Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 704
Filter
1.
Eur J Nutr ; 63(5): 1513-1528, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38478042

ABSTRACT

BACKGROUND: While folic acid (FA) is widely used to treat elevated total homocysteine (tHcy), promoting vascular health by reducing vascular oxidative stress and modulating endothelial nitric oxide synthase, the optimal daily dose and individual variation by MTHFR C677T genotypes have not been well studied. Therefore, this study aimed to explore the efficacy of eight different FA dosages on tHcy lowering in the overall sample and by MTHFR C677T genotypes. METHODS: This multicentered, randomized, double-blind, controlled clinical trial included 2697 eligible hypertensive adults with elevated tHcy (≥ 10 mmol/L) and without history of stroke and cardiovascular disease. Participants were randomized into eight dose groups of FA combined with 10 mg enalapril maleate, taken daily for 8 weeks of treatment. RESULTS: The intent to treat analysis included 2163 participants. In the overall sample, increasing FA dosage led to steady tHcy reduction within the FA dosing range of 0-1.2 mg. However, a plateau in tHcy lowering was observed in FA dose range of 1.2-1.6 mg, indicating a ceiling effect. In contrast, FA doses were positively and linearly associated with serum folate levels without signs of plateau. Among MTHFR genotype subgroups, participants with the TT genotype showed greater efficacy of FA in tHcy lowering. CONCLUSIONS: This randomized trial lent further support to the efficacy of FA in lowering tHcy; more importantly, it provided critically needed evidence to inform optimal FA dosage. We found that the efficacy of FA in lowering tHcy reaches a plateau if the daily dosage exceeds 1.2 mg, and only has a small gain by increasing the dosage from 0.8 to 1.2 mg. GOV IDENTIFIER: NCT03472508 (Registration Date: March 21, 2018).


Subject(s)
Folic Acid , Genotype , Homocysteine , Methylenetetrahydrofolate Reductase (NADPH2) , Humans , Folic Acid/administration & dosage , Folic Acid/blood , Homocysteine/blood , Female , Male , Double-Blind Method , Middle Aged , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Hypertension/drug therapy , Dose-Response Relationship, Drug , Aged , Enalapril/administration & dosage , Enalapril/pharmacology , Adult , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/blood
2.
BMC Cardiovasc Disord ; 24(1): 431, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39148029

ABSTRACT

BACKGROUND: Hyperhomocysteinemia (HHcy) is an independent risk factor for atherosclerosis (AS). Endothelial mesenchymal transition (EndMT) refers to the process in which endothelial cells lose endothelial cell morphology and characteristic gene expression, and acquire phenotypic characteristics and gene expression related to mesenchymal cells. Numerous studies have confirmed that EndMT is involved in the formation of atherosclerosis. Catalpol is one of the active components of Rehmannia, which has antioxidant, anti-inflammatory, anti-tumor, neuroprotective and other biological activities. Studies have shown that catalpol can reduce atherosclerotic plaque induced by high sugar or fat. However, the effect of catalpol on HHCY-induced EndMT is unclear. METHODS AND RESULTS: In vitro HHcy-treated primary human umbilical vein endothelial cells (HUVECs) were used to construct a cell model, and the antioxidants N-acetylcysteine (NAC) and catalase alcohol were administered. In vivo C57BL/6N mice were given a diet fed with 4.4% high methionine chow to construct a HHcy mice model and were treated with catalpol. The results showed that hhcy could induce morphological transformation of endothelial cells into mesenchymal cells, increase intracellular ROS content, up-regulate α-SMA, N-cadherin, p-p65 protein expression, down-regulate VE-cadherin, CD31 protein expression, induce pathological changes of aortic root endothelium, and increase aortic endothelial ROS content. Catalpol reversed these hhcy induced outcomes. CONCLUSIONS: Catalpol inhibits HHcy-induced EndMT, and the underlying mechanism may be related to the ROS/NF-κB signaling pathway. Catalpol may be a potential drug for the treatment of HHcy-related AS.


Subject(s)
Atherosclerosis , Endothelial-Mesenchymal Transition , Hyperhomocysteinemia , Iridoid Glucosides , NF-kappa B , Reactive Oxygen Species , Animals , Humans , Antigens, CD/metabolism , Antioxidants/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/pathology , Cadherins/metabolism , Cells, Cultured , Disease Models, Animal , Endothelial-Mesenchymal Transition/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/complications , Iridoid Glucosides/pharmacology , Iridoid Glucosides/therapeutic use , Mice, Inbred C57BL , NF-kappa B/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Mice
3.
J Integr Neurosci ; 23(1): 3, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38287840

ABSTRACT

BACKGROUND: The impact of the methylenetetrahydrofolate reductase (MTHFR) C677T mutation on the relationship between plasma homocysteine (Hcy) levels and stroke has been extensively studied and documented in previous study. However, it remains unclear whether the MTHFR C677T mutation can affect the response to Hcy lowering treatment in stroke patients with hyperhomocysteinemia (HHcy). Understanding the impact of genetic factors on treatment response can help optimize personalized treatment strategies for stroke patients with HHcy. We aimed to investigate the potential association between the MTHFR C677T gene polymorphisms and the effectiveness of Hcy lowering treatment using vitamin therapy in stroke patients with HHcy. METHODS: The MTHFR C677T genotype polymorphisms were identified using polymerase chain reaction-restriction fragment length polymorphism, and the distribution of three genotypes in the MTHFR C677T gene locus was compared. The treatment effects of Hcy lowering agents were compared among patients with different genotypes. RESULTS: Among the 320 stroke patients enrolled in the study, 258 (80.6%) were diagnosed with HHcy. Of these, 162 patients (Effective Group) responded well to the clinical Hcy lowering treatment, while 96 patients (Invalid Group) failed to achieve sufficient response even after taking combination supplements of folic acid, Vitamin B6, and methylcobalamin for one month. Significant differences were observed in terms of age (p < 0.001), hypertension (p = 0.034), dyslipidemia (p = 0.022), hyperuricemia (p = 0.013) and genotype distribution of MTHFR C677T gene polymorphism (p < 0.001) between the Invalid group and the Effective group. The multivariate regression analysis revealed that the T allele (odd rations [OR], 1.327; 95% confidence interval [CI], 1.114-1.580; p = 0.0015) was independently associated with an insufficient Hcy lowering treatment effect. Additionally, the TT genotype was independently associated with insufficient response in both the codominant model (OR, 1.645; 95% CI, 1.093-2.476; p = 0.017) and the recessive model (TT versus CC + CT; OR, 1.529; 95% CI, 1.145-2.042; p = 0.004). However, no relationship was observed between CT + TT genotypes and poor treatment effect in the dominate model. CONCLUSIONS: Our findings suggested that the TT genotype and T allele of MTHFR C677T polymorphism were independently associated with an insufficient Hcy lowering treatment effect in stroke patients with HHcy.


Subject(s)
Hyperhomocysteinemia , Stroke , Humans , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/genetics , Polymorphism, Genetic , Stroke/complications , Stroke/drug therapy , Stroke/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Homocysteine/genetics , Vitamins
4.
J Neuroinflammation ; 20(1): 199, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37658433

ABSTRACT

BACKGROUND: Epidemiological data suggests statins could reduce the risk of dementia, and more specifically, Alzheimer's disease (AD). Pre-clinical data suggests statins reduce the risk of dementia through their pleiotropic effects rather than their cholesterol lowering effects. While AD is a leading cause of dementia, it is frequently found co-morbidly with cerebral small vessel disease and other vascular contributions to cognitive impairment and dementia (VCID), which are another leading cause of dementia. In this study, we determined if atorvastatin ameliorated hyperhomocysteinemia (HHcy)-induced VCID. METHODS: Wild-type (C57Bl6/J) mice were placed on a diet to induce HHcy or a control diet each with or without atorvastatin for 14 weeks. Mice underwent novel object recognition testing before tissue collection. Plasma total cholesterol and total homocysteine as well as related metabolites were measured. Using qPCR and NanoString technology, we profiled glial cell-associated gene expression changes. Finally, microglial morphology, astrocyte end feet, and microhemorrhages were analyzed using histological methods. RESULTS: Atorvastatin treatment of HHcy in mice led to no changes in total cholesterol but decreases in total homocysteine in plasma. While HHcy decreased expression of many glial genes, atorvastatin rescued these gene changes, which mostly occurred in oligodendrocytes and microglia. Microglia in HHcy mice with atorvastatin were trending towards fewer processes compared to control with atorvastatin, but there were no atorvastatin effects on astrocyte end feet. While atorvastatin treatment was trending towards increasing the area of microhemorrhages in HHcy mice in the frontal cortex, it only slightly (non-significantly) reduced the number of microhemorrhages. Finally, atorvastatin treatment in HHcy mice led to improved cognition on the novel object recognition task. CONCLUSIONS: These data suggest that atorvastatin rescued cognitive changes induced by HHcy most likely through lowering plasma total homocysteine and rescuing gene expression changes rather than impacts on vascular integrity or microglial changes.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Dementia, Vascular , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hyperhomocysteinemia , Animals , Mice , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognition , Homocysteine/toxicity
5.
Clin Exp Hypertens ; 45(1): 2205058, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37154141

ABSTRACT

OBJECTIVES: To investigate the actions of amlodipine-folic acid (amlodipine-FA) preparation on hypertension and cardiovascular in renal hypertensive rats with hyperhomocysteinemia (HHcy), so as to provide experimental basis for clinical research of amlodipine folic acid tablets. METHODS: Rats model of renal hypertension with HHcy were established. The rats were randomly divided into groups of model, amlodipine, folic acid (FA) and amlodipine-FA of various dosages. Normal rats were used as normal control group. Blood pressure, Hcy as well as plasma NO, ET-1 and hemodynamics were assayed. Histological alterations of heart and abdominal aorta were also examined. RESULTS: Compared with the normal group, blood pressure, plasma Hcy, and NO of the rats in model group were significantly increased, while the plasma ET-1 was decreased. Compared with the normal group, the animals in the model group had reduced cardiac function, thickened wall of the aorta and narrowed lumen. In FA group and amlodipine group, the rat plasma NO was increased while ET-1 was decreased, the protective effect of amlodipine-FA group on endothelial cells was further enhanced. In amlodipine group, the rat hemodynamics (LVSP, LVEDP and ±dp/dtmax, et al.) and vascular damage were significantly reduced, while in amlodipine-FA group, the heart function were further improved, and myocardial and vascular hypertrophy were significantly reduced. CONCLUSIONS: As compared to amlodipine alone, amlodipine -FA can lower both blood pressure and plasma Hcy, significantly enhancing vascular endothelial function to protect the heart and blood vessel in renal hypertensive rats with HHcy.


Subject(s)
Hyperhomocysteinemia , Hypertension , Rats , Animals , Folic Acid/pharmacology , Amlodipine/pharmacology , Endothelial Cells , Hypertension/complications , Hypertension/drug therapy , Kidney , Homocysteine , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy
6.
Crit Rev Food Sci Nutr ; 62(20): 5462-5475, 2022.
Article in English | MEDLINE | ID: mdl-33724098

ABSTRACT

Elevated plasma levels of homocysteine (Hcy) are a recognized risk factor for stroke. This relationship represents one aspect of the debated `Hcy hypothesis'. Elevated Hcy may be an independent and treatable cause of atherosclerosis and thrombotic vascular diseases. Further observations indicate that proper dietary supplementation with B-vitamins decreases total plasma Hcy concentrations and may be an effective intervention for stroke prevention. Metabolic vitamin B12 deficiency is a nutritional determinant of total Hcy and stroke risk. Genetic factors may link B vitamins with stroke severity due to the impact on Hcy metabolism of polymorphism in the genes coding for methylenetetrahydrofolate reductase, methionine-synthase, methionine synthase reductase, and cystathionine ß-synthase. Several meta-analyses of large randomized controlled trials exist. However, they are not completely in agreement about B vitamins' role, particularly folic acid levels, vitamin B12, and B6, in lowering the homocysteine concentrations in people at high stroke risk. A very complex relationship exists between Hcy and B vitamins, and several factors appear to modify the preventive effects of B vitamins in stroke. This review highlights the regulating factors of the active role of B vitamins active in stroke prevention. Also, inputs for further large, well-designed studies, for specific, particularly sensitive subgroups are given.


Subject(s)
Hyperhomocysteinemia , Stroke , Vitamin B Complex , Folic Acid , Homocysteine , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy , Stroke/prevention & control , Vitamin B 12 , Vitamin B Complex/pharmacology , Vitamin B Complex/therapeutic use
7.
Vascular ; 30(5): 988-998, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34362270

ABSTRACT

BACKGROUND: Hypertension associated with hyperhomocysteinemia (HHcy) is correlated with a high risk of vascular diseases. Studies found that folic acid (FA) supplementation can reduce the risk of cardiovascular and cerebrovascular events. The aim of the present study was to explore the potential mechanisms of FA attenuating HHcy-related arterial injury in spontaneously hypertensive rats (SHRs). METHODS: 24 SHRs were randomized into the control group, the HHcy group, and the HHcy + FA group (8 per group). The SHRs in the HHcy group and the HHcy + FA group were given DL-Hcy intraperitoneally to mimic hypertension associated with HHcy. The SHRs in the HHcy + FA group were given FA by gavage to mimic an FA-fortified diet. The histopathology and immunohistochemistry of rat aorta and carotid artery were analyzed, and the relative expression levels of immune/inflammation and oxidative stress molecules in arterial tissue were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. RESULTS: FA significantly reduced the expression levels of nuclear factor-κ-gene binding (NF-κB) p65/Rela and interleukin-6 (IL-6) in rat arterial tissues, as well as the levels of plasma HHcy and serum malondialdehyde (MDA) in hypertension associated with HHcy rats (p < 0.05). At the same time, FA significantly increased the serum superoxide dismutase (SOD) level in hypertension associated with HHcy rats, and even the SOD level of the HHcy + FA group was higher than that of the control group (p < 0.05). However, HHcy induced the opposite results of the above indicators in SHRs compared with the control group (p < 0.05). CONCLUSIONS: The arterial protection mechanisms of FA are related to reducing the concentration of HHcy to eliminate the tissue toxicity of HHcy, inhibiting NF-κBp65/Rela/IL-6 pathway molecules to regulate inflammatory response, and promoting the potential anti-oxidative stress pathway molecules to reduce oxidative stress level.


Subject(s)
Arteritis , Hyperhomocysteinemia , Hypertension , Animals , Arteritis/complications , Folic Acid/pharmacology , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy , Hypertension/complications , Hypertension/drug therapy , Interleukin-6 , Malondialdehyde/metabolism , NF-kappa B , Rats , Rats, Inbred SHR , Superoxide Dismutase/metabolism
8.
Int J Neurosci ; 132(4): 384-396, 2022 Apr.
Article in English | MEDLINE | ID: mdl-32859137

ABSTRACT

AIM: The present study investigates the potential of Tadalafil, a phosphodiesterase-5 inhibitor, in a rat model of hyperhomocysteinemia induced vascular dementia. METHODS: Hyperhomocysteinemia induced vascular dementia in Wistar rats was produced by administering l-Methionine (1.7 g/kg/day; p.o.×8 weeks). Learning and memory was assessed by employing Morris water maze (MWM) test. Endothelial dysfunction was assessed through acetylcholine-induced endothelial-dependent vasorelaxation and serum nitrite levels. Various other biochemical and histopathological estimations were also performed. RESULTS: l-Methionine produced significant impairment in acetylcholine-induced endothelium-dependent vasorelaxation and a decrease in serum nitrite levels indicating endothelial dysfunction. Further, these animals performed poorly on Morris water maze, depicting impairment of learning and memory. There was a significant rise in brain oxidative stress level (indicated by an increase in brain thiobarbituric acid reactive species and a decrease in reduced glutathione levels). Increase in brain acetylcholinesterase activity; brain myeloperoxidase activity and brain neutrophil infiltration (a marker of inflammation) were also observed. Tadalafil (5 and 10 mg/kg, p.o.)/Donepezil (0.5 mg/kg, i.p., serving as standard) treatment ameliorated l-Methionine induced endothelial dysfunction; memory deficits; biochemical and histopathological changes in a significant manner. CONCLUSIONS: It may be concluded that tadalafil has shown efficacy in the rat model of l-Methionine induced vascular dementia and that phosphodiesterase-5 can be considered as an important therapeutic target for the treatment of vascular dementia.


Subject(s)
Dementia, Vascular , Hyperhomocysteinemia , Tadalafil , Acetylcholine , Acetylcholinesterase/metabolism , Animals , Brain/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5 , Dementia, Vascular/drug therapy , Dementia, Vascular/etiology , Hyperhomocysteinemia/chemically induced , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy , Maze Learning , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Methionine , Nitrites/blood , Oxidative Stress , Phosphodiesterase 5 Inhibitors , Rats , Rats, Wistar , Tadalafil/therapeutic use
9.
Int J Neurosci ; 132(11): 1061-1071, 2022 Nov.
Article in English | MEDLINE | ID: mdl-33287606

ABSTRACT

Backgroud: Hyperhomocysteinemia (HHcy) is implicated in various neurovascular disorders including vascular dementia, subarachnoid hemorrhage and stroke. Elevated homocysteine (Hcy) levels are associated with increased oxidative stress and compromised blood-brain barrier (BBB) integrity. Hydrogen sulfide (H2S) has recently emerged as potent neuroprotective molecule in various neurological conditions including those associated with HHcy. The present study evaluates the protective effect of sodium hydrogen sulfide (NaHS; a source of H2S) on HHcy-induced BBB dysfunction and underpin molecular mechanisms.Materials and methods: Supplementation of NaHS restored the increased BBB permeability in the cortex and hippocampus of HHcy animals assessed in terms of diffused sodium fluorescein and Evans blue tracer dyes in the brain. Activity of matrix metalloproteinases (MMPs) assessed by gelatinase activity and in situ gelatinase assay was restored to the normal in the cortex and hippocampus of HHcy animals supplemented with NaHS.Results: Application of gelatin zymography revealed that specifically MMP-9 activity was increased in the cortex and hippocampus of HHcy animals, which was inhibited by NaHS supplementation. Real-time RT-PCR analysis showed that NaHS administration also decreased mRNA expression of MMP-9 in the hippocampus of HHcy animals. NaHS supplementation was further observed to reduce water retention in the brain regions of Hcy treated animals.Conclusion: Taken together, these findings suggest that NaHS supplementation ameliorates HHcy-induced BBB permeability and brain edema by inhibiting the mRNA expression and activity of MMP-9. Therefore, H2S and H2S releasing drugs may be used as a novel therapeutic approach to treat HHcy-associated neurovascular disorders.


Subject(s)
Hydrogen Sulfide , Hyperhomocysteinemia , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/metabolism , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy , Blood-Brain Barrier , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Matrix Metalloproteinase 9/therapeutic use , Evans Blue/metabolism , Evans Blue/pharmacology , Evans Blue/therapeutic use , Fluorescein/metabolism , Fluorescein/pharmacology , Fluorescein/therapeutic use , Gelatin/metabolism , Gelatin/pharmacology , Gelatin/therapeutic use , Permeability , RNA, Messenger/metabolism , Sodium , Coloring Agents/metabolism , Coloring Agents/pharmacology , Coloring Agents/therapeutic use , Homocysteine , Water/metabolism , Water/pharmacology
10.
Metabolomics ; 17(5): 47, 2021 05 09.
Article in English | MEDLINE | ID: mdl-33966131

ABSTRACT

INTRODUCTION: Hyperhomocysteinemia (HHCys) is an independent risk factor for various diseases such as cardiovascular diseases, Alzheimer's, and cancers. Folate deficiency is one of the significant reasons for HHCys. However, it is not known whether folate deficiency with HHCys is associated with any serum metabolites. OBJECTIVES: Our objective was to identify the metabolic alterations in people having folate deficiency with HHCys and check whether a short-term folic acid therapy could reverse those metabolic changes. METHODS: The study enrolled 34 participants aged between 18 and 40 years having folate deficiency (< 4.6 ng/mL) with HHCys (> 15 µmol/L) and 21 normal healthy individuals. A short-term intervention of oral folic acid (5 mg/day) was done in the HHCys group for 30 days. Untargeted metabolomics analysis of serum was performed in all study subjects before and after the folic acid treatment. Different univariate methods and the multivariable-adjusted linear regression models were employed to determine an association between homocysteine level and metabolite profile. RESULTS: Metabolomics analysis data showed that many metabolites involved in the biochemical pathways of lipid metabolisms such as polyunsaturated fatty acids, glycerolipids, and phospholipids were downregulated in the HHCys group. Short-term oral folic acid therapy significantly reduced their serum homocysteine level. However, the metabolic pathway alterations observed in folate-deficient HHCys-condition were unaltered even after the folic acid treatment. CONCLUSIONS: Our study revealed that people who have a folic acid deficiency with HHCys have an altered metabolite profile related to lipid metabolism, which cannot be reversed by short-term folic acid therapy.


Subject(s)
Hyperhomocysteinemia , Adolescent , Adult , Folic Acid , Folic Acid Deficiency/drug therapy , Homocysteine , Humans , Hyperhomocysteinemia/drug therapy , Metabolome , Vitamin B 12 , Young Adult
11.
Sheng Li Xue Bao ; 73(4): 551-558, 2021 Aug 25.
Article in Zh | MEDLINE | ID: mdl-34405211

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) and hyperhomocysteinemia (HHcy) both are major health problems worldwide, whose incidence are closely related with each other. We previously reported the mechanism of HHcy-caused hepatic steatosis, but the role of n-3 polyunsaturated fatty acid (n-3 PUFA) in HHcy-induced hepatic steatosis remains unclear. In this study, 6-week-old C57BL/6 male mice were given a high methionine diet (HMD, 2% methionine diet), and plasma homocysteine levels were measured by ELISA to confirm the establishment of an HHcy model. Meantime, mice were fed HMD with or without n-3 PUFA supplement for 8 weeks to determine the role and mechanism of n-3 PUFA in hepatic steatosis induced by HHcy. Results showed that n-3 PUFA significantly improved hepatic lipid deposition induced by HHcy. qRT-PCR analysis demonstrated that n-3 PUFA inhibited the upregulation of Cd36, a key enzyme of fatty acid uptake, caused by HHcy. Further, the inhibition of hepatic Cd36 expression was associated with the inactivation of aryl hydrocarbon receptor (Ahr) induced by n-3 PUFA. Of note, mass spectrometry revealed that hepatic content of lipoxin A5 (LXA5) was significantly increased in HMD+n-3 PUFA-fed mice compared with that in HMD-fed mice. In primary cultured hepatocytes, LXA5 treatment markedly reversed homocysteine-evoked Cd36 upregulation and Ahr activation, which resulted in reduced lipid accumulation. In conclusion, we demonstrate that n-3 PUFA inactivates HHcy-induced Ahr-Cd36 pathway by increasing hepatic LXA5 content, which alleviates hepatic steatosis. Thus, our results may provide a potential strategy for treatment of NAFLD.


Subject(s)
Fatty Acids, Omega-3 , Fatty Liver , Hyperhomocysteinemia , Animals , Fatty Liver/drug therapy , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy , Liver , Male , Mice , Mice, Inbred C57BL
12.
J Gene Med ; 22(4): e3156, 2020 04.
Article in English | MEDLINE | ID: mdl-31864233

ABSTRACT

BACKGROUND: A decrease in cystathionine beta-synthase (CBS) enzyme activity could lead to hyperhomocysteinemia (HHcy). Studies have revealed that DNA methylation has a mediating effect on the development of diseases. The present study aimed to explore CBS promoter methylation-mediating effects on the efficacy of folate treatment for HHcy. METHODS: HHcy patients were treated with folate (5 mg/day) for 90 days and then divided into a failure group (Hcy ≥ 15 µmol/l) and a success group (Hcy < 15 µmol/l) according to post-treatment plasma Hcy levels. Genotyping of CBS gene (rs2851391 and rs706209) in patients (n = 638) was detected using a MassArray system (Sequenom, San Diego, CA, USA). The baseline DNA methylation levels of patients (n = 299) were detected using MethylTarget™ technology (Genesky Biotechnologies Inc., Shanghai, China). RESULTS: The CBS rs2851391 TC + CC genotype was related to a 57% reduction of failure risk in HHcy treatment compared to the TT genotype (95% confidence interval [CI] = 0.19-0.97). The CBS rs706209 CT + TT genotype had a 2.97-fold increased risk of failure to treatment compared to the CC genotype (95% CI = 1.52-5.80). After adjustment for confounding factors, the odds ratio (95% CI) for the risk of failure in HHcy treatment in total and male patients was 0.55 (0.32-0.93) and 0.34 (0.16-0.69), respectively, for patients with higher methylation levels (≥ methylation median). Additionally, baseline CBS promoter methylation mediated 33.39% of the effect of rs2851391 on the efficacy of folate treatment for HHcy (ACME [average causal mediation effects]: -0.05, 95% CI = -0.11 to 0.00, p = 0.046). CONCLUSIONS: The present study indicates that CBS gene polymorphism and promoter methylation could affect the efficacy of HHcy. There were potentially causal effects of genetic, epigenetic variations at the CBS rs2851391 locus on the efficacy of HHcy therapy with folate.


Subject(s)
Cystathionine beta-Synthase/genetics , DNA Methylation , Folic Acid/therapeutic use , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/genetics , Polymorphism, Genetic , Promoter Regions, Genetic , Adult , Aged , Alleles , Biomarkers , Female , Folic Acid/administration & dosage , Gene Frequency , Genotype , Humans , Male , Middle Aged , Odds Ratio , Pharmacogenetics/methods , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Treatment Outcome
13.
J Neurol Neurosurg Psychiatry ; 91(11): 1201-1209, 2020 11.
Article in English | MEDLINE | ID: mdl-32690803

ABSTRACT

BACKGROUND: Evidence on preventing Alzheimer's disease (AD) is challenging to interpret due to varying study designs with heterogeneous endpoints and credibility. We completed a systematic review and meta-analysis of current evidence with prospective designs to propose evidence-based suggestions on AD prevention. METHODS: Electronic databases and relevant websites were searched from inception to 1 March 2019. Both observational prospective studies (OPSs) and randomised controlled trials (RCTs) were included. The multivariable-adjusted effect estimates were pooled by random-effects models, with credibility assessment according to its risk of bias, inconsistency and imprecision. Levels of evidence and classes of suggestions were summarised. RESULTS: A total of 44 676 reports were identified, and 243 OPSs and 153 RCTs were eligible for analysis after exclusion based on pre-decided criteria, from which 104 modifiable factors and 11 interventions were included in the meta-analyses. Twenty-one suggestions are proposed based on the consolidated evidence, with Class I suggestions targeting 19 factors: 10 with Level A strong evidence (education, cognitive activity, high body mass index in latelife, hyperhomocysteinaemia, depression, stress, diabetes, head trauma, hypertension in midlife and orthostatic hypotension) and 9 with Level B weaker evidence (obesity in midlife, weight loss in late life, physical exercise, smoking, sleep, cerebrovascular disease, frailty, atrial fibrillation and vitamin C). In contrast, two interventions are not recommended: oestrogen replacement therapy (Level A2) and acetylcholinesterase inhibitors (Level B). INTERPRETATION: Evidence-based suggestions are proposed, offering clinicians and stakeholders current guidance for the prevention of AD.


Subject(s)
Alzheimer Disease/prevention & control , Evidence-Based Medicine , Antihypertensive Agents/therapeutic use , Cognition , Craniocerebral Trauma/prevention & control , Depression/therapy , Diabetes Mellitus/therapy , Education , Exercise , Humans , Hyperhomocysteinemia/drug therapy , Hypertension/drug therapy , Hypotension, Orthostatic/therapy , Life Style , Obesity/therapy , Observational Studies as Topic , Randomized Controlled Trials as Topic , Risk Reduction Behavior , Stress, Psychological/therapy
14.
Acta Pharmacol Sin ; 41(1): 47-55, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31607752

ABSTRACT

T cell metabolic activation plays a crucial role in inflammation of atherosclerosis. Shikonin (SKN), a natural naphthoquinone with anti-inflammatory activity, has shown to exert cardioprotective effects, but the effect of SKN on atherosclerosis is unclear. In addition, SKN was found to inhibit glycolysis via targeting pyruvate kinase muscle isozyme 2 (PKM2). In the present study, we investigated the effects of SKN on hyperhomocysteinemia (HHcy)-accelerated atherosclerosis and T cell inflammatory activation in ApoE-/- mice and the metabolic mechanisms in this process. Drinking water supplemented with Hcy (1.8 g/L) was administered to ApoE-/- mice for 2 weeks and the mice were injected with SKN (1.2 mg/kg, i.p.) or vehicle every 3 days. We showed that SKN treatment markedly attenuated HHcy-accelerated atherosclerosis in ApoE-/- mice and significantly decreased inflammatory activated CD4+ T cells and proinflammatory macrophages in plaques. In splenic CD4+ T cells isolated from HHcy-ApoE-/- mice, SKN treatment significantly inhibited HHcy-stimulated PKM2 activity, interferon-γ secretion and the capacity of these T cells to promote macrophage proinflammatory polarization. SKN treatment significantly inhibited HHcy-stimulated CD4+ T cell glycolysis and oxidative phosphorylation. Metabolic profiling analysis of CD4+ T cells revealed that Hcy administration significantly increased various glucose metabolites as well as lipids and acetyl-CoA carboxylase 1, which were reversed by SKN treatment. In conclusion, our results suggest that SKN is effective to ameliorate atherosclerosis in HHcy-ApoE-/- mice and this is at least partly associated with the inhibition of SKN on CD4+ T cell inflammatory activation via PKM2-dependent metabolic suppression.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Hyperhomocysteinemia/drug therapy , Inflammation/drug therapy , Naphthoquinones/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Dose-Response Relationship, Drug , Female , Hyperhomocysteinemia/metabolism , Inflammation/metabolism , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthoquinones/administration & dosage
15.
Nutr Metab Cardiovasc Dis ; 30(8): 1281-1288, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32522470

ABSTRACT

BACKGROUND AND AIMS: In Portugal, The Azores Archipelago has the highest standardized mortality rate for CAD. Therefore, the aim of this study was to evaluate conventional risk factors, as well as plasma and erythrocyte aminothiol concentration in high-risk Azorean patients undergoing elective coronary angiography and to investigate whether any aminothiol was associated with CAD risk and severity. METHODS AND RESULTS: 174 subjects with symptomatic CAD (age 56±9y; 68% men) submitted to coronary angiography were split into 2 groups: one formed by CAD patients (≥50% stenosis in at least one major coronary vessel) and the other by non-CAD patients (<50% stenosis). Both groups were age-, sex- and BMI-matched. Plasma and erythrocyte aminothiol profiles were evaluated by RP-HPLC/FLD. CAD patients significantly exhibited both higher concentrations of plasma Cys and hypercysteinemia (Cys ≥ 300 µM) prevalence than those in the non-CAD group (261 ± 58 µM vs. 243 ± 56 µM; 22% vs. 10%, respectively). No differences were observed between groups regarding plasma Hcy levels or hyperhomocysteinemia prevalence. After adjustment for several confounders (including Hcy), subjects in the highest quartile of plasma Cys had a 3.31 (95% CI, 1.32-8.30, p = 0.011) fold risk for CAD, compared with those in the lowest quartiles. Furthermore, plasma Cys levels (but not Hcy) tended to increase with the number of stenotic vessels (1VD: 253 ± 64 µM; 2VD: 262 ± 52 µM; 3VD: 279 ± 57 µM, p = 0.129). CONCLUSION: Hypercysteinemia revealed to be a better predictor of CAD than hyperhomocysteinemia. Moreover, plasma Cys showed to be a useful biomarker for CAD both in primary and secondary preventions, seeming to resist better than Hcy to oral medication therapy.


Subject(s)
Coronary Artery Disease/blood , Coronary Stenosis/blood , Cysteine/blood , Homocysteine/blood , Hyperhomocysteinemia/blood , Adult , Aged , Biomarkers/blood , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/epidemiology , Coronary Artery Disease/prevention & control , Coronary Stenosis/diagnostic imaging , Coronary Stenosis/epidemiology , Female , Humans , Hyperhomocysteinemia/diagnosis , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/epidemiology , Male , Middle Aged , Portugal/epidemiology , Predictive Value of Tests , Prevalence , Primary Prevention , Prognosis , Retrospective Studies , Risk Assessment , Risk Factors , Secondary Prevention , Severity of Illness Index
16.
J Stroke Cerebrovasc Dis ; 29(5): 104633, 2020 May.
Article in English | MEDLINE | ID: mdl-32122776

ABSTRACT

CASE: Two years ago, annual magnetic resonance imaging for unruptured right internal carotid artery aneurysm of a 47-year-old woman detected a cerebral infarct in her right occipital lobe which was unknown etiology and antiplatelet therapy was initiated. She presented with sensory disorder of her left fingers 4 months ago. Infarction in right parieto-occipital cortex and severe stenosis of right middle cerebral artery was revealed. Her laboratory test was normal except remarkably high homocysteine value. Regardless of dual anti-platelet therapy, she suffered from repeated minor stroke and the stenosis was progressing. Therefore, right superficial temporal artery - middle cerebral artery bypass was undertaken. Aspirin and clopidogrel were withdrawn 1 week before the surgery. Two branches were anastomosed with 2 separate frontal M4 branches. Although patency was confirmed immediately after the anastomosis, thrombus formation was revealed after 10 minutes. We needed to perform removal of the thrombus and re-anastomosis twice. Intraoperative administration of aspirin and ozagrel alleviated thrombotic tendency. After surgery, antiplatelet therapy and supplementation with folate and vitamin B were performed. Her postoperative course was uneventful and patency of both anastomoses was confirmed. DISCUSSION: Controversy still exists regarding preoperative antiplatelet therapy before superficial temporal artery-middle cerebral artery bypass, and folates and B6-12 vitamins supplementation for hyperhomocysteinemia. Considering intraoperative thrombo tendency in our case, it is recommended to evaluate the homocysteine level before bypass surgery for intracranial stenosis especially for young patients or patients with unknown etiology. Before bypass surgery of the patient with hyperhomocysteinemia, continuation of perioperative antiplatelet drugs and supplementation with folates and B6-12 vitamins are mandatory.


Subject(s)
Hyperhomocysteinemia/complications , Infarction, Middle Cerebral Artery/surgery , Middle Cerebral Artery/surgery , Temporal Arteries/surgery , Vascular Grafting/adverse effects , Venous Thrombosis/etiology , Dietary Supplements , Female , Fibrinolytic Agents/administration & dosage , Humans , Hyperhomocysteinemia/diagnosis , Hyperhomocysteinemia/drug therapy , Infarction, Middle Cerebral Artery/diagnostic imaging , Infarction, Middle Cerebral Artery/etiology , Middle Aged , Middle Cerebral Artery/diagnostic imaging , Recurrence , Risk Factors , Severity of Illness Index , Temporal Arteries/diagnostic imaging , Thrombectomy , Treatment Outcome , Venous Thrombosis/diagnostic imaging , Venous Thrombosis/therapy , Vitamin B Complex/administration & dosage
17.
Georgian Med News ; (309): 22-28, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33526724

ABSTRACT

Treatment of hereditary hyperhomocysteinemia and the achievement of optimal folate status is necessary for persons of reproductive age in order to increase live birth rate. Patients are usually advised to take folic acid, a key nutrient in homocysteine remethylation. The results of study showed risk factors for hyperhomocysteinemia development in investigated married couples: male gender, MTHFR, MTR1 genes variants, lower vitamin B12 blood serum and no additional intake of vitamin B12. Since MTHFR, MTR1 genes variants affect to decrease the efficiency of homocysteine metabolic transformations, to contribute also to endothelial dysfunction in one of patients group we used betargine combined with folic acids and vitamin B12 administration. Patients group with combined administration including betargine within 2 weeks, in comparison with the group without its supplement, had significantly decreased level of homocysteine in plasma, less than 12 µmol/l (81.03% and 50% of cases, respectively). Folic acid and vitamin B12 mean values in blood serum was significantly increased in patients after two week vitamins administration including betargin. Further research is needed to establish the duration of betaine-arginine intake until the target homocysteine level will be reached, as well as to estimate the durability of clinical effect achieved after consumption.


Subject(s)
Betaine , Hyperhomocysteinemia , Arginine , Betaine/therapeutic use , Folic Acid , Homocysteine , Humans , Hyperhomocysteinemia/drug therapy , Male , Vitamin B 12
18.
J Cell Mol Med ; 23(1): 464-475, 2019 01.
Article in English | MEDLINE | ID: mdl-30394648

ABSTRACT

Picroside II (P-II), one of the main active components of scrophularia extract, which have anti-oxidative, anti-inflammatory effects, but its effect on hyperhomocysteinemia (HHcy) induced endothelial injury remains to be determined. Here, we test whether P-II protects HHcy-induced endothelial dysfunction against oxidative stress, inflammation and cell apoptosis. In vitro study using HUVECs, and in hyperhomocysteinemia mouse models, we found that HHcy decreased endothelial SIRT1 expression and increased LOX-1 expression, subsequently causing reactive oxygen species generation, up-regulation of NADPH oxidase activity and NF-κB activation, thereby promoting pro-inflammatory response and cell apoptosis. Blockade of Sirt1 with Ex527 or siRNASIRT1 increased LOX-1 expression, whereas overexpression of SIRT1 decreased LOX-1 expression markedly. P-II treatment significantly increased SIRT1 expression and reduced LOX-1 expression, and protected against endothelial cells from Hcy-induced oxidative injury, inflammation and apoptosis. However, blockade of SIRT1 or overexpression of LOX-1 attenuated the therapeutic effects of P-II. In conclusion, our results suggest that P-II prevents the Hcy induced endothelial damage probably through regulating the SIRT1/LOX-1 signaling pathway.


Subject(s)
Apoptosis/drug effects , Cinnamates/pharmacology , Endothelium/drug effects , Hyperhomocysteinemia/drug therapy , Inflammation/drug therapy , Iridoid Glucosides/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Cell Line , Endothelium/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hyperhomocysteinemia/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sirtuin 1/metabolism , Up-Regulation/drug effects
19.
J Cell Physiol ; 234(3): 3007-3019, 2019 03.
Article in English | MEDLINE | ID: mdl-30206943

ABSTRACT

An elevated level of homocysteine (Hcy) leads to hyperhomocysteinemia (HHcy), which results in vascular dysfunction and pathological conditions identical to stroke symptoms. Hcy increases oxidative stress and leads to increase in blood-brain barrier permeability and leakage. Hydrogen sulfide (H2 S) production during the metabolism of Hcy has a cerebroprotective effect, although its effectiveness in Hcy-induced neurodegeneration and neurovascular permeability is less explored. Therefore, the current study was designed to perceive the neuroprotective effect of exogenous H 2 S against HHcy, a cause of neurodegeneration. To test this hypothesis, we used four groups of mice: control, Hcy, control + sodium hydrosulfide hydrate (NaHS), and Hcy + NaHS, and an HHcy mice model in Swiss albino mice by giving a dose of 1.8 g of dl-Hcy/L in drinking for 8-10 weeks. Mice that have 30 µmol/L Hcy were taken for the study, and a H 2 S supplementation of 20 µmol/L was given for 8 weeks to all groups of mice. HHcy results in the rise of the levels of superoxide and nitrite, although a concomitant decrease in the level of superoxide dismutase, catalase, glutathione peroxidase, reduced glutathione, and arginase in oxidative stress and a concomitant decrease in the endogenous level of H 2 S. Although H 2 S supplementation ameliorated, the effect of HHcy and the levels of H 2 S returned to the average level in HHcy animals supplemented with H 2 S. Interestingly, H 2 S supplementation ameliorated neurovascular remodeling and neurodegeneration. Thus, our study suggested that H 2 S could be a beneficial therapeutic candidate for the treatment of Hcy-associated neurodegeneration, such as stroke and neurovascular disorders.


Subject(s)
Cerebrovascular Disorders/drug therapy , Hydrogen Sulfide/pharmacology , Hyperhomocysteinemia/drug therapy , Nerve Degeneration/drug therapy , Animals , Blood-Brain Barrier/drug effects , Cerebrovascular Disorders/chemically induced , Cerebrovascular Disorders/metabolism , Cerebrovascular Disorders/pathology , Disease Models, Animal , Homocysteine/toxicity , Humans , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/pathology , Matrix Metalloproteinase 9/genetics , Mice , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Permeability/drug effects , Stroke/drug therapy , Stroke/metabolism , Stroke/pathology
20.
Ann Hum Genet ; 83(6): 434-444, 2019 11.
Article in English | MEDLINE | ID: mdl-31111486

ABSTRACT

Oral folate is currently the most common treatment for hyperhomocysteinemia (HHcy), which seriously threatens human health, but its efficacy is unsatisfactory. Betaine-homocysteine methyltransferase (BHMT) is a key enzyme that regulates Hcy metabolism. We investigated the association between the BHMT rs3733890 and the efficacy of oral folate therapy for HHcy in the Chinese Han population and analysed the effects of gene-environmental interactions on the efficacy. Blood samples were collected from 1071 eligible patients at baseline, and these individuals received subsequent folate treatment for 90 days. A total of 638 patients included in the final analysis were grouped into the treatment success group or the treatment failure group based on posttreatment Hcy levels. Hcy concentrations were measured by fluorescence polarization immunoassay. Time-of-flight mass spectrometry (MassArray system) was used to assess the genotype of BHMT rs3733890. Stratified analyses based on additive models and generalized multifactor dimensionality reduction were used to explore gene-environmental interactions. The genotype distribution presented distinct differences in the two groups. The mutant genotype and allele had significantly increased risk of treatment failure (p < 0.05). Furthermore, synergistic effects of the BHMT rs3733890 polymorphism with environmental risk factors (smoking, drinking, past history) on the efficacy of therapy were also found. However, future, large well-designed studies, as well as mechanistic studies, are still needed to validate our findings.


Subject(s)
Alleles , Betaine-Homocysteine S-Methyltransferase/genetics , Folic Acid/therapeutic use , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/genetics , Polymorphism, Single Nucleotide , Administration, Oral , Aged , Aged, 80 and over , Comorbidity , Environment , Female , Folic Acid/administration & dosage , Humans , Male , Middle Aged , Odds Ratio , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL