Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Biomol Struct Dyn ; 41(21): 11818-11831, 2023.
Article in English | MEDLINE | ID: mdl-36597898

ABSTRACT

MTHFR (Methylenetetrahydrofolate reductase) is a pivotal enzyme involved in one-carbon metabolism, which is critical for the proliferation of cancer cells. In line with this, published literature showed that MTHFR knockdown caused impaired growth of multiple types of cancer cells. Moreover, higher MTHFR expression levels were linked to shorter overall survival in hepatocellular carcinoma, adrenocortical carcinoma, and low-grade glioma, bringing the need to design MTHFR inhibitors as a possible treatment option. No competitive inhibitors of MTHFR have been reported as of today. This study aimed to identify potential competitive MTHFR inhibitor candidates using an in silico drug screen. A total of 30470 molecules containing biogenic compounds, FDA-approved drugs, and those in clinical trials were screened against the catalytic pocket of MTHFR in the presence and absence of cofactors. Binding energy and ADMET analysis revealed that Vilanterol (ß2-adrenergic agonist), Selexipag (prostacyclin receptor agonist), and Ramipril Diketopiperazine (ACE inhibitor) are potential competitive inhibitors of MTHFR. Molecular dynamics analyses and MM-PBSA calculations with these compounds particularly revealed the amino acids between 285-290 for ligand binding and highlighted Vilanterol as the strongest candidate for MTHFR inhibition. Our results could guide the development of novel MTHFR inhibitor compounds, which could be inspired by the drugs brought into the spotlight here. More importantly, these potential candidates could be quhickly tested as a repurposing strategy in pre-clinical and clinical studies of the cancers mentioned above.Communicated by Ramaswamy H. Sarma.


Subject(s)
Liver Neoplasms , Methylenetetrahydrofolate Reductase (NADPH2) , Humans , Amino Acids , Drug Repositioning , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Molecular Docking Simulation , Molecular Dynamics Simulation
2.
Int J Mol Sci ; 22(17)2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34502300

ABSTRACT

Folate depletion causes chromosomal instability by increasing DNA strand breakage, uracil misincorporation, and defective repair. Folate mediated one-carbon metabolism has been suggested to play a key role in the carcinogenesis and progression of hepatocellular carcinoma (HCC) through influencing DNA integrity. Methylenetetrahydrofolate reductase (MTHFR) is the enzyme catalyzing the irreversible conversion of 5,10-methylenetetrahydrofolate to 5-methyltetrahydrofolate that can control folate cofactor distributions and modulate the partitioning of intracellular one-carbon moieties. The association between MTHFR polymorphisms and HCC risk is inconsistent and remains controversial in populational studies. We aimed to establish an in vitro cell model of liver origin to elucidate the interactions between MTHFR function, folate status, and chromosome stability. In the present study, we (1) examined MTHFR expression in HCC patients; (2) established cell models of liver origin with stabilized inhibition of MTHFR using small hairpin RNA delivered by a lentiviral vector, and (3) investigated the impacts of reduced MTHFR and folate status on cell cycle, methyl group homeostasis, nucleotide biosynthesis, and DNA stability, all of which are pathways involved in DNA integrity and repair and are critical in human tumorigenesis. By analyzing the TCGA/GTEx datasets available within GEPIA2, we discovered that HCC cancer patients with higher MTHFR had a worse survival rate. The shRNA of MTHFR (shMTHFR) resulted in decreased MTHFR gene expression, MTHFR protein, and enzymatic activity in human hepatoma cell HepG2. shMTHFR tended to decrease intracellular S-adenosylmethionine (SAM) contents but folate depletion similarly decreased SAM in wildtype (WT), negative control (Neg), and shMTHFR cells, indicating that in cells of liver origin, shMTHFR does not exacerbate the methyl group supply in folate depletion. shMTHFR caused cell accumulations in the G2/M, and cell population in the G2/M was inversely correlated with MTHFR gene level (r = -0.81, p < 0.0001), MTHFR protein expression (r = -0.8; p = 0.01), and MTHFR enzyme activity (r = -0.842; p = 0.005). Folate depletion resulted in G2/M cell cycle arrest in WT and Neg but not in shMTHFR cells, indicating that shMTHFR does not exacerbate folate depletion-induced G2/M cell cycle arrest. In addition, shMTHFR promoted the expression and translocation of nuclei thymidine synthetic enzyme complex SHMT1/DHFR/TYMS and assisted folate-dependent de novo nucleotide biosynthesis under folate restriction. Finally, shMTHFR promoted nuclear MLH1/p53 expression under folate deficiency and further reduced micronuclei formation and DNA uracil misincorporation under folate deficiency. In conclusion, shMTHFR in HepG2 induces cell cycle arrest in G2/M that may promote nucleotide supply and assist cell defense against folate depletion-induced chromosome segregation and uracil misincorporation in the DNA. This study provided insight into the significant impact of MTHFR function on chromosome stability of hepatic tissues. Data from the present study may shed light on the potential regulatory mechanism by which MTHFR modulates the risk for hepatic malignancies.


Subject(s)
Carcinoma, Hepatocellular/pathology , Chromosome Segregation , DNA, Neoplasm/genetics , Folic Acid/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Uracil/metabolism , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Chromosomal Instability , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Polymorphism, Genetic , Prognosis , Survival Rate , Tumor Cells, Cultured
3.
Biochimie ; 183: 100-107, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33476699

ABSTRACT

The folate and methionine cycles, constituting one-carbon metabolism, are critical pathways for cell survival. Intersecting these two cycles, 5,10-methylenetetrahydrofolate reductase (MTHFR) directs one-carbon units from the folate to methionine cycle, to be exclusively used for methionine and S-adenosylmethionine (AdoMet) synthesis. MTHFR deficiency and upregulation result in diverse disease states, rendering it an attractive drug target. The activity of MTHFR is inhibited by the binding of AdoMet to an allosteric regulatory domain distal to the enzyme's active site, which we have previously identified to constitute a novel fold with a druggable pocket. Here, we screened 162 AdoMet mimetics using differential scanning fluorimetry, and identified 4 compounds that stabilized this regulatory domain. Three compounds were sinefungin analogues, closely related to AdoMet and S-adenosylhomocysteine (AdoHcy). The strongest thermal stabilisation was provided by (S)-SKI-72, a potent inhibitor originally developed for protein arginine methyltransferase 4 (PRMT4). Using surface plasmon resonance, we confirmed that (S)-SKI-72 binds MTHFR via its allosteric domain with nanomolar affinity. Assay of MTHFR activity in the presence of (S)-SKI-72 demonstrates inhibition of purified enzyme with sub-micromolar potency and endogenous MTHFR from HEK293 cell lysate in the low micromolar range, both of which are lower than AdoMet. Nevertheless, unlike AdoMet, (S)-SKI-72 is unable to completely abolish MTHFR activity, even at very high concentrations. Combining binding assays, kinetic characterization and compound docking, this work indicates the regulatory domain of MTHFR can be targeted by small molecules and presents (S)-SKI-72 as an excellent candidate for development of MTHFR inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/chemistry , S-Adenosylmethionine/chemistry , Allosteric Regulation , Humans , Protein Domains
4.
Cell Physiol Biochem ; 45(3): 1149-1155, 2018.
Article in English | MEDLINE | ID: mdl-29444504

ABSTRACT

BACKGROUND/AIMS: The association between the genetic polymorphisms located in either the exon or untranslated region of MTHFR and the risk of human atherosclerosis has been well-documented. This study analyzed MTHFR polymorphisms at the 3'-untranslated region for association with risk and outcome of atherosclerosis in a Chinese Han population. METHODS: The hospital based case-control study was conducted with 500 patients and 600 healthy volunteers as control enrolled. The genotyping was conducted by using Taqman probe. The potential interaction was predicted by multiple bioinformatics analysis. The relative expression of MTHFR was detected by qRT-PCR. Further confirmation was determined by dual-luciferase assay. The plasma homocysteine levels were assayed by ELISA. RESULTS: Cigarette smoking, alcohol consumption, diabetes, hypertension and low levels of serum high-density lipoprotein-C were associated with an increased risk of developing ischemic stroke. MTHFR rs915014 AG and GG genotypes were significantly associated with increased risk of rs915014 compared with the GG genotype. The qRT-PCR confirmed that MTHFR rs915014 AG or GG genotypes could facilitate miR-2861 binding leading to decreased MTHFR levels in cells. In addition, patients carrying the MTHFR rs915014 AG or GG genotypes were associated with accumulation of circulating tHcy volume and a poor atherosclerosis consequence. CONCLUSIONS: This study demonstrates that the MTHFR rs915014 is associated with increased risk of atherosclerosis and might be a shot term outcome biomarker for atherosclerosis patients.


Subject(s)
Atherosclerosis/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , MicroRNAs/metabolism , Aged , Alcohol Drinking , Antagomirs/metabolism , Asian People/genetics , Atherosclerosis/pathology , Case-Control Studies , Female , HEK293 Cells , Homocysteine/blood , Humans , Hypertension/complications , Lipoproteins, HDL/blood , Male , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , Polymorphism, Single Nucleotide , Smoking , Stroke/etiology
5.
Article in English | MEDLINE | ID: mdl-29367024

ABSTRACT

The experimental and theoretical investigations of structure of the 3-(1-(phenylamino)ethylidene)-chroman-2,4-dione were performed. X-ray structure analysis and spectroscopic methods (FTIR and FT-Raman, 1H and 13C NMR), along with the density functional theory calculations (B3LYP functional with empirical dispersion corrections D3BJ in combination with the 6-311 + G(d,p) basis set), were used in order to characterize the molecular structure and spectroscopic behavior of the investigated coumarin derivative. Molecular docking analysis was carried out to identify the potency of inhibition of the title molecule against human's Ubiquinol-Cytochrome C Reductase Binding Protein (UQCRB) and Methylenetetrahydrofolate reductase (MTHFR). The inhibition activity was obtained for ten conformations of ligand inside the proteins.


Subject(s)
Chromans/chemistry , Chromans/pharmacology , Magnetic Resonance Spectroscopy/methods , Molecular Docking Simulation , Quantum Theory , Spectroscopy, Fourier Transform Infrared/methods , Spectrum Analysis, Raman/methods , Carrier Proteins/antagonists & inhibitors , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Models, Molecular , Molecular Conformation , Molecular Structure , Thermodynamics
6.
Biochem Biophys Res Commun ; 465(3): 363-7, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26253470

ABSTRACT

Genistein (5, 7-dihydroxy-3- (4-hydroxyphenyl)-4H-1-benzopyran-4-one) is a natural isoflavone revealing many biological activities. Thus, it is considered as a therapeutic compound in as various disorders as cancer, infections and genetic diseases. Here, we demonstrate for the first time that genistein inhibits activities of bacterial methylenetetrahydrofolate reductase (MetF) and lactate dehydrogenase (LDH). Both enzymes use NADH as a substrate, and results of biochemical as well as molecular modeling studies with MetF suggest that genistein may interfere with binding of this dinucleotide to the enzyme. These results have implications for our understanding of biological functions of genistein and its effects on cellular metabolism.


Subject(s)
Genistein/chemistry , L-Lactate Dehydrogenase/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Models, Chemical , NAD/chemistry , Binding Sites , Enzyme Activation , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/chemistry , Substrate Specificity
7.
Am J Clin Nutr ; 101(3): 646-58, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25733650

ABSTRACT

BACKGROUND: Increased consumption of folic acid is prevalent, leading to concerns about negative consequences. The effects of folic acid on the liver, the primary organ for folate metabolism, are largely unknown. Methylenetetrahydrofolate reductase (MTHFR) provides methyl donors for S-adenosylmethionine (SAM) synthesis and methylation reactions. OBJECTIVE: Our goal was to investigate the impact of high folic acid intake on liver disease and methyl metabolism. DESIGN: Folic acid-supplemented diet (FASD, 10-fold higher than recommended) and control diet were fed to male Mthfr(+/+) and Mthfr(+/-) mice for 6 mo to assess gene-nutrient interactions. Liver pathology, folate and choline metabolites, and gene expression in folate and lipid pathways were examined. RESULTS: Liver and spleen weights were higher and hematologic profiles were altered in FASD-fed mice. Liver histology revealed unusually large, degenerating cells in FASD Mthfr(+/-) mice, consistent with nonalcoholic fatty liver disease. High folic acid inhibited MTHFR activity in vitro, and MTHFR protein was reduced in FASD-fed mice. 5-Methyltetrahydrofolate, SAM, and SAM/S-adenosylhomocysteine ratios were lower in FASD and Mthfr(+/-) livers. Choline metabolites, including phosphatidylcholine, were reduced due to genotype and/or diet in an attempt to restore methylation capacity through choline/betaine-dependent SAM synthesis. Expression changes in genes of one-carbon and lipid metabolism were particularly significant in FASD Mthfr(+/-) mice. The latter changes, which included higher nuclear sterol regulatory element-binding protein 1, higher Srepb2 messenger RNA (mRNA), lower farnesoid X receptor (Nr1h4) mRNA, and lower Cyp7a1 mRNA, would lead to greater lipogenesis and reduced cholesterol catabolism into bile. CONCLUSIONS: We suggest that high folic acid consumption reduces MTHFR protein and activity levels, creating a pseudo-MTHFR deficiency. This deficiency results in hepatocyte degeneration, suggesting a 2-hit mechanism whereby mutant hepatocytes cannot accommodate the lipid disturbances and altered membrane integrity arising from changes in phospholipid/lipid metabolism. These preliminary findings may have clinical implications for individuals consuming high-dose folic acid supplements, particularly those who are MTHFR deficient.


Subject(s)
Dietary Supplements/poisoning , Enzyme Inhibitors/poisoning , Folic Acid/poisoning , Homocystinuria/etiology , Lipid Metabolism , Liver/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity/etiology , Non-alcoholic Fatty Liver Disease/etiology , Animals , Gene Expression Regulation , Heterozygote , Homocystinuria/metabolism , Homocystinuria/pathology , Homocystinuria/physiopathology , Lipogenesis , Liver/pathology , Liver/physiopathology , Male , Methylation , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Mice, Inbred BALB C , Mice, Mutant Strains , Muscle Spasticity/metabolism , Muscle Spasticity/pathology , Muscle Spasticity/physiopathology , Mutation , Organ Size , Psychotic Disorders/etiology , Psychotic Disorders/metabolism , Psychotic Disorders/pathology , Psychotic Disorders/physiopathology , Specific Pathogen-Free Organisms
8.
Pharmacogenomics ; 9(9): 1195-206, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18781847

ABSTRACT

The 5,10-methylenetetrahydrofolate reductase (MTHFR) is a key enzyme for intracellular folate homeostasis and metabolism. Two common MTHFR polymorphisms, C677T and A1298C, which lead to an altered amino acid sequence, have been associated with a decreased enzyme activity and susceptibility to cancer suggesting that these genetic variants may modulate the risk of several malignancies. C667T, and to a lesser extent A1298C polymorphisms, are also reported to influence the cytotoxic effect of fluoropyrimidines and antifolates providing support for their pharmacogenetic role in predicting the efficacy and the toxicity in cancer and rheumatoid arthritis patients. A combined polymorphisms and haplotype analysis may result in a more effective approach than a single polymorphism one. Moreover gene-nutrient/environmental and gene-racial/ethnic interactions have been shown to affect the impact of these MTHFR genetic variants. Further well-designed studies are needed to clarify the role of MTHFR polymorphisms to derive dose adjustment recommendations on the basis of the patient's genotype.


Subject(s)
Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Pharmacogenetics/trends , Polymorphism, Genetic/genetics , Animals , Genetic Predisposition to Disease , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Pharmacogenetics/methods , Polymorphism, Genetic/drug effects , Risk Factors
9.
Cell Biol Int ; 32(8): 879-87, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18486501

ABSTRACT

The present study aimed to investigate the effect of knocking-down methylenetetrahydrofolate reductase (MTHFR) on the survival of the human gastric cancer cell line MKN45. Antisense and small interfering RNA (siRNA) plasmids were used to target MTHFR in MKN45. Meanwhile, we also constructed a wild-type MTHFR plasmid to assess the effect of over-expression of this protein on cell viability. The knock-down of MTHFR decreased cell survival by approximately 30% compared to the control and resulted in cell cycle arrest at the G2 phase. These cells also had lower levels of c-myc compared to control cells, while over-expression of MTHFR increased cell proliferation and induced the down-regulation of p21WAF1 and hMLH1. Inhibiting MTHFR with either antisense or siRNA decreases the viability of methionine-dependent transformed gastric cancer cells and suggests that MTHFR inhibition may be a novel anticancer approach.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Genetic Vectors , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , MutL Protein Homolog 1 , Plasmids , Stomach Neoplasms/therapy , Transfection
10.
Curr Pharm Des ; 14(11): 1143-50, 2008.
Article in English | MEDLINE | ID: mdl-18473861

ABSTRACT

Tumor cells have an enhanced requirement for glucose, amino acids and DNA precursors. Since folates are required for the synthesis of thymidine and purines, the metabolism of folate has been exploited as an anti-cancer target for over 6 decades, with emphasis on the inhibition of DNA synthesis. However, folate is also used to generate methionine, which is essential for proliferation by virtue of its role in protein synthesis, polyamine synthesis and transmethylation reactions. Tumor-derived cell lines and human tumor xenografts have been shown to be methionine dependent i.e., they are unable to survive without methionine and are unable to efficiently utilize homocysteine, the immediate metabolic precursor of methionine. Since non-transformed cells are methionine-independent, the targeting of methionine metabolism presents an opportunity to selectively disrupt the unique metabolic networks in cancer cells. This chapter provides an overview of the critical role of folate and methionine metabolism in tumor cells and summarizes the current anti-folate and anti-methionine strategies to inhibit growth of transformed lines and tumors. We also present our work on the development of a novel anti-cancer target, methylenetetrahydrofolate reductase (MTHFR), a key enzyme of both folate and methionine metabolism. Our data demonstrate that antisense-mediated inhibition of MTHFR is associated with increased cytotoxicity in vitro and with decreased growth of tumors in vivo. These findings warrant further investigation of this enzyme and the methionine biosynthetic pathway in exploring new strategies for cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Clinical Trials as Topic , Drug Delivery Systems , Folic Acid/metabolism , Humans , Methionine/metabolism , Oligonucleotides, Antisense/pharmacology
11.
Int J Hematol ; 87(5): 451-458, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18365141

ABSTRACT

Polymorphism in 5,10-methylenetetrahydrofolate reductase (MTHFR), a central enzyme in folate metabolism, has been shown to affect the sensitivity of patients to folate-based drugs such as methotrexate. In this study, we investigated whether a common single nucleotide polymorphism at position 677 in the donor or recipient's MTHFR gene affects the risk for acute graft-versus-host disease (GVHD) following allogeneic hematopoietic stem cell transplantation (HSCT) from HLA-identical sibling donors when the recipient receives prophylactic treatment with methotrexate for GVHD. MTHFR genotypes were determined in 159 recipients with a hematological disease and their donors using polymerase chain reaction-restriction fragment length polymorphism analysis of genomic DNA. The 677TT genotype, which encodes an enzyme with approximately 30% of the activity of the wild-type (677CC), was observed in 13% of patients and in 8% of normal donors. Multivariate analyses demonstrated a significant association between 677TT genotype in patients and a lower incidence of grade I-IV acute GVHD (relative risk, 0.35; 95% confidence interval, 0.13-0.95; P = 0.040). There was no association between the incidence of acute GVHD and the donor MTHFR genotypes. These results suggest that greater immunosuppression by methotrexate due to low MTHFR enzyme activity decreases the risk of acute GVHD in recipients of allogeneic HSCT.


Subject(s)
Graft vs Host Disease/genetics , Hematopoietic Stem Cell Transplantation , Living Donors , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Genetic , Acute Disease , Adolescent , Adult , Female , Genotype , Graft vs Host Disease/drug therapy , Graft vs Host Disease/enzymology , Hematologic Diseases/enzymology , Hematologic Diseases/genetics , Hematologic Diseases/therapy , Humans , Immunosuppression Therapy/methods , Immunosuppressive Agents/administration & dosage , Male , Methotrexate/administration & dosage , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Middle Aged , Risk Factors , Transplantation, Homologous
12.
Int J Oral Maxillofac Surg ; 36(5): 429-34, 2007 May.
Article in English | MEDLINE | ID: mdl-17467239

ABSTRACT

Non-syndromic cleft of the lip and/or palate is one of the most common birth defects in humans. Embryonic palatal mesenchymal (EPM) cells are an attractive source for investigating embryonic palatal development. In this study, we developed a highly efficient transfection method for murine EPM (MEPM) cells. MEPM cells were transfected with the plasmid pEGFP-N1 using two non-viral methods: nucleofection and lipofection. Nucleofection provided a much better rate of gene transfer than lipofection particularly in MEPM cells. The methylenetetrahydrofolate reductase (MTHFR) gene is an important candidate for involvement in the pathogenesis of this birth defect. The RNA interference plasmid of MTHFR was constructed and nucleofected into MEPM cells. Successful transfection resulted in a remarkable reduction in the expression of MTHFR. Taken together, the results indicate that nucleofection is highly efficient for MEPM cell transfection, and that this approach may be useful for investigating gene function in the process of palatogenesis.


Subject(s)
Mesenchymal Stem Cells/physiology , Palate/embryology , Transfection/methods , Animals , Blotting, Western , Cell Nucleus , Cells, Cultured , Cleft Palate/embryology , Cleft Palate/genetics , Female , Fluorescent Dyes , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins , Lipids , Liposomes , Male , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Plasmids/genetics , Polymerase Chain Reaction
13.
Mini Rev Med Chem ; 5(12): 1061-73, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16375752

ABSTRACT

1,4-Benzothiazine (1,4-BT) derivatives have been reported to exhibit a wide range of pharmacological properties including antifungal, immunostimulating, anti-aldoso-reductase, anti-rheumatic, anti-allergic, vasorelaxant, anti-arrhythmic, anti-hypertensive, neuroprotective and cytotoxic activities. These different effects indicate that 1,4-BT is a template potentially useful in medicinal chemistry research and therapeutic applications.


Subject(s)
Thiazines/chemistry , Thiazines/pharmacology , Anti-Allergic Agents/chemistry , Anti-Allergic Agents/pharmacology , Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Calcium Channels/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/pharmacology , Molecular Structure , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Potassium Channels/metabolism , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacology
14.
Clin Cancer Res ; 11(5): 2047-52, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15756031

ABSTRACT

PURPOSE: Many cancer lines are methionine dependent and decrease proliferation when methionine supply is limited. Methylenetetrahydrofolate reductase (MTHFR) generates the folate derivative for homocysteine remethylation to methionine. We investigated the effect of antisense-mediated inhibition of MTHFR on survival of human cancer cells. EXPERIMENTAL DESIGN: We examined the in vitro and in vivo anticancer effects of a combination of MTHFR antisense and standard cytotoxic drugs. RESULTS: Specific antisense against MTHFR (EX5) showed significant inhibitory effects on growth of human colon, lung, breast, prostate, and neuroblastoma tumor cells in vitro compared with that of the control oligonucleotide. Cytotoxic drugs (5-fluorouracil, cisplatin, or paclitaxel) potentiated the effect of EX5. In vivo, antisense alone or in combination with cytotoxic drugs inhibited the growth of human colon and lung carcinoma xenografts. In comparison with control oligonucleotide, treatment with EX5 inhibited growth of colon tumors and lung tumors by 60% and 45%, respectively. EX5 with 5-fluorouracil decreased growth of colon tumors by an additional 30% compared with EX5 alone, and EX5 with cisplatin decreased growth of lung tumors by an additional 40% compared with cisplatin alone. Growth inhibition by EX5 was associated with decreased amounts of MTHFR protein and with increased amounts of an apoptosis marker. CONCLUSIONS: Our results confirm that MTHFR inhibition decreases tumor growth and suggest that inhibition of MTHFR by antisense or small molecules may be a novel anticancer approach.


Subject(s)
Breast Neoplasms/pathology , Cell Survival , Colonic Neoplasms/pathology , Lung Neoplasms/pathology , Methylenetetrahydrofolate Reductase (NADPH2)/antagonists & inhibitors , Methylenetetrahydrofolate Reductase (NADPH2)/pharmacology , Neuroblastoma/pathology , Oligonucleotides, Antisense/pharmacology , Prostatic Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Proliferation , Cisplatin/pharmacology , Female , Humans , Male , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...