1
|
Rajpoot A, Aggarwal T, Sharma V. Unraveling the enigma of cardiac damage caused by lead: Understanding the intricate relationship between oxidative stress and other multifactorial mechanisms. Toxicology 2024; 509:153984. [PMID: 39481524 DOI: 10.1016/j.tox.2024.153984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Lead (Pb) exposure remains a pressing concern in the realm of public health, with a mounting body of evidence underscoring its adverse impact on cardiovascular well-being. The exposure to Lead instigates the production of reactive oxygen species (ROS), leading to consequential cellular and physiological damage and a perturbation in redox equilibrium. The resultant oxidative stress, induced by ROS, disrupts endothelial functionality, propagates inflammatory processes, and initiates vascular remodeling, collectively contributing to the advancement of cardiovascular diseases (CVDs). The objective of this current review is to comprehensively expound upon the intricate mechanisms through which Lead induced toxicity affects cardiac cells. Additionally, it briefly addresses the ramifications of Lead exposure on the development of three interconnected cardiovascular conditions: atherosclerosis, hypertension, and myocardial infarction. Furthermore, the discourse delves into the specific repercussions of Lead exposure on lipid metabolism, blood pressure regulation, and cardiac performance, culminating in the initiation and progression of atherosclerotic plaque formation, elevated blood pressure, and an augmented risk of myocardial infarction. By understanding these intricate mechanisms, targeted interventions may be devised to counteract the deleterious effects of Lead on cardiovascular health. Thus, this review offers novel avenues for preventive and therapeutic strategies, ultimately serving to alleviate the burden of cardiovascular diseases associated with Lead toxicity.
Collapse
Affiliation(s)
- Anjali Rajpoot
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India.
| | - Tanya Aggarwal
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India.
| | - Veena Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India.
| |
Collapse
|
2
|
Nucera S, Serra M, Caminiti R, Ruga S, Passacatini LC, Macrì R, Scarano F, Maiuolo J, Bulotta R, Mollace R, Bosco F, Guarnieri L, Oppedisano F, Ilari S, Muscoli C, Palma E, Mollace V. Non-essential heavy metal effects in cardiovascular diseases: an overview of systematic reviews. Front Cardiovasc Med 2024; 11:1332339. [PMID: 38322770 PMCID: PMC10844381 DOI: 10.3389/fcvm.2024.1332339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
Introduction Cardiovascular diseases (CVDs) are the most important cause of premature death and disability worldwide. Environmental degradation and cardiovascular diseases are two keys to health challenges, characterized by a constant evolution in an industrialized world that exploits natural resources regardless of the consequences for health. The etiological risk factors of CVDs are widely known and include dyslipidemia, obesity, diabetes, and chronic cigarette consumption. However, one component that is often underestimated is exposure to heavy metals. The biological perspective explains that different metals play different roles. They are therefore classified into essential heavy metals, which are present in organisms where they perform important vital functions, especially in various physiological processes, or non-essential heavy metals, with a no biological role but, nonetheless, remain in the environment in which they are absorbed. Although both types of metal ions are many times chemically similar and can bind to the same biological ligands, the attention given today to nonessential metals in several eukaryotic species is starting to raise strong concerns due to an exponential increase in their concentrations. The aim of this systematic review was to assess possible correlations between exposure to nonessential heavy metals and increased incidence of cardiovascular disease, reporting the results of studies published in the last 5 years through March 2023. Methods The studies includes reviews retrieved from PubMed, Medline, Embase, and Web of Science databases, in accordance with the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) statement and following the PICO (Population Intervention Comparison Outcome Population) framework. Results Eight reviews, including a total of 153 studies, were identified. Seven of these review enlighted the association between CVDs and non-essential heavy metals chronic exposure. Discussion It is evident that exposure to heavy metals represent a risk factor for CVDs onset. However, further studies are needed to better understand the effects caused by these metals.
Collapse
Affiliation(s)
- Saverio Nucera
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Maria Serra
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Rosamaria Caminiti
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Stefano Ruga
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | | | - Roberta Macrì
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Federica Scarano
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Jessica Maiuolo
- Department of Health Sciences, Laboratory of Pharmaceutical Biology, Institute of Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Rosamaria Bulotta
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Rocco Mollace
- Department of Systems Medicine, University “Tor Vergata” of Rome, Rome, Italy
| | - Francesca Bosco
- Science of Health Department, Section of Pharmacology, School of Medicine, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Lorenza Guarnieri
- Science of Health Department, Section of Pharmacology, School of Medicine, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Francesca Oppedisano
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Sara Ilari
- Physiology and Pharmacology of Pain, IRCCS San Raffaele Roma, Rome, Italy
| | - Carolina Muscoli
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Physiology and Pharmacology of Pain, IRCCS San Raffaele Roma, Rome, Italy
| | - Ernesto Palma
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Department of Health Sciences, Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health (IRC-FSH), University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Vincenzo Mollace
- Department of Health Sciences, Instituteof Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Renato Dulbecco Institute, Catanzaro, Italy
| |
Collapse
|
3
|
Keshawarz A, Joehanes R, Ma J, Lee GY, Costeira R, Tsai PC, Masachs OM, Bell JT, Wilson R, Thorand B, Winkelmann J, Peters A, Linseisen J, Waldenberger M, Lehtimäki T, Mishra PP, Kähönen M, Raitakari O, Helminen M, Wang CA, Melton PE, Huang RC, Pennell CE, O’Sullivan TA, Ochoa-Rosales C, Voortman T, van Meurs JB, Young KL, Graff M, Wang Y, Kiel DP, Smith CE, Jacques PF, Levy D. Dietary and supplemental intake of vitamins C and E is associated with altered DNA methylation in an epigenome-wide association study meta-analysis. Epigenetics 2023; 18:2211361. [PMID: 37233989 PMCID: PMC10228397 DOI: 10.1080/15592294.2023.2211361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Dietary intake of antioxidants such as vitamins C and E protect against oxidative stress, and may also be associated with altered DNA methylation patterns. METHODS We meta-analysed epigenome-wide association study (EWAS) results from 11,866 participants across eight population-based cohorts to evaluate the association between self-reported dietary and supplemental intake of vitamins C and E with DNA methylation. EWAS were adjusted for age, sex, BMI, caloric intake, blood cell type proportion, smoking status, alcohol consumption, and technical covariates. Significant results of the meta-analysis were subsequently evaluated in gene set enrichment analysis (GSEA) and expression quantitative trait methylation (eQTM) analysis. RESULTS In meta-analysis, methylation at 4,656 CpG sites was significantly associated with vitamin C intake at FDR ≤ 0.05. The most significant CpG sites associated with vitamin C (at FDR ≤ 0.01) were enriched for pathways associated with systems development and cell signalling in GSEA, and were associated with downstream expression of genes enriched in the immune response in eQTM analysis. Furthermore, methylation at 160 CpG sites was significantly associated with vitamin E intake at FDR ≤ 0.05, but GSEA and eQTM analysis of the top most significant CpG sites associated with vitamin E did not identify significant enrichment of any biological pathways investigated. CONCLUSIONS We identified significant associations of many CpG sites with vitamin C and E intake, and our results suggest that vitamin C intake may be associated with systems development and the immune response.
Collapse
Affiliation(s)
| | - Roby Joehanes
- Framingham Heart Study, Framingham, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiantao Ma
- Framingham Heart Study, Framingham, Framingham, MA, USA
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Gha Young Lee
- Framingham Heart Study, Framingham, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ricardo Costeira
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Olatz M. Masachs
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Rory Wilson
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, Neuherberg, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, München, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Chair of Neurogenetics, School of Medicine, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, München, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Chair of Epidemiology, Medical Faculty, Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), München Heart Alliance, Munich, Germany
| | - Jakob Linseisen
- Chair of Epidemiology, University Augsburg at University Hospital Augsburg, Augsburg, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), München Heart Alliance, Munich, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Pashupati P. Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Mika Helminen
- Tays Research Services, Tampere University Hospital, Tampere, Finland
- Faculty of Social Sciences, Health Sciences, Tampere University, Tampere, Finland
| | - Carol A. Wang
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Phillip E. Melton
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Rae-Chi Huang
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Perth, Australia
| | - Craig E. Pennell
- Faculty of Social Sciences, Health Sciences, Tampere University, Tampere, Finland
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | | | - Carolina Ochoa-Rosales
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Centro de Vida Saludable, Universidad de Concepción, Concepción, Chile
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Joyce B.J. van Meurs
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Kristin L. Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Hebrew Senior Life, Chapel Hill, North Carolina, USA
| | - Misa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Hebrew Senior Life, Chapel Hill, North Carolina, USA
| | - Yujie Wang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Hebrew Senior Life, Chapel Hill, North Carolina, USA
| | - Douglas P. Kiel
- Department of Medicine, Beth Israel Deaconess Medical Center, Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Caren E. Smith
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Paul F. Jacques
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Daniel Levy
- Framingham Heart Study, Framingham, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Renu K, Mukherjee AG, Wanjari UR, Vinayagam S, Veeraraghavan VP, Vellingiri B, George A, Lagoa R, Sattu K, Dey A, Gopalakrishnan AV. Misuse of Cardiac Lipid upon Exposure to Toxic Trace Elements-A Focused Review. Molecules 2022; 27:5657. [PMID: 36080424 PMCID: PMC9457865 DOI: 10.3390/molecules27175657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022] Open
Abstract
Heavy metals and metalloids like cadmium, arsenic, mercury, and lead are frequently found in the soil, water, food, and atmosphere; trace amounts can cause serious health issues to the human organism. These toxic trace elements (TTE) affect almost all the organs, mainly the heart, kidney, liver, lungs, and the nervous system, through increased free radical formation, DNA damage, lipid peroxidation, and protein sulfhydryl depletion. This work aims to advance our understanding of the mechanisms behind lipid accumulation via increased free fatty acid levels in circulation due to TTEs. The increased lipid level in the myocardium worsens the heart function. This dysregulation of the lipid metabolism leads to damage in the structure of the myocardium, inclusive fibrosis in cardiac tissue, myocyte apoptosis, and decreased contractility due to mitochondrial dysfunction. Additionally, it is discussed herein how exposure to cadmium decreases the heart rate, contractile tension, the conductivity of the atrioventricular node, and coronary flow rate. Arsenic may induce atherosclerosis by increasing platelet aggregation and reducing fibrinolysis, as exposure interferes with apolipoprotein (Apo) levels, resulting in the rise of the Apo-B/Apo-A1 ratio and an elevated risk of acute cardiovascular events. Concerning mercury and lead, these toxicants can cause hypertension, myocardial infarction, and carotid atherosclerosis, in association with the generation of free radicals and oxidative stress. This review offers a complete overview of the critical factors and biomarkers of lipid and TTE-induced cardiotoxicity useful for developing future protective interventions.
Collapse
Affiliation(s)
- Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Sathishkumar Vinayagam
- Department of Biotechnology, PG Extension Centre, Periyar University, Dharmapuri 636701, Tamil Nadu, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Ricardo Lagoa
- School of Technology and Management, Polytechnic Institute of Leiria, 2411-901 Leiria, Portugal
- Applied Molecular Biosciences Unit, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Kamaraj Sattu
- Department of Biotechnology, PG Extension Centre, Periyar University, Dharmapuri 636701, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
5
|
Pal P, De A, Roychowdhury T, Mukhopadhyay PK. Vitamin C and E supplementation can ameliorate NaF mediated testicular and spermatozoal DNA damages in adult Wistar rats. Biomarkers 2022; 27:361-374. [PMID: 35232301 DOI: 10.1080/1354750x.2022.2048891] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
OBJECTIVE Present study was designed to explore the efficacy of vitamin C and E (VC&VE) against fluoride mediated testicular, epididymal and spermatozoal anomalies. MATERIALS AND METHODS Thirty two adult Wistar rats were divided into four groups. Group-I was control; Group-II received sodium fluoride (NaF) at 15 mg/kg/day dose; Group-III was provided with VC (200 mg/kg/day) and VE (400 mg/kg/day) plus NaF; Group-IV received only VC&VE. Structural integrity and oxidative stress markers (superoxide dismutase, catalase, malondialdehyde and protein carbonyl) of testis and epididymis were assessed. Spermatozoal parameters (count, motility, viability and hypo-osmotic swelling) were evaluated. Testicular functional maker enzymes (acid phosphatase, alkaline phosphatase and lactate dehydrogenase) were also assessed. Integrity of testicular and spermatozoal DNA were evaluated. Testicular fluoride content was measured. RESULT Fluoride induced structural changes and alterations of oxidative stress markers were observed in testis and epididymis. Spermatozoal potentials were altered and reduced activities of testicular functional marker enzymes were observed. Fluoride caused testicular and spermatozoal DNA damages. VC&VE supplementation resulted in protectionfrom all fluoride mediated alterations and helped in attenuating testicular fluoride accumulation. CONCLUSION Antioxidant properties of VC&VE ameliorated fluoride mediated reproductive damages but only supplementation did not exhibit any notable effect compared to control rats.
Collapse
Affiliation(s)
- Priyankar Pal
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Ayan De
- School of Environmental Studies, Jadavpur University, Kolkata, India
| | | | | |
Collapse
|
6
|
Mabrouk A, Bel Hadj Salah I, Chaieb W, Ben Cheikh H. Protective effect of thymoquinone against lead-induced hepatic toxicity in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:12206-12215. [PMID: 26971798 DOI: 10.1007/s11356-016-6419-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 03/03/2016] [Indexed: 06/05/2023]
Abstract
Lead (Pb) intoxication is a worldwide health problem which frequently affects the liver. This study was carried out to investigate the potential protective effect of thymoquinone (TQ), the major active ingredient of volatile oil of Nigella sativa seeds, against Pb-induced liver damage. Adult male rats were randomized into four groups: Control group received no treatment, Pb group was exposed to 2000 ppm Pb acetate in drinking water, Pb-TQ group was cotreated with Pb plus TQ (5 mg/kg/day, per orally), and TQ group receiving only TQ. All treatments were applied for 5 weeks. Results indicated that Pb exposure increased hepatic Pb content, damaged hepatic histological structure (necrotic foci, hepatic strands disorganization, hypertrophied hepatocytes, cytoplasmic vacuolization, cytoplasmic loss, chromatin condensation, mononuclear cell infiltration, congestion, centrilobular swelling), and changed liver function investigated by plasma biochemical parameters (AST, ALT, ALP, γ-GT, LDH). Pb treatment also decreased total antioxidant status level and increased lipid peroxidation in the liver. Supplementation with TQ remarkably improved the Pb-induced adverse effects without significantly reducing the metal accumulation in the liver. In conclusion, our results indicate, for the first time, a protective effect of TQ against Pb-induced hepatotoxicity and suggest that this component might be clinically useful in Pb intoxication.
Collapse
Affiliation(s)
- Aymen Mabrouk
- Laboratory of Histology and Cytogenetic (Research Unit of Genetic, Genotoxicity and Child Disease UR 12 ES 10), Faculty of Medicine, University of Monastir, Street Avicenna, Monastir, 5019, Tunisia.
| | - Imen Bel Hadj Salah
- Laboratory of Histology and Cytogenetic (Research Unit of Genetic, Genotoxicity and Child Disease UR 12 ES 10), Faculty of Medicine, University of Monastir, Street Avicenna, Monastir, 5019, Tunisia
| | - Wafa Chaieb
- Laboratory of Histology and Cytogenetic (Research Unit of Genetic, Genotoxicity and Child Disease UR 12 ES 10), Faculty of Medicine, University of Monastir, Street Avicenna, Monastir, 5019, Tunisia
| | - Hassen Ben Cheikh
- Laboratory of Histology and Cytogenetic (Research Unit of Genetic, Genotoxicity and Child Disease UR 12 ES 10), Faculty of Medicine, University of Monastir, Street Avicenna, Monastir, 5019, Tunisia
| |
Collapse
|
7
|
Lee W, Yoon JH, Roh J, Lee S, Seok H, Lee JH, Jung PK, Rhie J, Won JU. The association between low blood lead levels and the prevalence of prehypertension among nonhypertensive adults in Korea. Am J Hum Biol 2016; 28:729-35. [DOI: 10.1002/ajhb.22857] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 12/10/2015] [Accepted: 03/20/2016] [Indexed: 11/11/2022] Open
Affiliation(s)
- Wanhyung Lee
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| | - Jin-ha Yoon
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| | - Jaehoon Roh
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| | - Seunghyun Lee
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| | - Hongdeok Seok
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| | - June-hee Lee
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| | - Pil Kyun Jung
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| | - Jeongbae Rhie
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| | - Jong-uk Won
- Department of Preventive Medicine; The Institute for Occupational Health; Seoul South Korea
| |
Collapse
|
8
|
Karamian R, Komaki A, Salehi I, Tahmasebi L, Komaki H, Shahidi S, Sarihi A. Vitamin C reverses lead-induced deficits in hippocampal synaptic plasticity in rats. Brain Res Bull 2015; 116:7-15. [DOI: 10.1016/j.brainresbull.2015.05.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 04/11/2015] [Accepted: 05/08/2015] [Indexed: 12/11/2022]
|
9
|
Light and electron microscopic study of the toxic effect of prolonged lead exposure on the seminiferous tubules of albino rats and the possible protective effect of ascorbic acid. Food Chem Toxicol 2011; 49:734-43. [DOI: 10.1016/j.fct.2010.11.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 11/07/2010] [Accepted: 11/23/2010] [Indexed: 11/20/2022]
|
10
|
Sharma I, Dhaliwal LK, Saha SC, Sangwan S, Dhawan V. Role of 8-iso-prostaglandin F2alpha and 25-hydroxycholesterol in the pathophysiology of endometriosis. Fertil Steril 2009; 94:63-70. [PMID: 19324352 DOI: 10.1016/j.fertnstert.2009.01.141] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Revised: 01/09/2009] [Accepted: 01/26/2009] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To investigate the involvement of 8-iso-PGF(2alpha) and 25-hydroxycholesterol (25-OH-Chol) in the pathophysiology of endometriosis. DESIGN Observational case-control study using enzyme immunoassay and high-performance liquid chromatography (HPLC). SETTING Postgraduate Institute of Medical Education and Research. PATIENT(S) Forty-five women undergoing laparoscopy (n = 25), laparotomy (n = 19), or tubal ligation (n =1). INTERVENTION(S) Venipuncture and laparoscopic peritoneal fluid (PF) collection. MAIN OUTCOME MEASURE(S) The levels of 8-iso-PGF(2alpha) were determined both in urine and PF of all the patients using enzyme immunoassay. The levels of 25-OH-Chol were determined by using reversed phase HPLC both in the plasma and PF samples. Oxidative damage to DNA was assessed by agarose gel electrophoresis. RESULT(S) Significantly increased levels of 8-iso-PGF(2alpha) were observed both in urine and PF of women with endometriosis compared with control women. Similarly, higher levels of 25-OH-Chol were observed both in plasma and PF of patients compared with controls and the difference was statistically significant. A clear-cut tailing pattern was observed in DNA of patients with endometriosis, indicating significant DNA damage. CONCLUSION(S) Our observations implicate oxidative stress in the pathophysiology of endometriosis. For the first time, we demonstrate that 8-iso-PGF(2alpha) and oxysterols (the known promoters of steroidogenesis) might be the culprits in this disease.
Collapse
Affiliation(s)
- Indu Sharma
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | | | |
Collapse
|
11
|
Caylak E, Aytekin M, Halifeoglu I. Antioxidant effects of methionine, alpha-lipoic acid, N-acetylcysteine and homocysteine on lead-induced oxidative stress to erythrocytes in rats. EXPERIMENTAL AND TOXICOLOGIC PATHOLOGY : OFFICIAL JOURNAL OF THE GESELLSCHAFT FUR TOXIKOLOGISCHE PATHOLOGIE 2008; 60:289-94. [PMID: 18407480 DOI: 10.1016/j.etp.2007.11.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Accepted: 11/20/2007] [Indexed: 02/08/2023]
Abstract
Lead, widely used in industry, is a great environmental health problem. Many studies have examined its effects on the health of both humans and animals. Experimental studies have shown that sulphur-containing antioxidants have beneficial effects against the detrimental properties of lead. The present study was designed to investigate markers of oxidative stress (hemoglobin (Hb) in whole blood, malondialdehyde (MDA) in sera; superoxidase dismutase (SOD) and glutathione peroxidise (GSH-Px) in erythrocyte hemolysate and vitamins A and E in plasma) in rats given lead (2000ppm) with or without sulphur-containing antioxidants (l-methionine (Met) (100mg/kg/day), N-acetylcysteine (NAC) (800mg/kg/day), l-homocysteine (Hcy) (25mg/kg/day), lipoic acid (LA) (50mg/kg/day)) in their water for 5 weeks. In the lead group, Hb and plasma vitamin E levels were significantly lower whereas MDA levels were significantly higher compared to controls (p<0.05). Hb levels in lead-methionine and lead-LA groups were significantly higher than the lead group (p<0.01). MDA levels were reduced in all groups compared to the lead group (p<0.01). There was a decrease below control values in erythrocyte SOD (p<0.01) and GSH-Px (p<0.05) levels in the lead-LA group. Plasma vitamin A levels were significantly high in lead-methionine group compared to lead group (p<0.01). In conclusion, the data suggests that oxidative stress induced by lead is reduced by sulphur-containing compounds.
Collapse
Affiliation(s)
- Emrah Caylak
- Department of Biochemistry and Clinical Biochemistry, Firat University, Medical School, Elazig, Turkey
| | | | | |
Collapse
|
12
|
O'Toole TE, Conklin DJ, Bhatnagar A. Environmental risk factors for heart disease. REVIEWS ON ENVIRONMENTAL HEALTH 2008; 23:167-202. [PMID: 19119685 DOI: 10.1515/reveh.2008.23.3.167] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
In this review, we discuss current evidence linking environmental pollutants to cardiovascular disease (CVD). Extensive evidence indicates that environmental factors contribute to CVD risk, incidence, and severity. Migrant studies show that changes in the environment could substantially alter CVD risk in a genetically stable population. Additionally, CVD risk is affected by changes in nutritional and lifestyle choices. Recent studies in the field of environmental cardiology suggest that environmental toxins also influence CVD. Exposure to tobacco smoke is paradigmatic of such environmental risk and is strongly and positively associated with increased cardiovascular morbidity and mortality. In animal models of exposure, tobacco smoke induces endothelial dysfunction and prothrombotic responses and exacerbates atherogenesis and myocardial ischemic injury. Similar mechanism may be engaged by other pollutants or food constituents. Several large population-based studies indicate that exposure to fine or ultrafine particulate air pollution increases CVD morbidity and mortality, and the plausibility of this association is supported by data from animal studies. Exposure to other chemicals such as polyaromatic hydrocarbons, aldehydes, and metals has also been reported to elevate CVD risk by affecting atherogenesis, thrombosis, or blood pressure regulation. Maternal exposure to drugs, toxins, and infection has been linked with cardiac birth defects and premature CVD in later life. Collectively, the data support the notion that chronic environmental stress is an important determinant of CVD risk. Further work is required to assess the magnitude of this risk fully and to delineate specific mechanisms by which environmental toxins affect CVD.
Collapse
Affiliation(s)
- Timothy E O'Toole
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | | | | |
Collapse
|
13
|
Evaluation of the ability of a battery of three in vitro genotoxicity tests to discriminate rodent carcinogens and non-carcinogens. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2008; 654:114-32. [DOI: 10.1016/j.mrgentox.2008.05.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Revised: 05/03/2008] [Accepted: 05/09/2008] [Indexed: 11/24/2022]
|
14
|
Abstract
Lead is a ubiquitous environmental toxin that is capable of causing numerous acute and chronic illnesses. Population studies have demonstrated a link between lead exposure and subsequent development of hypertension (HTN) and cardiovascular disease. In vivo and in vitro studies have shown that chronic lead exposure causes HTN and cardiovascular disease by promoting oxidative stress, limiting nitric oxide availability, impairing nitric oxide signaling, augmenting adrenergic activity, increasing endothelin production, altering the renin-angiotensin system, raising vasoconstrictor prostaglandins, lowering vasodilator prostaglandins, promoting inflammation, disturbing vascular smooth muscle Ca(2+) signaling, diminishing endothelium-dependent vasorelaxation, and modifying the vascular response to vasoactive agonists. Moreover, lead has been shown to cause endothelial injury, impede endothelial repair, inhibit angiogenesis, reduce endothelial cell growth, suppress proteoglycan production, stimulate vascular smooth muscle cell proliferation and phenotypic transformation, reduce tissue plasminogen activator, and raise plasminogen activator inhibitor-1 production. Via these and other actions, lead exposure causes HTN and promotes arteriosclerosis, atherosclerosis, thrombosis, and cardiovascular disease. In conclusion, studies performed in experimental animals, isolated tissues, and cultured cells have provided compelling evidence that chronic exposure to low levels of lead can cause HTN, endothelial injury/dysfunction, arteriosclerosis, and cardiovascular disease. More importantly, these studies have elucidated the cellular and molecular mechanisms of lead's action on cardiovascular/renal systems, a task that is impossible to accomplish using clinical and epidemiological investigations alone.
Collapse
Affiliation(s)
- Nosratola D Vaziri
- Division of Nephrology and Hypertension, UCI Medical Center, 101 The City Dr., Bldg. 53, Rm. 125, Rt. 81, Orange, CA 92868, USA.
| |
Collapse
|
15
|
Robles HV, Romo E, Sanchez-Mendoza A, Rios A, Soto V, Avila-Casado MC, Medina A, Escalante B. Lead exposure effect on angiotensin II renal vasoconstriction. Hum Exp Toxicol 2007; 26:499-507. [PMID: 17698945 DOI: 10.1177/0960327106077597] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Low levels of chronic lead exposure can produce hypertension and endothelial dysfunction, which could be associated with oxidative stress, changes in vascular tone and an imbalance of endothelial-derived vasoconstriction and vasodilator factors. The aim was to investigate the effect of chronic lead-exposure on angiotensin II-induced vasoconstriction in isolated perfused kidney and microvessels. Male Wistar rats (230-250 g) were treated for 12 weeks with lead acetate (100 ppm, Pbgroup) or pure water (control group). We evaluated the vascular reactivity in the kidneys and renal microvessels in the presence and absence of N(omega)-nitro-L-arginine methyl ester (L-NAME) in both groups. The nitrite concentration in renal perfusate was measured as an index of NO released, renal abundance of 3-nitrotyrosine was measured as well as endothelial NO synthase (eNOS) expression. Oxidative stress was measured by using the oxidative fluorescence dye dihydroethidium (DHE) to evaluate in situ production of superoxide and identified by confocal microscopy. Lead-exposure significantly increased blood pressure, eNOS protein expression, oxidative stress and vascular reactivity to angiotensin II. L-NAME potentiated vascular response to angiotensin II in control group but had no effect on the Pb-group. Nitrites released from the kidney of lead-group was lower compared to the control group while 3-nitrotyrosine was higher. This data suggest that lead-induced hypertension could be caused partially by an altered NOsystem.
Collapse
Affiliation(s)
- Hilda Vargas Robles
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), México City, México
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Vaziri ND, Khan M. Interplay of reactive oxygen species and nitric oxide in the pathogenesis of experimental lead-induced hypertension. Clin Exp Pharmacol Physiol 2007; 34:920-5. [PMID: 17645641 DOI: 10.1111/j.1440-1681.2007.04644.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
1. Lead is a common environmental and industrial toxin that can cause a variety of acute and chronic illnesses. For example, chronic exposure to low levels of lead has been shown to raise arterial pressure and promote renal and cardiovascular complications. 2. Several mechanisms have been identified by which chronic lead exposure can cause hypertension and cardiovascular disease. In recent years, increasing evidence has emerged pointing to the role of oxidative stress as a major mediator of lead-induced hypertension. 3. The present article provides an overview of the published studies on this subject.
Collapse
Affiliation(s)
- N D Vaziri
- Division of Nephrology and Hypertension, University of California, Irvine, California, USA.
| | | |
Collapse
|
17
|
Roncal C, Mu W, Reungjui S, Kim KM, Henderson GN, Ouyang X, Nakagawa T, Johnson RJ. Lead, at low levels, accelerates arteriolopathy and tubulointerstitial injury in chronic kidney disease. Am J Physiol Renal Physiol 2007; 293:F1391-6. [PMID: 17715263 DOI: 10.1152/ajprenal.00216.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chronic lead exposure has been epidemiologically linked with hypertension and renal disease. Clinical studies suggest that low lead levels may contribute to renal progression. However, experimental studies have not examined whether low levels of lead accelerate progression in experimental chronic renal disease. Sprague-Dawley rats were administered lead (L; 150 ppm in drinking water, n = 16) for 4 wk, followed by remnant kidney (RK) surgery with continuation of lead for an additional 12 wk; control rats (n = 9) were treated similarly but did not receive lead. Lead treatment was well tolerated and resulted in modest elevations in whole blood lead levels (26.4 +/- 4.5 vs. 1 +/- 0 mug/dl, week 16, P < 0.001). Lead treatment was associated with higher systolic blood pressure (P < 0.05) and worse renal function (creatinine clearance 1.4 +/- 0.4 vs. 1.8 +/- 0.5 ml/min, RK+L vs. RK, P < 0.05), and with a tendency for greater proteinuria (6.6 +/- 6.1 vs. 3.6 +/- 1.5 mg protein/mg creatinine, RK+L vs. RK, P = 0.08). While glomerulosclerosis tended to be worse in lead-treated rats (37.6 +/- 11 vs. 28.8 +/- 2.3%, RK+L vs. RK, P = 0.06), the most striking finding was the development of worse arteriolar disease (P < 0.05), peritubular capillary loss (P < 0.05), tubulointerstitial damage, and macrophage infiltration (P < 0.05) in association with significantly increased renal expression of monocyte chemoattractant protein-1 mRNA. In conclusion, lead accelerates chronic renal disease, primarily by raising blood pressure and accelerating microvascular and tubulointerstitial injury.
Collapse
Affiliation(s)
- Carlos Roncal
- Division of Nephrology, Hypertension, and Transplantation, University of Florida, Gainesville, FL 32610-0224, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Dhawan V, Jain S. Garlic supplementation prevents oxidative DNA damage in essential hypertension. Mol Cell Biochem 2006; 275:85-94. [PMID: 16335787 DOI: 10.1007/s11010-005-0824-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Oxygen-free radicals and other oxygen/nitrogen species are constantly generated in the human body. Most are intercepted by antioxidant defences and perform useful metabolic roles, whereas others escape to damage biomolecules like DNA, lipids and proteins. Garlic has been shown to contain antioxidant phytochemicals that prevent oxidative damage. These include unique water-soluble organosulphur compounds, lipid-soluble organosulphur compounds and flavonoids. Therefore, in the present study, we have tried to explore the antioxidant effect of garlic supplementation on oxidative stress-induced DNA damage, nitric oxide (NO) and superoxide generation and on the total antioxidant status (TAS) in patients of essential hypertension (EH). Twenty patients of EH as diagnosed by JNC VI criteria (Group I) and 20 age and sex-matched normotensive controls (Group II) were enrolled in the study. Both groups were given garlic pearls (GP) in a dose of 250 mg per day for 2 months. Baseline samples were taken at the start of the study, i.e. 0 day, and thereafter 2 months follow-up. 8-Hydroxy-2'-deoxyguanosine (8-OHdG), lipids, lipid peroxidation (MDA), NO and antioxidant vitamins A, E and C were determined. A moderate decline in blood pressure (BP) and a significant reduction in 8-OHdG, NO levels and lipid peroxidation were observed in Group I subjects with GP supplementation. Further, a significant increase in vitamin levels and TAS was also observed in this group as compared to the control subjects. These findings point out the beneficial effects of garlic supplementation in reducing blood pressure and counteracting oxidative stress, and thereby, offering cardioprotection in essential hypertensives.
Collapse
Affiliation(s)
- Veena Dhawan
- Department of Experimental Medicine and Biotechnology, Research Block B, IInd Floor, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
| | | |
Collapse
|
19
|
Affiliation(s)
- Lawrence S Cozma
- Department of Medicine, University Hospital of Wales B7, Cardiff CF14 4XW, UK
| |
Collapse
|