Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circ Res ; 130(10): 1565-1582, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35410483

ABSTRACT

BACKGROUND: S-adenosylhomocysteine (SAH) is a risk factor of cardiovascular disease; inhibition of SAH hydrolase (SAHH) results in SAH accumulation and induces endothelial dysfunction and atherosclerosis. However, the effect and mechanism of SAHH in atherosclerotic calcification is still unclear. We aimed to explore the role and mechanism of SAHH in atherosclerotic calcification. METHODS: The relationship between SAHH and atherosclerotic calcification was investigated in patients with coronary atherosclerotic calcification. Different in vivo genetic models were used to examine the effect of SAHH deficiency on atherosclerotic calcification. Human aortic and murine vascular smooth muscle cells (VSMCs) were cultured to explore the underlying mechanism of SAHH on osteoblastic differentiation of VSMCs. RESULTS: The expression and activity of SAHH were decreased in calcified human coronary arteries and inversely associated with coronary atherosclerotic calcification severity, whereas plasma SAH and total homocysteine levels were positively associated with coronary atherosclerotic calcification severity. Heterozygote knockout of SAHH promoted atherosclerotic calcification. Specifically, VSMC-deficient but not endothelial cell-deficient or macrophage-deficient SAHH promoted atherosclerotic calcification. Mechanistically, SAHH deficiency accumulated SAH levels and induced H19-mediated Runx2 (runt-related transcription factor 2)-dependent osteoblastic differentiation of VSMCs by inhibiting DNMT3b (DNA methyltransferase 3b) and leading to hypomethylation of the H19 promoter. On the contrary, SAHH deficiency resulted in lower intracellular levels of adenosine and reduced AMPK (AMP-activated protein kinase) activation. Adenosine supplementation activated AMPK and abolished SAHH deficiency-induced expression of H19 and Runx2 and osteoblastic differentiation of VSMCs. Finally, AMPK activation by adenosine inhibited H19 expression by inducing Sirt1 (sirtuin-1)-mediated histone H3 hypoacetylation and DNMT3b-mediated hypermethylation of the H19 promoter in SAHH deficiency VSMCs. CONCLUSIONS: We have confirmed a novel correlation between SAHH deficiency and atherosclerotic calcification and clarified a new mechanism that epigenetic upregulation of H19 and AMPK inhibition concurrently contribute to SAHH deficiency-promoted Runx2-dependent atherosclerotic calcification.


Subject(s)
Atherosclerosis , Calcinosis , Vascular Calcification , AMP-Activated Protein Kinases/metabolism , Adenosine/metabolism , Amino Acid Metabolism, Inborn Errors , Animals , Atherosclerosis/metabolism , Calcinosis/genetics , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Epigenesis, Genetic , Glycine N-Methyltransferase/deficiency , Humans , Mice , Myocytes, Smooth Muscle/metabolism , RNA, Long Noncoding , S-Adenosylhomocysteine/metabolism , Up-Regulation , Vascular Calcification/genetics , Vascular Calcification/metabolism
2.
Int J Mol Sci ; 25(12)2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38928097

ABSTRACT

Tissue hypoxia is associated with the development of organ dysfunction and death in critically ill patients commonly captured using blood lactate. The kinetic parameters of serial lactate evaluations are superior at predicting mortality compared with single values. S-adenosylhomocysteine (SAH), which is also associated with hypoxia, was recently established as a useful predictor of septic organ dysfunction and death. We evaluated the performance of kinetic SAH parameters for mortality prediction compared with lactate parameters in a cohort of critically ill patients. For lactate and SAH, maxima and means as well as the normalized area scores were calculated for two periods: the first 24 h and the total study period of up to five days following ICU admission. Their performance in predicting in-hospital mortality were compared in 99 patients. All evaluated parameters of lactate and SAH were significantly higher in non-survivors compared with survivors. In univariate analysis, the predictive power for mortality of SAH was higher compared with lactate in all forms of application. Multivariable models containing SAH parameters demonstrated higher predictive values for mortality than models based on lactate parameters. The optimal models for mortality prediction incorporated both lactate and SAH parameters. Compared with lactate, SAH displayed stronger predictive power for mortality in static and dynamic application in critically ill patients.


Subject(s)
Critical Illness , Lactic Acid , S-Adenosylhomocysteine , Humans , Critical Illness/mortality , Male , Female , Lactic Acid/blood , Middle Aged , Aged , S-Adenosylhomocysteine/blood , Hospital Mortality , Kinetics , Prognosis , Biomarkers/blood , Cohort Studies , Intensive Care Units , Adult
3.
Biochem Biophys Res Commun ; 686: 149152, 2023 12 17.
Article in English | MEDLINE | ID: mdl-37926042

ABSTRACT

S-adenosylhomocysteine (SAH) hydrolase is the enzyme responsible for breaking down SAH into adenosine and homocysteine. It has long been believed that a deficiency of this enzyme leads to SAH accumulation, subsequently inhibiting methyltransferases responsible for nucleic acids and proteins, which severely affects cell proliferation. To investigate whether targeting this enzyme could be a viable strategy to combat Trypanosoma brucei, the causative agent of human African trypanosomiasis, we created a null mutant of the SAH hydrolase gene in T. brucei using the Cre/loxP system and conducted a phenotype analysis. Surprisingly, the null mutant, where all five SAH hydrolase gene loci were deleted, exhibited normal proliferation despite the observed SAH accumulation. These findings suggest that inhibiting SAH hydrolase may not be an effective approach to suppressing T. brucei proliferation, making the enzyme a less promising target for antitrypanosome drug development.


Subject(s)
Trypanosoma brucei brucei , Humans , Adenosylhomocysteinase/genetics , Adenosylhomocysteinase/metabolism , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism , S-Adenosylhomocysteine/metabolism , Adenosine/genetics , Adenosine/pharmacology
4.
Cardiovasc Diabetol ; 22(1): 219, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37620823

ABSTRACT

BACKGROUND: Clinical observations suggest a complex relationship between obesity and coronary artery disease (CAD). This study aimed to characterize the intermediate metabolism phenotypes among obese patients with CAD and without CAD. METHODS: Sixty-two participants who consecutively underwent coronary angiography were enrolled in the discovery cohort. Transcriptional and untargeted metabolomics analyses were carried out to screen for key molecular changes between obese patients with CAD (CAD obese), without CAD (Non-CAD obese), and Non-CAD leans. A targeted GC-MS metabolomics approach was used to further identify differentially expressed metabolites in the validation cohorts. Regression and receiver operator curve analysis were performed to validate the risk model. RESULTS: We found common aberrantly expressed pathways both at the transcriptional and metabolomics levels. These pathways included cysteine and methionine metabolism and arginine and proline metabolism. Untargeted metabolomics revealed that S-adenosylhomocysteine (SAH), 3-hydroxybenzoic acid, 2-hydroxyhippuric acid, nicotinuric acid, and 2-arachidonoyl glycerol were significantly elevated in the CAD obese group compared to the other two groups. In the validation study, targeted cysteine and methionine metabolomics analyses showed that homocysteine (Hcy), SAH, and choline were significantly increased in the CAD obese group compared with the Non-CAD obese group, while betaine, 5-methylpropanedioic acid, S-adenosylmethionine, 4-PA, and vitamin B2 (VB2) showed no significant differences. Multivariate analyses showed that Hcy was an independent predictor of obesity with CAD (hazard ratio 1.7; 95%CI 1.2-2.6). The area under the curve based on the Hcy metabolomic (HCY-Mtb) index was 0.819, and up to 0.877 for the HCY-Mtb.index plus clinical variables. CONCLUSION: This is the first study to propose that obesity with hyperhomocysteinemia is a useful intermediate metabolism phenotype that could be used to identify obese patients at high risk for developing CAD.


Subject(s)
Coronary Artery Disease , Hyperhomocysteinemia , Obesity , Humans , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/etiology , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Cross-Sectional Studies , Cysteine , East Asian People , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , Metabolomics , Obesity/complications , Obesity/genetics , Obesity/metabolism , Prospective Studies , Risk Factors , Transcriptome , Coronary Angiography , Cardiometabolic Risk Factors , Adult , Middle Aged , Aged
5.
Anal Biochem ; 666: 115047, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36682579

ABSTRACT

Due to the emergence of multidrug resistant pathogens, it is imperative to identify new targets for antibiotic drug discovery. The S-adenosylhomocysteine (SAH) nucleosidase enzyme is a promising target for antimicrobial drug development due to its critical functions in multiple bacterial processes including recycling of toxic byproducts of S-adenosylmethionine (SAM)-mediated reactions and producing the precursor of the universal quorum sensing signal, autoinducer-2 (AI-2). Riboswitches are structured RNA elements typically used by bacteria to precisely monitor and respond to changes in essential bacterial processes, including metabolism. Natural riboswitches fused to a reporter gene can be exploited to detect changes in metabolism or in physiological signaling. We performed a high-throughput screen (HTS) using an SAH-riboswitch controlled ß-galactosidase reporter gene in Escherichia coli to discover small molecules that inhibit SAH recycling. We demonstrate that the assay strategy using SAH riboswitches to detect the effects of SAH nucleosidase inhibitors can quickly identify compounds that penetrate the barriers of Gram-negative bacterial cells and perturb pathways involving SAH.


Subject(s)
Riboswitch , S-Adenosylmethionine/metabolism , RNA/genetics , Bacteria/genetics , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism
6.
Int J Mol Sci ; 24(16)2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37628779

ABSTRACT

A common final pathway of pathogenetic mechanisms in septic organ dysfunction and death is a lack or non-utilization of oxygen. Plasma concentrations of lactate serve as surrogates for the oxygen-deficiency-induced imbalance between energy supply and demand. As S-adenosylhomocysteine (SAH) was shown to reflect tissue hypoxia, we compared the ability of SAH versus lactate to predict the progression of inflammatory and septic disease to septic organ dysfunction and death. Using univariate and multiple logistic regression, we found that SAH but not lactate, taken upon patients' inclusion in the study close to ICU admission, significantly and independently contributed to the prediction of disease progression and death. Due to the stronger increase in SAH in relation to S-adenosylmethionine (SAM), the ratio of SAM to SAH, representing methylation potential, was significantly decreased in patients with septic organ dysfunction and non-survivors compared with SIRS/sepsis patients (2.8 (IQR 2.3-3.9) vs. 8.8 (4.9-13.8); p = 0.003) or survivors (4.9 (2.8-9.5) vs. 8.9 (5.1-14.3); p = 0.026), respectively. Thus, SAH appears to be a better contributor to the prediction of septic organ dysfunction and death than lactate in critically ill patients. As SAH is a potent inhibitor of SAM-dependent methyltransferases involved in numerous vital biochemical processes, the impairment of the SAM-to-SAH ratio in severely critically ill septic patients and non-survivors warrants further studies on the pathogenetic role of SAH in septic multiple organ failure.


Subject(s)
Critical Illness , S-Adenosylhomocysteine , Humans , Multiple Organ Failure , Prospective Studies , Lactic Acid , Hypoxia , Oxygen , S-Adenosylmethionine , Disease Progression
7.
Int J Mol Sci ; 24(13)2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37446171

ABSTRACT

Maintaining optimal one-carbon metabolism (OCM) is essential for health and pregnancy. In this cross-sectional study, folate status was assessed based on 5-methyltetrahydrofolate (5-MTHF) levels, and the association between 5-MTHF and OCM-related metabolites was investigated in 227 female Japanese university students aged 18-25 years. The participants were divided into high and low 5-MTHF groups based on their folate status. Serum samples of the participants were collected while they were fasting, and 18 OCM-related metabolites were measured using stable-isotope dilution liquid chromatography-electrospray tandem mass spectrometry. The association between serum 5-MTHF and OCM-related metabolite concentrations was assessed using Spearman's rank correlation coefficient. Serum 5-MTHF concentrations were negatively correlated with total homocysteine (tHcy) concentrations and positively correlated with S-adenosylmethionine (SAM) and total cysteine (tCys) concentrations. Serum 5-MTHF concentrations demonstrated a stronger negative correlation with tHcy/tCys than with tHcy alone. The negative correlation between betaine and tHcy concentrations was stronger in the low 5-MTHF group than in the high 5-MTHF group. The 5-MTHF status could be linked to Hcy flux into the transsulfuration pathway via SAM. Therefore, the tHcy/tCys ratio may be a more sensitive indicator of the 5-MTHF status than tHcy alone. Furthermore, a low 5-MTHF status can enhance Hcy metabolism via betaine.


Subject(s)
Betaine , Folic Acid , Pregnancy , Humans , Female , Adolescent , Young Adult , Adult , Cross-Sectional Studies , S-Adenosylmethionine , Carbon , Homocysteine
8.
Int J Mol Sci ; 24(22)2023 Nov 08.
Article in English | MEDLINE | ID: mdl-38003292

ABSTRACT

S-adenosylhomocysteine hydrolase (AHCY) deficiency results mainly in hypermethioninemia, developmental delay, and is potentially fatal. In order to shed new light on molecular aspects of AHCY deficiency, in particular any changes at transcriptome level, we enabled knockdown of AHCY expression in the colon cancer cell line SW480 to simulate the environment occurring in AHCY deficient individuals. The SW480 cell line is well known for elevated AHCY expression, and thereby represents a suitable model system, in particular as AHCY expression is regulated by MYC, which, on the other hand, is involved in Wnt signaling and the regulation of Wnt-related genes, such as the ß-catenin co-transcription factor LEF1 (lymphoid enhancer-binding factor 1). We selected LEF1 as a potential target to investigate its association with S-adenosylhomocysteine hydrolase deficiency. This decision was prompted by our analysis of RNA-Seq data, which revealed significant changes in the expression of genes related to the Wnt signaling pathway and genes involved in processes responsible for epithelial-mesenchymal transition (EMT) and cell proliferation. Notably, LEF1 emerged as a common factor in these processes, showing increased expression both on mRNA and protein levels. Additionally, we show alterations in interconnected signaling pathways linked to LEF1, causing gene expression changes with broad effects on cell cycle regulation, tumor microenvironment, and implications to cell invasion and metastasis. In summary, we provide a new link between AHCY deficiency and LEF1 serving as a mediator of changes to the Wnt signaling pathway, thereby indicating potential connections of AHCY expression and cancer cell phenotype, as Wnt signaling is frequently associated with cancer development, including colorectal cancer (CRC).


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Humans , Adenosylhomocysteinase/genetics , Adenosylhomocysteinase/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/genetics , Colorectal Neoplasms/pathology , Down-Regulation , Gene Expression Regulation, Neoplastic , Tumor Microenvironment , Wnt Signaling Pathway/genetics
9.
BMC Cardiovasc Disord ; 22(1): 101, 2022 03 13.
Article in English | MEDLINE | ID: mdl-35282820

ABSTRACT

BACKGROUND: To investigate the expression of serum S-adenosylhomocysteine (SAH), interleukin-1ß (IL-1ß), serum homocysteine (Hcy), tumor necrosis factor-α (TNF-α) and brain derived neurotrophic factor (BDNF) in coronary heart disease and their relationship with the degree of coronary artery disease. METHODS: A total of 132 patients with coronary heart disease (CHD) from March 2020 to April 2021 were included in this retrospective study. The experimental group was composed of CHD patients, including single-vascular group (46 cases), dual-vascular group (49 cases), and multi-vascular group (37 cases). 145 healthy subjects during the same period for physical examination constituted the control group. RESULTS: The levels of SAH, IL-1ß, Hcy, TNF-α and BDNF in single-vascular group, dual-vascular group and multi-vascular group were higher than that in control group, and the differences were statistically significant (P < 0.05). The serum levels of SAH, IL-1ß, Hcy, TNF-α and BDNF in multi-vascular group were higher than those in single-vascular group and dual-vascular group, and the serum levels of SAH, IL-1ß, Hcy, TNF-α and BDNF in dual-vascular group were higher than those in single-vascular group, with statistical significance (P < 0.05). Kendall's tau-b correlation showed that the levels of SAH, IL-1ß, Hcy, TNF-α and BDNF were positively correlated with the number of stenosis vessels (r = 0.421, 0.533, 0.301, 0.265, 0.678, P = 0.016, 0.009, 0.023, 0.036, 0.004). CONCLUSION: SAH, IL-1ß, Hcy, TNF-α and BDNF in serum of patients with CHD can be used as effective biological indicators to monitor the degree of CHD and severity of coronary stenosis.


Subject(s)
Coronary Artery Disease , Coronary Stenosis , Brain-Derived Neurotrophic Factor , Coronary Stenosis/diagnostic imaging , Homocysteine , Humans , Interleukin-1beta , Retrospective Studies , Tumor Necrosis Factor-alpha
10.
Int J Mol Sci ; 23(8)2022 Apr 10.
Article in English | MEDLINE | ID: mdl-35456998

ABSTRACT

Dysregulation of one-carbon metabolism affects a wide range of biological processes and is associated with a number of diseases, including cardiovascular disease, dementia, neural tube defects, and cancer. Accumulating evidence suggests that one-carbon metabolism plays an important role in COVID-19. The symptoms of long COVID-19 are similar to those presented by subjects suffering from vitamin B12 deficiency (pernicious anemia). The metabolism of a cell infected by the SARS-CoV-2 virus is reshaped to fulfill the need for massive viral RNA synthesis, which requires de novo purine biosynthesis involving folate and one-carbon metabolism. Many aspects of host sulfur amino acid metabolism, particularly glutathione metabolism underlying antioxidant defenses, are also taken over by the SARS-CoV-2 virus. The purpose of this review is to summarize recent findings related to one-carbon metabolism and sulfur metabolites in COVID-19 and discuss how they inform strategies to combat the disease.


Subject(s)
COVID-19 , COVID-19/complications , Carbon/metabolism , Folic Acid/metabolism , Homocysteine , Humans , Methionine/metabolism , SARS-CoV-2 , Vitamin B 12/metabolism , Post-Acute COVID-19 Syndrome
11.
J Clin Biochem Nutr ; 71(3): 238-244, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36447492

ABSTRACT

The relationship between folic acid and S-adenosylhomocysteine (SAH) is controversial. This study aims to explore the effect of different doses of folic acid supplementation on SAH levels in hypertensive patients and the modification of methylene-tetrahydrofolate reductase (MTHFR) C677T gene polymorphism. A randomized, double-blind, controlled clinical trial was conducted. Hypertensive patients aged 45-75 years without a history of stroke and cardiovascular disease were selected, who were randomly assigned to one of 8 dose groups. This trial has been registered with Trial Number: ChiCTR1800016135. In the total population, folic acid supplementation of 0.4-2.0 mg/day had no effect on SAH level (ß = 0.47, 95% CI: -0.86-1.79, p = 0.491), while folic acid supplementation of 2.4 mg/day significantly increased SAH level (ß = 1.93, 95% CI: 0.22-3.64, p = 0.027). Stratified analysis found that MTHFR C677T genotype CC supplemented with 2.4 mg/day folic acid had no effect on SAH level (ß = 0.30, 95% CI: -2.74-3.34, p = 0.847), while CT and TT genotype supplemented with 2.4 mg/day folic acid showed a significant increase in SAH level (CT: ß = 2.98, 95% CI: 0.34-5.62, p = 0.027; TT: ß = 3.00, 95% CI: -0.51-6.51, p = 0.095; CT combined with TT: ß = 2.99, 95% CI: 0.90-5.09, p = 0.005). In conclusion, supplementation of 2.4 mg/day folic acid can lead to increased SAH levels, especially in MTHFR C677T genotype CT and TT.

12.
Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi ; 40(11): 801-806, 2022 Nov 20.
Article in Zh | MEDLINE | ID: mdl-36510712

ABSTRACT

Objective: To investigate the relationship of polycyclic aromatic hydrocarbons (PAHs) exposure, S-adenosylhomocysteine hydrolase (SAHH) activity and long noncoding RNA H19 gene expression in the urine of coke oven workers. Methods: In September 2019, in a coking plant in Taiyuan City, 146 male workers who had worked in coke oven operations for one year were selected through a completely random sampling method, and their basic personal information was collected by questionnaire survey, and blood and urine samples were collected. The levels of 4 PAHs metabolites 2-hydroxfluorene (2-FLU), 2- hydroxynaphthalene (2-NAP), 9-hydroxyphenanthren (9-PHE), and 1-hydroxypyrene (1-OHP) in urine were detected by high performance liquid chromatography (HPLC) -fluorescence detection method. HPLC-UV detection method was used to detect the content of S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) in plasma, and the SAHH activity value was obtained by calculating the ratio. Reverse transcription PCR method was used to determine the H19 gene expression level. Urine levels of 2-FLU, 2-NAP, 9-PHE, and 1-OHP were divided into Q(1), Q(2), Q(3), and Q(4) groups according to quartiles (P(25), P(50), P(75)). Regression, trend test and restricted cubic splines were used to analyze the relationship among PAHs metabolites, SAHH activity, H19 gene expression and their dose-response. Results: The median age of coke oven workers was 39.60 years old, the median length of service was 20.38 years, and the urinary levels of 2-FLU, 2-NAP, 9- PHE, and 1-OHP were 0.29, 0.74, 0.09, and 0.06 µg/mmol Cr, respectively. The levels of 2-FLU, 2-NAP and 9-PHE in the urine of workers were significantly different between groups with different 1-OHP levels (P<0.05). After adjusting for age, length of service, smoking, drinking, and levels of 2-FLU, 2-NAP and 9-PHE, SAHH activity decreased with the increase of urinary 1-OHP level (OR=0.63, 95%CI: 0.41-0.98, P=0.038), showing a nonlinear relationship (P(nonlinear)= 0.030). H19 gene expression increased with the increase of urinary 1- OHP level (OR=1.51, 95%CI: 1.03-2.19, P=0.033), there was a linear relationship (P(trend)= 0.058). The relationship between the other three metabolites in urine and SAHH activity and H19 gene expression was not statistically significant (P>0.05) . Conclusion: Urinary 1-OHP level may be a risk factor for decreased SAHH activity and increased H19 gene expression in coke oven workers.


Subject(s)
Coke , Occupational Exposure , Polycyclic Aromatic Hydrocarbons , Humans , Adult , Coke/analysis , Polycyclic Aromatic Hydrocarbons/analysis , Occupational Exposure/analysis , Pyrenes/analysis , Smoking/urine
13.
Circulation ; 139(19): 2260-2277, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30773021

ABSTRACT

BACKGROUND: Elevated levels of S-adenosylhomocysteine (SAH), the precursor of homocysteine, are positively associated with the risk of cardiovascular disease and with the development and progression of atherosclerosis. However, the role of SAH in endothelial dysfunction is unclear. METHODS: Apolipoprotein E-deficient ( apoE-/-) mice received dietary supplementation with the SAH hydrolase (SAHH) inhibitor adenosine dialdehyde or were intravenously injected with a retrovirus expressing SAHH shRNA. These 2 approaches, along with the heterozygous SAHH gene knockout ( SAHH+/-) mouse model, were used to elevate plasma SAH levels and to examine the role of SAH in aortic endothelial dysfunction. The relationship between plasma SAH levels and endothelial dysfunction was also investigated in human patients with coronary artery disease and healthy control subjects. RESULTS: Plasma SAH levels were increased in SAHH+/- mice and in apoE-/- mice after dietary administration of adenosine dialdehyde or intravenous injection with SAHH shRNA. SAHH+/- mice or apoE-/- mice with SAHH inhibition showed impaired endothelium-dependent vascular relaxation and decreased nitric oxide bioavailability after treatment with acetylcholine; this was completely abolished by the administration of the endothelial nitric oxide synthase inhibitor NG-nitro-l-arginine methyl ester. Furthermore, SAHH inhibition induced production of reactive oxygen species and p66shc expression in the mouse aorta and human aortic endothelial cells. Antioxidants and p66shc siRNA prevented SAHH inhibition-induced generation of reactive oxygen species and attenuated the impaired endothelial vasomotor responses in high-SAH mice. Moreover, inhibition of SAHH induced hypomethylation in the p66shc gene promoter and inhibited expression of DNA methyltransferase 1. Overexpression of DNA methyltransferase 1, induced by transduction of an adenovirus, was sufficient to abrogate SAHH inhibition-induced upregulation of p66shc expression. Finally, plasma SAH levels were inversely associated with flow-mediated dilation and hypomethylation of the p66shc gene promoter and positively associated with oxidative stress levels in patients with coronary artery disease and healthy control subjects. CONCLUSIONS: Our findings indicate that inhibition of SAHH results in elevated plasma SAH levels and induces endothelial dysfunction via epigenetic upregulation of the p66shc-mediated oxidative stress pathway. Our study provides novel molecular insight into mechanisms of SAH-associated endothelial injury that may contribute to the development of atherosclerosis. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov . Unique identifier: NCT03345927.


Subject(s)
Adenosylhomocysteinase/metabolism , Atherosclerosis/metabolism , Coronary Artery Disease/metabolism , Endothelium, Vascular/physiology , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosylhomocysteinase/antagonists & inhibitors , Adenosylhomocysteinase/genetics , Aged , Animals , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Middle Aged , Oxidative Stress , RNA, Small Interfering/genetics , S-Adenosylhomocysteine/blood , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
14.
Chembiochem ; 21(9): 1364-1371, 2020 05 04.
Article in English | MEDLINE | ID: mdl-31802575

ABSTRACT

Psychotropic Psilocybe mushrooms biosynthesize their principal natural product psilocybin in five steps, among them a phosphotransfer and two methyltransfer reactions, which consume one equivalent of 5'-adenosine triphosphate (ATP) and two equivalents of S-adenosyl-l-methionine (SAM). This short but co-substrate-intensive pathway requires nucleoside cofactor salvage to maintain high psilocybin production rates. We characterized the adenosine kinase (AdoK) and S-adenosyl-l-homocysteine (SAH) hydrolase (SahH) of Psilocybe cubensis. Both enzymes are directly or indirectly involved in regenerating SAM. qRT-PCR expression analysis revealed an induced expression of the genes in the fungal primordia and carpophores. A one-pot in vitro reaction with the N-methyltransferase PsiM of the psilocybin pathway demonstrates a concerted action with SahH to facilitate biosynthesis by removal of accumulating SAH.


Subject(s)
Adenosine Kinase/metabolism , Adenosine/metabolism , Adenosylhomocysteinase/metabolism , Psilocybe/enzymology , Psilocybin/biosynthesis , S-Adenosylmethionine/metabolism , Adenosine Kinase/genetics , Adenosylhomocysteinase/genetics , Gene Expression Profiling , Psilocybe/genetics
15.
Electrophoresis ; 41(3-4): 209-214, 2020 02.
Article in English | MEDLINE | ID: mdl-31779046

ABSTRACT

A new approach for direct determination of S-adenosylmethionine (SAM), S-adenosylhomocysteine (SAH), and methylthioadenosine (MTA) in urine was developed based on MEKC by using SDS modified with isobutanol in the presence of PEG-300. Analytes were first extracted with grafted phenylborononic acid. Using a 50 µm internal diameter silica capillary of 32 cm total length filled with 0.05 M SDS, 0.05 M H3 PO4 , 5% (v/v) isobutanol, and 10% (v/v) PEG-300, LOQ of 0.15 µM for SAM and SAH, and 0.2 µM for MTA was reached. Accuracy was 92% for MTA, 109% for SAH, and 105% for SAM, intra- and interday imprecision were <2.5 and ≤3%, respectively. The total time of analysis for one sample was 10 min. Analysis of 30 urine samples from healthy volunteers showed that the median SAM and SAH levels were 12.1 and 0.73 µM, respectively. MTA levels, which were determined in urine for the first time (according to our data), were 0.43 µM, and these values correlated well with the SAM level (r = 0.748, p < 0.01).


Subject(s)
Adenosine/analogs & derivatives , Chromatography, Micellar Electrokinetic Capillary/methods , S-Adenosylhomocysteine/urine , S-Adenosylmethionine/urine , Thionucleosides/urine , Adenosine/urine , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Limit of Detection , Linear Models , Male , Middle Aged , Reproducibility of Results , Young Adult
16.
J Nutr ; 150(Suppl 1): 2532S-2537S, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000156

ABSTRACT

Homocysteine (Hcy) is methylated by methionine synthase to form methionine with methyl-cobalamin as a cofactor. The reaction demethylates 5-methyltetrahydrofolate to tetrahydrofolate, which is required for DNA and RNA synthesis. Deficiency of either of the cobalamin (Cbl) and/or folate cofactors results in elevated Hcy and megaloblastic anemia. Elevated Hcy is a sensitive biomarker of Cbl and/or folate status and more specific than serum vitamin assays. Elevated Hcy normalizes when the correct vitamin is given. Elevated Hcy is associated with alcohol use disorder and drugs that target folate or Cbl metabolism, and is a risk factor for thrombotic vascular disease. Elevated methionine and cystathionine are associated with liver disease. Elevated Hcy, cystathionine, and cysteine, but not methionine, are common in patients with chronic renal failure. Higher cysteine predicts obesity and future weight gain. Serum S-adenosylhomocysteine (AdoHcy) is elevated in Cbl deficiency and chronic renal failure. Drugs that require methylation for catabolism may deplete liver S-adenosylmethionine and raise AdoHcy and Hcy. Deficiency of Cbl or folate or perturbations of their metabolism cause major changes in sulfur amino acids.


Subject(s)
Amino Acids, Sulfur/metabolism , Folic Acid Deficiency/complications , Folic Acid/blood , Hyperhomocysteinemia/blood , Nutritional Status , Vitamin B 12 Deficiency/complications , Vitamin B 12/blood , Alcoholism/blood , Amino Acids, Sulfur/blood , Anemia, Megaloblastic/blood , Biomarkers/blood , Cardiovascular Diseases/blood , Folic Acid Deficiency/blood , Humans , Hyperhomocysteinemia/complications , Kidney Failure, Chronic/blood , Liver Diseases/blood , Obesity/blood , S-Adenosylhomocysteine/blood , Vitamin B 12 Deficiency/blood
17.
J Nutr ; 150(Suppl 1): 2506S-2517S, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000152

ABSTRACT

The metabolism of sulfur-containing amino acids (SAAs) requires an orchestrated interplay among several dozen enzymes and transporters, and an adequate dietary intake of methionine (Met), cysteine (Cys), and B vitamins. Known human genetic disorders are due to defects in Met demethylation, homocysteine (Hcy) remethylation, or cobalamin and folate metabolism, in Hcy transsulfuration, and Cys and hydrogen sulfide (H2S) catabolism. These disorders may manifest between the newborn period and late adulthood by a combination of neuropsychiatric abnormalities, thromboembolism, megaloblastic anemia, hepatopathy, myopathy, and bone and connective tissue abnormalities. Biochemical features include metabolite deficiencies (e.g. Met, S-adenosylmethionine (AdoMet), intermediates in 1-carbon metabolism, Cys, or glutathione) and/or their accumulation (e.g. S-adenosylhomocysteine, Hcy, H2S, or sulfite). Treatment should be started as early as possible and may include a low-protein/low-Met diet with Cys-enriched amino acid supplements, pharmacological doses of B vitamins, betaine to stimulate Hcy remethylation, the provision of N-acetylcysteine or AdoMet, or experimental approaches such as liver transplantation or enzyme replacement therapy. In several disorders, patients are exposed to long-term markedly elevated Met concentrations. Although these conditions may inform on Met toxicity, interpretation is difficult due to the presence of additional metabolic changes. Two disorders seem to exhibit Met-associated toxicity in the brain. An increased risk of demyelination in patients with Met adenosyltransferase I/III (MATI/III) deficiency due to biallelic mutations in the MATIA gene has been attributed to very high blood Met concentrations (typically >800 µmol/L) and possibly also to decreased liver AdoMet synthesis. An excessively high Met concentration in some patients with cystathionine ß-synthase deficiency has been associated with encephalopathy and brain edema, and direct toxicity of Met has been postulated. In summary, studies in patients with various disorders of SAA metabolism showed complex metabolic changes with distant cellular consequences, most of which are not attributable to direct Met toxicity.


Subject(s)
Amino Acids, Sulfur/metabolism , Cysteine/metabolism , Homocysteine/metabolism , Metabolic Diseases/genetics , Methionine/metabolism , Sulfur Compounds/metabolism , Sulfur/metabolism , Animals , Brain Diseases/etiology , Brain Diseases/metabolism , Glutathione/metabolism , Homocystinuria/etiology , Homocystinuria/metabolism , Humans , Hydrogen Sulfide/metabolism , Liver/metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Metabolic Diseases/therapy , Metabolism, Inborn Errors/pathology , Metabolism, Inborn Errors/therapy , Methionine Adenosyltransferase/metabolism , Methylation , S-Adenosylmethionine/metabolism , Sulfites/metabolism
18.
Am J Med Genet A ; 182(4): 740-745, 2020 04.
Article in English | MEDLINE | ID: mdl-31957987

ABSTRACT

S-adenosylhomocysteine hydrolase deficiency is an autosomal recessive neurometabolic disorder affecting the muscles, liver, and nervous system. The disease occurs by pathogenic variants of AHCY gene encoding S-adenosylhomocysteine hydrolase (AHCY) enzyme. This article reports a patient with presumed AHCY deficiency who was diagnosed by whole exome sequencing due to compound heterozygosity of novel p.T57I (c.170C>T) and p.V217M (c.649G>A) variants of AHCY gene. The patient had diffuse edema, coagulopathy, central nervous system abnormalities, and hypotonia. She died in 3 months due to cardiovascular collapse. Clinical findings of the present case were compatible with previously reported AHCY deficiency patients and the novel variants we found are considered to be the cause of the symptoms. This article also compiles the previous reports and expands clinical spectrum of AHCY deficiency by adding new features.


Subject(s)
Adenosylhomocysteinase/genetics , Amino Acid Metabolism, Inborn Errors/diagnosis , Glycine N-Methyltransferase/deficiency , Mutation , Amino Acid Metabolism, Inborn Errors/genetics , Female , Glycine N-Methyltransferase/genetics , Humans , Infant, Newborn , Prognosis
19.
Nutr Metab Cardiovasc Dis ; 30(12): 2286-2295, 2020 11 27.
Article in English | MEDLINE | ID: mdl-32912785

ABSTRACT

BACKGROUND & AIMS: Patients with cystathionine ß-synthase deficiency (CBSD) exhibit high circulating levels of homocysteine and enhanced lipid peroxidation. We have characterized the plasma lipidome in CBSD patients and related lipid abnormalities with reactions underlying enhanced homocysteine levels. METHODS AND RESULTS: Using an ultra-high-performance liquid chromatography-electrospray ionization-quadrupole-time of flight-mass spectrometry method, plasma lipids were determined with an untargeted lipidomics approach in 11 CBSD patients and 11 matched healthy subjects (CTRL). Compared to CTRL, CBSD patients had a higher medium and long-chain polyunsaturated fatty acids (PUFA) content in phosphatidylethanolamine (PE) and lysophosphatidylethanolamine (LPE) species (p < 0.02), and depletion of phosphatidylcholine (PC; p = 0.02) and of lysophosphatidylcholine (LPC; p = 0.003) species containing docosahexaenoic acid (DHA), suggesting impaired phosphatidylethanolamine-N-methyltransferase (PEMT) activity. PEMT converts PE into PC using methyl group by S-adenosylmethionine (SAM) thus converted in S-adenosylhomocysteine (SAH). Whole blood SAM and SAH concentrations by liquid chromatography tandem mass spectrometry were 1.4-fold (p = 0.015) and 5.3-fold (p = 0.003) higher in CBSD patients than in CTRL. A positive correlation between SAM/SAH and PC/PE ratios (r = 0.520; p = 0.019) was found. CONCLUSIONS: A novel biochemical abnormality in CBSD patients consisting in depletion of PC and LPC species containing DHA and accumulation of PUFA in PE and LPE species is revealed by this lipidomic approach. Changes in plasma SAM and SAH concentrations are associated with such phospholipid dysregulation. Given the key role of DHA in thrombosis prevention, depletion of PC species containing DHA in CBSD patients provides a new direction to understand the poor cardiovascular outcome of patients with homocystinuria.


Subject(s)
Dyslipidemias/blood , Homocystinuria/complications , Phospholipids/blood , Adult , Aged , Biomarkers/blood , Case-Control Studies , Chromatography, High Pressure Liquid , Dyslipidemias/diagnosis , Dyslipidemias/etiology , Female , Homocystinuria/blood , Homocystinuria/diagnosis , Humans , Lipidomics , Male , Middle Aged , Spectrometry, Mass, Electrospray Ionization
20.
Int J Mol Sci ; 20(4)2019 Feb 17.
Article in English | MEDLINE | ID: mdl-30781581

ABSTRACT

Homocysteine (Hcy) is a sulfur-containing non-proteinogenic amino acid formed during the metabolism of the essential amino acid methionine. Hcy is considered a risk factor for atherosclerosis and cardiovascular disease (CVD), but the molecular basis of these associations remains elusive. The impairment of endothelial function, a key initial event in the setting of atherosclerosis and CVD, is recurrently observed in hyperhomocysteinemia (HHcy). Various observations may explain the vascular toxicity associated with HHcy. For instance, Hcy interferes with the production of nitric oxide (NO), a gaseous master regulator of endothelial homeostasis. Moreover, Hcy deregulates the signaling pathways associated with another essential endothelial gasotransmitter: hydrogen sulfide. Hcy also mediates the loss of critical endothelial antioxidant systems and increases the intracellular concentration of reactive oxygen species (ROS) yielding oxidative stress. ROS disturb lipoprotein metabolism, contributing to the growth of atherosclerotic vascular lesions. Moreover, excess Hcy maybe be indirectly incorporated into proteins, a process referred to as protein N-homocysteinylation, inducing vascular damage. Lastly, cellular hypomethylation caused by build-up of S-adenosylhomocysteine (AdoHcy) also contributes to the molecular basis of Hcy-induced vascular toxicity, a mechanism that has merited our attention in particular. AdoHcy is the metabolic precursor of Hcy, which accumulates in the setting of HHcy and is a negative regulator of most cell methyltransferases. In this review, we examine the biosynthesis and catabolism of Hcy and critically revise recent findings linking disruption of this metabolism and endothelial dysfunction, emphasizing the impact of HHcy on endothelial cell methylation status.


Subject(s)
Atherosclerosis/metabolism , Cardiovascular Diseases/metabolism , Homocysteine/metabolism , Hyperhomocysteinemia/metabolism , Atherosclerosis/pathology , Cardiovascular Diseases/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Homocysteine/toxicity , Humans , Hydrogen Sulfide/metabolism , Hyperhomocysteinemia/pathology , Methionine/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , S-Adenosylhomocysteine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL