracil in mtDNA may impair mitochondrial function.


Assuntos
Deficiência de Ácido Fólico , Ácido Fólico , Animais , DNA Mitocondrial/genética , Fibroblastos , Camundongos , Mitocôndrias , Respiração , Uracila
4.
J Nutr ; 150(Suppl 1): 2532S-2537S, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33000156

RESUMO

Homocysteine (Hcy) is methylated by methionine synthase to form methionine with methyl-cobalamin as a cofactor. The reaction demethylates 5-methyltetrahydrofolate to tetrahydrofolate, which is required for DNA and RNA synthesis. Deficiency of either of the cobalamin (Cbl) and/or folate cofactors results in elevated Hcy and megaloblastic anemia. Elevated Hcy is a sensitive biomarker of Cbl and/or folate status and more specific than serum vitamin assays. Elevated Hcy normalizes when the correct vitamin is given. Elevated Hcy is associated with alcohol use disorder and drugs that target folate or Cbl metabolism, and is a risk factor for thrombotic vascular disease. Elevated methionine and cystathionine are associated with liver disease. Elevated Hcy, cystathionine, and cysteine, but not methionine, are common in patients with chronic renal failure. Higher cysteine predicts obesity and future weight gain. Serum S-adenosylhomocysteine (AdoHcy) is elevated in Cbl deficiency and chronic renal failure. Drugs that require methylation for catabolism may deplete liver S-adenosylmethionine and raise AdoHcy and Hcy. Deficiency of Cbl or folate or perturbations of their metabolism cause major changes in sulfur amino acids.


Assuntos
Aminoácidos Sulfúricos/metabolismo , Deficiência de Ácido Fólico/complicações , Ácido Fólico/sangue , Hiper-Homocisteinemia/sangue , Estado Nutricional , Deficiência de Vitamina B 12/complicações , Vitamina B 12/sangue , Alcoolismo/sangue , Aminoácidos Sulfúricos/sangue , Anemia Megaloblástica/sangue , Biomarcadores/sangue , Doenças Cardiovasculares/sangue , Deficiência de Ácido Fólico/sangue , Humanos , Hiper-Homocisteinemia/complicações , Falência Renal Crônica/sangue , Hepatopatias/sangue , Obesidade/sangue , S-Adenosil-Homocisteína/sangue , Deficiência de Vitamina B 12/sangue
5.
J Nutr ; 150(7): 1705-1712, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32271909

RESUMO

BACKGROUND: Neural tube defects (NTDs) occur in nervous tissue during embryogenesis when the neural tube fails to close. Approximately 70% of all human NTDs can be prevented by folic acid (FA). Altered expression and/or function of the tumor suppressor protein p53 can lead to NTDs in mouse models. OBJECTIVES: The aim of this study was to determine if dietary FA could rescue p53-/--induced NTDs in mice, and to determine the effect loss of p53 has on pathways in folate 1-carbon metabolism. METHODS: p53+/- female mice were randomly allocated and weaned onto either an FA-sufficient diet (2 mg/kg folic acid; +FA), or an FA-deficient diet (-FA). After 8 wk, the females were time-mated to p53-/- males. Embryos were examined at E12.5 for NTDs. Folate enzyme concentrations, nucleotide synthesis, uracil accumulation in DNA, and proliferation were measured in primary murine embryonic fibroblasts (MEFs). The "n - 1" chi-square test was used to compare NTD percentages, whereas all other data were analyzed by Student t test, except where noted a multilevel-fit model was used. RESULTS: NTD rates of litters from dams consuming the +FA diet (20/46; 43%) did not differ from those of litters from dams consuming the -FA diet (14/35; 40%) (P > 0.05). p53-/- MEFs had 55% higher rates of folate-dependent de novo dTMP synthesis, a ∼2-fold higher accumulation of uracil in DNA, and a ∼30% higher rate of proliferation (P ≤ 0.05) than p53+/- MEFs independent of folate. CONCLUSIONS: p53-related NTDs are not FA responsive. Increased dTMP synthesis in p53-/- MEFs might not have been sufficient to meet the demands for thymidine triphosphate (dTTP) synthesis as evidenced by the elevated amounts of uracil in DNA. This study provides additional evidence that elevated uracil in DNA is a risk factor for NTDs.


Assuntos
DNA/química , Ácido Fólico/farmacologia , Defeitos do Tubo Neural/genética , Proteína Supressora de Tumor p53 , Uracila/metabolismo , Animais , DNA/metabolismo , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Camundongos Knockout , Complexo Vitamínico B/farmacologia
6.
FASEB J ; 33(5): 6339-6353, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30768359

RESUMO

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


Assuntos
Betaína-Homocisteína S-Metiltransferase/metabolismo , Betaína/farmacologia , Homocistinúria , Fígado/enzimologia , Taurina/farmacologia , Animais , Betaína-Homocisteína S-Metiltransferase/genética , Modelos Animais de Doenças , Homocistinúria/tratamento farmacológico , Homocistinúria/genética , Homocistinúria/metabolismo , Homocistinúria/patologia , Humanos , Fígado/patologia , Camundongos , Camundongos Knockout
7.
Am J Perinatol ; 37(11): 1084-1093, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32120425

RESUMO

OBJECTIVE: Fetuses measuring below the 10th percentile for gestational age may be either constitutionally small for gestational age (SGA) or have pathologic fetal growth restriction (FGR). FGR is associated with adverse outcomes; however, identification of low-risk SGA cases is difficult. We performed a pilot study evaluating maternal markers of pathologic FGR, hypothesizing there are distinct amino acid signatures that might be used for diagnosis and development of new interventions. STUDY DESIGN: This was a cohort study of healthy women with sonographic fetal estimated fetal weight <5th percentile divided into two groups based upon umbilical artery (UmA) Doppler studies or uterine artery (UtA) Doppler studies. We collected maternal blood samples prior to delivery and used ion pair reverse phase liquid chromatography-mass spectrometry or gas chromatography-mass spectrometry to assess 44 amino acids. RESULTS: Among 14 women included, five had abnormal UmA, and three had abnormal UtA Doppler results. Those with abnormal UmA showed elevated ornithine. Those with abnormal UtA had lower dimethylglycine, isoleucine, methionine, phenylalanine, and 1-methylhistidine. CONCLUSION: We found several amino acids that might identify pregnancies affected by pathologic FGR. These findings support the feasibility of future larger studies to identify maternal metabolic approaches to accurately stratify risk for small fetuses.


Assuntos
Aminoácidos/sangue , Retardo do Crescimento Fetal/diagnóstico , Artérias Umbilicais/diagnóstico por imagem , Artéria Uterina/diagnóstico por imagem , Adulto , Estudos de Coortes , Feminino , Retardo do Crescimento Fetal/sangue , Retardo do Crescimento Fetal/diagnóstico por imagem , Cromatografia Gasosa-Espectrometria de Massas , Idade Gestacional , Humanos , Recém-Nascido , Recém-Nascido Pequeno para a Idade Gestacional , Projetos Piloto , Gravidez , Resultado da Gravidez , Terceiro Trimestre da Gravidez , Ultrassonografia Doppler , Ultrassonografia Pré-Natal , Adulto Jovem
8.
FASEB J ; 32(3): 1265-1280, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29101223

RESUMO

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


Assuntos
Aminobutiratos/metabolismo , Homocistinúria/metabolismo , Fígado/metabolismo , Aldeído Pirúvico/metabolismo , Compostos de Sulfidrila/metabolismo , Ácidos Sulfínicos/metabolismo , Taurina/farmacologia , Aminoácidos/metabolismo , Animais , Cistationina beta-Sintase/metabolismo , Modelos Animais de Doenças , Feminino , Homocistinúria/tratamento farmacológico , Homocistinúria/patologia , Fígado/efeitos dos fármacos , Masculino , Metaboloma , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , gama-Glutamiltransferase/metabolismo
9.
J Inherit Metab Dis ; 42(3): 424-437, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30873612

RESUMO

STUDY OBJECTIVE: A phase 1/2 clinical trial was performed in individuals with cystathionine ß synthase (CBS) deficient homocystinuria with aims to: (a) assess pharmacokinetics and safety of taurine therapy, (b) evaluate oxidative stress, inflammation, and vascular function in CBS deficiency, and (c) evaluate the impact of short-term taurine treatment. METHODS: Individuals with pyridoxine-nonresponsive CBS deficiency with homocysteine >50 µM, without inflammatory disorder or on antioxidant therapy were enrolled. Biomarkers of oxidative stress and inflammation, endothelial function (brachial artery flow-mediated dilation [FMD]), and disease-related metabolites obtained at baseline were compared to normal values. While maintaining current treatment, patients were treated with 75 mg/kg taurine twice daily, and treatment response assessed after 4 hours and 4 days. RESULTS: Fourteen patients (8-35 years; 8 males, 6 females) were enrolled with baseline homocysteine levels 161 ± 67 µM. The study found high-dose taurine to be safe when excluding preexisting hypertriglyceridemia. Taurine pharmacokinetics showed a rapid peak level returning to near normal levels at 12 hours, but had slow accumulation and elevated predosing levels after 4 days of treatment. Only a single parameter of oxidative stress, 2,3-dinor-8-isoprostaglandin-F2α, was elevated at baseline, with no elevated inflammatory parameters, and no change in FMD values overall. Taurine had no effect on any of these parameters. However, the effect of taurine was strongly related to pretreatment FMD values; and taurine significantly improved FMD in the subset of individuals with pretreatment FMD values <10% and in individuals with homocysteine levels >125 µM, pertinent to endothelial function. CONCLUSION: Taurine improves endothelial function in CBS-deficient homocystinuria in patients with preexisting reduced function.


