Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 318
Filter
1.
eNeuro ; 11(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38729764

ABSTRACT

Intracerebral hemorrhage (ICH), the most common subtype of hemorrhagic stroke, leads to cognitive impairment and imposes significant psychological burdens on patients. Hippocampal neurogenesis has been shown to play an essential role in cognitive function. Our previous study has shown that tetrahydrofolate (THF) promotes the proliferation of neural stem cells (NSCs). However, the effect of THF on cognition after ICH and the underlying mechanisms remain unclear. Here, we demonstrated that administration of THF could restore cognition after ICH. Using Nestin-GFP mice, we further revealed that THF enhanced the proliferation of hippocampal NSCs and neurogenesis after ICH. Mechanistically, we found that THF could prevent ICH-induced elevated level of PTEN and decreased expressions of phosphorylated AKT and mTOR. Furthermore, conditional deletion of PTEN in NSCs of the hippocampus attenuated the inhibitory effect of ICH on the proliferation of NSCs and abnormal neurogenesis. Taken together, these results provide molecular insights into ICH-induced cognitive impairment and suggest translational clinical therapeutic strategy for hemorrhagic stroke.


Subject(s)
Cognitive Dysfunction , Hippocampus , Neural Stem Cells , Neurogenesis , PTEN Phosphohydrolase , Signal Transduction , Tetrahydrofolates , Animals , Neurogenesis/drug effects , Neurogenesis/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , PTEN Phosphohydrolase/metabolism , Male , Signal Transduction/drug effects , Signal Transduction/physiology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Tetrahydrofolates/pharmacology , Mice , Hemorrhagic Stroke , Mice, Inbred C57BL , Mice, Transgenic , Cell Proliferation/drug effects
2.
Food Funct ; 13(21): 10923-10936, 2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36205648

ABSTRACT

In the present study, mice with high-fat-diet-induced obesity were used in investigating the anti-obesity effects of an aqueous extract and isoquercitrin from Apocynum venetum L. The aqueous extract and the signal molecule isoquercitrin significantly reduced the body weight gain, food intake, water consumption, and fasting blood glucose, plasma triglyceride and total cholesterol levels of the obese mice. Furthermore, the mechanism of action of isoquercitrin was explored through RT-PCR analyses and uptake experiments of adenosine 5'-monophosphate-activated protein kinase (AMPK) and sterol regulatory-element binding protein (SREBP-1c) inhibitors and glucose. The indexes of SREBP-1c, fatty acid synthase (FAS), stearoyl-CoA desaturase-1 (SCD), and cluster of differentiation 36 (CD36) in obese mice significantly increased but returned to normal levels after the administration of isoquercitrin. Meanwhile, the anti-obesity effect of isoquercitrin was diminished by the inhibitors of AMPK and SREBP-1c. In addition, intestinal glucose uptake in normal mice was significantly inhibited after the oral administration of isoquercitrin. Moreover, 2D gel electrophoresis based proteome-wide cellular thermal shift assay (CETSA) showed that the potential target proteins of isoquercitrin were C-1-tetrahydrofolate synthase, carbonyl reductase, and glutathione S-transferase P. These results suggested that isoquercitrin produces an anti-obesity effect by targeting the above-mentioned proteins and regulating the AMPK/SREBP-1c signaling pathway and potentially prevents obesity and obesity-related metabolic disorders.


Subject(s)
Apocynum , Sterol Regulatory Element Binding Proteins , Mice , Animals , Mice, Obese , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Sterol Regulatory Element Binding Proteins/pharmacology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Apocynum/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Liver/metabolism , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Obesity/drug therapy , Obesity/metabolism , Signal Transduction , Tetrahydrofolates/metabolism , Tetrahydrofolates/pharmacology , Mice, Inbred C57BL , Lipid Metabolism
3.
Microb Biotechnol ; 15(11): 2758-2772, 2022 11.
Article in English | MEDLINE | ID: mdl-36070350

ABSTRACT

L-5-Methyltetrahydrofolate (L-5-MTHF) is the only biologically active form of folate in the human body. Production of L-5-MTHF by using microbes is an emerging consideration for green synthesis. However, microbes naturally produce only a small amount of L-5-MTHF. Here, Escherichia coli BL21(DE3) was engineered to increase the production of L-5-MTHF by overexpressing the intrinsic genes of dihydrofolate reductase and methylenetetrahydrofolate (methylene-THF) reductase, introducing the genes encoding formate-THF ligase, formyl-THF cyclohydrolase and methylene-THF dehydrogenase from the one-carbon metabolic pathway of Methylobacterium extorquens or Clostridium autoethanogenum and disrupting the gene of methionine synthase involved in the consumption and synthesis inhibition of the target product. Thus, upon its native pathway, an additional pathway for L-5-MTHF synthesis was developed in E. coli, which was further analysed and confirmed by qRT-PCR, enzyme assays and metabolite determination. After optimizing the conditions of induction time, temperature, cell density and concentration of IPTG and supplementing exogenous substances (folic acid, sodium formate and glucose) to the culture, the highest yield of 527.84 µg g-1 of dry cell weight for L-5-MTHF was obtained, which was about 11.8 folds of that of the original strain. This study paves the way for further metabolic engineering to improve the biosynthesis of L-5-MTHF in E. coli.


Subject(s)
Escherichia coli Infections , Escherichia coli , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Tetrahydrofolates/metabolism , Tetrahydrofolates/pharmacology , Folic Acid/metabolism , Folic Acid/pharmacology
4.
Oxid Med Cell Longev ; 2022: 9021474, 2022.
Article in English | MEDLINE | ID: mdl-35265266

ABSTRACT

Neural stem cell (NSC) proliferation is the initial step for NSC participating in neurorehabilitation after central nervous system (CNS) injury. During this process, oxidative stress is always involved in restricting the regenerative ability of NSC. Tetrahydrofolate (THF) is susceptible to oxidative stress and exhibits a high antioxidant activity. While its effect on NSC proliferation under oxidative stress condition remains obscure. Here, NSC were isolated from embryonic mice and identified using immunofluorescent staining. Meanwhile, the results showed that THF (5 µM and 10 µM) attenuated oxidative stress induced by 50 µM hydrogen peroxide (H2O2) in NSC using mitochondrial hydroxyl radical detection and Western blotting assays. Afterward, administration of THF markedly alleviated the inhibitory effect of oxidative stress on NSC proliferation, which was evidenced by Cell Counting Kit-8 (CCK8), neurosphere formation, and immunofluorescence of Ki67 assays. Thereafter, the results revealed that PTEN/Akt/mTOR signaling pathway played a pivotal role in counteracting oxidative stress to rescue the inhibitory effect of oxidative stress on NSC proliferation using Western blotting assays and gene knockdown techniques. Collectively, these results demonstrate that THF mitigates the inhibitory effect of oxidative stress on NSC proliferation via PTEN/Akt/mTOR signaling pathway, which provides evidence for administrating THF to potentiate the neuro-reparative capacity of NSC in the treatment of CNS diseases with the presence of oxidative stress.


Subject(s)
Neural Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Tetrahydrofolates/therapeutic use , Vitamin B Complex/therapeutic use , Animals , Cell Proliferation , Humans , Mice , Oxidative Stress , Tetrahydrofolates/pharmacology , Vitamin B Complex/pharmacology
5.
Food Chem Toxicol ; 156: 112518, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34418477

ABSTRACT

Elevated Homocysteine (Hcy) is associated with increased risk of vascular disease, but whether it induces genotoxicity to vascular endothelial cells remains unknown. Here, we conducted a comprehensive study of the genotoxicity, and unexpected anti-genotoxicity, of Hcy by cytokinesis-blocked micronucleus assay in HUVECs and erythrocyte micronucleus test in mouse bone marrow cells. Our experiments led to several important findings. First, while supraphysiological Hcy (SP-Hcy) exhibited remarkable genotoxicity, physiologically-relevant Hcy (PR-Hcy) reduced the basal genotoxicity. Second, among the metabolites of Hcy, cysteine phenocopied the anti-genotoxicity of PR-Hcy and, methionine, S-adenosylhomocysteine and H2S phenocopied the genotoxicity of SP-Hcy. Third, the genotoxicity of SP-Hcy was mitigated by vitamin B6, Fe2+ and Cu2+, but was exacerbated by N-acetylcysteine. Fourth, under pre-, co- or post-treatment protocol, both SP-Hcy and PR-Hcy attenuated the genotoxicity of cisplatin, mitomycin-C, nocodazole or deoxycholate. Finally, 100 and 250 mg/kg Hcy ameliorated cisplatin-induced genotoxicity in bone marrow cells of CF-1 and Kunming mice. Our results suggest that genotoxicity may be one mechanism through which Hcy confers an increased risk for vascular disease, but more importantly, they challenge the long-standing paradigm that Hcy is always harmful to human health. Our study calls for a more systematic effort in understanding the molecular mechanisms underlying the anti-genotoxicity of Hcy.


Subject(s)
Bone Marrow Cells/drug effects , Homocysteine/toxicity , Animals , Copper/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Iron/pharmacology , Male , Mice , Mutagenicity Tests , Tetrahydrofolates/pharmacology , Vitamin B 6/pharmacology
6.
Nutrients ; 13(5)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925570

ABSTRACT

Supplementation with [6S]-5-methyltetrahydrofolic acid (MTHF) is recommended as an alternative to folic acid (FA) in prenatal supplements. This study compared equimolar gestational FA and MTHF diets on energy regulation of female offspring. Wistar rats were fed an AIN-93G diet with recommended (2 mg/kg diet) or 5-fold (5X) intakes of MTHF or FA. At weaning, female offspring were fed a 45% fat diet until 19 weeks. The 5X-MTHF offspring had higher body weight (>15%), food intake (8%), light-cycle energy expenditure, and lower activity compared to 5X-FA offspring (p < 0.05). Both the 5X offspring had higher plasma levels of the anorectic hormone leptin at birth (60%) and at 19 weeks (40%), and lower liver weight and total liver lipids compared to the 1X offspring (p < 0.05). Hypothalamic mRNA expression of leptin receptor (ObRb) was lower, and of suppressor of cytokine signaling-3 (Socs3) was higher in the 5X-MTHF offspring (p < 0.05), suggesting central leptin dysregulation. In contrast, the 5X-FA offspring had higher expression of genes encoding for dopamine and GABA- neurotransmitter receptors (p < 0.01), consistent with their phenotype and reduced food intake. When fed folate diets at the requirement level, no differences were found due to form in the offspring. We conclude that MTHF compared to FA consumed at high levels in the gestational diets program central and peripheral mechanisms to favour increased weight gain in the offspring. These pre-clinical findings caution against high gestational intakes of folates of either form and encourage clinical trials examining their long-term health effects when consumed during pregnancy.


Subject(s)
Body Weight/drug effects , Diet/methods , Energy Intake/drug effects , Feeding Behavior/drug effects , Folic Acid/pharmacology , Tetrahydrofolates/pharmacology , Animals , Animals, Newborn , Energy Metabolism/drug effects , Female , Folic Acid/administration & dosage , Mice , Models, Animal , Pregnancy , Rats, Wistar , Tetrahydrofolates/administration & dosage , Vitamin B Complex/administration & dosage , Vitamin B Complex/pharmacology
7.
Oncol Rep ; 44(1): 77-90, 2020 07.
Article in English | MEDLINE | ID: mdl-32627002

ABSTRACT

Protein/nucleic acid deglycase DJ­1 (DJ­1) is a 20­kDa conserved protein, which belongs to the DJ­1/ThiJ/Pfp â…  protein superfamily. Immunohistochemistry was performed to investigate the expression of DJ­1 in a colorectal cancer (CRC) tissue microarray containing tumor and corresponding adjacent normal tissues. In the present study, DJ­1 expression was significantly upregulated in CRC cells and tissues, compared with that in normal colon cells and adjacent normal tissues, respectively. In addition, patients with high DJ­1 expression levels had a worse overall survival (OS) compared with patients with low expression levels. Multivariate Cox regression analysis revealed that high DJ­1 expression levels was an independent prognostic factor for patients with CRC. Moreover, DJ­1 was able to regulate the PI3K/Akt/p27/cyclin E and PI3K/Akt/mTOR signaling pathways to promote CRC cell growth and metastasis in vitro and in vivo. In addition, DJ­1 regulated the NF­κB/Snail signaling pathway to induce CRC cell epithelial­mesenchymal transition to promote migration and invasion. Notably, patients receiving LFP treatment (oxaliplatin, 5­FU and tetrahydrofolate) had an increased OS compared with patients who underwent only surgery and low DJ­1 expression levels. The findings from the present study suggest that DJ­1 may serve as a promising prognostic marker and predicts chemotherapy efficacy in patients with CRC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Protein Deglycase DJ-1/genetics , Protein Deglycase DJ-1/metabolism , Up-Regulation , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis , Oxaliplatin/administration & dosage , Oxaliplatin/pharmacology , Prognosis , Signal Transduction , Survival Analysis , Tetrahydrofolates/administration & dosage , Tetrahydrofolates/pharmacology , Xenograft Model Antitumor Assays
8.
J Cell Mol Med ; 24(12): 6704-6715, 2020 06.
Article in English | MEDLINE | ID: mdl-32391636

ABSTRACT

Patients with relapsed/refractory Burkitt's lymphoma (BL) have a dismal prognosis. Current research efforts aim to increase cure rates by identifying high-risk patients in need of more intensive or novel therapy. The 8q24 chromosomal translocation of the c-Myc gene, a main molecular marker of BL, is related to the metabolism by regulating phosphoribosyl pyrophosphate synthetase 2 (PRPS2). In our study, BL showed significant resistance to thiopurines. PRPS2 homologous isoenzyme, PRPS1, was demonstrated to play the main role in thiopurine resistance. c-Myc did not have direct effects on thiopurine resistance in BL for only driving PRPS2. PRPS1 wild type (WT) showed different resistance to 6-mercaptopurine (6-mp) in different metabolic cells because it could be inhibited by adenosine diphosphate or guanosine diphosphate negative feedback. PRPS1 A190T mutant could dramatically increase thiopurine resistance in BL. The interim analysis of the Treatment Regimen for Children or Adolescent with mature B cell non-Hodgkin's lymphoma in China (CCCG-B-NHL-2015 study) confirms the value of high-dose methotrexate (MTX) and cytarabine (ARA-C) in high-risk paediatric patients with BL. However, there remains a subgroup of patients with lactate dehydrogenase higher than four times of the normal value (4N) for whom novel treatments are needed. Notably, we found that the combination of thiopurines and the phosphoribosylglycinamide formyltransferase (GART) inhibitor lometrexol could serve as a therapeutic strategy to overcome thiopurine resistance in BL.


Subject(s)
Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Drug Resistance, Neoplasm/genetics , Mercaptopurine/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Ribose-Phosphate Pyrophosphokinase/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , HEK293 Cells , Humans , Mercaptopurine/pharmacology , Mutation/genetics , Nucleotides/metabolism , Tetrahydrofolates/pharmacology , Tetrahydrofolates/therapeutic use
9.
Neurobiol Dis ; 132: 104613, 2019 12.
Article in English | MEDLINE | ID: mdl-31525435

ABSTRACT

The prevalence of stroke increases with age and the ability to absorb all nutrients from our diets decreases with age. Nutrition is a modifiable risk factor for stroke, which is a leading cause of death and disability in world-wide. Deficiencies in one­carbon metabolism, including in methyltetrahydrofolate reductase (MTHFR), have been linked to increased risk of stroke. The Mthfr+/- mice mouse model mimic the phenotype of the MTHFR677C➔T polymorphism, such as elevated levels of homocystine. Using this mouse model, the aim of this study was to investigate the impact of dietary supplementation with 5-methylTHF, vitamin B12, and choline after ischemic stroke. Male Mthfr+/- and wildtype littermate control mice were aged (~1.5-year-old) and were placed on control diet (CD) 4-weeks prior to sensorimotor cortex damage using photothrombosis (PT), a model for ischemic stroke. Post-operatively, one group of Mthfr+/- and wildtype littermate mice were placed on 5-methylTHF, vitamin B12, and choline supplemented diet (SD). Four weeks after PT and SD motor function was assessed using the accelerating rotarod, forepaw asymmetry, and ladder beam walking tasks. Total homocysteine and cysteine levels were measured in blood. Brain tissue was processed to assess lesion volume and investigate biochemical and molecular changes. After PT and SD, Mthfr+/- mice were able to stay on the accelerating rotarod longer and used their impaired forepaw to explore more when compared to CD animals. Furthermore, total homocysteine levels in plasma and lesion volume were reduced in Mthfr+/+ and Mthfr+/- SD mice. Within the damage site, there were reduced levels of apoptotic cell death and increased neuroprotective cellular response in the brains of SD treated Mthfr+/- mice. This study reveals a critical role for one­carbon supplementation, with 5-methylTHF, vitamin B12, and choline, in supporting improvement after ischemic stroke damage.


Subject(s)
Choline/pharmacology , Dietary Supplements , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Stroke/physiopathology , Tetrahydrofolates/pharmacology , Vitamin B 12/pharmacology , Aging , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Mice, Inbred C57BL , Recovery of Function/drug effects
10.
J Nutr ; 148(6): 885-890, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29878267

ABSTRACT

Background: Folic acid fortification of grains is mandated in many countries to prevent neural tube defects. Concerns regarding excessive intakes of folic acid have been raised. A synthetic analog of the circulating form of folate, l-5-methyltetrahydrofolate (l-5-MTHF), may be a potential alternative. Objective: The objective of this study was to determine the effects of folic acid or l-5-MTHF supplementation on blood folate concentrations, methyl nutrient metabolites, and DNA methylation in women living in Malaysia, where there is no mandatory fortification policy. Methods: In a 12-wk, randomized, placebo-controlled intervention trial, healthy Malaysian women (n = 142, aged 20-45 y) were randomly assigned to receive 1 of the following supplements daily: 1 mg (2.27 µmol) folic acid, 1.13 mg (2.27 µmol) l-5-MTHF, or a placebo. The primary outcomes were plasma and RBC folate and vitamin B-12 concentrations. Secondary outcomes included plasma total homocysteine, total cysteine, methionine, betaine, and choline concentrations and monocyte long interspersed nuclear element-1 (LINE-1) methylation. Results: The folic acid and l-5-MTHF groups had higher (P < 0.001) RBC folate (mean ± SD: 1498 ± 580 and 1951 ± 496 nmol/L, respectively) and plasma folate [median (25th, 75th percentiles): 40.1 nmol/L (24.9, 52.7 nmol/L) and 52.0 nmol/L (42.7, 73.1 nmol/L), respectively] concentrations compared with RBC folate (958 ± 345 nmol/L) and plasma folate [12.6 nmol/L (8.80, 17.0 nmol/L)] concentrations in the placebo group at 12 wk. The l-5-MTHF group had higher RBC folate (1951 ± 496 nmol/L; P = 0.003) and plasma folate [52.0 nmol/L (42.7, 73.1 nmol/L); P = 0.023] at 12 wk than did the folic acid group [RBC folate, 1498 ± 580 nmol/L; plasma folate, 40.1 nmol/L (24.9, 52.7 nmol/L)]. The folic acid and l-5-MTHF groups had 17% and 15%, respectively, lower (P < 0.001) plasma total homocysteine concentrations than did the placebo group at 12 wk; there were no differences between the folic acid and l-5-MTHF groups. No differences in plasma vitamin B-12, total cysteine, methionine, betaine, and choline and monocyte LINE-1 methylation were observed. Conclusion: These findings suggest differential effects of l-5-MTHF compared with folic acid supplementation on blood folate concentrations but no differences on plasma total homocysteine lowering in Malaysian women. This trial was registered at clinicaltrials.gov as NCT01584050.


Subject(s)
Folic Acid/administration & dosage , Folic Acid/blood , Tetrahydrofolates/administration & dosage , Tetrahydrofolates/pharmacology , Adult , Dietary Supplements , Double-Blind Method , Female , Folic Acid/pharmacology , Humans , Malaysia , Young Adult
11.
Mol Cell ; 69(4): 610-621.e5, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29452640

ABSTRACT

Upon glucose restriction, eukaryotic cells upregulate oxidative metabolism to maintain homeostasis. Using genetic screens, we find that the mitochondrial serine hydroxymethyltransferase (SHMT2) is required for robust mitochondrial oxygen consumption and low glucose proliferation. SHMT2 catalyzes the first step in mitochondrial one-carbon metabolism, which, particularly in proliferating cells, produces tetrahydrofolate (THF)-conjugated one-carbon units used in cytoplasmic reactions despite the presence of a parallel cytoplasmic pathway. Impairing cytoplasmic one-carbon metabolism or blocking efflux of one-carbon units from mitochondria does not phenocopy SHMT2 loss, indicating that a mitochondrial THF cofactor is responsible for the observed phenotype. The enzyme MTFMT utilizes one such cofactor, 10-formyl THF, producing formylmethionyl-tRNAs, specialized initiator tRNAs necessary for proper translation of mitochondrially encoded proteins. Accordingly, SHMT2 null cells specifically fail to maintain formylmethionyl-tRNA pools and mitochondrially encoded proteins, phenotypes similar to those observed in MTFMT-deficient patients. These findings provide a rationale for maintaining a compartmentalized one-carbon pathway in mitochondria.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Glycine Hydroxymethyltransferase/metabolism , Mitochondria/genetics , Peptide Chain Initiation, Translational , RNA, Transfer, Met/chemistry , Serine/chemistry , Animals , Apoptosis , Breast Neoplasms/metabolism , CRISPR-Cas Systems , Cell Proliferation , Cytosol/metabolism , Female , Glycine Hydroxymethyltransferase/antagonists & inhibitors , Glycine Hydroxymethyltransferase/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/metabolism , Protein Processing, Post-Translational , RNA, Transfer, Met/genetics , RNA, Transfer, Met/metabolism , Serine/genetics , Serine/metabolism , Tetrahydrofolates/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Mol Psychiatry ; 23(2): 316-322, 2018 02.
Article in English | MEDLINE | ID: mdl-28289280

ABSTRACT

Folic acid supplementation confers modest benefit in schizophrenia, but its effectiveness is influenced by common genetic variants in the folate pathway that hinder conversion to its active form. We examined physiological and clinical effects of l-methylfolate, the fully reduced and bioactive form of folate, in schizophrenia. In this randomized, double-blind trial, outpatients with schizophrenia (n=55) received l-methylfolate 15 mg or placebo for 12 weeks. Patients were maintained on stable doses of antipsychotic medications. The pre-defined primary outcome was change in plasma methylfolate at 12 weeks. Secondary outcomes included change in symptoms (Positive and Negative Syndrome Scale (PANSS), Scale for Assessment of Negative Symptoms, Calgary Depression Scale for Schizophrenia), cognition (Measurement and Treatment Research to Improve Cognition in Schizophrenia composite) and three complementary magnetic resonance imaging measures (working memory-related activation, resting connectivity, cortical thickness). Primary, mixed model, intent-to-treat analyses covaried for six genetic variants in the folate pathway previously associated with symptom severity and/or response to folate supplementation. Analyses were repeated without covariates to evaluate dependence on genotype. Compared with placebo, l-methylfolate increased plasma methylfolate levels (d=1.00, P=0.0009) and improved PANSS Total (d=0.61, P=0.03) as well as PANSS Negative and General Psychopathology subscales. Although PANSS Total and General Psychopathology changes were influenced by genotype, significant PANSS Negative changes occurred regardless of genotype. No treatment differences were seen in other symptom rating scales or cognitive composite scores. Patients receiving l-methylfolate exhibited convergent changes in ventromedial prefrontal physiology, including increased task-induced deactivation, altered limbic connectivity and increased cortical thickness. In conclusion, l-methylfolate supplementation was associated with salutary physiological changes and selective symptomatic improvement in this study of schizophrenia patients, warranting larger clinical trials. ClinicalTrials.gov, NCT01091506.


Subject(s)
Schizophrenia/drug therapy , Tetrahydrofolates/pharmacology , Adult , Antipsychotic Agents/therapeutic use , Cognition/drug effects , Double-Blind Method , Female , Folic Acid/metabolism , Folic Acid/pharmacology , Humans , Male , Middle Aged , Psychiatric Status Rating Scales , Tetrahydrofolates/therapeutic use , Treatment Outcome
13.
Plant Physiol ; 174(2): 1274-1284, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28450424

ABSTRACT

Folates, termed from tetrahydrofolate (THF) and its derivatives, function as coenzymes in one-carbon transfer reactions and play a central role in synthesis of nucleotides and amino acids. Dysfunction of cellular folate metabolism leads to serious defects in plant development; however, the molecular mechanisms of folate-mediated cellular modifications and physiological responses in plants are still largely unclear. Here, we reported that THF controls flowering time by adjusting DNA methylation-regulated gene expression in Arabidopsis (Arabidopsis thaliana). Wild-type seedlings supplied with THF as well as the high endogenous THF content mutant dihydrofolate synthetase folypoly-Glu synthetase homolog B exhibited significant up-regulation of the flowering repressor of Flowering Wageningen and thereby delaying floral transition in a dose-dependent manner. Genome-wide transcripts and DNA methylation profiling revealed that THF reduces DNA methylation so as to manipulate gene expression activity. Moreover, in accompaniment with elevated cellular ratios between monoglutamylated and polyglutamylated folates under increased THF levels, the content of S-adenosylhomo-Cys, a competitive inhibitor of methyltransferases, was obviously higher, indicating that enhanced THF accumulation may disturb cellular homeostasis of the concerted reactions between folate polyglutamylation and folate-dependent DNA methylation. In addition, we found that the loss-of-function mutant of CG DNA methyltransferase MET1 displayed much less responsiveness to THF-associated flowering time alteration. Taken together, our studies revealed a novel regulatory role of THF on epigenetic silencing, which will shed lights on the understanding of interrelations in folate homeostasis, epigenetic variation, and flowering control in plants.


Subject(s)
Arabidopsis/genetics , Arabidopsis/physiology , Epigenesis, Genetic/drug effects , Flowers/genetics , Gene Silencing/drug effects , Tetrahydrofolates/pharmacology , DNA Methylation/drug effects , DNA Methylation/genetics , Flowers/drug effects , Gene Expression Profiling , Gene Expression Regulation, Plant/drug effects , Genome, Plant , Polyglutamic Acid/metabolism
14.
Psychiatr Genet ; 27(3): 116-119, 2017 06.
Article in English | MEDLINE | ID: mdl-28272116

ABSTRACT

Errors in folate metabolism may play a role in the pathology of autism spectrum disorders because of increased vulnerability to oxidative stress. We report a case where L-methylfolate supplementation improved symptoms of aggression and disruptive behavior in a child with autism who tested positive for the C677TT allele of the methyltetrahydrofolate reductase enzyme gene. To our knowledge, this is the first report of L-methylfolate administration in this situation. Further controlled studies of L-methylfolate in this population are warranted.


Subject(s)
Autistic Disorder/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Tetrahydrofolates/pharmacology , Autistic Disorder/complications , Autistic Disorder/metabolism , Child , Dietary Supplements , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Oxidoreductases , Selective Serotonin Reuptake Inhibitors/therapeutic use , Tetrahydrofolates/metabolism
15.
World J Microbiol Biotechnol ; 32(12): 196, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27734279

ABSTRACT

When exposed to mixtures of glucose and fructose, as occurs during the fermentation of grape juice into wine, Saccharomyces cerevisiae uses these sugars at different rates. Moreover, glucose and fructose are transported by the same hexose transporters (HXT), which present a greater affinity for glucose, so that late in fermentation, fructose becomes the predominant sugar. Only a few commercial fermentation activators are available to optimally solve the problems this entails. The aim of this study was to investigate the relation between HXT3 gene expression and fructose/glucose discrepancy in two different media inoculated with a commercial wine strain of S. cerevisiae in the presence of three metabolic activators. Fermentation kinetics, vitality and major metabolites were also measured. Rehydration with ergosterol improved the area under the curve and the growth rate (µ max ) in both studied media. Also, the fructose/glucose discrepancy values were improved with all activator treatments, highlighting rehydration in the presence of ascorbic acid. The yeast rehydration process was demonstrated to influence HXT3 expression under the studied conditions. Tetrahydrofolic acid treatment greatly influenced HXT3 gene expression, especially on the 12th day of the fermentation process. To a lesser extent, ergosterol and ascorbic acid also improved this parameter.


Subject(s)
Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Wine/microbiology , Ascorbic Acid/pharmacology , Ergosterol/pharmacology , Fermentation , Fructose/metabolism , Gene Expression Regulation, Fungal/drug effects , Glucose/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Tetrahydrofolates/pharmacology , Wine/analysis
16.
Biotechnol Prog ; 32(6): 1539-1546, 2016 11.
Article in English | MEDLINE | ID: mdl-27578320

ABSTRACT

Adaptation of dihydrofolate reductase (DHFR)-deficient Chinese hamster ovary (CHO) DG44 cells to chemically defined suspension culture conditions is a time-consuming and labor-intensive process because nonadapted DHFR-deficient CHO DG44 cells normally show poor growth in chemically defined medium (CDM). We examined the effects of folate derivatives, ribonucleotides, and nucleobases on the growth of suspension-adapted DHFR-deficient CHO DG44 cells in CDM. Among the tested additives, tetrahydrofolate (THF) was identified as an effective component for increasing cell growth. THF supplementation in the range of 0.2-359 µM enhanced cell growth in in-house CDM. Addition of 3.6 µM THF to in-house CDM resulted in a more than 2.5-fold increase in maximum viable cell density. Moreover, supplementation of six different commercial CDMs with 3.6 µM THF yielded up to 2.9-fold enhancement of maximum viable cell density. An anchorage- and serum-dependent DHFR-deficient CHO DG44 cell line was adapted within two consecutive passages to suspension growth in in-house CDM supplemented with 3.6 µM THF. These data indicate that supplementation of chemically defined cell culture media with greater than 0.2 µM THF can help achieve a high density of suspension-adapted DHFR-deficient CHO DG44 cells and may facilitate rapid adaptation of nonadapted DHFR-deficient CHO DG44 cells to suspension culture. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 32:1539-1546, 2016.


Subject(s)
Culture Media/pharmacology , Tetrahydrofolate Dehydrogenase/metabolism , Tetrahydrofolates/pharmacology , Animals , CHO Cells , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cricetulus , Culture Media/chemistry , Dose-Response Relationship, Drug , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/deficiency , Tetrahydrofolates/chemistry
17.
Asian J Psychiatr ; 22: 74-5, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27520898

ABSTRACT

The 5,10-methylenetetrahydrofolate reductase (MTHFR) gene plays a central role in folate metabolism. Many studies have demonstrated an association between MTHFR C677 T variant with depression, schizophrenia and bipolar disorder as one of them being comorbid to other. This has justified the use of folate supplement in psychiatric disorders mainly depression but still not in various other comorbid complex psychiatric disorders. Here we have tried to show how the l-methylfolate in conjunction with the conventional psychotropic drugs can be useful in a state of such complex psychiatric phenomenon and comorbid diagnosis with genetic polymorphism of MTHFR C677 T mutation.


Subject(s)
Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Bipolar Disorder/drug therapy , Depressive Disorder, Treatment-Resistant/drug therapy , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Obsessive-Compulsive Disorder/drug therapy , Tetrahydrofolates/pharmacology , Vitamin B Complex/pharmacology , Adult , Antidepressive Agents/administration & dosage , Antipsychotic Agents/administration & dosage , Aripiprazole/administration & dosage , Aripiprazole/pharmacology , Drug Therapy, Combination , Fluoxetine/administration & dosage , Fluoxetine/pharmacology , Humans , Lithium Compounds/administration & dosage , Lithium Compounds/pharmacology , Male , Polymorphism, Genetic , Tetrahydrofolates/administration & dosage , Vitamin B Complex/administration & dosage
18.
Nat Commun ; 7: 11640, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27291711

ABSTRACT

DNA methylation is an epigenetic mechanism that has important functions in transcriptional silencing and is associated with repressive histone methylation (H3K9me). To further investigate silencing mechanisms, we screened a mutagenized Arabidopsis thaliana population for expression of SDCpro-GFP, redundantly controlled by DNA methyltransferases DRM2 and CMT3. Here, we identify the hypomorphic mutant mthfd1-1, carrying a mutation (R175Q) in the cytoplasmic bifunctional methylenetetrahydrofolate dehydrogenase/methenyltetrahydrofolate cyclohydrolase (MTHFD1). Decreased levels of oxidized tetrahydrofolates in mthfd1-1 and lethality of loss-of-function demonstrate the essential enzymatic role of MTHFD1 in Arabidopsis. Accumulation of homocysteine and S-adenosylhomocysteine, genome-wide DNA hypomethylation, loss of H3K9me and transposon derepression indicate that S-adenosylmethionine-dependent transmethylation is inhibited in mthfd1-1. Comparative analysis of DNA methylation revealed that the CMT3 and CMT2 pathways involving positive feedback with H3K9me are mostly affected. Our work highlights the sensitivity of epigenetic networks to one-carbon metabolism due to their common S-adenosylmethionine-dependent transmethylation and has implications for human MTHFD1-associated diseases.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , DNA Methylation/genetics , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Arabidopsis Proteins/genetics , Cytoplasm/drug effects , Cytoplasm/metabolism , DNA Demethylation , Epigenesis, Genetic , Folic Acid/metabolism , Gene Expression Regulation, Plant/drug effects , Gene Silencing , Green Fluorescent Proteins/metabolism , Histones/metabolism , Homeostasis/drug effects , Lysine/metabolism , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methionine/pharmacology , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Models, Biological , Mutation/genetics , Protein Transport/drug effects , S-Adenosylmethionine/metabolism , Tetrahydrofolates/pharmacology
19.
Am J Clin Nutr ; 103(1): 115-27, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26561619

ABSTRACT

BACKGROUND: During fetal and perinatal periods, many nutrients, such as long-chain polyunsaturated fatty acids [contained in fish oil (FO)] and folate, are important in achieving normal brain development. Several studies have shown the benefits of early nutrition on children's neurocognitive development. However, the evidence with regard to the attention system is scarce. OBJECTIVES: The aim of this study was to analyze the long-term effects of FO, 5-methyltetrahydrofolate (5-MTHF), or FO+5-MTHF prenatal supplementation on attention networks. DESIGN: Participants were 136 children born to mothers from the NUHEAL (Nutraceuticals for a Healthy Life) project (randomly assigned to receive FO and/or 5-MTHF or placebo prenatal supplementation) who were recalled for a new examination 8.5 y later. The response conflict-resolution ability (using congruent and incongruent conditions)), alerting, and spatial orienting of attention were evaluated with behavioral measures (Attention Network Test), electroencephalography/event-related potentials (ERPs), and standardized low-resolution brain electromagnetic tomography (sLORETA). RESULTS: Children born to mothers supplemented with 5-MTHF alone solved the response conflict more quickly than did the placebo and the FO+5-MTHF groups (all P < 0.05). Differences between ERP amplitudes for the conflict conditions were also observed. sLORETA analysis showed higher activation of the right midcingulate cortex for the incongruent condition. In addition, a significant slowing down of response speed depending on the warning cue in the 5-MTHF and FO groups was observed. CONCLUSIONS: Folate supplementation during pregnancy, rather than FO or FO+5-MTHF supplementation, improves children's ability to solve response conflicts. This advantage seems to be based on the higher activation of the midcingulate cortex, indicating that early nutrition influences the functionality of specific brain areas involved in executive functions. This trial was registered at clinicaltrials.gov as NCT01180933.


Subject(s)
Attention/physiology , Brain/growth & development , Dietary Fats, Unsaturated/pharmacology , Dietary Supplements , Executive Function/physiology , Prenatal Nutritional Physiological Phenomena , Tetrahydrofolates/pharmacology , Adult , Brain/physiology , Child , Child Development , Double-Blind Method , Female , Fetal Development , Fish Oils/pharmacology , Folic Acid/pharmacology , Humans , Male , Pregnancy , Vitamin B Complex/pharmacology
20.
Nutr Cancer ; 67(4): 706-11, 2015.
Article in English | MEDLINE | ID: mdl-25802929

ABSTRACT

Folic acid (FA) consumption at high levels has been associated with colon cancer risk. Several mechanisms have been proposed to explain this association. The Notch signal pathway has been implicated in the regulation of cellular proliferation. Our aim was to demonstrate that high concentrations of FA or its reduced form, 5-methyltetrahydrofolic acid (5-MTHF), increase colorectal carcinoma HT29 cell proliferation through an increase of Notch1 activation and to prove if the inhibition of Notch1 activation by gamma secretase inhibitor, reduce the effect of folic acid. HT29 cells were cultured in high (400 nM), low (20 nM), or 0 nM FA or 5-MTHF concentrations during 96 h with or without DAPT (gamma secretase inhibitor). Cell proliferation was determined by the methylthiazole tetrazolium method, and Notch1-intracellular domain (NICD) was analyzed by flow cytometry. HT29 cells exposed to 400 nM FA or 5-MTHF showed higher proliferation rate than those exposed to 20 nM of FA or 5-MTHF (P < 0.01) during 96 h. NICD expression increased at higher FA or 5-MTHF concentrations compared with lower concentrations (P < 0.01). This effect on proliferation was partially reversible when we blocked Notch1 activation with the inhibitor of γ-secretase (P < 0.05).These data suggest that high concentration of FA and 5-MTHF induce HT29 cell proliferation activating Notch1 pathway.


Subject(s)
Cell Proliferation/drug effects , Folic Acid/pharmacology , Receptor, Notch1/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Colonic Neoplasms/pathology , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , HT29 Cells , Humans , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction , Tetrahydrofolates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL