51
|
Folate deficiency promotes differentiation of vascular smooth muscle cells without affecting the methylation status of regulated genes. Biochem J 2020; 476:2769-2795. [PMID: 31530711 DOI: 10.1042/bcj20190275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 01/10/2023]
Abstract
Elevated serum homocysteine, an intermediate of cellular one-carbon metabolism, is an independent risk factor for cardiovascular disease (CVD). Folate deficiency increases serum homocysteine and may contribute to CVD progression. Vascular smooth muscle cells (VSMCs) regulate vascular contractility, but also contribute to repair processes in response to vascular injury. Nutritional deficiencies, like folate deficiency, are thought to impact on this phenotypic plasticity, possibly by epigenetic mechanisms. We have investigated the effect of folate deficiency on VSMCs in two cell culture systems representing early and late stages of smooth muscle cells differentiation. We find that folate deficiency promotes differentiation towards a more contractile phenotype as indicated by increased expression of respective marker genes. However, microarray analysis identified markers of striated muscle as the predominant gene expression change elicited by folate deficiency. These changes are not merely a reflection of cell cycle arrest, as foetal calf serum restriction or iron deficiency do not replicate the gene expression changes observed in response to folate deficiency. Folate deficiency only has a marginal effect on global DNA methylation. DNA methylation of CpG islands associated with genes regulated by folate deficiency remains unaffected. This supports our earlier findings in a mouse model system which also did not show any changes in global DNA methylation in response to folate and vitamin B6/B12 deficiency. These data suggest that folate deficiency enhances the expression of smooth muscle marker gene expression, promotes a shift towards a skeletal muscle phenotype, and does not regulate gene expression via DNA methylation.
Collapse
|
52
|
Toya T, Sara JD, Lerman B, Ahmad A, Taher R, Godo S, Corban MT, Lerman LO, Lerman A. Elevated plasma homocysteine levels are associated with impaired peripheral microvascular vasomotor response. IJC HEART & VASCULATURE 2020; 28:100515. [PMID: 32322661 PMCID: PMC7171522 DOI: 10.1016/j.ijcha.2020.100515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/23/2022]
Abstract
Homocysteine > 10 μmol/L is associated with peripheral microvascular endothelial dysfunction (PMED). Homocysteine > 10 μmol/L was associated with PMED in older, obese, or hypertensive patients. The association of homocysteine and PMED was prominent in patients with B-vitamins. Homocysteine > 10 μmol/L was associated with higher major cardiovascular events in univariate analysis.
Background Hyperhomocysteinemia (HHcy) has been proposed as an important cardiovascular risk factor (cRF). However, little is known about the association between plasma homocysteine levels and peripheral microvascular endothelial dysfunction (PMED), which is an integrated index of vascular health. Methods This cross-sectional and retrospective cohort study included patients who underwent non-invasive PMED assessment using reactive hyperemia peripheral arterial tonometry (RH-PAT). The association between HHcy and PMED, and its impact on MACE (all-cause mortality and atherosclerotic cardiovascular events) was investigated. Results A total of 257 patients were enrolled (HHcy > 10.0 µmol/L, N = 51; lower levels of homocysteine [LHcy] ≤ 10 µmol/L, N = 206). Patients with HHcy were older, predominantly males, and with more comorbidities than patients with LHcy (p < 0.05 for all). RH-PAT index was lower in patients with HHcy versus LHcy (p = 0.01). A significant association between HHcy and PMED was observed in older (≥60 years), obese (≥30 kg/m2), present/past smokers and hypertensive patients. HHcy was significantly associated with PMED even after adjusting for other cRF and B-vitamins supplementation. HHcy was associated with an increased risk of MACE with a hazard ratio of 3.65 (95% CI 1.41–9.48, p = 0.01) and an adjusted hazard ratio of 2.44 (95% CI 0.91–6.51, p = 0.08) after adjustment for age (≥60 years). Conclusion HHcy was independently associated with PMED after adjusting for cRF and B-vitamins supplementation. Thus, the link between homocysteine and MACE could be mediated by endothelial dysfunction, and will require further clarification with future studies.
Collapse
Affiliation(s)
- Takumi Toya
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA.,Division of Cardiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Jaskanwal D Sara
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Ben Lerman
- School of Medicine, St. George's University, St George's, West Indies, Grenada
| | - Ali Ahmad
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Riad Taher
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Shigeo Godo
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Michel T Corban
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| |
Collapse
|
53
|
Yang Z, Shi J, He Z, Lü Y, Xu Q, Ye C, Chen S, Tang B, Yin K, Lu Y, Chen X. Predictors for imaging progression on chest CT from coronavirus disease 2019 (COVID-19) patients. Aging (Albany NY) 2020; 12:6037-6048. [PMID: 32275643 PMCID: PMC7185104 DOI: 10.18632/aging.102999] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/28/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study aimed to investigate the potential parameters associated with imaging progression on chest CT from coronavirus disease 19 (COVID-19) patients. RESULTS The average age of 273 COVID-19 patients enrolled with imaging progression were older than those without imaging progression (p = 0.006). The white blood cells, platelets, neutrophils and acid glycoprotein were all decreased in imaging progression patients (all p < 0.05), and monocytes were increased (p = 0.025). The parameters including homocysteine, urea, creatinine and serum cystatin C were significantly higher in imaging progression patients (all p < 0.05), while eGFR decreased (p < 0.001). Monocyte-lymphocyte ratio (MLR) was significantly higher in imaging progression patients compared to that in imaging progression-free ones (p < 0.001). Logistic models revealed that age, MLR, homocysteine and period from onset to admission were factors for predicting imaging progression on chest CT at first week from COVID-19 patients (all p < 0.05). CONCLUSION Age, MLR, homocysteine and period from onset to admission could predict imaging progression on chest CT from COVID-19 patients. METHODS The primary outcome was imaging progression on chest CT. Baseline parameters were collected at the first day of admission. Imaging manifestations on chest CT were followed-up at (6±1) days.
Collapse
Affiliation(s)
- Zongguo Yang
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Jia Shi
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zhang He
- Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Ying Lü
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Qingnian Xu
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Chen Ye
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Shishi Chen
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Bozong Tang
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Keshan Yin
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Yunfei Lu
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaorong Chen
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| |
Collapse
|
54
|
Mechanisms of homocysteine-induced damage to the endothelial, medial and adventitial layers of the arterial wall. Biochimie 2020; 173:100-106. [PMID: 32105811 DOI: 10.1016/j.biochi.2020.02.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/20/2020] [Indexed: 11/23/2022]
Abstract
Homocysteine (Hcy) is a non-protein forming amino acid which is the direct metabolic precursor of methionine. Increased concentration of serum Hcy is considered a risk factor for cardiovascular disease and is specifically linked to various diseases of the vasculature. Serum Hcy is associated with atherosclerosis, hypertension and aneurysms of the aorta in humans, though the precise mechanisms by which Hcy contributes to these conditions remain elusive. Results from clinical trials that successfully lowered serum Hcy without reducing features of vascular disease in cardiovascular patients have cast doubt on whether or not Hcy directly impacts the vasculature. However, studies in animals and in cell culture suggest that Hcy has a vast array of toxic effects on the vasculature, with demonstrated roles in endothelial dysfunction, medial remodeling and adventitial inflammation. It is hypothesized that rather than serum Hcy, tissue-bound Hcy and the incorporation of Hcy into proteins could underlie the toxic effects of Hcy on the vasculature. In this review, we present evidence for Hcy-associated vascular disease in humans, and we critically examine the possible mechanisms by which Hcy specifically impacts the endothelial, medial and adventitial layers of the arterial wall. Deciphering the mechanisms by which Hcy interacts with proteins in the arterial wall will allow for a better understanding of the pathomechanisms of hyperhomocysteinemia and will help to define a better means of prevention at the appropriate window of life.
Collapse
|
55
|
Zhang YM, Zhou XJ, Shi SF, Liu LJ, Lyu JC, Zhang H. Homocysteine and IgA nephropathy: observational and Mendelian randomization analyses. Chin Med J (Engl) 2020; 133:277-284. [PMID: 31929371 PMCID: PMC7004620 DOI: 10.1097/cm9.0000000000000613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND High levels of plasma homocysteine occur almost uniformly in patients with end-stage renal disease (ESRD). IgA nephropathy (IgAN) is the most common form of primary glomerulonephritis and a common cause of ESRD in young adults. Here, we aimed to detect whether homocysteine was elevated and associated with clinical-pathologic manifestations of IgAN patients and tested its causal effects using a two-sample Mendelian randomization (MR) approach. METHODS For observational analysis, 108 IgAN patients, 30 lupus nephritis (LN) patients, 50 minimal change disease (MCD) patients, and 206 healthy controls were recruited from April 2014 to April 2015. Their plasma homocysteine was measured and clinical-pathologic manifestations were collected from medical records. For MR analysis, we further included 1686 IgAN patients. The missense variant methylenetetrahydrofolate reductase C677T (rs1801133) was selected as an instrument, which was genotyped by TaqMan allele discrimination assays. RESULTS Majority of IgAN patients (93.52%, 101/108) showed elevated levels of plasma homocysteine (>10 μmol/L). Plasma homocysteine in IgAN patients was significantly higher than that in MCD patients (median: 18.32 vs. 11.15 μmol/L, Z = -5.29, P < 0.01) and in healthy controls (median: 18.32 vs. 10.00 μmol/L, Z = -8.76, P < 0.01), but comparable with those in LN patients (median: 18.32 L vs. 14.50 μmol/L, Z = -1.32, P = 0.19). Significant differences were observed in sub-groups of IgAN patients according to quartiles of plasma homocysteine for male ratio (22.22% vs. 51.85% vs. 70.37% vs. 70.37%, χ = 14.29, P < 0.01), serum creatinine (median: 77.00 vs. 100.00 vs. 129.00 vs. 150.00 μmol/L, χ = 34.06, P < 0.01), estimated glomerular filtration rate (median: 100.52 vs. 74.23 vs. 52.68 vs. 42.67 mL·min·1.73 m, χ = 21.75, P < 0.01), systolic blood pressure (median: 120.00 vs. 120.00 vs. 125.00 vs. 130.00 mmHg, χ = 2.97, P = 0.05), diastolic blood pressure (median 80.00 vs. 75.00 vs. 80.00 vs. 81.00 mmHg, χ = 11.47, P < 0.01), and pathologic tubular atrophy and interstitial fibrosis (T) (T0/T1/T2: 62.96%/33.33%/3.70% vs. 29.63%/40.74%/29.63% vs. 24.00%/48.00%/28.00% vs. 14.81%/37.04%/48.15%, χ = 17.66, P < 0.01). The coefficient of each rs1801133-T allele on homocysteine levels after controlling age and sex was 7.12 (P < 0.01). MR estimates showed causal positive effects of homocysteine on serum creatine (β = 0.76, P = 0.02), systolic blood pressure (β = 0.26, P = 0.02), diastolic blood pressure (β = 0.20, P = 0.01), and pathologic T lesion (β = 0.01, P = 0.01) in IgAN. CONCLUSIONS By observational and MR analyses, consistent results were observed for associations of plasma homocysteine with serum creatinine, blood pressures, and pathologic T lesion in IgAN patients.
Collapse
Affiliation(s)
- Yue-Miao Zhang
- Renal Division, Department of Medicine, Peking University First Hospital; Institute of Nephrology, Peking University; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing 100034, China
| | | | | | | | | | | |
Collapse
|
56
|
Salissou MTM, Mahaman YAR, Zhu F, Huang F, Wang Y, Xu Z, Ke D, Wang Q, Liu R, Wang JZ, Zhang B, Wang X. Methanolic extract of Tamarix Gallica attenuates hyperhomocysteinemia induced AD-like pathology and cognitive impairments in rats. Aging (Albany NY) 2019; 10:3229-3248. [PMID: 30425189 PMCID: PMC6286848 DOI: 10.18632/aging.101627] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/27/2018] [Indexed: 12/20/2022]
Abstract
Although few drugs are available today for the management of Alzheimer’s disease (AD) and many plants and their extracts are extensively employed in animals’ studies and AD patients, yet no drug or plant extract is able to reverse AD symptoms adequately. In the present study, Tamarix gallica (TG), a naturally occurring plant known for its strong antioxidative, anti-inflammatory and anti-amyloidogenic properties, was evaluated on homocysteine (Hcy) induced AD-like pathology and cognitive impairments in rats. We found that TG attenuated Hcy-induced oxidative stress and memory deficits. TG also improved neurodegeneration and neuroinflammation by upregulating synaptic proteins such as PSD95 and synapsin 1 and downregulating inflammatory markers including CD68 and GFAP with concomitant decrease in proinflammatory mediators interlukin-1β (IL1β) and tumor necrosis factor α (TNFα). TG attenuated tau hyperphosphorylation at multiple AD-related sites through decreasing some kinases and increasing phosphatase activities. Moreover, TG rescued amyloid-β (Aβ) pathology through downregulating BACE1. Our data for the first time provide evidence that TG attenuates Hcy-induced AD-like pathological changes and cognitive impairments, making TG a promising candidate for the treatment of AD-associated pathological changes.
Collapse
Affiliation(s)
- Maibouge Tanko Mahamane Salissou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yacoubou Abdoul Razak Mahaman
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518001, Guangdong Province, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518001, Guangdong Province, China
| | - Fang Huang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuman Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhendong Xu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, 226001, China
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, 226001, China
| |
Collapse
|
57
|
Choi JW, Chong S, Phi JH, Lee JY, Kim HS, Chae JH, Lee J, Kim SK. Postoperative Symptomatic Cerebral Infarction in Pediatric Moyamoya Disease: Risk Factors and Clinical Outcome. World Neurosurg 2019; 136:e158-e164. [PMID: 31870818 DOI: 10.1016/j.wneu.2019.12.072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Indirect bypass surgery is used to improve the hemodynamic status of pediatric patients with moyamoya disease (MMD). Symptomatic cerebral infarction during the early postoperative period may be the most frustrating complication. This study was conducted to investigate the factors associated with early postoperative symptomatic cerebral infarction. METHODS Between January 2000 and February 2014, we performed 1241 indirect bypass surgeries in 659 pediatric MMD patients. Symptomatic infarction during the early postoperative period was diagnosed in 63 operations in 61 patients. RESULTS The overall incidence of symptomatic cerebral infarction after indirect bypass surgery was 5.1%. The median age of the patients with postoperative infarction was 6 years (mean, 6.4 years; range, 1-15 years). The performance of 2 craniotomies in single operation was associated with a higher rate of cerebral infarction. Moreover, the incidence was higher in young patients (age <6 years) compared with older patients. In a matched analysis, an immediate postoperative hemoglobin level >13 g/dL was associated with decreased risk of infarction (odds ratio, 0.144; P = 0.003). Mutation of the methylenetetrahydrofolate reductase (MTHFR) gene occurred in a relatively high proportion of our infarction cohort. CONCLUSIONS Postoperative symptomatic infarctions can occur despite a unified surgical method and formulaic perioperative management protocol. Patient-centered factors, such as young age, genetic background of MTHFR, and certain medical conditions, including hyperthyroidism, renovascular hypertension, and hemolytic uremic syndrome, as well as management-related factors, including 2 craniotomies and low immediate postoperative hemoglobin level, could be risk factors for early postoperative symptomatic cerebral infarction.
Collapse
Affiliation(s)
- Jung Won Choi
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sangjoon Chong
- Department of Neurosurgery, Asan Medical Center, Seoul, Republic of Korea
| | - Ji Hoon Phi
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Yeoun Lee
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hee-Soo Kim
- Division of Pediatric Anesthesiology and Pain Medicine, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong Hee Chae
- Division of Pediatric Neurology, Department of Pediatrics, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joongyub Lee
- Division of Clinical Epidemiology, Medical Research Collaborating Center, Biomedical Research Institution, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
58
|
Fan X, Wang E, He J, Zhang L, Zeng X, Gui Y, Sun Q, Song Y, Yuan H. Ligustrazine Protects Homocysteine-Induced Apoptosis in Human Umbilical Vein Endothelial Cells by Modulating Mitochondrial Dysfunction. J Cardiovasc Transl Res 2019. [PMID: 31359360 DOI: 10.1007/s12265-019-09900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Ligustrazine is one of the alkaloid compounds isolated from the traditional Chinese herb, which shows protective effects on cardiovascular disorders. High homocysteine (Hcy) level can predict cardiovascular-related events including death. In this study, we used Hcy to stimulate the human umbilical vein endothelial cells (HUVECs) and investigated the protective effect of ligustrazine on endothelial dysfunction by assessing the cell apoptosis, oxidative damage, mitochondrial dysfunction, and the potential molecular pathways. Our results clearly showed that ligustrazine increased HUVEC cell viability, decreased the dehydrogenase (LDH) level, and inhibited HUVEC apoptosis, which was associated with the attenuation of attenuated oxidative damage. The mitochondrial-dependent pathway was closely related in the regulation of ligustrazine, reflected by the attenuated mitochondrial membrane potential change and decreased cytochrome c release from the mitochondria to the cytosol. Ligustrazine may protect Hcy-induced apoptosis in HUVECs by attenuating oxidative damage and modulating mitochondrial dysfunction.
Collapse
Affiliation(s)
- Xuesong Fan
- Department of Clinical Laboratory Center, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Enshi Wang
- Center for Pediatric Cardiac Surgery, National Center for Cardiovascular Diseases and Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jianxun He
- Department of Clinical Laboratory Center, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Lei Zhang
- Department of Clinical Laboratory Center, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Xiaoli Zeng
- Department of Clinical Laboratory Center, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yuan Gui
- Department of Clinical Laboratory Center, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Qi Sun
- Department of Clinical Laboratory Center, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yang Song
- Department of Clinical Laboratory Center, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Hui Yuan
- Department of Clinical Laboratory Center, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China.
| |
Collapse
|
59
|
Ligustrazine Protects Homocysteine-Induced Apoptosis in Human Umbilical Vein Endothelial Cells by Modulating Mitochondrial Dysfunction. J Cardiovasc Transl Res 2019; 12:591-599. [PMID: 31359360 DOI: 10.1007/s12265-019-09900-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/03/2019] [Indexed: 01/06/2023]
Abstract
Ligustrazine is one of the alkaloid compounds isolated from the traditional Chinese herb, which shows protective effects on cardiovascular disorders. High homocysteine (Hcy) level can predict cardiovascular-related events including death. In this study, we used Hcy to stimulate the human umbilical vein endothelial cells (HUVECs) and investigated the protective effect of ligustrazine on endothelial dysfunction by assessing the cell apoptosis, oxidative damage, mitochondrial dysfunction, and the potential molecular pathways. Our results clearly showed that ligustrazine increased HUVEC cell viability, decreased the dehydrogenase (LDH) level, and inhibited HUVEC apoptosis, which was associated with the attenuation of attenuated oxidative damage. The mitochondrial-dependent pathway was closely related in the regulation of ligustrazine, reflected by the attenuated mitochondrial membrane potential change and decreased cytochrome c release from the mitochondria to the cytosol. Ligustrazine may protect Hcy-induced apoptosis in HUVECs by attenuating oxidative damage and modulating mitochondrial dysfunction.
Collapse
|
60
|
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: 172] [Impact Index Per Article: 34.4] [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
|
61
|
Ma Y, Zhang Z, Chen R, Shi R, Zeng P, Chen R, Leng Y, Chen AF. NRP1 regulates HMGB1 in vascular endothelial cells under high homocysteine condition. Am J Physiol Heart Circ Physiol 2019; 316:H1039-H1046. [PMID: 30767669 DOI: 10.1152/ajpheart.00746.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endothelial inflammation plays an important role in hyperhomocysteinemia (HHcy)-associated vascular diseases. High mobility group box 1 (HMGB1) is a pro-inflammatory danger molecule produced by endothelial cells. However, whether HMGB1 is involved in vascular endothelial inflammation of HHcy is poorly understood. Neuropilin-1 (NRP1) mediates inflammatory response and activates mitogen-activated protein kinases (MAPKs) pathway that has been reported to be involved in regulation of HMGB1. The aim of this study was to determine the alteration of HMGB1 in HHcy, and the role of NRP1 in regulation of endothelial HMGB1 under high homocysteine (Hcy) condition. In the present study, we first observed that the plasma level of HMGB1 was elevated in HHcy patients and an experimental rat model, and increased HMGB1 was also observed in the thoracic aorta of an HHcy rat model. HMGB1 was induced by Hcy accompanied with upregulated NRP1 in vascular endothelial cells. Overexpression of NRP1 promoted expression and secretion of HMGB1 and endothelial inflammation; knockdown of NRP1 inhibited HMGB1 and endothelial inflammation induced by Hcy, which partially regulated through p38 MAPK pathway. Furthermore, NRP1 inhibitor ATWLPPR reduced plasma HMGB1 level and expression of HMGB1 in the thoracic aorta of HHcy rats. In conclusion, our data suggested that Hcy requires NRP1 to regulate expression and secretion of HMGB1. The present study provides the evidence for inhibition of NRP1 and HMGB1 to be the novel therapeutic targets of vascular endothelial inflammation in HHcy in the future. NEW & NOTEWORTHY This study shows for the first time to our knowledge that the plasma level of high mobility group box 1 (HMGB1) is elevated in hyperhomocysteinemia (HHcy) patients, and homocysteine promotes expression and secretion of HMGB1 partially regulated by neuropilin-1 in endothelial cells, which is involved in endothelial inflammation. Most importantly, these new findings will provide a potential therapeutic strategy for vascular endothelial inflammation in HHcy.
Collapse
Affiliation(s)
- Yeshuo Ma
- Department of Cardiology, The Third Xiangya Hospital of Central South University , Changsha , China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Zhen Zhang
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China.,Centre for Experimental Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Runtai Chen
- Department of Cardiology, The Third Xiangya Hospital of Central South University , Changsha , China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Rui Shi
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China.,Xiangya School of Pharmaceutical Sciences, Central South University , Changsha , China
| | - Pingyu Zeng
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China.,Centre for Experimental Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Ruifang Chen
- Department of Cardiology, The Third Xiangya Hospital of Central South University , Changsha , China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Yiping Leng
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Alex F Chen
- Department of Cardiology, The Third Xiangya Hospital of Central South University , Changsha , China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| |
Collapse
|
62
|
Tan B, Venketasubramanian N, Vrooman H, Cheng CY, Wong TY, Ikram MK, Chen C, Hilal S. Homocysteine and Cerebral Atrophy: The Epidemiology of Dementia in Singapore Study. J Alzheimers Dis 2019; 62:877-885. [PMID: 29480177 DOI: 10.3233/jad-170796] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Plasma homocysteine levels are increasingly studied as a potential risk factor for dementia. Elevated homocysteine levels have been linked with gray and white matter volume reduction among individuals with mild cognitive impairment and Alzheimer's disease. However, the effects of homocysteine on brain changes in preclinical stages of dementia remain unexplored. OBJECTIVE To examine the association of elevated homocysteine levels with markers of neurodegeneration, i.e., white and gray matter volume in an elderly population. METHODS The study included 768 participants (mean age: 69.6±6.5 years, 51.3% women) from the Epidemiology of Dementia In Singapore study. Participants underwent a brain MRI scan and blood tests. Serum homocysteine was measured using competitive immunoassay. Cortical thickness and subcortical structural volume were quantified using FreeSurfer whereas white matter volume was quantified using a previous validated method. RESULTS Higher homocysteine levels were significantly associated with decreased global white matter volume [mean difference (β) in volume (ml) per micromole per liter (μmol/l) increase in homocysteine levels: - 0.555, 95% Confidence Interval (CI): - 0.873; - 0.237], decreased parietal cortical thickness [β in thickness (μm) per μmol/l increase in homocysteine levels:- 1.429, 95% CI: - 2.781; - 0.077], and smaller volumes of the thalamus [β: - 0.017, 95% CI: - 0.026; - 0.008], brainstem [β: - 0.037, 95% CI: - 0.058; - 0.016], and accumbens [β: - 0.004, 95% CI: - 0.006; - 0.002]. CONCLUSION Higher homocysteine levels were associated with cerebral atrophy. Further studies are required to assess whether lowering plasma homocysteine levels may prevent neurodegenerative changes or delay progression of clinical symptoms before the development of dementia.
Collapse
Affiliation(s)
- Bryce Tan
- Memory Ageing and Cognition Center (MACC), National University Health System, Singapore
| | | | - Henri Vrooman
- Departments of Radiology and Medical Informatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore.,Academic Medicine Research Institute, Duke-NUS Graduate Medical School, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore.,Academic Medicine Research Institute, Duke-NUS Graduate Medical School, Singapore
| | | | - Christopher Chen
- Memory Ageing and Cognition Center (MACC), National University Health System, Singapore.,Department of Pharmacology, National University of Singapore, Singapore
| | - Saima Hilal
- Memory Ageing and Cognition Center (MACC), National University Health System, Singapore.,Department of Pharmacology, National University of Singapore, Singapore
| |
Collapse
|
63
|
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: 154] [Impact Index Per Article: 30.8] [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
|
64
|
Nam KW, Kwon HM, Jeong HY, Park JH, Kwon H, Jeong SM. Serum homocysteine level is related to cerebral small vessel disease in a healthy population. Neurology 2019; 92:e317-e325. [PMID: 30602466 DOI: 10.1212/wnl.0000000000006816] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/27/2018] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To evaluate the relationship between serum total homocysteine (tHcy) levels and cerebral small vessel disease (cSVD) in a healthy population. METHODS We included consecutive participants who visited our department for health checkups between 2006 and 2013. We rated white matter hyperintensity volumes using both the Fazekas score and semiautomated quantitative methods. We also evaluated lacunes, cerebral microbleeds, and enlarged perivascular spaces (EPVS), which are involved in cSVD. To assess the dose-dependent relationship between tHcy and cSVD parameters, we scored the burdens of each radiologic marker of cSVD. RESULTS A total of 1,578 participants were included (age 55 ± 8 years, male sex 57%). In the multivariable analysis, tHcy remained an independent predictor of the white matter hyperintensity volume (B = 0.209; 95% confidence interval [CI] = 0.033-0.385, p = 0.020), presence of cerebral microbleeds (adjusted odds ratio = 2.800; 95% CI = 1.104-7.105, p = 0.030), and moderate to severe EPVS (adjusted odds ratio = 5.906; 95% CI = 3.523-9.901, p < 0.001) after adjusting for confounders. Furthermore, tHcy had positive associations with periventricular Fazekas score (p = 0.001, p for trend <0.001), subcortical Fazekas score (p = 0.003, p for trend = 0.005), and moderate to severe EPVS lesion burden (p < 0.001, p for trend <0.001) in a dose-dependent manner. CONCLUSIONS Serum tHcy level is correlated with cSVD development in a dose-dependent manner. These findings provide us with clues for further studies of the pathophysiology of cSVD.
Collapse
Affiliation(s)
- Ki-Woong Nam
- From the Departments of Neurology (K.-W.N., H.-Y.J.) and Family Medicine (J.-H.P., H.K., S.-M.J.), Seoul National University College of Medicine and Seoul National University; and Department of Neurology (H.-M.K.), Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Hyung-Min Kwon
- From the Departments of Neurology (K.-W.N., H.-Y.J.) and Family Medicine (J.-H.P., H.K., S.-M.J.), Seoul National University College of Medicine and Seoul National University; and Department of Neurology (H.-M.K.), Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea.
| | - Han-Yeong Jeong
- From the Departments of Neurology (K.-W.N., H.-Y.J.) and Family Medicine (J.-H.P., H.K., S.-M.J.), Seoul National University College of Medicine and Seoul National University; and Department of Neurology (H.-M.K.), Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Jin-Ho Park
- From the Departments of Neurology (K.-W.N., H.-Y.J.) and Family Medicine (J.-H.P., H.K., S.-M.J.), Seoul National University College of Medicine and Seoul National University; and Department of Neurology (H.-M.K.), Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea.
| | - Hyuktae Kwon
- From the Departments of Neurology (K.-W.N., H.-Y.J.) and Family Medicine (J.-H.P., H.K., S.-M.J.), Seoul National University College of Medicine and Seoul National University; and Department of Neurology (H.-M.K.), Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Su-Min Jeong
- From the Departments of Neurology (K.-W.N., H.-Y.J.) and Family Medicine (J.-H.P., H.K., S.-M.J.), Seoul National University College of Medicine and Seoul National University; and Department of Neurology (H.-M.K.), Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| |
Collapse
|
65
|
Jana L, Maity PP, Perveen H, Dash M, Jana S, Dey A, De SK, Chattopadhyay S. Attenuation of utero-toxicity, metabolic dysfunction and inflammation by soy protein concentrate in rats exposed to fluoridated water: consequence of hyperlipidemia in parallel with hypohomocysteinemia. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:36462-36473. [PMID: 30374712 DOI: 10.1007/s11356-018-3542-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/19/2018] [Indexed: 06/08/2023]
Abstract
Lipid peroxidation and ROS generation are the pathogenesis of chronic fluoride toxicity, and its detrimental effects on human reproduction are noted drastically. The aim of the present study was to elucidate the defensive effects of soy protein concentrate (SPC) against sodium fluoride (NaF)-induced uterine dysfunction at biochemical and histological level. Rats were randomly distributed into four groups as control, NaF-treated (200 ppm), and SPC co-administered groups (20 mg and 40 mg/ 100 g body weight) for 16 days. SPC reversed the toxic effects of NaF. SPC significantly ameliorated the NaF-induced alterations of the antioxidant system in the uterus by decreasing lipid peroxidation products and by increasing antioxidant activities. SPC significantly counteracted the adverse effects of NaF on serum level of lactate dehydrogenase (LDH) and inflammatory markers Interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α) and nuclear factor kappa-B (NF-κB). Our results also explored that lipid profile was meaningfully altered due to NaF and also focused a diminution of circulating homocysteine (Hcy) and altered lipid profiles along with a diminished quantity of serum B12 and B9. However, both the doses of SPC reverted back serum levels of B12, B9, and Hcy status in similar fashion along with its corrective action on lipid profile. NaF-treated group exhibited a marked degree of reduction in the weights of ovary and uterus with an alteration of normal tissue histology and significant diminution in serum estradiol (ES) levels without fluctuating uterine estradiol receptor-α (ER-α). However, SPC restored the normal tissue histoarchitecture and also increased the functional efficiency and expression of the ER-α receptor by overturning the ES levels in NaF-treated rats. Moreover, both the doses of SPC were effective against NaF-induced alterations, although 40 mg SPC/100 g body weight had better efficacy in ameliorating the NaF-induced adverse effects on the uterus and ovary.
Collapse
Affiliation(s)
- Lipirani Jana
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Pikash Pratim Maity
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Hasina Perveen
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Moumita Dash
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Suryashis Jana
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Arindam Dey
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Subrata Kumar De
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Sandip Chattopadhyay
- Department of Biomedical Laboratory Science and Management, and Clinical Nutrition and Dietetics division (UGC Innovative Department), Vidyasagar University, Midnapore, West Bengal, 721102, India.
| |
Collapse
|
66
|
Wang Y, Hong Y, Zhang C, Shen Y, Pan YS, Chen RZ, Zhang Q, Chen YH. Picroside II attenuates hyperhomocysteinemia-induced endothelial injury by reducing inflammation, oxidative stress and cell apoptosis. J Cell Mol Med 2018; 23:464-475. [PMID: 30394648 PMCID: PMC6307770 DOI: 10.1111/jcmm.13949] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 09/03/2018] [Indexed: 12/27/2022] Open
Abstract
Picroside II (P‐II), one of the main active components of scrophularia extract, which have anti‐oxidative, anti‐inflammatory effects, but its effect on hyperhomocysteinemia (HHcy) induced endothelial injury remains to be determined. Here, we test whether P‐II protects HHcy‐induced endothelial dysfunction against oxidative stress, inflammation and cell apoptosis. In vitro study using HUVECs, and in hyperhomocysteinemia mouse models, we found that HHcy decreased endothelial SIRT1 expression and increased LOX‐1 expression, subsequently causing reactive oxygen species generation, up‐regulation of NADPH oxidase activity and NF‐κB activation, thereby promoting pro‐inflammatory response and cell apoptosis. Blockade of Sirt1 with Ex527 or siRNASIRT1 increased LOX‐1 expression, whereas overexpression of SIRT1 decreased LOX‐1 expression markedly. P‐II treatment significantly increased SIRT1 expression and reduced LOX‐1 expression, and protected against endothelial cells from Hcy‐induced oxidative injury, inflammation and apoptosis. However, blockade of SIRT1 or overexpression of LOX‐1 attenuated the therapeutic effects of P‐II. In conclusion, our results suggest that P‐II prevents the Hcy induced endothelial damage probably through regulating the SIRT1/LOX‐1 signaling pathway.
Collapse
Affiliation(s)
- Yunkai Wang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yajun Hong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chunyu Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunli Shen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ye Shen Pan
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rui Zhen Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Han Chen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
67
|
Kumar M, Goudihalli S, Mukherjee K, Dhandapani S, Sandhir R. Methylenetetrahydrofolate reductase C677T variant and hyperhomocysteinemia in subarachnoid hemorrhage patients from India. Metab Brain Dis 2018; 33:1617-1624. [PMID: 29926428 DOI: 10.1007/s11011-018-0268-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022]
Abstract
Methylenetetrahydrofolate reductase (MTHFR) polymorphism (C677T, A1298C) has been implicated in increased plasma homocysteine (Hcy) levels. The present study was designed to investigate the association between MTHFR polymorphism and increased Hcy levels in subarachnoid haemorrhage (SAH) patients. A total of 150 subjects from North India were included in the study, comprising of 100 SAH patients and 50 healthy controls. Plasma Hcy levels was determined and MTHFR polymorphism (C677T, A1298C) was screened by High resolution melting (HRM) analysis. Plasma Hcy levels were found to be significantly higher (p < 0.001) in SAH patients than in healthy controls. No significant difference in the genotype and allele frequency of MTHFR A1298C was observed. However, frequency of MTHFR C677T genotype, CT (53% vs. 20%; p < 0.001) and TT (15% vs. 2%; p < 0.05) was significantly higher in SAH group as compared to healthy controls. The frequency of T allele (41.5% vs. 12%; p < 0.001) was also found to be higher in SAH patients in comparison to healthy controls. Furthermore, Hcy levels were higher in SAH patients with TT genotype than in patients having CT genotype, whereas CC genotype had lower Hcy levels. The study suggests that higher frequency of MTHFR C677T allele may contribute to etiopathology of SAH through increase in Hcy levels.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Biochemistry, Basic Medical Sciences Block-II, Panjab University, Sector-25, Chandigarh, 160014, India
| | - Sachin Goudihalli
- Department of Neurosurgery, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Kanchan Mukherjee
- Department of Neurosurgery, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Sivashanmugam Dhandapani
- Department of Neurosurgery, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Rajat Sandhir
- Department of Biochemistry, Basic Medical Sciences Block-II, Panjab University, Sector-25, Chandigarh, 160014, India.
| |
Collapse
|
68
|
Lekawanvijit S. Cardiotoxicity of Uremic Toxins: A Driver of Cardiorenal Syndrome. Toxins (Basel) 2018; 10:toxins10090352. [PMID: 30200452 PMCID: PMC6162485 DOI: 10.3390/toxins10090352] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 08/19/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is highly prevalent in the setting of chronic kidney disease (CKD). Such coexistence of CVD and CKD—the so-called “cardiorenal or renocardiac syndrome”—contributes to exponentially increased risk of cardiovascular (CV) mortality. Uremic cardiomyopathy is a characteristic cardiac pathology commonly found in CKD. CKD patients are also predisposed to heart rhythm disorders especially atrial fibrillation. Traditional CV risk factors as well as known CKD-associated CV risk factors such as anemia are insufficient to explain CV complications in the CKD population. Accumulation of uremic retention solutes is a hallmark of impaired renal excretory function. Many of them have been considered inert solutes until their biological toxicity is unraveled and they become accepted as “uremic toxins”. Direct cardiotoxicity of uremic toxins has been increasingly demonstrated in recent years. This review offers a mechanistic insight into the pathological cardiac remodeling and dysfunction contributed by uremic toxins with a main focus on fibroblastic growth factor-23, an emerging toxin playing a central role in the chronic kidney disease–mineral bone disorder, and the two most investigated non-dialyzable protein-bound uremic toxins, indoxyl sulfate and p-cresyl sulfate. Potential therapeutic strategies that could address these toxins and their relevant mediated pathways since pre-dialysis stages are also discussed.
Collapse
Affiliation(s)
- Suree Lekawanvijit
- Department of Pathology, Faculty of Medicine, Chiang Mai University, 110 Intawaroros Rd, Sribhoom, Chiang Mai 50200, Thailand.
| |
Collapse
|
69
|
Chen X, Lei T. Glycerol fructose combined with vitamin B6 is beneficial to postoperative recovery of patients with cerebral aneurysm. Exp Ther Med 2018; 16:236-240. [PMID: 29977363 PMCID: PMC6030894 DOI: 10.3892/etm.2018.6133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/23/2018] [Indexed: 12/23/2022] Open
Abstract
The aim of the present study is to investigate whether glycerol fructose combined with vitamin B6 is beneficial to the postoperative recovery of patients with cerebral aneurysm (CA). A total of 134 patients receiving embolization of CA in the Central Hospital of Wuhan between February, 2013 and June, 2015 and were divided into observation and control groups according to the random number table method, with 67 cases in each group. The control was given vitamin B6 routine treatment, while the observation group received glycerol fructose on the basis of treatment in the control group. The incidence rate of postoperative complications after treatment, Glasgow coma scale (GCS) score, Barthel index score, and neurological fatigue index (NFI) score were compared between the two groups. After treatment, the GCS, Barthel index and NFI scores of patients in the observation were better than those in the control group (p<0.05), and the Barthel index score in the observation group was significantly higher than that in the control group (p<0.01). The mean flow velocity of middle cerebral artery (MCA) in the observation group after treatment was significantly different from that in the control group (p<0.05). As for complications, the incidence rates of postoperative cerebral vasospasm (1.49%), cerebral ischemia (1.49%), hematoma at puncture site (2.98%) and aneurysm rupture and hemorrhage (4.47%) in the observation group were lower than those of cerebral vasospasm (8.95%), cerebral ischemia (7.46%), hematoma at puncture site (8.95%) and aneurysm rupture and hemorrhage (10.44%) in the control group, and the differences were statistically significant (p<0.05). In conclusion, glycerol fructose combined with vitamin B6 can reduce the incidence rate of postoperative complications and improve patients self-care ability and quality of life. Therefore, it is beneficial to postoperative recovery and it is worthy of clinical application.
Collapse
Affiliation(s)
- Xiya Chen
- Department of Neurosurgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 431400, P.R. China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
70
|
Cueto R, Zhang L, Shan HM, Huang X, Li X, Li YF, Lopez J, Yang WY, Lavallee M, Yu C, Ji Y, Yang X, Wang H. Identification of homocysteine-suppressive mitochondrial ETC complex genes and tissue expression profile - Novel hypothesis establishment. Redox Biol 2018; 17:70-88. [PMID: 29679893 PMCID: PMC6006524 DOI: 10.1016/j.redox.2018.03.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 03/22/2018] [Indexed: 12/13/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) is an independent risk factor for cardiovascular disease (CVD) which has been implicated in matochondrial (Mt) function impairment. In this study, we characterized Hcy metabolism in mouse tissues by using LC-ESI-MS/MS analysis, established tissue expression profiles for 84 nuclear-encoded Mt electron transport chain complex (nMt-ETC-Com) genes in 20 human and 19 mouse tissues by database mining, and modeled the effect of HHcy on Mt-ETC function. Hcy levels were high in mouse kidney/lung/spleen/liver (24-14 nmol/g tissue) but low in brain/heart (~5 nmol/g). S-adenosylhomocysteine (SAH) levels were high in the liver/kidney (59-33 nmol/g), moderate in lung/heart/brain (7-4 nmol/g) and low in spleen (1 nmol/g). S-adenosylmethionine (SAM) was comparable in all tissues (42-18 nmol/g). SAM/SAH ratio was as high as 25.6 in the spleen but much lower in the heart/lung/brain/kidney/liver (7-0.6). The nMt-ETC-Com genes were highly expressed in muscle/pituitary gland/heart/BM in humans and in lymph node/heart/pancreas/brain in mice. We identified 15 Hcy-suppressive nMt-ETC-Com genes whose mRNA levels were negatively correlated with tissue Hcy levels, including 11 complex-I, one complex-IV and two complex-V genes. Among the 11 Hcy-suppressive complex-I genes, 4 are complex-I core subunits. Based on the pattern of tissue expression of these genes, we classified tissues into three tiers (high/mid/low-Hcy responsive), and defined heart/eye/pancreas/brain/kidney/liver/testis/embryonic tissues as tier 1 (high-Hcy responsive) tissues in both human and mice. Furthermore, through extensive literature mining, we found that most of the Hcy-suppressive nMt-ETC-Com genes were suppressed in HHcy conditions and related with Mt complex assembly/activity impairment in human disease and experimental models. We hypothesize that HHcy inhibits Mt complex I gene expression leading to Mt dysfunction.
Collapse
Affiliation(s)
- Ramon Cueto
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Lixiao Zhang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Hui Min Shan
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Xiao Huang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Xinyuan Li
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Ya-Feng Li
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Jahaira Lopez
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - William Y Yang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Muriel Lavallee
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Catherine Yu
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA; The Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China.
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA; Department of Pharmacology, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA; Thrombosis Research Center, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA; Cardiovascular Research Center, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University - Lewis Katz School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA; Department of Pharmacology, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA; Thrombosis Research Center, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA; Cardiovascular Research Center, Temple University - Lewis Katz School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
71
|
Impact of homocysteine on vasculogenic factors and bone formation in chicken embryos. Cell Biol Toxicol 2018; 35:49-58. [DOI: 10.1007/s10565-018-9436-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/21/2018] [Indexed: 12/21/2022]
|
72
|
Srećković B, Soldatovic I, Colak E, Mrdovic I, Sumarac-Dumanovic M, Janeski H, Janeski N, Gacic J, Dimitrijevic-Sreckovic V. Homocysteine is the confounding factor of metabolic syndrome-confirmed by siMS score. Drug Metab Pers Ther 2018; 33:99-103. [PMID: 29624500 DOI: 10.1515/dmpt-2017-0013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 02/09/2018] [Indexed: 11/15/2022]
Abstract
BACKGROUND Abdominal adiposity has a central role in developing insulin resistance (IR) by releasing pro-inflammatory cytokines. Patients with metabolic syndrome (MS) have higher values of homocysteine. Hyperhomocysteinemia correlates with IR, increasing the oxidative stress. Oxidative stress causes endothelial dysfunction, hypertension and atherosclerosis. The objective of the study was to examine the correlation of homocysteine with siMS score and siMS risk score and with other MS co-founding factors. METHODS The study included 69 obese individuals (age over 30, body mass index [BMI] >25 kg/m2), classified into two groups: I-with MS (33 patients); II-without MS (36 patients). Measurements included: anthropometric parameters, lipids, glucose regulation parameters and inflammation parameters. IR was determined by homeostatic model assessment for insulin resistance (HOMA-IR). ATP III classification was applied for diagnosing MS. SiMS score was used as continuous measure of metabolic syndrome. RESULTS A significant difference between groups was found for C-reactive protein (CRP) (p<0.01) apolipoprotein (Apo) B, HOMA-IR and acidum uricum (p<0.05). siMS risk score showed a positive correlation with homocysteine (p=0.023), while siMS score correlated positively with fibrinogen (p=0.013), CRP and acidum uricum (p=0.000) and homocysteine (p=0.08). Homocysteine correlated positively with ApoB (p=0.036), HbA1c (p=0.047), HOMA-IR (p=0.008) and negatively with ApoE (p=0.042). CONCLUSIONS Correlation of siMS score with homocysteine, fibrinogen, CRP and acidum uricum indicates that they are co-founding factors of MS. siMS risk score correlation with homocysteine indicates that hyperhomocysteinemia increases with age. Hyperhomocysteinemia is linked with genetic factors and family nutritional scheme, increasing the risk for atherosclerosis.
Collapse
Affiliation(s)
| | - Ivan Soldatovic
- Institute for Medical Statistics and Informatics, Belgrade, Serbia
| | - Emina Colak
- Institute of Medical Biochemistry, Clinical Center of Serbia, Belgrade, Serbia
| | - Igor Mrdovic
- Clinic for Cardiovascular Diseases, Emergency Center, Belgrade, Serbia
| | - Mirjana Sumarac-Dumanovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia and Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | | | - Jasna Gacic
- Clinical Center Bežanijska Kosa, Belgrade, Serbia
| | - Vesna Dimitrijevic-Sreckovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia and Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
73
|
Kostić S, Mićovic Ž, Andrejević L, Cvetković S, Stamenković A, Stanković S, Obrenović R, Labudović-Borović M, Hrnčić D, Jakovljević V, Djurić D. The effects of L-cysteine and N-acetyl-L-cysteine on homocysteine metabolism and haemostatic markers, and on cardiac and aortic histology in subchronically methionine-treated Wistar male rats. Mol Cell Biochem 2018; 451:43-54. [PMID: 29936684 DOI: 10.1007/s11010-018-3391-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/17/2018] [Indexed: 11/24/2022]
Abstract
Methionine is the precursor of homocysteine, a sulfur amino acid intermediate in the methylation and transsulfuration pathways; methionine-rich diets were used to induce hyperhomocysteinemia, and cardiovascular pathology was often observed. Other sulfur amino acids interfere with this metabolism, i.e., L-cysteine (Cys) and N-aceyl-L-cysteine (NAC), and probably also affect cardiovascular system. Their effects are controversial due to their ability to act both as anti- or pro-oxidant. Thus, this study aimed to elucidate their influence on levels of homocysteine, folate and vitamin B12, levels of different haemostatic parameters (fibrinogen, D-dimer, vWF Ag, vWF Ac) in rat serum or plasma as well as their effects on cardiac and aortic tissue histology in subchronically methionine-treated rats. Wistar albino rats were divided into 4 experimental groups: (a) control group (0.9% sodium chloride 0.1-0.2 mL/day) (n = 10) (K); (b) DL-methionine (0.8 mmol/kg/bw/day) (n = 10) (M); (c) DL-methionine (0.8 mmol/kg/bw/day) + L-cysteine (7 mg/kg/bw/day) (n = 8) (C); (d) DL-methionine (0.8 mmol/ kg/bw/day) + N-acetyl-L-cysteine (50 mg/kg/bw/day) (n = 8) (N). All substances were applied i.p., treatment duration 3 weeks. Lower levels of vitamin B12 in all the groups were found. Folate was reduced only in N group. Decreased fibrinogen was noted in C and N groups and increased D-dimer only in C. VWF activity was reduced in M and C groups. Deleterious effects in heart were observed, especially after Cys and NAC application. Aortic tissue remained unchanged. In conclusion, it could be said that sulfur amino acids have the significant impact on cardiovascular system in subchronically methionine-treated rats. This study points out the relevance of their complex interactions and deleterious effects mediated by either direct influence or procoagulant properties.
Collapse
Affiliation(s)
- Sanja Kostić
- Faculty of Medicine, Institute of Medical Physiology "Richard Burian", University of Belgrade, Visegradska 26/II, Belgrade, 11000, Serbia
| | - Žarko Mićovic
- Military Health Department, Ministry of Defence, Belgrade, Serbia
| | - Lazar Andrejević
- Clinic of Gynecology and Obstetrics, Faculty of Medical Science University of Pristina - Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Saša Cvetković
- Clinic of Gynecology and Obstetrics, Faculty of Medical Science University of Pristina - Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Aleksandra Stamenković
- St. Boniface Hospital Research Center, Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Canada
| | - Sanja Stanković
- Centre of Medical Biochemistry, Clinical Centre of Serbia, Belgrade, Serbia
| | - Radmila Obrenović
- Centre of Medical Biochemistry, Clinical Centre of Serbia, Belgrade, Serbia
| | - Milica Labudović-Borović
- Faculty of Medicine, Institute of Histology and Embryology "Aleksandar Dj. Kostic", University of Belgrade, Belgrade, Serbia
| | - Dragan Hrnčić
- Faculty of Medicine, Institute of Medical Physiology "Richard Burian", University of Belgrade, Visegradska 26/II, Belgrade, 11000, Serbia
| | - Vladimir Jakovljević
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Department of Human Pathology, 1st Moscow State Medical University IM Sechenov, Moscow, Russian Federation
| | - Dragan Djurić
- Faculty of Medicine, Institute of Medical Physiology "Richard Burian", University of Belgrade, Visegradska 26/II, Belgrade, 11000, Serbia.
| |
Collapse
|
74
|
Hendrix P, Foreman PM, Harrigan MR, Fisher WS, Vyas NA, Lipsky RH, Lin M, Walters BC, Tubbs RS, Shoja MM, Pittet JF, Mathru M, Griessenauer CJ. Association of cystathionine beta-synthase polymorphisms and aneurysmal subarachnoid hemorrhage. J Neurosurg 2018; 128:1771-1777. [DOI: 10.3171/2017.2.jns162933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVECystathionine β-synthase (CBS) is involved in homocysteine and hydrogen sulfide (H2S) metabolism. Both products have been implicated in the pathophysiology of cerebrovascular diseases. The impact of CBS polymorphisms on aneurysmal subarachnoid hemorrhage (aSAH) and its clinical sequelae is poorly understood.METHODSBlood samples from all patients enrolled in the CARAS (Cerebral Aneurysm Renin Angiotensin System) study were used for genetic evaluation. The CARAS study prospectively enrolled aSAH patients at 2 academic institutions in the United States from 2012 to 2015. Common CBS polymorphisms were detected using 5′exonuclease genotyping assays. Analysis of associations between CBS polymorphisms and aSAH was performed.RESULTSSamples from 149 aSAH patients and 50 controls were available for analysis. In multivariate logistic regression analysis, the insertion allele of the 844ins68 CBS insertion polymorphism showed a dominant effect on aSAH. The GG genotype of the CBS G/A single nucleotide polymorphism (rs234706) was independently associated with unfavorable functional outcome (modified Rankin Scale Score 3–6) at discharge and last follow-up, but not clinical vasospasm or delayed cerebral ischemia (DCI).CONCLUSIONSThe insertion allele of the 844ins68 CBS insertion polymorphism was independently associated with aSAH while the GG genotype of rs234706 was associated with an unfavorable outcome both at discharge and last follow-up. Increased CBS activity may exert its neuroprotective effects through alteration of H2S levels, and independent of clinical vasospasm and DCI.
Collapse
Affiliation(s)
- Philipp Hendrix
- 1Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg/Saar, Germany
| | - Paul M. Foreman
- 2Department of Neurosurgery, University of Alabama at Birmingham, Alabama
| | - Mark R. Harrigan
- 2Department of Neurosurgery, University of Alabama at Birmingham, Alabama
| | - Winfield S. Fisher
- 2Department of Neurosurgery, University of Alabama at Birmingham, Alabama
| | - Nilesh A. Vyas
- 3Department of Neurosciences, Inova Health System, Falls Church
| | - Robert H. Lipsky
- 3Department of Neurosciences, Inova Health System, Falls Church
- 4Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia
| | - Mingkuan Lin
- 4Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia
| | - Beverly C. Walters
- 2Department of Neurosurgery, University of Alabama at Birmingham, Alabama
- 4Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia
| | | | - Mohammadali M. Shoja
- 6Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mali Mathru
- 7Department of Anesthesiology, University of Alabama at Birmingham, Alabama
| | - Christoph J. Griessenauer
- 8Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; and
- 9Department of Neurosurgery, Geisinger Health System, Danville, Pennsylvania
| |
Collapse
|
75
|
Huang A, Patel S, McAlpine CS, Werstuck GH. The Role of Endoplasmic Reticulum Stress-Glycogen Synthase Kinase-3 Signaling in Atherogenesis. Int J Mol Sci 2018; 19:ijms19061607. [PMID: 29848965 PMCID: PMC6032052 DOI: 10.3390/ijms19061607] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 01/31/2023] Open
Abstract
Cardiovascular disease (CVD) is the number one cause of global mortality and atherosclerosis is the underlying cause of most CVD. However, the molecular mechanisms by which cardiovascular risk factors promote the development of atherosclerosis are not well understood. The development of new efficient therapies to directly block or slow disease progression will require a better understanding of these mechanisms. Accumulating evidence supports a role for endoplasmic reticulum (ER) stress in all stages of the developing atherosclerotic lesion however, it was not clear how ER stress may contribute to disease progression. Recent findings have shown that ER stress signaling through glycogen synthase kinase (GSK)-3α may significantly contribute to macrophage lipid accumulation, inflammatory cytokine production and M1macrophage polarization. In this review we summarize our knowledge of the potential role of ER stress-GSK3 signaling in the development and progression of atherosclerosis as well as the possible therapeutic implications of this pathway.
Collapse
Affiliation(s)
- Aric Huang
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L9L 2X2, Canada.
| | - Sarvatit Patel
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L9L 2X2, Canada.
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada.
| | - Cameron S McAlpine
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L9L 2X2, Canada.
| | - Geoff H Werstuck
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L9L 2X2, Canada.
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada.
- Department of Medicine, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
76
|
Vanholder R, Argilés A, Baurmeister U, Brunet P, Clark W, Cohen G, Dedeyn P, Deppisch R, Descamps-Latscha B, Henle T, Jörres A, Massy Z, Rodriguez M, Stegmayr B, Stenvinkel P, Wratten M. Uremic Toxicity: Present State of the Art. Int J Artif Organs 2018. [DOI: 10.1177/039139880102401004] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The uremic syndrome is a complex mixture of organ dysfunctions, which is attributed to the retention of a myriad of compounds that under normal condition are excreted by the healthy kidneys (uremic toxins). In the area of identification and characterization of uremic toxins and in the knowledge of their pathophysiologic importance, major steps forward have been made during recent years. The present article is a review of several of these steps, especially in the area of information about the compounds that could play a role in the development of cardiovascular complications. It is written by those members of the Uremic Toxins Group, which has been created by the European Society for Artificial Organs (ESAO). Each of the 16 authors has written a state of the art in his/her major area of interest.
Collapse
Affiliation(s)
- R. Vanholder
- The Nephrology Section, Department of Internal Medicine, University Hospital, Gent - Belgium
| | - A. Argilés
- Institute of Human Genetics, IGH-CNRS UPR 1142, Montpellier - France
| | | | - P. Brunet
- Nephrology, Internal Medicine, Ste Marguerite Hospital, Marseille - France
| | - W. Clark
- Baxter Healthcare Corporation, Lessines - Belgium
| | - G. Cohen
- Division of Nephrology, Department of Medicine, University of Vienna, Vienna - Austria
| | - P.P. Dedeyn
- Department of Neurology, Middelheim Hospital, Laboratory of Neurochemistry and Behaviour, University of Antwerp - Belgium
| | - R. Deppisch
- Gambro Corporate Research, Hechingen - Germany
| | | | - T. Henle
- Institute of Food Chemistry, Technical University, Dresden - Germany
| | - A. Jörres
- Nephrology and Medical Intensive Care, UK Charité, Campus Virchow-Klinikum, Medical Faculty of Humboldt-University, Berlin - Germany
| | - Z.A. Massy
- Division of Nephrology, CH-Beauvais, and INSERM Unit 507, Necker Hospital, Paris - France
| | - M. Rodriguez
- University Hospital Reina Sofia, Research Institute, Cordoba - Spain
| | - B. Stegmayr
- Norrlands University Hospital, Medical Clinic, Umea - Sweden
| | - P. Stenvinkel
- Nephrology Department, University Hospital, Huddinge - Sweden
| | | |
Collapse
|
77
|
Cronje PB. Essential role of methyl donors in animal productivity. ANIMAL PRODUCTION SCIENCE 2018. [DOI: 10.1071/an15729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dietary requirements for the methyl donors, choline, betaine and folate, in livestock species are poorly defined and have not been included in diet formulation software or simulation models for animals. A deficiency of methyl donors may promote an inflammatory state, which is significant for the livestock industry because chronic low-grade inflammation is widespread among livestock under commercial conditions. Furthermore, recent evidence showing that methyl donors activate adenosine monophosphate-activated protein kinase, an anti-inflammatory master switch, indicates that dietary methyl-donor supplementation could be used to prevent or ameliorate chronic inflammation and its sequelae in livestock, which include fatty liver disease in dairy cows, fatty liver and kidney syndrome in broilers, fatty liver haemorrhagic syndrome in layers, gut ulcers in pigs, liver abscesses in feedlot cattle, enteritis in poultry and susceptibility to heat stress in all species. Because of the complexity of interactions among methyl donors, a modelling approach inclusive of a supporting research effort will be required to harness the potential of methyl-donor supplementation in livestock production.
Collapse
|
78
|
Azad MAK, Huang P, Liu G, Ren W, Teklebrh T, Yan W, Zhou X, Yin Y. Hyperhomocysteinemia and cardiovascular disease in animal model. Amino Acids 2017; 50:3-9. [DOI: 10.1007/s00726-017-2503-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/04/2017] [Indexed: 01/16/2023]
|
79
|
Zhou F, Wang N, Zhou L, Zhou Y, Guo T, Yu D. Methylenetetrahydrofolate reductase gene A1298C polymorphism and gene-environment interactions are associated with carotid plaque in a south Chinese population. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:9744-9752. [PMID: 31966857 PMCID: PMC6965971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/25/2017] [Indexed: 06/10/2023]
Abstract
OBJECTIVE Our primary objective was to evaluate the associations of conventional risk factors and methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms with the risk of carotid plaque in a south Chinese population. Our secondary objective was to explore gene-environment interactions and potential relationship with carotid plaque. METHOD We enrolled 229 patients suffer from carotid plaque and 180 controls in this case-control study. We measured the carotid intima-media thickness by ultrasound and investigated conventional risk factors, biomarkers and C677T/A1298C MTHFR genotypes. Logistic analysis was used to evaluate the association between conventional risk factors and carotid plaque. The SNPstats platform was used to investigate the association between MTHFR gene polymorphisms and carotid plaque under 5 genetic models (dominant, recessive, codominant, over dominant and additive models). Gene-environment interactions analysis was then performed by multifactor dimensionality reduction. RESULTS Age and hypertension were identified as independent risk factors of carotid plaque. C677T and A1298C demonstrated associations with carotid plaque under the recessive model (C677T: P = 0.03, odds ratio = 3.14, 95% confidence interval = 1.04-11.21; A1298C: P = 0.018, odds ratio = 2.40, 95% confidence interval = 1.13-5.10). Neither C677T nor A1298C polymorphism was associated with stable or vulnerable plaques. Additionally, Significant multiplicative and additive interactions were observed in terms of carotid plaque between A1298C polymorphism and diabetes, age, and smoking (P = 0.013). CONCLUSION MTHFR gene C677T and A1298C polymorphisms may act as modifiers of carotid plaque risk in south Chinese population. In addition, the combined effect of gene-environment interactions between A1298C polymorphism and conventional risk factors may promote the progression of carotid plaque.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Neurology, Affiliated Haikou Hospital at Xiangya Medical College of Central South University Haikou, Hainan Province, China
| | - Nianzhen Wang
- Department of Neurology, Affiliated Haikou Hospital at Xiangya Medical College of Central South University Haikou, Hainan Province, China
| | - Lv Zhou
- Department of Neurology, Affiliated Haikou Hospital at Xiangya Medical College of Central South University Haikou, Hainan Province, China
| | - Yanhui Zhou
- Department of Neurology, Affiliated Haikou Hospital at Xiangya Medical College of Central South University Haikou, Hainan Province, China
| | - Tie Guo
- Department of Neurology, Affiliated Haikou Hospital at Xiangya Medical College of Central South University Haikou, Hainan Province, China
| | - Dan Yu
- Department of Neurology, Affiliated Haikou Hospital at Xiangya Medical College of Central South University Haikou, Hainan Province, China
| |
Collapse
|
80
|
Homocysteine enhances the predictive value of the GRACE risk score in patients with ST-elevation myocardial infarction. Anatol J Cardiol 2017; 18:182-193. [PMID: 28782750 PMCID: PMC5689049 DOI: 10.14744/anatoljcardiol.2017.7798] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Objective: The present study aims to investigate whether the addition of homocysteine level to the Global Registry of Acute Coronary Events (GRACE) risk score enhances its predictive value for clinical outcomes in ST-elevation myocardial infarction (STEMI). Methods: A total of 1143 consecutive patients with STEMI were included in this prospective cohort study. Homocysteine was detected, and the GRACE score was calculated. The predictive power of the GRACE score alone or combined with homocysteine was assessed by the receiver operating characteristic (ROC) analysis, methods of net reclassification improvement (NRI) and integrated discrimination improvement (IDI). Results: During a median follow-up period of 36.7 months, 271 (23.7%) patients reached the clinical endpoints. It showed that the GRACE score and homocysteine could independently predict all-cause death [GRACE: HR=1.031 (1.024–1.039), p<0.001; homocysteine: HR=1.023 (1.018–1.028), p<0.001] and MACE [GRACE: HR=1.008 (1.005–1.011), p<0.001; homocysteine: HR=1.022 (1.018–1.025), p<0.001]. When they were used in combination to assess the clinical outcomes, the area under the ROC curve significantly increased from 0.786 to 0.884 (95% CI=0.067–0.128, Z=6.307, p<0.001) for all-cause death and from 0.678 to 0.759 (95% CI=0.055–0.108, Z=5.943, p<0.001) for MACE. The addition of homocysteine to the GRACE model improved NRI (all-cause death: 0.575, p<0.001; MACE: 0.621, p=0.008) and IDI (all-cause death: 0.083, p<0.001; MACE: 0.130, p=0.016), indicating effective discrimination and reclassification. Conclusion: Both the GRACE score and homocysteine are significant and independent predictors for clinical outcomes in patients with STEMI. A combination of them can develop a more predominant prediction for clinical outcomes in these patients.
Collapse
|
81
|
Lind M, Hayes A, Caprnda M, Petrovic D, Rodrigo L, Kruzliak P, Zulli A. Inducible nitric oxide synthase: Good or bad? Biomed Pharmacother 2017. [PMID: 28651238 DOI: 10.1016/j.biopha.2017.06.036] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Nitric oxide synthases (NOS) are a family of isoforms responsible for the synthesis of the potent dilator nitric oxide (NO). Expression of inducible NOS (iNOS) occurs in conditions of inflammation, and produces large amounts of NO. In pathological conditions iNOS is regarded as a harmful enzyme and is proposed to be a major contributor to diseases of the cardiovascular system such as atherosclerosis. In this review, we address the notion that iNOS is a detrimental enzyme in disease and discuss its potentially beneficial roles. Additionally, we describe other molecules associated with iNOS in diseases such as atherosclerosis, and current research on therapeutic inhibitors tested to reduced pathology associated with cardiovascular diseases (CVD).
Collapse
Affiliation(s)
- Maggie Lind
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Alan Hayes
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Daniel Petrovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Peter Kruzliak
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic; 2nd Department of Surgery, Centre of Vascular Diseases, Faculty of Medicine, Masaryk University and St. Anne´s Faculty Hospital, Brno, Czech Republic.
| | - Anthony Zulli
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia.
| |
Collapse
|
82
|
Liang Y, Zhen X, Wang K, Ma J. Folic acid attenuates cobalt chloride-induced PGE 2 production in HUVECs via the NO/HIF-1alpha/COX-2 pathway. Biochem Biophys Res Commun 2017. [PMID: 28624453 DOI: 10.1016/j.bbrc.2017.06.079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prostaglandin E2 (PGE2), an important lipid inflammatory mediator involved in the progression of vascular diseases, can be induced by hypoxia in many cell types. While folic acid has been shown to protect against inflammation in THP-1 cells during hypoxia and hypoxia-induced endothelial cell injury, whether it might do so by attenuating PGE2 production remains unclear. To investigate this we constructed a hypoxia-induced injury model by treating human umbilical vein endothelial cells (HUVECs) with cobalt chloride (CoCl2), which mimics the effects of hypoxia. In CoCl2-treated HUVECs, folic acid significantly attenuated PGE2 production and increased vasoprotective nitric oxide (NO) content. Folic acid also decreased cyclooxygenase-2 (COX-2) and hypoxia-inducible factor 1-alpha (HIF-1α) expression and altered endothelial nitric oxide synthase (eNOS) signaling by increasing p-eNOS(Ser1177) and decreasing p-eNOS(Thr495) in a dose-dependent manner. Further investigation of the pathway demonstrated that treatment with 2-Methoxyestradiol (2-MeOE2) and celecoxib both decreased CoCl2-induced COX-2 expression but only 2-MeOE2 decreased HIF-1α expression. The ability of folic acid to down-regulate HIF-1α and COX-2 protein levels was dramatically abrogated by L-NAME treatment, which also decreased eNOS mRNA and NO production. The NO donor sodium nitroprusside also dose-dependently down-regulated HIF-1α and COX-2 protein levels. Overall, these findings suggest a novel application for folic acid in attenuating CoCl2-induced PGE2 production in HUVECs via regulation of the NO/HIF-1α/COX-2 pathway.
Collapse
Affiliation(s)
- Yuming Liang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, PR China
| | - Xiaozhou Zhen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, PR China
| | - Kaiwen Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, PR China
| | - Jing Ma
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
83
|
Homocysteine Induces Apoptosis of Human Umbilical Vein Endothelial Cells via Mitochondrial Dysfunction and Endoplasmic Reticulum Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017. [PMID: 28630659 PMCID: PMC5467318 DOI: 10.1155/2017/5736506] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Homocysteine- (Hcy-) induced endothelial cell apoptosis has been suggested as a cause of Hcy-dependent vascular injury, while the proposed molecular pathways underlying this process are unclear. In this study, we investigated the adverse effects of Hcy on human umbilical vein endothelial cells (HUVEC) and the underlying mechanisms. Our results demonstrated that moderate-dose Hcy treatment induced HUVEC apoptosis in a time-dependent manner. Furthermore, prolonged Hcy treatment increased the expression of NOX4 and the production of intracellular ROS but decreased the ratio of Bcl-2/Bax and mitochondrial membrane potential (MMP), resulting in the leakage of cytochrome c and activation of caspase-3. Prolonged Hcy treatment also upregulated glucose-regulated protein 78 (GRP78), activated protein kinase RNA-like ER kinase (PERK), and induced the expression of C/EBP homologous protein (CHOP) and the phosphorylation of NF-κb. The inhibition of NOX4 decreased the production of ROS and alleviated the Hcy-induced HUVEC apoptosis and ER stress. Blocking the PERK pathway partly alleviated Hcy-induced HUVEC apoptosis and the activation of NF-κb. Taken together, our results suggest that Hcy-induced mitochondrial dysfunction crucially modulated apoptosis and contributed to the activation of ER stress in HUVEC. The excessive activation of the PERK pathway partly contributed to Hcy-induced HUVEC apoptosis and the phosphorylation of NF-κb.
Collapse
|
84
|
Hildebrandt W, Schwarzbach H, Pardun A, Hannemann L, Bogs B, König AM, Mahnken AH, Hildebrandt O, Koehler U, Kinscherf R. Age-related differences in skeletal muscle microvascular response to exercise as detected by contrast-enhanced ultrasound (CEUS). PLoS One 2017; 12:e0172771. [PMID: 28273102 PMCID: PMC5342194 DOI: 10.1371/journal.pone.0172771] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022] Open
Abstract
Background Aging involves reductions in exercise total limb blood flow and exercise capacity. We hypothesized that this may involve early age-related impairments of skeletal muscle microvascular responsiveness as previously reported for insulin but not for exercise stimuli in humans. Methods Using an isometric exercise model, we studied the effect of age on contrast-enhanced ultrasound (CEUS) parameters, i.e. microvascular blood volume (MBV), flow velocity (MFV) and blood flow (MBF) calculated from replenishment of Sonovue contrast-agent microbubbles after their destruction. CEUS was applied to the vastus lateralis (VLat) and intermedius (VInt) muscle in 15 middle-aged (MA, 43.6±1.5 years) and 11 young (YG, 24.1±0.6 years) healthy males before, during, and after 2 min of isometric knee extension at 15% of peak torque (PT). In addition, total leg blood flow as recorded by femoral artery Doppler-flow. Moreover, fiber-type-specific and overall capillarisation as well as fiber composition were additionally assessed in Vlat biopsies obtained from CEUS site. MA and YG had similar quadriceps muscle MRT-volume or PT and maximal oxygen uptake as well as a normal cardiovascular risk factors and intima-media-thickness. Results During isometric exercise MA compared to YG reached significantly lower levels in MFV (0.123±0.016 vs. 0.208±0.036 a.u.) and MBF (0.007±0.001 vs. 0.012±0.002 a.u.). In the VInt the (post-occlusive hyperemia) post-exercise peaks in MBV and MBF were significantly lower in MA vs. YG. Capillary density, capillary fiber contacts and femoral artery Doppler were similar between MA and YG. Conclusions In the absence of significant age-related reductions in capillarisation, total leg blood flow or muscle mass, healthy middle-aged males reveal impaired skeletal muscle microcirculatory responses to isometric exercise. Whether this limits isometric muscle performance remains to be assessed.
Collapse
Affiliation(s)
- Wulf Hildebrandt
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Marburg, Marburg, Germany
- * E-mail:
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Anita Pardun
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Lena Hannemann
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Björn Bogs
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Alexander M. König
- Department of Diagnostic and Interventional Radiology, University Hospital of Giessen and Marburg (UKGM) University, Baldingerstraße, Marburg, Germany
| | - Andreas H. Mahnken
- Department of Diagnostic and Interventional Radiology, University Hospital of Giessen and Marburg (UKGM) University, Baldingerstraße, Marburg, Germany
| | - Olaf Hildebrandt
- Department of Sleep Medicine, Division of Pneumology, Internal Medicine, University Hospital of Giessen and Marburg (UKGM) Baldingerstraße, Marburg, Germany
| | - Ulrich Koehler
- Department of Sleep Medicine, Division of Pneumology, Internal Medicine, University Hospital of Giessen and Marburg (UKGM) Baldingerstraße, Marburg, Germany
| | - Ralf Kinscherf
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| |
Collapse
|
85
|
Steger CM, Mayr T, Bonaros N, Bonatti J, Schachner T. Vein graft disease in a knockout mouse model of hyperhomocysteinaemia. Int J Exp Pathol 2016; 97:447-456. [PMID: 28004436 DOI: 10.1111/iep.12215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 11/15/2016] [Indexed: 12/28/2022] Open
Abstract
A major reason for vein graft failure after coronary artery bypass grafting is neointimal hyperplasia and thrombosis. Elevated serum levels of homocysteine (Hcy) are associated with higher incidence of cardiovascular disease, but homocysteine levels also tend to increase during the first weeks or months after cardiac surgery. To investigate this further, C57BL/6J mice (WT) and cystathionine-beta-synthase heterozygous knockout mice (CBS+/-), a mouse model for hyperhomocysteinaemia, underwent interposition of the vena cava of donor mice into the carotid artery of recipient mice. Two experimental groups were examined: 20 mice of each group underwent bypass surgery (group 1: WT donor and WT recipient; group 2: CBS+/- donor and CBS+/- recipient). After 4 weeks, the veins were harvested, dehydrated, paraffin-embedded, stained and analysed by histomorphology and immunohistochemistry. Additionally, serum Hcy levels in CBS knockout animals and in WT animals before and after bypass surgery were measured. At 4 weeks postoperatively, group 2 mice showed a higher percentage of thrombosis compared to controls, a threefold increase in neointima formation, higher general vascularization, a lower percentage of elastic fibres with shortage and fragmentation in the neointima, a lower percentage of acid mucopolysaccharides in the neointima and a more intense fibrosis in the neointima and media. In conclusion, hyperhomocysteinaemic cystathionine-beta-synthase knockout mice can play an important role in the study of mechanisms of vein graft failure. But further in vitro and in vivo studies are necessary to answer the question whether or not homocysteine itself or a related metabolic factor is the key aetiologic agent for accelerated vein graft disease.
Collapse
Affiliation(s)
- Christina Maria Steger
- Department of Pathology, Academic Teaching Hospital Feldkirch (Affiliation of the Innsbruck Medical University), Feldkirch, Austria
| | - Tobias Mayr
- Department of Surgery, State Hospital Kufstein, Kufstein, Austria
| | - Nikolaos Bonaros
- Department of Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Johannes Bonatti
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Thomas Schachner
- Department of Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
86
|
Klafke JZ, Pereira RLD, Hirsch GE, Parisi MM, Porto FG, de Almeida AS, Rubin FH, Schmidt A, Beutler H, Nascimento S, Trevisan G, Brusco I, de Oliveira SM, Duarte MMMF, Duarte T, Viecili PRN. Study of oxidative and inflammatory parameters in LDLr-KO mice treated with a hypercholesterolemic diet: Comparison between the use of Campomanesia xanthocarpa and acetylsalicylic acid. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1227-1234. [PMID: 26776955 DOI: 10.1016/j.phymed.2015.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/11/2015] [Accepted: 11/14/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Atherosclerosis is an inflammatory disease that affects the arterial wall leading to myocardial, cerebral, and peripheral ischemic syndromes. The use of low doses of aspirin inhibits platelet aggregation and inflammation and prevents cardiovascular mortality. However, ASA may produce hemorrhagic events. Thus, several studies have sought new natural compounds to suppress platelet aggregation without causing serious adverse effects. PURPOSE In this sense, this study aims to compare the effects of Campomanesia xanthocarpa plant extract with those of acetylsalicylic acid (ASA) on inflammatory parameters observed in homozygous mice knockout for the low-density lipoprotein receptor (LDLr-KO) treated with a hypercholesterolemic diet. MATERIAL AND METHODS In this study, 28 male LDLr-KO mice were divided into three groups and fed a hypercholesterolemic diet for 4 weeks. Thereafter, the animals that received the hypercholesterolemic diet were treated for 5 days with (1) distilled water, (2) C. xanthocarpa extract, or (3) acetylsalicylic acid. The levels of inflammatory markers were assessed in the blood samples. The gastric tolerability of the animals after oral administration of the treatments was assessed through quantification of the lesions in the gastric mucosa. RESULTS The levels of proinflammatory cytokines IL-1, IL-6, TNF-α, and INF-γ were reduced to 19.2 ± 3%, 20.4 + 1.3%, 24.7 ± 1.2%, and 20.8 ± 1.7%, respectively, in the group treated with C. xanthocarpa, when compared to control group. Furthermore, treatment with plant extract significantly increased the levels of the anti-inflammatory cytokine IL-10 by 27.3 ± 5.9%, but ASA showed no significant effect on the same cytokines when compared to the control group, with the exception of IL-10, which presented an increase of 8.6 ± 3.5%. Treatments with C. xanthocarpa and ASA also caused significant reductions of 26.4 ± 3% and 38.4± 6% in the serum levels of oxLDL, respectively. However, only treatment with C. xanthocarpa reduced the levels of anti-oxLDL antibodies when compared with the control (25.8 ± 6%). In addition, the analyzed extract did not induce ulcerogenic activity, while ASA induced the formation of lesions. CONCLUSION In conclusion, treatment with C. xanthocarpa causes anti-inflammatory activity in hypercholesterolemic animals, with results superior to those obtained with the use of ASA.
Collapse
Affiliation(s)
- Jonatas Zeni Klafke
- Programa de Pós-Graduação em Atenção Integral à Saúde (PPGAIS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil.
| | - Roberta Lelis Dias Pereira
- Programa de Pós-Graduação em Atenção Integral à Saúde (PPGAIS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil
| | - Gabriela Elisa Hirsch
- Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil
| | - Mariana Migliorini Parisi
- Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil
| | - Fernando Garcez Porto
- Programa de Pós-Graduação em Atenção Integral à Saúde (PPGAIS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil
| | - Amanda Spring de Almeida
- Programa de Pós-Graduação em Atenção Integral à Saúde (PPGAIS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil
| | - Fabiane Horbach Rubin
- Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil
| | - Aline Schmidt
- Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil
| | - Henrique Beutler
- Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil
| | - Sabrina Nascimento
- Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil
| | - Gabriela Trevisan
- Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), 88006-000 Cricíuma, SC, Brazil
| | - Indiara Brusco
- Programa de Pós-Graduacão em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Sara Marchesan de Oliveira
- Programa de Pós-Graduacão em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | | | - Thiago Duarte
- Centro de ciências da saúde, Pós-Graduação em farmacologia Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Paulo Ricardo Nazário Viecili
- Programa de Pós-Graduação em Atenção Integral à Saúde (PPGAIS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Grupo Multidisciplinar de Saúde (GMS), Universidade de Cruz Alta (UNICRUZ), 98020-290 Cruz Alta, RS, Brazil; Centro de Ensino e Pesquisa (CEP), Instituto de Cardiologia de Cruz Alta (ICCA), 98010-110 Cruz Alta, RS, Brazil.
| |
Collapse
|
87
|
Cao C, Zhang H, Zhao L, Zhou L, Zhang M, Xu H, Han X, Li G, Yang X, Jiang Y. miR-125b targets DNMT3b and mediates p53 DNA methylation involving in the vascular smooth muscle cells proliferation induced by homocysteine. Exp Cell Res 2016; 347:95-104. [PMID: 27426728 DOI: 10.1016/j.yexcr.2016.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are short non-coding RNA and play crucial roles in a wide array of biological processes, including cell proliferation, differentiation and apoptosis. Our previous studies found that homocysteine(Hcy) can stimulate the proliferation of vascular smooth muscle cells (VSMCs), however, the underlying mechanisms were not fully elucidated. Here, we found proliferation of VSMCs induced by Hcy was of correspondence to the miR-125b expression reduced both in vitro and in the ApoE knockout mice, the hypermethylation of p53, its decreased expression, and DNA (cytosine-5)-methyltransferase 3b (DNMT3b) up-regulated. And, we found DNMT3b is a target of miR-125b, which was verified by the Dual-Luciferase reporter assay and western blotting. Besides, the siRNA interference for DNMT3b significantly decreased the methylation level of p53, which unveiled the causative role of DNMT3b in p53 hypermethylation. miR-125b transfection further confirmed its regulative roles on p53 gene methylation status and the VSMCs proliferation. Our data suggested that a miR-125b-DNMT3b-p53 signal pathway may exist in the VSMCs proliferation induced by Hcy.
Collapse
Affiliation(s)
- ChengJian Cao
- Key Laboratory of Basic Research in Cardio-Cerebral Vascular Diseases, Ningxia Medical University, Yinchuan, China
| | - HuiPing Zhang
- Department of Prenatal Diagnosis Center, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Li Zhao
- Department of Medical Laboratory, Ningxia Medical University, Yinchuan, China
| | - Longxia Zhou
- Department of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Minghao Zhang
- Key Laboratory of Basic Research in Cardio-Cerebral Vascular Diseases, Ningxia Medical University, Yinchuan, China; Department of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Hua Xu
- Key Laboratory of Basic Research in Cardio-Cerebral Vascular Diseases, Ningxia Medical University, Yinchuan, China; Department of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Xuebo Han
- Department of Medical Laboratory, Ningxia Medical University, Yinchuan, China
| | - Guizhong Li
- Key Laboratory of Basic Research in Cardio-Cerebral Vascular Diseases, Ningxia Medical University, Yinchuan, China; Department of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Xiaoling Yang
- Key Laboratory of Basic Research in Cardio-Cerebral Vascular Diseases, Ningxia Medical University, Yinchuan, China; Department of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - YiDeng Jiang
- Key Laboratory of Basic Research in Cardio-Cerebral Vascular Diseases, Ningxia Medical University, Yinchuan, China; Department of Basic Medicine, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
88
|
Weber GJ, Pushpakumar S, Tyagi SC, Sen U. Homocysteine and hydrogen sulfide in epigenetic, metabolic and microbiota related renovascular hypertension. Pharmacol Res 2016; 113:300-312. [PMID: 27602985 DOI: 10.1016/j.phrs.2016.09.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 12/18/2022]
Abstract
Over the past several years, hydrogen sulfide (H2S) has been shown to be an important player in a variety of physiological functions, including neuromodulation, vasodilation, oxidant regulation, inflammation, and angiogenesis. H2S is synthesized primarily through metabolic processes from the amino acid cysteine and homocysteine in various organ systems including neuronal, cardiovascular, gastrointestinal, and kidney. Derangement of cysteine and homocysteine metabolism and clearance, particularly in the renal vasculature, leads to H2S biosynthesis deregulation causing or contributing to existing high blood pressure. While a variety of environmental influences, such as diet can have an effect on H2S regulation and function, genetic factors, and more recently epigenetics, also have a vital role in H2S regulation and function, and therefore disease initiation and progression. In addition, new research into the role of gut microbiota in the development of hypertension has highlighted the need to further explore these microorganisms and how they influence the levels of H2S throughout the body and possibly exploiting microbiota for use of hypertension treatment. In this review, we summarize recent advances in the field of hypertension research emphasizing renal contribution and how H2S physiology can be exploited as a possible therapeutic strategy to ameliorate kidney dysfunction as well as to control blood pressure.
Collapse
Affiliation(s)
- Gregory J Weber
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY 40202, United States
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY 40202, United States
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY 40202, United States
| | - Utpal Sen
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY 40202, United States.
| |
Collapse
|
89
|
Jamison RL, Hartigan P, Gaziano JM, Fortmann SP, Goldfarb DS, Haroldson JA, Kaufman J, Lavori P, McCully KS, Robinson K. Design and statistical issues in the homocysteinemia in kidney and end stage renal disease (HOST) study. Clin Trials 2016; 1:451-60. [PMID: 16279283 DOI: 10.1191/1740774504cn038oa] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The Homocysteine Study (HOST) Veterans Affairs Cooperative Studies Program No. 453, is a prospective, randomized, two arm, double blind study of patients with end stage renal disease (ESRD) or advanced chronic kidney disease (ACKD, defined as an estimated creatinine clearance of 30 ml/min or less). Its primary objective is to determine whether administration of high doses of three vitamins, folic acid, vitamin B6 and vitamin B12, to lower the high plasma homocysteine levels, will reduce all cause mortality. The secondary objectives are to examine whether the treatment lowers the incidence of myocardial infarction, stroke, amputation of a lower extremity, a composite of death and the foregoing three events, the plasma homocysteine level, and, in ESRD patients undergoing hemodialysis, thrombosis of the vascular access. A unique feature of this trial is that after initial evaluation at enrollment and one return visit the follow up is exclusively by phone (or, if necessary, by mail). The subject is contacted every three months throughout the duration of the study from a central location. The study drug is shipped to the patient from a central location rather supplied locally. In a two year enrollment period, 2006 patients are to be enrolled. The duration of the observation period is four to six years. Data will be stored and analyzed at a coordinating center. The study design has the power to detect a reduction in all cause mortality rate of 17%. Issues related to the unique features of the design of this study are discussed.
Collapse
Affiliation(s)
- Rex L Jamison
- Division of Nephrology, Stanford University School of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Korai M, Kitazato KT, Tada Y, Miyamoto T, Shimada K, Matsushita N, Kanematsu Y, Satomi J, Hashimoto T, Nagahiro S. Hyperhomocysteinemia induced by excessive methionine intake promotes rupture of cerebral aneurysms in ovariectomized rats. J Neuroinflammation 2016; 13:165. [PMID: 27349749 PMCID: PMC4924228 DOI: 10.1186/s12974-016-0634-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 06/21/2016] [Indexed: 12/31/2022] Open
Abstract
Background Hyperhomocysteinemia (HHcy) is associated with inflammation and a rise in the expression of matrix metalloproteinase-9 (MMP-9) in the vascular wall. However, the role of HHcy in the growth and rupture of cerebral aneurysms remains unclear. Methods Thirteen-week-old female Sprague-Dawley rats were subject to bilateral ovariectomy and ligation of the right common carotid artery and fed an 8 % high-salt diet to induce cerebral aneurysms. Two weeks later, they underwent ligation of the bilateral posterior renal arteries. They were divided into two groups and methionine (MET) was or was not added to their drinking water. In another set of experiments, the role of folic acid (FA) against cerebral aneurysms was assessed. Results During a 12-week observation period, subarachnoid hemorrhage due to aneurysm rupture was observed at the anterior communicating artery (AcomA) or the posterior half of the circle of Willis. HHcy induced by excessive MET intake significantly increased the incidence of ruptured aneurysms at 6–8 weeks. At the AcomA of rats treated with MET, we observed the promotion of aneurysmal growth and infiltration by M1 macrophages. Furthermore, the mRNA level of MMP-9, the ratio of MMP-9 to the tissue inhibitor of metalloproteinase-2, and the level of interleukin-6 were higher in these rats. Treatment with FA abolished the effect of MET, suggesting that the inflammatory response and vascular degradation at the AcomA is attributable to HHcy due to excessive MET intake. Conclusions We first demonstrate that in hypertensive ovariectomized rats, HHcy induced by excessive MET intake may be associated with the propensity of the aneurysm wall to rupture. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0634-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masaaki Korai
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan. .,Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA.
| | - Keiko T Kitazato
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Yoshiteru Tada
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Takeshi Miyamoto
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Kenji Shimada
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Nobuhisa Matsushita
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Yasuhisa Kanematsu
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Junichiro Satomi
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Tomoki Hashimoto
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Shinji Nagahiro
- Department of Neurosurgery, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| |
Collapse
|
91
|
Daulatzai MA. Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer's disease. J Neurosci Res 2016; 95:943-972. [PMID: 27350397 DOI: 10.1002/jnr.23777] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 02/06/2023]
Abstract
Aging, hypertension, diabetes, hypoxia/obstructive sleep apnea (OSA), obesity, vitamin B12/folate deficiency, depression, and traumatic brain injury synergistically promote diverse pathological mechanisms including cerebral hypoperfusion and glucose hypometabolism. These risk factors trigger neuroinflammation and oxidative-nitrosative stress that in turn decrease nitric oxide and enhance endothelin, Amyloid-β deposition, cerebral amyloid angiopathy, and blood-brain barrier disruption. Proinflammatory cytokines, endothelin-1, and oxidative-nitrosative stress trigger several pathological feedforward and feedback loops. These upstream factors persist in the brain for decades, upregulating amyloid and tau, before the cognitive decline. These cascades lead to neuronal Ca2+ increase, neurodegeneration, cognitive/memory decline, and Alzheimer's disease (AD). However, strategies are available to attenuate cerebral hypoperfusion and glucose hypometabolism and ameliorate cognitive decline. AD is the leading cause of dementia among the elderly. There is significant evidence that pathways involving inflammation and oxidative-nitrosative stress (ONS) play a key pathophysiological role in promoting cognitive dysfunction. Aging and several comorbid conditions mentioned above promote diverse pathologies. These include inflammation, ONS, hypoperfusion, and hypometabolism in the brain. In AD, chronic cerebral hypoperfusion and glucose hypometabolism precede decades before the cognitive decline. These comorbid disease conditions may share and synergistically activate these pathophysiological pathways. Inflammation upregulates cerebrovascular pathology through proinflammatory cytokines, endothelin-1, and nitric oxide (NO). Inflammation-triggered ONS promotes long-term damage involving fatty acids, proteins, DNA, and mitochondria; these amplify and perpetuate several feedforward and feedback pathological loops. The latter includes dysfunctional energy metabolism (compromised mitochondrial ATP production), amyloid-β generation, endothelial dysfunction, and blood-brain-barrier disruption. These lead to decreased cerebral blood flow and chronic cerebral hypoperfusion- that would modulate metabolic dysfunction and neurodegeneration. In essence, hypoperfusion deprives the brain from its two paramount trophic substances, viz., oxygen and nutrients. Consequently, the brain suffers from synaptic dysfunction and neuronal degeneration/loss, leading to both gray and white matter atrophy, cognitive dysfunction, and AD. This Review underscores the importance of treating the above-mentioned comorbid disease conditions to attenuate inflammation and ONS and ameliorate decreased cerebral blood flow and hypometabolism. Additionally, several strategies are described here to control chronic hypoperfusion of the brain and enhance cognition. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mak Adam Daulatzai
- Sleep Disorders Group, EEE Dept/MSE, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
92
|
Deminice R, Ribeiro DF, Frajacomo FTT. The Effects of Acute Exercise and Exercise Training on Plasma Homocysteine: A Meta-Analysis. PLoS One 2016; 11:e0151653. [PMID: 26986570 PMCID: PMC4795785 DOI: 10.1371/journal.pone.0151653] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/02/2016] [Indexed: 12/29/2022] Open
Abstract
Background Although studies have demonstrated that physical exercise alters homocysteine levels in the blood, meta-analyses of the effects of acute exercise and exercise training on homocysteine blood concentration have not been performed, especially regarding the duration and intensity of exercise, which could affect homocysteine levels differently. Objective The aim of this meta-analysis was to ascertain the effects of acute exercise and exercise training on homocysteine levels in the blood. Method A review was conducted according to the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses using the online databases PubMed, SPORTDiscus, and SciELO to identify relevant studies published through June 2015. Review Manager was used to calculate the effect size of acute exercise and exercise training using the change in Hcy plasmaserum concentration from baseline to post-acute exercise and trained vs. sedentary control groups, respectively. Weighted mean differences were calculated using random effect models. Results Given the abundance of studies, acute exercise trials were divided into two subgroups according to exercise volume and intensity, whereas the effects of exercise training were analyzed together. Overall, 22 studies with a total of 520 participants indicated increased plasma homocysteine concentration after acute exercise (1.18 μmol/L, 95% CI: 0.71 to 1.65, p < .01). Results of a subgroup analysis indicated that either long-term exercise of low-to-moderate intensity (1.39 μmol/L, 95% CI: 0.9 to 1.89, p < .01) or short-term exercise of high intensity (0.83 μmol/L, 95% CI: 0.19 to 1.40, p < .01) elevated homocysteine levels in the blood. Increased homocysteine induced by exercise was significantly associated with volume of exercise, but not intensity. By contrast, resistance training reduced plasma homocysteine concentration (-1.53 μmol/L, 95% CI: -2.77 to -0.28, p = .02), though aerobic training did not. The cumulative results of the seven studies with a total of 230 participants in exercise training analysis did not demonstrate a significant impact on homocysteine levels in the blood (-0.56 μmol/L, 95% CI: -1.61 to 0.50, p = .23). Conclusions Current evidence demonstrates that acute exercise increases homocysteine levels in the blood independent of exercise duration and intensity. Resistance, but not aerobic training decreases plasma homocysteine levels.
Collapse
Affiliation(s)
- Rafael Deminice
- Department of Physical Education, State University of Londrina, Londrina-PR, Brazil
- * E-mail:
| | - Diogo Farias Ribeiro
- Department of Physical Education, State University of Londrina, Londrina-PR, Brazil
| | | |
Collapse
|
93
|
Chernyavskiy I, Veeranki S, Sen U, Tyagi SC. Atherogenesis: hyperhomocysteinemia interactions with LDL, macrophage function, paraoxonase 1, and exercise. Ann N Y Acad Sci 2016; 1363:138-54. [PMID: 26849408 DOI: 10.1111/nyas.13009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/23/2015] [Accepted: 01/05/2016] [Indexed: 12/13/2022]
Abstract
Despite great strides in understanding the atherogenesis process, the mechanisms are not entirely known. In addition to diet, cigarette smoking, genetic predisposition, and hypertension, hyperhomocysteinemia (HHcy), an accumulation of the noncoding sulfur-containing amino acid homocysteine (Hcy), is a significant contributor to atherogenesis. Although exercise decreases HHcy and increases longevity, the complete mechanism is unclear. In light of recent evidence, in this review, we focus on the effects of HHcy on macrophage function, differentiation, and polarization. Though there is need for further evidence, it is most likely that HHcy-mediated alterations in macrophage function are important contributors to atherogenesis, and HHcy-countering strategies, such as nutrition and exercise, should be included in the combinatorial regimens for effective prevention and regression of atherosclerotic plaques. Therefore, we also included a discussion on the effects of exercise on the HHcy-mediated atherogenic process.
Collapse
Affiliation(s)
- Ilya Chernyavskiy
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Sudhakar Veeranki
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Utpal Sen
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
94
|
Selhub J, Troen AM. Sulfur amino acids and atherosclerosis: a role for excess dietary methionine. Ann N Y Acad Sci 2015; 1363:18-25. [PMID: 26647293 DOI: 10.1111/nyas.12962] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The homocysteine theory of arteriosclerosis received credence when it was shown that after a methionine load, circulating homocysteine-cysteine concentrations were higher in cardiovascular disease patients than in healthy controls. Subsequent studies showing associations between homocysteine and coronary artery disease, stroke and cognitive impairment, relied on small increases in homocysteine concentration unlike the very high homocysteine seen in the rare genetic disorders that lead to homocystinuria and much higher homocysteine levels. Subsequent studies in cell culture, animals, and humans showed that a variety of cardiovascular adverse effects of "high homocysteine" introduced either as a nonphysiological bolus or as a methionine load led to high homocysteine. We fed apolipoprotein E-deficient mice diets designed to achieve three conditions: (1) high methionine intake with normal blood homocysteine, (2) high methionine intake with B vitamin deficiency and hyperhomocysteinemia, and (3) normal methionine intake with both B vitamin deficiency and hyperhomocysteinemia. We found that the mice fed methionine-rich diets had significant atheromatous pathology in the aortic arch even with normal plasma homocysteine levels. Mice fed B vitamin-deficient diets developed severe hyperhomocysteinemia but without any increase in vascular pathology. Our findings suggest that even moderate increases in methionine intake are atherogenic in susceptible mice while high plasma homocysteine is not.
Collapse
Affiliation(s)
- Jacob Selhub
- Jean Mayer Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Aron M Troen
- Nutrition and Brain Health Laboratory, The Institute of Biochemistry Food and Nutrition Science, The Robert H. Smith Faculty of Agriculture Food and the Environment, The Hebrew University of Jerusalem, Israel
| |
Collapse
|
95
|
Griffin JL, Wang X, Stanley E. Does our gut microbiome predict cardiovascular risk? A review of the evidence from metabolomics. ACTA ACUST UNITED AC 2015; 8:187-91. [PMID: 25691688 DOI: 10.1161/circgenetics.114.000219] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Millions of microbes are found in the human gut, and are collectively referred as the gut microbiota. Recent studies have estimated that the microbiota genome contains 100-fold more genes than the host genome. These microbiota contribute to digestion by processing energy substrates unutilized by the host, with about half of the total genome of the gut microbiota being related to central carbon and amino acid metabolism as well as the biosynthesis of secondary metabolites. Therefore, the gut microbiome and its interaction with the host influences many aspects of health and disease, including the composition of biofluids such as urine and blood plasma. Metabolomics is uniquely suited to capture these functional host-microbe interactions. This review aims at providing an overview of recent metabolomics evidence of gut microbiota-host metabolic interactions with a specific focus on cardiovascular disease and related aspects of the metabolic syndrome. Furthermore, the emphasis is given on the complexities of translating these metabolite signatures as potential clinical biomarkers, as the composition and activity of gut microbiome change with many factors, particularly with diet, with special reference to trimethylamine-oxide.
Collapse
Affiliation(s)
- Julian L Griffin
- From the Department of Lipid Profiling and Signalling, MRC Human Nutrition Research, Elsie Widdowson Laboratory (J.L.G., X.W., E.S.); and Department of Biochemistry, Cambridge Systems Biology Centre (J.L.G., X.W.), University of Cambridge, Cambridge, United Kingdom.
| | - Xinzhu Wang
- From the Department of Lipid Profiling and Signalling, MRC Human Nutrition Research, Elsie Widdowson Laboratory (J.L.G., X.W., E.S.); and Department of Biochemistry, Cambridge Systems Biology Centre (J.L.G., X.W.), University of Cambridge, Cambridge, United Kingdom
| | - Elizabeth Stanley
- From the Department of Lipid Profiling and Signalling, MRC Human Nutrition Research, Elsie Widdowson Laboratory (J.L.G., X.W., E.S.); and Department of Biochemistry, Cambridge Systems Biology Centre (J.L.G., X.W.), University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
96
|
Hildebrandt W, Sauer R, Bonaterra G, Dugi KA, Edler L, Kinscherf R. Oral N-acetylcysteine reduces plasma homocysteine concentrations regardless of lipid or smoking status. Am J Clin Nutr 2015; 102:1014-24. [PMID: 26447155 DOI: 10.3945/ajcn.114.101964] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 08/27/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Elevated total plasma homocysteine (tHcy) is considered to be an independent cardiovascular disease risk factor, although tHcy lowering by B-vitamins improves only certain clinical endpoints. N-acetylcysteine (NAC), a thiol-containing antioxidant, acutely lowers tHcy and possibly also blood pressure. However, to our knowledge, at present no conclusive long-term evaluation exists that controls for factors such as hyperlipidemia, smoking, medication, and disease stage, all of which affect the thiol redox state, including tHcy. OBJECTIVE We reanalyzed 2 double-blind, placebo-controlled trials in unmedicated middle-aged men, one in a hyperlipidemic group (HYL group; n = 40) and one in a normolipidemic group (NOL group; n = 42), each stratified for smokers and nonsmokers. DESIGN We evaluated the effect of 4 wk of oral NAC (1.8 g/d) on tHcy (primary endpoint), plasma thiol (cysteine), and intracellular glutathione concentrations as well as on blood pressure. The HYL group had total cholesterol >220 mg/dL or triglycerides >150 mg/dL. RESULTS NAC treatment significantly (P = 0.001, multivariate analysis of variance for repeated measures) lowered postabsorptive plasma concentrations of tHcy by -11.7% ± 3.0% (placebo: 4.1% ± 3.6%) while increasing those of cysteine by 28.1% ± 5.7% (placebo: 4.0% ± 3.4%) with no significant impact of hyperlipidemia or smoking. Moreover, NAC significantly decreased systolic (P = 0.003) and diastolic (P = 0.017) blood pressure within all subjects with a significant reduction in diastolic pressure in the HYL group (P = 0.008) but not in the NOL group. An explorative stepwise multiple regression analysis identified 1) post-treatment cysteine as well as 2) pretreatment tHcy and 3) albumin plasma concentrations as being significant contributors to tHcy reduction. CONCLUSIONS Four weeks of oral NAC treatment significantly decreased plasma tHcy concentrations, irrespective of lipid or smoking status, and lowered systolic blood pressure in both normolipidemic and hyperlipidemic men, with significant diastolic blood pressure reductions in the HYL group only. Increased oral intake of cysteine may therefore be considered for primary or secondary prevention of vascular events with regard to the 2 independent risk factors of hyperhomocysteinemia and arterial hypertension.
Collapse
Affiliation(s)
| | - Roland Sauer
- Immunochemistry and Department of Neurology, University Hospital Erlangen, Erlangen, Germany
| | | | - Klaus A Dugi
- Internal Medicine I, University of Heidelberg, Heidelberg, Germany; Departments of
| | - Lutz Edler
- Biostatistics, Deutsches Krebsforschungszentrum, Heidelberg, Germany; and
| | | |
Collapse
|
97
|
Vilas-Boas W, Veloso Cerqueira BA, Figueiredo CVB, Santiago RP, da Guarda CC, Pitanga TN, Santana SS, Dias Zanette AM, Goncalves MDS. Association of homocysteine and inflammatory-related molecules in sickle cell anemia. ACTA ACUST UNITED AC 2015; 21:126-31. [PMID: 26334689 DOI: 10.1179/1607845415y.0000000048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Investigate the role of homocysteine (Hcy), Th17-related cytokines, and adhesion molecules in the inflammatory state seen in the sickle cell anemia (SCA). METHODS We studied the Hcy, interleukin (IL)-17, and transforming growth factor β (TGF-β) cytokine levels of 62 SCA patients, as well as the expression levels of inflammatory and endothelial activation markers. RESULTS We found significant associations between Hcy levels and increased expression of IL-17 and TGF-β among SCA patients, and a positive significant correlation between Hcy and soluble vascular cellular adhesion molecules (sVCAM). SCA individuals had raised IL-17 levels when compared with controls. DISCUSSION These results suggest a possible role of Hyc in the induction of TGF-β and IL-17. Other authors proposed that Hcy may contribute to the initiation and progression of vascular disease by monocyte activation, resulting in the secretion of cytokines that amplify the inflammatory response. The role of Hcy in cytokine production and oxidative stress in the endothelium may explain the increase of sVCAM expression and, the vascular activation currently described among the SCA individuals with the highest Hcy serum levels. The chronic inflammation was observed in hyperhomocysteinemic mice, with an increased expression of VCAM-1 and plasma levels of tumor necrosis factor-alpha, showing an association of this inflammatory molecule and vascular changes. CONCLUSION Our findings suggest that the increased levels of IL-17,Hcy and sVCAM contributes contributes to the vascular inflammation and activation presented by SCA patients, which probably have an important role in vaso-occlusion. On the basis of the presented data, IL-17 and Hcy might be considered as important components in the pathogenesis of SCA.
Collapse
Affiliation(s)
| | - Bruno Antonio Veloso Cerqueira
- a Centro de Pesquisas Gonçalo Moniz/FIOCRUZ , Salvador , Brazil.,b Universidade do Estado da Bahia , Salvador , Bahia , Brazil
| | - Camylla V B Figueiredo
- a Centro de Pesquisas Gonçalo Moniz/FIOCRUZ , Salvador , Brazil.,c Universidade Federal da Bahia , Salvador , Brazil
| | - Rayra Pereira Santiago
- a Centro de Pesquisas Gonçalo Moniz/FIOCRUZ , Salvador , Brazil.,c Universidade Federal da Bahia , Salvador , Brazil
| | - Caroline C da Guarda
- a Centro de Pesquisas Gonçalo Moniz/FIOCRUZ , Salvador , Brazil.,c Universidade Federal da Bahia , Salvador , Brazil
| | - Thassila Nogueira Pitanga
- a Centro de Pesquisas Gonçalo Moniz/FIOCRUZ , Salvador , Brazil.,c Universidade Federal da Bahia , Salvador , Brazil
| | - Sanzio Silva Santana
- a Centro de Pesquisas Gonçalo Moniz/FIOCRUZ , Salvador , Brazil.,c Universidade Federal da Bahia , Salvador , Brazil
| | | | - Marilda de Souza Goncalves
- a Centro de Pesquisas Gonçalo Moniz/FIOCRUZ , Salvador , Brazil.,c Universidade Federal da Bahia , Salvador , Brazil
| |
Collapse
|
98
|
Lai WKC, Kan MY. Homocysteine-Induced Endothelial Dysfunction. ANNALS OF NUTRITION AND METABOLISM 2015. [DOI: 10.1159/000437098] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This review discussed and in particular emphasis the potential cellular pathways and the biological processes involved that lead to homocysteine-induced endothelial dysfunction, in particular in the impaired endothelial dependent dilatation aspect. Hyperhomocysteinemia is an independent cardiovascular risk factor that has been associated with atherosclerotic vascular diseases and ischemic heart attacks. The potential mechanisms by which elevated plasma homocysteine level leads to reduction in nitric oxide bioavailability include the disruptive uncoupling of nitric oxide synthase activity and quenching of nitric oxide by oxidative stress, the enzymatic inhibition by asymmetric dimethylarginine, endoplasmic reticulum stress with eventual endothelial cell apoptosis, and chronic inflammation/prothrombotic conditions. Homocysteine-induced endothelial dysfunction presumably affecting the bioavailability of the potent vasodilator ‘nitric oxide', and such dysfunction can easily be monitor by flow-mediated dilation method using ultrasound. Understanding the mechanisms by which plasma homocysteine alter endothelial nitric oxide production is therefore essential in the comprehension of homocysteine-induced impairment of endothelial dependent dilatation, and its association of cardiovascular risk and its pathophysiology.
Collapse
|
99
|
Homocysteine facilitates LOX-1 activation and endothelial death through the PKCβ and SIRT1/HSF1 mechanism: relevance to human hyperhomocysteinaemia. Clin Sci (Lond) 2015; 129:477-87. [DOI: 10.1042/cs20150127] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/15/2015] [Indexed: 12/20/2022]
Abstract
Our study demonstrates a new direction for LOX-1 regulation by modulating the PKCβ/NADPH oxidase/SIRT1/HSF1 pathway, which affects hyperhomocysteinaemia-induced endothelial cell dysfunction and apoptosis.
Collapse
|
100
|
Rane G, Koh WP, Kanchi MM, Wang R, Yuan JM, Wang X. Association Between Leukocyte Telomere Length and Plasma Homocysteine in a Singapore Chinese Population. Rejuvenation Res 2015; 18:203-10. [PMID: 25546508 DOI: 10.1089/rej.2014.1617] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
RATIONALE Leukocyte telomere length (LTL) and plasma homocysteine (HCY) have been independently associated with cardiovascular disease (CVD) morbidity and mortality. However, few studies have investigated the association between LTL and HCY levels. OBJECTIVE This study investigated the association of LTL with CVD risk factors, including HCY, in an overt CVD-free Singapore Chinese population comprised of middle aged and elderly, the age group at risk of developing CVD. APPROACH The association of plasma HCY and other CVD biomarkers with LTL were assessed in 100 samples drawn from the Singapore Chinese Health Study (SCHS). SCHS, a population-based cohort, recruited Chinese individuals, aged 45-74 years, between 1993 and 1998. Questionnaire data were collected via face-to-face interviews. Known CVD biomarkers were measured from the blood collected at the time of recruitment, and LTL was measured using the conventional Southern blot method. RESULTS After adjustment for age, gender, smoking status, education, and dialect, LTL was found to be inversely associated with plasma HCY levels (p for trend=0.014). Serum urate showed a weak association (p for trend=0.056). Other CVD risk factors and nutrients, namely total cholesterol, low-density lipoprotein (LDL), triglycerides and creatinine, high-density lipoprotein (HDL), folate, and vitamin B6 showed the expected trend with LTL, but did not reach statistical significance. CONCLUSION LTL displayed an inverse association with plasma HCY. This LTL-HCY inverse association in subjects lacking obvious cardiovascular events suggests that telomere length may be an intermediary in the biological mechanism by which elevated HCY leads to CVD.
Collapse
Affiliation(s)
- Grishma Rane
- 1 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System , Singapore, Singapore
| | - Woon-Puay Koh
- 2 Duke-National University of Singapore Graduate Medical School , Singapore, Singapore .,3 Saw Swee Hock School of Public Health, National University of Singapore , Singapore
| | - Madhu Mathi Kanchi
- 1 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System , Singapore, Singapore
| | - Renwei Wang
- 4 Cancer Epidemiology, Prevention and Control Program, The University of Pittsburgh Cancer Institute , Pittsburgh, Pennsylvania
| | - Jian-Min Yuan
- 4 Cancer Epidemiology, Prevention and Control Program, The University of Pittsburgh Cancer Institute , Pittsburgh, Pennsylvania.,5 Department of Epidemiology, University of Pittsburgh Graduate School of Public Health , Pittsburgh, Pennsylvania
| | - Xueying Wang
- 1 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System , Singapore, Singapore .,6 National University Cancer Institute of Singapore (NCIS) , Singapore
| |
Collapse
|