Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 143
1.
Development ; 151(10)2024 May 15.
Article En | MEDLINE | ID: mdl-38682273

Neurulation is a highly synchronized biomechanical process leading to the formation of the brain and spinal cord, and its failure leads to neural tube defects (NTDs). Although we are rapidly learning the genetic mechanisms underlying NTDs, the biomechanical aspects are largely unknown. To understand the correlation between NTDs and tissue stiffness during neural tube closure (NTC), we imaged an NTD murine model using optical coherence tomography (OCT), Brillouin microscopy and confocal fluorescence microscopy. Here, we associate structural information from OCT with local stiffness from the Brillouin signal of embryos undergoing neurulation. The stiffness of neuroepithelial tissues in Mthfd1l null embryos was significantly lower than that of wild-type embryos. Additionally, exogenous formate supplementation improved tissue stiffness and gross embryonic morphology in nullizygous and heterozygous embryos. Our results demonstrate the significance of proper tissue stiffness in normal NTC and pave the way for future studies on the mechanobiology of normal and abnormal embryonic development.


Neural Tube Defects , Neural Tube , Neurulation , Tomography, Optical Coherence , Animals , Tomography, Optical Coherence/methods , Mice , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Neural Tube Defects/pathology , Neural Tube/metabolism , Neurulation/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Formates/metabolism , Embryo, Mammalian/metabolism , Female , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Mutation/genetics , Biomechanical Phenomena , Microscopy, Confocal , Mice, Knockout
2.
Int J Mol Sci ; 24(16)2023 Aug 08.
Article En | MEDLINE | ID: mdl-37628752

We investigated the association between methylenetetrahydrofolate reductase (gene MTHFR 677C>T, rs1801133), 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR 2756A>G, rs1805087), and methylenetetrahydrofolate dehydrogenase, cyclohydrolase and formyltetrahydrofolate synthetase 1 (gene MTHFD1 1958G>A, rs2236225)-well-studied functional variants involved in one-carbon metabolism-and gynecologic cancer risk, and the interaction between these polymorphisms and depression. A total of 200 gynecologic cancer cases and 240 healthy controls were recruited to participate in this study. Three single-nucleotide variants (SNVs) (rs1801133, rs1805087, rs2236225) were genotyped using the PCR-restriction fragment length polymorphism method. Depression was assessed in all patients using the Hamilton Depression Scale. Depression was statistically significantly more frequent in women with gynecologic cancers (69.5% vs. 34.2% in controls, p < 0.001). MTHFD1 rs2236225 was associated with an increased risk of gynecologic cancers (in dominant OR = 1.53, p = 0.033, and in log-additive models OR = 1.37, p = 0.024). Moreover, an association was found between depression risk and MTHFR rs1801133 genotypes in the controls but not in women with gynecologic cancers (in codominant model CC vs. TT: OR = 3.39, 95%: 1.49-7.74, p = 0.011). Cancers of the female reproductive system are associated with the occurrence of depression, and ovarian cancer may be associated with the rs2236225 variant of the MTHFD1 gene. In addition, in healthy aging women in the Polish population, the rs1801133 variant of the MTHFR gene is associated with depression.


Formate-Tetrahydrofolate Ligase , Genital Neoplasms, Female , Female , Humans , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Depression , Genital Neoplasms, Female/genetics , Carbon , Minor Histocompatibility Antigens/genetics , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase
3.
Database (Oxford) ; 20222022 06 16.
Article En | MEDLINE | ID: mdl-35708586

AcetoBase is a public repository and database of formyltetrahydrofolate synthetase (FTHFS) sequences. It is the first systematic collection of bacterial FTHFS nucleotide and protein sequences from genomes and metagenome-assembled genomes and of sequences generated by clone library sequencing. At its publication in 2019, AcetoBase (Version 1) was also the first database to establish connections between the FTHFS gene, the Wood-Ljungdahl pathway and 16S ribosomal RNA genes. Since the publication of AcetoBase, there have been significant improvements in the taxonomy of many bacterial lineages and accessibility/availability of public genomics and metagenomics data. The update to the AcetoBase reference database described here (Version 2) provides new sequence data and taxonomy, along with improvements in web functionality and user interface. The evaluation of this latest update by re-analysis of publicly accessible FTHFS amplicon sequencing data previously analysed with AcetoBase Version 1 revealed significant improvements in the taxonomic assignment of FTHFS sequences. Database URL: https://acetobase.molbio.slu.se.


Formate-Tetrahydrofolate Ligase , Anaerobiosis , Bacteria/genetics , Formate-Tetrahydrofolate Ligase/genetics , Metagenome , Metagenomics , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
4.
Article En | MEDLINE | ID: mdl-35346016

Hyperhomocysteinemia is an independent risk factor for atherosclerosis, even in early childhood. A mutation in genes that code homocysteine metabolism enzymes or deficiency of specific vitamin cofactors may cause hyperhomocysteinemia. Vitamin B complex has been correlated with serum homocysteine levels. Any abnormality in its metabolism or nutritional deficiency may lead to hyperhomocysteinemia. Both vitamin B complex and homocysteine levels are partly genetically determined. Specifically, the most studied polymorphism is 677T-C in exon 5 of the 5,10- methylenetetrahydrofolate reductase (MTHFR) gene, which plays an important role in folate's metabolism. This polymorphism has been shown to be correlated with hypertension and cardiovascular disease. Polymorphisms in methylenetetrahydrofolate dehydrogenase (NADP+ dependent) 1-like (MTHFD1L) gene have also been correlated with increased risk for coronary artery disease. Other common serious polymorphisms regard the area with high linkage disequilibrium, including the neuroblastoma breakpoint family, NBPF3 gene, and ~ 12-50 kb upstream of the tissue nonspecific alkaline phosphatase gene. Finally, the polymorphisms which have been mostly associated with vitamin B12 concentration are the rs11254363 polymorphism at intron 52 of the intrinsic factor vitamin B12 receptor of the CUBN and the rs526934 polymorphism at intron 8 of transcobalamin I. To sum up, several polymorphisms have already been associated with vitamin B complexes and therefore homocysteine level, highlighting the complex nature of vitamin B genetics.


Cardiovascular Diseases , Hyperhomocysteinemia , Vitamin B Complex , Aminohydrolases/genetics , Cardiovascular Diseases/complications , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/genetics , Homocysteine , Humans , Hyperhomocysteinemia/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Multienzyme Complexes/genetics , Neoplasm Proteins/genetics , Polymorphism, Genetic , Receptors, Cell Surface/genetics , Vitamin B 12/metabolism
5.
Biosci Rep ; 42(1)2022 01 28.
Article En | MEDLINE | ID: mdl-34908119

BACKGROUND: Defects in methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L) expression have earlier been examined in only a few human cancers. OBJECTIVES: Multi-omics profiling of MTHFD1L as a shared biomarker in distinct subtypes of human cancers. METHODS: In the current study, for the multi-omics analysis of MTHFD1L in 24 major subtypes of human cancers, a comprehensive in silico approach was adopted to mine different open access online databases including UALCAN, Kaplan-Meier (KM) plotter, LOGpc, GEPIA, Human Protein Atlas (HPA), Gene Expression across Normal and Tumor tissue (GENT2), MEXPRESS, cBioportal, STRING, DAVID, TIMER, and Comparative Toxicogenomics Database (CTD). RESULTS: We noticed that the expression of MTHFD1L was significantly higher in all the analyzed 24 subtypes of human cancers as compared with the normal controls. Moreover, MTHDF1L overexpression was also found to be significantly associated with the reduced overall survival (OS) duration of Bladder urothelial cancer (BLCA), Head and neck cancer (HNSC), Kidney renal papillary cell carcinoma (KIRP), Lung adenocarcinoma (LUAD), and Uterine corpus endometrial carcinoma (UCEC). This implies that MTHFD1L plays a significant role in the development and progression of these cancers. We further noticed that MTHFD1L was also overexpressed in BLCA, HNSC, KIRP, LUAD, and UCEC patients of different clinicopathological features. Pathways enrichment analysis revealed the involvement of MTHFD1L-associated genes in five diverse pathways. We also explored few interesting correlations between MTHFD1L expression and its promoter methylation, genetic alterations, CNVs, and between CD8+ T immune cells level. CONCLUSION: In conclusion, our results elucidated that MTHFD1L can serve as a shared diagnostic and prognostic biomarker in BLCA, HNSC, KIRP, LUAD, and UCEC patients of different clinicopathological features.


Aminohydrolases/genetics , Biomarkers, Tumor/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Male , Middle Aged , Neoplasms/enzymology , Neoplasms/mortality , Neoplasms/pathology , Predictive Value of Tests , Prognosis , Protein Interaction Maps , Signal Transduction , Up-Regulation , Young Adult
6.
J Pineal Res ; 71(4): e12767, 2021 Dec.
Article En | MEDLINE | ID: mdl-34533844

Metabolic remodeling is now widely recognized as a hallmark of cancer, yet its role in head and neck squamous cell carcinoma (HNSCC) remains largely unknown. In this study, metabolomic analysis of melatonin-treated HNSCC cell lines revealed that exogenous melatonin inhibited many important metabolic pathways including folate cycle in HNSCC cells. Methylenetetrahydrofolate dehydrogenase 1 like (MTHFD1L), a metabolic enzyme of the folate cycle regulating the production of formate, was identified as a downstream target of melatonin. MTHFD1L was found to be markedly upregulated in HNSCC, and MTHFD1L overexpression was significantly associated with unfavorable clinical outcome of HNSCC patients. In addition, MTHFD1L promoted HNSCC progression in vitro and in vivo and reversed the oncostatic effects of exogenous melatonin. More importantly, the malignant phenotypes suppressed by knockdown of MTHFD1L or exogenous melatonin could be partially rescued by formate. Furthermore, we found that melatonin inhibited the expression of MTHFD1L in HNSCC cells through the downregulation of cyclic AMP-responsive element-binding protein 1 (CREB1) phosphorylation. Lastly, this novel regulatory axis of melatonin-p-CREB1-MTHFD1L-formate was also verified in HNSCC tissues. Collectively, our findings have demonstrated that MTHFD1L-formate axis promotes HNSCC progression and melatonin inhibits HNSCC progression through CREB1-mediated downregulation of MTHFD1L and formate. These findings have revealed new metabolic mechanisms in HNSCC and may provide novel insights on the therapeutic intervention of HNSCC.


Formate-Tetrahydrofolate Ligase , Head and Neck Neoplasms , Melatonin , Aminohydrolases/genetics , Aminohydrolases/metabolism , Cell Line, Tumor , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Formates , Gene Expression Regulation, Neoplastic , Humans , Melatonin/pharmacology , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics
7.
Amino Acids ; 53(10): 1589-1595, 2021 Oct.
Article En | MEDLINE | ID: mdl-34550462

One-carbon units, critical intermediates for cell growth, may be produced by a variety of means, one of which is via the production of formate. Excessive formate accumulation, known as formate overflow and a characteristic of oxidative cancer, has been observed in cancer cells. However, the basis for this high rate of formate production is unknown. We examined the effect of elevated expression of oncogenic Ras (RasV12), on formate production in NIH-3T3 cells (mouse fibroblasts) cultured with either labelled 13C-serine or 13C-glycine. Formate accumulation by the fibroblasts transformed by RasV12 was increased two-threefold over those by vector control (Babe) cells. The production of formate exceeded the rate of utilization in both cell types. 13C-formate was produced almost exclusively from the #3 carbon of 13C-serine. Virtually no labelled formate was produced from either the #2 carbon of serine or the #2 carbon of glycine. The increased formate production by RasV12 cells was associated with increased mRNA abundances for enzymes of formate production in both the mitochondria and the cytosol. Thus, we find the oncogenic RasV12 significantly increases formate overflow and may be one way for tumor cells to produce one-carbon units required for enhanced proliferation of these cells and/or for other processes which have not been identified.


Formates/metabolism , Genes, ras , ras Proteins , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Gene Expression Regulation , Glycine/metabolism , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , NIH 3T3 Cells , RNA, Messenger/genetics , Serine/metabolism , ras Proteins/genetics , ras Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article En | MEDLINE | ID: mdl-34544865

Bats are responsible for the zoonotic transmission of several major viral diseases, including those leading to the 2003 SARS outbreak and likely the ongoing COVID-19 pandemic. While comparative genomics studies have revealed characteristic adaptations of the bat innate immune system, functional genomic studies are urgently needed to provide a foundation for the molecular dissection of the viral tolerance in bats. Here we report the establishment of genome-wide RNA interference (RNAi) and CRISPR libraries for the screening of the model megabat, Pteropus alecto. We used the complementary RNAi and CRISPR libraries to interrogate P. alecto cells for infection with two different viruses: mumps virus and influenza A virus, respectively. Independent screening results converged on the endocytosis pathway and the protein secretory pathway as required for both viral infections. Additionally, we revealed a general dependence of the C1-tetrahydrofolate synthase gene, MTHFD1, for viral replication in bat cells and human cells. The MTHFD1 inhibitor, carolacton, potently blocked replication of several RNA viruses, including SARS-CoV-2. We also discovered that bats have lower expression levels of MTHFD1 than humans. Our studies provide a resource for systematic inquiry into the genetic underpinnings of bat biology and a potential target for developing broad-spectrum antiviral therapy.


Aminohydrolases/genetics , COVID-19/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Pandemics , Aminohydrolases/antagonists & inhibitors , Animals , Antiviral Agents/therapeutic use , COVID-19/virology , Cell Line , Chiroptera/genetics , Chiroptera/virology , Formate-Tetrahydrofolate Ligase/antagonists & inhibitors , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Minor Histocompatibility Antigens , Multienzyme Complexes/antagonists & inhibitors , RNA Viruses/genetics , SARS-CoV-2/pathogenicity , Virus Replication/genetics , COVID-19 Drug Treatment
9.
J Pharm Pharmacol ; 73(11): 1480-1490, 2021 Oct 07.
Article En | MEDLINE | ID: mdl-34254644

OBJECTIVES: This study aims to evaluate the association between polymorphisms of methotrexate pathway genes and high-dose methotrexate-related hepatotoxicity in Chinese patients with primary central nervous system lymphoma. METHODS: Sixty-five patients in 411 treatment courses were enrolled and their toxicities were evaluated. The association between 30 candidate SNPs from 20 methotrexate pathway genes and high-dose methotrexate-related hepatotoxicity was analysed by PLINK and logistic regression. KEY FINDINGS: TYMS 6 bp DI + II (rs151264360; OR, 0.41; 95% CI, 0.25-0.66; P = 0.00029), MTHFD1 1958 GA + AA (rs2236225; OR, 0.55; 95% CI, 0.33-0.91; P = 0.020) and CCND1 870 GA + GG (rs9344; OR, 0.42; 95% CI, 0.24-0.73; P = 0.0024) had less risk of hepatotoxicity compared with their homozygotes (DD, GG and AA, respectively), while ABCC2 intron 29 GA + GG (rs3740065; OR, 3.14; 95% CI, 1.89-5.20; P = 0.00001) was more prevalent in patients with hepatotoxicity than TT. CONCLUSIONS: TYMS 6 bp DI + II, MTHFD1 1958 GA + AA, CCND1 870 GA + GG genotypes were associated with a lower probability of hepatotoxicity in patients with primary central nervous system lymphoma on high-dose methotrexate therapy, and ABCC2 intron 29 GA + GG was correlated with increased risk of hepatotoxicity.


Aminohydrolases/genetics , Chemical and Drug Induced Liver Injury/genetics , Cyclin D1/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methotrexate/administration & dosage , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multidrug Resistance-Associated Protein 2/genetics , Multienzyme Complexes/genetics , Polymorphism, Single Nucleotide , Thymidylate Synthase/genetics , Adult , Aged , Aminohydrolases/metabolism , Asian People/genetics , Central Nervous System/pathology , Chemical and Drug Induced Liver Injury/drug therapy , Cyclin D1/metabolism , Female , Formate-Tetrahydrofolate Ligase/metabolism , Genotype , Humans , Introns , Liver/drug effects , Lymphoma/drug therapy , Male , Methotrexate/therapeutic use , Methotrexate/toxicity , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Middle Aged , Multidrug Resistance-Associated Protein 2/metabolism , Multienzyme Complexes/metabolism , Odds Ratio , Thymidylate Synthase/metabolism
10.
J Biosci ; 462021.
Article En | MEDLINE | ID: mdl-34168100

Obesity has been followed with interest as a risk factor for COVID-19, with triglycerides as one of four common criteria used to define obesity, which have been used to study the mechanism of obesity. In this study, we showed that angiotensin-converting enzyme-2 (ACE2) is widely expressed in the mouse body, including the kidney, spleen, brain, heart, lung, liver, and testis, and that ACE2 levels increased after a high-fat diet. The ACE2 levels were recorded at 0 days, 3 days, 7 days, and 14 days after a high-fat diet, and they increased at 14 days after high-fat diet initiation. In addition, triglyceride levels were also significantly increased at 14 days after high-fat diet initiation, but body weight was not changed. Furthermore, we examined the ACE2 levels in Calu3 cells (a lung cancer cell line) after triglyceride treatment, and the results indicated that ACE2 levels were increased at 25 µM and reached their peak at 200 µM. Finally, we found that the mRNA level of mthfd1 was significantly increased in the high-fat diet group. Given these findings, we hypothesize that triglycerides can regulate the expression of ACE2 and Mthfd1.


Angiotensin-Converting Enzyme 2/metabolism , Diet, High-Fat/adverse effects , Formate-Tetrahydrofolate Ligase/metabolism , Gene Expression Regulation/drug effects , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multifunctional Enzymes/metabolism , Triglycerides/pharmacology , Angiotensin-Converting Enzyme 2/genetics , Animals , Biomarkers/blood , COVID-19/etiology , COVID-19/virology , Cell Line , Cell Survival , Formate-Tetrahydrofolate Ligase/genetics , Humans , Male , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Mice, Inbred C57BL , Multifunctional Enzymes/genetics , Obesity/complications , Obesity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors , SARS-CoV-2 , Triglycerides/administration & dosage
11.
Anal Bioanal Chem ; 413(13): 3573-3582, 2021 May.
Article En | MEDLINE | ID: mdl-33829277

Mammalian folate-dependent one-carbon (1C) metabolism provides the building blocks essential during development via amino acid interconversion, methyl-donor production, regeneration of redox factors, and de novo purine and thymidylate synthesis. Folate supplementation prevents many neural tube defects (NTDs) that occur during the embryonic process of neurulation. The mechanism by which folate functions during neurulation is not well understood, and not all NTDs are preventable by folate supplementation. Mthfd1l is a mitochondrial 1C metabolism enzyme that produces formate, a 1C donor that fuels biosynthesis and the methyl cycle in the cytoplasm. Homozygous deletion of the Mthfd1l gene in mice (Mthfd1lz/z) causes embryonic lethality, developmental delay, and folate-resistant NTDs. These mice also have defects in cranial mesenchyme formation. In this work, mass spectrometry imaging was used to obtain ion maps of the cranial mesenchyme that identified the spatial distribution and relative abundance of metabolites in wild-type and Mthfd1lz/z embryos. The relative abundances of purine and thymidylate derivatives, as well as amino acids, were diminished in the cranial mesenchyme of Mthfd1lz/z embryos. Loss of Mthfd1l activity in this region also led to abnormal levels of methionine and dysregulated energy metabolism. These alterations in metabolism suggest possible approaches to preventing NTDs in humans.


Aminohydrolases/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Mice , Mice, Knockout
12.
Biosci Rep ; 41(2)2021 02 26.
Article En | MEDLINE | ID: mdl-33605411

Liver hepatocellular carcinoma (LIHC) is one of the most frequently occurring primary malignant liver tumors and seriously harms people's health in the world. Methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L) has been shown to be associated with colon cancer cell proliferation, colony formation and invasion. In the present study, a total of 370 LIHC and 51 normal samples data were downloaded from The Cancer Genome Atlas (TCGA) database. Bioinformatics and immunohistochemistry (IHC) analysis showed that MTHFD1L is highly expressed in liver tumors. Correlation analysis suggested the differences of vital status between high- and low-expression MTHFD1L groups of LIHC. Univariate and multivariate Cox proportional hazards regression were performed to identify the relationship between clinical characteristics and overall survival (OS). In addition, to explore whether MTHFD1L has an effect on the immune infiltration of LIHC. The correlation between MTHFD1L expression and 24 immune cells were analyzed by ImmuneCellAI database. Furthermore, we combined three databases CIBERSORT, TIMER and ImmuneCellAI to do a comprehensive validation and determined that dendritic cells (DCs) resting, macrophage M0 and macrophage M2 closely related to the expression of MTHFD1L. The results showed that MTHFD1L was a potential prognostic biomarker for LIHC, and could help to elucidate that how the immune microenvironment promotes liver cancer development.


Aminohydrolases/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Computational Biology , Formate-Tetrahydrofolate Ligase/metabolism , Liver Neoplasms/pathology , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multienzyme Complexes/metabolism , Aminohydrolases/genetics , Carcinoma, Hepatocellular/metabolism , Cohort Studies , Formate-Tetrahydrofolate Ligase/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Prognosis , Tumor Microenvironment/genetics
13.
Nutrients ; 14(1)2021 Dec 28.
Article En | MEDLINE | ID: mdl-35011003

Folate and choline are interconnected metabolically. The MTHFD1 R653Q SNP is a risk factor for birth defects and there are concerns that choline deficiency may interact with this SNP and exacerbate health risks. 80-90% of women do not meet the Adequate Intake (AI) for choline. The objective of this study was to assess the effects of choline deficiency on maternal one-carbon metabolism and reproductive outcomes in the MTHFD1-synthetase deficient mouse (Mthfd1S), a model for MTHFD1 R653Q. Mthfd1S+/+ and Mthfd1S+/- females were fed control (CD) or choline-deficient diets (ChDD; 1/3 the amount of choline) before mating and during pregnancy. Embryos were evaluated for delays and defects at 10.5 days gestation. Choline metabolites were measured in the maternal liver, and total folate measured in maternal plasma and liver. ChDD significantly decreased choline, betaine, phosphocholine, and dimethylglycine in maternal liver (p < 0.05, ANOVA), and altered phosphatidylcholine metabolism. Maternal and embryonic genotype, and diet-genotype interactions had significant effects on defect incidence. Mild choline deficiency and Mthfd1S+/- genotype alter maternal one-carbon metabolism and increase incidence of developmental defects. Further study is required to determine if low choline intakes contribute to developmental defects in humans, particularly in 653QQ women.


Aminohydrolases/genetics , Choline Deficiency/genetics , Developmental Disabilities/genetics , Formate-Tetrahydrofolate Ligase/deficiency , Formate-Tetrahydrofolate Ligase/genetics , Maternal Nutritional Physiological Phenomena/genetics , Methenyltetrahydrofolate Cyclohydrolase/deficiency , Methylenetetrahydrofolate Dehydrogenase (NADP)/deficiency , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Multifunctional Enzymes/deficiency , Animals , Choline/analysis , Developmental Disabilities/epidemiology , Disease Models, Animal , Embryonic Development/genetics , Female , Folic Acid/metabolism , Genotype , Incidence , Liver/metabolism , Mice , Polymorphism, Single Nucleotide , Pregnancy
14.
FEBS Open Bio ; 11(1): 289-299, 2021 01.
Article En | MEDLINE | ID: mdl-33146000

Arbutin, a glycoside extracted from the plant Arctostaphylos uva-ursi, has been previously reported to possess antioxidant, anti-inflammatory and anticancer effects. Here, we investigated whether arbutin affects the proliferation of the cells of the osteosarcoma (OS) cell lines MG-63 and SW1353. Arbutin suppressed OS cell viability in a dose- and time-dependent manner, as shown by Cell Counting Kit-8 assay. Furthermore, arbutin exposure decreased the protein levels of MTHFD1L, CCND1 and phosphorylated-protein kinase B (AKT)/phosphorylated-mammalian target of rapamycin (mTOR). Potential upstream miRNAs of MTHFD1L were predicted using TargetScan, PICTAR5, miRanda and miRWalk. We performed luciferase activity assays to show that miR-338-3p directly targets and negatively regulates the expression of MTHFD1L. Knockdown of miR-338-3p promoted cell invasion, migration and proliferation in arbutin-treated OS cells via MTHFD1L. In summary, our data suggest that arbutin inhibits OS cell proliferation, migration and invasion via miR-338-3p/MTHFD1L and by inactivating the AKT/mTOR pathway.


Arbutin/pharmacology , Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , Aminohydrolases/genetics , Arbutin/therapeutic use , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Formate-Tetrahydrofolate Ligase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Multienzyme Complexes/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism
15.
Biochem Biophys Res Commun ; 528(3): 426-431, 2020 07 30.
Article En | MEDLINE | ID: mdl-32505353

Methylobacterium extorquens is a methylotroph model organism that has the ability to assimilate formate using the tetrahydrofolate (THF) pathway. The formate-tetrahydrofolate ligase from M. extorquens (MeFtfL) is an enzyme involved in the THF pathway that catalyzes the conversion of formate, THF, and ATP into formyltetrahydrofolate and ADP. To investigate the biochemical properties of MeFtfL, we evaluated the metal usage and enzyme kinetics of the enzyme. MeFtfL uses the Mg ion for catalytic activity, but also has activity for Mn and Ca ions. The enzyme kinetics analysis revealed that Km value of farmate was much higher than THF and ATP, which shows that the ligation activity of MeFtfL is highly dependent on formation concentration. We also determined the crystal structure of MeFtfL at 2.8 Å resolution. MeFtfL functions as a tetramer, and each monomer consists of three domains. The structural superposition of MeFtfL with FtfL from Moorella thermoacetica allowed us to predict the substrate binding site of the enzyme.


Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Formate-Tetrahydrofolate Ligase/chemistry , Formate-Tetrahydrofolate Ligase/metabolism , Methylobacterium extorquens/enzymology , Bacterial Proteins/genetics , Catalytic Domain , Crystallography, X-Ray , Formate-Tetrahydrofolate Ligase/genetics , Formates/metabolism , Kinetics , Metabolic Networks and Pathways , Methylobacterium extorquens/genetics , Models, Molecular , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
16.
J Recept Signal Transduct Res ; 40(6): 584-590, 2020 Dec.
Article En | MEDLINE | ID: mdl-32456526

Osteosarcoma (OS) is the most frequent primary malignancy initially in bone with multiple genomic aberrations. Methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L) is linked with the progression of diverse tumors. However, its function in OS is not understood completely. The expression pattern and prognostic significance of MTHFD1L in OS tissues were analyzed based on GEO database. The expression level of MTHFD1L in OS cell lines was explored by qRT-PCR. The cell proliferation, colony formation ability, invasion as well as migration in OS cells after MTHFD1L knockdown were determined using cell counting kit 8 (CCK-8) assay, colony formation and transwell methods. GSEA analysis was performed to predict the underlying mechanisms of MTHFD1L in OS development. Furthermore, the western blot was utilized to study the influence of MTHFD1L on AKT/mTOR pathway. Our results indicated that MTHFD1L expression was significantly up-regulated in OS tissues and cells compared with normal tissues and cells. High expression of MTHFD1L could lead to poor prognosis of OS patients. Cell proliferation, colony formation ability, migration and invasion were blocked because of reduced MTHFD1L in vitro. Moreover, cell cycle and AKT/mTOR pathway were all associated with MTHFD1L expression. In conclusion, the findings revealed that MTHFD1L might promote the development of OS via mediating cell cycle and AKT/mTOR pathway, indicating that MTHFD1L might act as a promising therapeutic target for OS treatment.


Aminohydrolases/metabolism , Biomarkers, Tumor/metabolism , Bone Neoplasms/pathology , Formate-Tetrahydrofolate Ligase/metabolism , Gene Expression Regulation, Neoplastic , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multienzyme Complexes/metabolism , Osteosarcoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Aminohydrolases/genetics , Apoptosis , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/genetics , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Neoplasm Invasiveness , Osteosarcoma/genetics , Osteosarcoma/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured
17.
Database (Oxford) ; 20192019 01 01.
Article En | MEDLINE | ID: mdl-31832668

Acetogenic bacteria are imperative to environmental carbon cycling and diverse biotechnological applications, but their extensive physiological and taxonomical diversity is an impediment to systematic taxonomic studies. Acetogens are chemolithoautotrophic bacteria that perform reductive carbon fixation under anaerobic conditions through the Wood-Ljungdahl pathway (WLP)/acetyl-coenzyme A pathway. The gene-encoding formyltetrahydrofolate synthetase (FTHFS), a key enzyme of this pathway, is highly conserved and can be used as a molecular marker to probe acetogenic communities. However, there is a lack of systematic collection of FTHFS sequence data at nucleotide and protein levels. In an attempt to streamline investigations on acetogens, we developed AcetoBase - a repository and database for systematically collecting and organizing information related to FTHFS sequences. AcetoBase also provides an opportunity to submit data and obtain accession numbers, perform homology searches for sequence identification and access a customized blast database of submitted sequences. AcetoBase provides the prospect to identify potential acetogenic bacteria, based on metadata information related to genome content and the WLP, supplemented with FTHFS sequence accessions, and can be an important tool in the study of acetogenic communities. AcetoBase can be publicly accessed at https://acetobase.molbio.slu.se.


Databases, Genetic , Formate-Tetrahydrofolate Ligase/genetics , Acetates/metabolism , Amino Acid Sequence , Base Sequence , Databases, Protein , Genome , Phylogeny
18.
Birth Defects Res ; 111(19): 1520-1534, 2019 11 15.
Article En | MEDLINE | ID: mdl-31518072

BACKGROUND: Periconceptional intake of supplemental folic acid can reduce the incidence of neural tube defects by as much as 70%, but the mechanisms by which folic acid supports cellular processes during neural tube closure are unknown. The mitochondrial 10-formyl-tetrahydrofolate synthetase MTHFD1L catalyzes production of formate, thus generating one-carbon units for cytoplasmic processes. Deletion of Mthfd1l causes embryonic lethality, developmental delay, and neural tube defects in mice. METHODS: To investigate the role of mitochondrial one-carbon metabolism during cranial neural tube closure, we have analyzed cellular morphology and function in neural tissues in Mthfd1l knockout embryos. RESULTS: The head mesenchyme showed significantly lower cellular density in Mthfd1l nullizygous embryos compared to wildtype embryos during the process of neural tube closure. Apoptosis and neural crest cell specification were not affected by deletion of Mthfd1l. Sections from the cranial region of Mthfd1l knockout embryos exhibited decreased cellular proliferation, but only after completion of neural tube closure. Supplementation of pregnant dams with formate improved mesenchymal density and corrected cell proliferation in the nullizygous embryos. CONCLUSIONS: Deletion of Mthfd1l causes decreased density in the cranial mesenchyme and this defect is improved with formate supplementation. This study reveals a mechanistic link between folate-dependent mitochondrially produced formate, head mesenchyme formation and neural tube defects.


Formate-Tetrahydrofolate Ligase/genetics , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multifunctional Enzymes/genetics , Neural Tube Defects/genetics , Animals , Embryo, Mammalian/metabolism , Female , Folic Acid/genetics , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/metabolism , Formates/metabolism , Male , Mesoderm/metabolism , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Multifunctional Enzymes/metabolism , Neural Crest/metabolism , Neural Tube Defects/metabolism , Neurulation , Sequence Deletion
20.
Nucleic Acids Res ; 47(14): 7580-7591, 2019 08 22.
Article En | MEDLINE | ID: mdl-31147722

Alternative cleavage and polyadenylation (APA) can occur at more than half of all human genes, greatly enhancing the cellular repertoire of mRNA isoforms. As these isoforms can have altered stability, localisation and coding potential, deregulation of APA can disrupt gene expression and this has been linked to many diseases including cancer progression. How APA generates cancer-specific isoform profiles and what their physiological consequences are, however, is largely unclear. Here we use a subcellular fractionation approach to determine the nuclear and cytoplasmic APA profiles of successive stages of colon cancer using a cell line-based model. Using this approach, we show that during cancer progression specific APA profiles are established. We identify that overexpression of hnRNPC has a critical role in the establishment of APA profiles characteristic for metastatic colon cancer cells, by regulating poly(A) site selection in a subset of genes that have been implicated in cancer progression including MTHFD1L.


Alternative Splicing , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Neoplasms/genetics , Polyadenylation , RNA Isoforms/genetics , Aminohydrolases/genetics , Aminohydrolases/metabolism , Cell Line, Transformed , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Poly A/genetics , Poly A/metabolism , RNA Interference
...