Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Mol Microbiol ; 119(2): 143-150, 2023 02.
Article in English | MEDLINE | ID: mdl-36350090

ABSTRACT

Methionine is a sulfur-containing residue found in most proteins which are particularly susceptible to oxidation. Although methionine oxidation causes protein damage, it can in some cases activate protein function. Enzymatic systems reducing oxidized methionine have evolved in most bacterial species and methionine oxidation proves to be a reversible post-translational modification regulating protein activity. In this review, we inspect recent examples of methionine oxidation provoking protein loss and gain of function. We further speculate on the role of methionine oxidation as a multilayer endogenous antioxidant system and consider its potential consequences for bacterial virulence.


Subject(s)
Methionine Sulfoxide Reductases , Methionine , Methionine/metabolism , Methionine Sulfoxide Reductases/genetics , Methionine Sulfoxide Reductases/metabolism , Oxidation-Reduction , Racemethionine/metabolism , Bacteria/metabolism , Protein Processing, Post-Translational
2.
Biochem Biophys Res Commun ; 701: 149589, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38309152

ABSTRACT

OBJECTIVE: To evaluate the role of PRDX2 in nonalcoholic steatohepatitis (NASH). METHODS: NASH was induced in wild-type (WT) mice and liver-specific PRDX2 knockout (PRDX2 LKO) mice that were fed a methionine-choline deficient diet (MCD) for 5 weeks. Assessments of PRDX2 LKO's impact on the pathogenesis of NASH include histological analyses, quantitative PCR (q-PCR), western blotting (WB), and RNA sequencing (RNA-Seq). RESULTS: PRDX2 LKO mice exhibited a significant increase in hepatic lipid accumulation and inflammation compared to WT mice after MCD feeding. PRDX2 KO markedly elevated circulating levels of aspartate aminotransferase (AST) and the pro-inflammatory signaling pathways within the liver. There was a notable increase in the activities of signal transducer and activator of transcription 1 (STAT1) and nuclear factor kappa B (NF-кB). We also found that PRDX2 KO significantly increased the extent of lipid peroxidation in the liver, most likely owing to the impaired peroxidase activity of PRDX2. Of interest, these findings were observed only in MCD-fed female mice, suggesting the sexual dimorphism of PRDX2 KO in MCD-induced NASH. CONCLUSION: PRDX2 deficiency increases MCD-induced NASH in female mice, suggesting a protective role for PRDX2.


Subject(s)
Choline Deficiency , Non-alcoholic Fatty Liver Disease , Mice , Female , Animals , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Choline/metabolism , Methionine/metabolism , Choline Deficiency/metabolism , Liver/metabolism , Racemethionine/metabolism , Diet , Mice, Knockout , Mice, Inbred C57BL
3.
Biol Chem ; 405(4): 257-265, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-37943731

ABSTRACT

The prevention and treatment of gastric cancer has been the focus and difficulty of medical research. We aimed to explore the mechanism of inhibiting migration and invasion of gastric cancer cells by methionine restriction (MR). The human gastric cancer cell lines AGS and MKN45 cultured with complete medium (CM) or medium without methionine were used for in vitro experiments. MKN45 cells were injected tail vein into BALB/c nude mice and then fed with normal diet or methionine diet for in vivo experiments. MR treatment decreased cell migration and invasion, increased E-cadherin expression, decreased N-cadherin and p-p65 expressions, and inhibited nuclear p65 translocation of AGS and MKN45 cells when compared with CM group. MR treatment increased IκBα protein expression and protein stability, and decreased IκBα protein ubiquitination level and TRIM47 expression. TRIM47 interacted with IκBα protein, and overexpression of TRIM47 reversed the regulatory effects of MR. TRIM47 promoted lung metastasis formation and partially attenuated the effect of MR on metastasis formation in vivo compared to normal diet group mice. MR reduces TRIM47 expression, leads to the degradation of IκBα, and then inhibits the translocation of nuclear p65 and the migration and invasion of gastric cancer cells.


Subject(s)
Stomach Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Methionine/metabolism , Methionine/pharmacology , Mice, Nude , Neoplasm Proteins/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-KappaB Inhibitor alpha/pharmacology , Nuclear Proteins/metabolism , Racemethionine/metabolism , Racemethionine/pharmacology , Stomach Neoplasms/metabolism , Tripartite Motif Proteins/metabolism
4.
Magn Reson Med ; 91(4): 1625-1636, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38115605

ABSTRACT

PURPOSE: Nonalcoholic fatty liver disease is an important cause of chronic liver disease. There are limited methods for monitoring metabolic changes during progression to steatohepatitis. Hyperpolarized 13 C MRSI (HP 13 C MRSI) was used to measure metabolic changes in a rodent model of fatty liver disease. METHODS: Fifteen Wistar rats were placed on a methionine- and choline-deficient (MCD) diet for 1-18 weeks. HP 13 C MRSI, T2 -weighted imaging, and fat-fraction measurements were obtained at 3 T. Serum aspartate aminotransaminase, alanine aminotransaminase, and triglycerides were measured. Animals were sacrificed for histology and measurement of tissue lactate dehydrogenase (LDH) activity. RESULTS: Animals lost significant weight (13.6% ± 2.34%), an expected characteristic of the MCD diet. Steatosis, inflammation, and mild fibrosis were observed. Liver fat fraction was 31.7% ± 4.5% after 4 weeks and 22.2% ± 4.3% after 9 weeks. Lactate-to-pyruvate and alanine-to-pyruvate ratios decreased significantly over the study course; were negatively correlated with aspartate aminotransaminase and alanine aminotransaminase (r = -[0.39-0.61]); and were positively correlated with triglycerides (r = 0.59-0.60). Despite observed decreases in hyperpolarized lactate signal, LDH activity increased by a factor of 3 in MCD diet-fed animals. Observed decreases in lactate and alanine hyperpolarized signals on the MCD diet stand in contrast to other studies of liver injury, where lactate and alanine increased. Observed hyperpolarized metabolite changes were not explained by alterations in LDH activity, suggesting that changes may reflect co-factor depletion known to occur as a result of oxidative stress in the MCD diet. CONCLUSION: HP 13 C MRSI can noninvasively measure metabolic changes in the MCD model of chronic liver disease.


Subject(s)
Choline Deficiency , Non-alcoholic Fatty Liver Disease , Rats , Animals , Mice , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Methionine/metabolism , Choline/metabolism , Pyruvic Acid/metabolism , Aspartic Acid/metabolism , Choline Deficiency/complications , Choline Deficiency/metabolism , Choline Deficiency/pathology , Rats, Wistar , Liver/metabolism , Racemethionine/metabolism , Diet , Triglycerides , Alanine/metabolism , Lactates/metabolism , Mice, Inbred C57BL , Disease Models, Animal
5.
Mol Genet Metab ; 141(1): 108111, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38103461

ABSTRACT

Methionine dependence, the inability to grow in culture when methionine in the medium is replaced by its metabolic precursor homocysteine, occurs in many tumor cell lines. In most affected lines, the cause of methionine dependence is not known. An exception is the melanoma-derived cell line MeWo-LC1, in which hypermethylation of the MMACHC gene is associated with decreased MMACHC expression. Decreased expression results in decreased provision of the methylcobalamin cofactor required for activity of methionine synthase and thus decreased conversion of homocysteine to methionine. Analysis of data in the Cancer Cell Line Encyclopedia Archive demonstrated that MMACHC hypermethylation and decreased MMACHC expression occurred more frequently in melanoma cell lines when compared to other tumor cell lines. We further investigated methionine dependence and aspects of MMACHC function in a panel of six melanoma lines, including both melanoma lines with known methionine dependence status (MeWo, which is methionine independent, and A375, which is methionine dependent). We found that the previously unclassified melanoma lines HMCB, Colo829 and SH-4 were methionine dependent, while SK-Mel-28 was methionine independent. However, despite varying levels of MMACHC methylation and expression, none of the tested lines had decreased methylcobalamin and adenosylcobalamin synthesis as seen in MeWo-LC1, and the functions of both cobalamin-dependent enzymes methionine synthase and methylmalonyl-CoA mutase were intact. Thus, while melanoma lines were characterized by relatively high levels of MMACHC methylation and low expression, the defect in metabolism observed in MeWo-LC1 was unique, and decreased MMACHC expression was not a cause of methionine dependence in the other melanoma lines.


Subject(s)
Melanoma , Methionine , Humans , Methionine/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Racemethionine/metabolism , Cell Line, Tumor , DNA Methylation , Homocysteine/metabolism , Vitamin B 12/metabolism , Oxidoreductases/metabolism
6.
Stem Cells ; 41(3): 271-286, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36472570

ABSTRACT

Human induced pluripotent stem cells (iPSCs) require high levels of methionine (Met). Met deprivation results in a rapid decrease in intracellular S-adenosyl-methionine (SAM), poising human iPSCs for differentiation and leading to the apoptosis of undifferentiated cells. Met deprivation triggers rapid metabolic changes, including SAM, followed by reversible epigenetic modifications. Here, we show that short-term Met deprivation impairs the pluripotency network through epigenetic modification in a 3D suspension culture. The trimethylation of lysine 4 on histone H3 (H3K4me3) was drastically affected compared with other histone modifications. Short-term Met deprivation specifically affects the transcription start site (TSS) region of genes, such as those involved in the transforming growth factor ß pathway and cholesterol biosynthetic process, besides key pluripotent genes such as NANOG and POU5F1. The expression levels of these genes decreased, correlating with the loss of H3K4me3 marks. Upon differentiation, Met deprivation triggers the upregulation of various lineage-specific genes, including key definitive endoderm genes, such as GATA6. Upon differentiation, loss of H3K27me3 occurs in many endodermal genes, switching from a bivalent to a monovalent (H3K4me3) state. In conclusion, Met metabolism maintains the pluripotent network with histone marks, and their loss potentiates differentiation.


Subject(s)
Induced Pluripotent Stem Cells , Methionine , Humans , Methionine/genetics , Methionine/metabolism , Induced Pluripotent Stem Cells/metabolism , Histone Code , Embryonic Stem Cells/metabolism , Cell Differentiation/genetics , Epigenesis, Genetic , Racemethionine/metabolism , S-Adenosylmethionine/metabolism
7.
Anal Bioanal Chem ; 416(4): 1011-1022, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38108841

ABSTRACT

Methionine and choline both are essential nutrients which are needed for methyl group metabolism. A methionine-choline-deficient (MCD) diet leads to pathological changes in the kidney. The mechanism of the MCD diet is complex, and fundamental research is still required to provide a better understanding of the driving forces behind it. We evaluated the regional effects of the MCD diet on the metabolites of mouse kidney tissue using desorption electrospray ionization mass spectrometry imaging technology. A total of 20, 17, and 13 metabolites were significantly changed in the cortex, outer medulla, and inner medulla, respectively, of the mouse kidney tissue after the administration of the MCD diet. Among the discriminating metabolites, only three metabolites (guanidoacetic acid, serine, and nicotinamide riboside) were significantly increased, and all the other metabolites showed a significant decrease. The results showed that there were significant region-specific changes in the serine metabolism, carnitine metabolism, choline metabolism, and arginine metabolism. This study presents unique regional metabolic data, providing a more comprehensive understanding of the molecular characteristics of the MCD diet in the kidney.


Subject(s)
Choline , Non-alcoholic Fatty Liver Disease , Mice , Animals , Choline/analysis , Methionine/metabolism , Racemethionine/metabolism , Racemethionine/pharmacology , Diet , Mass Spectrometry , Kidney/metabolism , Serine/metabolism , Liver/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism
8.
Acta Biochim Biophys Sin (Shanghai) ; 56(1): 62-70, 2024 01 25.
Article in English | MEDLINE | ID: mdl-38143381

ABSTRACT

Invasion and metastasis are the leading causes of death in individuals with malignant tumors, including gastric cancer. In this study, we aim to explore the effect and related mechanisms of methionine restriction (MR) on gastric carcinoma metastasis. In the MR cell model, gastric carcinoma cells are cultured in the MR medium, and in the animal model, BALB/c nude rodents are administered with a methionine-free diet after receiving injections of MKN45 cells into the caudal vein. Transwell assay is used to detect cell invasion and migration. Chromatin immunoprecipitation is performed to investigate the levels of H3K9me2, H3K27Ac, and H3K27me3 in the E-cadherin promoter. The results show that MR inhibits gastric carcinoma cell migration, invasion, and lung metastasis. MR increases E-cadherin while reducing the H3K27me3 level in the E-cadherin promoter. E-cadherin expression in gastric carcinoma cells is adversely regulated by HDAC2. Overexpressing HDAC2 reduces the H3K27Ac level in the E-cadherin promoter, while interfering with HDAC2 increases the H3K27Ac level. HDAC2 interference under MR conditions further upregulates E-cadherin expression and inhibits gastric carcinoma cell migration, invasion, and lung metastasis. MR combined with HDAC2 interference promotes E-cadherin expression by mediating the methylation and acetylation of E-cadherin, thus inhibiting the invasion, migration, and lung metastasis of gastric carcinoma cells. Our study provides a new theoretical basis for the inhibitory effect of MR on gastric cancer.


Subject(s)
Carcinoma , Lung Neoplasms , Stomach Neoplasms , Animals , Stomach Neoplasms/pathology , Up-Regulation , Histones/metabolism , Methionine/metabolism , Cadherins/genetics , Cadherins/metabolism , Lung Neoplasms/genetics , Racemethionine/metabolism , Cell Line, Tumor , Cell Movement , Neoplasm Invasiveness/genetics , Gene Expression Regulation, Neoplastic
9.
Biomed Chromatogr ; 38(1): e5763, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37858975

ABSTRACT

Alisol B 23-acetate (AB23A) has been demonstrated to have beneficial effects on nonalcoholic steatohepatitis (NASH). However, the mechanisms of AB23A on NASH remain unclear. This study aimed to investigate the mechanisms underlying the metabolic regulatory effects of AB23A on NASH. We used AB23A to treat mice with NASH, which was induced by a methionine and choline deficient (MCD) diet. We initially investigated therapeutic effect and resistance to oxidation and inflammation of AB23A on NASH. Subsequently, we performed untargeted metabolomic analyses and relative validation assessments to evaluate the metabolic regulatory effects of AB23A. AB23A reduced lipid accumulation, ameliorated oxidative stress and decreased pro-inflammatory cytokines in the liver. Untargeted metabolomic analysis found that AB23A altered the metabolites of liver. A total of 55 differential metabolites and three common changed pathways were screened among the control, model and AB23A treatment groups. Further tests validated the effects of AB23A on modulating common changed pathway-involved factors. AB23A treatment can ameliorate NASH by inhibiting oxidative stress and inflammation. The mechanism of AB23A on NASH may be related to the regulation of alanine, aspartate and glutamate metabolism, d-glutamine and d-glutamate metabolism, and arginine biosynthesis pathways.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Methionine/metabolism , Methionine/pharmacology , Choline , Liver/metabolism , Racemethionine/metabolism , Racemethionine/pharmacology , Diet , Inflammation/metabolism , Mice, Inbred C57BL , Disease Models, Animal
10.
J Dairy Sci ; 107(4): 2087-2098, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37923213

ABSTRACT

Low crude protein (CP) diets might be fed to dairy cows without affecting productivity if the balance of absorbed AA were improved, which would decrease the environmental effect of dairy farms. The aim of this study was to investigate the effects of supplementing ruminally protected Lys (RPL) and Met (RPM) at 2 levels of dietary CP on nutrient intake, milk production, milk composition, milk N efficiency (MNE), and plasma concentrations of AA in lactating Holstein cows and to evaluate these effects against the predictions of the new NASEM (2021) model. Fifteen multiparous cows were used in a replicated 3 × 3 Latin square design with 21-d periods. The 3 treatments were (1) a high-protein (HP) basal diet containing 16.4% CP (metabolizable protein [MP] balance of -130 g/d; 95% of target values), (2) a medium-protein diet containing 15% CP plus RPL (60 g/cow per day) and RPM (25 g/cow per day; MPLM; MP balance of -314 g/d; 87% of target values), and (3) a low-protein diet containing 13.6% CP plus RPL (60 g/cow per day) and RPM (25 g/cow per day; LPLM; MP balance of -479 g/d; 80% of target values). Dry matter intake was less for cows fed MPLM and LPLM diets compared with those fed the HP diet. Compared with the HP diet, the intake of CP, neutral detergent fiber, acid detergent fiber, and organic matter, but not starch, was lower for cows fed MPLM and LPLM diets. Milk production and composition were not affected by MPLM or LPLM diets relative to the HP diet. Milk urea N concentrations were reduced for the MPLM and LPLM diets compared with the HP diet, indicating that providing a low-protein diet supplemented with rumen-protected AA led to greater N efficiency. There was no significant effect of treatment on plasma AA concentrations except for proline, which significantly increased for the MPLM treatment compared with the other 2 treatments. Overall, the results supported the concept that milk performance might be maintained when feeding lactating dairy cows with low CP diets if the absorbed AA balance is maintained through RPL and RPM feeding. Further investigations are needed to evaluate responses over a longer time period with consideration of all AA rather than on the more aggregated MP and the ratio between Lys and Met.


Subject(s)
Lysine , Methionine , Female , Cattle , Animals , Diet, Protein-Restricted/veterinary , Lactation/physiology , Rumen/metabolism , Nitrogen/metabolism , Detergents/metabolism , Milk Proteins/metabolism , Diet/veterinary , Dietary Supplements , Milk/chemistry , Racemethionine/metabolism , Racemethionine/pharmacology , Dietary Proteins/metabolism
11.
Int J Mol Sci ; 25(3)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38338668

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is an inflammatory form of non-alcoholic fatty liver disease (NAFLD), closely associated with disease progression, cirrhosis, liver failure, and hepatocellular carcinoma. Time-restricted feeding (TRF) has been shown to decrease body weight and adiposity and improve metabolic outcomes; however, the effect of TRF on NASH has not yet been fully understood. We had previously reported that inositol polyphosphate multikinase (IPMK) mediates hepatic insulin signaling. Importantly, we have found that TRF increases hepatic IPMK levels. Therefore, we investigated whether there is a causal link between TRF and IPMK in a mouse model of NASH, i.e., methionine- and choline-deficient diet (MCDD)-induced steatohepatitis. Here, we show that TRF alleviated markers of NASH, i.e., reduced hepatic steatosis, liver triglycerides (TG), serum alanine transaminase (ALT) and aspartate aminotransferase (AST), inflammation, and fibrosis in MCDD mice. Interestingly, MCDD led to a significant reduction in IPMK levels, and the deletion of hepatic IPMK exacerbates the NASH phenotype induced by MCDD, accompanied by increased gene expression of pro-inflammatory chemokines. Conversely, TRF restored IPMK levels and significantly reduced gene expression of proinflammatory cytokines and chemokines. Our results demonstrate that TRF attenuates MCDD-induced NASH via IPMK-mediated changes in hepatic steatosis and inflammation.


Subject(s)
Choline Deficiency , Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Methionine/metabolism , Choline/metabolism , Choline Deficiency/complications , Choline Deficiency/metabolism , Liver/metabolism , Racemethionine/metabolism , Diet , Inflammation/metabolism , Chemokines/metabolism , Mice, Inbred C57BL , Disease Models, Animal
12.
Zhongguo Zhong Yao Za Zhi ; 49(5): 1310-1317, 2024 Mar.
Article in Zh | MEDLINE | ID: mdl-38621978

ABSTRACT

This study investigated the effect of Erchen Decoction(ECD) on the prevention of non-alcoholic steatohepatitis(NASH) in mice and explored its possible mechanism, so as to provide scientific data for the clinical application of ECD in the prevention of NASH. C57BL/6 male mice were randomly divided into normal group(methionine and choline supplement, MCS), model group(methionine and choline deficient, MCD), low-dose ECD group(ECD_L, 6 g·kg~(-1)), medium-dose ECD group(ECD_M, 12 g·kg~(-1)), and high-dose ECD group(ECD_H, 24 g·kg~(-1)), with eight mice in each group. The MCS group was fed with an MCS diet, and the other groups were fed with an MCD diet. The mice in each group were given corresponding diets, but the drug intervention group was given low-, medium-, and high-dose ECD(10 mL·kg~(-1)·d~(-1)) by intragastric administration for six weeks on the basis of MCD diet feeding, and the mice could eat and drink freely during the whole experiment. At the end of the experiment, mice were fasted overnight(12 h) and were anesthetized with 20% urethane. Thereafter, the blood and liver tissue were collected. The serum was used to detect the levels of alanine aminotransferase(ALT), aspartate aminotransaminase(AST), interleukin-1ß(IL-1ß), interleukin-6(IL-6), interleukin-10(IL-10), and tumor necrosis factor-α(TNF-α). Liver tissue was processed by hematoxylin-eosin(HE) staining and used for hepatic histological analysis and detection of the expression levels of genes and proteins related to nuclear factor erythroid 2-related factor 2/glutathione peroxidase 4(Nrf2/GPX4) pathway by real-time quantitative reverse transcriptase-polymerase chain reaction(RT-qPCR) and Western blot analysis, respectively. The results showed that compared with the MCS group, the MCD group showed higher serum ALT and AST levels; the HE staining exhibited fat vacuoles and obvious inflammatory cell infiltration in liver tissue; serum IL-1ß, IL-6, and TNF-α levels were significantly increased, and the serum IL-10 level was significantly decreased. The mRNA expressions of fatty acid synthase(FASN), monocyte chemoattractant protein-1(MCP-1), and IL-1ß in liver tissue were significantly up-regulated, while those of GPX4, Nrf2, and NAD(P)H:quinine oxidoreductase(NQO1) were significantly down-regulated. Compared with the MCD group, the serum ALT and AST levels of ECD_M and ECD_H groups were significantly decreased, and the AST level in the ECD_L group was significantly decreased. The number of fat vacuoles and the degree of inflammatory cell infiltration in liver tissue were improved; serum IL-1ß, IL-6, and TNF-α levels were significantly decreased, but the serum IL-10 level was significantly increased only in the ECD_H group. The mRNA expressions of FASN, MCP-1, and IL-1ß in liver tissue were significantly down-regulated, and those of GPX4 and NQO1 were significantly up-regulated. The mRNA expressions of Nrf2 in ECD_M and ECD_H groups were significantly up-regulated. Western blot results showed that compared with the MCD group, the protein expression levels of Nrf2 and GPX4 in each group were significantly increased after ECD administration, and the protein expression level of FASN was significantly decreased; the protein expression of NQO1 was increased in ECD_M and ECD_H groups. In summary, ECD can reduce hepatic lipid accumulation, oxidative stress, liver inflammation, and liver injury in NASH mice, which may be related to the activation of the Nrf2/GPX4 pathway.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Male , Animals , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Methionine/metabolism , Methionine/pharmacology , Interleukin-10/genetics , Choline/metabolism , Choline/pharmacology , Choline/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , NF-E2-Related Factor 2/metabolism , Mice, Inbred C57BL , Liver , Racemethionine/metabolism , Racemethionine/pharmacology , Diet , RNA, Messenger/metabolism
13.
Gut ; 72(3): 501-511, 2023 03.
Article in English | MEDLINE | ID: mdl-35803704

ABSTRACT

OBJECTIVE: Methionine metabolism is involved in a myriad of cellular functions, including methylation reactions and redox maintenance. Nevertheless, it remains unclear whether methionine metabolism, RNA methylation and antitumour immunity are molecularly intertwined. DESIGN: The antitumour immunity effect of methionine-restricted diet (MRD) feeding was assessed in murine models. The mechanisms of methionine and YTH domain-containing family protein 1 (YTHDF1) in tumour immune escape were determined in vitro and in vivo. The synergistic effects of MRD or YTHDF1 depletion with PD-1 blockade were also investigated. RESULTS: We found that dietary methionine restriction reduced tumour growth and enhanced antitumour immunity by increasing the number and cytotoxicity of tumour-infiltrating CD8+ T cells in different mouse models. Mechanistically, the S-adenosylmethionine derived from methionine metabolism promoted the N6-methyladenosine (m6A) methylation and translation of immune checkpoints, including PD-L1 and V-domain Ig suppressor of T cell activation (VISTA), in tumour cells. Furthermore, MRD or m6A-specific binding protein YTHDF1 depletion inhibited tumour growth by restoring the infiltration of CD8+ T cells, and synergised with PD-1 blockade for better tumour control. Clinically, YTHDF1 expression correlated with poor prognosis and immunotherapy outcomes for cancer patients. CONCLUSIONS: Methionine and YTHDF1 play a critical role in anticancer immunity through regulating the functions of T cells. Targeting methionine metabolism or YTHDF1 could be a potential new strategy for cancer immunotherapy.


Subject(s)
Methionine , Neoplasms , Mice , Animals , Methionine/metabolism , CD8-Positive T-Lymphocytes , Methylation , Programmed Cell Death 1 Receptor , Racemethionine/metabolism
14.
J Mol Cell Cardiol ; 175: 49-61, 2023 02.
Article in English | MEDLINE | ID: mdl-36528076

ABSTRACT

BACKGROUND: Available evidence suggest that Ca2+/calmodulin-dependent protein kinase type IIδ (CaMKIIδ) and reactive oxygen species (ROS) are important in early ischemia-reperfusion arrhythmias (IRA). Since ROS can activate CaMKIIδ by oxidation of two methionines at positions 281/282, oxidized-CaMKIIδ (Ox-CaMKIIδ) has been proposed to be important for IRA. However, direct evidence for this is missing. METHODS: We exposed Langendorff-perfused hearts and ventricular cardiomyocytes from C57BL/6 mice to global and simulated ischemia, respectively, and recorded arrhythmic events during early reperfusion. Hearts were collected for immunoblotting of key phosphoproteins. We evaluated the effects of beta-adrenoceptor stimulation, inhibition of CaMKII, and reduced ROS levels with isoprenaline, KN93/AIP and N-acetylcysteine (NAC), respectively. We further tested the importance of Ox-CaMKIIδ by using hearts and cardiomyocytes from mice with CaMKIIδ resistant to oxidation of methionines 281 and 282 (MMVV). RESULTS: Hearts treated with KN93, AIP or NAC had lower incidence of early IRA, and NAC-treated cardiomyocytes had lower incidence of arrhythmogenic events. However, hearts from MMVV mice had a similar incidence of early IRA to wild type mice (WT), and MMVV and WT cardiomyocytes had a similar frequency of Ca2+ waves and Ca2+ sparks. Immunoblotting confirmed high levels of oxidation in early reperfusion, but revealed no significant differences in the phosphorylation levels of Ca2+-handling proteins in MMVV and WT hearts. CONCLUSIONS: Although CaMKII and ROS both contribute to early IRA, hearts from mice with CaMKII resistant to oxidation at methionines 281/282 were not protected from such arrhythmias, suggesting that oxidation at these sites is not a determining factor.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Methionine , Mice , Animals , Reactive Oxygen Species/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Methionine/metabolism , Mice, Inbred C57BL , Arrhythmias, Cardiac/metabolism , Myocytes, Cardiac/metabolism , Racemethionine/metabolism , Reperfusion/adverse effects , Phosphorylation
15.
Biochemistry ; 62(23): 3343-3346, 2023 12 05.
Article in English | MEDLINE | ID: mdl-38009918

ABSTRACT

Family 1 glycosyltransferases (GT1s, UGTs) catalyze the regioselective glycosylation of natural products in a single step. We identified GmUGT88E3 as a particularly promising biocatalyst able to produce a variety of pure, single glycosidic products from polyphenols with high chemical yields. We investigated this particularly desirable duality toward specificity, i.e., promiscuous toward acceptors while regiospecific. Using high-field NMR, kinetic characterization, molecular dynamics simulations, and mutagenesis studies, we uncovered that the main molecular determinant of GmUGT88E3 specificity is a methionine-aromatic bridge, an interaction often present in protein structures but never reported for enzyme-substrate interactions. Here, mutating Met127 led to inactive proteins or 100-fold reduced activity.


Subject(s)
Glycine max , Glycosyltransferases , Glycosyltransferases/metabolism , Glycine max/genetics , Methionine/metabolism , Glycosylation , Glycosides , Racemethionine/metabolism , Substrate Specificity
16.
Mol Microbiol ; 118(4): 387-402, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36271735

ABSTRACT

Repairing oxidative-targeted macromolecules is a central mechanism necessary for living organisms to adapt to oxidative stress. Reactive oxygen and chlorine species preferentially oxidize sulfur-containing amino acids in proteins. Among these amino acids, methionine can be converted into methionine sulfoxide. This post-translational oxidation can be reversed by methionine sulfoxide reductases, Msr enzymes. In Gram-negative bacteria, the antioxidant MsrPQ system is involved in the repair of periplasmic oxidized proteins. Surprisingly, in this study, we observed in Escherichia coli that msrPQ was highly expressed in the absence of oxygen. We have demonstrated that the anaerobic induction of msrPQ was due to chlorate (ClO3 - ) contamination of the Casamino Acids. Molecular investigation led us to determine that the reduction of chlorate to the toxic oxidizing agent chlorite (ClO2 - ) by the three nitrate reductases (NarA, NarZ, and Nap) led to methionine oxidation of periplasmic proteins. In response to this stress, the E. coli HprSR two-component system was activated, leading to the over-production of MsrPQ. This study, therefore, supports the idea that methionine oxidation in proteins is part of chlorate toxicity, and that MsrPQ can be considered as an anti-chlorate/chlorite defense system in bacteria. Finally, this study challenges the traditional view of the absence of Met-oxidation during anaerobiosis.


Subject(s)
Escherichia coli , Periplasmic Proteins , Escherichia coli/metabolism , Methionine Sulfoxide Reductases/metabolism , Periplasmic Proteins/metabolism , Anaerobiosis , Chlorine/metabolism , Antioxidants/metabolism , Oxidation-Reduction , Methionine/metabolism , Racemethionine/metabolism , Oxygen/metabolism , Oxidants/metabolism , Sulfur/metabolism
17.
Amino Acids ; 55(1): 139-152, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36371728

ABSTRACT

Sulfur-containing amino acids such as methionine and cysteine play critical roles in immune system and redox status. A body of evidence shows that metabolic aspects of supplemented Met and Cys may differ in the body. Therefore, the study aimed to investigate the effects of dietary Met and Cys supplementation in immunologically challenged weaned pigs. Forty weaned piglets (6.5 ± 0.3 kg) were randomly allocated to five treatment groups. The treatment included: (1) sham-challenged control (SCC), (2) challenged control (CC), (3) MET (CC + 0.1% DL-Met), (4) CYS (CC + 0.1% L-Cys), and (5) MET + CYS (CC + 0.1% DL-Met + 0.1% L-Cys). On day 7, all pigs were intramuscularly injected with either Escherichia coli O55:B5 lipopolysaccharides (LPS) or phosphate-buffered saline. Blood, liver, and jejunum samples were analyzed for immune response and redox status. The CC group had lower (P < 0.05) villus surface area and higher (P < 0.05) flux of 4-kDa fluorescein isothiocyanate dextran (FD4) than the SCC group. A lower (P < 0.05) glutathione (GSH) concentration was observed in the jejunum of pigs in the CC group than those in the SCC group. Dietary Cys supplementation increased (P < 0.05) villus surface area, GSH levels, and reduced (P < 0.05) the flux of FD4 in the jejunum of LPS-challenged pigs. Dietary Met supplementation enhanced (P < 0.05) hepatic GSH content. Pigs challenged with LPS in the MET group had lower serum IL-8 concentration than those in the CC group. There was a Met × Cys interaction (P < 0.05) in serum IL-4 and IL-8 concentrations, and Trolox equivalent antioxidant capacity. Dietary L-Cys supplementation restored intestinal integrity and GSH levels that were damaged by lipopolysaccharides administration. Dietary DL-Met supplementation improved hepatic GSH and reduced systemic inflammatory response, but antagonistic interaction with dietary L-Cys supplementation was observed in the inflammatory response and redox status.


Subject(s)
Cysteine , Methionine , Swine , Animals , Methionine/metabolism , Cysteine/pharmacology , Cysteine/metabolism , Interleukin-8 , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Diet/veterinary , Dietary Supplements , Glutathione/metabolism , Oxidation-Reduction , Racemethionine/metabolism , Animal Feed/analysis , Weaning
18.
Amino Acids ; 55(8): 1039-1048, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37326859

ABSTRACT

Dietary supplementation with methionine and threonine spares body protein in rats fed a low protein diet, but the effect is not observed for other essential amino acids. Although the requirement for sulfur amino acids is relatively high in rodents, the precise mechanisms underlying protein retention are not fully understood. The aim of this study was to explore whether the activation of mammalian target of rapamycin complex 1 (mTORC1) downstream factors in skeletal muscle by supplementation with threonine and/or methionine contributes to protein retention under sufficient cystine requirement. Male Sprague-Dawley rats were freely fed a 0% protein diet for 2 weeks. These experimental rats were then fed a restricted diet (14.5 g/day) containing 12% soy protein supplemented with both cystine and, methionine and threonine (MT), methionine (M), threonine (T), or neither (NA) (n = 8) for an additional 12 days. Two additional groups were freely fed a diet containing 0% protein or 20% casein as controls (n = 6). Body weight and gastrocnemius muscle weight were higher, and blood urea nitrogen and urinary nitrogen excretion were lower, in the M and MT groups than in the T and NA groups, respectively. p70 S6 kinase 1 abundance was higher, and eukaryotic translation initiation factor 4E-binding protein 1 abundance and mRNA levels were lower, in the skeletal muscles of the M and MT groups. These results suggest that methionine regulates mTORC1 downstream factors in skeletal muscle, leading to spare body protein in rats fed a low protein diet meeting cystine requirements.


Subject(s)
Amino Acids, Sulfur , Methionine , Rats , Male , Animals , Methionine/metabolism , Amino Acids, Sulfur/analysis , Amino Acids, Sulfur/metabolism , Soybean Proteins/pharmacology , Pilot Projects , Cystine , Rats, Sprague-Dawley , Liver/metabolism , Diet , Racemethionine/metabolism , Dietary Supplements , Mechanistic Target of Rapamycin Complex 1/metabolism , Threonine/metabolism , Mammals/metabolism
19.
Microb Cell Fact ; 22(1): 151, 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37568230

ABSTRACT

Methionine is an essential amino acid in mammals and a precursor for vital metabolites required for the survival of all organisms. Consequently, its inclusion is required in diverse applications, such as food, feed, and pharmaceuticals. Although amino acids and other metabolites are commonly produced through microbial fermentation, high-yield biosynthesis of L-methionine remains a significant challenge due to the strict cellular regulation of the biosynthesis pathway. As a result, methionine is produced primarily synthetically, resulting in a racemic mixture of D,L-methionine. This study explores methionine bio-production in E. coli by replacing its native trans-sulfurylation pathway with the more common direct-sulfurylation pathway used by other bacteria. To this end, we generated a methionine auxotroph E. coli strain (MG1655) by simultaneously deleting metA and metB genes and complementing them with metX and metY from different bacteria. Complementation of the genetically modified E. coli with metX/metY from Cyclobacterium marinum or Deinococcus geothermalis, together with the deletion of the global repressor metJ and overexpression of the transporter yjeH, resulted in a substantial increase of up to 126 and 160-fold methionine relative to the wild-type strain, respectively, and accumulation of up to 700 mg/L using minimal MOPS medium and 2 ml culture. Our findings provide a method to study methionine biosynthesis and a chassis for enhancing L-methionine production by fermentation.


Subject(s)
Escherichia coli , Methionine , Escherichia coli/genetics , Escherichia coli/metabolism , Methionine/metabolism , Bacteria/metabolism , Fermentation , Racemethionine/metabolism , Metabolic Engineering/methods
20.
J Pept Sci ; 29(3): e3454, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36181422

ABSTRACT

Considering the fact that site-selective late-stage diversification of peptides and proteins remains a challenge for biochemistry, strategies targeting low-abundance natural amino acids need to be further developed. As an extremely oxidation-sensitive and low-abundance amino acid, methionine emerges as a promising target for chemo- and site-selective modification. Herein we report an efficient and highly selective modification on methionine residues by one-pot O- and N-transfer reaction, generating sulfoximine-modified peptides with near-perfect conversion within 10 min. Moreover, the great tolerance to other natural amino acids has been demonstrated in reactions with various peptide substrates. To demonstrate the generality of this protocol, we have modified natural peptides and obtained sulfoximination products with high conversion rates. This methodology provides a novel strategy as the expansion of the methionine-based peptide functionalization toolbox.


Subject(s)
Methionine , Proteins , Methionine/chemistry , Methionine/metabolism , Proteins/chemistry , Peptides/chemistry , Racemethionine/metabolism , Oxidative Stress
SELECTION OF CITATIONS
SEARCH DETAIL