Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Signal Transduct Target Ther ; 9(1): 214, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117631

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a serious threat to public health, but its underlying mechanism remains poorly understood. In screening important genes using Gene Importance Calculator (GIC) we developed previously, ribosomal modification protein rimK-like family member A (RIMKLA) was predicted as one essential gene but its functions remained largely unknown. The current study determined the roles of RIMKLA in regulating glucose and lipid metabolism. RIMKLA expression was reduced in livers of human and mouse with NAFLD. Hepatic RIMKLA overexpression ameliorated steatosis and hyperglycemia in obese mice. Hepatocyte-specific RIMKLA knockout aggravated high-fat diet (HFD)-induced dysregulated glucose/lipid metabolism in mice. Mechanistically, RIMKLA is a new protein kinase that phosphorylates betaine-homocysteine S-methyltransferase 1 (BHMT1) at threonine 45 (Thr45) site. Upon phosphorylation at Thr45 and activation, BHMT1 eliminated homocysteine (Hcy) to inhibit the activity of transcription factor activator protein 1 (AP1) and its induction on fatty acid synthase (FASn) and cluster of differentiation 36 (CD36) gene transcriptions, concurrently repressing lipid synthesis and uptake in hepatocytes. Thr45 to alanine (T45A) mutation inactivated BHMT1 to abolish RIMKLA's repression on Hcy level, AP1 activity, FASn/CD36 expressions, and lipid deposition. BHMT1 overexpression rescued the dysregulated lipid metabolism in RIMKLA-deficient hepatocytes. In summary, RIMKLA is a novel protein kinase that phosphorylates BHMT1 at Thr45 to repress lipid synthesis and uptake. Under obese condition, inhibition of RIMKLA impairs BHMT1 activity to promote hepatic lipid deposition.


Subject(s)
Betaine-Homocysteine S-Methyltransferase , Lipid Metabolism , Non-alcoholic Fatty Liver Disease , Animals , Mice , Humans , Betaine-Homocysteine S-Methyltransferase/genetics , Betaine-Homocysteine S-Methyltransferase/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Lipid Metabolism/genetics , Diet, High-Fat/adverse effects , Hepatocytes/metabolism , Male , Mice, Knockout , Phosphorylation/genetics
2.
Genes (Basel) ; 15(7)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39062651

ABSTRACT

Congenital heart disease is one of the most common congenital malformations and thus represents a considerable public health burden. Hence, the identification of individuals and families with an increased genetic predisposition to congenital heart disease (CHD) and its possible prevention is important. Even though CHD is associated with the lack of folate during early pregnancy, the genetic background of folate and methionine metabolism perturbations and their influence on CHD risk is not clear. While some genes, such as those coding for cytosolic enzymes of folate/methionine cycles, have been extensively studied, genetic studies of folate transporters (de)glutamation enzymes and mitochondrial enzymes of the folate cycle are lacking. Among genes coding for cytoplasmic enzymes of the folate cycle, MTHFR, MTHFD1, MTR, and MTRR have the strongest association with CHD, while among genes for enzymes of the methionine cycle BHMT and BHMT2 are the most prominent. Among mitochondrial folate cycle enzymes, MTHFD2 plays the most important role in CHD formation, while FPGS was identified as important in the group of (de)glutamation enzymes. Among transporters, the strongest association with CHD was demonstrated for SLC19A1.


Subject(s)
Folic Acid , Heart Defects, Congenital , Methionine , Methylenetetrahydrofolate Dehydrogenase (NADP) , Humans , Folic Acid/metabolism , Heart Defects, Congenital/genetics , Methionine/metabolism , Methionine/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Ferredoxin-NADP Reductase/genetics , Ferredoxin-NADP Reductase/metabolism , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Genetic Predisposition to Disease , Betaine-Homocysteine S-Methyltransferase/genetics , Betaine-Homocysteine S-Methyltransferase/metabolism , Aminohydrolases , Multifunctional Enzymes
3.
Mol Cell Biol ; 44(6): 245-258, 2024.
Article in English | MEDLINE | ID: mdl-38804232

ABSTRACT

Betaine-homocysteine S-methyltransferase (BHMT) is one of the most abundant proteins in the liver and regulates homocysteine metabolism. However, the molecular mechanisms underlying Bhmt transcription have not yet been elucidated. This study aimed to assess the molecular mechanisms underlying Bhmt transcription and the effect of BHMT deficiency on metabolic functions in the liver mediated by liver receptor homolog-1 (LRH-1). During fasting, both Bhmt and Lrh-1 expression increased in the liver of Lrh-1f/f mice; however, Bhmt expression was decreased in LRH-1 liver specific knockout mice. Promoter activity analysis confirmed that LRH-1 binds to a specific site in the Bhmt promoter region. LRH-1 deficiency was associated with elevated production of reactive oxygen species (ROS), lipid peroxidation, and mitochondrial stress in hepatocytes, contributing to hepatic triglyceride (TG) accumulation. In conclusion, this study suggests that the absence of an LRH-1-mediated decrease in Bhmt expression promotes TG accumulation by increasing ROS levels and inducing mitochondrial stress. Therefore, LRH-1 deficiency not only leads to excess ROS production and mitochondrial stress in hepatocytes, but also disrupts the methionine cycle. Understanding these regulatory pathways may pave the way for novel therapeutic interventions against metabolic disorders associated with hepatic lipid accumulation.


Subject(s)
Betaine-Homocysteine S-Methyltransferase , Hepatocytes , Liver , Methionine , Mice, Knockout , Reactive Oxygen Species , Receptors, Cytoplasmic and Nuclear , Triglycerides , Animals , Liver/metabolism , Mice , Reactive Oxygen Species/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Betaine-Homocysteine S-Methyltransferase/metabolism , Betaine-Homocysteine S-Methyltransferase/genetics , Hepatocytes/metabolism , Methionine/metabolism , Triglycerides/metabolism , Promoter Regions, Genetic/genetics , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Lipid Peroxidation
4.
Sci China Life Sci ; 67(8): 1648-1665, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38679670

ABSTRACT

Betaine-homocysteine methyltransferase (BHMT) regulates protein methylation and is correlated with tumorigenesis; however, the effects and regulation of BHMT in hepatocarcinogenesis remain largely unexplored. Here, we determined the clinical significance of BHMT in the occurrence and progression of hepatocellular carcinoma (HCC) using tissue samples from 198 patients. BHMT was to be frequently found (86.6%) expressed at relatively low levels in HCC tissues and was positively correlated with the overall survival of patients with HCC. Bhmt overexpression effectively suppressed several malignant phenotypes in hepatoma cells in vitro and in vivo, whereas complete knockout of Bhmt (Bhmt-/-) produced the opposite effect. We combined proteomics, metabolomics, and molecular biological strategies and detected that Bhmt-/- promoted hepatocarcinogenesis and tumor progression by enhancing the activity of glucose-6-phosphate dehydrogenase (G6PD) and PPP metabolism in DEN-induced HCC mouse and subcutaneous tumor-bearing models. In contrast, restoration of Bhmt with an AAV8-Bhmt injection or pharmacological inhibition of G6PD attenuated hepatocarcinogenesis. Additionally, coimmunoprecipitation identified monomethylated modifications of the G6PD, and BHMT regulated the methylation of G6PD. Protein sequence analysis, generation and application of specific antibodies, and site-directed mutagenesis indicated G6PD methylation at the arginine residue 246. Furthermore, we established bidirectionally regulated BHMT cellular models combined with methylation-deficient G6PD mutants to demonstrate that BHMT potentiated arginine methylation of G6PD, thereby inhibiting G6PD activity, which in turn suppressed hepatocarcinogenesis. Taken together, this study reveals a new methylation-regulatory mechanism in hepatocarcinogenesis owing to BHMT deficiency, suggesting a potential therapeutic strategy for HCC treatment.


Subject(s)
Betaine-Homocysteine S-Methyltransferase , Carcinogenesis , Carcinoma, Hepatocellular , Glucosephosphate Dehydrogenase , Liver Neoplasms , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Animals , Humans , Methylation , Betaine-Homocysteine S-Methyltransferase/metabolism , Betaine-Homocysteine S-Methyltransferase/genetics , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase/genetics , Mice , Carcinogenesis/genetics , Carcinogenesis/metabolism , Arginine/metabolism , Male , Cell Line, Tumor , Mice, Knockout , Gene Expression Regulation, Neoplastic
5.
Obesity (Silver Spring) ; 31(6): 1569-1583, 2023 06.
Article in English | MEDLINE | ID: mdl-37203331

ABSTRACT

OBJECTIVE: Betaine-homocysteine methyltransferase (Bhmt) belongs to the family of methyltransferases and is involved in the one-carbon metabolic cycle, which is associated with the risk of diabetes and adiposity. This study aimed to explore whether Bhmt participated in the development of obesity or its associated diabetes, as well as the mechanism involved. METHODS: The expression levels of Bhmt were examined in stromal vascular fraction cells and mature adipocytes in obesity and nonobesity. Knockdown and overexpression of Bhmt in C3H10T1/2 cells were used to investigate Bhmt's function in adipogenesis. Bhmt's role in vivo was analyzed using an adenovirus-expressing system and a high-fat diet-induced obesity mouse model. RESULTS: Bhmt was highly expressed in stromal vascular fraction cells rather than mature adipocytes of adipose tissue and was upregulated in adipose tissue in obesity and C3H10T1/2-commited preadipocytes. Overexpression of Bhmt promoted adipocyte commitment and differentiation in vitro and exacerbated adipose tissue expansion in vivo, with a concomitant increase in insulin resistance, whereas Bhmt silencing exhibited opposite effects. Mechanistically, Bhmt-induced adipose expansion was mediated by stimulating the p38 MAPK/Smad pathway. CONCLUSIONS: The findings of this study highlight the obesogenic and diabetogenic role of adipocytic Bhmt and propose Bhmt as a promising therapeutic target for obesity and obesity-related diabetes.


Subject(s)
Betaine-Homocysteine S-Methyltransferase , Insulin Resistance , Animals , Mice , Adipocytes/metabolism , Betaine-Homocysteine S-Methyltransferase/metabolism , Obesity/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Nutrients ; 15(8)2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37111122

ABSTRACT

Hsp70.1 has a dual function as a chaperone protein and lysosomal stabilizer. In 2009, we reported that calpain-mediated cleavage of carbonylated Hsp70.1 causes neuronal death by inducing lysosomal rupture in the hippocampal CA1 neurons of monkeys after transient brain ischemia. Recently, we also reported that consecutive injections of the vegetable oil-peroxidation product 'hydroxynonenal' induce hepatocyte death via a similar cascade in monkeys. As Hsp70.1 is also related to fatty acid ß-oxidation in the liver, its deficiency causes fat accumulation. The genetic deletion of betaine-homocysteine S-methyltransferase (BHMT) was reported to perturb choline metabolism, inducing a decrease in phosphatidylcholine and resulting in hepatic steatosis. Here, focusing on Hsp70.1 and BHMT disorders, we studied the mechanisms of hepatocyte degeneration and steatosis. Monkey liver tissues with and without hydroxynonenal injections were compared using proteomics, immunoblotting, immunohistochemical, and electron microscopy-based analyses. Western blotting showed that neither Hsp70.1 nor BHMT were upregulated, but an increased cleavage was observed in both. Proteomics showed a marked downregulation of Hsp70.1, albeit a two-fold increase in the carbonylated BHMT. Hsp70.1 carbonylation was negligible, in contrast to the ischemic hippocampus, which was associated with ~10-fold increments. Although histologically, the control liver showed very little lipid deposition, numerous tiny lipid droplets were seen within and around the degenerating/dying hepatocytes in monkeys after the hydroxynonenal injections. Electron microscopy showed permeabilization/rupture of lysosomal membranes, dissolution of the mitochondria and rough ER membranes, and proliferation of abnormal peroxisomes. It is probable that the disruption of the rough ER caused impaired synthesis of the Hsp70.1 and BHMT proteins, while impairment of the mitochondria and peroxisomes contributed to the sustained generation of reactive oxygen species. In addition, hydroxynonenal-induced disorders facilitated degeneration and steatosis in the hepatocytes.


Subject(s)
Betaine-Homocysteine S-Methyltransferase , Fatty Liver , Animals , Betaine-Homocysteine S-Methyltransferase/metabolism , Haplorhini/metabolism , Cell Death , Hepatocytes/metabolism , Ischemia , Liver/metabolism
7.
J Enzyme Inhib Med Chem ; 38(1): 2163242, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36629431

ABSTRACT

Androgen deprivation therapy (ADT) is a common treatment for recurrent prostate cancer (PC). However, after a certain period of responsiveness, ADT resistance occurs virtually in all patients and the disease progresses to lethal metastatic castration-resistant prostate cancer (mCRPC). Aberrant expression and function of the epigenetic modifiers EZH2 and BET over activates c-myc, an oncogenic transcription factor critically contributing to mCRPC. In the present work, we tested, for the first time, the combination of an EZH2 inhibitor with a BET inhibitor in metastatic PC cells. The combination outperformed single drugs in inhibiting cell viability, cell proliferation and clonogenic ability, and concomitantly reduced both c-myc and NF-kB expression. Although these promising results will warrant further in vivo validation, they represent the first step to establishing the rationale that the proposed combination might be suitable for mCRPC treatment, by exploiting molecular targets different from androgen receptor.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Transcription Factors , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/metabolism
8.
Nutr Rev ; 81(6): 716-726, 2023 05 10.
Article in English | MEDLINE | ID: mdl-36164833

ABSTRACT

The most important pathway in the development of folate-related pathologies is an increase in the level of homocysteine (HC). HC, a cytotoxic and neurotoxic amino acid (when its level is ≥12 µmol/L), is 1 of the most widely studied compounds in cardiology, neurobiology, oncology, and embryology for the last 20 years. Given its toxicity, the processes of endogenous detoxification of HC are of particular interest to medicine. To date, the most studied pathway is that of remethylation (the conversion of HC to methionine), with the participation of B12- and B9-dependent methionine synthase. Less studied is remethylation with the participation of the choline derivatives betaine and betaine-HC-S-methyltransferase (BHMT). Therefore, the aim of this review was to conduct a theoretical analysis of available information regarding the contribution of betaine metabolism, its enzyme, and its genetic polymorphism to folate metabolism disturbances, and the development of folate-related pathologies. This review emphasizes the potential clinical significance of 2 factors that can influence the remethylation reaction of HC: the use of betaine and identifying the BHMT gene variants and their impact on the risk for developing certain folate-related pathologies, and treatment options. Moreover, with a high level of methylation of the BHMT gene and in the presence of its low-function variants (eg, rs3733890), it is necessary to use betaine as an additional methyl donor, especially during folate therapy. More clinical research is needed to identify the effects of the different BHMT gene variants on the individual risk for folate-related pathologies to better assess the clinical significance, the need for genetic testing, and betaine consumption.


Subject(s)
Betaine , Folic Acid , Humans , Betaine/therapeutic use , Betaine/metabolism , Betaine-Homocysteine S-Methyltransferase/genetics , Betaine-Homocysteine S-Methyltransferase/metabolism , Methionine/pharmacology , Amino Acids , Homocysteine
9.
Int J Mol Sci ; 23(19)2022 Sep 25.
Article in English | MEDLINE | ID: mdl-36232598

ABSTRACT

Nutritional status and gene polymorphisms of one-carbon metabolism confer a well-known interaction that in pregnant women may affect embryo viability and the health of the newborn. Folate metabolism directly impacts nucleotide synthesis and methylation, which is of increasing interest in the reproductive medicine field. Studies assessing the genetic influence of folate metabolism on IVF treatments have currently been performed in women using their own oocytes. Most of these patients seeking to have a child or undergoing IVF treatments are advised to preventively intake folate supplies that restore known metabolic imbalances, but the treatments could lead to the promotion of specific enzymes in specific women, depending on their genetic variance. In the present study, we assess the influence of candidate gene variants related to folate metabolism, such as Serine Hydroxymethyltransferase 1 SHMT1 (rs1979276 and rs1979277), Betaine-Homocysteine S-Methyltransferase BHMT (rs3733890), Methionine synthase reductase MTRR (rs1801394), Methylenetetrahydrofolate reductase MTHFR (rs1801131 and rs1801133), methionine synthase MTR (rs12749581), ATP Binding Cassette Subfamily B Member 1 ABCB1 (rs1045642) and folate receptor alpha FOLR1 (rs2071010) on the success of IVF treatment performed in women being recipients of donated oocytes. The implication of such gene variants seems to have no direct impact on pregnancy consecution after IVF; however, several gene variants could influence pregnancy loss events or pregnancy maintenance, as consequence of folic acid fortification.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase , Methylenetetrahydrofolate Reductase (NADPH2) , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Adenosine Triphosphate , Betaine-Homocysteine S-Methyltransferase/genetics , Betaine-Homocysteine S-Methyltransferase/metabolism , Carbon/metabolism , Female , Ferredoxin-NADP Reductase/genetics , Ferredoxin-NADP Reductase/metabolism , Fertilization in Vitro , Folate Receptor 1/genetics , Folic Acid/metabolism , Genotype , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Nucleotides/metabolism , Oocytes/metabolism , Polymorphism, Single Nucleotide , Pregnancy
10.
Hum Cell ; 34(6): 1671-1681, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34410622

ABSTRACT

Down syndrome (DS) is the most common chromosomal disorder, resulting from the failure of normal chromosome 21 segregation. Studies have suggested that impairments within the one-carbon metabolic pathway can be of relevance for the global genome instability observed in mothers of individuals with DS. Based on the association between global DNA hypomethylation, genome instability, and impairments within the one-carbon metabolic pathway, the present study aimed to identify possible predictors, within the one-carbon metabolism, of global DNA methylation, measured by methylation patterns of LINE-1 and Alu repetitive sequences, in mothers of individuals with DS and mothers of individuals without the syndrome. In addition, we investigated one-carbon genetic polymorphisms and metabolites as maternal predisposing factors for the occurrence of trisomy 21 in children. Eighty-three samples of mothers of children with DS with karyotypically confirmed free trisomy 21 (case group) and 84 of mothers who had at least one child without DS or any other aneuploidy were included in the study. Pyrosequencing assays were performed to access global methylation. The results showed that group affiliation (case or control), betaine-homocysteine methyltransferase (BHMT) G742A and transcobalamin 2 (TCN2) C776G polymorphisms, and folate concentration were identified as predictors of global Alu DNA methylation values. In addition, thymidylate synthase (TYMS) 28-bp repeats 2R/3R or 3R/3R genotypes are independent maternal predisposing factors for having a child with DS. This study adds evidence that supports the association of impairments in the one-carbon metabolism, global DNA methylation, and the possibility of having a child with DS.


Subject(s)
Carbon/metabolism , DNA Methylation/genetics , Down Syndrome/genetics , Down Syndrome/metabolism , Genome-Wide Association Study , Genomic Instability/genetics , Mother-Child Relations , Mothers , Adolescent , Adult , Aged , Alu Elements/genetics , Betaine-Homocysteine S-Methyltransferase/genetics , Betaine-Homocysteine S-Methyltransferase/metabolism , Female , Folic Acid/metabolism , Genetic Predisposition to Disease/genetics , Humans , Long Interspersed Nucleotide Elements/genetics , Male , Middle Aged , Polymorphism, Genetic , Signal Transduction/genetics , Signal Transduction/physiology , Thymidylate Synthase/genetics , Transcobalamins/genetics , Transcobalamins/metabolism , Young Adult
11.
PLoS One ; 16(5): e0250486, 2021.
Article in English | MEDLINE | ID: mdl-33975330

ABSTRACT

Research into the epigenome is of growing importance as a loss of epigenetic control has been implicated in the development of neurodegenerative diseases. Previous studies have implicated aberrant DNA and histone methylation in multiple sclerosis (MS) disease pathogenesis. We have previously reported that the methyl donor betaine is depleted in MS and is linked to changes in histone H3 trimethylation (H3K4me3) in neurons. We have also shown that betaine increases histone methyltransferase activity by activating chromatin bound betaine homocysteine S-methyltransferase (BHMT). Here, we investigated the role of the BHMT-betaine methylation pathway in oligodendrocytes. Immunocytochemistry in the human MO3.13 cell line, primary rat oligodendrocytes, and tissue from MS postmortem brain confirmed the presence of the BHMT enzyme in the nucleus in oligodendrocytes. BHMT expression is increased 2-fold following oxidative insult, and qRT-PCR demonstrated that betaine can promote an increase in expression of oligodendrocyte maturation genes SOX10 and NKX-2.2 under oxidative conditions. Chromatin fractionation provided evidence of a direct interaction of BHMT on chromatin and co-IP analysis indicates an interaction between BHMT and DNMT3a. Our data show that both histone and DNA methyltransferase activity are increased following betaine administration. Betaine effects were shown to be dependent on BHMT expression following siRNA knockdown of BHMT. This is the first report of BHMT expression in oligodendrocytes and suggests that betaine acts through BHMT to modulate histone and DNA methyltransferase activity on chromatin. These data suggest that methyl donor availability can impact epigenetic changes and maturation in oligodendrocytes.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/metabolism , Betaine/metabolism , Multiple Sclerosis/pathology , Oligodendroglia/drug effects , Animals , Betaine/pharmacology , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/genetics , Brain/metabolism , Brain/pathology , Cells, Cultured , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic , Gene Expression/drug effects , Histones/metabolism , Humans , Methionine/metabolism , Methylation , Multiple Sclerosis/genetics , Nitroprusside/pharmacology , Oligodendroglia/cytology , Oligodendroglia/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , SOXE Transcription Factors/metabolism
12.
Nat Chem Biol ; 17(3): 326-334, 2021 03.
Article in English | MEDLINE | ID: mdl-33199915

ABSTRACT

Secreted polypeptides are a fundamental axis of intercellular and endocrine communication. However, a global understanding of the composition and dynamics of cellular secretomes in intact mammalian organisms has been lacking. Here, we introduce a proximity biotinylation strategy that enables labeling, detection and enrichment of secreted polypeptides in a cell type-selective manner in mice. We generate a proteomic atlas of hepatocyte, myocyte, pericyte and myeloid cell secretomes by direct purification of biotinylated secreted proteins from blood plasma. Our secretome dataset validates known cell type-protein pairs, reveals secreted polypeptides that distinguish between cell types and identifies new cellular sources for classical plasma proteins. Lastly, we uncover a dynamic and previously undescribed nutrient-dependent reprogramming of the hepatocyte secretome characterized by the increased unconventional secretion of the cytosolic enzyme betaine-homocysteine S-methyltransferase (BHMT). This secretome profiling strategy enables dynamic and cell type-specific dissection of the plasma proteome and the secreted polypeptides that mediate intercellular signaling.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/genetics , Biotin/chemistry , Blood Proteins/genetics , Hepatocytes/metabolism , Proteome/genetics , Staining and Labeling/methods , Animals , Betaine-Homocysteine S-Methyltransferase/metabolism , Biotin/administration & dosage , Biotinylation , Blood Proteins/metabolism , Gene Expression , HEK293 Cells , Hepatocytes/cytology , Humans , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Muscle Cells/cytology , Muscle Cells/metabolism , Myeloid Cells/cytology , Myeloid Cells/metabolism , Organ Specificity , Pericytes/cytology , Pericytes/metabolism , Proteome/metabolism , Proteomics/methods
13.
Eur J Pharmacol ; 891: 173697, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33144068

ABSTRACT

We investigated the role of protein arginine methylation (PAM) in estrogen receptor (ER)-positive breast cancer cells through pharmacological intervention. Tamoxifen (TAM) or adenosine dialdehyde (ADOX), independently, triggered cell cycle arrest and down-regulated PAM, as reduced protein arginine methyltransferase1 (PRMT1) mRNA and asymmetric dimethylarginine (ADMA) levels. Synergistic effect of these compounds elicited potent anti-cancer effect. However, reduction in ADMA was not proportionate with the compound-induced down-regulation of PRMT1 mRNA. We hypothesized that the disproportionate effect is due to the influence of the compounds on other methyltransferases, which catalyze the arginine dimethylation reaction and the diversity in the degree of drug-protein interaction among these methyltransferases. In silico analyses revealed that independently, ADOX or TAM, binds with phosphatidylethanolamine-methyltransferase (PEMT) or betaine homocysteine-methyl transferase (BHMT); and that the binding affinity of ADOX with PEMT or BHMT is prominent than TAM. These observations suggest that in breast cancer, synergistic effect of ADOX + TAM elicits impressive protective function by regulating PAM; and plausibly, restoration of normal enzyme activities of methyltransferases catalyzing arginine dimethylation could have clinical benefits.


Subject(s)
Adenosine/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Protein Processing, Post-Translational/drug effects , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Tamoxifen/pharmacology , Adenosine/metabolism , Adenosine/pharmacology , Antineoplastic Combined Chemotherapy Protocols/metabolism , Arginine/analogs & derivatives , Arginine/metabolism , Betaine-Homocysteine S-Methyltransferase/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Down-Regulation , Drug Synergism , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Methylation , Molecular Docking Simulation , Oxidative Stress/drug effects , Phosphatidylethanolamine N-Methyltransferase/metabolism , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Signal Transduction , Tamoxifen/metabolism
14.
Sci Rep ; 10(1): 14831, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32908189

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), a persistent environmental contaminant, induces steatosis that can progress to steatohepatitis with fibrosis, pathologies that parallel stages in the development of non-alcoholic fatty liver disease (NAFLD). Coincidently, one carbon metabolism (OCM) gene expression and metabolites are often altered during NAFLD progression. In this study, the time- and dose-dependent effects of TCDD were examined on hepatic OCM in mice. Despite AhR ChIP-seq enrichment at 2 h, OCM gene expression was not changed within 72 h following a bolus dose of TCDD. Dose-dependent repression of methionine adenosyltransferase 1A (Mat1a), adenosylhomocysteinase (Achy) and betaine-homocysteine S-methyltransferase (Bhmt) mRNA and protein levels following repeated treatments were greater at 28 days compared to 8 days. Accordingly, levels of methionine, betaine, and homocysteic acid were dose-dependently increased, while S-adenosylmethionine, S-adenosylhomocysteine, and cystathionine exhibited non-monotonic dose-dependent responses consistent with regulation by OCM intermediates and repression of glycine N-methyltransferase (Gnmt). However, the dose-dependent effects on SAM-dependent metabolism of polyamines and creatine could not be directly attributed to alterations in SAM levels. Collectively, these results demonstrate persistent AhR activation disrupts hepatic OCM metabolism at the transcript, protein and metabolite levels within context of TCDD-elicited progression of steatosis to steatohepatitis with fibrosis.


Subject(s)
Folic Acid/metabolism , Liver , Methionine/metabolism , Non-alcoholic Fatty Liver Disease , Polychlorinated Dibenzodioxins/toxicity , Adenosylhomocysteinase/metabolism , Animals , Betaine-Homocysteine S-Methyltransferase/metabolism , Carbon/metabolism , Disease Progression , Fibrosis , Glycine N-Methyltransferase/metabolism , Liver/metabolism , Liver/pathology , Methionine Adenosyltransferase/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology
15.
Epigenetics ; 15(8): 871-886, 2020 08.
Article in English | MEDLINE | ID: mdl-32096676

ABSTRACT

Methionine metabolism is dysregulated in multiple sclerosis (MS). The methyl donor betaine is depleted in the MS brain where it is linked to changes in levels of histone H3 trimethylated on lysine 4 (H3K4me3) and mitochondrial impairment. We investigated the effects of replacing this depleted betaine in the cuprizone mouse model of MS. Supplementation with betaine restored epigenetic control and alleviated neurological disability in cuprizone mice. Betaine increased the methylation potential (SAM/SAH ratio), levels of H3K4me3, enhanced neuronal respiration, and prevented axonal damage. We show that the methyl donor betaine and the betaine homocysteine methyltransferase (BHMT) enzyme can act in the nucleus to repair epigenetic control and activate neuroprotective transcriptional programmes. ChIP-seq data suggest that BHMT acts on chromatin to increase the SAM/SAH ratio and histone methyltransferase activity locally to increase H3K4me3 and activate gene expression that supports neuronal energetics. These data suggest that the methyl donor betaine may provide neuroprotection in MS where mitochondrial impairment damages axons and causes disability.


Subject(s)
Betaine/pharmacology , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Mitochondria/metabolism , Multiple Sclerosis/genetics , Animals , Betaine-Homocysteine S-Methyltransferase/metabolism , Cell Respiration , Cells, Cultured , Cuprizone/toxicity , Histone Code , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Multiple Sclerosis/etiology , Multiple Sclerosis/metabolism , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley
16.
J Dairy Sci ; 103(3): 2662-2676, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31864735

ABSTRACT

Precalving feeding level and body condition score (BCS) alter postcalving energy balance and oxidant status of dairy cows. We hypothesized that the reported benefits of a controlled restriction precalving depend on precalving BCS. The objective was to identify alterations in activity and intermediates of the hepatic one-carbon metabolism, transsulfuration, and tricarboxylic acid pathways. Twenty-eight pregnant and nonlactating grazing dairy cows of mixed age and breed (Friesian, Friesian × Jersey) were randomly allocated to 1 of 4 treatment groups in a 2 × 2 factorial design: 2 prepartum BCS categories [4.0 (thin, BCS4) and 5.0 (optimal, BCS5); 10-point scale], by managing cows in late lactation to achieve the 2 groups at dry-off, and 2 levels of energy intake during the 3 wk preceding calving (75 or 125% of estimated requirements), obtained via allowance (m2/cow) of fresh pasture composed of mostly perennial ryegrass and white cover. Average (± standard deviation) age was 6 ± 2, 6 ± 3, 5 ± 1, and 7 ± 3 yr for BCS4 fed 75 and 125%, and BCS5 fed 75 and 125%, respectively. Breed distribution (average ± standard deviation) for the 4 groups was 79 ± 21, 92 ± 11, 87 ± 31, and 74 ± 23% Friesian, and 17 ± 20, 8 ± 11, 13 ± 31, and 25 ± 23% Jersey. Liver tissue was collected by biopsy at -7, 7, and 28 d relative to calving. Tissue was used for 14C radio-labeling assays to measure betaine-homocysteine S-methyltransferase, 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR), and cystathionine-ß-synthase (CBS) activity. Liver metabolomics was undertaken using a targeted liquid chromatography with tandem mass spectrometry-based profiling approach. After initial liquid chromatography separation, mass spectra were acquired under both positive and negative ionization, whereas multiple reaction monitoring was used to measure target compound signal response (peak area count). Enzyme activity and metabolite peak area count were normalized with the homogenate protein concentration. Repeated measures analysis of variance via PROC MIXED in SAS (SAS Institute Inc., Cary, NC), with BCS, feeding, and time as fixed effects, and cow as random effect was used. All enzyme activities were affected by time, with betaine-homocysteine S-methyltransferase activity peaking at 7 d, whereas CBS and MTR activity decreased postpartum. Overall, thin cows had greater MTR activity, whereas cows fed 125% requirements had greater CBS activity. An interaction was detected between BCS and feeding for CBS activity, as thin cows fed 125% of requirements had greater overall activity. Compared with liver from BCS4 cows, BCS5 cows had overall greater betaine, glycine, butyrobetaine/acetylcholine, serine, and taurine concentrations. The same metabolites, plus choline and N-N-dimethylglycine, were overall greater in liver of cows fed 75% compared with those fed 125% of requirements. An interaction of BCS and feeding level was detected for the aforementioned metabolites plus methionine, cystathionine, cysteinesulfinate, and hypotaurine, due to greater overall concentrations in BCS5 cows fed 75% of requirements compared with other groups. Overall, differences in hepatic enzyme activity and intermediate metabolites suggest that both BCS and feeding level can alter the internal antioxidant system (e.g., glutathione and taurine) throughout the periparturient period. Further studies are needed to better understand potential mechanisms involved.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Betaine-Homocysteine S-Methyltransferase/metabolism , Cattle/physiology , Cystathionine beta-Synthase/metabolism , Energy Intake , Energy Metabolism , Animals , Antioxidants/metabolism , Carbon/metabolism , Cattle/genetics , Choline/metabolism , Diet/veterinary , Female , Homocysteine/metabolism , Lactation , Liver/enzymology , Metabolomics , Methionine/metabolism , Nutritional Status , Postpartum Period , Pregnancy
17.
Asia Pac J Clin Nutr ; 28(4): 879-887, 2019.
Article in English | MEDLINE | ID: mdl-31826386

ABSTRACT

BACKGROUND AND OBJECTIVES: Hyperhomocysteinaemia (HHcy) is an independent risk factors for several disorders, including cardiovascular disease. The understanding of the relationship among genetic, epigenetic and the efficacy of folate therapy for HHcy remain unclear. This study aim to investigate whether betaine-homocysteine methyltransferase (BHMT) single-nucleotide polymorphisms (SNPs) and DNA methylation are related to the efficacy of folate therapy for HHcy and whether BHMT DNA methylation mediates the SNP-folate therapy efficacy association. METHODS AND STUDY DESIGN: A total of 638 patients with HHcy were involved in this prospective cohort study. Logistic and linear regression was used to explore associations among SNPs, DNA methylation, and folate therapy efficacy. Finally, mediation analysis was performed to investigate whether DNA methylation of BHMT mediates the association between SNPs and folate therapy efficacy. RESULTS: BHMT rs3733890 was significantly associated with folate therapy efficacy (p<0.05). BHMT and BHMT_1 DNA methylation level was significantly associated with folate therapy efficacy (p=0.017 and p=0.028). DNA methylation of BHMT and BHMT_1 mediated 34.84% and 33.06% of the effect of rs3733890 on folate therapy efficacy, respectively. CONCLUSIONS: There has a consistent interrelationship among BHMT genetic variants, methylation levels of BHMT, and folate therapy efficacy. BHMT and BHMT_1 DNA methylation proportionally mediated the effects of rs3733890 SNPs on the efficacy of folate therapy for HHcy.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/metabolism , Epigenesis, Genetic , Folic Acid/therapeutic use , Gene Expression Regulation/drug effects , Hyperhomocysteinemia/drug therapy , Aged , Betaine-Homocysteine S-Methyltransferase/genetics , Cohort Studies , Female , Gene Expression Regulation/physiology , Genotype , Humans , Male , Middle Aged , Prospective Studies
18.
J Dairy Sci ; 102(11): 10291-10303, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31477291

ABSTRACT

Maternal supply of methyl donors such as methionine (Met) during late pregnancy can affect offspring growth and development. The objective was to investigate the effect of postruminal Met supply during late pregnancy on 1-carbon, Met cycle, and transsulfuration pathways in the calf liver. During the last 28 d of pregnancy, cows were individually fed a control diet or the control diet plus rumen-protected dl-Met (MET; 0.09% dry matter intake). Liver samples obtained from calves (n = 14/group) at 4, 14, 28, and 50 d of age were used for metabolomics, real-time PCR, and enzyme activity analyses. Genes associated with 1-carbon metabolism, DNA methylation, and the cytidine 5'-diphosphocholine-choline pathway were analyzed via real-time PCR. Activity of betaine homocysteine methyltransferase, cystathionine ß-synthase, and 5-methyltetrahydrofolate homocysteine methyltransferase (MTR) was analyzed using 14C isotopes. Data were analyzed using a mixed model that included the fixed effects of maternal treatment, day, and their interaction, and the random effect was calf within maternal diet. Calves born to dams offered MET tended to have greater birth body weight and had overall greater body weight during the first 9 wk of life. However, no differences were detected for daily feed intake and average daily gain between groups. Concentrations of betaine and choline, reflecting Met cycle activity, at d 14 through 28 were greater in MET calves. Transsulfuration pathway intermediates also were altered in MET calves, with concentrations of cysteine sulfinic acid and hypotaurine (d 4 and 14) and taurine being greater (d 4, 14, 28, and 50). Despite the lack of differences in daily feed intake, the greater concentrations of the tricarboxylic acid cycle intermediates fumarate and glutamate along with NAD/NADH in MET calves indicated enhanced rates of energy metabolism. Although activity of betaine homocysteine methyltransferase was greater in MET calves at d 14, cystathionine ß-synthase was lower and increased at d 14 and 28, where it was greater compared with the control diet. Activity of MTR was lower at d 4 and 50 in MET calves. Among gene targets measured, MET calves had greater overall expression of MTR, phosphatidylethanolamine N-methyltransferase, and choline kinase α and ß. An interaction of maternal diet by time was detected for mRNA abundance of DNA methyltransferase 3α (involved in de novo methylation) due to greater values at d 4 and 14 in MET calves. Overall, the data indicate that enhanced postruminal supply of Met to cows during late pregnancy may program hepatic metabolism of the calf in the context of maintaining Met homeostasis, phosphatidylcholine and taurine synthesis, DNA methylation, and energy metabolism. These alterations potentially result in better efficiency of nutrient use, hence conferring the calf a physiologic advantage during a period of rapid growth and development. The precise biologic mechanisms remain to be established.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/metabolism , Carbon/metabolism , Cattle/physiology , Energy Metabolism , Gene Expression Regulation, Enzymologic/drug effects , Methionine/administration & dosage , Animals , Animals, Newborn , Betaine/metabolism , Betaine-Homocysteine S-Methyltransferase/genetics , Biomarkers/metabolism , Cattle/genetics , Cattle/growth & development , Choline/metabolism , Diet/veterinary , Epigenesis, Genetic , Female , Liver/enzymology , Parturition , Pregnancy , Prenatal Nutritional Physiological Phenomena , RNA, Messenger/metabolism , Rumen/metabolism
19.
J Dairy Sci ; 102(9): 8305-8318, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31301838

ABSTRACT

Although choline requirements are unknown, enhanced postruminal supply may decrease liver triacylglycerol (TAG) storage and increase flux through the methionine cycle, helping cows during a negative energy balance (NEB). The objective was to investigate effects of postruminal choline supply during NEB on hepatic activity of betaine-homocysteine methyltransferase (BHMT), methionine synthase (MTR), methionine adenosyltransferase, transcription of enzymes, and metabolite concentrations in the methionine cycle. Ten primiparous rumen-cannulated Holstein cows (158 ± 24 d postpartum) were used in a replicated 5 × 5 Latin square design with 4-d treatment periods and 10 d of recovery (14 d/period). Treatments were unrestricted intake with abomasal infusion of water (A0), restricted intake (R; 60% of net energy for lactation requirements to induce NEB) with abomasal infusion of water (R0) or R plus abomasal infusion of 6.25, 12.5, or 25 g/d of choline ion. Liver tissue was collected on d 5 after the infusions ended, blood on d 1 to 5, and milk on d 1 to 4. Statistical contrasts were A0 versus R0 (CONT1) and tests of linear (L), quadratic (Q), and cubic (C) effects of choline dose. Plasma choline increased with R (CONT1) and choline (L). Although R decreased milk yield (CONT1), choline increased milk yield and liver phosphatidylcholine (PC), but decreased TAG (L). No differences were observed in plasma PC or very-low-density lipoprotein concentrations with R or choline. Activity and mRNA abundance of BHMT were greater with R (CONT1) and increased with choline (L). Although activity of MTR was lower with R (CONT1), it tended to increase with choline (L). No effect of R was detected for activity of methionine adenosyltransferase, but it changed cubically across dose of choline. Those responses were associated with linear increases in the concentrations of liver tissue (+13%) and plasma methionine concentrations. The mRNA abundance of CPT1A, SLC22A5, APOA5, and APOB, genes associated with fatty acid oxidation and lipoprotein metabolism, was upregulated by choline (Q). Overall, enhanced supply of choline during NEB increases hepatic activity of BHMT and MTR to regenerate methionine and PC, partly to help clear TAG. The relevance of these effects during the periparturient period merits further research.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Betaine-Homocysteine S-Methyltransferase/metabolism , Cattle/metabolism , Choline/administration & dosage , Energy Metabolism/drug effects , Liver/metabolism , Methionine/metabolism , Abomasum/drug effects , Animals , Betaine-Homocysteine S-Methyltransferase/genetics , Choline/blood , Fatty Acids/metabolism , Female , Lactation/drug effects , Lipid Metabolism/drug effects , Lipoproteins/metabolism , Liver/drug effects , Liver/enzymology , Methionine/blood , Oxidation-Reduction , Parturition/metabolism , Pregnancy , RNA, Messenger/analysis
20.
J Nutr ; 149(8): 1369-1376, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31111947

ABSTRACT

BACKGROUND: Hyperhomocysteinemia is associated with increased cardiovascular disease risk. Whole eggs contain several nutrients known to affect homocysteine regulation, including sulfur amino acids, choline, and B vitamins. OBJECTIVE: The aim of this study was to determine the effect of whole eggs and egg components (i.e., egg protein and choline) with respect to 1) homocysteine balance and 2) the hepatic expression and activity of betaine-homocysteine S-methyltransferase (BHMT) and cystathionine ß-synthase (CBS) in a folate-restricted (FR) rat model of hyperhomocysteinemia. METHODS: Male Sprague Dawley rats (n = 48; 6 wk of age) were randomly assigned to a casein-based diet (C; n = 12), a casein-based diet supplemented with choline (C + Cho; 1.3%, wt:wt; n = 12), an egg protein-based diet (EP; n = 12), or a whole egg-based diet (WE; n = 12). At week 2, half of the rats in each of the 4 dietary groups were provided an FR (0 g folic acid/kg) diet and half continued on the folate-sufficient (FS; 0.2 g folic acid/kg) diet for an additional 6 wk. All diets contained 20% (wt:wt) total protein. Serum homocysteine was measured by HPLC and BHMT and CBS expression and activity were evaluated using real-time quantitative polymerase chain reaction, Western blot, and enzyme activity. A 2-factor ANOVA was used for statistical comparisons. RESULTS: Rats fed FR-C exhibited a 53% increase in circulating homocysteine concentrations compared with rats fed FS-C (P < 0.001). In contrast, serum homocysteine did not differ between rats fed FS-C and FR-EP (P = 0.078). Hepatic BHMT activity was increased by 45% and 40% by the EP (P < 0.001) and WE (P = 0.002) diets compared with the C diets, respectively. CONCLUSIONS: Dietary intervention with egg protein prevented elevated circulating homocysteine concentrations in a rat model of hyperhomocysteinemia, due in part to upregulation of hepatic BHMT. These data may support the inclusion of egg protein for dietary recommendations targeting hyperhomocysteinemia prevention.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/metabolism , Egg Proteins, Dietary/administration & dosage , Folic Acid Deficiency/metabolism , Hyperhomocysteinemia/prevention & control , Liver/enzymology , Up-Regulation , Animals , Betaine-Homocysteine S-Methyltransferase/genetics , Body Weight , Cysteine/blood , Egg Proteins, Dietary/metabolism , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL