Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 169(3): 442-456.e18, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431245

RESUMEN

Fluoropyrimidines are the first-line treatment for colorectal cancer, but their efficacy is highly variable between patients. We queried whether gut microbes, a known source of inter-individual variability, impacted drug efficacy. Combining two tractable genetic models, the bacterium E. coli and the nematode C. elegans, we performed three-way high-throughput screens that unraveled the complexity underlying host-microbe-drug interactions. We report that microbes can bolster or suppress the effects of fluoropyrimidines through metabolic drug interconversion involving bacterial vitamin B6, B9, and ribonucleotide metabolism. Also, disturbances in bacterial deoxynucleotide pools amplify 5-FU-induced autophagy and cell death in host cells, an effect regulated by the nucleoside diphosphate kinase ndk-1. Our data suggest a two-way bacterial mediation of fluoropyrimidine effects on host metabolism, which contributes to drug efficacy. These findings highlight the potential therapeutic power of manipulating intestinal microbiota to ensure host metabolic health and treat disease.


Asunto(s)
Antineoplásicos/metabolismo , Escherichia coli/metabolismo , Fluorouracilo/metabolismo , Microbioma Gastrointestinal , Animales , Autofagia , Caenorhabditis elegans , Muerte Celular , Neoplasias Colorrectales/tratamiento farmacológico , Dieta , Escherichia coli/enzimología , Escherichia coli/genética , Humanos , Modelos Animales , Pentosiltransferasa/genética
2.
Cell ; 153(1): 228-39, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23540700

RESUMEN

The biguanide drug metformin is widely prescribed to treat type 2 diabetes and metabolic syndrome, but its mode of action remains uncertain. Metformin also increases lifespan in Caenorhabditis elegans cocultured with Escherichia coli. This bacterium exerts complex nutritional and pathogenic effects on its nematode predator/host that impact health and aging. We report that metformin increases lifespan by altering microbial folate and methionine metabolism. Alterations in metformin-induced longevity by mutation of worm methionine synthase (metr-1) and S-adenosylmethionine synthase (sams-1) imply metformin-induced methionine restriction in the host, consistent with action of this drug as a dietary restriction mimetic. Metformin increases or decreases worm lifespan, depending on E. coli strain metformin sensitivity and glucose concentration. In mammals, the intestinal microbiome influences host metabolism, including development of metabolic disease. Thus, metformin-induced alteration of microbial metabolism could contribute to therapeutic efficacy-and also to its side effects, which include folate deficiency and gastrointestinal upset.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/microbiología , Ácido Fólico/metabolismo , Hipoglucemiantes/farmacología , Longevidad/efectos de los fármacos , Metformina/farmacología , Metionina/metabolismo , Adenilato Quinasa/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Biguanidas/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Restricción Calórica , Proteínas de Unión al ADN/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Escherichia coli/metabolismo , Humanos , Hipoglucemiantes/metabolismo , Metagenoma , Metformina/metabolismo , Factores de Transcripción/metabolismo
3.
Mol Cell Proteomics ; 23(3): 100718, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38224738

RESUMEN

A functional role has been ascribed to the human dihydrofolate reductase 2 (DHFR2) gene based on the enzymatic activity of recombinant versions of the predicted translated protein. However, the in vivo function is still unclear. The high amino acid sequence identity (92%) between DHFR2 and its parental homolog, DHFR, makes analysis of the endogenous protein challenging. This paper describes a targeted mass spectrometry proteomics approach in several human cell lines and tissue types to identify DHFR2-specific peptides as evidence of its translation. We show definitive evidence that the DHFR2 activity in the mitochondria is in fact mediated by DHFR, and not DHFR2. Analysis of Ribo-seq data and an experimental assessment of ribosome association using a sucrose cushion showed that the two main Ensembl annotated mRNA isoforms of DHFR2, 201 and 202, are differentially associated with the ribosome. This indicates a functional role at both the RNA and protein level. However, we were unable to detect DHFR2 protein at a detectable level in most cell types examined despite various RNA isoforms of DHFR2 being relatively abundant. We did detect a DHFR2-specific peptide in embryonic heart, indicating that the protein may have a specific role during embryogenesis. We propose that the main functionality of the DHFR2 gene in adult cells is likely to arise at the RNA level.


Asunto(s)
ARN , Tetrahidrofolato Deshidrogenasa , Humanos , Línea Celular , Péptidos/metabolismo , Biosíntesis de Proteínas , Ribosomas/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo
4.
FASEB J ; 38(11): e23738, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38855924

RESUMEN

Maternal nutrition contributes to gene-environment interactions that influence susceptibility to common congenital anomalies such as neural tube defects (NTDs). Supplemental myo-inositol (MI) can prevent NTDs in some mouse models and shows potential for prevention of human NTDs. We investigated effects of maternal MI intake on embryonic MI status and metabolism in curly tail mice, which are genetically predisposed to NTDs that are inositol-responsive but folic acid resistant. Dietary MI deficiency caused diminished MI in maternal plasma and embryos, showing that de novo synthesis is insufficient to maintain MI levels in either adult or embryonic mice. Under normal maternal dietary conditions, curly tail embryos that developed cranial NTDs had significantly lower MI content than unaffected embryos, revealing an association between diminished MI status and failure of cranial neurulation. Expression of inositol-3-phosphate synthase 1, required for inositol biosynthesis, was less abundant in the cranial neural tube than at other axial levels. Supplemental MI or d-chiro-inositol (DCI) have previously been found to prevent NTDs in curly tail embryos. Here, we investigated the metabolic effects of MI and DCI treatments by mass spectrometry-based metabolome analysis. Among inositol-responsive metabolites, we noted a disproportionate effect on nucleotides, especially purines. We also found altered proportions of 5-methyltetrahydrolate and tetrahydrofolate in MI-treated embryos suggesting altered folate metabolism. Treatment with nucleotides or the one-carbon donor formate has also been found to prevent NTDs in curly tail embryos. Together, these findings suggest that the protective effect of inositol may be mediated through the enhanced supply of nucleotides during neural tube closure.


Asunto(s)
Inositol , Defectos del Tubo Neural , Inositol/metabolismo , Inositol/farmacología , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/prevención & control , Animales , Femenino , Ratones , Embarazo , Embrión de Mamíferos/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Metaboloma , Ácido Fólico/metabolismo
5.
Mol Genet Metab ; 142(3): 108496, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38761651

RESUMEN

Non-Ketotic Hyperglycinemia (NKH) is a rare inborn error of metabolism caused by impaired function of the glycine cleavage system (GCS) and characterised by accumulation of glycine in body fluids and tissues. NKH is an autosomal recessive condition and the majority of affected individuals carry mutations in GLDC (glycine decarboxylase). Current treatments for NKH have limited effect and are not curative. As a monogenic condition with known genetic causation, NKH is potentially amenable to gene therapy. An AAV9-based expression vector was designed to target sites of GCS activity. Using a ubiquitous promoter to drive expression of a GFP reporter, transduction of liver and brain was confirmed following intra-venous and/or intra-cerebroventricular administration to neonatal mice. Using the same capsid and promoter with transgenes to express mouse or human GLDC, vectors were then tested in GLDC-deficient mice that provide a model of NKH. GLDC-deficient mice exhibited elevated plasma glycine concentration and accumulation of glycine in liver and brain tissues as previously observed. Moreover, the folate profile indicated suppression of folate one­carbon metabolism (FOCM) in brain tissue, as found at embryonic stages, and reduced abundance of FOCM metabolites including betaine and choline. Neonatal administration of vector achieved reinstatement of GLDC mRNA and protein expression in GLDC-deficient mice. Treated GLDC-deficient mice showed significant lowering of plasma glycine, confirming functionality of vector expressed protein. AAV9-GLDC treatment also led to lowering of brain tissue glycine, and normalisation of the folate profile indicating restoration of glycine-derived one­carbon supply. These findings support the hypothesis that AAV-mediated gene therapy may offer potential in treatment of NKH.


Asunto(s)
Encéfalo , Dependovirus , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos , Glicina-Deshidrogenasa (Descarboxilante) , Glicina , Hiperglicinemia no Cetósica , Hígado , Animales , Hiperglicinemia no Cetósica/genética , Hiperglicinemia no Cetósica/metabolismo , Hiperglicinemia no Cetósica/terapia , Glicina-Deshidrogenasa (Descarboxilante)/genética , Glicina-Deshidrogenasa (Descarboxilante)/metabolismo , Dependovirus/genética , Ratones , Humanos , Vectores Genéticos/genética , Glicina/metabolismo , Hígado/metabolismo , Encéfalo/metabolismo , Biomarcadores/metabolismo , Ácido Fólico/metabolismo
6.
J Nutr ; 152(11): 2333-2342, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36774100

RESUMEN

BACKGROUND: Myo-inositol (MI) is incorporated into numerous biomolecules, including phosphoinositides and inositol phosphates. Disturbance of inositol availability or metabolism is associated with various disorders, including neurological conditions and cancers, whereas supplemental MI has therapeutic potential in conditions such as depression, polycystic ovary syndrome, and congenital anomalies. Inositol status can be influenced by diet, synthesis, transport, utilization, and catabolism. OBJECTIVES: We aimed to investigate potential genetic regulation of circulating MI status and to evaluate correlation of MI concentration with other metabolites. METHODS: GC-MS was used to determine plasma MI concentration of >2000 healthy, young adults (aged 18-28 y) from the Trinity Student Study. Genotyping data were used to test association of plasma MI with single nucleotide polymorphisms (SNPs) in candidate genes, encoding inositol transporters and synthesizing enzymes, and test for genome-wide association. We evaluated potential correlation of plasma MI with d-chiro-inositol (DCI), glucose, and other metabolites by Spearman rank correlation. RESULTS: Mean plasma MI showed a small but significant difference between males and females (28.5 and 26.9 µM, respectively). Candidate gene analysis revealed several nominally significant associations with plasma MI, most notably for SLC5A11 (solute carrier family 5 member 11), encoding a sodium-coupled inositol transporter, also known as SMIT2 (sodium-dependent myo-inositol transporter 2). However, these did not survive correction for multiple testing. Subsequent testing for genome-wide association with plasma MI did not identify associations of genome-wide significance (P < 5 × 10-8). However, 8 SNPs exceeded the threshold for suggestive significant association with plasma MI concentration (P < 1 × 10-5), 3 of which were located within or close to genes: MTDH (metadherin), LAPTM4B (lysosomal protein transmembrane 4 ß), and ZP2 (zona pellucida 2). We found significant positive correlation of plasma MI concentration with concentration of dci and several other biochemicals including glucose, methionine, betaine, sarcosine, and tryptophan. CONCLUSIONS: Our findings suggest potential for modulation of plasma MI in young adults by variation in SLC5A11, which is worthy of further investigation.


Asunto(s)
Inositol , Síndrome del Ovario Poliquístico , Femenino , Humanos , Masculino , Adulto Joven , Dieta , Estudio de Asociación del Genoma Completo , Glucosa , Inositol/sangre , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana , Proteínas Oncogénicas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Transporte de Sodio-Glucosa/uso terapéutico
7.
Cereb Cortex ; 31(1): 635-649, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32995858

RESUMEN

Folate is an essential micronutrient required for both cellular proliferation through de novo nucleotide synthesis and epigenetic regulation of gene expression through methylation. This dual requirement places a particular demand on folate availability during pregnancy when both rapid cell generation and programmed differentiation of maternal, extraembryonic, and embryonic/fetal tissues are required. Accordingly, prenatal neurodevelopment is particularly susceptible to folate deficiency, which can predispose to neural tube defects, or when effective transport into the brain is impaired, cerebral folate deficiency. Consequently, adequate folate consumption, in the form of folic acid (FA) fortification and supplement use, is widely recommended and has led to a substantial increase in the amount of FA intake during pregnancy in some populations. Here, we show that either maternal folate deficiency or FA excess in mice results in disruptions in folate metabolism of the offspring, suggesting diversion of the folate cycle from methylation to DNA synthesis. Paradoxically, either intervention causes comparable neurodevelopmental changes by delaying prenatal cerebral cortical neurogenesis in favor of late-born neurons. These cytoarchitectural and biochemical alterations are accompanied by behavioral abnormalities in FA test groups compared with controls. Our findings point to overlooked potential neurodevelopmental risks associated with excessively high levels of prenatal FA intake.


Asunto(s)
Conducta Animal/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Ácido Fólico/farmacología , Embarazo/efectos de los fármacos , Animales , Metilación de ADN/efectos de los fármacos , Suplementos Dietéticos/efectos adversos , Femenino , Deficiencia de Ácido Fólico/complicaciones , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Ratones Endogámicos C57BL
8.
J Inherit Metab Dis ; 43(6): 1186-1198, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32743799

RESUMEN

Glycine abundance is modulated in a tissue-specific manner by use in biosynthetic reactions, catabolism by the glycine cleavage system (GCS), and excretion via glycine conjugation. Dysregulation of glycine metabolism is associated with multiple disorders including epilepsy, developmental delay, and birth defects. Mutation of the GCS component glycine decarboxylase (GLDC) in non-ketotic hyperglycinemia (NKH) causes accumulation of glycine in body fluids, but there is a gap in our knowledge regarding the effects on glycine metabolism in tissues. Here, we analysed mice carrying mutations in Gldc that result in severe or mild elevations of plasma glycine and model NKH. Liver of Gldc-deficient mice accumulated glycine and numerous glycine derivatives, including multiple acylglycines, indicating increased flux through reactions mediated by enzymes including glycine-N-acyltransferase and arginine: glycine amidinotransferase. Levels of dysregulated metabolites increased with age and were normalised by liver-specific rescue of Gldc expression. Brain tissue exhibited increased abundance of glycine, as well as derivatives including guanidinoacetate, which may itself be epileptogenic. Elevation of brain tissue glycine occurred even in the presence of only mildly elevated plasma glycine in mice carrying a missense allele of Gldc. Treatment with benzoate enhanced hepatic glycine conjugation thereby lowering plasma and tissue glycine. Moreover, administration of a glycine conjugation pathway intermediate, cinnamate, similarly achieved normalisation of liver glycine derivatives and circulating glycine. Although exogenous benzoate and cinnamate impact glycine levels via activity of glycine-N-acyltransferase, that is not expressed in brain, they are sufficient to lower levels of glycine and derivatives in brain tissue of treated Gldc-deficient mice.


Asunto(s)
Encéfalo/metabolismo , Glicina-Deshidrogenasa (Descarboxilante)/genética , Glicina/metabolismo , Hiperglicinemia no Cetósica/enzimología , Alelos , Animales , Encéfalo/patología , Hiperglicinemia no Cetósica/patología , Ratones , Mutación Missense
9.
Hum Mol Genet ; 26(5): 888-900, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28069796

RESUMEN

Methylenetetrahydrofolate reductase (MTHFR) generates methyltetrahydrofolate for methylation reactions. Severe MTHFR deficiency results in homocystinuria and neurologic impairment. Mild MTHFR deficiency (677C > T polymorphism) increases risk for complex traits, including neuropsychiatric disorders. Although low dietary folate impacts brain development, recent concerns have focused on high folate intake following food fortification and increased vitamin use. Our goal was to determine whether high dietary folate during pregnancy affects brain development in murine offspring. Female mice were placed on control diet (CD) or folic acid-supplemented diet (FASD) throughout mating, pregnancy and lactation. Three-week-old male pups were evaluated for motor and cognitive function. Tissues from E17.5 embryos, pups and dams were collected for choline/methyl metabolite measurements, immunoblotting or gene expression of relevant enzymes. Brains were examined for morphology of hippocampus and cortex. Pups of FASD mothers displayed short-term memory impairment, decreased hippocampal size and decreased thickness of the dentate gyrus. MTHFR protein levels were reduced in FASD pup livers, with lower concentrations of phosphocholine and glycerophosphocholine in liver and hippocampus, respectively. FASD pup brains showed evidence of altered acetylcholine availability and Dnmt3a mRNA was reduced in cortex and hippocampus. E17.5 embryos and placentas from FASD dams were smaller. MTHFR protein and mRNA were reduced in embryonic liver, with lower concentrations of choline, betaine and phosphocholine. Embryonic brain displayed altered development of cortical layers. In summary, high folate intake during pregnancy leads to pseudo-MTHFR deficiency, disturbed choline/methyl metabolism, embryonic growth delay and memory impairment in offspring. These findings highlight the unintended negative consequences of supplemental folic acid.


Asunto(s)
Ácido Fólico/efectos adversos , Homocistinuria/genética , Memoria a Corto Plazo/efectos de los fármacos , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Espasticidad Muscular/genética , Acetilcolina/genética , Acetilcolina/metabolismo , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Dieta/efectos adversos , Femenino , Ácido Fólico/administración & dosificación , Homocistinuria/inducido químicamente , Homocistinuria/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/fisiopatología , Metilación , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Ratones , Espasticidad Muscular/inducido químicamente , Espasticidad Muscular/patología , Embarazo , Trastornos Psicóticos/genética , Trastornos Psicóticos/patología
10.
FASEB J ; 29(3): 1069-79, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25466894

RESUMEN

The embryonic pattern of global DNA methylation is first established in the inner cell mass (ICM) of the mouse blastocyst. The methyl donor S-adenosylmethionine (SAM) is produced in most cells through the folate cycle, but only a few cell types generate SAM from betaine (N,N,N-trimethylglycine) via betaine-homocysteine methyltransferase (BHMT), which is expressed in the mouse ICM. Here, mean ICM cell numbers decreased from 18-19 in controls to 11-13 when the folate cycle was inhibited by the antifolate methotrexate and to 12-14 when BHMT expression was knocked down by antisense morpholinos. Inhibiting both pathways, however, much more severely affected ICM development (7-8 cells). Total SAM levels in mouse blastocysts decreased significantly only when both pathways were inhibited (from 3.1 to 1.6 pmol/100 blastocysts). DNA methylation, detected as 5-methylcytosine (5-MeC) immunofluorescence in isolated ICMs, was minimally affected by inhibition of either pathway alone but decreased by at least 45-55% when both BHMT and the folate cycle were inhibited simultaneously. Effects on cell numbers and 5-MeC levels in the ICM were completely rescued by methionine (immediate SAM precursor) or SAM. Both the folate cycle and betaine/BHMT appear to contribute to a methyl pool required for normal ICM development and establishing initial embryonic DNA methylation.


Asunto(s)
Betaína-Homocisteína S-Metiltransferasa/metabolismo , Blastocisto/metabolismo , Metilación de ADN , Embrión de Mamíferos/metabolismo , Ácido Fólico/metabolismo , Regulación Enzimológica de la Expresión Génica , S-Adenosilmetionina/metabolismo , 5-Metilcitosina/análisis , Animales , Antimetabolitos Antineoplásicos/farmacología , Betaína-Homocisteína S-Metiltransferasa/antagonistas & inhibidores , Blastocisto/citología , Blastocisto/efectos de los fármacos , Linaje de la Célula , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Metotrexato/farmacología , Ratones , Proteínas Nucleares snRNP/metabolismo
11.
Br J Nutr ; 115(6): 974-83, 2016 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-26847388

RESUMEN

Although peri-conceptional folic acid (FA) supplementation can prevent a proportion of neural tube defects (NTD), there is increasing evidence that many NTD are FA non-responsive. The vitamin-like molecule inositol may offer a novel approach to preventing FA-non-responsive NTD. Inositol prevented NTD in a genetic mouse model, and was well tolerated by women in a small study of NTD recurrence. In the present study, we report the Prevention of Neural Tube Defects by Inositol (PONTI) pilot study designed to gain further experience of inositol usage in human pregnancy as a preliminary trial to a future large-scale controlled trial to evaluate efficacy of inositol in NTD prevention. Study subjects were UK women with a previous NTD pregnancy who planned to become pregnant again. Of 117 women who made contact, ninety-nine proved eligible and forty-seven agreed to be randomised (double-blind) to peri-conceptional supplementation with inositol plus FA or placebo plus FA. In total, thirty-three randomised pregnancies produced one NTD recurrence in the placebo plus FA group (n 19) and no recurrences in the inositol plus FA group (n 14). Of fifty-two women who declined randomisation, the peri-conceptional supplementation regimen and outcomes of twenty-two further pregnancies were documented. Two NTD recurred, both in women who took only FA in their next pregnancy. No adverse pregnancy events were associated with inositol supplementation. The findings of the PONTI pilot study encourage a large-scale controlled trial of inositol for NTD prevention, but indicate the need for a careful study design in view of the unwillingness of many high-risk women to be randomised.


Asunto(s)
Suplementos Dietéticos , Ácido Fólico/uso terapéutico , Inositol/uso terapéutico , Fenómenos Fisiologicos Nutricionales Maternos , Defectos del Tubo Neural/prevención & control , Atención Preconceptiva , Adulto , Estudios de Cohortes , Suplementos Dietéticos/efectos adversos , Método Doble Ciego , Estudios de Factibilidad , Femenino , Ácido Fólico/efectos adversos , Estudios de Seguimiento , Humanos , Inositol/efectos adversos , Inositol/sangre , Inositol/orina , Defectos del Tubo Neural/sangre , Defectos del Tubo Neural/epidemiología , Defectos del Tubo Neural/orina , Cooperación del Paciente , Proyectos Piloto , Embarazo , Recurrencia , Riesgo , Reino Unido/epidemiología , Adulto Joven
12.
PLoS Genet ; 8(11): e1003059, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166514

RESUMEN

Neural tube defects (NTDs), including spina bifida and anencephaly, are common birth defects whose complex multigenic causation has hampered efforts to delineate their molecular basis. The effect of putative modifier genes in determining NTD susceptibility may be investigated in mouse models, particularly those that display partial penetrance such as curly tail, a strain in which NTDs result from a hypomorphic allele of the grainyhead-like-3 gene. Through proteomic analysis, we found that the curly tail genetic background harbours a polymorphic variant of lamin B1, lacking one of a series of nine glutamic acid residues. Lamins are intermediate filament proteins of the nuclear lamina with multiple functions that influence nuclear structure, cell cycle properties, and transcriptional regulation. Fluorescence loss in photobleaching showed that the variant lamin B1 exhibited reduced stability in the nuclear lamina. Genetic analysis demonstrated that the variant also affects neural tube closure: the frequency of spina bifida and anencephaly was reduced three-fold when wild-type lamin B1 was bred into the curly tail strain background. Cultured fibroblasts expressing variant lamin B1 show significantly increased nuclear dysmorphology and diminished proliferative capacity, as well as premature senescence, associated with reduced expression of cyclins and Smc2, and increased expression of p16. The cellular basis of spinal NTDs in curly tail embryos involves a proliferation defect localised to the hindgut epithelium, and S-phase progression was diminished in the hindgut of embryos expressing variant lamin B1. These observations indicate a mechanistic link between altered lamin B1 function, exacerbation of the Grhl3-mediated cell proliferation defect, and enhanced susceptibility to NTDs. We conclude that lamin B1 is a modifier gene of major effect for NTDs resulting from loss of Grhl3 function, a role that is likely mediated via the key function of lamin B1 in maintaining integrity of the nuclear envelope and ensuring normal cell cycle progression.


Asunto(s)
Ciclo Celular , Lamina Tipo B , Defectos del Tubo Neural , Membrana Nuclear , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular , División Celular , Proliferación Celular , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Ratones , Mutación , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Polimorfismo Genético , Proteómica , Disrafia Espinal/genética , Disrafia Espinal/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Hum Mol Genet ; 21(7): 1496-503, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22171071

RESUMEN

Neural tube defects (NTDs), including spina bifida and anencephaly, are common birth defects of the central nervous system. The complex multigenic causation of human NTDs, together with the large number of possible candidate genes, has hampered efforts to delineate their molecular basis. Function of folate one-carbon metabolism (FOCM) has been implicated as a key determinant of susceptibility to NTDs. The glycine cleavage system (GCS) is a multi-enzyme component of mitochondrial folate metabolism, and GCS-encoding genes therefore represent candidates for involvement in NTDs. To investigate this possibility, we sequenced the coding regions of the GCS genes: AMT, GCSH and GLDC in NTD patients and controls. Two unique non-synonymous changes were identified in the AMT gene that were absent from controls. We also identified a splice acceptor site mutation and five different non-synonymous variants in GLDC, which were found to significantly impair enzymatic activity and represent putative causative mutations. In order to functionally test the requirement for GCS activity in neural tube closure, we generated mice that lack GCS activity, through mutation of AMT. Homozygous Amt(-/-) mice developed NTDs at high frequency. Although these NTDs were not preventable by supplemental folic acid, there was a partial rescue by methionine. Overall, our findings suggest that loss-of-function mutations in GCS genes predispose to NTDs in mice and humans. These data highlight the importance of adequate function of mitochondrial folate metabolism in neural tube closure.


Asunto(s)
Aminometiltransferasa/genética , Proteína H del Complejo de la Glicina Descarboxilasa/genética , Glicina-Deshidrogenasa (Descarboxilante)/genética , Mutación , Defectos del Tubo Neural/genética , Animales , Complejo Glicina-Descarboxilasa/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación Missense
14.
Brain ; 136(Pt 9): 2836-41, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23935126

RESUMEN

Closure of the neural tube during embryogenesis is a crucial step in development of the central nervous system. Failure of this process results in neural tube defects, including spina bifida and anencephaly, which are among the most common birth defects worldwide. Maternal use of folic acid supplements reduces risk of neural tube defects but a proportion of cases are not preventable. Folic acid is thought to act through folate one-carbon metabolism, which transfers one-carbon units for methylation reactions and nucleotide biosynthesis. Hence suboptimal performance of the intervening reactions could limit the efficacy of folic acid. We hypothesized that direct supplementation with nucleotides, downstream of folate metabolism, has the potential to support neural tube closure. Therefore, in a mouse model that exhibits folic acid-resistant neural tube defects, we tested the effect of specific combinations of pyrimidine and purine nucleotide precursors and observed a significant protective effect. Labelling in whole embryo culture showed that nucleotides are taken up by the neurulating embryo and incorporated into genomic DNA. Furthermore, the mitotic index was elevated in neural folds and hindgut of treated embryos, consistent with a proposed mechanism of neural tube defect prevention through stimulation of cellular proliferation. These findings may provide an impetus for future investigations of supplemental nucleotides as a means to prevent a greater proportion of human neural tube defects than can be achieved by folic acid alone.


Asunto(s)
Ácido Fólico/efectos adversos , Defectos del Tubo Neural/prevención & control , Nucleósidos de Purina/uso terapéutico , Nucleósidos de Pirimidina/uso terapéutico , Animales , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/fisiología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Ácido Fólico/metabolismo , Histonas/metabolismo , Tamaño de la Camada/efectos de los fármacos , Masculino , Exposición Materna , Ratones , Ratones Mutantes , Defectos del Tubo Neural/tratamiento farmacológico , Defectos del Tubo Neural/genética , Embarazo , Estadísticas no Paramétricas , Timidina/uso terapéutico
15.
Mol Cell Biochem ; 378(1-2): 229-36, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23483428

RESUMEN

Folates act as co-factors for transfer of one-carbon units for nucleotide production, methylation and other biosynthetic reactions. Comprehensive profiling of multiple folates can be achieved using liquid chromatography tandem mass spectrometry, enabling determination of their relative abundance that may provide an indication of metabolic differences between cell types. For example, cell lines exposed to methotrexate showed a dose-dependent elevation of dihydrofolate, consistent with inhibition of dihydrofolate reductase. We analysed the folate profile of E. coli sub-types as well as cell lines and embryonic tissue from both human and mouse. The folate profile of bacteria differed markedly from those of all the mammalian samples, most notably in the greater abundance of formyl tetrahydrofolate. The overall profiles of mouse and human fibroblasts and mid-gestation mouse embryos were broadly similar, with specific differences. The major folate species in these cell types was 5-methyl tetrahydrofolate, in contrast to lymphoblastoid cell lines in which the predominant form was tetrahydrofolate. Analysis of embryonic human brain revealed a shift in folate profile with increasing developmental stage, with a decline in relative abundance of dihydrofolate and increase in 5-methyl tetrahydrofolate. These cell type-specific and developmental changes in folate profile may indicate differential requirements for the various outputs of folate metabolism.


Asunto(s)
Encéfalo/metabolismo , Tetrahidrofolatos/metabolismo , Animales , Encéfalo/embriología , Línea Celular , Escherichia coli/metabolismo , Ácido Fólico/metabolismo , Humanos , Metotrexato/química , Metotrexato/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Estándares de Referencia , Especificidad de la Especie , Espectrometría de Masas en Tándem/métodos , Espectrometría de Masas en Tándem/normas , Tetrahidrofolatos/química
16.
Front Immunol ; 14: 1209490, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37457690

RESUMEN

Objectives: The disease-modifying anti-rheumatic drug methotrexate (MTX) is recognized to reduce cardiovascular risk in patients with systemic inflammatory diseases. However, the molecular basis for these cardioprotective effects remains incompletely understood. This study evaluated the actions of low-dose MTX on the vascular endothelium. Methods: Human endothelial cells (EC) were studied under in vitro conditions relevant to inflammatory arthritis. These included culture in a pro-inflammatory microenvironment and exposure to fluid shear stress (FSS) using a parallel plate model. Respectively treated cells were analyzed by RNA sequencing and quantitative real-time PCR for gene expression, by immunoblotting for protein expression, by phosphokinase activity arrays, by flow cytometry for cell cycle analyses and by mass spectrometry to assess folate metabolite levels. Results: In static conditions, MTX was efficiently taken up by EC and caused cell cycle arrest concurrent with modulation of cell signaling pathways. These responses were reversed by folinic acid (FA), suggesting that OCM is a predominant target of MTX. Under FSS, MTX did not affect cell proliferation or pro-inflammatory gene expression. Exposure to FSS downregulated endothelial one carbon metabolism (OCM) as evidenced by decreased expression of key OCM genes and metabolites. Conclusion: We found that FSS significantly downregulated OCM and thereby rendered EC less susceptible to the effects of MTX treatment. The impact of shear stress on OCM suggested that MTX does not directly modulate endothelial function. The cardioprotective actions of MTX likely reflect direct actions on inflammatory cells and indirect benefit on the vascular endothelium.


Asunto(s)
Antirreumáticos , Metotrexato , Humanos , Metotrexato/uso terapéutico , Células Endoteliales , Antirreumáticos/efectos adversos , Ácido Fólico , Carbono
17.
EBioMedicine ; 95: 104762, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37586112

RESUMEN

BACKGROUND: Dolutegravir (DTG) is a recommended first-line regimen for all people with Human Immunodeficiency Virus (HIV) infection. Initial findings from Botswana, a country with no folate fortification program, showed an elevated prevalence of neural tube defects (NTDs) with peri-conceptional exposure to DTG. Here we explore whether a low folate diet influences the risk of DTG-associated foetal anomalies in a mouse model. METHODS: C57BL/6 mice fed a folate-deficient diet for 2 weeks, were mated and then randomly allocated to control (water), or 1xDTG (2.5 mg/kg), or 5xDTG (12.5 mg/kg) both administered orally with 50 mg/kg tenofovir disoproxil fumarate 33.3 mg/kg emtricitabine. Treatment was administered once daily from gestational day (GD) 0.5 to sacrifice (GD15.5). Foetuses were assessed for gross anomalies. Maternal and foetal folate levels were quantified. FINDINGS: 313 litters (103 control, 106 1xDTG, 104 5xDTG) were assessed. Viability, placental weight, and foetal weight did not differ between groups. NTDs were only observed in the DTG groups (litter rate: 0% control; 1.0% 1xDTG; 1.3% 5xDTG). Tail, abdominal wall, limb, craniofacial, and bleeding defects all occurred at higher rates in the DTG groups versus control. Compared with our previous findings on DTG usage in folate-replete mouse pregnancies, folate deficiency was associated with higher rates of several defects, including NTDs, but in the DTG groups only. We observed a severe left-right asymmetry phenotype that was more frequent in DTG groups than controls. INTERPRETATION: Maternal folate deficiency may increase the risk for DTG-associated foetal defects. Periconceptional folic acid supplementation could be considered for women with HIV taking DTG during pregnancy, particularly in countries lacking folate fortification programs. FUNDING: This project has been funded by Federal funds from the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, under Contract No. HHSN275201800001I and award #R01HD104553. LS is supported by a Tier 1 Canada Research Chair in Maternal-Child Health and HIV. HM is supported by a Junior Investigator award from the Ontario HIV Treatment Network.


Asunto(s)
Deficiencia de Ácido Fólico , Infecciones por VIH , Defectos del Tubo Neural , Femenino , Embarazo , Humanos , Ratones , Animales , Incidencia , Placenta , Ratones Endogámicos C57BL , Ácido Fólico , Deficiencia de Ácido Fólico/complicaciones , Defectos del Tubo Neural/etiología , Modelos Animales de Enfermedad , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/complicaciones , Intercambio Materno-Fetal , Feto , Ontario
18.
Dev Biol ; 355(2): 205-14, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21539830

RESUMEN

Spinal cord regenerative ability is lost with development, but the mechanisms underlying this loss are still poorly understood. In chick embryos, effective regeneration does not occur after E13, when spinal cord injury induces extensive apoptotic response and tissue damage. As initial experiments showed that treatment with a calcium chelator after spinal cord injury reduced apoptosis and cavitation, we hypothesized that developmentally regulated mediators of calcium-dependent processes in secondary injury response may contribute to loss of regenerative ability. To this purpose we screened for such changes in chick spinal cords at stages of development permissive (E11) and non-permissive (E15) for regeneration. Among the developmentally regulated calcium-dependent proteins identified was PAD3, a member of the peptidylarginine deiminase (PAD) enzyme family that converts protein arginine residues to citrulline, a process known as deimination or citrullination. This post-translational modification has not been previously associated with response to injury. Following injury, PAD3 up-regulation was greater in spinal cords injured at E15 than at E11. Consistent with these differences in gene expression, deimination was more extensive at the non-regenerating stage, E15, both in the gray and white matter. As deimination paralleled the extent of apoptosis, we investigated the effect of blocking PAD activity on cell death and deiminated-histone 3, one of the PAD targets we identified by mass-spectrometry analysis of spinal cord deiminated proteins. Treatment with the PAD inhibitor, Cl-amidine, reduced the abundance of deiminated-histone 3, consistent with inhibition of PAD activity, and significantly reduced apoptosis and tissue loss following injury at E15. Altogether, our findings identify PADs and deimination as developmentally regulated modulators of secondary injury response, and suggest that PADs might be valuable therapeutic targets for spinal cord injury.


Asunto(s)
Calcio/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Hidrolasas/metabolismo , Traumatismos de la Médula Espinal/embriología , Regeneración de la Medula Espinal/fisiología , Factores de Edad , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting , Embrión de Pollo , Cartilla de ADN/genética , Humanos , Hidrolasas/antagonistas & inhibidores , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Espectrometría de Masas , Análisis de Secuencia por Matrices de Oligonucleótidos , Ornitina/análogos & derivados , Ornitina/farmacología , Desiminasas de la Arginina Proteica , ARN Mensajero/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología
19.
Circ Res ; 105(9): 921-9, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19745165

RESUMEN

RATIONALE: Atherosclerotic lesions express matrix metalloproteinase (MMP)8, which possesses proteolytic activity on matrix proteins particularly fibrillar collagens and on nonmatrix proteins such as angiotensin (Ang) I. OBJECTIVE: We studied whether MMP8 plays a role in atherogenesis. METHODS AND RESULTS: In atherosclerosis-prone apolipoprotein E-deficient mice, inactivating MMP8 resulted in a substantial reduction in atherosclerotic lesion formation. Immunohistochemical examinations showed that atherosclerotic lesions in MMP8-deficient mice had significantly fewer macrophages but increased collagen content. In line with results of in vitro assays showing that Ang I cleavage by MMP8 generated Ang II, MMP8 knockout mice had lower Ang II levels and lower blood pressure. In addition, we found that products of Ang I cleavage by MMP8 increased vascular cell adhesion molecule (VCAM)-1 expression and that MMP8-deficient mice had reduced VCAM-1 expression in atherosclerotic lesions. Intravital microscopy analysis showed that leukocyte rolling and adhesion on vascular endothelium was reduced in MMP8 knockout mice. Furthermore, we detected an association between MMP8 gene variation and extent of coronary atherosclerosis in patients with coronary artery disease. A relationship among MMP8 gene variation, plasma VCAM-1 level, and atherosclerosis progression was also observed in a population-based, prospective study. CONCLUSIONS: These results indicate that MMP8 is an important player in atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Enfermedades de las Arterias Carótidas/enzimología , Estenosis Coronaria/enzimología , Metaloproteinasa 8 de la Matriz/metabolismo , Angiotensina II/metabolismo , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/fisiopatología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/fisiopatología , Aterosclerosis/prevención & control , Presión Sanguínea , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/prevención & control , Colágeno/metabolismo , Estenosis Coronaria/genética , Estenosis Coronaria/prevención & control , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endotelio Vascular/metabolismo , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Rodamiento de Leucocito , Macrófagos/metabolismo , Masculino , Metaloproteinasa 8 de la Matriz/deficiencia , Metaloproteinasa 8 de la Matriz/genética , Ratones , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Estudios Prospectivos , Índice de Severidad de la Enfermedad , Molécula 1 de Adhesión Celular Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA