1
|
Ocharán-Mercado A, Loaeza-Loaeza J, Hernández-Sotelo D, Cid L, Hernández-Kelly LC, Felder-Shmittbuhl MP, Ortega A. Fluoride Exposure Increases the Activity of the Cystine/Glutamate Exchanger in Glia Cells. Neurochem Res 2025; 50:105. [PMID: 39998572 PMCID: PMC11861166 DOI: 10.1007/s11064-025-04358-2] [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: 11/14/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Fluoride exposure in drinking water has been widely related to impairment of cognitive function. Even though this ion has been described as neurotoxic for more than two decades, the molecular mechanisms of fluoride neurotoxicity are not fully understood, however, increasing evidence suggests that glial cells are the site of early injury in fluoride neurotoxicity. Nevertheless, a convergence point of many studies is the effect on glutamatergic neurotransmission and the generation of reactive oxygen species. In this context, we evaluated here the expression and regulation of the cystine/glutamate exchanger upon fluoride exposure since this transporter is in the interface between excitotoxicity and the antioxidant response. We demonstrate here the functional expression of the cystine /glutamate exchanger in both the U373 human glioblastoma cells and chick cerebellar Bergmann glia cells. Using a [3H]-L-Glutamate uptake assay, we demonstrate that fluoride increases the activity of the exchanger in a time and dose-dependent manner. This augmentation is mitigated by the antioxidant Trolox. To gain insight into fluoride neurotoxicity mechanisms, we evaluated its effect on human antigen R, a RNA binding protein, that binds to the 3'-UTR region of exchanger mRNA increasing its half time life. An increase in human antigen R protein was recorded after a 6 h fluoride exposure, suggesting that this ion regulates the exchanger through this RNA-binding protein. Furthermore, we show that fluoride exposure increases both the exchanger and human antigen R mRNAs half-life. These results provide insights into fluoride neurotoxicity mechanisms and support the notion of a central role of glial cells in neuronal glutamatergic transmission disruption that leads to neuronal cell death.
Collapse
Affiliation(s)
- Andrea Ocharán-Mercado
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro, Zacatenco, 07360, México
| | - Jaqueline Loaeza-Loaeza
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro, Zacatenco, 07360, México
| | - Daniel Hernández-Sotelo
- Laboratorio de Epigenética del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas 88, 39086, Chilpancingo, Guerrero, México
| | - Luis Cid
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro, Zacatenco, 07360, México
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro, Zacatenco, 07360, México
| | - Marie-Paule Felder-Shmittbuhl
- Centre National de La Recherche Scientifique, Institut Des Neurosciences Cellulaires Et Intégratives (UPR 3212), Université de Strasbourg, Strasbourg, France
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro, Zacatenco, 07360, México.
| |
Collapse
|
2
|
Nasr MM, Wahdan SA, El-Naga RN, Salama RM. Neuroprotective effect of empagliflozin against doxorubicin-induced chemobrain in rats: Interplay between SIRT-1/MuRF-1/PARP-1/NLRP3 signaling pathways and enhanced expression of miRNA-34a and LncRNA HOTAIR. Neurotoxicology 2024; 105:216-230. [PMID: 39426736 DOI: 10.1016/j.neuro.2024.10.006] [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: 08/24/2024] [Revised: 10/05/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Chemobrain, a challenging side effect of doxorubicin (DOX)-based chemotherapy, impairs cognitive abilities in cancer survivors. DOX triggers chemobrain via oxidative stress, leading to inflammation and apoptosis. Empagliflozin (EMPA), a sodium glucose co-transporter-2 inhibitor, demonstrated neuroprotective effects by reducing reactive oxygen species (ROS) and inflammation, but its protective mechanisms against DOX-induced chemobrain is not fully known. Thus, this study aimed to investigate EMPA's neuroprotective effects on DOX-induced chemobrain in rats and to uncover the underlying protective mechanisms. Fifty male Wistar rats were divided into control, EMPA, DOX (2 mg/kg, IP, once/week for 4 weeks), and two treated groups (DOX+ EMPA 5 and 10 mg/kg/day, PO, for 4 weeks). Behavioral tests showed improved memory, motor performance, and reduced anxiety in EMPA-treated groups compared to DOX, with superior results at the higher dose. Histopathological analysis revealed increased intact neurons in the cortex and hippocampus in EMPA-treated groups, with 346.4 % increase in CA3 (p < 0.0001), 19.1 % in dentate gyrus (p = 0.0006), and 362.6 % in cortex (p < 0.0001) in the high-dose EMPA group. Biochemical investigations of the high-dose EMPA group revealed significant decreases in inflammatory and apoptotic markers (JNK/PARP-1/NLRP3/MuRF-1/FOXO-1), increased SIRT-1 protein expression by 389.9 % (p < 0.0001), and reduced miRNA-34a and LncRNA HOTAIR gene expression (50.4 % and 53.4 % respectively, p < 0.0001) relative to DOX group. Conclusively, EMPA demonstrated superior behavioral and histopathological outcomes particularly at higher dose, positioning it as a promising neuroprotective candidate against DOX-induced chemobrain, possibly through modulating SIRT-1, NF-κb, NLRP3, and oxidative stress pathways.
Collapse
Affiliation(s)
- Merihane M Nasr
- Clinical Pharmacy Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Rania M Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| |
Collapse
|
3
|
Zhang Y, Gao Y, Liu X. Focus on cognitive impairment induced by excessive fluoride: An update review. Neuroscience 2024; 558:22-29. [PMID: 39137871 DOI: 10.1016/j.neuroscience.2024.08.011] [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: 10/04/2023] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Fluorosis is a global public health concern. Prolonged exposure to excessive fluoride causes fluoride accumulation in the hippocampus, resulting in cognitive dysfunction. Cell death is necessary for maintaining tissue function and morphology, and changes in the external morphology of nerve cells and the function of many internal organelles are typical features of cell death; however, it is also a typical feature of cognitive impairment caused by fluorosis. However, the pathogenesis of cognitive impairment caused by different degrees of fluoride exposure varies. Herein, we provide an overview of cognitive impairment caused by excessive fluoride exposure in different age groups, and the underlying mechanisms for cognitive impairment in various model organisms. The mechanisms underlying these impairments include oxidative stress, synaptic and neurotransmission dysfunction, disruption of mitochondrial and energy metabolism, and calcium channel dysregulation. This study aims to provide potential insights that serve as a reference for subsequent research on the cognitive function caused by excessive fluoride.
Collapse
Affiliation(s)
- Yuhang Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province and Ministry of Health (23618504), Harbin, Heilongjiang 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health, Harbin, Heilongjiang 150081, China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province and Ministry of Health (23618504), Harbin, Heilongjiang 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health, Harbin, Heilongjiang 150081, China.
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province and Ministry of Health (23618504), Harbin, Heilongjiang 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
4
|
Lee WS, Kim JH, Han B, Lee GC, Jung HR, Shin YJ, Ha EK, Lee E, Lee S, Han MY. Association of fluoride exposure with disease burden and neurodevelopment outcomes in children in South Korea. World J Pediatr 2024; 20:1029-1042. [PMID: 38937407 DOI: 10.1007/s12519-024-00820-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Community water fluoridation is an effective public health strategy for preventing dental caries, yet. Concerns exist about potential health problems. This study explores associations between tap water fluoride levels and pediatric disease burden, as well as neurodevelopmental outcomes at 6 years of age. METHODS This nationwide population-based cohort study included children born in Korean cities with and without tap water fluoridation projects, between 2006 and 2012, aiming for a fluoride concentration of 0.8 ± 0.2 mg/L in treated tap water. Data from the National Health Insurance Service were used, spanning from birth to 2018. The relationship between exposure to fluoridated tap water and incidence of 16 childhood diseases that were previously identified as potentially linked to fluoride exposure were examined. Additionally, we evaluated the neurodevelopmental outcomes across various domains, including gross motor, fine motor, cognition, language, social skills, and self-help functions. These assessments were performed using data from a comprehensive national health screening program for children aged six years. RESULTS A fluoride-unexposed group included 22,881 children, whereas a fluoride-exposed group comprised 29,991 children (52% males). Children in the fluoride-exposed group had a decreased risk of dental caries and bone fractures [hazard ratio (95% confidence interval, CI), 0.76 (0.63-0.93) and 0.89 (0.82-0.93), respectively] and increased risk of hepatic failures [1.85, (1.14-2.98)] compared to those in the unexposed group. Additionally, the risk ratio of abnormal neurodevelopmental screening outcomes increased by 9%, but this was statistically uncertain (95% CI, 0.95-1.26). CONCLUSIONS Fluoridated tap water was associated with an increased risk of hepatic failure but a decreased risk of bone fractures in children. The association between fluoridated tap water and neurodevelopmental screening outcomes at 6 years remains unclear, highlighting the need for further studies to clarify this association.
Collapse
Affiliation(s)
- Won Seok Lee
- Department of Pediatrics, CHA Ilsan Medical Center, CHA University School of Medicine, Goyang, Republic of Korea
| | - Ju Hee Kim
- Department of Pediatrics, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Boeun Han
- Department of Pediatrics, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
- Multi-Omics Research Center, CHA Future Medicine Research Institute, Seongnam, Republic of Korea
| | - Gi Chun Lee
- Department of Computer Science and Engineering, Konkuk University, Seoul, Republic of Korea
| | - Hye Ri Jung
- School of Medicine, CHA University, Pocheon, Republic of Korea
| | - Ye Jin Shin
- School of Medicine, CHA University, Pocheon, Republic of Korea
| | - Eun Kyo Ha
- Department of Pediatrics, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Eun Lee
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Man Yong Han
- Department of Pediatrics, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Bundang-Gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
5
|
Ba Y, Feng Z, Fu X, Chen R, Jiao X, Du Y, Liu X, Huang H, Yu F, Zhou G. Mediation of mitochondrial DNA copy number and oxidative stress in fluoride-related bone mineral density alteration in Chinese farmers. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2024; 46:184. [PMID: 38695941 DOI: 10.1007/s10653-024-01970-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/25/2024] [Indexed: 06/17/2024]
Abstract
Excessive fluoride can adversely affect bone mineral density (BMD). Oxidative stress and mitochondrial dysfunction are crucial mechanisms of health damage induced by fluoride. Here, a cross-sectional survey involving 907 Chinese farmers (aged 18-60) was carried out in Tongxu County in 2017, aiming to investigate the significance of mitochondrial DNA copy number (mtDNAcn) and oxidative stress in fluoride-related BMD change. Concentrations of urinary fluoride (UF), serum oxidative stress biomarkers, including total antioxidant capacity (T-AOC), total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA), as well as relative mtDNAcn in peripheral blood were determined. The multivariable linear model and mediation analysis were performed to assess associations between UF, oxidative stress, and relative mtDNAcn with BMD. Results showed that GSH-Px levels increased by 6.98 U/mL [95% confidence interval (CI) 3.41-10.56)] with each 1.0 mg/L increment of UF. After stratification, the T-AOC, relative mtDNAcn, and BMD decreased by 0.04 mmol/L (-0.08 ~ -0.01), 0.29-unit (-0.55 ~ -0.04), and 0.18-unit (-0.33 ~ -0.03) with every 1.0 mg/L elevation of UF in the excessive fluoride group (EFG, adults with UF > 1.6 mg/L), respectively. Furthermore, T-AOC and relative mtDNAcn were favorably related to the BMD in the EFG (β = 0.82, 95%CI 0.16-1.48 for T-AOC; β = 0.11, 95%CI 0.02-0.19 for relative mtDNAcn). Mediation analysis showed that relative mtDNAcn and T-AOC mediated 15.4% and 17.1% of the connection between excessive fluoride and reduced BMD, respectively. Findings suggested that excessive fluoride was related to lower BMD in adults, and the decrement of T-AOC and relative mtDNAcn partially mediate this relationship.
Collapse
Affiliation(s)
- Yue Ba
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
- Yellow River Institute for Ecological Protection & Regional Coordinated Development, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zichen Feng
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiaoli Fu
- Department of Health Management, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ruiqin Chen
- Department of Epidemiology, Jinshui District Center for Disease Control and Prevention, Zhengzhou, 450053, Henan, China
| | - Xuecheng Jiao
- Department of Epidemic Prevention and Control, Puyang Center for Disease Control and Prevention, Puyang, 457001, Henan, China
| | - Yuhui Du
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiaoxue Liu
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Hui Huang
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Fangfang Yu
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
- Yellow River Institute for Ecological Protection & Regional Coordinated Development, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Guoyu Zhou
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.
- Yellow River Institute for Ecological Protection & Regional Coordinated Development, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
6
|
Kumar S, Chhabra V, Mehra M, K S, Kumar B H, Shenoy S, Swamy RS, Murti K, Pai KSR, Kumar N. The fluorosis conundrum: bridging the gap between science and public health. Toxicol Mech Methods 2024; 34:214-235. [PMID: 37921264 DOI: 10.1080/15376516.2023.2268722] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/04/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023]
Abstract
Fluorosis, a chronic condition brought on by excessive fluoride ingestion which, has drawn much scientific attention and public health concern. It is a complex and multifaceted issue that affects millions of people worldwide. Despite decades of scientific research elucidating the causes, mechanisms, and prevention strategies for fluorosis, there remains a significant gap between scientific understanding and public health implementation. While the scientific community has made significant strides in understanding the etiology and prevention of fluorosis, effectively translating this knowledge into public health policies and practices remains challenging. This review explores the gap between scientific research on fluorosis and its practical implementation in public health initiatives. It suggests developing evidence-based guidelines for fluoride exposure and recommends comprehensive educational campaigns targeting the public and healthcare providers. Furthermore, it emphasizes the need for further research to fill the existing knowledge gaps and promote evidence-based decision-making. By fostering collaboration, communication, and evidence-based practices, policymakers, healthcare professionals, and the public can work together to implement preventive measures and mitigate the burden of fluorosis on affected communities. This review highlighted several vital strategies to bridge the gap between science and public health in the context of fluorosis. It emphasizes the importance of translating scientific evidence into actionable guidelines, raising public awareness about fluoride consumption, and promoting preventive measures at individual and community levels.
Collapse
Affiliation(s)
- Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Vishal Chhabra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Manmeet Mehra
- Department of Pharmacology, Guru Nanak Dev University, Amritsar, India
| | - Saranya K
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Harish Kumar B
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Smita Shenoy
- Department of Pharmacology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Ravindra Shantakumar Swamy
- Division of Anatomy, Department of Basic Medical Sciences (DBMS), Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, India
| |
Collapse
|
7
|
Dhapola R, Kumari S, Sharma P, HariKrishnaReddy D. Insight into the emerging and common experimental in-vivo models of Alzheimer's disease. Lab Anim Res 2023; 39:33. [PMID: 38082453 PMCID: PMC10712122 DOI: 10.1186/s42826-023-00184-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 05/30/2024] Open
Abstract
Alzheimer's disease (AD) is a multifactorial, rapidly progressing neurodegenerative disorder. As the exact cause of the disease is still unclear, the drug development is very challenging. This review encompasses the commonly used AD models involving various chemicals, heavy metals and endogenous substances induced models and the transgenic models. It also provides insight into the reliable emerging models of AD that may overcome the shortcomings associated with available models. Chemicals like streptozotocin, scopolamine, colchicine and okadaic acid render the animal susceptible to neuroinflammation and oxidative stress induced neurodegeneration along with amyloid-β deposition and tau hyperphosphorylation. Similarly, endogenous substances like acrolein and amyloid-β 1-42 are efficient in inducing the major pathologies of AD. Heavy metals like aluminum and fluoride and mixture of these have been reported to induce neurotoxicity therefore are used as animal models for AD. Transgenic models developed as a result of knock-in or knock-out of certain genes associated with AD including PDAPP, APP23, Tg2576, APP/PS1, 3 × Tg and 5 × FAD have also been incorporated in this study. Further, emerging and advanced pathomimetic models of AD are provided particular interest here which will add on to the current knowledge of animal models and may aid in the drug development process and deepen our understanding related to AD pathogenesis. These newly discovered models include oAβ25-35 model, transgenic model expressing 82-kDa ChAT, oDGal mouse and APP knock-in rat. This study may aid in the selection of suitable model for development of novel potent therapeutics and for exploring detailed pathogenic mechanism of AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Sneha Kumari
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Prajjwal Sharma
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Dibbanti HariKrishnaReddy
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
8
|
Kocak Y, Oto G, Huyut Z, Alp HH, Turkan F, Onay E. Effects of fluoride on oxidative DNA damage, nitric oxide level, lipid peroxidation and cholinesterase enzyme activity in a rotenone-induced experimental Parkinson's model. Neurol Res 2023; 45:979-987. [PMID: 37699078 DOI: 10.1080/01616412.2023.2257452] [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: 03/31/2023] [Accepted: 07/29/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVE Environmental toxins are known to be one of the important factors in the development of Parkinson's disease (PD). This study was designed to investigate the possible contribution of fluoride (F) exposure to oxidative stress and neurodegeneration in rats with PD induced by rotenone (ROT). MATERIALS AND METHODS A total of 72 Wistar albino male rats were used in the experiment and 9 groups were formed with 8 animals in each group. ROT (2 mg/kg) was administered subcutaneously (sc) for 28 days. Different doses of sodium fluoride (NaF) (25, 50 and 100 ug/mL) were given orally (po) for 4 weeks. Malondialdehyde (MDA), glutathione (GSH), nitric oxide (NO), oxidative DNA damage (8-OHdG) and cholinesterase (AChE/BChE) enzyme activities were evaluated in serum and brain tissue homogenates. RESULTS Rats treated with ROT and NaF had significant increases in serum and brain MDA, NO content, and decreases in GSH. In addition, the combination of ROT and NaF triggered oxidative DNA damage and resulted in increased AChE/BChE activity. CONCLUSIONS Findings suggest that NaF and ROT may interact synergistically leading to oxidative damage and neuronal cell loss. As a result, we believe that exposure to pesticides in combination with NaF is one of the environmental factors that should not be ignored in the etiology of neurological diseases such as PD in populations in areas with endemic fluorosis.
Collapse
Affiliation(s)
- Yilmaz Kocak
- Department of Physical therapy and rehabilitation, Faculty of Health Sciences, Van Yuzuncu Yil University, Van, Turkey
- Department of Pharmacology-Toxicology, Van Yuzuncu Yil University, Van, Turkey
| | - Gokhan Oto
- Department of Pharmacology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yıl University, Van, Turkey
| | - Hamit Hakan Alp
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yıl University, Van, Turkey
| | - Fikret Turkan
- Department of Basic Sciences Faculty of Dentistry, Igdir University, Iğdır, Turkey
| | - Ezgi Onay
- Department of Pharmacology, Faculty of Medicine, Van Yüzüncü Yıl University, Van, Turkey
| |
Collapse
|
9
|
Ma Y, Meng X, Sowanou A, Wang J, Li H, Li A, Zhong N, Yao Y, Pei J. Effect of Fluoride on the Expression of 8-Hydroxy-2'-Deoxyguanosine in the Blood, Kidney, Liver, and Brain of Rats. Biol Trace Elem Res 2023; 201:2904-2916. [PMID: 35984601 DOI: 10.1007/s12011-022-03394-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022]
Abstract
Excessive exposure of fluoride not only leads to damage on bone, but also has an adverse effect on soft tissues. Oxidative DNA damage induced by fluoride is thought to be one of the toxic mechanisms of fluoride effect. However, the dose-response of fluoride on oxidative DNA damage is barely studied in organisms. This study investigated the concentration of fluoride in rat blood, kidney, liver, and brain as well as the dose-time effect of fluoride on the expression of 8-hydroxy-2'-deoxyguanosine (8-OHdG) in the above tissues. Rats were exposed to 0 mg/L, 25 mg/L, 50 mg/L, and 100 mg/L of fluorine ion and treated for one and three months. The results showed that the accumulation of fluoride in soft tissues was very different. At the first month, blood fluoride was increased, liver and brain fluoride showed a U-shaped change, and kidney fluoride was not significant. At the third month, blood fluoride was altered with an inverted U-shaped change, kidney and brain fluoride increased, but liver fluoride decreased. Both the exposure concentration and the time of exposure had a significant effect on the expression of 8-OHdG in the above tissues. However, the effect patterns of fluoride on these tissues were notably different at different times. At the first month of fluoride treatment, blood, kidney, and liver 8-OHdG decreased with the increasing fluoride concentration. At the third month, blood 8-OHdG showed a U-shaped change, but kidney 8-OHdG altered with an inverted U-shaped change. Liver 8-OHdG increased, while brain 8-OHdG decreased at the third month. Correlation analysis showed that only blood 8-OHdG was significantly inversely correlated with blood fluoride and dental fluorosis grade in both the first and third months. Liver 8-OHdG was negatively and significantly correlated with liver fluoride. There was a weak but nonsignificant correlation between kidney and brain 8-OHdG and fluoride in both tissues. Additionally, blood 8-OHdG was positively correlated with kidney and liver 8-OHdG at the first month and positively correlated with brain 8-OHdG at the third month. Taken together, our data suggests that concentration and time of fluoride exposure had a significant effect on 8-OHdG, but the effect patterns of fluoride on 8-OHdG were different in the tissues, which suggests that the impact of fluoride on 8-OHdG may be a tissue-specific, as well as a non-monotonic positive correlation.
Collapse
Affiliation(s)
- Yongzheng Ma
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Xinyue Meng
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Alphonse Sowanou
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Jian Wang
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Hanying Li
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Ailin Li
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Nan Zhong
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Yingjie Yao
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Junrui Pei
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province (23618504), Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
10
|
Xin J, Zhu B, Wang H, Zhang Y, Sun N, Cao X, Zheng L, Zhou Y, Fang J, Jing B, Pan K, Zeng Y, Zeng D, Li F, Xia Y, Xu P, Ni X. Prolonged fluoride exposure induces spatial-memory deficit and hippocampal dysfunction by inhibiting small heat shock protein 22 in mice. JOURNAL OF HAZARDOUS MATERIALS 2023; 456:131595. [PMID: 37224709 DOI: 10.1016/j.jhazmat.2023.131595] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/08/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
Millions of residents in areas with high-fluoride drinking water supply ingest excessive levels of fluoride for long periods. This study investigated the mechanisms and impacts of lifelong exposure to naturally occurring moderate-high-fluoride drinking water on spatial-memory function by studying mice in controlled experiments. Spatial-memory deficits and disorders of hippocampal neuronal electrical activity were observed in mice exposed to 25-ppm or 50-ppm-fluoride drinking water for 56 weeks, but not in adult or old mice exposed to 50 ppm fluoride for 12 weeks. Ultrastructural analysis showed severely damaged hippocampal mitochondria, evidenced by reduced mitochondrial membrane potential and ATP content. Mitochondrial biogenesis was impaired in fluoride-exposed mice, manifesting as a significantly reduced mtDNA content, mtDNA-encoded subunits mtND6 and mtCO1, and respiratory complex activities. Fluoride reduced expression of Hsp22, a beneficial mediator of mitochondrial homeostasis, and decreased levels of signaling for the PGC-1α/TFAM pathway-which regulates mitochondrial biogenesis-and the NF-κβ/STAT3 pathway-which regulates mitochondrial respiratory chain enzyme activity. Hippocampus-specific Hsp22-overexpression improved fluoride-induced spatial-memory deficits by activating the PGC-1α/TFAM and STAT3 signaling pathways, while Hsp22-silencing aggravated the deficits by inhibiting both pathways. Downregulation of Hsp22 plays a vital role in fluoride-induced spatial-memory deficits by impacting mtDNA-encoding subsets and mitochondrial respiratory chain enzyme activity.
Collapse
Affiliation(s)
- Jinge Xin
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bin Zhu
- Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hesong Wang
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Zhang
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xi Cao
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Liqin Zheng
- Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yanxi Zhou
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jing Fang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Fali Li
- Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yang Xia
- Department of Neurosurgery, Sichuan Provincial People's Hospital, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Peng Xu
- Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Ottappilakkil H, Babu S, Balasubramanian S, Manoharan S, Perumal E. Fluoride Induced Neurobehavioral Impairments in Experimental Animals: a Brief Review. Biol Trace Elem Res 2023; 201:1214-1236. [PMID: 35488996 DOI: 10.1007/s12011-022-03242-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/09/2022] [Indexed: 02/07/2023]
Abstract
Fluoride is one of the major toxicants in the environment and is often found in drinking water at higher concentrations. Living organisms including humans exposed to high fluoride levels are found to develop mild-to-severe detrimental pathological conditions called fluorosis. Fluoride can cross the hematoencephalic barrier and settle in various brain regions. This accumulation affects the structure and function of both the central and peripheral nervous systems. The neural ultrastructure damages are reflected in metabolic and cognitive activities. Hindrances in synaptic plasticity and signal transmission, early neuronal apoptosis, functional alterations of the intercellular signaling pathway components, improper protein synthesis, dyshomeostasis of the transcriptional and neurotrophic factors, oxidative stress, and inflammatory responses are accounted for the fluoride neurotoxicity. Fluoride causes a decline in brain functions that directly influence the overall quality of life in both humans and animals. Animal studies are widely used to explore the etiology of fluoride-induced neurotoxicity. A good number of these studies support a positive correlation between fluoride intake and toxicity phenotypes closely associated with neurotoxicity. However, the experimental dosages highly surpass the normal environmental concentrations and are difficult to compare with human exposures. The treatment procedures are highly dependent on the dosage, duration of exposure, sex, and age of specimens among other factors which make it difficult to arrive at general conclusions. Our review aims to explore fluoride-induced neuronal damage along with associated histopathological, behavioral, and cognitive effects in experimental models. Furthermore, the correlation of various molecular mechanisms upon fluoride intoxication and associated neurobehavioral deficits has been discussed. Since there is no well-established mechanism to prevent fluorosis, phytochemical-based alleviation of its characteristic indications has been proposed as a possible remedial measure.
Collapse
Affiliation(s)
| | - Srija Babu
- Bharathiar University, Coimbatore, Tamilnadu, India
| | | | | | | |
Collapse
|
12
|
Żwierełło W, Maruszewska A, Skórka-Majewicz M, Gutowska I. Fluoride in the Central Nervous System and Its Potential Influence on the Development and Invasiveness of Brain Tumours-A Research Hypothesis. Int J Mol Sci 2023; 24:1558. [PMID: 36675073 PMCID: PMC9866357 DOI: 10.3390/ijms24021558] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The purpose of this review is to attempt to outline the potential role of fluoride in the pathogenesis of brain tumours, including glioblastoma (GBM). In this paper, we show for the first time that fluoride can potentially affect the generally accepted signalling pathways implicated in the formation and clinical course of GBM. Fluorine compounds easily cross the blood-brain barrier. Enhanced oxidative stress, disruption of multiple cellular pathways, and microglial activation are just a few examples of recent reports on the role of fluoride in the central nervous system (CNS). We sought to present the key mechanisms underlying the development and invasiveness of GBM, as well as evidence on the current state of knowledge about the pleiotropic, direct, or indirect involvement of fluoride in the regulation of these mechanisms in various tissues, including neural and tumour tissue. The effects of fluoride on the human body are still a matter of controversy. However, given the growing incidence of brain tumours, especially in children, and numerous reports on the effects of fluoride on the CNS, it is worth taking a closer look at these mechanisms in the context of brain tumours, including gliomas.
Collapse
Affiliation(s)
- Wojciech Żwierełło
- Department of Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 71 St., 70-111 Szczecin, Poland
| | - Agnieszka Maruszewska
- Department of Physiology and Biochemistry, Institute of Biology, University of Szczecin, Felczaka 3c St., 71-412 Szczecin, Poland
- Molecular Biology and Biotechnology Centre, Institute of Biology, University of Szczecin, Wąska 13 St., 71-415 Szczecin, Poland
| | - Marta Skórka-Majewicz
- Department of Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 71 St., 70-111 Szczecin, Poland
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 71 St., 70-111 Szczecin, Poland
| |
Collapse
|
13
|
Wu Y, Zhang X, Chen J, Cao J, Feng C, Luo Y, Lin Y. Self-recovery study of fluoride-induced ferroptosis in the liver of zebrafish (Danio rerio). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 251:106275. [PMID: 36007351 DOI: 10.1016/j.aquatox.2022.106275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/29/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Ferroptosis plays a key role in fluorosis in aquatic organisms, but whether it is involved in fluoride-induced liver damage remains unclear. Previous studies have indicated that fluoride toxicity has the reversible tendency, but the mechanism of self-recovery after fluorosis in aquatic animals has not been elucidated. In this study, adult zebrafish and embryos were exposed to 0, 20, 40, 80 mg/L of fluoride for 30, 60 and 90 d and 3, 4 and 5 d post-fertilization (dpf), respectively. After 90 d, adult zebrafish were transferred to clean water for self-recovery of 30 d. The results showed that fluoride induced the prominent histopathologial changes in liver of adults, and the developmental delay and dark liver area in larvae. Fluoride significantly increased the iron overload, while decreased the expression levels of transferrin (tf), transferrin receptor (tfr), ferroportin (fpn), membrane iron transporter (fpn), and ferritin heavy chain (fth) in adults and larvae. Fluoride also induced the oxidative stress in adults and larvae by increasing the levels of reactive oxygen species (ROS) and malondialdehyde (MDA), while decreasing the glutathione (GSH) content and the levels of glutathione peroxidase 4 (gpx4) and solute carrier family 7 member 11 (slc7a11). Self-recovery relieved fluoride-induced ferroptosis by reducing the histopathological damage and oxidative stress, reversing the expression levels of fth and slc7a11, Fe2+ metabolism and GSH synthesis. Lipid peroxidation and Fe2+ metabolism may be the key factor in alleviating effects of self-recovery on fluoride toxicity. Moreover, males are more sensitive than females. Our results provide a theoretical basis for studying the alleviating effects of self-recovery on fluoride toxicity and the underlying mechanism of its protective effect.
Collapse
Affiliation(s)
- Yijie Wu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xiuling Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jianjie Chen
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jinling Cao
- College of Food Science and Technology, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| | - Cuiping Feng
- College of Food Science and Technology, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Yongju Luo
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Science, Nanning 530021, Guangxi, China.
| | - Yong Lin
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Science, Nanning 530021, Guangxi, China.
| |
Collapse
|
14
|
Redox and biometal status in Wistar rats after subacute exposure to fluoride and selenium counter-effects. Arh Hig Rada Toksikol 2022; 73:207-222. [PMID: 36226821 PMCID: PMC9837529 DOI: 10.2478/aiht-2022-73-3650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/01/2022] [Indexed: 11/05/2022] Open
Abstract
This study aimed to investigate the effect of 150 mg/L sodium fluoride (NaF) on redox status parameters and essential metals [copper (Cu), iron (Fe), and zinc (Zn)] in the blood, liver, kidney, brain, and spleen of Wistar rats and to determine the protective potential of selenium (Se) against fluoride (F-) toxicity. Male Wistar rats were randomly distributed in groups of five (n=5) receiving tap water (control) or water with NaF 150 mg/L, NaF 150 mg/L + Se 1.5 mg/L, and Se 1.5 mg/L solutions ad libitum for 28 days. Fluorides caused an imbalance in the redox and biometal (Cu, Fe, and Zn) status, leading to high superoxide anion (O2 .-) and malondialdehyde (MDA) levels in the blood and brain and a drop in superoxide dismutase (SOD1) activity in the liver and its increase in the brain and kidneys. Se given with NaF improved MDA, SOD1, and O2 .- in the blood, brain, and kidneys, while alone it decreased SH group levels in the liver and kidney. Biometals both reduced and increased F- toxicity. Further research is needed before Se should be considered as a promising strategy for mitigating F- toxicity.
Collapse
|
15
|
Xu K, Feng Z, Afrim FK, Ma J, Yang S, Zhang X, Niu Z, An N, Du Y, Yu F, Zhou G, Ba Y. Interaction of fluoride exposure and CREB1 gene polymorphisms on thyroid function in school-age children. CHEMOSPHERE 2022; 303:135156. [PMID: 35640685 DOI: 10.1016/j.chemosphere.2022.135156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/13/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES To evaluate the effects of CREB1 gene polymorphisms and long-term exposure to fluoride on thyroid function of children. STUDY DESIGN A total of 424 children (including 226 boys and 198 girls) aged 7-12 years old were enrolled in Kaifeng, China by cross-sectional study in 2017. The concentrations of urinary fluoride (UF) and creatinine (UCr) were measured using fluoride ion-selective electrode assay and creatinine assay kit (picric acid method), respectively. The concentration of UCr-adjusted UF (CUF) was calculated. Children were divided into high fluoride exposure group (HFG, CUF >1.41 mg/L) and low fluoride exposure group (LFG, CUF ≤1.41 mg/L) according to the median concentration of CUF (1.41 mg/L). The serum thyroid-stimulating hormone (TSH), total triiodothyronine (TT3) and total thyronine (TT4) levels were detected by the radiation immunoassay. The B-mode ultrasound was performed to test the thyroid volume (Tvol). Genotyping of CREB1 gene was conducted by a custom-by-design 48-plex SNPscan™ Kit. Associations between CUF concentration, CREB1 single nucleotide polymorphisms (SNPs) and thyroid function were assessed by multiple linear regression models. RESULTS Negative and positive associations between serum TT4 level (β = -0.721, 95%CI: -1.209, -0.234) and Tvol (β = 0.031, 95%CI: 0.011, 0.050) and CUF concentration were observed respectively. Children carrying CREB1 rs11904814 TG and rs2254137 AC genotypes had lower TT3 levels (P < 0.05). Children in HFG carrying rs11904814 TT, rs2253206 GG genotypes and rs6740584 C allele easily manifested lower serum TT4 levels (P < 0.05). Moreover, interactions between excessive fluoride exposure and CREB1 SNPs on Tvol were observed, and the interaction among different loci of CREB1 gene could modify serum TT3 level (P < 0.05, respectively). CONCLUSIONS Fluoride could alter children's serum TT4 levels and Tvol. Interactions between fluoride exposure and CREB1 polymorphisms may modify thyroid volume of children.
Collapse
Affiliation(s)
- Kaihong Xu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zichen Feng
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Francis Kojo Afrim
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jun Ma
- Department of Endemic Disease, Kaifeng Center for Disease Control and Prevention, Kaifeng, Henan, 475000, China
| | - Shuo Yang
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xuanyin Zhang
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zeyuan Niu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Ning An
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yuhui Du
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Fangfang Yu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Guoyu Zhou
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yue Ba
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
16
|
Zhou W, Luo W, Liu D, Canavese F, Li L, Zhao Q. Fluoride increases the susceptibility of developmental dysplasia of the hip via increasing capsular laxity triggered by cell apoptosis and oxidative stress in vivo and in vitro. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 234:113408. [PMID: 35298972 DOI: 10.1016/j.ecoenv.2022.113408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/26/2022] [Accepted: 03/08/2022] [Indexed: 06/14/2023]
Abstract
The etiology of developmental dysplasia of the hip (DDH) is multifactorial, including breech presentation and hip capsular laxity. In particular, hip laxity is the main contributor to DDH by changing the ratio and distribution of collagens. Also, fluoride (F) affects collagens from various tissue besides bone and tooth. To investigate the association of DDH and excessive F intake, we conducted this research in lab on cell and animal model simultaneously. We established animal model of combination of DDH and F toxicity. The incidence of DDH in each group was calculated, and hip capsules were collected for testing histopathological and ultrastructural changes. The primary fibroblasts were further extracted from hip capsule and treated with F. The expression of collagen type I and III was both examined in vivo and in vitro, and the level of oxidative stress and apoptosis was also tested identically. We revealed that the incidence of DDH increased with F concentration. Furthermore, the oxidative stress and apoptosis levels of hip capsules and fibroblasts both increased after F exposure. Therefore, this study shows that excessive F intake increases susceptibility to DDH by altering hip capsular laxity in vivo and in vitro respectively. We believe that F might be a risk factor for DDH by increasing hip laxity induced by triggering fibroblast oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Weizheng Zhou
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Dan Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Federico Canavese
- Department of Pediatric Orthopedics, Lille University Center, Jeanne de Flandres Hospital, Avenue Eugène-Avinée, Lille 59037, France
| | - Lianyong Li
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China.
| | - Qun Zhao
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| |
Collapse
|
17
|
Impacts of Fluoride Neurotoxicity and Mitochondrial Dysfunction on Cognition and Mental Health: A Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182412884. [PMID: 34948493 PMCID: PMC8700808 DOI: 10.3390/ijerph182412884] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 12/21/2022]
Abstract
This review focuses on the synthesis of current experimental and observational data regarding the effect of fluoride exposure on childhood mental health and the role of mitochondrial function as a mechanism of action. We aggregated data on the relationships between fluoride neurotoxicity, mitochondrial function, and cognitive and mental health using PubMed. Current animal and human research suggest that prenatal and perinatal fluoride exposure might have neurotoxic effects. These studies observed physical changes (fur loss and delayed reflex development in animals), intelligence loss, increased hyperactivity, and irregular moods associated with fluoride exposure. Two gaps in the literature were identified: (1) there is limited research on the mental and emotional impacts of fluoride exposure compared to research on cognitive outcomes, and (2) human studies primarily focus on prenatal and perinatal exposure, with little research conducted at other time points (e.g., adolescence). Furthermore, there is no agreed-upon mechanism for the neurotoxic effects of fluoride; however, fluoride can induce mitochondrial damage, including decreasing circulating mitochondrial DNA content, dysregulating biogenesis, and circular structure loss. Additionally, many neurodevelopmental conditions have mitochondrial underpinnings. More work is needed to elucidate the impact and timing of fluoride exposure on mental health and the role of mitochondrial function as a biological mechanism
Collapse
|
18
|
Haddad M, Hervé V, Ben Khedher MR, Rabanel JM, Ramassamy C. Glutathione: An Old and Small Molecule with Great Functions and New Applications in the Brain and in Alzheimer's Disease. Antioxid Redox Signal 2021; 35:270-292. [PMID: 33637005 DOI: 10.1089/ars.2020.8129] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Significance: Glutathione (GSH) represents the most abundant and the main antioxidant in the body with important functions in the brain related to Alzheimer's disease (AD). Recent Advances: Oxidative stress is one of the central mechanisms in AD. We and others have demonstrated the alteration of GSH levels in the AD brain, its important role in the detoxification of advanced glycation end-products and of acrolein, a by-product of lipid peroxidation. Recent in vivo studies found a decrease of GSH in several areas of the brain from control, mild cognitive impairment, and AD subjects, which are correlated with cognitive decline. Critical Issues: Several strategies were developed to restore its intracellular level with the l-cysteine prodrugs or the oral administration of γ-glutamylcysteine to prevent alterations observed in AD. To date, no benefit on GSH level or on oxidative biomarkers has been reported in clinical trials. Thus, it remains uncertain if GSH could be considered a potential preventive or therapeutic approach or a biomarker for AD. Future Directions: We address how GSH-coupled nanocarriers represent a promising approach for the functionalization of nanocarriers to overcome the blood/brain barrier (BBB) for the brain delivery of GSH while avoiding cellular toxicity. It is also important to address the presence of GSH in exosomes for its potential intercellular transfer or its shuttle across the BBB under certain conditions. Antioxid. Redox Signal. 35, 270-292.
Collapse
Affiliation(s)
- Mohamed Haddad
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada.,Institute on Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - Vincent Hervé
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada.,Institute on Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - Mohamed Raâfet Ben Khedher
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada.,Institute on Nutrition and Functional Foods, Université Laval, Québec, Canada
| | | | - Charles Ramassamy
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada.,Institute on Nutrition and Functional Foods, Université Laval, Québec, Canada
| |
Collapse
|
19
|
Xu K, An N, Huang H, Duan L, Ma J, Ding J, He T, Zhu J, Li Z, Cheng X, Zhou G, Ba Y. Fluoride exposure and intelligence in school-age children: evidence from different windows of exposure susceptibility. BMC Public Health 2020; 20:1657. [PMID: 33148225 PMCID: PMC7640398 DOI: 10.1186/s12889-020-09765-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/26/2020] [Indexed: 12/25/2022] Open
Abstract
Background The intellectual loss induced by fluoride exposure has been extensively studied, but the association between fluoride exposure in different susceptibility windows and children’s intelligence is rarely reported. Hence, we conducted a cross-sectional study to explore the association between fluoride exposure in prenatal and childhood periods and intelligence quotient (IQ). Methods We recruited 633 local children aged 7–13 years old randomly from four primary schools in Kaifeng, China in 2017. The children were divided into four groups, of which included: control group (CG, n = 228), only prenatal excessive fluoride exposure group (PFG, n = 107), only childhood excessive fluoride exposure group (CFG, n = 157), both prenatal and childhood excessive fluoride exposure group (BFG, n = 141). The concentrations of urinary fluoride (UF) and urinary creatinine (UCr) were determined by fluoride ion-selective electrode assay and a creatinine assay kit (picric acid method), respectively. The concentration of UCr-adjusted urinary fluoride (CUF) was calculated. IQ score was assessed using the second revision of the Combined Raven’s Test-The Rural in China (CRT-RC2). Threshold and saturation effects analysis, multiple linear regression analysis and logistic regression analysis were conducted to analyze the association between fluoride exposure and IQ. Results The mean IQ score in PFG was respectively lower than those in CG, CFG and BFG (P < 0.05). The odds of developing excellent intelligence among children in PFG decreased by 51.1% compared with children in CG (OR = 0.489, 95% CI: 0.279, 0.858). For all the children, CUF concentration of ≥1.7 mg/L was negatively associated with IQ scores (β = − 4.965, 95% CI: − 9.198, − 0.732, P = 0.022). In children without prenatal fluoride exposure, every 1.0 mg/L increment in the CUF concentration of ≥2.1 mg/L was related to a reduction of 11.4 points in children’s IQ scores (95% CI: − 19.2, − 3.5, P = 0.005). Conclusions Prenatal and childhood excessive fluoride exposures may impair the intelligence development of school children. Furthermore, children with prenatal fluoride exposure had lower IQ scores than children who were not prenatally exposed; therefore the reduction of IQ scores at higher levels of fluoride exposure in childhood does not become that evident.
Collapse
Affiliation(s)
- Kaihong Xu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.,Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ning An
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Hui Huang
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.,Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Leizhen Duan
- Department of Medical Services, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jun Ma
- Department of Endemic Disease, Kaifeng Center for Disease Control and Prevention, Kaifeng, 475000, Henan, China
| | - Jizhe Ding
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Tongkun He
- The Medical Section, The Eighth People Hospital of Zhengzhou, Zhengzhou, 450000, Henan, China
| | - Jingyuan Zhu
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zhiyuan Li
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xuemin Cheng
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.,Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Guoyu Zhou
- Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,Yellow River Institute for Ecological Protection & Regional Coordinated Development, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Yue Ba
- Department of Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China. .,Yellow River Institute for Ecological Protection & Regional Coordinated Development, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
20
|
Lopes GO, Martins Ferreira MK, Davis L, Bittencourt LO, Bragança Aragão WA, Dionizio A, Rabelo Buzalaf MA, Crespo-Lopez ME, Maia CSF, Lima RR. Effects of Fluoride Long-Term Exposure over the Cerebellum: Global Proteomic Profile, Oxidative Biochemistry, Cell Density, and Motor Behavior Evaluation. Int J Mol Sci 2020; 21:E7297. [PMID: 33023249 PMCID: PMC7582550 DOI: 10.3390/ijms21197297] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/22/2020] [Accepted: 08/28/2020] [Indexed: 12/16/2022] Open
Abstract
Although the literature does not provide evidence of health risks from exposure to fluoride (F) in therapeutic doses, questions remain about the effects of long-term and high-dose use on the function of the central nervous system. The objective of this study was to investigate the effect of long-term exposure to F at levels similar to those found in areas of artificial water fluoridation and in areas of endemic fluorosis on biochemical, proteomic, cell density, and functional parameters associated with the cerebellum. For this, mice were exposed to water containing 10 mg F/L or 50 mg F/L (as sodium fluoride) for 60 days. After the exposure period, the animals were submitted to motor tests and the cerebellum was evaluated for fluoride levels, antioxidant capacity against peroxyl radicals (ACAP), lipid peroxidation (MDA), and nitrite levels (NO). The proteomic profile and morphological integrity were also evaluated. The results showed that the 10 mg F/L dose was able to decrease the ACAP levels, and the animals exposed to 50 mg F/L presented lower levels of ACAP and higher levels of MDA and NO. The cerebellar proteomic profile in both groups was modulated, highlighting proteins related to the antioxidant system, energy production, and cell death, however no neuronal density change in cerebellum was observed. Functionally, the horizontal exploratory activity of both exposed groups was impaired, while only the 50 mg F/L group showed significant changes in postural stability. No motor coordination and balance impairments were observed in both groups. Our results suggest that fluoride may impair the cerebellar oxidative biochemistry, which is associated with the proteomic modulation and, although no morphological impairment was observed, only the highest concentration of fluoride was able to impair some cerebellar motor functions.
Collapse
Affiliation(s)
- Géssica Oliveira Lopes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA 66075-110, Brazil; (G.O.L.); (M.K.M.F.); (L.D.); (L.O.B.); (W.A.B.A.)
| | - Maria Karolina Martins Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA 66075-110, Brazil; (G.O.L.); (M.K.M.F.); (L.D.); (L.O.B.); (W.A.B.A.)
| | - Lodinikki Davis
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA 66075-110, Brazil; (G.O.L.); (M.K.M.F.); (L.D.); (L.O.B.); (W.A.B.A.)
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA 66075-110, Brazil; (G.O.L.); (M.K.M.F.); (L.D.); (L.O.B.); (W.A.B.A.)
| | - Walessa Alana Bragança Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA 66075-110, Brazil; (G.O.L.); (M.K.M.F.); (L.D.); (L.O.B.); (W.A.B.A.)
| | - Aline Dionizio
- Bauru School of Dentistry, Department of Biological Sciences, University of São Paulo, Bauru, SP 17012-90, Brazil; (A.D.); (M.A.R.B.)
| | - Marília Afonso Rabelo Buzalaf
- Bauru School of Dentistry, Department of Biological Sciences, University of São Paulo, Bauru, SP 17012-90, Brazil; (A.D.); (M.A.R.B.)
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém, PA 66075-110, Brazil;
| | - Cristiane Socorro Ferraz Maia
- Laboratory of Inflammation and Behavior Pharmacology, Pharmacy Faculty, Institute of Health Science, Federal University of Pará, Belém, PA 66075-110, Brazil;
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, PA 66075-110, Brazil; (G.O.L.); (M.K.M.F.); (L.D.); (L.O.B.); (W.A.B.A.)
| |
Collapse
|