1
|
Cestonaro LV, Conte AM, Goldoni FC, Quintão NLM, Garcia SC, Santin JR, Arbo MD. In vitro immunotoxic evaluation of herbicides in RAW 264.7 cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:433-446. [PMID: 39812396 DOI: 10.1080/15287394.2025.2450418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Weeds are a concern in agriculture and the use of herbicides constitutes an effective, efficient, and economical way to control their growth. Recent discoveries of herbicides are promising for the management of resistant weeds. However, there is a gap in the knowledge of the toxic effects of some herbicides previously reported on immune cells. The present study aimed to examine cellular immunotoxicity of three herbicides (clomazone, glyphosate, and sulfentrazone) after 96 hr incubation utilizing RAW 264.7 BALB/c mouse monocyte/macrophage-like cell line to elucidate the role of some toxicological pathways. Data demonstrated the herbicides clomazone, glyphosate, and sulfentrazone initiated a cytotoxic effect as evidenced by EC50 values of 429.2; 53.7; 866.6 mg/L, respectively. Clomazone and sulfentrazone, at all concentrations, induced excess production of reactive oxygen (ROS) and reactive nitrogen (RNS) free radicals. An immunosuppression was observed in RAW 264.7 cells after incubation with 50 or 100 mg/L glyphosate and 500 or 1000 mg/L sulfentrazone. In addition, all herbicides produced mitochondrial depolarization and decreased tumor necrosis factor-α (TNF-α) levels. This constitutes the first report of the effects of clomazone and sulfentrazone on RAW 264.7 cells, including reduced TNF-α levels, indicating the adverse influence of herbicides on the immune system.
Collapse
Affiliation(s)
- Larissa Vivan Cestonaro
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Aline Mocellin Conte
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - Fernanda Capitanio Goldoni
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Vale do Itajaí - UNIVALI, Itajaí, SC, Brazil
| | - Nara Lins Meira Quintão
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Vale do Itajaí - UNIVALI, Itajaí, SC, Brazil
| | - Solange Cristina Garcia
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| | - José Roberto Santin
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Vale do Itajaí - UNIVALI, Itajaí, SC, Brazil
| | - Marcelo Dutra Arbo
- Laboratório de Toxicologia, Departamento de Análises, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
2
|
Jung YH, Jo HY, Kim DH, Oh YJ, Kim M, Na S, Song HY, Lee HJ. Exosome-Mediated Mitochondrial Regulation: A Promising Therapeutic Tool for Alzheimer's Disease and Parkinson's Disease. Int J Nanomedicine 2025; 20:4903-4917. [PMID: 40259919 PMCID: PMC12011032 DOI: 10.2147/ijn.s513816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are representative neurodegenerative diseases with abnormal energy metabolism and altered distribution and deformation of mitochondria within neurons, particularly in brain regions such as the hippocampus and substantia nigra. Neurons have high energy demands; thus, maintaining a healthy mitochondrial population is important for their biological function. Recently, exosomes have been reported to have mitochondrial regulatory potential and antineurodegenerative properties. This review presents the mitochondrial abnormalities in brain cells associated with AD and PD and the potential of exosomes to treat these diseases. Specifically, it recapitulates research on the molecular mechanisms whereby exosomes regulate mitochondrial biogenesis, fusion/fission dynamics, mitochondrial transport, and mitophagy. Furthermore, this review discusses exosome-triggered signaling pathways that regulate nuclear factor (erythroid-derived 2)-like 2-dependent mitochondrial antioxidation and hypoxia inducible factor 1α-dependent metabolic reprogramming. In summary, this review aims to provide a profound understanding of the regulatory effect of exosomes on mitochondrial function in neurons and to propose exosome-mediated mitochondrial regulation as a promising strategy for AD and PD.
Collapse
Affiliation(s)
- Young Hyun Jung
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Hyo Youn Jo
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Dae Hyun Kim
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Yeon Ju Oh
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Minsoo Kim
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Seunghyun Na
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| |
Collapse
|
3
|
Ye J, Dai X, Zhang C, Duan Z, Zhou G, Wang J. Investigating the causal relationships between mitochondrial proteins and dementia with Lewy bodies. J Alzheimers Dis 2025:13872877251328882. [PMID: 40111912 DOI: 10.1177/13872877251328882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BackgroundDisruptions in mitochondrial function have been implicated in various neurodegenerative diseases. However, the specific role of mitochondrial proteins in the pathogenesis of dementia with Lewy bodies (DLB) remains poorly understood.ObjectiveThis study aims to investigate potential causal relationships between mitochondrial proteins and DLB risk using Mendelian randomization (MR) analysis.MethodsCausal associations between 66 mitochondrial proteins (MPs) and DLB were assessed by MR analysis, utilizing data from comprehensive genome-wide association studies (GWAS), with various analytical methods, including the inverse variance weighted, MR-Egger, and weighted median. Cochran's Q statistics assessed the heterogeneity of instrumental variables.ResultsGenetic predispositions to increased levels of ES1 protein homolog and apoptosis-inducing factor 1 (AIF-1) were associated with an elevated risk of DLB. Conversely, genetic predispositions to increased levels of glutaredoxin-2 (GLRX-2), complement component 1 Q subcomponent-binding protein (C1QBP), and mitochondrial glutamate carrier 2 (GC2) were found to be protective against DLB. Sensitivity analyses revealed no heterogeneity or horizontal pleiotropy among the selected instrumental variables.ConclusionsOur MR study identifies specific MPs potentially causally linked to DLB risk. These findings offer new insights into the MP-related mechanisms underlying DLB pathogenesis and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Jingna Ye
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Xuelian Dai
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Canwen Zhang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Zhihui Duan
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Guoqing Zhou
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Juan Wang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| |
Collapse
|
4
|
Feng B, Lu J, Jiang W, Xu N, Sun W. Chlorpyrifos-oxon induced neuronal cell death via endoplasmic reticulum stress-triggered apoptosis pathways. Toxicol In Vitro 2024; 101:105939. [PMID: 39251113 DOI: 10.1016/j.tiv.2024.105939] [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: 11/14/2023] [Revised: 08/21/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Chlorpyrifos (CPF) is one of the organophosphorus pesticides widely used throughout the world. Epidemiological studies suggested a link between CPF exposure and neurologic disorders, while the molecular mechanisms remain inconclusive. In the present study, we investigated the impacts of chlorpyrifos-oxon (CPO), the major toxic CPF metabolite, on cell apoptosis, and explored possible mechanism associated with endoplasmic reticulum (ER) stress in SH-SY5Y cells. Results showed that CPO exposure induced dose-dependent apoptosis and expression of ER stress-related proteins in SH-SY5Y cells. Pretreatment with 4-PBA (an ER stress inhibitor) effectively inhibited the expression of GRP78, GRP94, p-IRE1α, and XBP1-s, and apoptotic events. Pretreatment with STF-083010 (an IRE1α inhibitor) partially attenuated CPO-induced apoptosis. In addition, CPO exposure significantly evoked the generation of reactive oxygen species (ROS) which could be eliminated by pretreatment of 4-PBA. Of note, buffering the ROS generation with antioxidant NAC had little impact on the expression of p-IRE1α, and only partially attenuated CPO-induced apoptosis. In contrast, co-pretreatment with NAC and STF-083010 effectively inhibited CPO-induced apoptotic events. Collectively, our results indicate that CPO exposure exerts neuronal cytotoxicity via ER stress downstream-regulated IRE1α/XBP1 signaling pathway and ROS generation-triggered apoptosis. These findings highlight the role of ER stress in CPF-induced neurotoxicity, and provide a promising target for the intervention of organophosphate-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Baihuan Feng
- Department of Infection Prevention and Control, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Jingchun Lu
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wei Jiang
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Nani Xu
- Xihu District Center for Disease Control and Prevention, Hangzhou, Zhejiang 310013, China.
| | - Wenjun Sun
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
5
|
Hosen MHA, Whitworth DJ, Leusch FDL, Yuen N, Bengtson Nash SM. Bioenergetic Shifts in Humpback Whale Fibroblasts Upon Chemical Exposure. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:12313-12319. [PMID: 38958666 DOI: 10.1021/acs.est.3c10595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Southern Hemisphere humpback whales accumulate persistent and toxic chemicals, which are transported to Antarctica through distant sources and in situ usage. The extreme seasonal migration-associated fast of humpback whales results in the remobilization of persistent and lipophilic environmental contaminants from liberated fat stores. Mitochondria play a key role in lipid metabolism, and any disruption to mitochondrial function is expected to influence whole-organism bioenergetics. It is therefore of interest to advance understanding of the impact of known contaminants of the Antarctic sea-ice ecosystem upon humpback whale cellular bioenergetics. Using cell line-based in vitro testing, this study employed the Seahorse Extracellular Flux Analyzer to study cellular metabolic activity in live humpback whale fibroblast cells. The assay, based on oxygen consumption rate, provides insights into the cause of cellular bioenergetic disruption. Immortalized skin fibroblasts were exposed to four priority environmental chemicals found in the Antarctic sea-ice ecosystem. Our findings reveal chemical-dependent functional alterations and varying bioenergetic profile responses. Chlorpyrifos was observed to decrease mitochondrial basal oxygen consumption; dieldrin increased basal oxygen consumption; trifluralin's impact was dose-specific, and endosulfan displayed no effect. Our results provide unique insights into environmental chemical mechanisms of action on cellular bioenergetics, generating much-needed taxa-specific chemical effect data in support of evidence-based conservation policy and management.
Collapse
Affiliation(s)
- Md Hafiz All Hosen
- Centre for Planetary Health and Food Security, School of Environment and Science, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| | - Deanne J Whitworth
- The School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Frederic D L Leusch
- Australian Rivers Institute, School of Environment and Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Nicholas Yuen
- The School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Susan M Bengtson Nash
- Centre for Planetary Health and Food Security, School of Environment and Science, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| |
Collapse
|
6
|
Lința AV, Lolescu BM, Ilie CA, Vlad M, Blidișel A, Sturza A, Borza C, Muntean DM, Crețu OM. Liver and Pancreatic Toxicity of Endocrine-Disruptive Chemicals: Focus on Mitochondrial Dysfunction and Oxidative Stress. Int J Mol Sci 2024; 25:7420. [PMID: 39000526 PMCID: PMC11242905 DOI: 10.3390/ijms25137420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
In recent years, the worldwide epidemic of metabolic diseases, namely obesity, metabolic syndrome, diabetes and metabolic-associated fatty liver disease (MAFLD) has been strongly associated with constant exposure to endocrine-disruptive chemicals (EDCs), in particular, the ones able to disrupt various metabolic pathways. EDCs have a negative impact on several human tissues/systems, including metabolically active organs, such as the liver and pancreas. Among their deleterious effects, EDCs induce mitochondrial dysfunction and oxidative stress, which are also the major pathophysiological mechanisms underlying metabolic diseases. In this narrative review, we delve into the current literature on EDC toxicity effects on the liver and pancreatic tissues in terms of impaired mitochondrial function and redox homeostasis.
Collapse
Affiliation(s)
- Adina V. Lința
- Department of Functional Sciences—Chair of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.S.); (C.B.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania
| | - Bogdan M. Lolescu
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania
| | - Cosmin A. Ilie
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
- Department of Functional Sciences—Chair of Public Health & Sanitary Management, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Mihaela Vlad
- Department of Internal Medicine II—Chair of Endocrinology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq., No. 2, 300041 Timișoara, Romania;
| | - Alexandru Blidișel
- Department of Surgery I—Chair of Surgical Semiotics & Thoracic Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timişoara, Romania; (A.B.); (O.M.C.)
- Centre for Hepato-Biliary and Pancreatic Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timişoara, Romania
| | - Adrian Sturza
- Department of Functional Sciences—Chair of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.S.); (C.B.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
| | - Claudia Borza
- Department of Functional Sciences—Chair of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.S.); (C.B.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
| | - Danina M. Muntean
- Department of Functional Sciences—Chair of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.V.L.); (A.S.); (C.B.)
- Centre for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timișoara, Romania; (B.M.L.); (C.A.I.)
| | - Octavian M. Crețu
- Department of Surgery I—Chair of Surgical Semiotics & Thoracic Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timişoara, Romania; (A.B.); (O.M.C.)
- Centre for Hepato-Biliary and Pancreatic Surgery, “Victor Babeș” University of Medicine and Pharmacy of Timișoara, E. Murgu Sq. No. 2, 300041 Timişoara, Romania
| |
Collapse
|
7
|
Afsheen S, Rehman AS, Jamal A, Khan N, Parvez S. Understanding role of pesticides in development of Parkinson's disease: Insights from Drosophila and rodent models. Ageing Res Rev 2024; 98:102340. [PMID: 38759892 DOI: 10.1016/j.arr.2024.102340] [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: 03/10/2024] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/19/2024]
Abstract
Parkinson's disease is a neurodegenerative illness linked to ageing, marked by the gradual decline of dopaminergic neurons in the midbrain. The exact aetiology of Parkinson's disease (PD) remains uncertain, with genetic predisposition and environmental variables playing significant roles in the disease's frequency. Epidemiological data indicates a possible connection between pesticide exposure and brain degeneration. Specific pesticides have been associated with important characteristics of Parkinson's disease, such as mitochondrial dysfunction, oxidative stress, and α-synuclein aggregation, which are crucial for the advancement of the disease. Recently, many animal models have been developed for Parkinson's disease study. Although these models do not perfectly replicate the disease's pathology, they provide valuable insights that improve our understanding of the condition and the limitations of current treatment methods. Drosophila, in particular, has been useful in studying Parkinson's disease induced by toxins or genetic factors. The review thoroughly analyses many animal models utilised in Parkinson's research, with an emphasis on issues including pesticides, genetic and epigenetic changes, proteasome failure, oxidative damage, α-synuclein inoculation, and mitochondrial dysfunction. The text highlights the important impact of pesticides on the onset of Parkinson's disease (PD) and stresses the need for more research on genetic and mechanistic alterations linked to the condition.
Collapse
Affiliation(s)
- Saba Afsheen
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Ahmed Shaney Rehman
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Nazia Khan
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
8
|
Abdelhafez HEDH, Abdallah AA, Abdel-Razik RK, Hamed NA, Elshatory A, Awad W, Khalaf AAA, Mekkawy AM. Sex comparison of oxidative stress, mitochondrial dysfunction, and apoptosis triggers induced by single-dose Abamectin in albino rats. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 201:105903. [PMID: 38685225 DOI: 10.1016/j.pestbp.2024.105903] [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: 02/05/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 05/02/2024]
Abstract
Abamectin (AB) is widely used in agriculture and has been employed as an insecticide, nematicide, and livestock pest control agent. However, it may also pose a serious threat to mammals. The primary purpose of this research was to compare the sex variations between male and female rats during exposure and to assess the risk of toxicity of abamectin, which are still largely unknown. The twenty albino rats were divided randomly into four groups (n = 5): 1) the male control group; 2) the male treatment group treated with AB (1 mg/kg B.W.); 3) the female control group; and 4) the female treatment group treated with AB (1 mg/kg B.W.). AB administration caused a drop in body weight in females more than males with showing oxidative stress in both sexes of animals, as characterized by an increase in MDA content and a decrease in glutathione (GSH) content and superoxide dismutase (SOD) activity. Reported sex-specific effects suggested that females are more susceptible from males in brain tissues for alteration of antioxidant markers while females' liver and kidney tissues showed more level of lipid peroxidation than males. In addition, mitochondrial dysfunction was associated with a significant decrease in NADH dehydrogenase (Complex I) and a significant decrease in mitochondrial ATPase, which led to apoptosis and histopathological alterations in the targeted tissues, indicating that females are higher sensitive than males to these biological events. In brief, the results of this study led to female rats are generally more sensitive than male rats to neurobehavioral and hepatic complications associated with abamectin treatment. Further evaluation should be performed to determine the adverse outcome pathways involved and to determine the effects of sex on improving the risk assessment of abamectin in both sexes.
Collapse
Affiliation(s)
- Hossam El Din H Abdelhafez
- Mammalian and Aquatic Toxicology Department, Central Agricultural Pesticides Laboratory, Agricultural Research Center, P.O. Box 12618, Dokki, Giza, Egypt.
| | - Amr A Abdallah
- Mammalian and Aquatic Toxicology Department, Central Agricultural Pesticides Laboratory, Agricultural Research Center, P.O. Box 12618, Dokki, Giza, Egypt
| | - Reda K Abdel-Razik
- Mammalian and Aquatic Toxicology Department, Central Agricultural Pesticides Laboratory, Agricultural Research Center, P.O. Box 12618, Dokki, Giza, Egypt
| | - Nadia A Hamed
- Mammalian and Aquatic Toxicology Department, Central Agricultural Pesticides Laboratory, Agricultural Research Center, P.O. Box 12618, Dokki, Giza, Egypt
| | - Ahmed Elshatory
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Cairo University, Egypt
| | - Walaa Awad
- Clinical Pharmacy Department, Abo El-Reesh Al Mounira Hospital, Cairo University, Cairo, Egypt
| | - Abdel Azeim A Khalaf
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Aya M Mekkawy
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
9
|
Luo S, Wang D, Zhang Z. Post-translational modification and mitochondrial function in Parkinson's disease. Front Mol Neurosci 2024; 16:1329554. [PMID: 38273938 PMCID: PMC10808367 DOI: 10.3389/fnmol.2023.1329554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/21/2023] [Indexed: 01/27/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease with currently no cure. Most PD cases are sporadic, and about 5-10% of PD cases present a monogenic inheritance pattern. Mutations in more than 20 genes are associated with genetic forms of PD. Mitochondrial dysfunction is considered a prominent player in PD pathogenesis. Post-translational modifications (PTMs) allow rapid switching of protein functions and therefore impact various cellular functions including those related to mitochondria. Among the PD-associated genes, Parkin, PINK1, and LRRK2 encode enzymes that directly involved in catalyzing PTM modifications of target proteins, while others like α-synuclein, FBXO7, HTRA2, VPS35, CHCHD2, and DJ-1, undergo substantial PTM modification, subsequently altering mitochondrial functions. Here, we summarize recent findings on major PTMs associated with PD-related proteins, as enzymes or substrates, that are shown to regulate important mitochondrial functions and discuss their involvement in PD pathogenesis. We will further highlight the significance of PTM-regulated mitochondrial functions in understanding PD etiology. Furthermore, we emphasize the potential for developing important biomarkers for PD through extensive research into PTMs.
Collapse
Affiliation(s)
- Shishi Luo
- Institute for Future Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, Hengyang, Hunan, China
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Danling Wang
- Institute for Future Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, Hengyang, Hunan, China
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Zhuohua Zhang
- Institute for Future Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, Hengyang, Hunan, China
- Institute of Molecular Precision Medicine, Xiangya Hospital, Key Laboratory of Molecular Precision Medicine of Hunan Province and Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Bustamante-Barrientos FA, Luque-Campos N, Araya MJ, Lara-Barba E, de Solminihac J, Pradenas C, Molina L, Herrera-Luna Y, Utreras-Mendoza Y, Elizondo-Vega R, Vega-Letter AM, Luz-Crawford P. Mitochondrial dysfunction in neurodegenerative disorders: Potential therapeutic application of mitochondrial transfer to central nervous system-residing cells. J Transl Med 2023; 21:613. [PMID: 37689642 PMCID: PMC10493034 DOI: 10.1186/s12967-023-04493-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
Mitochondrial dysfunction is reiteratively involved in the pathogenesis of diverse neurodegenerative diseases. Current in vitro and in vivo approaches support that mitochondrial dysfunction is branded by several molecular and cellular defects, whose impact at different levels including the calcium and iron homeostasis, energetic balance and/or oxidative stress, makes it difficult to resolve them collectively given their multifactorial nature. Mitochondrial transfer offers an overall solution since it contains the replacement of damage mitochondria by healthy units. Therefore, this review provides an introducing view on the structure and energy-related functions of mitochondria as well as their dynamics. In turn, we summarize current knowledge on how these features are deregulated in different neurodegenerative diseases, including frontotemporal dementia, multiple sclerosis, amyotrophic lateral sclerosis, Friedreich ataxia, Alzheimer´s disease, Parkinson´s disease, and Huntington's disease. Finally, we analyzed current advances in mitochondrial transfer between diverse cell types that actively participate in neurodegenerative processes, and how they might be projected toward developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliana Lara-Barba
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Javiera de Solminihac
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Carolina Pradenas
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Luis Molina
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ana María Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaiso, Valparaiso, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| |
Collapse
|
11
|
Melanis K, Stefanou MI, Themistoklis KM, Papasilekas T. mTOR pathway - a potential therapeutic target in stroke. Ther Adv Neurol Disord 2023; 16:17562864231187770. [PMID: 37576547 PMCID: PMC10413897 DOI: 10.1177/17562864231187770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/27/2023] [Indexed: 08/15/2023] Open
Abstract
Stroke is ranked as the second leading cause of death worldwide and a major cause of long-term disability. A potential therapeutic target that could offer favorable outcomes in stroke is the mammalian target of rapamycin (mTOR) pathway. mTOR is a serine/threonine kinase that composes two protein complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), and is regulated by other proteins such as the tuberous sclerosis complex. Through a significant number of signaling pathways, the mTOR pathway can modulate the processes of post-ischemic inflammation and autophagy, both of which play an integral part in the pathophysiological cascade of stroke. Promoting or inhibiting such processes under ischemic conditions can lead to apoptosis or instead sustained viability of neurons. The purpose of this review is to examine the pathophysiological role of mTOR in acute ischemic stroke, while highlighting promising neuroprotective agents such as hamartin for therapeutic modulation of this pathway. The therapeutic potential of mTOR is also discussed, with emphasis on implicated molecules and pathway steps that warrant further elucidation in order for their neuroprotective properties to be efficiently tested in future clinical trials.
Collapse
Affiliation(s)
- Konstantinos Melanis
- Second Department of Neurology, School of Medicine and ‘Attikon’ University Hospital, National and Kapodistrian University of Athens, Rimini 1 Chaidari, Athens 12462, Greece
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, School of Medicine and ‘Attikon’ University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos M. Themistoklis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Neurosurgery, ‘Korgialenio, Benakio, H.R.C’. General Hospital of Athens, Athens, Greece
| | - Themistoklis Papasilekas
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Neurosurgery, ‘Korgialenio, Benakio, H.R.C’. General Hospital of Athens, Athens, Greece
| |
Collapse
|
12
|
Moyano P, Sola E, Naval MV, Guerra-Menéndez L, Fernández MDLC, del Pino J. Neurodegenerative Proteinopathies Induced by Environmental Pollutants: Heat Shock Proteins and Proteasome as Promising Therapeutic Tools. Pharmaceutics 2023; 15:2048. [PMID: 37631262 PMCID: PMC10458078 DOI: 10.3390/pharmaceutics15082048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Environmental pollutants' (EPs) amount and diversity have increased in recent years due to anthropogenic activity. Several neurodegenerative diseases (NDs) are theorized to be related to EPs, as their incidence has increased in a similar way to human EPs exposure and they reproduce the main ND hallmarks. EPs induce several neurotoxic effects, including accumulation and gradual deposition of misfolded toxic proteins, producing neuronal malfunction and cell death. Cells possess different mechanisms to eliminate these toxic proteins, including heat shock proteins (HSPs) and the proteasome system. The accumulation and deleterious effects of toxic proteins are induced through HSPs and disruption of proteasome proteins' homeostatic function by exposure to EPs. A therapeutic approach has been proposed to reduce accumulation of toxic proteins through treatment with recombinant HSPs/proteasome or the use of compounds that increase their expression or activity. Our aim is to review the current literature on NDs related to EP exposure and their relationship with the disruption of the proteasome system and HSPs, as well as to discuss the toxic effects of dysfunction of HSPs and proteasome and the contradictory effects described in the literature. Lastly, we cover the therapeutic use of developed drugs and recombinant proteasome/HSPs to eliminate toxic proteins and prevent/treat EP-induced neurodegeneration.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Emma Sola
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - María Victoria Naval
- Department of Pharmacology, Pharmacognosy and Bothanic, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Lucia Guerra-Menéndez
- Department of Physiology, Medicine School, San Pablo CEU University, 28003 Madrid, Spain
| | - Maria De la Cabeza Fernández
- Department of Chemistry and Pharmaceutical Sciences, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Javier del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
13
|
Luque-Campos N, Riquelme R, Molina L, Canedo-Marroquín G, Vega-Letter AM, Luz-Crawford P, Bustamante-Barrientos FA. Exploring the therapeutic potential of the mitochondrial transfer-associated enzymatic machinery in brain degeneration. Front Physiol 2023; 14:1217815. [PMID: 37576343 PMCID: PMC10416799 DOI: 10.3389/fphys.2023.1217815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Mitochondrial dysfunction is a central event in the pathogenesis of several degenerative brain disorders. It entails fission and fusion dynamics disruption, progressive decline in mitochondrial clearance, and uncontrolled oxidative stress. Many therapeutic strategies have been formulated to reverse these alterations, including replacing damaged mitochondria with healthy ones. Spontaneous mitochondrial transfer is a naturally occurring process with different biological functions. It comprises mitochondrial donation from one cell to another, carried out through different pathways, such as the formation and stabilization of tunneling nanotubules and Gap junctions and the release of extracellular vesicles with mitochondrial cargoes. Even though many aspects of regulating these mechanisms still need to be discovered, some key enzymatic regulators have been identified. This review summarizes the current knowledge on mitochondrial dysfunction in different neurodegenerative disorders. Besides, we analyzed the usage of mitochondrial transfer as an endogenous revitalization tool, emphasizing the enzyme regulators that govern this mechanism. Going deeper into this matter would be helpful to take advantage of the therapeutic potential of mitochondrial transfer.
Collapse
Affiliation(s)
- Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
- IMPACT-Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ricardo Riquelme
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Luis Molina
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | - Gisela Canedo-Marroquín
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
- Faculty of Dentistry, Universidad de los Andes, Santiago, Chile
| | - Ana María Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaiso, Valparaiso, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
- IMPACT-Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Felipe A. Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
- IMPACT-Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
14
|
Mesnage R, Omriouate H, Antoniou MN. Comparison of transcriptome alterations induced by pendimethalin or its commercial formulation Stomp Aqua in human MCF-7, MCF-10 A and MCF-12 A mammary epithelial cells. BMC Res Notes 2023; 16:62. [PMID: 37098576 PMCID: PMC10131388 DOI: 10.1186/s13104-023-06327-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 04/06/2023] [Indexed: 04/27/2023] Open
Abstract
OBJECTIVE The toxicology of herbicides, which are currently in use is under-explored. One highly used but under investigated herbicide is pendimethalin. Here we mined high-throughput data from the US National Toxicology Program (NTP) to identify whether pendimethalin possesses an estrogenic capability in human cells. We also evaluated effects of pendimethalin and its reference commercial formulated herbicide Stomp Aqua on the transcriptome profile of three human mammary epithelial cell lines, cancerous MCF-7 and non-cancerous MCF-10 A and MCF-12 A to see whether this compound could have endocrine disrupting effects and if co-formulants present in the commercial formulation could amplify its toxicity. RESULTS The data mined from the US NTP database suggests that pendimethalin activates estrogen receptors at a concentration of approximately 10?M. MCF-7, MCF-10A and MCF-12A cells were exposed to 10 ?M pendimethalin and Stomp Aqua at an equivalent concentration. Transcriptome analysis showed changes in gene expression patterns implying that pendimethalin affected ubiquitin-mediated proteolysis and the function of the spliceosome. The formulated pendimethalin product Stomp Aqua gave comparable effects suggesting pendimethalin was responsible for the observed transcriptome alterations. Given the lack of information on the exposure to this pesticide, our study prompts the need for biomonitoring studies, especially under occupational use scenarios, to understand if low level exposure to pendimethalin could have endocrine disrupting effects on populations exposed to this compound. A deeper understanding of the exposure and mechanisms of action of this endocrine-disrupting pesticide is needed.
Collapse
Affiliation(s)
- Robin Mesnage
- Gene Expression and Therapy Group, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, SE1 9RT, UK
- Buchinger Wilhelmi Clinic, Wilhelmi-Beck-Straße 27, 88662, Überlingen, Germany
| | - Helin Omriouate
- Gene Expression and Therapy Group, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, SE1 9RT, UK
| | - Michael N Antoniou
- Gene Expression and Therapy Group, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, SE1 9RT, UK.
| |
Collapse
|
15
|
Roy T, Chatterjee A, Swarnakar S. Rotenone induced neurodegeneration is mediated via cytoskeleton degradation and necroptosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119417. [PMID: 36581087 DOI: 10.1016/j.bbamcr.2022.119417] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/07/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022]
Abstract
Rotenone has widespread beneficial effects in agriculture, fisheries and animal husbandries; however prolonged exposure causes a detrimental effect on the health of personnel working in such industries. Rotenone during its extraction, formulation or usage may cross the blood brain barrier leading to neurodegeneration and the development of Parkinson's disease like symptoms. It is a known inhibitor of the mitochondrial ETC complex I and responsible for impairing the OXPHOS system. Our study showed that rotenone exposure results in an increased production of ROS and decreased ATP level along with a conspicuous loss of mitochondrial membrane potential in N2A cells. The transcription and expression pattern of cofilin, a key component of actin cytoskeleton, was also altered after rotenone exposure; leading to the actin cytoskeleton degradation. We further observed an increased expression, as well as activity of matrix metalloproteinase9 (MMP9) in rotenone exposed N2A cells; suggesting the involvement of inflammation upon rotenone exposure. Simultaneously, an opposite pattern was noticed for the tissue inhibitors of metalloproteinases-1 (TIMP-1) protein, which is a known modulator of MMP9 activity. Additionally, the localization of MMP9 along with alpha-synuclein, UCHL1 and cofilin suggested their close proximity and cross interaction upon rotenone treatment. Furthermore, we observed significant increase in the level of TNF-α upon rotenone exposure along with the phosphorylation of RIPK1, RIPK3 and MLKL that has been identified as the necroptosis markers leading to programmed necroptotic death.
Collapse
Affiliation(s)
- Tapasi Roy
- CSIR-Indian Institute of Chemical Biology, Infectious Diseases and Immunology Division, Kolkata, West Bengal, India
| | - Abhishek Chatterjee
- CSIR-Indian Institute of Chemical Biology, Infectious Diseases and Immunology Division, Kolkata, West Bengal, India
| | - Snehasikta Swarnakar
- CSIR-Indian Institute of Chemical Biology, Infectious Diseases and Immunology Division, Kolkata, West Bengal, India.
| |
Collapse
|
16
|
Neurodegeneration in a Regulatory Context: The Need for Speed. CURRENT OPINION IN TOXICOLOGY 2022. [DOI: 10.1016/j.cotox.2022.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
17
|
Piel S, Janowska JI, Ward JL, McManus MJ, Jose JS, Starr J, Sheldon M, Clayman CL, Elmér E, Hansson MJ, Jang DH, Karlsson M, Ehinger JK, Kilbaugh TJ. Succinate prodrugs in combination with atropine and pralidoxime protect cerebral mitochondrial function in a rodent model of acute organophosphate poisoning. Sci Rep 2022; 12:20329. [PMID: 36434021 PMCID: PMC9700731 DOI: 10.1038/s41598-022-24472-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Pesticides account for hundreds of millions of cases of acute poisoning worldwide each year, with organophosphates (OPs) being responsible for the majority of all pesticide-related deaths. OPs inhibit the enzyme acetylcholinesterase (AChE), which leads to impairment of the central- and peripheral nervous system. Current standard of care (SOC) alleviates acute neurologic-, cardiovascular- and respiratory symptoms and reduces short term mortality. However, survivors often demonstrate significant neurologic sequelae. This highlights the critical need for further development of adjunctive therapies with novel targets. While the inhibition of AChE is thought to be the main mechanism of injury, mitochondrial dysfunction and resulting metabolic crisis may contribute to the overall toxicity of these agents. We hypothesized that the mitochondrially targeted succinate prodrug NV354 would support mitochondrial function and reduce brain injury during acute intoxication with the OP diisopropylfluorophosphate (DFP). To this end, we developed a rat model of acute DFP intoxication and evaluated the efficacy of NV354 as adjunctive therapy to SOC treatment with atropine and pralidoxime. We demonstrate that NV354, in combination with atropine and pralidoxime therapy, significantly improved cerebral mitochondrial complex IV-linked respiration and reduced signs of brain injury in a rodent model of acute DFP exposure.
Collapse
Affiliation(s)
- Sarah Piel
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| | - Joanna I. Janowska
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| | - J. Laurenson Ward
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| | - Meagan J. McManus
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| | - Joshua S. Jose
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| | - Jonathan Starr
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| | - Malkah Sheldon
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| | - Carly L. Clayman
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| | - Eskil Elmér
- grid.4514.40000 0001 0930 2361Mitochondrial Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden ,Abliva AB, Lund, Sweden
| | - Magnus J. Hansson
- grid.4514.40000 0001 0930 2361Mitochondrial Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden ,Abliva AB, Lund, Sweden
| | - David H. Jang
- grid.25879.310000 0004 1936 8972Division of Medical Toxicology, Department of Emergency Medicine, University of Pennsylvania School of Medicine, Philadelphia, USA
| | - Michael Karlsson
- grid.475435.4Department of Neurosurgery, Rigshospitalet, Copenhagen, Denmark
| | - Johannes K. Ehinger
- grid.4514.40000 0001 0930 2361Mitochondrial Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden ,grid.4514.40000 0001 0930 2361Otorhinolaryngology, Head and Neck Surgery, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Todd J. Kilbaugh
- grid.239552.a0000 0001 0680 8770Resuscitation Science Center of Emphasis, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia, Philadelphia, USA
| |
Collapse
|
18
|
Zaki MSA, El-Kott AF, AlGwaiz HIM, Sideeg AM, Andarawi M, Eid RA. The effectiveness of vitamin C on quinalphos ileal toxicity: a study of histological, ultrastructural, and oxidative stress markers. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:57896-57904. [PMID: 35359206 DOI: 10.1007/s11356-022-19820-9] [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: 01/12/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
There is a significant hazard of human exposure to the organophosphates which is a constant threat, and they are responsible for numerous cases of poisoning and mammalian toxicity annually in non-target wildlife. The antioxidants, including the vitamin C (Vit C), have a protective effect on some organophosphorus compounds-induced organ damage. Quinalphos (QP) is one of these compounds. The investigation's objective is to see if there was any effect of QP on the rat ileum which could be rectified by using Vit C. Three groups of 24 animals were created. As a control, the first group was given pure water. Second group subjected to oral gavages of QPs. Third group rats were given oral gavages of Vit C plus QPs for 10 days. The reaction of ileal enterocytes to food-borne QPs was marked by poorly organized microvilli, numerous vacuoles within them, disrupted nuclei with chromatin margination, disoriented mitochondria, and an expanded intercellular space. The absorptive columnar cell illustrated many vacuoles inside with herniation of microvilli, and normal goblet cells were also seen. Many Paneth cells towards the lumen of intestinal gland contained secretory granules of different sizes and shapes. The histological architecture of the ileal mucosa in the QP plus Vit C group was found to be close to those of healthy controls. The outcomes of this study suggest that administering Vit C in rats treated with QPs protects them from ill dysfunction caused by QP.
Collapse
Affiliation(s)
- Mohamed Samir Ahmed Zaki
- Department of Anatomy, College of Medicine, King Khalid University, P.O. 62529, Abha, Saudi Arabia.
- College of Medicine, Zagazig University, Zagazig, Egypt.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Saudi Arabia
- Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Hussah I M AlGwaiz
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, 11474, Saudi Arabia
| | - Abulqasim M Sideeg
- Department of Anatomy, College of Medicine, King Khalid University, P.O. 62529, Abha, Saudi Arabia
| | - Mohamed Andarawi
- Department of Pathology, College of Medicine, King Khalid University, P.O. 62529, Abha, Saudi Arabia
| | - Refaat A Eid
- Department of Pathology, College of Medicine, King Khalid University, P.O. 62529, Abha, Saudi Arabia
| |
Collapse
|
19
|
Palanisamy BN, Sarkar S, Malovic E, Samidurai M, Charli A, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Environmental neurotoxic pesticide exposure induces gut inflammation and enteric neuronal degeneration by impairing enteric glial mitochondrial function in pesticide models of Parkinson's disease: Potential relevance to gut-brain axis inflammation in Parkinson's disease pathogenesis. Int J Biochem Cell Biol 2022; 147:106225. [PMID: 35550926 PMCID: PMC10411482 DOI: 10.1016/j.biocel.2022.106225] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022]
Abstract
Despite the growing recognition that gastrointestinal (GI) dysfunction is prevalent in Parkinson's disease (PD) and occurs as a major prodromal symptom of PD, its cellular and molecular mechanisms remain largely unknown. Among the various types of GI cells, enteric glial cells (EGCs), which resemble astrocytes in structure and function, play a critical role in the pathophysiology of many GI diseases including PD. Thus, we investigated how EGCs respond to the environmental pesticides rotenone (Rot) and tebufenpyrad (Tebu) in cell and animal models to better understand the mechanism underlying GI abnormalities. Both Rot and Tebu induce dopaminergic neuronal cell death through complex 1 inhibition of the mitochondrial respiratory chain. We report that exposing a rat enteric glial cell model (CRL-2690 cells) to these pesticides increased mitochondrial fission and reduced mitochondrial fusion by impairing MFN2 function. Furthermore, they also increased mitochondrial superoxide generation and impaired mitochondrial ATP levels and basal respiratory rate. Measurement of LC3, p62 and lysosomal assays revealed impaired autolysosomal function in ECGs during mitochondrial stress. Consistent with our recent findings that mitochondrial dysfunction augments inflammation in astrocytes and microglia, we found that neurotoxic pesticide exposure also enhanced the production of pro-inflammatory factors in EGCs in direct correlation with the loss in mitochondrial mass. Finally, we show that pesticide-induced mitochondrial defects functionally impaired smooth muscle velocity, acceleration, and total kinetic energy in a mixed primary culture of the enteric nervous system (ENS). Collectively, our studies demonstrate for the first time that exposure to environmental neurotoxic pesticides impairs mitochondrial bioenergetics and activates inflammatory pathways in EGCs, further augmenting mitochondrial dysfunction and pro-inflammatory events to induce gut dysfunction. Our findings have major implications in understanding the GI-related pathogenesis and progression of environmentally linked PD.
Collapse
Affiliation(s)
- Bharathi N Palanisamy
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Souvarish Sarkar
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; Brigham and Women's Hospital, Harvard Medical School, USA
| | - Emir Malovic
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Manikandan Samidurai
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Adhithiya Charli
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; Charles River Laboratories International, Inc., USA
| | - Gary Zenitsky
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; Center for Brain Sciences and Neurodegenerative Diseases, Department of Physiology and Pharmacology, University of Georgia, GA 30602, USA
| | - Huajun Jin
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; Center for Brain Sciences and Neurodegenerative Diseases, Department of Physiology and Pharmacology, University of Georgia, GA 30602, USA
| | - Vellareddy Anantharam
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; Center for Brain Sciences and Neurodegenerative Diseases, Department of Physiology and Pharmacology, University of Georgia, GA 30602, USA
| | - Arthi Kanthasamy
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; Center for Brain Sciences and Neurodegenerative Diseases, Department of Physiology and Pharmacology, University of Georgia, GA 30602, USA
| | - Anumantha G Kanthasamy
- Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA; Center for Brain Sciences and Neurodegenerative Diseases, Department of Physiology and Pharmacology, University of Georgia, GA 30602, USA.
| |
Collapse
|
20
|
Coppola L, Tait S, Fabbrizi E, Perugini M, La Rocca C. Comparison of the Toxicological Effects of Pesticides in Non-Tumorigenic MCF-12A and Tumorigenic MCF-7 Human Breast Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:4453. [PMID: 35457321 PMCID: PMC9030493 DOI: 10.3390/ijerph19084453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022]
Abstract
Humans are exposed to residues of organophosphate and neonicotinoid pesticides, commonly used in agriculture. Children are particularly vulnerable and, among possible adverse outcomes, the increased incidence of premature mammary gland development (thelarche) has raised concern. We evaluated the toxicological effects of chlorpyrifos (CPF), imidacloprid (IMI) and glyphosate (GLY) at exposure concentrations occurring in children on the tumorigenic MCF-7 and non-tumorigenic MCF-12A breast cell lines, as representative of the target organ model, assessing cytotoxicity, apoptosis, necrosis, intracellular reactive oxygen species (ROS) and ATP levels, 17β-estradiol secretion and gene expression of nuclear receptors involved in mammary gland development. The pesticides decreased cell vitality in MCF-7 and cell proliferation in MCF-12A cells. ATP levels were decreased in MCF-7 cells by pesticides and apoptosis was increased in MCF-12A cells only by GLY (2.3 nM). ROS production was decreased by pesticides in both cell lines, except IMI (1.6 nM) in MCF-7 cells. Endocrine disrupting activity was highlighted by induction of 17β-estradiol secretion and modulation of the gene expression of estrogen alpha and beta, progesterone, androgen, and aryl hydrocarbon receptors in both cell lines. The use of MCF-7 and MCF-12A cells highlighted dissimilar modes of action of each pesticide at low human relevant concentrations.
Collapse
Affiliation(s)
- Lucia Coppola
- Center for Gender-Specific Medicine, Italian National Institute of Health, 00161 Rome, Italy; (L.C.); (S.T.)
- Department of Physiology and Pharmacology V. Erspamer, Sapienza University of Rome, 00185 Rome, Italy
| | - Sabrina Tait
- Center for Gender-Specific Medicine, Italian National Institute of Health, 00161 Rome, Italy; (L.C.); (S.T.)
| | - Enrica Fabbrizi
- Pediatric Departmental Simple Operative Unit, Civitanova Marche Hospital, ASUR Marche Area Vasta n. 3, 62100 Macerata, Italy;
| | - Monia Perugini
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Cinzia La Rocca
- Center for Gender-Specific Medicine, Italian National Institute of Health, 00161 Rome, Italy; (L.C.); (S.T.)
| |
Collapse
|
21
|
Costas-Ferreira C, Faro LRF. Systematic Review of Calcium Channels and Intracellular Calcium Signaling: Relevance to Pesticide Neurotoxicity. Int J Mol Sci 2021; 22:13376. [PMID: 34948173 PMCID: PMC8704302 DOI: 10.3390/ijms222413376] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/25/2022] Open
Abstract
Pesticides of different chemical classes exert their toxic effects on the nervous system by acting on the different regulatory mechanisms of calcium (Ca2+) homeostasis. Pesticides have been shown to alter Ca2+ homeostasis, mainly by increasing its intracellular concentration above physiological levels. The pesticide-induced Ca2+ overload occurs through two main mechanisms: the entry of Ca2+ from the extracellular medium through the different types of Ca2+ channels present in the plasma membrane or its release into the cytoplasm from intracellular stocks, mainly from the endoplasmic reticulum. It has also been observed that intracellular increases in the Ca2+ concentrations are maintained over time, because pesticides inhibit the enzymes involved in reducing its levels. Thus, the alteration of Ca2+ levels can lead to the activation of various signaling pathways that generate oxidative stress, neuroinflammation and, finally, neuronal death. In this review, we also discuss some proposed strategies to counteract the detrimental effects of pesticides on Ca2+ homeostasis.
Collapse
Affiliation(s)
| | - Lilian R. F. Faro
- Departamento de Biología Funcional y Ciencias de la Salud, Facultad de Biología, Universidade de Vigo, Campus Universitario As Lagoas Marcosende, 36310 Vigo, Spain;
| |
Collapse
|
22
|
Andrew A, Zhou J, Gui J, Harrison A, Shi X, Li M, Guetti B, Nathan R, Tischbein M, Pioro EP, Stommel E, Bradley W. Pesticides applied to crops and amyotrophic lateral sclerosis risk in the U.S. Neurotoxicology 2021; 87:128-135. [PMID: 34562505 PMCID: PMC10756230 DOI: 10.1016/j.neuro.2021.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Environmental exposures are implicated in the etiology of amyotrophic lateral sclerosis (ALS). Application of insecticides, herbicides, and fungicides with neurotoxic properties to crops is permitted in the U.S., however reporting of the quantities is government mandated. OBJECTIVE To identify pesticides that may be associated with ALS etiology for future study. METHODS We geospatially estimated exposure to crop-applied pesticides as risk factors for ALS in a large de-identified medical claims database, the SYMPHONY Integrated Dataverse®. We extracted residence at diagnosis of ∼26,000 nationally distributed ALS patients, and matched non-ALS controls. We mapped county-level U.S. Geological Survey data on applications of 423 pesticides to estimate local residential exposure. We randomly broke the SYMPHONY dataset into two groups to form independent discovery and validation cohorts, then confirmed top hits using residential history information from a study of NH, VT, and OH. RESULTS Pesticides with the largest positive statistically significant associations in both the discovery and the validation studies and evidence of neurotoxicity in the literature were the herbicides 2,4-D (OR 1.25 95 % CI 1.17-1.34) and glyphosate (OR 1.29 95 %CI 1.19-1.39), and the insecticides carbaryl (OR 1.32 95 %CI 1.23-1.42) and chlorpyrifos (OR 1.25 95 %CI 1.17-1.33). SIGNIFICANCE Our geospatial analysis results support potential neurotoxic pesticide exposures as risk factors for sporadic ALS. Focused studies to assess these identified potential relationships are warranted.
Collapse
Affiliation(s)
- Angeline Andrew
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States.
| | - Jie Zhou
- Dartmouth College, Hanover, NH, United States
| | - Jiang Gui
- Dartmouth College, Hanover, NH, United States
| | | | - Xun Shi
- Dartmouth College, Hanover, NH, United States
| | - Meifang Li
- Dartmouth College, Hanover, NH, United States
| | - Bart Guetti
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | | | - Maeve Tischbein
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Erik P Pioro
- Center for ALS and Related Disorders, Cleveland Clinic, Cleveland, OH, United States
| | - Elijah Stommel
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Walter Bradley
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
23
|
Hirano T, Suzuki N, Ikenaka Y, Hoshi N, Tabuchi Y. Neurotoxicity of a pyrethroid pesticide deltamethrin is associated with the imbalance in proteolytic systems caused by mitophagy activation and proteasome inhibition. Toxicol Appl Pharmacol 2021; 430:115723. [PMID: 34520793 DOI: 10.1016/j.taap.2021.115723] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/23/2022]
Abstract
Pyrethroids are one of the most commonly used classes of synthetic pesticides in the world. Recent laboratory and epidemiological evidence suggested that pyrethroids have potential adverse effects in the mammalian brain; however, the underlying mechanisms of the neurotoxic effects of pyrethroids have not been fully elucidated. In the present study, we investigated the mechanisms of effects of a type II pyrethroid deltamethrin (DM) in a neuronal cell model focusing on the proteolytic function, including autophagy and the ubiquitin-proteasome system. We confirmed that a micromolar concentration of DM dose-dependently decreased the cell viability and induced apoptotic cell death. Our results showed that DM enhanced autophagy in association with an accumulation of autophagosomes and increase in the levels of autophagy markers LC3-II/LC3-I ratio and p62 which were much elevated in the presence of lysosomal inhibitors bafilomycin A1 and chloroquine. We also found that DM caused a dysfunction of mitochondria with a decrease of mitochondrial membrane potential and mitochondrial DNA copy number as well as colocalization with autophagosomes. Moreover, a decrease in the activities of three major proteasomal enzymes and an accumulation of ubiquitinated proteins were observed by the exposure to DM. Transcriptome analysis revealed that up-regulated genes supported the activation of autophagy with induction of cellular stress responses including oxidative stress and endoplasmic reticulum stress, while down-regulated genes related to the cell cycle and DNA replication. These findings provide novel insights into the neurotoxicity of DM which underlie the imbalance in proteolytic function caused by mitophagy activation and proteasome inhibition.
Collapse
Affiliation(s)
- Tetsushi Hirano
- Life Science Research Center, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan.
| | - Nihei Suzuki
- Life Science Research Center, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Yoshinori Ikenaka
- Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan; Water Research Group, Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa; One Health Research Center, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan; Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Nobuhiko Hoshi
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Kobe, Hyogo 657-8501, Japan
| | - Yoshiaki Tabuchi
- Life Science Research Center, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| |
Collapse
|
24
|
Oxidative Stress, Mitochondrial Dysfunction, and Neuroprotection of Polyphenols with Respect to Resveratrol in Parkinson's Disease. Biomedicines 2021; 9:biomedicines9080918. [PMID: 34440122 PMCID: PMC8389563 DOI: 10.3390/biomedicines9080918] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/24/2021] [Accepted: 07/25/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease and is characterized by dopaminergic neuronal loss. The exact pathogenesis of PD is complex and not yet completely understood, but research has established the critical role mitochondrial dysfunction plays in the development of PD. As the main producer of cytosolic reactive oxygen species (ROS), mitochondria are particularly susceptible to oxidative stress once an imbalance between ROS generation and the organelle’s antioxidative system occurs. An overabundance of ROS in the mitochondria can lead to mitochondrial dysfunction and further vicious cycles. Once enough damage accumulates, the cell may undergo mitochondria-dependent apoptosis or necrosis, resulting in the neuronal loss of PD. Polyphenols are a group of natural compounds that have been shown to offer protection against various diseases, including PD. Among these, the plant-derived polyphenol, resveratrol, exhibits neuroprotective effects through its antioxidative capabilities and provides mitochondria protection. Resveratrol also modulates crucial genes involved in antioxidative enzymes regulation, mitochondrial dynamics, and cellular survival. Additionally, resveratrol offers neuroprotective effects by upregulating mitophagy through multiple pathways, including SIRT-1 and AMPK/ERK pathways. This compound may provide potential neuroprotective effects, and more clinical research is needed to establish the efficacy of resveratrol in clinical settings.
Collapse
|
25
|
Lestón Pinilla L, Ugun-Klusek A, Rutella S, De Girolamo LA. Hypoxia Signaling in Parkinson's Disease: There Is Use in Asking "What HIF?". BIOLOGY 2021; 10:723. [PMID: 34439955 PMCID: PMC8389254 DOI: 10.3390/biology10080723] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022]
Abstract
Hypoxia is a condition characterized by insufficient tissue oxygenation, which results in impaired oxidative energy production. A reduction in cellular oxygen levels induces the stabilization of hypoxia inducible factor α (HIF-1α), master regulator of the molecular response to hypoxia, involved in maintaining cellular homeostasis and driving hypoxic adaptation through the control of gene expression. Due to its high energy requirement, the brain is particularly vulnerable to oxygen shortage. Thus, hypoxic injury can cause significant metabolic changes in neural cell populations, which are associated with neurodegeneration. Recent evidence suggests that regulating HIF-1α may ameliorate the cellular damage in neurodegenerative diseases. Indeed, the hypoxia/HIF-1α signaling pathway has been associated to several processes linked to Parkinson's disease (PD) including gene mutations, risk factors and molecular pathways such as mitochondrial dysfunction, oxidative stress and protein degradation impairment. This review will explore the impact of hypoxia and HIF-1α signaling on these specific molecular pathways that influence PD development and will evaluate different novel neuroprotective strategies involving HIF-1α stabilization.
Collapse
Affiliation(s)
- Laura Lestón Pinilla
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Aslihan Ugun-Klusek
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Luigi A. De Girolamo
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| |
Collapse
|
26
|
Kalyabina VP, Esimbekova EN, Kopylova KV, Kratasyuk VA. Pesticides: formulants, distribution pathways and effects on human health - a review. Toxicol Rep 2021; 8:1179-1192. [PMID: 34150527 PMCID: PMC8193068 DOI: 10.1016/j.toxrep.2021.06.004] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Pesticides are commonly used in agriculture to enhance crop production and control pests. Therefore, pesticide residues can persist in the environment and agricultural crops. Although modern formulations are relatively safe to non-target species, numerous theoretical and experimental data demonstrate that pesticide residues can produce long-term negative effects on the health of humans and animals and stability of ecosystems. Of particular interest are molecular mechanisms that mediate the start of a cascade of adverse effects. This is a review of the latest literature data on the effects and consequences of contamination of agricultural crops by pesticide residues. In addition, we address the issue of implicit risks associated with pesticide formulations. The effects of pesticides are considered in the context of the Adverse Outcome Pathway concept.
Collapse
Affiliation(s)
- Valeriya P. Kalyabina
- Siberian Federal University, 79 Svobodny Prospect, Krasnoyarsk, 660041, Russia
- Institute of Biophysics SB RAS, 50/50 Akademgorodok, Krasnoyarsk, 660036, Russia
| | - Elena N. Esimbekova
- Siberian Federal University, 79 Svobodny Prospect, Krasnoyarsk, 660041, Russia
- Institute of Biophysics SB RAS, 50/50 Akademgorodok, Krasnoyarsk, 660036, Russia
| | - Kseniya V. Kopylova
- Siberian Federal University, 79 Svobodny Prospect, Krasnoyarsk, 660041, Russia
| | - Valentina A. Kratasyuk
- Siberian Federal University, 79 Svobodny Prospect, Krasnoyarsk, 660041, Russia
- Institute of Biophysics SB RAS, 50/50 Akademgorodok, Krasnoyarsk, 660036, Russia
| |
Collapse
|
27
|
Wu L, Zhou M, Liu C, Chen X, Chen Y. Double-enzymes-mediated Fe 2+/Fe 3+ conversion as magnetic relaxation switch for pesticide residues sensing. JOURNAL OF HAZARDOUS MATERIALS 2021; 403:123619. [PMID: 32827859 DOI: 10.1016/j.jhazmat.2020.123619] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 05/25/2023]
Abstract
It is a great challenge to develop a newly rapid and accurate detection method for pesticide residues. In this work, based on acetylcholinesterase (AChE) and choline oxidase (CHO), a double-enzymes-mediated Fe2+/Fe3+ conversion as magnetic relaxation switch was explored for the measurement of acetamiprid residue. In the double-enzymes reactions, acetylcholine chloride (ACh) can be catalyzed to produce choline by AChE, which is successively hydrolyzed to betaine and hydrogen peroxide (H2O2) by CHO. According to the enzyme inhibition principle, AChE activity will be inactivated in the presence of acetamiprid, thus leading to the less production of H2O2. Wherein, Fe2+, ACh, AChE and CHO were optimized as the reaction substrates. In the reaction system, acetamiprid can be reflected by the transverse relaxation time (T2) that related with H2O2 mediated Fe2+ variations, which was further developed as an enzyme cascade amplification method. The detection linear range is 0.01∼1000 μg mL-1 (R2 = 0.99), and the limit of detection (LOD) is 2.66 ng mL-1 (S/N = 3, n = 3), behaving a 335-fold improvement in LOD than that of traditional enzyme inhibition method (0.89 μg mL-1). This method can realize "one-step mixing" detection of acetamiprid, which makes it a promising analytical tool for monitoring pesticide residue in complicated samples.
Collapse
Affiliation(s)
- Long Wu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, PR China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering and Food, Hubei University of Technology, Wuhan, Hubei, 430068, PR China
| | - Min Zhou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, PR China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering and Food, Hubei University of Technology, Wuhan, Hubei, 430068, PR China
| | - Chen Liu
- Leibniz Institute of Photonic Technology, Jena-Member of the research alliance Leibniz Health Technologies, Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Albert-Einstein-Street 9, 07745, Jena, Germany; Leibniz Institute of Photonic Technology Jena - Member of the research alliance, Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745, Jena, Germany
| | - Xiaoqiang Chen
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering and Food, Hubei University of Technology, Wuhan, Hubei, 430068, PR China.
| | - Yiping Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, PR China.
| |
Collapse
|
28
|
Devi S, Kim JJ, Singh AP, Kumar S, Dubey AK, Singh SK, Singh RS, Kumar V. Proteotoxicity: A Fatal Consequence of Environmental Pollutants-Induced Impairments in Protein Clearance Machinery. J Pers Med 2021; 11:69. [PMID: 33503824 PMCID: PMC7912547 DOI: 10.3390/jpm11020069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/08/2023] Open
Abstract
A tightly regulated protein quality control (PQC) system maintains a healthy balance between correctly folded and misfolded protein species. This PQC system work with the help of a complex network comprised of molecular chaperones and proteostasis. Any intruder, especially environmental pollutants, disrupt the PQC network and lead to PQCs disruption, thus generating damaged and infectious protein. These misfolded/unfolded proteins are linked to several diseases such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and cataracts. Numerous studies on proteins misfolding and disruption of PQCs by environmental pollutants highlight the necessity of detailed knowledge. This review represents the PQCs network and environmental pollutants' impact on the PQC network, especially through the protein clearance system.
Collapse
Affiliation(s)
- Shweta Devi
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, India;
| | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea;
| | - Anand Prakash Singh
- Division of Cardiovascular Disease, The University of Alabama at Birmingham (UAB), 1720 2nd Ave South, Birmingham, AL 35294-1913, USA;
| | - Surendra Kumar
- Cytogenetics Lab, Department of Anatomy, All India Institute of Medical Sciences, New Delhi 110029, India;
| | | | | | - Ravi Shankar Singh
- Department of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Room 4D40, Health Sciences Building, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea;
| |
Collapse
|
29
|
Afewerky HK, Ayodeji AE, Tiamiyu BB, Orege JI, Okeke ES, Oyejobi AO, Bate PNN, Adeyemi SB. Critical review of the Withania somnifera (L.) Dunal: ethnobotany, pharmacological efficacy, and commercialization significance in Africa. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2021; 45:176. [PMID: 34697529 PMCID: PMC8529567 DOI: 10.1186/s42269-021-00635-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/08/2021] [Indexed: 05/10/2023]
Abstract
BACKGROUND Withania somnifera (L.) Dunal (W. somnifera) is a herb commonly known by its English name as Winter Cherry. Africa is indigenous to many medicinal plants and natural products. However, there is inadequate documentation of medicinal plants, including W. somnifera, in Africa. There is, therefore, a need for a comprehensive compilation of research outcomes of this reviewed plant as used in traditional medicine in different regions of Africa. METHODOLOGY Scientific articles and publications were scooped and sourced from high-impact factor journals and filtered with relevant keywords on W. somnifera. Scientific databases, including GBIF, PubMed, NCBI, Google Scholar, Research Gate, Science Direct, SciFinder, and Web of Science, were accessed to identify the most influential articles and recent breakthroughs published on the contexts of ethnography, ethnomedicinal uses, phytochemistry, pharmacology, and commercialization of W. somnifera. RESULTS This critical review covers the W. somnifera ethnography, phytochemistry, and ethnomedicinal usage to demonstrate the use of the plant in Africa and elsewhere to prevent or alleviate several pathophysiological conditions, including cardiovascular, neurodegenerative, reproductive impotence, as well as other chronic diseases. CONCLUSION W. somnifera is reportedly safe for administration in ethnomedicine as several research outcomes confirmed its safety status. The significance of commercializing this plant in Africa for drug development is herein thoroughly covered to provide the much-needed highlights towards its cultivations economic benefit to Africa.
Collapse
Affiliation(s)
- Henok Kessete Afewerky
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- School of Allied Health Professions, Asmara College of Health Sciences, 00291 Asmara, Eritrea
- Organization of African Academic Doctors, Nairobi, 00100 Kenya
| | - Ayeni Emmanuel Ayodeji
- Organization of African Academic Doctors, Nairobi, 00100 Kenya
- Department of Pharmacognosy and Drug Development, Ahmadu Bello University Zaria, PMB 1044, Kaduna, 800211 Nigeria
| | - Bashir Bolaji Tiamiyu
- Organization of African Academic Doctors, Nairobi, 00100 Kenya
- Department of Plant Biology, Faculty of Life Sciences, University of Ilorin, Ilorin, 240001 Nigeria
| | - Joshua Iseoluwa Orege
- Organization of African Academic Doctors, Nairobi, 00100 Kenya
- Department of Industrial Chemistry, Ekiti State University, PMB 5363, Ado-Ekiti, 362001 Nigeria
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023 China
| | - Emmanuel Sunday Okeke
- Organization of African Academic Doctors, Nairobi, 00100 Kenya
- Department of Biochemistry, FBS and Natural Science Unit, SGS, University of Nigeria, Nsukka, 410001 Nigeria
- School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013 China
| | - Aanuoluwapo Opeyemi Oyejobi
- Organization of African Academic Doctors, Nairobi, 00100 Kenya
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Petuel Ndip Ndip Bate
- Organization of African Academic Doctors, Nairobi, 00100 Kenya
- Guangzhou Institute of Biomedicine and Health, Guangzhou, 510530 China
| | - Sherif Babatunde Adeyemi
- Organization of African Academic Doctors, Nairobi, 00100 Kenya
- Department of Plant Biology, Faculty of Life Sciences, University of Ilorin, Ilorin, 240001 Nigeria
- C.G. Bhakta Institute of Biotechnology, Uka Tarsadia University, Bardoli-Mahuva Road, Bardoli, Surat, Gujarat 394350 India
| |
Collapse
|
30
|
Moyano P, Sanjuna J, Garcia JM, Garcia J, Frejo MT, Naval MV, Del Pino J. Paraquat Treatment Compromises the Clearance of β-Amyloid and Tau Proteins and Induces Primary Hippocampal Neuronal Cell Death through HSP70, P20S, and TFEB Disruption. Chem Res Toxicol 2020; 34:1240-1244. [PMID: 33156613 DOI: 10.1021/acs.chemrestox.0c00370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The herbicide paraquat (PQ) induces hippocampal neuronal cell loss and cognitive dysfunction after one and repeated treatment. All the mechanisms involved in these effects are not well understood. Single and repeated PQ treatment increased Aβ and tau protein levels, through HSP70 and TFEB downregulation and proteasome 20S inhibition, producing cell death in primary hippocampal neurons associated with cognitive decline. Our results reveal the mechanisms through which PQ could induce the accumulation of abnormal proteins and neurodegeneration that could originate the cognitive decline produced by it and could help managing its degenerative effects.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Javier Sanjuna
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - José Manuel Garcia
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jimena Garcia
- Department of Pharmacology, Health Sciences School, Alfonso X University, 28691 Madrid, Spain
| | - María Teresa Frejo
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - María Victoria Naval
- Department of Pharmacology, Pharmacognosy and Botany, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
31
|
Fernandes LC, Santos AG, Sampaio TB, Sborgi S, Prediger R, Ferro MM, Franco G, Lipinski L, Miyoshi E. Exposure to paraquat associated with periodontal disease causes motor damage and neurochemical changes in rats. Hum Exp Toxicol 2020; 40:81-89. [PMID: 32748713 DOI: 10.1177/0960327120938851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exposure to paraquat is possibly involved with the development of several conditions, including neurodegenerative diseases, such as Parkinson's disease (PD). This condition is mainly characterized by the loss of dopaminergic neurons in the nigrostriatal pathway and the development of classical motor symptoms. Etiology includes exposure to environmental factors, such as the paraquat exposure, and inflammatory diseases may exacerbate paraquat neurotoxicity. The aim of the study was to investigate whether the exposure to paraquat associated with the presence of periodontal disease is able to induce motor and biochemical changes in rats similar to that observed in PD. Adult male Wistar rats were sent to ligature. After 48 h, they were sent to daily treatment paraquat (1 mg/kg/day; 2 mL/kg; intragastric) or vehicle for 4 weeks. Twenty-four hours after the last administration, the open field test was performed. The rats were euthanized and the left hemimandibles and striatum were dissected for the analysis of dopaminergic and inflammatory markers. Only the combination of periodontal disease model plus paraquat exposure induced motor impairments. Remarkably, the paraquat exposure increased the ligature-induced alveolar bone loss in hemimandibles. Moreover, only the combination of periodontal disease and paraquat exposure induced the loss of dopaminergic neurons and astrocyte activation in the striatum.
Collapse
Affiliation(s)
- L C Fernandes
- 549253Department of Pharmaceutical Sciences, State University of Ponta Grossa (UEPG), Ponta Grossa, PR, Brazil
| | - A G Santos
- 549253Department of Pharmaceutical Sciences, State University of Ponta Grossa (UEPG), Ponta Grossa, PR, Brazil
| | - T B Sampaio
- Department of Pharmacology, 28117Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Sms Sborgi
- 549253Department of Pharmaceutical Sciences, State University of Ponta Grossa (UEPG), Ponta Grossa, PR, Brazil
| | - Rds Prediger
- Department of Pharmacology, 28117Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - M M Ferro
- Department of Biology, 67883State University of Ponta Grossa (UEPG), Ponta Grossa, PR, Brazil
| | - Gcn Franco
- Department of Odontology, 67883State University of Ponta Grossa (UEPG), Ponta Grossa, PR, Brazil
| | - L Lipinski
- Department of Medicine, 67883State University of Ponta Grossa (UEPG), Ponta Grossa, PR, Brazil
| | - E Miyoshi
- 549253Department of Pharmaceutical Sciences, State University of Ponta Grossa (UEPG), Ponta Grossa, PR, Brazil
| |
Collapse
|
32
|
He H, Liang L, Tang T, Luo J, Wang Y, Cui H. Progressive brain changes in Parkinson’s disease: A meta-analysis of structural magnetic resonance imaging studies. Brain Res 2020; 1740:146847. [DOI: 10.1016/j.brainres.2020.146847] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022]
|
33
|
Hallinger DR, Lindsay HB, Friedman KP, Suarez DA, Simmons SO. Respirometric Screening and Characterization of Mitochondrial Toxicants Within the ToxCast Phase I and II Chemical Libraries. Toxicol Sci 2020; 176:175-192. [PMID: 32374859 PMCID: PMC10626520 DOI: 10.1093/toxsci/kfaa059] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Mitochondrial toxicity drives several adverse health outcomes. Current high-throughput screening assays for chemically induced mitochondrial toxicity typically measure changes to mitochondrial structure and may not detect known mitochondrial toxicants. We adapted a respirometric screening assay (RSA) measuring mitochondrial function to screen ToxCast chemicals in HepG2 cells using a tiered testing strategy. Of 1042 chemicals initially screened at a singlemaximal concentration, 243 actives were identified and rescreened at 7 concentrations. Concentration-response data for 3 respiration phases confirmed activity and indicated a mechanism for 193 mitochondrial toxicants: 149 electron transport chain inhibitors (ETCi), 15 uncouplers and 29 adenosine triphosphate synthase inhibitors. Subsequently, an electron flow assay was used to identify the target complex for 84 of the 149 ETCi. Sixty reference chemicals were used to compare the RSA to existing ToxCast and Tox21 mitochondrial toxicity assays. The RSA was most predictive (accuracy = 90%) of mitochondrial toxicity. The Tox21 mitochondrial membrane potential assay was also highly predictive (accuracy = 87%) of bioactivity but underestimated the potency of well-known ETCi and provided no mechanistic information. The tiered RSA approach accurately identifies and characterizes mitochondrial toxicants acting through diverse mechanisms and at a throughput sufficient to screen large chemical inventories. The electron flow assay provides additional confirmation and detailed mechanistic understanding for ETCi, the most common type of mitochondrial toxicants among ToxCast chemicals. The mitochondrial toxicity screening approach described herein may inform hazard assessment and the in vitro bioactive concentrations used to derive relevant doses for screening level chemical assessment using new approach methodologies.
Collapse
Affiliation(s)
| | | | | | - Danielle A. Suarez
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | | |
Collapse
|
34
|
Buratta S, Chiaradia E, Tognoloni A, Gambelunghe A, Meschini C, Palmieri L, Muzi G, Urbanelli L, Emiliani C, Tancini B. Effect of Curcumin on Protein Damage Induced by Rotenone in Dopaminergic PC12 Cells. Int J Mol Sci 2020; 21:E2761. [PMID: 32316110 PMCID: PMC7215629 DOI: 10.3390/ijms21082761] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress is considered to be a key factor of the pathogenesis of Parkinson's disease, a multifactorial neurodegenerative disorder characterized by reduced dopaminergic neurons in the substantia nigra pars compacta and accumulated protein aggregates. Rotenone is a worldwide-used pesticide that induces the most common features of Parkinson's by direct inhibition of the mitochondrial complex I. Rotenone-induced Parkinson's models, as well as brain tissues from Parkinson's patients, are characterized by the presence of both lipid peroxidation and protein oxidation markers resulting from the increased level of free radical species. Oxidation introduces several modifications in protein structure, including carbonylation and nitrotyrosine formation, which severely compromise cell function. Due to the link existing between oxidative stress and Parkinson's disease, antioxidant molecules could represent possible therapeutic tools for this disease. In this study, we evaluated the effect of curcumin, a natural compound known for its antioxidant properties, in dopaminergic PC12 cells treated with rotenone, a cell model of Parkinsonism. Our results demonstrate that the treatment of PC12 cells with rotenone causes severe protein damage, with formation of both carbonylated and nitrotyrosine-derived proteins, whereas curcumin (10 µM) co-exposure exerts protective effects by reducing the levels of oxidized proteins. Curcumin also promotes proteasome activation, abolishing the inhibitory effect exerted by rotenone on this degradative system.
Collapse
Affiliation(s)
- Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (S.B.); (C.M.); (L.P.); (L.U.); (C.E.)
| | - Elisabetta Chiaradia
- Department of Veterinary Medicine, University of Perugia, 06126 Perugia, Italy; (E.C.); (A.T.)
| | - Alessia Tognoloni
- Department of Veterinary Medicine, University of Perugia, 06126 Perugia, Italy; (E.C.); (A.T.)
| | - Angela Gambelunghe
- Department of Medicine, University of Perugia, 06132 Perugia, Italy; (A.G.); (G.M.)
| | - Consuelo Meschini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (S.B.); (C.M.); (L.P.); (L.U.); (C.E.)
| | - Luigi Palmieri
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (S.B.); (C.M.); (L.P.); (L.U.); (C.E.)
| | - Giacomo Muzi
- Department of Medicine, University of Perugia, 06132 Perugia, Italy; (A.G.); (G.M.)
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (S.B.); (C.M.); (L.P.); (L.U.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (S.B.); (C.M.); (L.P.); (L.U.); (C.E.)
| | - Brunella Tancini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (S.B.); (C.M.); (L.P.); (L.U.); (C.E.)
| |
Collapse
|
35
|
Seydi E, Mehrpouya L, Sadeghi H, Rahimi S, Pourahmad J. Toxicity of fipronil on rat heart mitochondria. TOXIN REV 2019. [DOI: 10.1080/15569543.2019.1700382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Enayatollah Seydi
- Department of Occupational Health and Safety Engineering, School of Health, Alborz University of Medical Sciences, Karaj, Iran
- Research Center for Health, Safety and Environment, Alborz University of Medical Sciences, Karaj, Iran
| | - Leila Mehrpouya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadiseh Sadeghi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Rahimi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Lee S, Lee H, Kim KT. Optimization of experimental conditions and measurement of oxygen consumption rate (OCR) in zebrafish embryos exposed to organophosphate flame retardants (OPFRs). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 182:109377. [PMID: 31254858 DOI: 10.1016/j.ecoenv.2019.109377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 05/18/2023]
Abstract
The measurement of oxygen consumption rate (OCR) provides a comprehensive understanding of mitochondrial metabolism. However, no study has been conducted to investigate the mitochondrial dysfunction caused by organophosphate flame retardants (OPFRs). The objectives of this study were to optimize the experimental conditions to measure OCR in zebrafish embryos using the Seahorse XFe 24 Extracellular Flux Analyzer, and to investigate the changes of OCR in zebrafish embryos exposed to OPFRs. We first optimized the experimental conditions such as the number of embryos, concentrations of inhibitors, and time points. We determined the factors, i.e., three embryos, 12.5 μM of oligomycin, 8 μM of carbonyl cyanaide 4-(trifluoromethoxy) phenylhydrazone (FCCP), and 24 hpf (hours post-fertilization) time point, for obtaining the typical pattern of OCR in dechorinated zebrafish embryos. After confirming the determinants upon exposure of triclosan, the inhibition of OCR was measured in zebrafish embryos exposed to two major OPFRs, triphenyl phosphate (TPHP) and tris (1,3-dichloro-2-propyl) phosphate (TDCIPP). We found that significant inhibition of OCR was observed in basal respiration for TPHP, and in basal and maximal respiration for TDCIPP exposure, respectively. We suggest the optimum conditions of the Seahorse XFe 24 analyzer to better evaluate OCR in zebrafish embryos, and demonstrate the potential of TPHP and TDCIPP to cause the disruption of energy metabolism associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Sunjin Lee
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
| | - Hyojin Lee
- Department of Environmental Energy Engineering, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
| | - Ki-Tae Kim
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea; Department of Environmental Energy Engineering, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea.
| |
Collapse
|
37
|
Moyano P, Garcia JM, Frejo MT, Lobo M, Garcia J, Del Pino J. Proteasome 20S and Rab5 Alteration after 24 h and 14 Days Chlorpyrifos Exposure Lead to β-Amyloid and Tau Protein Level Increases and SN56 Neuronal Cell Death. Chem Res Toxicol 2019; 32:1920-1924. [PMID: 31580065 DOI: 10.1021/acs.chemrestox.9b00216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The biocide chlorpyrifos (CPF) was shown to produce cognition impairment following single and long-term exposure. The complete mechanisms that lead to the CPF induced cognitive disorders remain to be discovered. Aβ and tau proteins production was induced in basal forebrain SN56 cholinergic cells, by CPF, through proteasome 20S inhibition and Rab5 overexpression, leading to cell death both after acute and repeated administration, which was related with cognitive disorders induction. The results obtained in our study procure novel information related to the mechanisms involved in CPF neurodegeneration, which could be responsible for cognitive dysfunction and may lead to a promising alternative treatment of these effects.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School , Complutense University of Madrid , 28040 Madrid , Spain
| | - José Manuel Garcia
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School , Complutense University of Madrid , 28040 Madrid , Spain
| | - María Teresa Frejo
- Department of Pharmacology and Toxicology, Veterinary School , Complutense University of Madrid , 28040 Madrid , Spain
| | - Margarita Lobo
- Department of Pharmacology and Toxicology, Veterinary School , Complutense University of Madrid , 28040 Madrid , Spain
| | - Jimena Garcia
- Department of Pharmacology, Health Sciences School , Alfonso X University , 28691 Madrid , Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School , Complutense University of Madrid , 28040 Madrid , Spain
| |
Collapse
|
38
|
Zhou PK, Huang RX. Targeting of the respiratory chain by toxicants: beyond the toxicities to mitochondrial morphology. Toxicol Res (Camb) 2018; 7:1008-1011. [PMID: 30542598 PMCID: PMC6249626 DOI: 10.1039/c8tx00207j] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022] Open
Abstract
The mitochondrion is an important subcellular target of environmental toxicants. With environmental stress, a series of toxic effects on mitochondria are induced, which originate from the dynamic changes of mitochondrial fusion and fission, structure/membrane damage, and respiratory chain dysfunction. The toxic effects of various toxicants on mitochondrial morphology and intact membranes, and their determination of cell fate, have already been broadly studied and reported on. However, their effects on the integrity and function of the mitochondrial respiratory chain (RC) remain incompletely understood. Recently, Fan et al. and Yu et al. approached this topic by closely examining the mitochondrial toxicities, including the effect on the respiratory chain, induced by organic arsenical chemical 2-methoxy-4-(((4-(oxoarsanyl)phenyl)imino)methyl)phenol and thiourea gold(i) complexes (AuTuCl). Obviously, toxicant-induced dysfunction of the respiratory chain can hinder ATP production, and may elevate ROS generation. The increased ROS can further damage mtDNA, and consequently leads to inactivation of some RC protein-encoding mtDNA, generating a vicious circle of amplifying mitochondrial damage. We hope that these studies focused on RC structure and activity will broaden our view of mitochondrial toxicology and draw forth more profound mechanistic studies on the respiratory chain toxicity of environmental toxicants and their application in risk assessment.
Collapse
Affiliation(s)
- P K Zhou
- Department of Radiation Biology , Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , AMMS , Beijing 100850 , P. R. China .
| | - R X Huang
- Department of Occupational and Environmental Health , Central south University , Changsha , 410078 , China .
| |
Collapse
|