1
|
Abdi F, Farhangi MA, Mohammadzadeh M. Habitual dietary methyl donor's intake and metabolic profile in obese individuals: a cross-sectional study. Sci Rep 2024; 14:30046. [PMID: 39627237 PMCID: PMC11615318 DOI: 10.1038/s41598-024-75388-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/04/2024] [Indexed: 12/06/2024] Open
Abstract
Considering the role of dietary methyl donor (DMD) in numerous biochemical processes, we hypothesized that DMD could play an important role in metabolic syndrome such as hyperlipidemia, hypertension, insulin resistance, and appetite in obese individuals. This cross-sectional study was conducted on 335 obese people. We collected dietary data using a valid and reliable 147-question Food Frequency Questionnaire (FFQ). Multivariate multinomial logistic regression was used to estimate the odds ratio (OR) and 95% confidence interval (CI) for the association between dietary methyl intake and cardio-metabolic risk factors. After adjusting for confounding variables, individuals at the fourth and third quartile of DMD, were more likely to have lower low-density lipoprotein cholesterol (LDL-C) (OR = 0.968, CI = 0.943-0.994, P = 0.015 and OR = 0.978, CI = 0.957-0.998, P = 0.03 respectively) versus first quartile. Also, total cholesterol (TC) showed a significant decrease in forth quartile of DMD in model III (OR = 0.974, CI = 0.951-0.997, P = 0.029). Current results suggested that, high DMDs' consumption, significantly associated with decreased risk of cardiometabolic risk factors.
Collapse
Affiliation(s)
- Fatemeh Abdi
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Milad Mohammadzadeh
- Department of Clinical Nutrition and Dietetic, School of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Fan X, Liu Y, Chen X, Xu Y, Wu W, Li F, Liu G, Chen X, Zhang C, Zhou Y. Synergies between diabetes and hyperhomocysteinaemia: New insights to predict and prevent adverse cardiovascular effects. Diabetes Obes Metab 2024; 26:5776-5785. [PMID: 39434446 DOI: 10.1111/dom.15947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
AIM To explore the association of hyperhomocysteinaemia (HHcy) and diabetes synergies with cardiovascular events in the adult population of northern China. METHODS Data were collected from the Asymptomatic Polyvascular Abnomalities Community study for 2010 to 2019. Serum homocysteine (Hcy) levels were determined by enzyme-linked immunosorbent assay. The participants were categorized into four groups based on their Hcy levels and diabetes status: non-diabetes/non-HHcy, non-diabetes/HHcy, diabetes/non-HHcy and diabetes/HHcy. The composite endpoint consisted of the occurrence of first-ever stroke, myocardial infraction (MI) or all-cause mortality. Cox regression analyses were performed to evaluate the associations of diabetes and HHcy with cardiovascular disease (CVD) events. RESULTS In total, 5278 participants were eligible (average age 55.1 years, 60% male). Over a follow-up of 9.1 years, 618 events were identified, 202 stroke, 52 MI and 406 all-cause deaths. Compared with the non-diabetes/non-HHcy group, hazard ratios with 95% confidence intervals in the diabetes/HHcy group for stroke, MI, major adverse cardiovascular event (MACE), all-cause death and composite endpoint were 1.85 (1.12-3.04), 1.33 (0.42-4.23), 1.78 (1.13-2.80), 2.24 (1.56-3.23) and 1.97 (1.47-2.65), respectively. Significant interactions between HHcy and diabetes status were found for stroke, MI and MACE (P for interaction = .002, .027 and .044, respectively). In addition, the association of diabetes/HHcy with stroke was modified by age (< 60 and ≥ 60 years; P for interaction = .016). CONCLUSIONS The findings highlight the synergistic impact of diabetes and HHcy on CVD. Joint assessments of diabetes and Hcy levels should be emphasized for risk stratification and primary prevention of CVD.
Collapse
Affiliation(s)
- Xue Fan
- Department of Research Center for Cardiovascular and Cerebrovascular Disease, Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhe Liu
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, China
| | - Xueyu Chen
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuehao Xu
- Department of Research Center for Cardiovascular and Cerebrovascular Disease, Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqian Wu
- Department of Research Center for Cardiovascular and Cerebrovascular Disease, Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengchang Li
- Department of Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute for Nutrition and Health, Chinese Academy of sciences, Shanghai, China
| | - Gang Liu
- Department of Internal Medicine, Tangshan, China
| | - Xiaoli Chen
- Department of Internal Medicine, Tangshan, China
| | - Caiping Zhang
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, China
| | - Yong Zhou
- Department of Research Center for Cardiovascular and Cerebrovascular Disease, Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Tate BN, Van Guilder GP, Aly M, Spence LA, Diaz-Rubio ME, Le HH, Johnson EL, McFadden JW, Perry CA. Changes in Choline Metabolites and Ceramides in Response to a DASH-Style Diet in Older Adults. Nutrients 2023; 15:3687. [PMID: 37686719 PMCID: PMC10489641 DOI: 10.3390/nu15173687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
This feeding trial evaluated the impact of the Dietary Approaches to Stop Hypertension diet on changes in plasma choline, choline metabolites, and ceramides in obese older adults; 28 adults consumed 3oz (n = 15) or 6oz (n = 13) of beef within a standardized DASH diet for 12 weeks. Plasma choline, betaine, methionine, dimethylglycine (DMG), phosphatidylcholine (PC), lysophosphotidylcholine (LPC), sphingomyelin, trimethylamine-N-oxide (TMAO), L-carnitine, ceramide, and triglycerides were measured in fasted blood samples. Plasma LPC, sphingomyelin, and ceramide species were also quantified. In response to the study diet, with beef intake groups combined, plasma choline decreased by 9.6% (p = 0.012); DMG decreased by 10% (p = 0.042); PC decreased by 51% (p < 0.001); total LPC increased by 281% (p < 0.001); TMAO increased by 26.5% (p < 0.001); total ceramide decreased by 22.1% (p < 0.001); and triglycerides decreased by 18% (p = 0.021). All 20 LPC species measured increased (p < 0.01) with LPC 16:0 having the greatest response. Sphingomyelin 16:0, 18:0, and 18:1 increased (all p < 0.001) by 10.4%, 22.5%, and 24%, respectively. In contrast, we observed that sphingomyelin 24:0 significantly decreased by 10%. Ceramide 22:0 and 24:0 decreased by 27.6% and 10.9% (p < 0.001), respectively, and ceramide 24:1 increased by 36.8% (p = 0.013). Changes in choline and choline metabolites were in association with anthropometric and cardiometabolic outcomes. These findings show the impact of the DASH diet on choline metabolism in older adults and demonstrate the influence of diet to modify circulating LPC, sphingomyelin, and ceramide species.
Collapse
Affiliation(s)
- Brianna N. Tate
- Department of Animal Science, Cornell University, Ithaca, NY 14853, USA; (B.N.T.); (J.W.M.)
| | - Gary P. Van Guilder
- High Altitude Exercise Physiology Department, Western Colorado University, Gunnison, CO 81231, USA;
| | - Marwa Aly
- Department of Applied Health Science, Indiana University School of Public Health, Bloomington, IN 47405, USA; (M.A.); (L.A.S.)
| | - Lisa A. Spence
- Department of Applied Health Science, Indiana University School of Public Health, Bloomington, IN 47405, USA; (M.A.); (L.A.S.)
| | - M. Elena Diaz-Rubio
- Proteomic and Metabolomics Facility, Cornell University, Ithaca, NY 14853, USA;
| | - Henry H. Le
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (H.H.L.); (E.L.J.)
| | - Elizabeth L. Johnson
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (H.H.L.); (E.L.J.)
| | - Joseph W. McFadden
- Department of Animal Science, Cornell University, Ithaca, NY 14853, USA; (B.N.T.); (J.W.M.)
| | - Cydne A. Perry
- Department of Applied Health Science, Indiana University School of Public Health, Bloomington, IN 47405, USA; (M.A.); (L.A.S.)
| |
Collapse
|
4
|
Chan CW, Lin BF. Folate Deficiency Enhanced Inflammation and Exacerbated Renal Fibrosis in High-Fat High-Fructose Diet-Fed Mice. Nutrients 2023; 15:3616. [PMID: 37630806 PMCID: PMC10458828 DOI: 10.3390/nu15163616] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The prevalence of obesity and chronic kidney disease (CKD) is increasing simultaneously and rapidly worldwide. Our previous study showed that folate deficiency increased lipid accumulation and leptin production of adipocytes. Whether folate plays a role in CKD, particularly obesity-related nephropathy remains unclear. To investigate the effects of folate deficiency on CKD in diet-induced obese mice, four groups of male C57BL/6 mice were fed either a normal-fat diet (NF) with folate (NF+f); NF without folate (NF-f); high-fat high-fructose diet (HFF) with folate (HFF+f); or HFF without folate (HFF-f) for 12 months during the study. The results showed that HFF increased not only body weight, fasting blood glucose, total cholesterol (TC), low-density lipoprotein (LDL)-cholesterol, and blood pressure, but also cytokines levels, such as interleukin (IL)-2, interferon (IFN)-γ, IL-17A/F, IL-6, monocyte chemoattractant protein (MCP)-1, and transforming growth factor (TGF)-β1. The indicators of kidney failure including urinary protein, neutrophil gelatinase-associated lipocalin (NGAL), renal type I and IV collagen deposits and leptin content, and serum creatinine were also increased by HFF. Folate-deficient diets further elevated serum TC, LDL-cholesterol, IL-6, tumor necrosis factor (TNF)-α, MCP-1, TGF-β1, and leptin, but decreased IL-10 level, and thus exacerbated renal fibrosis. To investigate the possible mechanisms of folate deficiency on renal injury, phosphorylation of pro-fibrosis signaling molecules, including signal transducer and activator of transcription (STAT)3 and small mothers against decapentaplegic (Smad)2/3, were assayed. Both HFF and folate deficiency significantly increased the phosphorylation of STAT3 and Smad2/3, suggesting synergistic effects of HFF-f on chronic renal inflammation and fibrosis. In conclusion, the results demonstrated that folate deficiency might aggravate inflammatory status and enhance renal fibrosis.
Collapse
Affiliation(s)
| | - Bi-Fong Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan;
| |
Collapse
|
5
|
Wu HHL, McDonnell T, Chinnadurai R. Physiological Associations between Vitamin B Deficiency and Diabetic Kidney Disease. Biomedicines 2023; 11:biomedicines11041153. [PMID: 37189771 DOI: 10.3390/biomedicines11041153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
The number of people living with chronic kidney disease (CKD) is growing as our global population continues to expand. With aging, diabetes, and cardiovascular disease being major harbingers of kidney disease, the number of people diagnosed with diabetic kidney disease (DKD) has grown concurrently. Poor clinical outcomes in DKD could be influenced by an array of factors-inadequate glycemic control, obesity, metabolic acidosis, anemia, cellular senescence, infection and inflammation, cognitive impairment, reduced physical exercise threshold, and, importantly, malnutrition contributing to protein-energy wasting, sarcopenia, and frailty. Amongst the various causes of malnutrition in DKD, the metabolic mechanisms of vitamin B (B1 (Thiamine), B2 (Riboflavin), B3 (Niacin/Nicotinamide), B5 (Pantothenic Acid), B6 (Pyridoxine), B8 (Biotin), B9 (Folate), and B12 (Cobalamin)) deficiency and its clinical impact has garnered greater scientific interest over the past decade. There remains extensive debate on the biochemical intricacies of vitamin B metabolic pathways and how their deficiencies may affect the development of CKD, diabetes, and subsequently DKD, and vice-versa. Our article provides a review of updated evidence on the biochemical and physiological properties of the vitamin B sub-forms in normal states, and how vitamin B deficiency and defects in their metabolic pathways may influence CKD/DKD pathophysiology, and in reverse how CKD/DKD progression may affect vitamin B metabolism. We hope our article increases awareness of vitamin B deficiency in DKD and the complex physiological associations that exist between vitamin B deficiency, diabetes, and CKD. Further research efforts are needed going forward to address the knowledge gaps on this topic.
Collapse
Affiliation(s)
- Henry H L Wu
- Renal Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, The University of Sydney, Sydney, NSW 2065, Australia
| | - Thomas McDonnell
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - Rajkumar Chinnadurai
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
- Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M1 7HR, UK
| |
Collapse
|
6
|
Chakraborty P, Mitra I, Kalapahar S, Sharma S, Chattopadhyay R, Chakravarty B. Significance of Hyperhomocysteinemia in Immediate As Well As Long-Term Health Risk/s in Women with Polycystic Ovary Syndrome: a Probabilistic Model Using Dynamic Bayesian Network Analysis. Reprod Sci 2023; 30:1207-1216. [PMID: 35802293 DOI: 10.1007/s43032-022-01028-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous entity comprising broad spectra of ovarian disorders with trademark features of metabolic syndrome like insulin resistance, obesity, and dyslipidaemia to name a few. Hyperhomocysteinemia, an independent risk factor of metabolic syndrome, has been suggested as a causative factor in spontaneous miscarriage in PCOS. However, it is yet to be resolved whether hyperhomocysteinemia has a contributory role in the pathogenesis or could direct long-term sequences of the syndrome. A total of 2355 women with history of one or more first trimester abortions were screened and 1539 were selected for the study. Selected patients were initially divided by the presence or absence of PCOS, while subsequent stratification was based on hyperhomocysteinemia, insulin resistance, and/or obesity. The miscarriage population/s was mostly represented by hyperhomocysteinemia in both the cohorts (PCOS: 69.08% vs. non-PCOS: 56.68%). ROC-AUC values suggest increased predisposition of hyperhomocysteinemia-mediated miscarriage (hyperhomocysteinemia: 0.778; insulin resistance: 0.601; BMI: 0.548). A probabilistic causal model was designed using dynamic Bayesian network to evaluate the time-series data points before, during, and after pregnancy which revealed a possibility of 32.24% (n = 79) of PCOS cohort developing hypertension, 26.94% (n = 66) of onset of diabetes and 4.49% cardiovascular disease 3 years following pregnancy. We conclude hyperhomocysteinemia may possibly contribute to spontaneous miscarriage and related to metabolic derailments later in life.
Collapse
Affiliation(s)
- Pratip Chakraborty
- Department of Assisted Reproduction, Institute of Reproductive Medicine, HB-36/A/3, Sector-III, Saltlake City, Kolkata-700106, India.
| | - Imon Mitra
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, India
| | - Shovandeb Kalapahar
- Department of Assisted Reproduction, Institute of Reproductive Medicine, HB-36/A/3, Sector-III, Saltlake City, Kolkata-700106, India
| | - Sunita Sharma
- Department of Assisted Reproduction, Institute of Reproductive Medicine, HB-36/A/3, Sector-III, Saltlake City, Kolkata-700106, India
| | - Ratna Chattopadhyay
- Department of Assisted Reproduction, Institute of Reproductive Medicine, HB-36/A/3, Sector-III, Saltlake City, Kolkata-700106, India
| | - Baidyanath Chakravarty
- Department of Assisted Reproduction, Institute of Reproductive Medicine, HB-36/A/3, Sector-III, Saltlake City, Kolkata-700106, India
| |
Collapse
|
7
|
Development of inhibitors targeting glycogen synthase kinase-3β for human diseases: Strategies to improve selectivity. Eur J Med Chem 2022; 236:114301. [PMID: 35390715 DOI: 10.1016/j.ejmech.2022.114301] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023]
Abstract
Glycogen synthase kinase-3β (GSK-3β) is a conserved serine/threonine kinase that participates in the transmission of multiple signaling pathways and plays an important role in the occurrence and development of human diseases, such as metabolic diseases, neurological diseases and cancer, making it to be a potential and promising drug target. To date, copious GSK-3β inhibitors have been synthesized, but only few have entered clinical trials. Most of them exerts poor selectivity, concomitant off-target effects and side effects. This review summarizes the structural characteristics, biological functions and relationship with diseases of GSK-3β, as well as the selectivity profile and therapeutic potential of different categories of GSK-3β inhibitors. Strategies for increasing selectivity and reducing adverse effects are proposed for the future design of GSK-3β inhibitors.
Collapse
|
8
|
Liu C, Liu L, Wang Y, Chen X, Liu J, Peng S, Pi J, Zhang Q, Tomlinson B, Chan P, Zhang L, Fan H, Zheng L, Liu Z, Zhang Y. Hyperhomocysteinemia Increases Risk of Metabolic Syndrome and Cardiovascular Death in an Elderly Chinese Community Population of a 7-Year Follow-Up Study. Front Cardiovasc Med 2022; 8:811670. [PMID: 35224027 PMCID: PMC8870623 DOI: 10.3389/fcvm.2021.811670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022] Open
Abstract
Background Hyperhomocysteinemia (HHcy) and abdominal obesity are risk factors for metabolic syndrome (MetS) and death from cardiovascular disease (CVD). Recent studies have shown a correlation between HHcy and abdominal obesity, suggesting that they may have a combined effect on the risk of MetS and CVD mortality. However, this suspicion remains to be confirmed, particularly in the elderly population. We explored their combined effects on the risk of MetS and CVD mortality among the community population aged 65 and above in China. Methods and Results This prospective study enrolled 3,675 Chinese community residents aged 65 and above in May 2013 with 7-year follow-up of all-cause and CVD mortality. HHcy was defined as the blood homocysteine (Hcy) level >15 μmol/L and abdominal obesity as waist circumference (WC) ≥90 cm for men and ≥80 cm for women (HWC). All participants were grouped into four categories by WC and the blood level of Hcy: NWC (normal WC) /HHcy(–), NWC/HHcy(+), HWC/HHcy(–), and HWC/HHcy(+). The relationship of combined HHcy and abdominal obesity with MetS and metabolic profile was evaluated by logistic regression analysis and the association of combined HHcy and abdominal obesity with CVD and all-cause mortality evaluated by Cox regression analysis. The prevalence of HHcy, abdominal obesity and MetS in elderly Chinese community residents was 40.1, 59.3, and 41.4%, respectively. Using group without HHcy and abdominal obesity [NWC/HHcy(–)] as reference, the participants of other three groups had significantly higher risk of MetS and its component abnormalities, with HWC/HHcy(+) group having the highest risk (OR = 13.52; 95% CI = 8.61–14.55). After a median of 6.94 (±1.48) years follow-up, 454 deaths occurred with 135 CVD deaths. Compared with NWC/HHcy(–) group, the risk of 7-year follow-up CVD mortality (HR = 1.75; 95% CI = 1.02–3.03) and all-cause mortality (HR = 1.23; 95% CI = 1.04–2.18) of HWC/HHcy(+) group increased considerably after adjustment for major MetS and CVD risk factors. Conclusions There is high prevalence of HHcy, abdominal obesity, and MetS in the elderly Chinese community population. HHcy increases risk of MetS, CVD, and all-cause mortality, especially in the populations with abdominal obesity.
Collapse
Affiliation(s)
- Chang Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liping Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yinglu Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoli Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sheng Peng
- Department of Emergency Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingjiang Pi
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qi Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Brain Tomlinson
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Lin Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huimin Fan
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Zheng
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Liang Zheng
| | - Zhongmin Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Zhongmin Liu
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Yuzhen Zhang
| |
Collapse
|
9
|
Human Serum Betaine and Associated Biomarker Concentrations Following a 14 Day Supplemental Betaine Loading Protocol and during a 28 Day Washout Period: A Pilot Investigation. Nutrients 2022; 14:nu14030498. [PMID: 35276860 PMCID: PMC8839982 DOI: 10.3390/nu14030498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/05/2023] Open
Abstract
Several previous investigations have employed betaine supplementation in randomized controlled crossover designs to assess its ostensible ergogenic potential. Nevertheless, prior methodology is predicated on limited pharmacokinetic data and an appropriate betaine-specific washout period is hitherto undescribed. The purpose of the present pilot investigation was therein to determine whether a 28 day washout period was sufficient to return serum betaine concentrations to baseline following a supplementation protocol. Five resistance-trained men (26 ± 6 y) supplemented with 6 g/day betaine anhydrous for 14 days and subsequently visited the lab 10 additional times during a 28 day washout period. Participants underwent venipuncture to assess serum betaine and several other parameters before (PRE) and periodically throughout the washout timeframe (POST0, -4, -7, -10, -13, -16, -19, -22, -25 and -28). All analyses were performed at a significance level of p < 0.05. While analyses failed to detect any differences in any other serum biomarker (p > 0.05), serum betaine was significantly elevated from PRE-to-POST0 (p = 0.047; 2.31 ± 1.05 to 11.1 ± 4.91 µg·mL−1) and was statistically indistinguishable from baseline at POST4 (p = 1.00). Nevertheless, visual data assessment and an inability to assess skeletal muscle concentrations would otherwise suggest that a more conservative 7 day washout period is sufficient to truly return both serum-and-skeletal muscle betaine content to pre-supplementation levels.
Collapse
|
10
|
Zhang X, Qu YY, Liu L, Qiao YN, Geng HR, Lin Y, Xu W, Cao J, Zhao JY. Homocysteine inhibits pro-insulin receptor cleavage and causes insulin resistance via protein cysteine-homocysteinylation. Cell Rep 2021; 37:109821. [PMID: 34644569 DOI: 10.1016/j.celrep.2021.109821] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/06/2021] [Accepted: 09/21/2021] [Indexed: 01/10/2023] Open
Abstract
Elevation in homocysteine (Hcy) level is associated with insulin resistance; however, the causality between them and the underlying mechanism remain elusive. Here, we show that Hcy induces insulin resistance and causes diabetic phenotypes by protein cysteine-homocysteinylation (C-Hcy) of the pro-insulin receptor (pro-IR). Mechanistically, Hcy reacts and modifies cysteine-825 of pro-IR in the endoplasmic reticulum (ER) and abrogates the formation of the original disulfide bond. C-Hcy impairs the interaction between pro-IR and the Furin protease in the Golgi apparatus, thereby hindering the cleavage of pro-IR. In mice, an increase in Hcy level decreases the mature IR level in various tissues, thereby inducing insulin resistance and the type 2 diabetes phenotype. Furthermore, inhibition of C-Hcy in vivo and in vitro by overexpressing protein disulfide isomerase rescues the Hcy-induced phenotypes. In conclusion, C-Hcy in the ER can serve as a potential pharmacological target for developing drugs to prevent insulin resistance and increase insulin sensitivity.
Collapse
Affiliation(s)
- Xuan Zhang
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital of Fudan University, School of Life Sciences, Children's Hospital of Fudan University, Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Yuan-Yuan Qu
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital of Fudan University, School of Life Sciences, Children's Hospital of Fudan University, Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Lian Liu
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital of Fudan University, School of Life Sciences, Children's Hospital of Fudan University, Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Ya-Nan Qiao
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital of Fudan University, School of Life Sciences, Children's Hospital of Fudan University, Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Hao-Ran Geng
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital of Fudan University, School of Life Sciences, Children's Hospital of Fudan University, Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Yan Lin
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital of Fudan University, School of Life Sciences, Children's Hospital of Fudan University, Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Wei Xu
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital of Fudan University, School of Life Sciences, Children's Hospital of Fudan University, Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Jing Cao
- Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jian-Yuan Zhao
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital of Fudan University, School of Life Sciences, Children's Hospital of Fudan University, Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, P.R. China; Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
11
|
He R, Du S, Lei T, Xie X, Wang Y. Glycogen synthase kinase 3β in tumorigenesis and oncotherapy (Review). Oncol Rep 2020; 44:2373-2385. [PMID: 33125126 PMCID: PMC7610307 DOI: 10.3892/or.2020.7817] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/17/2020] [Indexed: 02/05/2023] Open
Abstract
Glycogen synthase kinase 3β (GSK 3β), a multifunctional serine and threonine kinase, plays a critical role in a variety of cellular activities, including signaling transduction, protein and glycogen metabolism, cell proliferation, cell differentiation, and apoptosis. Therefore, aberrant regulation of GSK 3β results in a broad range of human diseases, such as tumors, diabetes, inflammation and neurodegenerative diseases. Accumulating evidence has suggested that GSK 3β is correlated with tumorigenesis and progression. However, GSK 3β is controversial due to its bifacial roles of tumor suppression and activation. In addition, overexpression of GSK 3β is involved in tumor growth, whereas it contributes to the cell sensitivity to chemotherapy. However, the underlying regulatory mechanisms of GSK 3β in tumorigenesis remain obscure and require further in‑depth investigation. In this review, we comprehensively summarize the roles of GSK 3β in tumorigenesis and oncotherapy, and focus on its potentials as an available target in oncotherapy.
Collapse
Affiliation(s)
- Rui He
- Department of Union, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Tiantian Lei
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing 400013, P.R. China
| | - Xiaofang Xie
- Department of Medicine, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Yi Wang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
- Center of Translational Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
12
|
Machek SB, Cardaci TD, Willoughby DS. Blood Flow Restriction Training and Betaine Supplementation as a Novel Combined Modality to Augment Skeletal Muscle Adaptation: A Short Review. Strength Cond J 2020. [DOI: 10.1519/ssc.0000000000000556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
13
|
Dietary betaine intake is associated with skeletal muscle mass change over 3 years in middle-aged adults: the Guangzhou Nutrition and Health Study. Br J Nutr 2020; 125:440-447. [PMID: 32616104 DOI: 10.1017/s0007114520002433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A higher dietary intake or serum concentration of betaine has been associated with greater lean body mass in middle-aged and older adults. However, it remains unknown whether betaine intake is associated with age-related loss of skeletal muscle mass (SMM). We assessed the association between dietary betaine intake and relative changes in SMM after 3 years in middle-aged adults. A total of 1242 participants aged 41-60 years from the Guangzhou Nutrition and Health Study 2011-2013 and 2014-2017 with body composition measurements by dual-energy X-ray absorptiometry were included. A face-to-face questionnaire was used to collect general baseline information. After adjustment for potential confounders, multiple linear regression found that energy-adjusted dietary betaine intake was significantly and positively associated with relative changes (i.e. percentage loss or increase) in SMM of legs, limbs and appendicular skeletal mass index (ASMI) over 3 years of follow-up (β 0·322 (se 0·157), 0·309 (se 0·142) and 0·303 (se 0·145), respectively; P < 0·05). The ANCOVA models revealed that participants in the highest betaine tertile had significantly less loss in SMM of limbs and ASMI and more increase in SMM of legs over 3 years of follow-up, compared with those in the bottom betaine tertile (all Ptrend < 0·05). In conclusion, our findings suggest that elevated higher dietary betaine intake may be associated with less loss of SMM of legs, limbs and ASMI in middle-aged adults.
Collapse
|
14
|
Gao X, Zhang H, Guo XF, Li K, Li S, Li D. Effect of Betaine on Reducing Body Fat-A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2019; 11:nu11102480. [PMID: 31623137 PMCID: PMC6835719 DOI: 10.3390/nu11102480] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/08/2019] [Accepted: 10/12/2019] [Indexed: 12/22/2022] Open
Abstract
Animal studies have shown the beneficial effect of betaine supplementation on reducing body fat, while the data from human studies are controversial and inconsistent. The objective of the present systematic review was to investigate the effects of betaine intervention on treating obesity in humans and quantitatively evaluate the pooled effects based on randomized controlled trials with a meta-analysis. The PubMed and Scopus databases, and the Cochrane Library, were searched up to September 2019. Weighted mean differences were calculated for net changes in obesity-related indices by using a random-effects model. Publication bias was estimated using Begg’s test. Six studies with 195 participants were identified. Betaine supplementation significantly reduced the total body fat mass (−2.53 kg; 95% CI: −3.93, −0.54 kg; I2 = 6.6%, P = 0.36) and body fat percentage (−2.44%; 95% CI: −4.20, −0.68%; I2 = 0.0%, P = 0.44). No changes were observed regarding body weight (−0.29 kg; 95% CI: −1.48, 0.89 kg; I2 = 0.00%, P = 0.99) and body mass index (−0.10 kg/m2; 95% CI: −5.13, 0.31 kg/m2; I2 = 0.00%, P = 0.84). The results suggested that dietary betaine supplementation might be an effective approach for reducing body fat.
Collapse
Affiliation(s)
- Xiang Gao
- Institute of Nutrition and Health, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| | - Huijun Zhang
- Institute of Nutrition and Health, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| | - Xiao-Fei Guo
- Institute of Nutrition and Health, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| | - Kelei Li
- Institute of Nutrition and Health, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| | - Shan Li
- Institute of Nutrition and Health, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| | - Duo Li
- Institute of Nutrition and Health, College of Life Sciences, Qingdao University, Qingdao 266071, China.
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
15
|
Esse R, Barroso M, Tavares de Almeida I, Castro R. The Contribution of Homocysteine Metabolism Disruption to Endothelial Dysfunction: State-of-the-Art. Int J Mol Sci 2019; 20:E867. [PMID: 30781581 PMCID: PMC6412520 DOI: 10.3390/ijms20040867] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/05/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Ruben Esse
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Madalena Barroso
- University Children's Research@Kinder-UKE, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Isabel Tavares de Almeida
- Laboratory of Metabolism and Genetics, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
| | - Rita Castro
- Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
16
|
Jakubowski H. Homocysteine Modification in Protein Structure/Function and Human Disease. Physiol Rev 2019; 99:555-604. [PMID: 30427275 DOI: 10.1152/physrev.00003.2018] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epidemiological studies established that elevated homocysteine, an important intermediate in folate, vitamin B12, and one carbon metabolism, is associated with poor health, including heart and brain diseases. Earlier studies show that patients with severe hyperhomocysteinemia, first identified in the 1960s, exhibit neurological and cardiovascular abnormalities and premature death due to vascular complications. Although homocysteine is considered to be a nonprotein amino acid, studies over the past 2 decades have led to discoveries of protein-related homocysteine metabolism and mechanisms by which homocysteine can become a component of proteins. Homocysteine-containing proteins lose their biological function and acquire cytotoxic, proinflammatory, proatherothrombotic, and proneuropathic properties, which can account for the various disease phenotypes associated with hyperhomocysteinemia. This review describes mechanisms by which hyperhomocysteinemia affects cellular proteostasis, provides a comprehensive account of the biological chemistry of homocysteine-containing proteins, and discusses pathophysiological consequences and clinical implications of their formation.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers-New Jersey Medical School, International Center for Public Health , Newark, New Jersey ; and Department of Biochemistry and Biotechnology, Poznań University of Life Sciences , Poznań , Poland
| |
Collapse
|
17
|
Lind MV, Lauritzen L, Kristensen M, Ross AB, Eriksen JN. Effect of folate supplementation on insulin sensitivity and type 2 diabetes: a meta-analysis of randomized controlled trials. Am J Clin Nutr 2019; 109:29-42. [PMID: 30615110 DOI: 10.1093/ajcn/nqy234] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 08/06/2018] [Indexed: 11/14/2022] Open
Abstract
Background Various mechanisms link higher total homocysteine to higher insulin resistance (IR) and risk of type 2 diabetes (T2D). Folate supplementation is recognized as a way to lower homocysteine. However, randomized controlled trials (RCTs) show inconsistent results on IR and T2D outcomes. Objective The aim of this study was to examine the effect of folate supplementation on IR and T2D outcomes. Design We conducted a systematic literature search in PubMed, Web of Science, and EMBASE and prior systematic reviews and meta-analyses and identified 29 RCTs (22,250 participants) that assessed the effect of placebo-controlled folate supplementation alone or in combination with other B vitamins on fasting glucose, insulin, homeostasis model assessment for insulin resistance (HOMA-IR), glycated hemoglobin (HbA1c), or risk of T2D. The meta-analysis was conducted using both random- and fixed-effects models to calculate weighted mean differences (WMDs) or risk ratios with 95% CIs. Subgroup analyses were conducted based on intervention type (folate alone or in combination with other B vitamins), as well as analysis based on population characteristics, duration, dose, and change in homocysteine. Results When compared with placebo, folate supplementation lowered fasting insulin (WMD: -13.47 pmol/L; 95% CI: -21.41, -5.53 pmol/L; P < 0.001) and HOMA-IR (WMD: -0.57 units; 95% CI: -0.76, -0.37 units; P < 0.0001), but no overall effects were observed for fasting glucose or HbA1c. Heterogeneity was low in all meta-analyses, and subgroup analysis showed no signs of effect modification except for change in homocysteine, with the most pronounced effects in trials with a change of >2.5 µmol/L. Changes in homocysteine after folate supplementation correlated with changes in fasting glucose (β = 0.07; 95% CI: 0.01, 0.14; P = 0.025) and HbA1c (β = 0.46; 95% CI: 0.06, 0.85; P = 0.02). Only 2 studies examined folate supplementation on risk of T2D, and they found no change in RR (pooled RR: 0.91; 95% CI: 0.80, 1.04; P = 0.16). Conclusion Folate supplementation might be beneficial for glucose homeostasis and lowering IR, but at present there are insufficient data to conclusively determine the effect on development of T2D. This trial was registered on the Prospero database as CRD42016048254.
Collapse
Affiliation(s)
- Mads Vendelbo Lind
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark.,Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Lotte Lauritzen
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Mette Kristensen
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Alastair B Ross
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Jane Nygaard Eriksen
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
18
|
Zhao M, Yuan MM, Yuan L, Huang LL, Liao JH, Yu XL, Su C, Chen YH, Yang YY, Yu H, Xu DX. Chronic folate deficiency induces glucose and lipid metabolism disorders and subsequent cognitive dysfunction in mice. PLoS One 2018; 13:e0202910. [PMID: 30153273 PMCID: PMC6112663 DOI: 10.1371/journal.pone.0202910] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022] Open
Abstract
Previous studies have shown that folate levels were decreased in patients with type 2 diabetes (T2D) and further lowered in T2D patients with cognitive impairment. However, whether folate deficiency could cause T2D and subsequent cognitive dysfunction is still unknown. The present study aimed to explore the effects of chronic folate deficiency (CFD) on glucose and lipid metabolism and cognitive function in mice. Seven-week-old mice were fed with either a CFD or control diet for 25 weeks. Serum folate was significantly reduced, whereas serum total homocysteine was significantly increased in the CFD group. Moreover, CFD induced obesity after a 6-week diet treatment, glucose intolerance and insulin resistance after a 16-week-diet treatment. In addition, CFD reduced the hepatic p-Akt/Akt ratio in response to acute insulin administration. Moreover, CFD increased serum triglyceride levels, upregulated hepatic Acc1 and Fasn mRNA expression, and downregulated hepatic Cd36 and ApoB mRNA expression. After a 24-week diet treatment, CFD induced anxiety-related activities and impairment of spatial learning and memory performance. This study demonstrates that folate deficiency could induce obesity, glucose and lipid metabolism disorders and subsequent cognitive dysfunction.
Collapse
Affiliation(s)
- Mei Zhao
- School of Nursing, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, China
| | - Man Man Yuan
- School of Nursing, Anhui Medical University, Hefei, China
| | - Li Yuan
- School of Nursing, Anhui Medical University, Hefei, China
| | - Li Li Huang
- School of Nursing, Anhui Medical University, Hefei, China
| | - Jian Hong Liao
- School of Nursing, Anhui Medical University, Hefei, China
| | - Xiao Ling Yu
- School of Nursing, Anhui Medical University, Hefei, China
| | - Chang Su
- School of Nursing, Anhui Medical University, Hefei, China
| | - Yuan Hua Chen
- Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, China
- Department of Histology and Embryology, Anhui Medical University, Hefei, China
| | - Yu Ying Yang
- School of Nursing, Anhui Medical University, Hefei, China
| | - Huan Yu
- School of Nursing, Anhui Medical University, Hefei, China
| | - De Xiang Xu
- Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, China
- Department of Toxicology, Anhui Medical University, Hefei, China
| |
Collapse
|
19
|
Stracquadanio M, Ciotta L, Palumbo MA. Effects of myo-inositol, gymnemic acid, and L-methylfolate in polycystic ovary syndrome patients. Gynecol Endocrinol 2018; 34:495-501. [PMID: 29265900 DOI: 10.1080/09513590.2017.1418852] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous endocrine and metabolic disorder, characterized by chronic anovulation/oligomenorrhea, hyperandrogenism, and insulin-resistance. Moreover, some studies propose a possible association between insulin resistance and hyperhomocysteinemia, which is a significant long-term risk for factor for atherogenesis and chronic vascular damage, especially in situations where insulin levels are increased. Insulin-sensitizing agents are used in the treatment of PCOS: in fact, inositols were shown to have insulin-mimetic properties. Synergic action to myo-inositol is that of gymnemic acids that have antidiabetic, anti-sweetener, and anti-inflammatory activities. Gymnemic acid formulations have also been found useful against obesity due to their ability to delay the glucose absorption in the blood. L-methyl-folate increases peripheral sensitivity to insulin, maintaining folatemia stable, and thus restoring normal homocysteine levels. Unlike folic acid, L-methyl folate has a higher bioavailability, no drug/food interferences, high absorption, and it is stable to UV-A exposure. The aim of our study is to compare the clinical, endocrine, and metabolic parameters in 100 PCOS women treated with myo-inositol, gymnemic acid, and l-methylfolate (Group A) or myo inositol and folic acid only (Group B), continuously for 6 months. From a clinical point of view, it was noticed a more significant improvement of the menstrual cycle regularity and a more significant reduction of BMI in Group A. Moreover, a more significant decrease of total testosterone and increase of SHBG serum levels were noticed in Group A. The metabolic assessment found a more significant decrease of total cholesterol and homocysteine levels; OGTT glycemia and insulinemia values were significantly more improved after treatment with myo-inositol + gymnemic acid. In conclusion, we can state that a good option for the treatment of PCOS is the combined administration of myo-inositol + gymnemic acid + l-methyl-folate, especially for overweight/obese patients with marked insulin resistance and with associated hyperhomocysteinemia.
Collapse
Affiliation(s)
- M Stracquadanio
- a Institute of Obstetric and Gynecological Pathology , Santo Bambino Hospital, University of Catania , Catania , Italy
| | - L Ciotta
- a Institute of Obstetric and Gynecological Pathology , Santo Bambino Hospital, University of Catania , Catania , Italy
| | - M A Palumbo
- a Institute of Obstetric and Gynecological Pathology , Santo Bambino Hospital, University of Catania , Catania , Italy
| |
Collapse
|
20
|
Lind MV, Lauritzen L, Vestergaard H, Hansen T, Pedersen O, Kristensen M, Ross AB. One-carbon metabolism markers are associated with cardiometabolic risk factors. Nutr Metab Cardiovasc Dis 2018; 28:402-410. [PMID: 29499850 DOI: 10.1016/j.numecd.2018.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/03/2018] [Accepted: 01/16/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Alterations to one-carbon metabolism, especially elevated plasma homocysteine (Hcy), have been suggested to be both a cause and a consequence of the metabolic syndrome (MS). A deeper understanding of the role of other one-carbon metabolites in MS, including s-adenosylmethionine (SAM), s-adenosylhomocysteine (SAH), and the methylation capacity index (SAM:SAH ratio) is required. METHODS AND RESULTS 118 men and women with MS-risk factors were included in this cross-sectional study and cardiometabolic outcomes along with markers of one-carbon metabolism, including fasting plasma SAM, SAH, Hcy and vitamin B12 concentrations, were analysed. Multiple linear regression models were also used to examine the association between plasma one-carbon metabolites and cardiometabolic health features. We found that fasting plasma concentrations of Hcy, SAM and SAH were all positively correlated with markers of adiposity, including BMI (increase in BMI per 1-SD increase in one-carbon metabolite: 0.92 kg/m2 95% CI (0.28; 1.56), p = 0.005; 0.81 (0.15; 1.47), p = 0.02; 0.67 (-0.01; 1.36), p = 0.05, respectively). Hcy, but not SAM, SAH or SAM:SAH ratio was associated with BMI and body fat percentage after mutual adjustments. SAM concentrations were associated with higher fasting insulin (9.5% 95% CI (0.3; 19.5) per SD increase in SAM, p = 0.04), HOMA-IR (10.8% (0.8; 21.9), p = 0.03) and TNF-α (11.8% (5.0; 19.0), p < 0.001). CONCLUSION We found little evidence for associations between SAM:SAH ratio and cardiometabolic variables, but higher plasma concentrations of SAM, SAH and Hcy are related to an overall higher risk of metabolic dysfunctions. The studies were registered at www.clinicaltrials.gov (NCT01719913 &NCT01731366).
Collapse
Affiliation(s)
- M V Lind
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark; Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| | - L Lauritzen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - H Vestergaard
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center, Gentofte, Denmark
| | - T Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - O Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M Kristensen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - A B Ross
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
21
|
Ismaeel A. Effects of Betaine Supplementation on Muscle Strength and Power: A Systematic Review. J Strength Cond Res 2018; 31:2338-2346. [PMID: 28426517 DOI: 10.1519/jsc.0000000000001959] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Ismaeel, A. Effects of betaine supplementation on muscle strength and power: a systematic review. J Strength Cond Res 31(8): 2338-2346, 2017-Betaine (BET) has recently gotten much attention for its potential role as an ergogenic aid and has become a common ingredient in peri-workout dietary supplements. The purpose of this systematic review of the literature was to investigate the effects of BET supplementation on muscular strength and power. A computerized literature search of 3 databases (PubMed, MEDLINE, and Scopus) was performed. Included in the review were randomized controlled trials that measured muscle strength, power, or both measures in healthy participants. Quality was assessed using the PEDro scale. Seven trials were identified, and quality assessment showed that all were of "excellent quality." Only 2 of the studies reported increases in strength or power after supplementation with BET. The remaining 5 studies showed no change in any strength or power outcome measures with BET supplementation. This review highlights the lack of evidence for a clear ergogenic effect of BET supplementation on strength and power performance. However, because 2 studies did identify increases of up to 24.61% on strength and power as a result of BET supplementation, additional studies are necessary to evaluate its effectiveness.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Department of Family and Consumer Sciences, Nutrition Sciences, Baylor University, Waco, Texas
| |
Collapse
|
22
|
Fu S, Yao Y, Zhao Y, Luan F. Relationships of Hyperhomocysteinemia and Hyperuricemia With Metabolic Syndrome and Renal Function in Chinese Centenarians. Front Endocrinol (Lausanne) 2018; 9:502. [PMID: 30271378 PMCID: PMC6142902 DOI: 10.3389/fendo.2018.00502] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/09/2018] [Indexed: 11/30/2022] Open
Abstract
As the first time worldwide, this study aimed to investigate the relationships of hyperhomocysteinemia and hyperuricemia with metabolic syndrome (MetS) and renal function in Chinese centenarians. The China Hainan Centenarian Cohort Study was performed in 18 cities and counties of the Hainan Province. Home interview, physical examination, and blood analysis were performed on 808 centenarians following standard procedures. All centenarians had a median age of 102 (100-115) years. Prevalence of hyperhomocysteinemia and hyperuricemia was 91.6% (740 centenarians) and 28.5% (230 centenarians), respectively. The MetS was present in 117 centenarians (14.5%). In simple correlation analyses, hyperhomocysteinemia and hyperuricemia were significantly correlated with MetS and glomerular filtration rate (GFR) < 60 ml/min/1.73 m2 (P < 0.05 for all). Serum homocysteine levels were significantly correlated with GFR, waist circumference (WC), and triglyceride levels, while serum uric acid levels were significantly correlated with these variables plus high-density lipoprotein cholesterol (HDL-C) levels (P < 0.05 for all). In logistic regression analyses, hyperhomocysteinemia and hyperuricemia were significantly associated with MetS and GFR < 60 ml/min/1.73 m2 (P < 0.05 for all). In linear regression analyses, serum homocysteine levels were significantly associated with GFR, WC, and triglyceride, while serum uric acid levels were significantly associated with these variables plus HDL-C (P < 0.05 for all). Both hyperhomocysteinemia and hyperuricemia had important relationships with MetS and renal function in Chinese centenarians. Hyperuricemia and hyperhomocysteinemia that could help identify, while also affecting, the development of MetS and renal function may unfold complex relationships between MetS, renal function, and cardiovascular risk and provide effective prevention strategies for these conditions.
Collapse
Affiliation(s)
- Shihui Fu
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, China
- Department of Cardiology and Hainan Branch, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yao Yao
- Institute of Geriatrics, Chinese People's Liberation Army General Hospital, Beijing, China
- Beijing Key Laboratory of Geriatrics, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yali Zhao
- Central Laboratory, Hainan Branch of Chinese People's Liberation Army General Hospital, Sanya, China
- *Correspondence: Yali Zhao
| | - Fuxin Luan
- Central Laboratory, Hainan Branch of Chinese People's Liberation Army General Hospital, Sanya, China
- Fuxin Luan
| |
Collapse
|
23
|
Cholewa JM, Dardevet D, Lima-Soares F, de Araújo Pessôa K, Oliveira PH, Dos Santos Pinho JR, Nicastro H, Xia Z, Cabido CET, Zanchi NE. Dietary proteins and amino acids in the control of the muscle mass during immobilization and aging: role of the MPS response. Amino Acids 2017; 49:811-820. [PMID: 28175999 DOI: 10.1007/s00726-017-2390-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 01/28/2017] [Indexed: 12/14/2022]
Abstract
Dietary proteins/essential amino acids (EAAs) are nutrients with anabolic properties that may increase muscle mass or attenuate muscle loss during immobilization and aging via the stimulation of muscle protein synthesis (MPS). An EAA's anabolic threshold, capable to maximize the stimulation of MPS has been hypothesized, but during certain conditions associated with muscle loss, this anabolic threshold seems to increase which reduces the efficacy of dietary EAAs to stimulate MPS. Preliminary studies have demonstrated that acute ingestion of dietary proteins/EAA (with a sufficient amount of leucine) was capable to restore the postprandial MPS during bed rest, immobilization or aging; however, whether these improvements translate into chronic increases (or attenuates loss) of muscle mass is equivocal. For example, although free leucine supplementation acutely increases MPS and muscle mass in some chronic studies, other studies have reported no increases in muscle mass following chronic leucine supplementation. In contrast, chronically increasing leucine intake via the consumption of an overall increase in dietary protein appears to be the most effective dietary intervention toward increasing or attenuating lean mass during aging; however, more research investigating the optimal dose and timing of protein ingestion is necessary. Several studies have demonstrated that decreases in postprandial MPS as a result of increased circulating oxidative and inflammatory are more responsible than muscle protein breakdown for the decreases in muscle mass during disuse and health aging. Therefore, nutritional interventions that reduce oxidation or inflammation in conjunction with higher protein intakes that overcome the anabolic resistance may enhance the MPS response to feeding and either increase muscle mass or attenuate loss. In preliminary studies, antioxidant vitamins and amino acids with antioxidant or anti-inflammatory properties show potential to restore the anabolic response associated with protein ingestion. More research, however, is required to investigate if these nutrients translate to increases in MPS and, ultimately, increased lean mass in aging humans. The purpose of the present review is to discuss the role of protein/EAA intake to enhance postprandial MPS during conditions associated with muscle loss, and bring new perspectives and challenges associated nutritional interventions aimed to optimize the anabolic effects of dietary protein/EAAs ingestion.
Collapse
Affiliation(s)
- Jason M Cholewa
- Department of Kinesiology, Coastal Carolina University, Conway, SC, 29528, USA
| | | | - Fernanda Lima-Soares
- Federal University of Maranhão (UFMA), Department of Physical Education, São Luis, Maranhão, Brazil.,Laboratory of Cellular and Molecular Biology of Skeletal Muscle (LABCEMME), São Luis, Maranhão, Brazil
| | - Kassiana de Araújo Pessôa
- Federal University of Maranhão (UFMA), Department of Physical Education, São Luis, Maranhão, Brazil.,Laboratory of Cellular and Molecular Biology of Skeletal Muscle (LABCEMME), São Luis, Maranhão, Brazil
| | - Paulo Henrique Oliveira
- Federal University of Maranhão (UFMA), Department of Physical Education, São Luis, Maranhão, Brazil.,Laboratory of Cellular and Molecular Biology of Skeletal Muscle (LABCEMME), São Luis, Maranhão, Brazil
| | - João Ricardo Dos Santos Pinho
- Federal University of Maranhão (UFMA), Department of Physical Education, São Luis, Maranhão, Brazil.,Laboratory of Cellular and Molecular Biology of Skeletal Muscle (LABCEMME), São Luis, Maranhão, Brazil
| | - Humberto Nicastro
- Laboratory of Cellular and Molecular Biology of Skeletal Muscle (LABCEMME), São Luis, Maranhão, Brazil
| | - Zhi Xia
- Exercise Physiology and Biochemistry Laboratory, College of Physical Education, Jinggangshan University, Ji'an, China.,Department of Sports Medicine, Chengdu Sport Institute, Chengdu, China
| | - Christian Emmanuel Torres Cabido
- Federal University of Maranhão (UFMA), Department of Physical Education, São Luis, Maranhão, Brazil.,Laboratory of Cellular and Molecular Biology of Skeletal Muscle (LABCEMME), São Luis, Maranhão, Brazil
| | - Nelo Eidy Zanchi
- Federal University of Maranhão (UFMA), Department of Physical Education, São Luis, Maranhão, Brazil. .,Laboratory of Cellular and Molecular Biology of Skeletal Muscle (LABCEMME), São Luis, Maranhão, Brazil.
| |
Collapse
|
24
|
Hu Y, Liu J, Dong X, Xu Y, Leng S, Wang G. Clinical Study of Serum Homocysteine and Non-Alcoholic Fatty Liver Disease in Euglycemic Patients. Med Sci Monit 2016; 22:4146-4151. [PMID: 27803497 PMCID: PMC5103834 DOI: 10.12659/msm.897924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease. NAFLD includes a spectrum of hepatic pathologies: simple fatty liver, steatohepatitis and cirrhosis. Insulin resistance may contribute to NAFLD. The liver plays an important role in the production and metabolism of homocysteine (HCY), which is known to be an independent risk factor for cardiovascular disease. High HCY level can aggravate NAFLD by increasing the reactive oxygen species and activating oxidative stress. In this study, we investigated the relationship between HCY and NAFLD in euglycemic patients. MATERIAL AND METHODS A total of 1143 euglycemic patients were recruited: 519 patients with non-alcoholic fatty liver disease (NAFLD) and 624 sex and age-matched controls without NAFLD. RESULTS The NAFLD group had significantly higher HCY level (13.78±5.84 vs. 11.96±3.58 mmol/L, p<0.001), as well as higher body mass index (BMI), total cholesterol (TC), low density lipoprotein cholesterol (LDL-C), triglyceride (TG), glutamic-pyruvic transaminase (ALT), glutamic-oxalacetic transaminase (AST), fasting plasma glucose (FPG), fasting insulin (FINS), homeostasis model assessment for insulin resistance (HOMA-IR), homeostasis model assessment for beta cell function (HOMA-B), and lower high density lipoprotein cholesterol (HDL-C). HCY level was positively correlated with HOMA-IR (r=0.239, p<0.001), TG (r=0.356, p<0.001) and negatively correlated with HDL-C (r=-0.161, p<0.001). In the logistic regression analysis, BMI (beta=0.345, p<0.001), HOMA-IR (beta=0.654, p<0.01), TG (beta=0.881, p<0.001), and HCY (beta=0.04, p=0.044) were the predictors of NAFLD. CONCLUSIONS Higher HCY level existed in NAFLD patients and was correlated with the severity of insulin resistance. HCY is an independent risk factor for NAFLD.
Collapse
Affiliation(s)
- Yanjin Hu
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Xuejie Dong
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Yuan Xu
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Song Leng
- Health Management Center, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China (mainland)
| |
Collapse
|
25
|
Gao X, Wang Y, Randell E, Pedram P, Yi Y, Gulliver W, Sun G. Higher Dietary Choline and Betaine Intakes Are Associated with Better Body Composition in the Adult Population of Newfoundland, Canada. PLoS One 2016; 11:e0155403. [PMID: 27166611 PMCID: PMC4863971 DOI: 10.1371/journal.pone.0155403] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/28/2016] [Indexed: 02/07/2023] Open
Abstract
Background Choline is an essential nutrient and betaine is an osmolyte and methyl donor. Both are important to maintain health including adequate lipid metabolism. Supplementation of dietary choline and betaine increase muscle mass and reduce body fat in animals. However, little data is available regarding the role of dietary choline and betaine on body composition in humans. Objective To investigate the association between dietary choline and betaine intakes with body composition in a large population based cross-sectional study. Design A total of 3214 subjects from the CODING (Complex Disease in Newfoundland population: Environment and Genetics) study were assessed. Dietary choline and betaine intakes were computed from the Willett Food Frequency questionnaire. Body composition was measured using dual-energy X-ray absorptiometry following a 12-hour fast. Major confounding factors including age, sex, total calorie intake and physical activity level were controlled in all analyses. Result Significantly inverse correlations were found between dietary choline and betaine intakes, with all obesity measurements: total percent body fat (%BF), percent trunk fat (%TF), percent android fat (%AF), percent gynoid fat (%GF) and anthropometrics: weight, body mass index, waist circumference, waist-to-hip ratio in both women and men (r range from -0.13 to -0.47 for choline and -0.09 to -0.26 for betaine, p<0.001 for all). Dietary choline intake had stronger association than betaine. Moreover, obese subjects had the lowest dietary choline and betaine intakes, with overweight subjects in the middle, and normal weight subjects consumed the highest dietary choline and betaine (p<0.001). Vice versa, when subjects were ranked according to dietary choline and betaine intakes, subjects with the highest intake of both had the lowest %TF, %AF, %GF, %BF and highest %LM among the groups in both sexes. Conclusion Our findings indicate that high dietary choline and betaine intakes are significantly associated with favorable body composition in humans.
Collapse
Affiliation(s)
- Xiang Gao
- College of Food Science and Engineering, Ocean University of China, No.5, Yu Shan Road, Qingdao, Shandong Province, China
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John’s, NL, Canada
| | - Yongbo Wang
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John’s, NL, Canada
- The Department of Endocrinology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Edward Randell
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John’s, NL, Canada
| | - Pardis Pedram
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John’s, NL, Canada
| | - Yanqing Yi
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John’s, NL, Canada
| | - Wayne Gulliver
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John’s, NL, Canada
| | - Guang Sun
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John’s, NL, Canada
- * E-mail:
| |
Collapse
|
26
|
Serum betaine is inversely associated with low lean mass mainly in men in a Chinese middle-aged and elderly community-dwelling population. Br J Nutr 2016; 115:2181-8. [DOI: 10.1017/s0007114516001380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AbstractPrevious studies have demonstrated that betaine supplements increase lean body mass in livestock and improve muscle performance in human beings, but evidence for its effect on human lean mass is limited. Our study assessed the association of circulating betaine with lean mass and its composition in Chinese adults. A community-based study was conducted on 1996 Guangzhou residents (weight/mass: 1381/615) aged 50–75 years between 2008 and 2010. An interviewer-administered questionnaire was used to collect general baseline information. Fasting serum betaine was assessed using HPLC-MS. A total of 1590 participants completed the body composition analysis performed using dual-energy X-ray absorptiometry during a mean of 3·2 years of follow-up. After adjustment for age, regression analyses demonstrated a positive association of serum betaine with percentage of lean mass (LM%) of the entire body, trunk and limbs in men (all P<0·05) and LM% of the trunk in women (P=0·016). Each sd increase in serum betaine was associated with increases in LM% of 0·609 (whole body), 0·811 (trunk), 0·422 (limbs), 0·632 (arms) and 0·346 (legs) in men and 0·350 (trunk) in women. Multiple logistic regression analysis revealed that the prevalence of lower LM% decreased by 17 % (whole body) and 14 % (trunk) in women and 23 % (whole body), 28 % (trunk), 22 % (arms) and 26 % (percentage skeletal muscle index) in men with each sd increment in serum betaine. Elevated circulating betaine was associated with a higher LM% and lower prevalence of lower LM% in middle-aged and elderly Chinese adults, particularly men.
Collapse
|
27
|
Associations of MTHFR C677T and MTRR A66G gene polymorphisms with metabolic syndrome: a case-control study in Northern China. Int J Mol Sci 2014; 15:21687-702. [PMID: 25429430 PMCID: PMC4284672 DOI: 10.3390/ijms151221687] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 11/03/2014] [Accepted: 11/12/2014] [Indexed: 11/17/2022] Open
Abstract
Prior evidence indicates that homocysteine plays a role in the development of metabolic syndrome (MetS). Methylenetetrahydrofolate reductase (MTHFR) C677T and methionine synthase reductase (MTRR) A66G polymorphisms are common genetic determinants of homocysteine levels. To investigate the associations of the MTHFR C677T and MTRR A66G polymorphisms with MetS, 692 Chinese Han subjects with MetS and 878 controls were recruited. The component traits of MetS and the MTHFR C677T and MTRR A66G genotypes were determined. A significant association was observed between the MTHFR 677T allele and increased risk of MetS, high fasting blood glucose, high waist circumference, and increasing number of MetS components. The MTRR A66G polymorphism was associated with an increased risk of MetS when combined with the MTHFR 677TT genotype, although there was no association found between MetS and MTRR A66G alone. Furthermore, the MTRR 66GG genotype was associated with high fasting blood glucose and triglycerides. Our data suggest that the MTHFR 677T allele may contribute to an increased risk of MetS in the northern Chinese Han population. The MTRR A66G polymorphism is not associated with MetS. However, it may exacerbate the effect of the MTHFR C677T variant alone. Further large prospective population-based studies are required to confirm our findings.
Collapse
|
28
|
Chen FP, Wang HM, Chiang FF, Lin CC, Huang SC, Huang YC. The metabolic syndrome is associated with an increased risk of colorectal polyps independent of plasma homocysteine. ANNALS OF NUTRITION AND METABOLISM 2014; 64:106-12. [PMID: 24969661 DOI: 10.1159/000363418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 05/05/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS The links between the metabolic syndrome and homocysteine in relation to the risk of colorectal polyps are not understood. The purpose of this study was to investigate the association between the metabolic syndrome and homocysteine and further analyze the relationship between these two factors and the risk of colorectal polyps. METHODS This was a case-control study. A total of 135 participants with colorectal polyps (cases) and 110 participants without polyps (controls) were recruited. RESULTS There were 59 participants with the metabolic syndrome in the case group and 36 participants with the metabolic syndrome in the control group. The metabolic syndrome and its individual components, except for serum triglycerides, and homocysteine were associated with the risk of colorectal polyps. When the association of the metabolic syndrome and homocysteine with the risk of colorectal polyps was simultaneously considered, the association between homocysteine and the risk of colorectal polyps disappeared, but waist circumference, systolic and diastolic blood pressure, high-density lipoprotein cholesterol, and the metabolic syndrome itself were still significant risk factors for the development of colorectal polyps. CONCLUSION Although the metabolic syndrome and plasma homocysteine were individually related to the risk of colorectal polyps, the metabolic syndrome was a major contributing factor in relation to the risk of colorectal polyps independent of plasma homocysteine.
Collapse
Affiliation(s)
- Fang-Pei Chen
- School of Nutrition, Chung Shan Medical University, Taichung, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
29
|
Saddiqa A, Raza AR, Black DS, Kumar N. Chiron based synthesis of isocoumarins: reactivity of α-substituted carboxylic acids. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.tetasy.2014.03.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
30
|
Cholewa JM, Guimarães-Ferreira L, Zanchi NE. Effects of betaine on performance and body composition: a review of recent findings and potential mechanisms. Amino Acids 2014; 46:1785-93. [DOI: 10.1007/s00726-014-1748-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 04/08/2014] [Indexed: 01/22/2023]
|
31
|
Cholewa JM, Wyszczelska-Rokiel M, Glowacki R, Jakubowski H, Matthews T, Wood R, Craig SA, Paolone V. Effects of betaine on body composition, performance, and homocysteine thiolactone. J Int Soc Sports Nutr 2013; 10:39. [PMID: 23967897 PMCID: PMC3844502 DOI: 10.1186/1550-2783-10-39] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 08/09/2013] [Indexed: 11/17/2022] Open
Abstract
Background This study investigated the effects of long term betaine supplementation on body composition, performance, and homocysteine thiolactone (HCTL) in experienced strength trained men. Methods Twenty-three subjects were matched for training experience (4.8 ± 2.3 years) and body fat percentage (BF%: 16.9 ± 8.0%), randomly assigned to either a placebo (PL; n = 12) or betaine group (BET; n = 11; 2.5 g/day), and completed a 6 week periodized training program consisting of 3 two-week micro-cycles. Bench press and back squat training volumes were recorded and changes in training volume were assessed at each micro-cycle. Fasting urine was collected at baseline (BL), weeks 2, 4 and 6, and assayed for HCTL. Subjects were tested prior to and following 6 weeks of treatment. Arm and thigh cross sectional area (CSA) was estimated via girth and skin fold measurements. Body density was estimated via skin fold calipers and used to estimate BF%, fat mass (FM), and lean body mass (LBM). Performance was assessed via vertical jump (VJ), bench press 1 RM (BP), and back squat 1 RM (BS). Results Arm CSA increased significantly (p < .05) in BET but not PL. No differences existed between group and time for changes in thigh CSA. Back squat training volume increased significantly (p < .05) for both groups throughout training. Bench press training volume was significantly (p < .05) improved for BET compared to PL at microcycles one and three. Body composition (BF%, FM, LBM) improved significantly (p < .05) in BET but not PL. No differences were found in performance variables (BP, BS, VJ) between groups, except there was a trend (p = .07) for increased VJ power in BET versus PL. A significant interaction (p < .05) existed for HCTL, with increases from BL to week 2 in PL, but not BET. Additionally, HCTL remained elevated at week 4 in PL, but not BET. Conclusion Six-weeks of betaine supplementation improved body composition, arm size, bench press work capacity, attenuated the rise in urinary HCTL, and tended to improve power (p = .07) but not strength.
Collapse
Affiliation(s)
- Jason M Cholewa
- Department of Kinesiology, Recreation, and Sport Studies, Coastal Carolina University, Conway, SC, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Lambrinoudaki I, Papadimitriou D, Kaparos G, Rizos D, Panoulis C, Deligeoroglou E, Alexandrou A, Auguolea A, Apostolakis M, Creatsa M, Kouskouni E. MTHFR C677T polymorphism modifies the effect of HRT on metabolic parameters in postmenopausal women. Climacteric 2012; 16:568-75. [PMID: 23145891 DOI: 10.3109/13697137.2012.738722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To assess the interaction of the MTHFR C677T polymorphism with changes in lipid and glucose metabolism effected by oral hormone replacement therapy (HRT) in postmenopausal women. METHODS In this open-label, prospective, interventional study, parameters of lipid and glucose metabolism, as well as homocysteine, were assessed in 97 postmenopausal women at baseline and 1 year after the initiation of HRT. Participants were stratified into three subgroups, according to the MTHFR C677T polymorphism (wild-type: CC genotype; heterozygous: CT genotype; homozygous for the mutant variable: TT genotype). RESULTS The TT genotype was associated with an elevation of total and low density lipoprotein (LDL) cholesterol, while CT and CC genotypes were associated with a reduction of total cholesterol and LDL cholesterol after 1 year of HRT (p = 0.032 for total cholesterol and p = 0.002 for LDL cholesterol). Women with the TT genotype had higher glucose levels in contrast to women with the CC genotype who had lower glucose levels after 1 year of HRT (p = 0.011). Additionally, CC carriers under HRT had a significant elevation of apolipoprotein A1 levels (p = 0.018), contrarily to CT and TT genotypes. CONCLUSION While HRT was associated with favorable changes in lipid and metabolic parameters in carriers of the CC genotype, this effect was not evident in carriers of the T allele. The MTHFR C677T polymorphism may modify the effect of HRT on lipid and metabolic parameters in postmenopausal women.
Collapse
Affiliation(s)
- I Lambrinoudaki
- 2nd Department of Obstetrics and Gynecology, University of Athens , Aretaieio Hospital, Athens
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yilmaz N. Relationship between paraoxonase and homocysteine: crossroads of oxidative diseases. Arch Med Sci 2012; 8:138-53. [PMID: 22457688 PMCID: PMC3309450 DOI: 10.5114/aoms.2012.27294] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 02/17/2011] [Accepted: 04/07/2011] [Indexed: 01/14/2023] Open
Abstract
Homocysteine (Hcy) is an accepted independent risk factor for several major pathologies including cardiovascular disease, birth defects, osteoporosis, Alzheimer's disease, and renal failure. Interestingly, many of the pathologies associated with homocysteine are also linked to oxidative stress. The enzyme paraoxonase (PON1) - so named because of its ability to hydrolyse the toxic metabolite of parathion, paraoxon - was also shown early after its identification to manifest arylesterase activity. Although the preferred endogenous substrate of PON1 remains unknown, lactones comprise one possible candidate class. Homocysteine-thiolactone can be disposed of by enzymatic hydrolysis by the serum Hcy-thiolactonase/paraoxonase carried on high-density lipoprotein (HDL). In this review, Hcy and the PON1 enzyme family were scrutinized from different points of view in the literature and the recent articles on these subjects were examined to determine whether these two molecular groups are related to each other like a coin with two different sides, so close and yet so different and so opposite.
Collapse
Affiliation(s)
- Necat Yilmaz
- Central Laboratories of Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| |
Collapse
|
34
|
Pansuria M, Xi H, Li L, Yang XF, Wang H. Insulin resistance, metabolic stress, and atherosclerosis. Front Biosci (Schol Ed) 2012; 4:916-31. [PMID: 22202099 PMCID: PMC3319745 DOI: 10.2741/s308] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Atherosclerosis, a pathological process that underlies the development of cardiovascular disease, is the primary cause of morbidity and mortality in patients with type 2 diabetes mellitus (T2DM). T2DM is characterized by hyperglycemia and insulin resistance (IR), in which target tissues fail to respond to insulin. Systemic IR is associated with impaired insulin signaling in the metabolic tissues and vasculature. Insulin receptor is highly expressed in the liver, muscle, pancreas, and adipose tissue. It is also expressed in vascular cells. It has been suggested that insulin signaling in vascular cells regulates cell proliferation and vascular function. In this review, we discuss the association between IR, metabolic stress, and atherosclerosis with focus on 1) tissue and cell distribution of insulin receptor and its differential signaling transduction and 2) potential mechanism of insulin signaling impairment and its role in the development of atherosclerosis and vascular function in metabolic disorders including hyperglycemia, hypertension, dyslipidemia, and hyperhomocysteinemia. We propose that insulin signaling impairment is the foremost biochemical mechanism underlying increased cardiovascular morbidity and mortality in atherosclerosis, T2DM, and metabolic syndrome.
Collapse
Affiliation(s)
- Meghana Pansuria
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, 19140
| | - Hang Xi
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, 19140
| | - Le Li
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- School of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR, China
| | - Xiao-Feng Yang
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, 19140
| | - Hong Wang
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, 19140
- Thrombosis Research Center of Temple University School of Medicine, Philadelphia, PA, 19140
| |
Collapse
|
35
|
Jakubowski H. The role of paraoxonase 1 in the detoxification of homocysteine thiolactone. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 660:113-27. [PMID: 20221875 DOI: 10.1007/978-1-60761-350-3_11] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The thioester homocysteine (Hcy)-thiolactone, product of an error-editing reaction in protein biosynthesis, forms when Hcy is mistakenly selected by methionyl-tRNA synthetase. Accumulating evidence suggests that Hcy-thiolactone plays an important role in atherothrombosis. The thioester chemistry of Hcy-thiolactone underlies its ability to form isopeptide bonds with protein lysine residues, which impairs or alters protein function and has pathophysiological consequences including activation of an autoimmune response and enhanced thrombosis. Mammalian organisms, including human, have evolved the ability to eliminate Hcy-thiolactone. One such mechanism involves paraoxonase 1 (PON1), which has the ability to hydrolyze Hcy-thiolactone. This article outlines Hcy-thiolactone pathobiology and reviews evidence documenting the role of PON1 in minimizing Hcy-thiolactone and N-Hcy-protein accumulation.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, University of Life Sciences, Poznań, Poland.
| |
Collapse
|
36
|
Liu WJ, Ma LQ, Liu WH, Zhou W, Zhang KQ, Zou CG. Inhibition of hepatic glycogen synthesis by hyperhomocysteinemia mediated by TRB3. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1489-99. [PMID: 21435438 DOI: 10.1016/j.ajpath.2010.12.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 12/21/2010] [Accepted: 12/30/2010] [Indexed: 01/13/2023]
Abstract
Recently, epidemiological and experimental studies have linked hyperhomocysteinemia (HHcy) to insulin resistance. However, whether HHcy impairs glucose homeostasis by affecting glycogenesis in the liver is not clear. In the present study, we investigated the effect of HHcy on hepatic glycogen synthesis. Hyperhomocysteinemia was induced in mice by drinking water containing two percent methionine. Mice with HHcy showed an increase in the phosphorylation of glycogen synthase and a significant decrease in hepatic glycogen content and the rate of glycogen synthesis. The expression of TRB3 (tribbles-related protein 3) was up-regulated in the liver of mice with HHcy, concomitantly with the dephosphorylation of glycogen synthase kinase-3β and Akt. The knockdown of TRB3 by short hairpin RNA suppressed the dephosphorylation of these two kinases. Homocysteine induced an increase in the levels of hepatic cAMP and cAMP response element-binding protein phosphorylation, which in turn up-regulated the expression of peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1α and TRB3. The inhibition of PPAR-α by its inhibitor, MK886, or knockdown of PPAR-α by small interfering RNA significantly inhibited the expression of TRB3 induced by homocysteine. The current study demonstrates that HHcy impairs hepatic glycogen synthesis by inducing the expression of TRB3. These results provide a novel explanation for the development and progression of insulin resistance in HHcy.
Collapse
Affiliation(s)
- Wen-Jing Liu
- Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Yunnan, China
| | | | | | | | | | | |
Collapse
|
37
|
Shimomura T, Anan F, Masaki T, Umeno Y, Eshima N, Saikawa T, Yoshimatsu H, Fujiki M, Kobayashi H. Homocysteine levels are associated with hippocampus volume in type 2 diabetic patients. Eur J Clin Invest 2011; 41:751-8. [PMID: 21250986 DOI: 10.1111/j.1365-2362.2010.02464.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Elevated total plasma homocysteine (tHcy) levels are associated with cognitive dysfunction, in which changes in the hippocampus plausibly play a pivotal role. We tested the hypothesis that elevated tHcy levels are correlated with hippocampus volume and insulin resistance in nondementia patients with type 2 diabetes. MATERIALS AND METHODS The study included 43 nondementia patients with type 2 diabetes, who were divided into two groups: a high tHcy group (age: 65 ± 8 years, mean ± standard deviation, n = 16) and a normal tHcy group (64 ± 9 years, n = 27). Hippocampus volume was quantified with a computer-assisted analysis using a magnetic resonance imaging (MRI) voxel-based specific regional analysis system developed for the study of Alzheimer's disease (VSRAD), which yields a Z-score as the end point for the assessment of hippocampal volume. Results The Z-score was higher in the high tHcy group compared to the normal tHcy group (P < 0·0001). The fasting plasma glucose (P < 0·01) and insulin (P < 0·0001) concentrations and the homoeostasis model assessment (HOMA) index (P < 0·0001) were higher in the high tHcy group than in the normal tHcy group. Multiple regression analysis showed that the main factors that influenced tHcy levels may be the Z-score and the HOMA index. CONCLUSIONS Our results indicate that the elevated levels of tHcy in Japanese nondementia patients with type 2 diabetes are characterised by hippocampal atrophy and insulin resistance and that the Z-score and HOMA index may be the primary factors that influence tHcy levels.
Collapse
Affiliation(s)
- Tsuyoshi Shimomura
- Department of Neurosurgery, Faculty of Medicine, Oita University, Oita, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Liu Z, Luo Y, Jeppesen PB, Gregersen S, Hermansen K. Amino acid-induced gene expression profiling in clonal β-cell line INS-1E cells. Diabetes Metab Res Rev 2011; 27:120-76. [PMID: 21294238 DOI: 10.1002/dmrr.1153] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND There is abundant evidence that glucotoxicity and lipotoxicity contribute to impaired β-cell function in type 2 diabetes. Interestingly, amino acid (AA) derangement is also a characteristic part of the diabetic state. The acute effects of AA on pancreatic β-cell function have been widely explored; however, to our knowledge, the chronic effects of AA, e.g. proline (Pro), homocysteine (Hcy), and leucine (Leu), on pancreatic β-cell function and integrity have not yet been studied. We aimed to investigate global alterations in β-cell gene expression after long-term exposure of clonal INS-1E cells to elevated level of specific AA in vitro. METHODS Global gene expression profiling was performed to characterize genes differently modified by Pro, Hcy, and Leu, respectively, in INS-1E cells. RESULTS Gene expression profiling revealed significant changes in INS-1E cell mRNAs involved in the control of several aspects of β-cell function, e.g. epigenetic regulation of gene expression, metabolism, innate and adaptive immune responses, cellular signalling, protein synthesis, apoptosis, and cellular stress response. After 72 h, INS-1E cells were differentially regulated (≥1.5- or ≤ -1.5-fold) by Pro (295 transcripts), Hcy (301 transcripts), and Leu (701 transcripts). It appears that Hcy effects changes opposite to those induced by Leu and/or Pro. CONCLUSIONS AA appears to participate in and to influence many physiological processes including those involved in cholesterol metabolism, immune responses, and oxidative phosphorylation. Whether such events promote the β-cell dysfunction and the β-cell failure in diabetes remains to be elucidated. Our data strongly indicate that AA elevation may take part in the progressive development of type 2 diabetes.
Collapse
Affiliation(s)
- Zhenping Liu
- Department of Endocrinology and Metabolism MEA, Aarhus University Hospital, Aarhus Sygehus THG, Tage-Hansens Gade 2, Aarhus C, Denmark
| | | | | | | | | |
Collapse
|
39
|
El Mesallamy HO, El-Demerdash E, Hammad LN, El Magdoub HM. Effect of taurine supplementation on hyperhomocysteinemia and markers of oxidative stress in high fructose diet induced insulin resistance. Diabetol Metab Syndr 2010; 2:46. [PMID: 20591133 PMCID: PMC2907312 DOI: 10.1186/1758-5996-2-46] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 06/30/2010] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND High intake of dietary fructose is accused of being responsible for the development of the insulin resistance (IR) syndrome. Concern has arisen because of the realization that fructose, at elevated concentrations, can promote metabolic changes that are potentially deleterious. Among these changes is IR which manifests as a decreased biological response to normal levels of plasma insulin. METHODS Oral glucose tolerance tests (OGTT) were carried out, homeostasis model assessment of insulin resistance (HOMA) was calculated, homocysteine (Hcy), lipid concentrations and markers of oxidative stress were measured in male Wistar rats weighing 170-190 g. The rats were divided into four groups, kept on either control diet or high fructose diet (HFD), and simultaneously supplemented with 300 mg/kg/day taurine via intra-peritoneal (i.p.) route for 35 days. RESULTS Fructose-fed rats showed significantly impaired glucose tolerance, impaired insulin sensitivity, hypertriglyceridemia, hypercholesterolemia, hyperhomocysteinemia (HHcy), lower total antioxidant capacity (TAC), lower paraoxonase (PON) activity, and higher nitric oxide metabolites (NOx) concentration, when compared to rats fed on control diet. Supplementing the fructose-fed rats with taurine has ameliorated the rise in HOMA by 56%, triglycerides (TGs) by 22.5%, total cholesterol (T-Chol) by 11%, and low density lipoprotein cholesterol (LDL-C) by 21.4%. Taurine also abolished any significant difference of TAC, PON activity and NOx concentration among treated and control groups. TAC positively correlated with PON in both rats fed on the HFD and those received taurine in addition to the HFD. Fructose-fed rats showed 34.7% increase in Hcy level. Taurine administration failed to prevent the observed HHcy in the current dosage and duration. CONCLUSION Our results indicate that HFD could induce IR which could further result in metabolic syndrome (MS), and that taurine has a protective role against the metabolic abnormalities induced by this diet model except for HHcy.
Collapse
Affiliation(s)
- Hala O El Mesallamy
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Lamiaa N Hammad
- Department of Biochemistry, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Hekmat M El Magdoub
- Department of Biochemistry, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| |
Collapse
|
40
|
Homocysteine and Hypertension in Diabetes: Does PPARgamma Have a Regulatory Role? PPAR Res 2010; 2010:806538. [PMID: 20613990 PMCID: PMC2895301 DOI: 10.1155/2010/806538] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 11/11/2009] [Accepted: 05/10/2010] [Indexed: 12/31/2022] Open
Abstract
Dysfunction of macro- and microvessels is a major cause of morbidity and mortality in patients with cardio-renovascular diseases such as atherosclerosis, hypertension, and diabetes. Renal failure and impairment of renal function due to vasoconstriction of the glomerular arteriole in diabetic nephropathy leads to renal volume retention and increase in plasma homocysteine level. Homocysteine, which is a nonprotein amino acid, at elevated levels is an independent cardio-renovascular risk factor. Homocysteine induces oxidative injury of vascular endothelial cells, involved in matrix remodeling through modulation of the matrix metalloproteinase (MMP)/tissue inhibitor of metalloproteinase (TIMP) axis, and increased formation and accumulation of extracellular matrix protein, such as collagen. In heart this leads to increased endothelial-myocyte uncoupling resulting in diastolic dysfunction and hypertension. In the kidney, increased matrix accumulation in the glomerulus causes glomerulosclerosis resulting in hypofiltration, increased renal volume retention, and hypertension. PPARγ agonist reduces tissue homocysteine levels and is reported to ameliorate homocysteine-induced deleterious vascular effects in diabetes. This review, in light of current information, focuses on the beneficial effects of PPARγ agonist in homocysteine-associated hypertension and vascular remodeling in diabetes.
Collapse
|
41
|
Zhuo JM, Portugal GS, Kruger WD, Wang H, Gould TJ, Pratico D. Diet-induced hyperhomocysteinemia increases amyloid-beta formation and deposition in a mouse model of Alzheimer's disease. Curr Alzheimer Res 2010; 7:140-9. [PMID: 19939226 DOI: 10.2174/156720510790691326] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 10/25/2009] [Indexed: 11/22/2022]
Abstract
Hyperhomocysteinemia (HHcy) has been recognized as a risk factor for developing Alzheimer's disease (AD). However, its underlying molecular mechanisms are still elusive. Here we show that HHcy induces an elevation of amyloid beta (Abeta) levels and deposition, as well as behavioral impairments, in a mouse model of AD-like amyloidosis, the Tg2576 mice. This elevation is not associated with significant change of the steady state levels of the Abeta precursor protein (APP), beta- or alpha-secretase pathways, nor with the Abeta catabolic pathways. By contrast, HHcy significantly reduces glycogen synthase kinase 3 (GSK3) Ser21/9 phosphorylation, but not total GSK3 protein levels. Similar results are obtained in brains homogenates from a genetic mouse model of HHcy. In vitro studies show that homocysteine increases Abeta formation, reduces phosphorylated GSK3 levels, without changes in total APP and its metabolism, and these effects are prevented by selective GSK3 inhibition. Overall, these data support a potential link between GSK3 and the pro-amyloidotic effect of HHcy in vivo and in vitro.
Collapse
Affiliation(s)
- J-M Zhuo
- Department of Pharmacology, Neuroscience Program Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|
42
|
Yu X, Huang Y, Hu Q, Ma L. Hyperhomocysteinemia stimulates hepatic glucose output and PEPCK expression. Acta Biochim Biophys Sin (Shanghai) 2009; 41:1027-32. [PMID: 20011977 DOI: 10.1093/abbs/gmp097] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Homocysteine is an intermediate in the sulfur amino acid metabolism. Recent studies suggested that there might be links between hyperhomocysteinemia and insulin resistance. In the present study, we investigated the effect of homocysteine on glucose metabolism. We demonstrated that the levels of insulin were significantly higher in mice with hyperhomocysteinemia than those in the normal mice after administration of glucose. The effect of insulin on glucose output was significantly blocked in the homocysteine-treated hepatocytes. In addition, the expression of phosphoenolpyruvate carboxykinase (PEPCK) gene was elevated in the liver of mice with hyperhomocysteinemia and primary mouse hepatocytes treated with homocysteine. The action of homocysteine was suppressed by H89, a protein kinase A (PKA) inhibitor. Thus, hyperhomocysteinemia may be considered as a risk factor that contributes to the development of insulin resistance with respect to elevated glucose output and upregulation of PEPCK, probably via the PKA pathway. Our study provides a novel mechanistic explanation for the development of insulin resistance in hyperhomocysteinemia.
Collapse
Affiliation(s)
- Xue Yu
- Department of Cardiology, Beijing Hospital, Beijing 100730, China
| | | | | | | |
Collapse
|
43
|
Nabipour I, Ebrahimi A, Jafari SM, Vahdat K, Assadi M, Movahed A, Moradhaseli F, Obeidi N, Sanjdideh Z. The metabolic syndrome is not associated with homocysteinemia: the Persian Gulf Healthy Heart Study. J Endocrinol Invest 2009; 32:406-10. [PMID: 19794288 DOI: 10.1007/bf03346476] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND It is uncertain whether homocysteine and the metabolic syndrome or its components are related in the general population, as studies investigating the association between homocysteine levels and insulin resistance have shown conflicting results. METHODS In an ancillary study to the Persian Gulf Healthy Heart Study, a cohort study of Iranian men and women aged >or=25 yr, a random sample of 1754 subjects were evaluated for the association of plasma homocysteine levels and the metabolic syndrome using National Cholesterol Education Program (NCEP)-Adult Treatment Panel (ATP)-III criteria. Total homocysteine levels and high sensitivity C-reactive protein (CRP) were determined by enzyme-linked immunosorbent assays. RESULTS Subjects with lower HDL-cholesterol and higher blood pressure showed significantly higher homocysteine levels (p=0.001 and p<0.0001; respectively). There was no significant difference in serum levels of homocysteine between subjects with and without the metabolic syndrome. In multiple logistic regression analysis, the metabolic syndrome did not show a significant association with serum homocysteine levels after adjusting for sex, age, smoking, fruit and vegetable intake pattern, body mass index, and physical inactivity. Concurrent elevated CRP levels and the metabolic syndrome also did not show a significant association with serum homocysteine levels after adjusting for sex, age, and lifestyle cardiovascular risk factors. CONCLUSIONS There was no association between the metabolic syndrome using NCEP-ATPIII criteria and homocysteinemia in this study. These data refute the hypothesis that homocysteine levels are influenced by the metabolic syndrome, at least in general healthy population.
Collapse
Affiliation(s)
- I Nabipour
- Department of Endocrine and Metabolic Diseases, The Persian Gulf Tropical Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, I.R. Iran.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li Y, Jiang C, Xu G, Wang N, Zhu Y, Tang C, Wang X. Homocysteine upregulates resistin production from adipocytes in vivo and in vitro. Diabetes 2008; 57:817-27. [PMID: 18192543 DOI: 10.2337/db07-0617] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Homocysteine (Hcy) is epidemiologically related to insulin resistance, which has been speculated to be a low-grade systemic inflammatory condition. Resistin acts as a critical mediator of insulin resistance associated with inflammatory conditions. We aimed to determine whether Hcy can induce insulin resistance by directly regulating the expression and secretion of resistin from adipose tissue. RESEARCH DESIGN AND METHODS The effect of Hcy on the expression and secretion of resistin and insulin resistance was investigated using primary rat adipocytes and mice with hyperhomocysteinemia (HHcy). RESULTS Hcy impaired glucose transport and, particularly, the insulin signaling pathway as shown by decreased insulin-stimulated tyrosine phosphorylation of insulin receptor and insulin receptor substrate (IRS)-1, increased serine phosphorylation of IRS-1, and inhibited Akt phosphorylation both in vitro and in vivo, and these impairments were accompanied by an increase in resistin expression. Compared with normal mice, HHcy mice with a clinically relevant level of plasma Hcy (19 micromol/l) showed significantly increased resistin production from adipose tissue (33.38 +/- 3.08 vs. 19.27 +/- 1.71 ng/ml, P < 0.01). Hcy (300-1000 micromol/l) also increased mRNA expression of resistin in primary rat adipocytes in a time- and concentration-dependent manner, with maximal induction at 24 h of approximately fourfold with 1,000 micromol/l. In addition, Hcy-induced resistin expression attenuated by treatment with reactive oxygen species (ROS) scavengers, protein kinase C (PKC), and nuclear factor (NF)-kappaB inhibitors implies a role in the process for ROS, PKC, and NF-kappaB. CONCLUSIONS HHcy may promote insulin resistance through the induction of resistin expression and secretion from adipocytes via the activation of the ROS-PKC-NF-kappaB pathway.
Collapse
Affiliation(s)
- Yin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Jakubowski H. The molecular basis of homocysteine thiolactone-mediated vascular disease. Clin Chem Lab Med 2008; 45:1704-16. [PMID: 17937605 DOI: 10.1515/cclm.2007.338] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Accumulating evidence suggests that a metabolite of homocysteine (Hcy), the thioester Hcy-thiolactone, plays an important role in atherogenesis and thrombosis. Hcy-thiolactone levels are elevated in hyperhomocysteinemic humans and mice. The thioester chemistry of Hcy-thiolactone underlies its ability to form isopeptide bonds with protein lysine residues, which impairs or alters the protein's function. Protein targets for the modification by Hcy-thiolactone in human blood include fibrinogen, low-density lipoprotein, and high-density lipoprotein. Protein N-homocysteinylation leads to pathophysiological responses, including increased susceptibility to thrombogenesis caused by N-Hcy-fibrinogen, and an autoimmune response elicited by N-Hcy-proteins. Chronic activation of these responses in hyperhomocysteinemia over many years could lead to vascular disease. This article reviews recent evidence supporting the hypothesis that Hcy-thiolactone contributes to pathophysiological effects of Hcy on the vascular system.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Microbiology and Molecular Genetics, UMDNJ-New Jersey Medical School, International Center for Public Health, Newark, NJ 07101-1709, USA.
| |
Collapse
|
46
|
Golbahar J, Aminzadeh MA, Kassab SE, Omrani GR. Hyperhomocysteinemia induces insulin resistance in male Sprague-Dawley rats. Diabetes Res Clin Pract 2007; 76:1-5. [PMID: 16963146 DOI: 10.1016/j.diabres.2006.07.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2006] [Accepted: 07/28/2006] [Indexed: 11/21/2022]
Abstract
Association between elevated plasma homocysteine levels and insulin resistance has been reported, however, whether hyperhomocysteinemia induces insulin resistance or it is actually hyperinsulinemia that causes elevated plasma homocysteine levels, the direction of causality in this association is not still clear. In this study, we examined the hypothesis that hyperhomocysteinemia may cause hyperinsulinemia leading to insulin resistance in rats. Plasma glucose, insulin and total homocysteine concentrations were determined in two groups of male Sprague-Dawley rats, a test group that administered with homocysteine and a control group with no homocysteine in daily drinking water before and after 50 days. Oral glucose tolerance tests were also performed in control and test groups before and after 50 days. Mean fasting plasma insulin level was significantly higher (42.5+/-20.4 mU/L versus 23.2+/-5.9 mU/L, p=0.01), whereas mean glucose: insulin ratio was significantly lower in test rats than in control rats (0.12+/-0.07 versus 0.17+/-0.05, p=0.04) after 50 days. In addition, mean homeostasis assessment insulin resistance index was significantly higher in test rats than in control rats (7.5+/-3.5 versus 4.0+/-1.6, p=0.02) after 50 days. The mean plasma glucose level was not significantly different (4.1+/-1.1 mmol/L versus 3.9+/-0.8 mmol/L, p=0.57) between controls and test rats, however, the results from oral glucose tolerance tests showed the development of insulin resistance in test rats after 50 days administration of homocysteine. Results from this in vivo study suggest that homocysteine can cause insulin resistance and this relationship may need to be considered when evaluating the role of plasma homocysteine as a risk factor in patients with obesity and type II diabetes.
Collapse
Affiliation(s)
- Jamal Golbahar
- Al-Jawhara Center for Molecular Medicine and Gentices, College of Medicine and Medical Sciences, Arabian Gulf University, P.O. Box 22979, Manama, Bahrain.
| | | | | | | |
Collapse
|
47
|
Hajer GR, van der Graaf Y, Olijhoek JK, Verhaar MC, Visseren FLJ. Levels of homocysteine are increased in metabolic syndrome patients but are not associated with an increased cardiovascular risk, in contrast to patients without the metabolic syndrome. Heart 2007; 93:216-20. [PMID: 16952974 PMCID: PMC1861402 DOI: 10.1136/hrt.2006.093971] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/15/2006] [Indexed: 12/31/2022] Open
Abstract
AIM The metabolic syndrome is associated with increased cardiovascular risk. Elevated plasma homocysteine may cause or result from insulin resistance, and may indicate vascular risk or be actively involved in atherogenesis. The aim of the study was to investigate the relationship between homocysteine, the metabolic syndrome and the incidence of cardiovascular events in patients with manifest vascular disease. METHODS A cohort of 2169 patients with manifest vascular disease was followed for a mean period of 2.8 years. Plasma homocysteine was measured at baseline. Metabolic syndrome was defined by NCEP criteria. RESULTS Homocysteine levels were higher in metabolic syndrome patients compared to patients without the metabolic syndrome (14.9+/-0.2 v 14.1+/-0.2 micromol/l; p = 0.002) and increased with the presence of its components (from 0 to 5) (12.7 to 15.9 micromol/l; p<0.001). During follow-up, 52 strokes, 67 myocardial infarctions, 5 fatal ruptures of aortic aneurysms and 53 vascular deaths occurred. Patients without the metabolic syndrome and homocysteine levels in the highest tertile had increased risk for events (HR 1.9; 95% CI 1.0 to 3.5) compared to patients without the metabolic syndrome and homocysteine levels in the lowest tertile. The presence of the metabolic syndrome increased the risk (HR 2.2; 95% CI 1.2 to 4.2), but elevated homocysteine levels further increased the risk only marginally (2.5; 95% CI 1.4 to 4.6). CONCLUSIONS Metabolic syndrome patients have elevated homocysteine levels, but these higher levels are not associated with an increased risk for new cardiovascular events. In contrast, elevated homocysteine levels confer increased risk in patients without the metabolic syndrome.
Collapse
Affiliation(s)
- Gideon R Hajer
- Department of Internal Medicine, Section of Vascular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | | | | | |
Collapse
|
48
|
Association between homocysteine, vitamin B6 concentrations and inflammation. ACTA ACUST UNITED AC 2007; 45:1728-36. [DOI: 10.1515/cclm.2007.347] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AbstractDuring the last years, a growing body of evidence has been accumulated on the role of hyperhomocysteinemia in the occurrence of coronary artery disease and other arterial occlusive diseases. The mechanism by which high circulating homocysteine concentrations are a risk factor for atherothrombosis is incompletely understood. The present review is aimed to evaluate the role of inflammation in influencing homocysteine (Hcy) and vitamin BClin Chem Lab Med 2007;45:1728–36.
Collapse
|
49
|
Holven KB, Halvorsen B, Bjerkeli V, Damås JK, Retterstøl K, Mørkrid L, Ose L, Aukrust P, Nenseter MS. Impaired Inhibitory Effect of Interleukin-10 on the Balance Between Matrix Metalloproteinase-9 and Its Inhibitor in Mononuclear Cells From Hyperhomocysteinemic Subjects. Stroke 2006; 37:1731-6. [PMID: 16728689 DOI: 10.1161/01.str.0000226465.84561.cb] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Homocysteine has been linked to increased risk of ischemic stroke and other cardiovascular events, but the mechanism by which elevated plasma levels of homocysteine promotes atherogenesis remains unclear. Matrix degradation, partly regulated by the balance between matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs), plays an important role in atherogenesis and plaque destabilization, and we hypothesized an imbalance between MMPs and TIMPs in hyperhomocysteinemia. METHODS Serum MMP-9 and TIMP-1 was measured in 12 hyperhomocysteinemic and 12 control subjects. The release of MMP-9 and TIMP-1, with and without interleukin-10 (IL-10), and the effect of IL-10 on signal transducer and activator of transcription 3 (STAT3) phosphorylation were measured in peripheral blood mononuclear cells (PBMCs) from hyperhomocysteinemic and control subjects. RESULTS Our main findings were: (1) hyperhomocysteinemic subjects had raised serum levels of MMP-9 and MMP-9/TIMP-1 ratio comparing healthy controls; (2) although IL-10 markedly suppressed MMP-9 release from PBMCs in controls, no or only minor effect was seen in hyperhomocysteinemic subjects; (3) although IL-10 enhanced TIMP-1 levels in PBMCs from both hyperhomocysteinemic and control subjects, the increase was more prominent in controls, resulting in a marked difference in IL-10-induced changes in MMP-9/TIMP-1 ratio between these 2 groups; and (4) comparing PBMCs from controls, cells from hyperhomocysteinemic individuals had impaired IL-10-induced STAT3 phosphorylation. CONCLUSIONS Our findings suggest an attenuated inhibitory response to IL-10 on MMP-9 activity in hyperhomocysteinemic subjects, potentially promoting atherogenesis and plaque instability, representing a novel explanation for increased risk for atherosclerotic disease in these individuals.
Collapse
|
50
|
Abstract
Elevated level of the nonprotein amino acid homocysteine (Hcy) is a risk factor for cardiovascular diseases, neurodegenerative diseases, and neural tube defects. However, it is not clear why excess Hcy is harmful. To explain Hcy toxicity, the "Hcy-thiolactone hypothesis" has been proposed. According to this hypothesis, metabolic conversion of Hcy to a chemically reactive metabolite, Hcy-thiolactone, catalyzed by methionyl-tRNA synthetase is the first step in a pathway that contributes to Hcy toxicity in humans. Plasma Hcy-thiolactone levels are elevated in human subjects with hyperhomocysteinemia caused by mutations in CBS or MTHFR genes. Plasma and urinary Hcy-thiolactone levels are also elevated in mice fed a high-methionine diet. Hcy-thiolactone can be detrimental because of its intrinsic ability to form N-Hcy-protein adducts, in which a carboxyl group of Hcy is N-linked to epsilon-amino group of a protein lysine residue. This article reviews recent studies of Hcy-thiolactone and N-Hcy-protein in the human body, including their roles in autoimmune response, cellular toxicity, and atherosclerosis. Potential utility of Hcy-thiolactone, N-Hcy-protein, or anti-N-Hcy-protein autoantibodies as markers of Hcy excess is discussed.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Microbiology & Molecular Genetics, UMDNJ-New Jersey Medical School, International Center for Public Health, Newark, NJ 07101, USA.
| |
Collapse
|