1
|
Kou X, Canals J, Bulló M, Becerra-Tomás N, Jardí C, Arija V. Association of Prenatal Dietary Toxicants and Inorganic Arsenic Exposure with Children's Emotional and Behavioral Problems: ECLIPSES Study. TOXICS 2024; 12:398. [PMID: 38922078 PMCID: PMC11209564 DOI: 10.3390/toxics12060398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024]
Abstract
Prenatal exposure to dietary toxicants is linked to neurocognitive issues, but its effect on early emotional and behavioral development in children is less clear. To explore the relationship between prenatal intake of As, iAs, Cd, MeHg, Pb, PCDD/Fs, DL-PCBs, and NDL-PCBs and emotional and behavioral issues in four-year-old children. This study included 192 mother-child pairs from the ECLIPSES study, assessing prenatal dietary toxicant exposure through a food-frequency questionnaire and Catalan Food Safety Agency data. Children's emotional and behavioral scores were evaluated using the Child Behavior Checklist for ages 1.5-5 years. Multivariable regression and logistic models were used, focusing on iAs after finding significant preliminary associations. Increased prenatal dietary intake of iAs was associated with internalizing, externalizing, and attention-deficit/hyperactivity problems. Higher iAs levels (>4.16 μg/day) significantly increased the risk of total problems (OR = 2.94) and specific issues like anxious/depressed (OR = 4.88), anxiety (OR = 3.27), and oppositional defiant problems (OR = 4.30). High iAs consumption correlated with the intake of meat, eggs, cereals, tubers, fruits, and pulses Prenatal dietary iAs exposure is associated with various emotional and behavioral problems in children. Monitoring and reducing iAs levels in food are crucial for public health.
Collapse
Affiliation(s)
- Xiruo Kou
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43204 Reus, Spain (J.C.); (N.B.-T.); (C.J.)
- Institut d’Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain;
| | - Josefa Canals
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43204 Reus, Spain (J.C.); (N.B.-T.); (C.J.)
- Institut d’Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain;
- Centre de Recerca en Avaluació i Mesura de la Conducta (CRAMC), Department of Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
- University Research Institute on Sustainablility, Climate Change and Energy Transition (IU-RESCAT), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Monica Bulló
- Institut d’Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain;
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, 43201 Reus, Spain
| | - Nerea Becerra-Tomás
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43204 Reus, Spain (J.C.); (N.B.-T.); (C.J.)
- Institut d’Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain;
| | - Cristina Jardí
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43204 Reus, Spain (J.C.); (N.B.-T.); (C.J.)
- Institut d’Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain;
| | - Victoria Arija
- Nutrition and Mental Health (NUTRISAM) Research Group, Universitat Rovira i Virgili, 43204 Reus, Spain (J.C.); (N.B.-T.); (C.J.)
- Institut d’Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain;
- University Research Institute on Sustainablility, Climate Change and Energy Transition (IU-RESCAT), Universitat Rovira i Virgili, 43003 Tarragona, Spain
- Collaborative Research Group on Lifestyles, Nutrition and Smoking (CENIT), Tarragona-Reus Research Support Unit, Jordi Gol Primary Care Research Institute, 43003 Tarragona, Spain
| |
Collapse
|
2
|
González-Alfonso WL, Petrosyan P, Del Razo LM, Sánchez-Peña LC, Tapia-Rodríguez M, Hernández-Muñoz R, Gonsebatt ME. Chronic Exposure to Arsenic and Fluoride Starting at Gestation Alters Liver Mitochondrial Protein Expression and Induces Early Onset of Liver Fibrosis in Male Mouse Offspring. Biol Trace Elem Res 2024:10.1007/s12011-024-04198-1. [PMID: 38676876 DOI: 10.1007/s12011-024-04198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 04/21/2024] [Indexed: 04/29/2024]
Abstract
The presence of arsenic (As) and fluoride (F-) in drinking water is of concern due to the enormous number of individuals exposed to this condition worldwide. Studies in cultured cells and animal models have shown that As- or F-induced hepatotoxicity is primarily associated with redox disturbance and altered mitochondrial homeostasis. To explore the hepatotoxic effects of chronic combined exposure to As and F- in drinking water, pregnant CD-1 mice were exposed to 2 mg/L As (sodium arsenite) and/or 25 mg/L F- (sodium fluoride). The male offspring continued the exposure treatment up to 30 (P30) or 90 (P90) postnatal days. GSH levels, cysteine synthesis enzyme activities, and cysteine transporter levels were investigated in liver homogenates, as well as the expression of biomarkers of ferroptosis and mitochondrial biogenesis-related proteins. Serum transaminase levels and Hematoxylin-Eosin and Masson trichrome-stained liver tissue slices were examined. Combined exposure at P30 significantly reduced GSH levels and the mitochondrial transcription factor A (TFAM) expression while increasing lipid peroxidation, free Fe 2+, p53 expression, and serum ALT activity. At P90, the upregulation of cysteine uptake and synthesis was associated with a recovery of GSH levels. Nevertheless, the downregulation of TFAM continued and was now associated with a downstream inhibition of the expression of MT-CO2 and reduced levels of mtDNA and fibrotic liver damage. Our experimental approach using human-relevant doses gives evidence of the increased risk for early liver damage associated with elevated levels of As and F- in the diet during intrauterine and postnatal period.
Collapse
Affiliation(s)
- Wendy L González-Alfonso
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, México
| | - Pavel Petrosyan
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, México
| | - Luz M Del Razo
- Departamento de Toxicología, Centro de Investigación y Estudios Avanzados, 07360, Mexico City, Mexico
| | - Luz C Sánchez-Peña
- Departamento de Toxicología, Centro de Investigación y Estudios Avanzados, 07360, Mexico City, Mexico
| | - Miguel Tapia-Rodríguez
- Unidad de Microscopia, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Rolando Hernández-Muñoz
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, 04510, México
| | - María E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, México.
| |
Collapse
|
3
|
Ma M, Zhang J, Li S, Zhang M, Chen W, Li L, Li S. LINC00942 Alleviates NaAsO 2-induced Apoptosis by Promoting GSH Synthesis Through Targeting miR-214-5p. Biol Trace Elem Res 2024:10.1007/s12011-024-04167-8. [PMID: 38578483 DOI: 10.1007/s12011-024-04167-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
The mechanism of arsenic-induced liver toxicity is not fully understood. This study aimed to investigate the role of LINC00942 in arsenic-induced hepatotoxicity by regulating miR-214-5p. As the exposure dose of NaAsO2 gradually increases, cell viability, intracellular GSH content, ΔΨm, and the protein levels of GCLC and GCLM were reduced significantly. Apoptosis rate, ROS, and expression of apoptosis-related and NF-κB pathway proteins increased. The expression of LINC00942 was increased, while the expression of miR-214-5p was decreased. After suppressing LINC00942 levels, NaAsO2 exposure further decreased cell viability, intracellular GSH content, ΔΨm, GCLC protein, and miR-214-5p expression. The apoptosis rate, ROS, and apoptosis-related and NF-κB pathway proteins further increased. miR-214-5p is targeted and negatively regulated by LINC00942. After miR-214-5p was overexpressed, NaAsO2 further decreased cell viability, intracellular GSH content, ΔΨm, and GCLC protein expression compared to NaAsO2 exposure. The apoptosis rate, ROS, apoptosis-related and NF-κB pathway proteins p65, and IKKβ were higher than those exposed to NaAsO2. LINC00942 inhibitor along with miR-214-5p inhibitor combined with NaAsO2 treatment resulted in increased cell viability, GSH, Bcl-2, and GCLC protein expression and decreased apoptosis rate, apoptosis related, p65, IKKβ protein, and ΔΨm, as compared to the combined NaAsO2 and si LINC00942 group. NaAsO2 exposure induces oxidative damage and apoptosis in LX-2 cells by activating NF-κB and inhibiting GSH synthesis. During this process, the expression level of LINC00942 increases, targeting to reduce the level of miR-214-5p, then weakening the effect of NaAsO2 on NF-κB, thereby alleviating cellular oxidative damage and playing a protective role.
Collapse
Affiliation(s)
- Mingxiao Ma
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, China
| | - Jingyi Zhang
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, China
| | - Sheng Li
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, China
| | - Mengyao Zhang
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, China
| | - Weixin Chen
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, China
| | - Linzhi Li
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, China
| | - Shugang Li
- Department of Maternal and Children Health, School of Public Health, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
4
|
Li Y, Liang K, Yuan L, Gao J, Wei L, Zhao L. The role of thioredoxin and glutathione systems in arsenic-induced liver injury in rats under glutathione depletion. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:547-563. [PMID: 36528894 DOI: 10.1080/09603123.2022.2159016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Antioxidant systems like thioredoxin (Trx) and glutaredoxin (Grx) maintain oxidative stress balance. These systems have cross-talk supported by some in vitro studies. We investigated the underlying mechanisms of arsenic-induced liver injury in glutathione-deficient rats and whether there was any cross-talk between the Trx and Grx systems. The rats in arsenic-treated groups were administered with sodium arsenite (10, 20 mg/kg b w/d) for four weeks. In buthionine sulfoximine (BSO, an inhibitor of GSH) and 20 mg/kg arsenic combined groups, rats were injected with 2 mmol/kg BSO intraperitoneally twice per week. BSO exacerbated arsenic-induced liver injury by increasing arsenic accumulation in urine, serum, and liver while decreasing glutathione activity and resulting in upregulated mRNA expression of the Trx system and downregulation of Grx mRNA expression. The impact of Trx lasted longer than that of the Grx. The Trx system remained highly expressed, while GSH, Grx1, and Grx2 levels were decreased. The inhibitory effect of only BSO treatment on Grx1 and Grx2 was not pronounced. However, the combined impact of arsenic and BSO upregulated Trx expression, primarily related to further reduction of GSH. As a result, the suppressed Grxs were protected by the upregulated Trxs, which serve as a backup antioxidant defense system in the liver.
Collapse
Affiliation(s)
- Yuanyuan Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health and Family Planning Commission (23618504), Harbin, China
| | - Kun Liang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health and Family Planning Commission (23618504), Harbin, China
- Department of Science and Education, Bayan Nur Hospital, Bayan Nur, China
| | - Lin Yuan
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health and Family Planning Commission (23618504), Harbin, China
| | - Jing Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health and Family Planning Commission (23618504), Harbin, China
- Department of Public Health, Dalian Health Development Center, Dalian, China
| | - Linquan Wei
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health and Family Planning Commission (23618504), Harbin, China
| | - Lijun Zhao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health and Family Planning Commission (23618504), Harbin, China
| |
Collapse
|
5
|
Yamauchi H, Hitomi T, Takata A. Evaluation of arsenic metabolism and tight junction injury after exposure to arsenite and monomethylarsonous acid using a rat in vitro blood-Brain barrier model. PLoS One 2023; 18:e0295154. [PMID: 38032905 PMCID: PMC10688625 DOI: 10.1371/journal.pone.0295154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023] Open
Abstract
Experimental verification of impairment to cognitive abilities and cognitive dysfunction resulting from inorganic arsenic (iAs) exposure in children and adults is challenging. This study aimed to elucidate the effects of arsenite (iAsIII; 1, 10 and 20 μM) or monomethylarsonous acid (MMAIII; 0.1, 1 and 2 μM) exposure on arsenic metabolism and tight junction (TJ) function in the blood-brain barrier (BBB) using a rat in vitro-BBB model. The results showed that a small percentage (~15%) of iAsIII was oxidized or methylated within the BBB, suggesting the persistence of toxicity as iAsIII. Approximately 65% of MMAIII was converted to low-toxicity monomethylarsonic acid and dimethylarsenic acid via oxidation and methylation. Therefore, it is estimated that MMAIII causes TJ injury to the BBB at approximately 35% of the unconverted level. TJ injury of BBB after iAsIII or MMAIII exposure could be significantly assessed from decreased expression of claudin-5 and decreased transepithelial electrical resistance values. TJ injury in BBB was found to be significantly affected by MMAIII than iAsIII. Relatedly, the penetration rate in the BBB by 24 h of exposure was higher for MMAIII (53.1% ± 2.72%) than for iAsIII (43.3% ± 0.71%) (p < 0.01). Exposure to iAsIII or MMAIII induced an antioxidant stress response, with concentration-dependent increases in the expression of nuclear factor-erythroid 2-related factor 2 in astrocytes and heme oxygenase-1 in a group of vascular endothelial cells and pericytes, respectively. This study found that TJ injury at the BBB is closely related to the chemical form and species of arsenic; we believe that elucidation of methylation in the brain is essential to verify the impairment of cognitive abilities and cognitive dysfunction caused by iAs exposure.
Collapse
Affiliation(s)
- Hiroshi Yamauchi
- Department of Preventive Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Toshiaki Hitomi
- Department of Preventive Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Ayako Takata
- Department of Preventive Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
6
|
González-Alfonso WL, Pavel P, Karina HM, Del Razo LM, Sanchez-Peña LC, Zepeda A, Gonsebatt ME. Chronic exposure to inorganic arsenic and fluoride induces redox imbalance, inhibits the transsulfuration pathway, and alters glutamate receptor expression in the brain, resulting in memory impairment in adult male mouse offspring. Arch Toxicol 2023; 97:2371-2383. [PMID: 37482551 PMCID: PMC10404204 DOI: 10.1007/s00204-023-03556-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Exposure to toxic elements in drinking water, such as arsenic (As) and fluoride (F), starts at gestation and has been associated with memory and learning deficits in children. Studies in which rodents underwent mechanistic single exposure to As or F showed that the neurotoxic effects are associated with their capacity to disrupt redox balance, mainly by diminishing glutathione (GSH) levels, altering glutamate disposal, and altering glutamate receptor expression, which disrupts synaptic transmission. Elevated levels of As and F are common in groundwater worldwide. To explore the neurotoxicity of chronic exposure to As and F in drinking water, pregnant CD-1 mice were exposed to 2 mg/L As (sodium arsenite) and 25 mg/L F (sodium fluoride) alone or in combination. The male litter continued to receive exposure up to 30 or 90 days after birth. The effects of chronic exposure on GSH levels, transsulfuration pathway enzymatic activity, expression of cysteine/cystine transporters, glutamate transporters, and ionotropic glutamate receptor subunits as well as behavioral performance in the object recognition memory task were assessed. Combined exposure resulted in a significant reduction in GSH levels in the cortex and hippocampus at different times, decreased transsulfuration pathway enzyme activity, as well as diminished xCT protein expression. Altered glutamate receptor expression in the cortex and hippocampus and decreased transaminase enzyme activity were observed. These molecular alterations were associated with memory impairment in the object recognition task, which relies on these brain regions.
Collapse
Affiliation(s)
- Wendy L González-Alfonso
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México
| | - Petrosyan Pavel
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México
| | - Hernández-Mercado Karina
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México
| | - Luz M Del Razo
- Departamento de Toxicología, Centro de Investigación Y Estudios Avanzados, Mexico, DF, Mexico
| | - Luz C Sanchez-Peña
- Departamento de Toxicología, Centro de Investigación Y Estudios Avanzados, Mexico, DF, Mexico
| | - Angélica Zepeda
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México
| | - María E Gonsebatt
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México.
| |
Collapse
|
7
|
Chen Y, Liu X, Zhang Q, Wang H, Zhang R, Ge Y, Liang H, Li W, Fan J, Liu H, Lv Z, Dou W, Wang Y, Li X. Arsenic induced autophagy-dependent apoptosis in hippocampal neurons via AMPK/mTOR signaling pathway. Food Chem Toxicol 2023; 179:113954. [PMID: 37481228 DOI: 10.1016/j.fct.2023.113954] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Arsenic contamination of groundwater remains a serious public health problem worldwide. Arsenic-induced neurotoxicity receives increasing attention, however, the mechanism remains unclear. Hippocampal neuronal death is regarded as the main event of arsenic-induced cognitive dysfunction. Mitochondria lesion is closely related to cell death, however, the effects of arsenic on PGAM5-regulated mitochondrial dynamics has not been documented. Crosstalk between autophagy and apoptosis is complicated and autophagy has a dual role in the apoptosis pathways in neuronal cells. In this study, arsenic exposure resulted in mitochondrial PGAM5 activation and subsequent activation of apoptosis and AMPK-mTOR dependent autophagy. Intervention by autophagy activator Rapamycin or inhibitor 3-MA, both targeting at mTOR, accordingly induced activation or inhibition of apoptosis. Intervention by MK-3903 or dorsomorphin, activator or inhibitor of AMPK, received similar results. Our findings suggested that arsenic-induced PGAM5 activation played a role in AMPK-mTOR dependent autophagy and arsenic induced autophagy-dependent apoptosis in hippocampal neurons via AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yao Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Xudan Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Qianhui Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Huanhuan Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Ruo Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Yanhong Ge
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Huning Liang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Wanying Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Juanjun Fan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Huimin Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Zhengyang Lv
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Wenting Dou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China
| | - Yi Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China.
| | - Xin Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, China.
| |
Collapse
|
8
|
Aschner M, Skalny AV, Ke T, da Rocha JBT, Paoliello MMB, Santamaria A, Bornhorst J, Rongzhu L, Svistunov AA, Djordevic AB, Tinkov AA. Hydrogen Sulfide (H 2S) Signaling as a Protective Mechanism against Endogenous and Exogenous Neurotoxicants. Curr Neuropharmacol 2022; 20:1908-1924. [PMID: 35236265 PMCID: PMC9886801 DOI: 10.2174/1570159x20666220302101854] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/10/2022] [Accepted: 02/27/2022] [Indexed: 11/22/2022] Open
Abstract
In view of the significant role of H2S in brain functioning, it is proposed that H2S may also possess protective effects against adverse effects of neurotoxicants. Therefore, the objective of the present review is to discuss the neuroprotective effects of H2S against toxicity of a wide spectrum of endogenous and exogenous agents involved in the pathogenesis of neurological diseases as etiological factors or key players in disease pathogenesis. Generally, the existing data demonstrate that H2S possesses neuroprotective effects upon exposure to endogenous (amyloid β, glucose, and advanced-glycation end-products, homocysteine, lipopolysaccharide, and ammonia) and exogenous (alcohol, formaldehyde, acrylonitrile, metals, 6-hydroxydopamine, as well as 1-methyl-4-phenyl- 1,2,3,6- tetrahydropyridine (MPTP) and its metabolite 1-methyl-4-phenyl pyridine ion (MPP)) neurotoxicants. On the one hand, neuroprotective effects are mediated by S-sulfhydration of key regulators of antioxidant (Sirt1, Nrf2) and inflammatory response (NF-κB), resulting in the modulation of the downstream signaling, such as SIRT1/TORC1/CREB/BDNF-TrkB, Nrf2/ARE/HO-1, or other pathways. On the other hand, H2S appears to possess a direct detoxicative effect by binding endogenous (ROS, AGEs, Aβ) and exogenous (MeHg) neurotoxicants, thus reducing their toxicity. Moreover, the alteration of H2S metabolism through the inhibition of H2S-synthetizing enzymes in the brain (CBS, 3-MST) may be considered a significant mechanism of neurotoxicity. Taken together, the existing data indicate that the modulation of cerebral H2S metabolism may be used as a neuroprotective strategy to counteract neurotoxicity of a wide spectrum of endogenous and exogenous neurotoxicants associated with neurodegeneration (Alzheimer's and Parkinson's disease), fetal alcohol syndrome, hepatic encephalopathy, environmental neurotoxicant exposure, etc. In this particular case, modulation of H2S-synthetizing enzymes or the use of H2S-releasing drugs should be considered as the potential tools, although the particular efficiency and safety of such interventions are to be addressed in further studies.
Collapse
Affiliation(s)
- Michael Aschner
- Address correspondence to this author at the Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; E-mail
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Siracusa R, Voltarelli VA, Trovato Salinaro A, Modafferi S, Cuzzocrea S, Calabrese EJ, Di Paola R, Otterbein LE, Calabrese V. NO, CO and H 2S: A Trinacrium of Bioactive Gases in the Brain. Biochem Pharmacol 2022; 202:115122. [PMID: 35679892 DOI: 10.1016/j.bcp.2022.115122] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/02/2022]
Abstract
Oxygen and carbon dioxide are time honored gases that have direct bearing on almost all life forms, but over the past thirty years, and in large part due to the Nobel Prize Award in Medicine for the elucidation of nitric oxide (NO) as a bioactive gas, the research and medical communities now recognize other gases as critical for survival. In addition to NO, hydrogen sulfide (H2S) and carbon monoxide (CO) have emerged as a triumvirate or Trinacrium of gases with analogous importance and that serve important homeostatic functions. Perhaps, one of the most intriguing aspects of these gases is the functional interaction between them, which is intimately linked by the enzyme systems that produce them. Despite the need to better understand NO, H2S and CO biology, the notion that these are environmental pollutants remains ever present. For this reason, incorporating the concept of hormesis becomes imperative and must be included in discussions when considering developing new therapeutics that involve these gases. While there is now an enormous literature base for each of these gasotransmitters, we provide here an overview of their respective physiologic roles in the brain.
Collapse
Affiliation(s)
- Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, 98166, Italy
| | - Vanessa A Voltarelli
- Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, 98166, Italy
| | - Edward J Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA
| | - Rosanna Di Paola
- Department of Veterinary Science, University of Messina, 98168, Messina, Italy
| | - Leo E Otterbein
- Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA.
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
10
|
Du X, Luo L, Huang Q, Zhang J. Cortex metabolome and proteome analysis reveals chronic arsenic exposure via drinking water induces developmental neurotoxicity through hnRNP L mediated mitochondrial dysfunction in male rats. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 820:153325. [PMID: 35074374 DOI: 10.1016/j.scitotenv.2022.153325] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/09/2022] [Accepted: 01/18/2022] [Indexed: 06/14/2023]
Abstract
Lots of people are at the risk of arsenic-contaminated drinking water. Arsenic exposure was confirmed to be closely linked to neurocognitive deficits, particularly during childhood. The multi-omics approaches are known be well suitable for toxicological research. Thus, this study aimed to explore the molecular mechanisms of arsenic-induced learning and memory function impairments through the integrative proteome and metabolome analysis of cortex in rats. The weaned rats were exposed to arsenic-contaminated drinking water for six months to mimic the developmental exposure. 220 differential proteins and 19 differential metabolites were identified in the cortex, and nine potential biomarkers were found to be related to impaired Morris water maze (MWM) indicators. Chronic arsenic exposure affected the cognitive function by inducing the overproduction of amyloid-β (Aβ) peptides and the redox imbalance in the mitochondria. Glycolysis and tricarboxylic acid (TCA) cycle enhancement driven by the increased heterogeneous nuclear ribonucleoprotein L (hnRNP L) is a low-dose protective mechanism against arsenic-induced ATP deficiency and oxidative stress. Moreover, apoptosis is another important pathway of arsenic-induced neurotoxicity. This study provides new evidence about the alterations of proteins and metabolites in the cortex of the exposed rats under arsenic toxicity. These findings suggest hnRNP L could be a potential target for the treatment of arsenic-induced neurotoxicity.
Collapse
Affiliation(s)
- Xiaoyan Du
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China; Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Lianzhong Luo
- Department of Pharmacy, Xiamen Medical College, China
| | - Qingyu Huang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, China
| | - Jie Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China.
| |
Collapse
|
11
|
Yang B, Yin C, Zhang Y, Xing G, Wang S, Li F, Aschner M, Lu R. Differential effects of subchronic acrylonitrile exposure on hydrogen sulfide levels in rat blood, brain, and liver. Toxicol Res (Camb) 2022; 11:374-384. [PMID: 35510234 PMCID: PMC9052317 DOI: 10.1093/toxres/tfac011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/14/2022] [Accepted: 02/26/2022] [Indexed: 11/14/2022] Open
Abstract
Background Hydrogen sulfide (H2S), as the third gasotransmitter participates in both cellular physiological and pathological processes, including chemical-induced injuries. We recently reported acute acrylonitrile (AN) treatment inhibited endogenous H2S biosynthesis pathway in rat and astrocyte models. However, there is still no evidence to address the correlation between endogenous H2S and sub-chronic AN exposure. Objectives This study aims to explore the modulatory effects of prolonged AN exposure on endogenous H2S levels and its biosynthetic enzymes in rat blood, brain and liver. Methods A total of 50 male Sprague-Dawley rats were randomly divided into 5 groups, including the control group and AN-treated groups at dosages of 6.25, 12.5, 25 or 50 mg/kg. Rats received one exposure/day, 5 days/week, for 4 consecutive weeks. The rat bodyweight and brain/liver organ coefficient were detected, along with liver cytochrome P450 2E1(CYP2E1) expression. In addition, the H2S contents in rat serum and plasma, and in cerebral cortex and liver tissues were measured by methylene blue method. The expression of H2S-generating enzymes, including cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MPST) was also measured with Western blot both in rat cerebral cortex and liver. Results Subchronic exposure to AN significantly inhibited bodyweight-gain and increased the liver CYP2E1 expression compared with the control. In addition, AN significantly increased H2S levels in rat plasma and serum, but not in liver. The endogenous H2S level in rat cerebral cortex was also significantly increased upon AN treatment, when expression of the major H2S-generating enzymes, CBS and 3-MPST were significantly enhanced. However, hepatic protein levels of CBS and CSE were significantly increased, whereas hepatic levels of 3-MPST were significantly decreased. Conclusion This study showed that sub-chronic AN exposure increased endogenous H2S contents in rat blood and brain tissues, but not liver, which may be resulted from the distinct expression profile of H2S-producing enzymes in response to AN. The blood H2S contents may be applied as a potential novel biomarker for surveillance of chronically AN-exposed populations. Highlights Subchronic intraperitoneal exposure to acrylonitrile increased H2S content in rat blood and cerebral cortex, but not in liver.Distinct tissue expression profiles of H2S-producing enzymes contribute to the acrylonitrile-induced differential effects on the H2S level.Blood H2S level may be a biomarker for subchronic exposure to acrylonitrile.
Collapse
Affiliation(s)
- Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Changsheng Yin
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Yu Zhang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Guangwei Xing
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Suhua Wang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Fang Li
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
- Center for Experimental Research, Affiliated Kunshan Hospital to Jiangsu University School of Medicine, Kunshan, Suzhou, Jiangsu 215300, China
| |
Collapse
|
12
|
Mehta K, Kaur B, Pandey KK, Dhar P, Kaler S. Resveratrol protects against inorganic arsenic-induced oxidative damage and cytoarchitectural alterations in female mouse hippocampus. Acta Histochem 2021; 123:151792. [PMID: 34634674 DOI: 10.1016/j.acthis.2021.151792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/04/2023]
Abstract
Prolonged inorganic arsenic (iAs) exposure is widely associated with brain damage particularly in the hippocampus via oxidative and apoptotic pathways. Resveratrol (RES) has gained considerable attention because of its benefits to human health. However, its neuroprotective potential against iAs-induced toxicity in CA1 region of hippocampus remains unexplored. Therefore, we investigated the neuroprotective efficacy of RES against arsenic trioxide (As2O3)-induced adverse effects on neuronal morphology, apoptotic markers and oxidative stress parameters in mouse CA1 region (hippocampus). Adult female Swiss albino mice of reproductive maturity were orally exposed to either As2O3 (2 and 4 mg/kg bw) alone or in combination with RES (40 mg/kg bw) for a period of 45 days. After animal sacrifice on day 46, the perfusion fixed brain samples were used for the observation of neuronal morphology and studying the morphometric features. While the freshly dissected hippocampi were processed for biochemical estimation of oxidative stress markers and western blotting of apoptosis-associated proteins. Chronic iAs exposure led to significant decrease in Stratum Pyramidale layer thickness along with reduction in cell density and area of Pyramidal neurons in contrast to the controls. Biochemical analysis showed reduced hippocampal GSH content but no change in total nitrite (NO) levels following iAs exposure. Western blotting showed apparent changes in the expression levels of Bax and Bcl-2 proteins following iAs exposure, however the change was statistically insignificant. Contrastingly, iAs +RES co-treatment exhibited substantial reversal in morphological and biochemical observations. Together, these findings provide preliminary evidence of neuroprotective role of RES on structural and biochemical alterations pertaining to mouse hippocampus following chronic iAs exposure.
Collapse
Affiliation(s)
- K Mehta
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - B Kaur
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - K K Pandey
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - P Dhar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - S Kaler
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
13
|
Carmona A, Roudeau S, Ortega R. Molecular Mechanisms of Environmental Metal Neurotoxicity: A Focus on the Interactions of Metals with Synapse Structure and Function. TOXICS 2021; 9:toxics9090198. [PMID: 34564349 PMCID: PMC8471991 DOI: 10.3390/toxics9090198] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023]
Abstract
Environmental exposure to neurotoxic metals and metalloids such as arsenic, cadmium, lead, mercury, or manganese is a global health concern affecting millions of people worldwide. Depending on the period of exposure over a lifetime, environmental metals can alter neurodevelopment, neurobehavior, and cognition and cause neurodegeneration. There is increasing evidence linking environmental exposure to metal contaminants to the etiology of neurological diseases in early life (e.g., autism spectrum disorder) or late life (e.g., Alzheimer’s disease). The known main molecular mechanisms of metal-induced toxicity in cells are the generation of reactive oxygen species, the interaction with sulfhydryl chemical groups in proteins (e.g., cysteine), and the competition of toxic metals with binding sites of essential metals (e.g., Fe, Cu, Zn). In neurons, these molecular interactions can alter the functions of neurotransmitter receptors, the cytoskeleton and scaffolding synaptic proteins, thereby disrupting synaptic structure and function. Loss of synaptic connectivity may precede more drastic alterations such as neurodegeneration. In this article, we will review the molecular mechanisms of metal-induced synaptic neurotoxicity.
Collapse
|
14
|
SLC7A11 regulated by NRF2 modulates esophageal squamous cell carcinoma radiosensitivity by inhibiting ferroptosis. J Transl Med 2021; 19:367. [PMID: 34446045 PMCID: PMC8393811 DOI: 10.1186/s12967-021-03042-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/14/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Solute carrier family 7 member 11(SLC7A11) is a component of cysteine/glutamate transporter, which plays a key role in tumor growth; however, its underlying effect on radiosensitivity in esophageal squamous cell carcinoma (ESCC) remains unclear. This study aimed to clarify SLC7A11's expression and correlation with nuclear expression of nuclear factor erythroid-2 (NRF2)-associated radioresistance in ESCC. METHODS We included 127 ESCC patients who received radical chemoradiotherapy. Immunohistochemical staining was used to detect SLC7A11 and NRF2 nuclear expression, and the relationship between clinicopathological characteristics and survival rates or therapy response were evaluated. Western blot, dual-reporter assays and Chromatin immunoprecipitation (ChIP)-sequencing were used to analyze their relationship in vitro. Their roles in radioresistance were then investigated through multiple validation steps. RESULTS NRF2 nuclear expression and SLC7A11 expression were overexpressed in ESCC tissues and were positively correlated with one another. NRF2 nuclear expression was significantly associated with tumor length, lymph node metastasis, and TNM stage, while SLC7A11 expression was associated with lymph node metastasis. Patients with high NRF2 nuclear expression and SLC7A11 expression had significantly shorter overall and progression-free survival, and poor treatment response. The multivariate model showed that NRF2 nuclear expression and SLC7A11 expression, sex and tumor location are independent prognostic factors. In vitro analysis confirmed that hyperactivation of NRF2 induced SLC7A11 expression by directly binding to its promoter region, promoting radioresistance, reducing radiotherapy-induced lipid peroxidation levels, PTGS2 expression, and radiotherapy-related ferroptosis morphologic features. CONCLUSION Our study reveals a connection between high SLC7A11 expression and NRF2 nuclear expression in patients with ESCC that was related to worse survival and poorer therapy outcomes. SLC7A11-mediated ferroptosis inhibition induced NRF2-associated radioresistance, highlighting potential of NRF2/SLC7A11/ferroptosis axis as future therapeutic targets against therapy resistance biomarker.
Collapse
|
15
|
Hammad AM, Swiss GMS, Hall FS, Hikmat S, Sari Y, Al-Qirim TM, Amawi HA. Ceftriaxone Reduces Waterpipe Tobacco Smoke Withdrawal-induced Anxiety in rats via Modulating the Expression of TNF-α/NFĸB, Nrf2, and GLT-1. Neuroscience 2021; 463:128-142. [PMID: 33836247 DOI: 10.1016/j.neuroscience.2021.03.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 01/01/2023]
Abstract
Tobacco exposure has been linked to neuroinflammation and adaptive/maladaptive changes in neurotransmitter systems, including in glutamatergic systems. We examined the effects of waterpipe tobacco smoke (WTS) on inflammatory mediators and astroglial glutamate transporters in mesocorticolimbic brain regions including the prefrontal cortex (PFC), nucleus accumbens (NAc) and ventral tegmental area (VTA). The behavioral consequences of WTS exposure on withdrawal-induced anxiety-like behavior were assessed using elevated plus maze (EPM) and open field (OF) tests. Male Sprague-Dawley rats were randomly assigned to 3 experimental groups: a control group exposed only to standard room air, a WTS exposed group treated with saline vehicle, and a WTS exposed group treated with ceftriaxone. WTS exposure was performed for 2 h/day, 5 days/week, for 4 weeks. Behavioral tests (EPM and OF) were conducted weekly 24 h after WTS exposure, during acute withdrawal. During week 4, rats were given either saline or ceftriaxone (200 mg/kg i.p.) 30 min before WTS exposure. WTS increased withdrawal-induced anxiety, and ceftriaxone attenuated this effect. WTS exposure increased the relative mRNA levels for nuclear factor ĸB (NFĸB), tumor necrosis factor-α (TNF-α), and brain-derived neurotrophic factor (BDNF) in the PFC, NAc and VTA, and ceftriaxone treatment reversed these effects. In addition, WTS decreased the relative mRNA of nuclear factor erythroid 2 related factor 2 (Nrf2), glutamate transporter 1 (GLT-1) and cystine-glutamate transporter (xCT) in PFC, NAc and VTA, and ceftriaxone treatment normalized their expression. WTS caused neuroinflammation, alteration in relative mRNA glutamate transport expression, and increased anxiety-like behavior, and these effects were attenuated by ceftriaxone treatment.
Collapse
Affiliation(s)
- Alaa M Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan.
| | - Ghadeer M S Swiss
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Suhair Hikmat
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - T M Al-Qirim
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - H A Amawi
- Faculty of Pharmacy, Yarmouk University, Irbid 21110, Jordan
| |
Collapse
|
16
|
Li C, Zhang S, Li L, Hu Q, Ji S. Ursodeoxycholic Acid Protects Against Arsenic Induced Hepatotoxicity by the Nrf2 Signaling Pathway. Front Pharmacol 2020; 11:594496. [PMID: 33178028 PMCID: PMC7596389 DOI: 10.3389/fphar.2020.594496] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Arsenic is ubiquitous toxic metalloid responsible for many human diseases all over the world. Contrastingly, Ursodeoxycholic acid (UDCA) has been suggested as efficient antioxidant in various liver diseases. However, there are no reports of the effects of UDCA on arsenious acid [As(III)]-induced hepatotoxicity. The objective of this study is to elucidate the protective actions of UDCA on As(III)-induced hepatotoxicity and explore its controlling role in biomolecular mechanisms in vivo and in vitro. The remarkable liver damage induced by As(III) was ameliorated by treatment with UDCA, as reflected by reduced histopathological changes of liver and elevation of serum AST, ALT levels. UDCA play a critical role in stabilization of cellular membrane potential, inhibition of apoptosis and LDH leakage in LO2 cells. Meanwhile, the activities of SOD, CAT and GSH-Px and the level of TSH, GSH were enhanced with UDCA administration, while the accumulations of intracellular ROS, MDA and rate of GSSG/GSH were decreased in vivo and in vitro. Further study disclosed that UDCA significantly inhibited As(III)-induced apoptosis through increasing the expression of Bcl-2 and decreasing the expression of Bax, p53, Cyt C, Cleaved caspase-3 and 9. Moreover, UDCA promoted the expression of nuclear Nrf2, HO-1, and NQO1, although arsenic regulated nuclear translocation of Nrf2 positively. When Nrf2 was silenced, the protective effect of UDCA was abolished. Collectively, the results of this study showed that UDCA protects hepatocytes antagonize As(III)-induced cytotoxicity, and its mechanism may be related to activation of Nrf2 signaling.
Collapse
Affiliation(s)
- Chao Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Sheng Zhang
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China.,School of Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Liming Li
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Qing Hu
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Shen Ji
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| |
Collapse
|