Assuntos
Biomarcadores/metabolismo , Cistationina beta-Sintase/metabolismo , Homocistinúria/tratamento farmacológico , Taurina/farmacocinética , Taurina/uso terapêutico , Adolescente , Adulto , Artéria Braquial/efeitos dos fármacos , Criança , Cistationina beta-Sintase/deficiência , Feminino , Homocisteína/metabolismo , Homocistinúria/genética , Humanos , Inflamação/tratamento farmacológico , Masculino , Estresse Oxidativo/efeitos dos fármacos , Estados Unidos , Adulto Jovem
10.
J Nutr ; 148(3): 389-400, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29546304

RESUMO

Background: Epidemiologic and in vitro studies suggest independent linkages between poor folate and/or vitamin B-12 nutrition, genomic human papillomavirus (HPV) type 16 viral integration, and cancer. However, there is no direct evidence in vivo to support the causative role of poor folate nutrition in HPV16 integration into the cellular genome. Objective: We tested the hypothesis that folate deficiency enables the integration of HPV16 into the genome of HPV16-harboring keratinocytes, and could thereby influence earlier transformation of these cells to cancer in an animal model. Methods: HPV16-harboring human keratinocytes [(HPV16)BC-1-Ep/SL] were differentiated into 3-dimensional HPV16-organotypic rafts under either folate-replete or folate-deficient conditions in vitro. These were then subcutaneously implanted in severely immunocompromised female Beige Nude XID (Hsd: NIHS-LystbgFoxn1nuBtkxid) mice (4-6 wk old, 16-18 g) fed either a folate-replete diet (1200 nmol folate/kg diet) or a progressively folate-deficient diet (600 or 400 nmol folate/kg diet) for 2 mo prior to raft-implantation surgery, and indefinitely thereafter. The tumors that subsequently developed were characterized for onset, pattern of growth, morphology, HPV16 oncogene expression, and HPV16-genomic integration. Results: All HPV16-organotypic rafts developed in either folate-replete or physiologic low-folate media in vitro and subsequently implanted in folate-replete mice eventually transformed into aggressive malignancies within weeks. When compared to HPV16-high folate-organotypic raft-derived tumors from mice fed either a 1200 or 600 nmol folate/kg diet, those raft-derived cancers that developed in mice fed a 400 nmol folate/kg diet expressed significantly more HPV16 E6 (1.8-fold more) and E7 (2.8-fold more) oncogenic proteins (P = 0.001), and revealed significantly more HPV16-integration sites in genomic DNA (2-fold more), either directly into, or in the vicinity of, cellular genes (P < 0.05). Conclusions: This unprecedented animal model for the consistent rapid transformation of differentiated (HPV16)BC-1-Ep/SL-derived organotypic raft-keratinocytes to cancer in Beige Nude XID mice confirms that dietary folate deficiency can profoundly influence and modulate events leading to HPV16-induced carcinogenesis, and facilitates genomic integration of HPV16 DNA in vivo.


Assuntos
Carcinogênese/genética , Deficiência de Ácido Fólico/complicações , Ácido Fólico/administração & dosagem , Genoma , Papillomavirus Humano 16/genética , Neoplasias/etiologia , Integração Viral , Animais , DNA , Modelos Animais de Doenças , Feminino , Humanos , Queratinócitos/virologia , Camundongos Nus , Neoplasias/genética , Neoplasias/virologia , Estado Nutricional , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecções por Papillomavirus/etiologia , Infecções por Papillomavirus/virologia , Proteínas Repressoras/metabolismo , Neoplasias do Colo do Útero
11.
Mol Genet Metab ; 120(4): 325-336, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28291718

RESUMO

Classical homocystinuria (HCU) due to inactivating mutation of cystathionine ß-synthase (CBS) is a poorly understood life-threatening inborn error of sulfur metabolism. A previously described cbs-/- mouse model exhibits a semi-lethal phenotype due to neonatal liver failure. The transgenic HO mouse model of HCU exhibits only mild liver injury and recapitulates multiple aspects of the disease as it occurs in humans. Disruption of the methionine cycle in HCU has the potential to impact multiple aspect of phospholipid (PL) metabolism by disruption of both the Kennedy pathway and phosphatidylethanolamine N-methyltransferase (PEMT) mediated synthesis of phosphatidylcholine (PC). Comparative metabolomic analysis of HO mouse liver revealed decreased levels of choline, and choline phosphate indicating disruption of the Kennedy pathway. Alterations in the relative levels of multiple species of PL included significant increases in PL degradation products consistent with enhanced membrane PL turnover. A significant decrease in PC containing 20:4n6 which primarily formed by the methylation of phosphatidylethanolamine to PC was consistent with decreased flux through PEMT. Hepatic expression of PEMT in both the cbs-/- and HO models is post-translationally repressed with decreased levels of PEMT protein and activity that inversely-correlates with the scale of liver injury. Failure to induce further repression of PEMT in HO mice by increased homocysteine, methionine and S-adenosylhomocysteine or depletion of glutathione combined with examination of multiple homocysteine-independent models of liver injury indicated that repression of PEMT in HCU is a consequence rather than a cause of liver injury. Collectively, our data show significant alteration of a broad range of hepatic PL and choline metabolism in HCU with the potential to contribute to multiple aspects of pathogenesis in this disease.


Assuntos
Colina/metabolismo , Homocistinúria/enzimologia , Fígado/química , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Fosfolipídeos/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Fígado/lesões , Metabolômica , Camundongos , Camundongos Knockout , Fosfatidiletanolamina N-Metiltransferase/genética , Processamento de Proteína Pós-Traducional
12.
Mol Genet Metab ; 122(4): 160-171, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29153845

RESUMO

Vitamin B12 (cobalamin, Cbl) is a micronutrient essential to human health. Cbl is not utilized as is but must go through complex subcellular and metabolic processing to generate two cofactor forms: methyl-Cbl for methionine synthase, a cytosolic enzyme; and adenosyl-Cbl for methylmalonyl-CoA mutase, a mitochondrial enzyme. Some 10-12 human genes have been identified responsible for the intracellular conversion of Cbl to cofactor forms, including genes that code for ATP-binding cassette (ABC) transporters acting at the lysosomal and plasma membranes. However, the gene for mitochondrial uptake is not known. We hypothesized that ABC transporters should be candidates for other uptake and efflux functions, including mitochondrial transport, and set out to screen ABC transporter mutants for blocks in Cbl utilization using the nematode roundworm Caenorhabditis elegans. Thirty-seven mutant ABC transporters were screened for the excretion of methylmalonic acid (MMA), which should result from loss of Cbl transport into the mitochondria. One mutant, wht-6, showed elevated MMA excretion and reduced [14C]-propionate incorporation, pointing to a functional block in methylmalonyl-CoA mutase. In contrast, the wht-6 mutant appeared to have a normal cytosolic pathway based on analysis of cystathionine excretion, suggesting that cytosolic methionine synthase was functioning properly. Further, the MMA excretion in wht-6 could be partially reversed by including vitamin B12 in the assay medium. The human ortholog of wht-6 is a member of the G family of ABC transporters. We propose wht-6 as a candidate for the transport of Cbl into mitochondria and suggest that a member of the corresponding ABCG family of ABC transporters has this role in humans. Our ABC transporter screen also revealed that mrp-1 and mrp-2 mutants excreted lower MMA than wild type, suggesting they were concentrating intracellular Cbl, consistent with the cellular efflux defect proposed for the mammalian MRP1 ABC transporter.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Caenorhabditis elegans/metabolismo , Vitamina B 12/metabolismo , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Animais , Transporte Biológico , Caenorhabditis elegans/genética , Citosol/enzimologia , Citosol/metabolismo , Humanos , Lisossomos/metabolismo , Espectrometria de Massas , Ácido Metilmalônico/metabolismo , Metilmalonil-CoA Mutase/metabolismo , Mitocôndrias/enzimologia , Proteína 2 Associada à Farmacorresistência Múltipla , Mutação , Propionatos/metabolismo
13.
J Nutr ; 147(4): 482-498, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28250194

RESUMO

Background: Previously, we determined that heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) functions as an intracellular physiologic sensor of folate deficiency. In this model, l-homocysteine, which accumulates intracellularly in proportion to the extent of folate deficiency, covalently binds to and thereby activates homocysteinylated hnRNP-E1 to interact with folate receptor-α mRNA; this high-affinity interaction triggers the translational upregulation of cell surface folate receptors, which enables cells to optimize folate uptake from the external milieu. However, integral to this model is the need for ongoing generation of hnRNP-E1 to replenish homocysteinylated hnRNP-E1 that is degraded.Objective: We searched for an interrelated physiologic mechanism that could also maintain the steady-state concentration of hnRNP-E1 during prolonged folate deficiency.Methods: A novel RNA-protein interaction was functionally characterized by using molecular and biochemical approaches in vitro and in vivo.Results: l-homocysteine triggered a dose-dependent high-affinity interaction between hnRNP-E1 and a 25-nucleotide cis element within the 5'-untranslated region of hnRNP-E1 mRNA; this led to a proportionate increase in these RNA-protein complexes, and translation of hnRNP-E1 both in vitro and within placental cells. Targeted perturbation of this RNA-protein interaction either by specific 25-nucleotide antisense oligonucleotides or mutation within this cis element or by small interfering RNA to hnRNP-E1 mRNA significantly reduced cellular biosynthesis of hnRNP-E1. Conversely, transfection of hnRNP-E1 mutant proteins that mimicked homocysteinylated hnRNP-E1 stimulated both cellular hnRNP-E1 and folate receptor biosynthesis. In addition, ferrous sulfate heptahydrate [iron(II)], which also binds hnRNP-E1, significantly perturbed this l-homocysteine-triggered RNA-protein interaction in a dose-dependent manner. Finally, folate deficiency induced dual upregulation of hnRNP-E1 and folate receptors in cultured human cells and tumor xenografts, and more selectively in various fetal tissues of folate-deficient dams.Conclusions: This novel positive feedback loop amplifies hnRNP-E1 during prolonged folate deficiency and thereby maximizes upregulation of folate receptors in order to restore folate homeostasis toward normalcy in placental cells. It will also functionally impact several other mRNAs of the nutrition-sensitive, folate-responsive posttranscriptional RNA operon that is orchestrated by homocysteinylated hnRNP-E1.


Assuntos
Receptor 2 de Folato/metabolismo , Deficiência de Ácido Fólico/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Placenta/citologia , Regulação para Cima/efeitos dos fármacos , Animais , Linhagem Celular , Proteínas de Ligação a DNA , Feminino , Receptor 2 de Folato/genética , Ácido Fólico/farmacologia , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Neoplasias do Colo do Útero/metabolismo
14.
Mol Genet Metab ; 117(3): 344-50, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26750749

RESUMO

A discrepancy has been identified between numbers of expected and identified patients with homocystinuria due to cystathionine beta-synthase (CBS) deficiency. Patients homozygous for the frequent c.833T>C (p.I278T) are most often responsive to vitamin B6, and can present with a total-homocysteine (tHcy) <100 µM on a normal diet. In Denmark, patients with tHcy <100 µM are not routinely sequenced for CBS(2) mutations. This study investigated the prevalence of CBS mutations and the common methylenetetrahydrofolate reductase (MTHFR) c.677C>T polymorphism in patients with tHcy ≥ 50 µM and the association with clinical manifestations. We studied a cohort of patients with intermediate and severe hyperhomocysteinemia (tHcy ≥ 50 µM) determined between 1996 and 2011. Among the 413 eligible patients, 184 (45%) patients agreed to participate in the present follow-up study. A MTHFR(3)c.677TT genotype was found in 49% of the patients. Eight patients were found to have mutations in CBS(2). Of those, two were homozygous for c.833T>C (p.I278T), and four were compound heterozygous for c.833T>C. One c.833T>C (p.I278T) compound heterozygote was identified by lowering the threshold for sequencing from tHcy at 100 µM to 50 µM. The most prominent clinical presentation among patients with a CBS(2) mutation was thrombosis presenting at a median age of 25 years. In case of arterial or venous thrombosis without any explanation in individuals below 40 years, tHcy should be part of the thrombophilia screening. When tHcy is between 50 and 100 µM genotyping for the MTHFR(3) c.677TT is relevant, and when tHcy >100 µM CBS should be genotyped.


Assuntos
Densidade Óssea , Cistationina beta-Sintase/genética , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Mutação , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Cistationina beta-Sintase/sangue , Cistationina beta-Sintase/deficiência , Dinamarca/epidemiologia , Feminino , Seguimentos , Genótipo , Heterozigoto , Homocisteína/sangue , Homocistinúria/etiologia , Homocistinúria/metabolismo , Homozigoto , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/epidemiologia , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Pessoa de Meia-Idade , Polimorfismo Genético , Prevalência , Tromboembolia/etiologia , Adulto Jovem
15.
Dev Biol ; 396(1): 94-106, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25281006

RESUMO

Mutations in HCFC1 (MIM300019), have been recently associated with cblX (MIM309541), an X-linked, recessive disorder characterized by multiple congenital anomalies including craniofacial abnormalities. HCFC1 is a transcriptional co-regulator that modulates the expression of numerous downstream target genes including MMACHC, but it is not clear how these HCFC1 targets play a role in the clinical manifestations of cblX. To begin to elucidate the mechanism by which HCFC1 modulates disease phenotypes, we have carried out loss of function analyses in the developing zebrafish. Of the two HCFC1 orthologs in zebrafish, hcfc1a and hcfc1b, the loss of hcfc1b specifically results in defects in craniofacial development. Subsequent analysis revealed that hcfc1b regulates cranial neural crest cell differentiation and proliferation within the posterior pharyngeal arches. Further, the hcfc1b-mediated craniofacial abnormalities were rescued by expression of human MMACHC, a downstream target of HCFC1 that is aberrantly expressed in cblX. Furthermore, we tested distinct human HCFC1 mutations for their role in craniofacial development and demonstrated variable effects on MMACHC expression in humans and craniofacial development in zebrafish. Notably, several individuals with mutations in either HCFC1 or MMACHC have been reported to have mild to moderate facial dysmorphia. Thus, our data demonstrates that HCFC1 plays a role in craniofacial development, which is in part mediated through the regulation of MMACHC expression.


Assuntos
Proteínas de Transporte/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Fator C1 de Célula Hospedeira/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Padronização Corporal/genética , Região Branquial/fisiologia , Proteínas de Transporte/genética , Diferenciação Celular , Movimento Celular , Condrócitos/citologia , Anormalidades Craniofaciais/genética , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Fator C1 de Célula Hospedeira/genética , Humanos , Camundongos Transgênicos , Mutação , Crista Neural/citologia , Crista Neural/fisiologia , Oxirredutases , Fenótipo , Células-Tronco/citologia , Vitamina B 12/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
16.
J Nutr ; 145(7): 1507-14, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25995278

RESUMO

BACKGROUND: Limited data are available from controlled studies on biomarkers of maternal vitamin B-12 status. OBJECTIVE: We sought to quantify the effects of pregnancy and lactation on the vitamin B-12 status response to a known and highly controlled vitamin B-12 intake. METHODS: As part of a 10-12 wk feeding trial, pregnant (26-29 wk gestation; n = 26), lactating (5 wk postpartum; n = 28), and control (nonpregnant, nonlactating; n = 21) women consumed vitamin B-12 amounts of ∼8.6 µg/d [mixed diet (∼6 µg/d) plus a prenatal multivitamin supplement (2.6 µg/d)]. Serum vitamin B-12, holotranscobalamin (bioactive form of vitamin B-12), methylmalonic acid (MMA), and homocysteine were measured at baseline and study-end. RESULTS: All participants achieved adequate vitamin B-12 status in response to the study dose. Compared with control women, pregnant women had lower serum vitamin B-12 (-21%; P = 0.02) at study-end, whereas lactating women had higher (P = 0.04) serum vitamin B-12 throughout the study (+26% at study-end). Consumption of the study vitamin B-12 dose increased serum holotranscobalamin in all reproductive groups (+16-42%; P ≤ 0.009). At study-end, pregnant (vs. control) women had a higher holotranscobalamin-to-vitamin B-12 ratio (P = 0.04) with ∼30% (vs. 20%) of total vitamin B-12 in the bioactive form. Serum MMA increased during pregnancy (+50%; P < 0.001) but did not differ by reproductive state at study-end. Serum homocysteine increased in pregnant women (+15%; P = 0.009) but decreased in control and lactating women (-16-17%; P < 0.001). Despite these changes, pregnant women had ∼20% lower serum homocysteine than the other 2 groups at study-end (P ≤ 0.02). CONCLUSION: Pregnancy and lactation alter vitamin B-12 status in a manner consistent with enhanced vitamin B-12 supply to the child. Consumption of the study vitamin B-12 dose (∼3 times the RDA) increased the bioactive form of vitamin B-12, suggesting that women in these reproductive states may benefit from vitamin B-12 intakes exceeding current recommendations. This trial was registered at clinicaltrials.gov as NCT01127022.


Assuntos
Ingestão de Energia , Micronutrientes/administração & dosagem , Vitamina B 12/sangue , Adulto , Biomarcadores/sangue , Aleitamento Materno , Colina/administração & dosagem , Colina/sangue , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Homocisteína/sangue , Homocisteína/urina , Humanos , Lactação/sangue , Ácido Metilmalônico/sangue , Período Pós-Parto , Gravidez , Recomendações Nutricionais , Vitamina B 12/administração & dosagem , Adulto Jovem
17.
FASEB J ; 28(9): 4044-54, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24891521

RESUMO

Cystathionine ß-synthase-deficient homocystinuria (HCU) is a serious life-threatening inborn error of sulfur metabolism with poorly understood pathogenic mechanisms. We investigated the effect of HCU on hepatic cysteine oxidation in a transgenic mouse model of the disease. Cysteine dioxygenase (CDO) protein levels were 90% repressed without any change in mRNA levels. Cysteinesulfinic acid decarboxylase (CSAD) was induced at both the mRNA (8-fold) and protein (15-fold) levels. Cysteine supplementation normalized CDO protein levels without reversing the induction of CSAD. Regulatory changes in CDO and CSAD expression were proportional to homocysteine elevation, indicating a possible threshold effect. Hepatic and blood taurine levels in HCU animals were decreased by 21 and 35%, respectively, and normalized by cysteine supplementation. Expression of the cytoplasmic (GOT1) and mitochondrial (GOT2) isoforms of glutamic-oxaloacetic transaminase were repressed in HCU animals by 86 and 30%, respectively. HCU induced regulatory changes in CSAD, CDO, and GOT1 expression were normalized by taurine supplementation, indicating that cysteine is not the only sulfur compound that regulates hepatic cysteine oxidation. Collectively, our results indicate that HCU induces significant alterations of sulfur metabolism with the potential to contribute to pathogenesis and that cysteine and taurine have the potential to serve as adjunctive treatments in this disease.


Assuntos
Cistationina beta-Sintase/fisiologia , Cisteína/metabolismo , Homocistinúria/fisiopatologia , Fígado/metabolismo , Enxofre/metabolismo , Taurina/farmacologia , Animais , Western Blotting , Carboxiliases/genética , Carboxiliases/metabolismo , Cisteína/química , Cisteína Dioxigenase/genética , Cisteína Dioxigenase/metabolismo , Suplementos Nutricionais , Feminino , Homocistinúria/tratamento farmacológico , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Metionina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oxirredução , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Biol Chem ; 287(15): 12559-77, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22351779

RESUMO

Although HPV16 transforms infected epithelial tissues to cancer in the presence of several co-factors, there is insufficient molecular evidence that poor nutrition has any such role. Because physiological folate deficiency led to the intracellular homocysteinylation of heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) and activated a nutrition-sensitive (homocysteine-responsive) posttranscriptional RNA operon that included interaction with HPV16 L2 mRNA, we investigated the functional consequences of folate deficiency on HPV16 in immortalized HPV16-harboring human (BC-1-Ep/SL) keratinocytes and HPV16-organotypic rafts. Although homocysteinylated hnRNP-E1 interacted with HPV16 L2 mRNA cis-element, it also specifically bound another HPV16 57-nucleotide poly(U)-rich cis-element in the early polyadenylation element (upstream of L2L1 genes) with greater affinity. Together, these interactions led to a profound reduction of both L1 and L2 mRNA and proteins without effects on HPV16 E6 and E7 in vitro, and in cultured keratinocyte monolayers and HPV16-low folate-organotypic rafts developed in physiological low folate medium. In addition, HPV16-low folate-organotypic rafts contained fewer HPV16 viral particles, a similar HPV16 DNA viral load, and a much greater extent of integration of HPV16 DNA into genomic DNA when compared with HPV16-high folate-organotypic rafts. Subcutaneous implantation of 18-day old HPV16-low folate-organotypic rafts into folate-replete immunodeficient mice transformed this benign keratinocyte-derived raft tissue into an aggressive HPV16-induced cancer within 12 weeks. Collectively, these studies establish a likely molecular linkage between poor folate nutrition and HPV16 and predict that nutritional folate and/or vitamin-B(12) deficiency, which are both common worldwide, will alter the natural history of HPV16 infections and also warrant serious consideration as reversible co-factors in oncogenic transformation of HPV16-infected tissues to cancer.


Assuntos
Transformação Celular Viral , Deficiência de Ácido Fólico , Papillomavirus Humano 16/fisiologia , Queratinócitos/virologia , Neoplasias Experimentais/virologia , Infecções por Papillomavirus/virologia , Animais , Sequência de Bases , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Catalase/biossíntese , Catalase/genética , Técnicas de Cultura de Células , Proliferação de Células , Células Cultivadas , Proteínas de Ligação a DNA , Feminino , Ácido Fólico/metabolismo , Genes Reporter , Ribonucleoproteínas Nucleares Heterogêneas/química , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Homocisteína/química , Homocisteína/metabolismo , Interações Hospedeiro-Patógeno , Papillomavirus Humano 16/genética , Humanos , Queratinócitos/metabolismo , Queratinócitos/transplante , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Neoplasias Experimentais/patologia , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Ligação Proteica , Proteólise , RNA Mensageiro/biossíntese , RNA Mensageiro/química , RNA Viral/química , RNA Viral/metabolismo , Proteínas de Ligação a RNA , Compostos de Sulfidrila/metabolismo , Carga Tumoral , Integração Viral
19.
J Biol Chem ; 287(38): 31994-2005, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22854956

RESUMO

Cystathionine (R-S-(2-amino-2-carboxyethyl)-l-homocysteine) is a non-proteinogenic thioether containing amino acid. In mammals, cystathionine is formed as an intermediate of the transsulfuration pathway by the condensation of serine and homocysteine (Hcy) in a reaction catalyzed by cystathionine ß-synthase (CBS). Cystathionine is subsequently converted to cysteine plus ammonia and α-ketobutyrate by the action of cystathionine γ-lyase (CGL). Pathogenic mutations in CBS result in CBS-deficient homocystinuria (HCU) which, if untreated, results in mental retardation, thromboembolic complications and connective tissue disorders. Currently there is no known function for cystathionine other than serving as an intermediate in transsulfuration and to date, the possible contribution of the abolition of cystathionine synthesis to pathogenesis in HCU has not been investigated. Using both mouse and cell-culture models, we have found that cystathionine is capable of blocking the induction of hepatic steatosis and kidney injury, acute tubular necrosis, and apoptotic cell death by the endoplasmic reticulum stress inducing agent tunicamycin. Northern and Western blotting analysis indicate that the protective effects of cystathionine occur without any obvious alteration of the induction of the unfolded protein response. Our data constitute the first experimental evidence that the abolition of cystathionine synthesis may contribute to the pathology of HCU and that this compound has therapeutic potential for disease states where ER stress is implicated as a primary initiating pathogenic factor.


Assuntos
Apoptose , Cistationina/química , Retículo Endoplasmático/metabolismo , Homocistinúria/metabolismo , Lipídeos/química , Animais , Antibacterianos/farmacologia , Cistationina beta-Sintase/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mutação , Necrose/patologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Desnaturação Proteica , Tunicamicina/farmacologia
20.
J Nutr ; 143(7): 1028-35, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23700346

RESUMO

Impaired folate-mediated one-carbon metabolism (OCM) has emerged as a risk factor for several diseases associated with age-related cognitive decline, but the underlying mechanisms remain unknown and thus hinder the identification of subpopulations most vulnerable to OCM disruption. Here we investigated the role of serine hydroxymethyltransferase 1 (SHMT1), a folate-dependent enzyme regulating de novo thymidylate biosynthesis, in influencing neuronal and cognitive function in the adult mouse. We observed Shmt1 expression in the hippocampus, including the granule cell layer of the dentate gyrus (DG), and examined hippocampal neurogenesis and hippocampal-dependent fear conditioning in mice deficient for Shmt1. We used a 3 × 3 design in which adult male Shmt1(+/+), Shmt1(+/-), and Shmt1(-/-) mice were fed folic acid control (2 mg/kg), folic acid-deficient (0 mg/kg), or folic acid-supplemented (8 mg/kg) diets from weaning through the duration of the study. Proliferation within the DG was elevated by 70% in Shmt1(+/-) mice, yet the number of newborn mature neurons was reduced by 98% compared with that in Shmt1(+/+) mice. Concomitant with these alterations, Shmt1(+/-) mice showed a 45% reduction in mnemonic recall during trace fear conditioning. Dietary folate manipulations alone did not influence neural outcomes. Together, these data identify SHMT1 as one of the first enzymes within the OCM pathway to regulate neuronal and cognitive profiles and implicate impaired thymidylate biosynthesis in the etiology of folate-related neuropathogenesis.


Assuntos
Glicina Hidroximetiltransferase/genética , Hipocampo/patologia , Memória/fisiologia , Neurogênese/genética , Animais , Proliferação de Células , Giro Denteado/metabolismo , Medo/fisiologia , Ácido Fólico/administração & dosagem , Ácido Fólico/sangue , Deficiência de Ácido Fólico/patologia , Regulação da Expressão Gênica , Glicina Hidroximetiltransferase/metabolismo , Hipocampo/metabolismo , Homocisteína/metabolismo , Hibridização In Situ , Masculino , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Fatores de Risco , Timidina Monofosfato/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA