1
|
Tizabi Y, Bennani S, El Kouhen N, Getachew B, Aschner M. Heavy Metal Interactions with Neuroglia and Gut Microbiota: Implications for Huntington's Disease. Cells 2024; 13:1144. [PMID: 38994995 PMCID: PMC11240758 DOI: 10.3390/cells13131144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/01/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
Huntington's disease (HD) is a rare but progressive and devastating neurodegenerative disease characterized by involuntary movements, cognitive decline, executive dysfunction, and neuropsychiatric conditions such as anxiety and depression. It follows an autosomal dominant inheritance pattern. Thus, a child who has a parent with the mutated huntingtin (mHTT) gene has a 50% chance of developing the disease. Since the HTT protein is involved in many critical cellular processes, including neurogenesis, brain development, energy metabolism, transcriptional regulation, synaptic activity, vesicle trafficking, cell signaling, and autophagy, its aberrant aggregates lead to the disruption of numerous cellular pathways and neurodegeneration. Essential heavy metals are vital at low concentrations; however, at higher concentrations, they can exacerbate HD by disrupting glial-neuronal communication and/or causing dysbiosis (disturbance in the gut microbiota, GM), both of which can lead to neuroinflammation and further neurodegeneration. Here, we discuss in detail the interactions of iron, manganese, and copper with glial-neuron communication and GM and indicate how this knowledge may pave the way for the development of a new generation of disease-modifying therapies in HD.
Collapse
Affiliation(s)
- Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Samia Bennani
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20670, Morocco
| | - Nacer El Kouhen
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20670, Morocco
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
2
|
Pang Y, Li M, Li F, Lei J, Zhang T. Preliminary study on the E-liquid and aerosol on the neurobehavior of C. elegans. ENVIRONMENT INTERNATIONAL 2023; 179:108180. [PMID: 37690220 DOI: 10.1016/j.envint.2023.108180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/27/2023] [Accepted: 08/31/2023] [Indexed: 09/12/2023]
Abstract
E-cigarettes, also known as electronic nicotine delivery systems (ENDS), are mainly used among adolescents and young adults. Similar to traditional cigarettes, different concentrations of nicotine are also added to E-cigarette's liquid (E-liquid), but due to the supplementation of chemicals such as propylene glycol (PG), vegetable glycerin (VG) and flavors, it is difficult to determine the risk after using E-cigarettes. And given to the specificity of the aerosol particle composition and atomization process of E-cigarettes, it is necessary to assess the neurotoxic effects of long-term E-cigarettes use. In this study, two commercial nicotine-containing (5%) and nicotine-free E-liquids were diluted to investigate the neurobehavioral changes and addictive tendencies of developing C. elegans after sub-chronic exposure to E-liquid. The results showed that sub-chronic exposure of E-liquid could lead to impaired growth and development of nematodes, abnormal general neuromotor behavior and advanced learning and memory behavior, and nicotine-containing E-liquid could also lead to increased addiction tendency of nematodes. Although the damage effect of nicotine free E-liquid is smaller than that of the nicotine-containing group, its toxic effect cannot be ignored. Further analysis of the neurotoxicity mechanism found that redox imbalance-mediated mitochondrial stress and aging may be important causes of E-liquid-induced biological damage. The biosafety of e-cigarette aerosols was also included in the assessment. The study found that the heated atomization process did not alter the E-liquid components, and E-cigarette aerosols still have the effect of interfering with the growth and development of nematodes and neurobehavior, and its addictive nature is also of concern. This study can provide new ideas for future studies on the neurotoxic effects and safety assessment of the E-cigarettes, and provide theoretical reference for the study on the injury mechanism of E-cigarettes.
Collapse
Affiliation(s)
- Yanting Pang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Menghan Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Fuxian Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jialin Lei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
3
|
Ferlazzo GM, Gambetta AM, Amato S, Cannizzaro N, Angiolillo S, Arboit M, Diamante L, Carbognin E, Romani P, La Torre F, Galimberti E, Pflug F, Luoni M, Giannelli S, Pepe G, Capocci L, Di Pardo A, Vanzani P, Zennaro L, Broccoli V, Leeb M, Moro E, Maglione V, Martello G. Genome-wide screening in pluripotent cells identifies Mtf1 as a suppressor of mutant huntingtin toxicity. Nat Commun 2023; 14:3962. [PMID: 37407555 DOI: 10.1038/s41467-023-39552-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/19/2023] [Indexed: 07/07/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by CAG-repeat expansions in the huntingtin (HTT) gene. The resulting mutant HTT (mHTT) protein induces toxicity and cell death via multiple mechanisms and no effective therapy is available. Here, we employ a genome-wide screening in pluripotent mouse embryonic stem cells (ESCs) to identify suppressors of mHTT toxicity. Among the identified suppressors, linked to HD-associated processes, we focus on Metal response element binding transcription factor 1 (Mtf1). Forced expression of Mtf1 counteracts cell death and oxidative stress caused by mHTT in mouse ESCs and in human neuronal precursor cells. In zebrafish, Mtf1 reduces malformations and apoptosis induced by mHTT. In R6/2 mice, Mtf1 ablates motor defects and reduces mHTT aggregates and oxidative stress. Our screening strategy enables a quick in vitro identification of promising suppressor genes and their validation in vivo, and it can be applied to other monogenic diseases.
Collapse
Affiliation(s)
- Giorgia Maria Ferlazzo
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
- Aptuit (Verona) S.r.l., an Evotec Company, Campus Levi-Montalcini, 37135, Verona, Italy
| | - Anna Maria Gambetta
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Sonia Amato
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
- Department of Neuroscience, University of Padova, Via Belzoni, 160, 35131, Padua, Italy
| | - Noemi Cannizzaro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Silvia Angiolillo
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Mattia Arboit
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Linda Diamante
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Elena Carbognin
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Patrizia Romani
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Federico La Torre
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Elena Galimberti
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Florian Pflug
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Mirko Luoni
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Serena Giannelli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
| | | | | | | | - Paola Vanzani
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Lucio Zennaro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Vania Broccoli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
- CNR Institute of Neuroscience, 20854, Vedrano al Lambro, Italy
| | - Martin Leeb
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Enrico Moro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | | | - Graziano Martello
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy.
| |
Collapse
|
4
|
Kamitsuka PJ, Ghanem MM, Ziar R, McDonald SE, Thomas MG, Kwakye GF. Defective Mitochondrial Dynamics and Protein Degradation Pathways Underlie Cadmium-Induced Neurotoxicity and Cell Death in Huntington's Disease Striatal Cells. Int J Mol Sci 2023; 24:ijms24087178. [PMID: 37108341 PMCID: PMC10139096 DOI: 10.3390/ijms24087178] [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: 03/18/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Exposure to heavy metals, including cadmium (Cd), can induce neurotoxicity and cell death. Cd is abundant in the environment and accumulates in the striatum, the primary brain region selectively affected by Huntington's disease (HD). We have previously reported that mutant huntingtin protein (mHTT) combined with chronic Cd exposure induces oxidative stress and promotes metal dyshomeostasis, resulting in cell death in a striatal cell model of HD. To understand the effect of acute Cd exposure on mitochondrial health and protein degradation pathways, we hypothesized that expression of mHTT coupled with acute Cd exposure would cooperatively alter mitochondrial bioenergetics and protein degradation mechanisms in striatal STHdh cells to reveal novel pathways that augment Cd cytotoxicity and HD pathogenicity. We report that mHTT cells are significantly more susceptible to acute Cd-induced cell death as early as 6 h after 40 µM CdCl2 exposure compared with wild-type (WT). Confocal microscopy, biochemical assays, and immunoblotting analysis revealed that mHTT and acute Cd exposure synergistically impair mitochondrial bioenergetics by reducing mitochondrial potential and cellular ATP levels and down-regulating the essential pro-fusion proteins MFN1 and MFN2. These pathogenic effects triggered cell death. Furthermore, Cd exposure increases the expression of autophagic markers, such as p62, LC3, and ATG5, and reduces the activity of the ubiquitin-proteasome system to promote neurodegeneration in HD striatal cells. Overall, these results reveal a novel mechanism to further establish Cd as a pathogenic neuromodulator in striatal HD cells via Cd-triggered neurotoxicity and cell death mediated by an impairment in mitochondrial bioenergetics and autophagy with subsequent alteration in protein degradation pathways.
Collapse
Affiliation(s)
- Paul J Kamitsuka
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| | - Marwan M Ghanem
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| | - Rania Ziar
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| | - Sarah E McDonald
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| | - Morgan G Thomas
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| | - Gunnar F Kwakye
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| |
Collapse
|
5
|
Liu C, Zhang R, Yang L, Ji T, Zhu C, Liu B, Zhang H, Xu C, Zhang N, Huang S, Chen L. Neuroprotection of resveratrol against cadmium-poisoning acts through dual inhibition of mTORC1/2 signaling. Neuropharmacology 2022; 219:109236. [PMID: 36049535 PMCID: PMC9524506 DOI: 10.1016/j.neuropharm.2022.109236] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/07/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022]
Abstract
Resveratrol is a natural polyphenol with neuroprotective function. The underlying mechanism is not well understood. Our previous studies have identified that resveratrol antagonizes cadmium (Cd) neurotoxicity via targeting PP2A/PP5-mediated Erk1/2 and JNK pathways. Here we show that resveratrol protected against Cd-poisoning also by blocking Cd-induced activation of mTORC1 and mTORC2 pathways in PC12 cells and murine primary neurons. Co-treatment with inhibitors of mTORC1 (rapamycin), mTORC1/2 (PP242), Erk1/2 (U0126) and/or JNK (SP600125), knockdown of mTOR, or disruption of mTORC1 and/or mTORC2 by silencing raptor, rictor or raptor/rictor, respectively, markedly potentiated the inhibitory effects of resveratrol on Cd-induced phosphorylation of S6K1/4E-BP1 (mTORC1 substrates), Akt (mTORC2 substrate), Erk1/2 and/or JNK/c-Jun, cleavage of caspase-3 and cell death in PC12 cells and/or primary neurons. Knockdown of S6K1 or 4E-BP1, or ectopic expression of constitutively hypophosphorylated 4E-BP1 (4E-BP1-5A) reinforced the resveratrol's inhibition on Cd-evoked cell death, whereas ectopic expression of constitutively active S6K1 or knockdown of 4E-BP1 attenuated the resveratrol's inhibition on Cd-induced cell death. Co-treatment with Akt inhibitor or overexpression of dominant negative Akt (dn-Akt) strengthened the resveratrol's suppression on Cd-induced ROS, Erk1/2 activation and apoptosis, whereas overexpression of constitutively active Akt (myr-Akt) conferred high resistance to the resveratrol's inhibitory effects in the neuronal cells. Taken together, the results indicate that resveratrol attenuates Cd-induced neuronal apoptosis partly through inhibition of mTORC1/2 pathways. Our studies highlight that resveratrol can be exploited for the prevention of Cd toxicity related to neurodegenerative diseases.
Collapse
Affiliation(s)
- Chunxiao Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China; Department of Medical Technology, Suzhou Vocational Health College, Suzhou, 215009, PR China
| | - Ruijie Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China; College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Liu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China
| | - Tong Ji
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China
| | - Cuilan Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China
| | - Beibei Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China
| | - Hai Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China
| | - Chong Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China
| | - Nana Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Shreveport, LA, 71130-3932, USA; Department of Hematology and Oncology, Shreveport, LA, 71130-3932, USA; Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, PR China.
| |
Collapse
|
6
|
Qin M, Xie Z, Cao T, Wang Z, Zhang X, Wang F, Wei W, Jin M, Ma J, Zeng L, Wang Y, Pei S, Zhang X. Autophagy in Rat Müller Glial Cells Is Modulated by the Sirtuin 4/AMPK/mTOR Pathway and Induces Apoptosis under Oxidative Stress. Cells 2022; 11:cells11172645. [PMID: 36078054 PMCID: PMC9454555 DOI: 10.3390/cells11172645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/14/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022] Open
Abstract
Müller glial cells (MGCs) are a group of glial cells in the retina that provide essential support to retinal neurons; however, the understanding of MGC apoptosis and autophagy remains limited. This study was aimed at investigating the role of autophagy in MGCs under normal and oxidative conditions, and identifying the underlying mechanisms. In addition, the sirtuin 4 (SIRT4)-mediated signaling pathway was observed to regulate the autophagic process in MGCs. To assess the effect of autophagy on MGC mitochondrial function and survival, we treated rMC-1 cells—rat-derived Müller glial cells—with rapamycin and 3-methyladenine (3-MA), and found that MGC death was not induced by such treatment, while autophagic dysfunction could increase MGC apoptosis under oxidative stress, as reflected by the expression level of cleaved caspase 3 and PI staining. In addition, the downregulation of autophagy by 3-MA could influence the morphology of the mitochondrial network structure, the mitochondrial membrane potential, and generation of reactive oxygen species (ROS) under oxidative stress. Moreover, SIRT4 depletion enhanced autophagosome formation, as verified by an increase in the LC3 II/I ratio and a decrease in the expression of SQSTM1/p62, and vice versa. The inhibition of AMPK phosphorylation by compound C could reverse these changes in LC3 II/I and SQSTM1/p62 caused by SIRT4 knockdown. Our research concludes that MGCs can endure autophagic dysfunction in the absence of oxidative stress, while the downregulation of autophagy can cause MGCs to become more sensitized to oxidative stress. Simultaneous exposure to oxidative stress and autophagic dysfunction in MGCs can result in a pronounced impairment of cell survival. Mechanically, SIRT4 depletion can activate the autophagic process in MGCs by regulating the AMPK–mTOR signaling pathway.
Collapse
Affiliation(s)
- Mengqi Qin
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Zhi Xie
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Ting Cao
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Zhiruo Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaoyu Zhang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Feifei Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Wei Wei
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Ming Jin
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Jingyuan Ma
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
- Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Ling Zeng
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Yanan Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Shaonan Pei
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Xu Zhang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
- Correspondence:
| |
Collapse
|
7
|
Du H, Zheng Y, Zhang W, Tang H, Jing B, Li H, Xu F, Lin J, Fu H, Chang L, Shu G. Nano-Selenium Alleviates Cadmium-Induced Acute Hepatic Toxicity by Decreasing Oxidative Stress and Activating the Nrf2 Pathway in Male Kunming Mice. Front Vet Sci 2022; 9:942189. [PMID: 35958302 PMCID: PMC9362431 DOI: 10.3389/fvets.2022.942189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/20/2022] [Indexed: 01/11/2023] Open
Abstract
Cadmium (Cd) is known as a highly toxic heavy metal and has been reported to induce hepatotoxicity in animals. Nano-selenium (NSe) is an antioxidant that plays many biological roles such as oxidative stress alleviation. The purpose of this study is to explore the mechanism of action by which NSe inhibits Cd-induced hepatic toxicity and oxidative stress. Sixty eight-week-old male Kunming mice were randomly divided into four groups (15 mice per group). The control group and cadmium groups received distilled water, whereas the sodium-selenite group received 0.2 mg/kg SSe and the NSe group received 0.2 mg/kg NSe intragastrically for 2 weeks. On the last day, all the other groups were treated with Cd (126 mg/kg) except for the control group. The results obtained in this study showed that NSe alleviated Cd-induced hepatic pathological changes. Furthermore, NSe reduced the activities of ALT and AST as well as the content of MDA, while elevated the activities of T-AOC, T-SOD and GSH (P < 0.05). In addition, the NSe group significantly increased mRNA expressions of Nrf2 pathway related molecules (Nrf2, HO-1, NQO-1, GST, GSH-Px, CAT and SOD) compared to the Cd group (P < 0.05). In conclusion, NSe shows its potentiality to reduce Cd-induced liver injury by inhibiting oxidative stress and activating the Nrf2 pathway.
Collapse
Affiliation(s)
- Hong Du
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yilei Zheng
- College of Veterinary Medicine, University of Minnesota, St Paul, MN, United States
| | - Wei Zhang
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Huaqiao Tang
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bo Jing
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Haohuan Li
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Funeng Xu
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juchun Lin
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hualin Fu
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lijen Chang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, United States
- *Correspondence: Lijen Chang
| | - Gang Shu
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Gang Shu
| |
Collapse
|
8
|
Targeting Molecular Mediators of Ferroptosis and Oxidative Stress for Neurological Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3999083. [PMID: 35910843 PMCID: PMC9337979 DOI: 10.1155/2022/3999083] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022]
Abstract
With the acceleration of population aging, nervous system diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), anxiety, depression, stroke, and traumatic brain injury (TBI) have become a huge burden on families and society. The mechanism of neurological disorders is complex, which also lacks effective treatment, so relevant research is required to solve these problems urgently. Given that oxidative stress-induced lipid peroxidation eventually leads to ferroptosis, both oxidative stress and ferroptosis are important mechanisms causing neurological disorders, targeting mediators of oxidative stress and ferroptosis have become a hot research direction at present. Our review provides a current view of the mechanisms underlying ferroptosis and oxidative stress participate in neurological disorders, the potential application of molecular mediators targeting ferroptosis and oxidative stress in neurological disorders. The target of molecular mediators or agents of oxidative stress and ferroptosis associated with neurological disorders, such as reactive oxygen species (ROS), nuclear factor erythroid 2–related factor-antioxidant response element (Nrf2-ARE), n-acetylcysteine (NAC), Fe2+, NADPH, and its oxidases NOX, has been described in this article. Given that oxidative stress-induced ferroptosis plays a pivotal role in neurological disorders, further research on the mechanisms of ferroptosis caused by oxidative stress will help provide new targets for the treatment of neurological disorders.
Collapse
|
9
|
A review on neurodegenerative diseases associated with oxidative stress and mitochondria. Int J Health Sci (Qassim) 2022. [DOI: 10.53730/ijhs.v6ns1.6130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease, Parkinson's disease, and other neurological diseases afflict people of all ages. Neuronal loss and cognitive dysfunction are common symptoms of these disorders. Overproduction of reactive oxygen species has been demonstrated to aggravate disease progression in previous investigations (ROS). Because of the large quantities of polyunsaturated fatty acids in their membranes and their fast oxygen consumption rate, neurons are especially susceptible to oxidative damage. The molecular aetiology of neurodegeneration produced by changes in redox balance has not yet been established. New antioxidants have shown considerable potential in modifying disease characteristics. For the treatment of Alzheimer's disease and other neurodegenerative illnesses such as Parkinson's disease, ALS and spinocerebellar ataxia and Huntington's disease, antioxidant-based therapies are examined extensively in the literature.
Collapse
|
10
|
Devi S, Chaturvedi M, Fatima S, Priya S. Environmental factors modulating protein conformations and their role in protein aggregation diseases. Toxicology 2022; 465:153049. [PMID: 34818560 DOI: 10.1016/j.tox.2021.153049] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/12/2021] [Accepted: 11/20/2021] [Indexed: 12/13/2022]
Abstract
The adverse physiological conditions have been long known to impact protein synthesis, folding and functionality. Major physiological factors such as the effect of pH, temperature, salt and pressure are extensively studied for their impact on protein structure and homeostasis. However, in the current scenario, the environmental risk factors (pollutants) have gained impetus in research because of their increasing concentrations in the environment and strong epidemiologic link with protein aggregation disorders. Here, we review the physiological and environmental risk factors for their impact on protein conformational changes, misfolding, aggregation, and associated pathological conditions, especially environmental risk factors associated pathologies.
Collapse
Affiliation(s)
- Shweta Devi
- Systems Toxicology and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Minal Chaturvedi
- Systems Toxicology and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Siraj Fatima
- Systems Toxicology and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Smriti Priya
- Systems Toxicology and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
11
|
Alam SI, Kim MW, Shah FA, Saeed K, Ullah R, Kim MO. Alpha-Linolenic Acid Impedes Cadmium-Induced Oxidative Stress, Neuroinflammation, and Neurodegeneration in Mouse Brain. Cells 2021; 10:cells10092274. [PMID: 34571925 PMCID: PMC8467071 DOI: 10.3390/cells10092274] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 12/27/2022] Open
Abstract
Alpha-Linolenic acid (ALA), an omega-3 polyunsaturated fatty acid, is extracted from plant sources and has been shown to be one of the anti-inflammatory and antioxidant agents. Herein, we revealed the molecular mechanism underlying the anti-inflammatory and antioxidant potential of (ALA), against cadmium in the adult mouse brain. We evaluated the neuroprotective effect of ALA (60 mg/kg per oral for 6 weeks) against CdCl2 (5 mg/kg)-induced oxidative stress, neuroinflammation, and neuronal apoptosis. According to our findings, ALA markedly reduced ROS production and nitric oxide synthase 2 (NOS2) and enhanced the expression of nuclear factor-2 erythroid-2 (Nrf-2) and heme oxygenase-1 (HO-1) in mice treated with CdCl2. Most importantly, the molecular docking study revealed that ALA allosterically decreases the overexpression of c-Jun N-terminal kinase (JNK) activity and inhibited the detrimental effect against CdCl2. Moreover, ALA suppressed CdCl2-induced glial fibrillary acidic protein (GFAP), nuclear factor-kappa b (NF-κB), and interleukin-1β (IL-1β) in the mouse brain. Further, we also checked the pro- and anti-apoptotic proteins markers such as Bax, Bcl-2, and caspase-3, which were regulated in the cortex of ALA co-treated mouse brain. Overall, our study suggests that oral administration of ALA can impede oxidative stress, neuroinflammation, and increase neuronal apoptosis in the cortex of Cd-injected mouse brain.
Collapse
Affiliation(s)
- Sayed-Ibrar Alam
- Division of Life Sciences and Applied Life Science (BK 21 PLUS), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (S.-I.A.); (M.-W.K.); (K.S.); (R.U.)
| | - Min-Woo Kim
- Division of Life Sciences and Applied Life Science (BK 21 PLUS), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (S.-I.A.); (M.-W.K.); (K.S.); (R.U.)
| | - Fawad Ali Shah
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan;
| | - Kamran Saeed
- Division of Life Sciences and Applied Life Science (BK 21 PLUS), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (S.-I.A.); (M.-W.K.); (K.S.); (R.U.)
| | - Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21 PLUS), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (S.-I.A.); (M.-W.K.); (K.S.); (R.U.)
| | - Myeong-Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21 PLUS), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (S.-I.A.); (M.-W.K.); (K.S.); (R.U.)
- Correspondence: ; Tel.: +82-55-772-1345
| |
Collapse
|
12
|
Cheng H, Yang B, Ke T, Li S, Yang X, Aschner M, Chen P. Mechanisms of Metal-Induced Mitochondrial Dysfunction in Neurological Disorders. TOXICS 2021; 9:142. [PMID: 34204190 PMCID: PMC8235163 DOI: 10.3390/toxics9060142] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 01/31/2023]
Abstract
Metals are actively involved in multiple catalytic physiological activities. However, metal overload may result in neurotoxicity as it increases formation of reactive oxygen species (ROS) and elevates oxidative stress in the nervous system. Mitochondria are a key target of metal-induced toxicity, given their role in energy production. As the brain consumes a large amount of energy, mitochondrial dysfunction and the subsequent decrease in levels of ATP may significantly disrupt brain function, resulting in neuronal cell death and ensuing neurological disorders. Here, we address contemporary studies on metal-induced mitochondrial dysfunction and its impact on the nervous system.
Collapse
Affiliation(s)
- Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China; (H.C.); (X.Y.)
| | - Bobo Yang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China;
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China; (H.C.); (X.Y.)
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou 545006, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| |
Collapse
|
13
|
Aminzadeh A, Salarinejad A. Effects of myricetin against cadmium-induced neurotoxicity in PC12 cells. Toxicol Res (Camb) 2021; 10:84-90. [PMID: 33613976 DOI: 10.1093/toxres/tfaa104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/27/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023] Open
Abstract
Cadmium (Cd) is one of the most prevalent toxic metals widely found in the environment. Cd induces toxicity and apoptosis in various organs and cells. The nervous system is one of the primary organs targeted by Cd. Cd toxicity is correlated with induction of severe oxidative stress. Myricetin, a natural product, has been found to exert protective effects against various disease conditions. The present study aimed to evaluate the potential protective effects of myricetin on Cd-induced neurotoxicity in PC12 cells. The cells were pretreated with myricetin in the absence and presence of Cd. The viability of cells was assessed using the MTT assay. Markers of oxidative stress were investigated by the lipid peroxidation (LPO), glutathione (GSH) content, and total antioxidant capacity (TAC). Moreover, activation of caspase 3 was examined by Western blot analysis. Myricetin could significantly enhance the viability of PC12 cells. Pretreatment of the cells with myricetin, prior to Cd exposure, showed a significant decrease in the levels of LPO whereas GSH and TAC levels were increased. In addition, the activity of caspase-3 was notably prevented by myricetin. These findings revealed that myricetin has protective effects on Cd-induced neurotoxicity in PC12 cells, which can be linked to its antioxidant potential, inhibition of LPO, and prevention of caspase-3 activation.
Collapse
Affiliation(s)
- Azadeh Aminzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Haft-Bagh Blvd., P.O. Box 7616911319, Kerman, Iran.,Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Haft-Bagh Blvd., P.O. Box 7616911319, Kerman, Iran
| | - Ayda Salarinejad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Haft-Bagh Blvd., P.O. Box 7616911319, Kerman, Iran
| |
Collapse
|
14
|
Fang J, Yin H, Yang Z, Tan M, Wang F, Chen K, Zuo Z, Shu G, Cui H, Ouyang P, Guo H, Chen Z, Huang C, Geng Y, Liu W. Vitamin E protects against cadmium-induced sub-chronic liver injury associated with the inhibition of oxidative stress and activation of Nrf2 pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111610. [PMID: 33396130 DOI: 10.1016/j.ecoenv.2020.111610] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 06/12/2023]
Abstract
Hepatic oxidative stress, as one important mechanism of cadmium (Cd)-induced hepatic toxicity, could, as known, be ameliorated by vitamin E (VE). However, the underlying mechanism remains to be elucidated. To investigate whether the antioxidant vitamin E can protect against Cd-induced sub-chronic liver injury associated with oxidative stress and nuclear factor erythrocyte 2-related factor 2 (Nrf2) pathway, male Sprague-Dawley rats (nine-week-old) were randomly divided into four groups (eight rats/group), namely, control, VE (100 mg/kg VE), Cd (5 mg/kg CdCl2) and VE+Cd (100 mg/kg VE+5 mg/kg CdCl2), and received intragastric administration of Cd and/or VE for four weeks. Cd-exposure alone resulted in reduced liver weight, liver histological alteration and oxidative stress, accumulation of Cd in the liver, elevated ALT and AST concentrations in serum together with decreased mRNA and protein expressions of Nrf2 pathway related molecules (Nrf2, HO-1, NQO-1, GCLC, GCLM and GST). However, the co-treatment of Cd and VE significantly ameliorated the changes mentioned above, and promoted the expression of genes and proteins of Nrf2 pathway related molecules in comparison to the Cd-exposure alone. Our results indicate that the protective effect of VE against Cd-induced sub-chronic hepatic damage in rats is associated with the inhibition of oxidative stress and activation of Nrf2 pathway.
Collapse
Affiliation(s)
- Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Heng Yin
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Zhuangzhi Yang
- Chengdu Academy of Agriculture and Forestry Sciences, Chengdu, Sichuan 611130, PR China
| | - Maoyun Tan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Fengyuan Wang
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chendu, Sichuan 610041, PR China
| | - Kejie Chen
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, PR China.
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.
| | - Gang Shu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Ping Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Zhengli Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Yi Geng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Wentao Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| |
Collapse
|
15
|
Li B, Xia M, Zorec R, Parpura V, Verkhratsky A. Astrocytes in heavy metal neurotoxicity and neurodegeneration. Brain Res 2021; 1752:147234. [PMID: 33412145 DOI: 10.1016/j.brainres.2020.147234] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/15/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
With the industrial development and progressive increase in environmental pollution, the mankind overexposure to heavy metals emerges as a pressing public health issue. Excessive intake of heavy metals, such as arsenic (As), manganese (Mn), mercury (Hg), aluminium (Al), lead (Pb), nickel (Ni), bismuth (Bi), cadmium (Cd), copper (Cu), zinc (Zn), and iron (Fe), is neurotoxic and it promotes neurodegeneration. Astrocytes are primary homeostatic cells in the central nervous system. They protect neurons against all types of insults, in particular by accumulating heavy metals. However, this makes astrocytes the main target for heavy metals neurotoxicity. Intake of heavy metals affects astroglial homeostatic and neuroprotective cascades including glutamate/GABA-glutamine shuttle, antioxidative machinery and energy metabolism. Deficits in these astroglial pathways facilitate or even instigate neurodegeneration. In this review, we provide a concise outlook on heavy metal-induced astrogliopathies and their association with major neurodegenerative disorders. In particular, we focus on astroglial mechanisms of iron-induced neurotoxicity. Iron deposits in the brain are detected in main neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Accumulation of iron in the brain is associated with motor and cognitive impairments and iron-induced histopathological manifestations may be considered as the potential diagnostic biomarker of neurodegenerative diseases. Effective management of heavy metal neurotoxicity can be regarded as a potential strategy to prevent or retard neurodegenerative pathologies.
Collapse
Affiliation(s)
- Baoman Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China.
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang, People's Republic of China
| | - Robert Zorec
- Celica BIOMEDICAL, Tehnološki Park 24, 1000 Ljubljana, Slovenia; Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alexei Verkhratsky
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain.
| |
Collapse
|
16
|
Werder EJ, Engel LS, Curry MD, Sandler DP. Selenium modifies associations between multiple metals and neurologic symptoms in Gulf states residents. Environ Epidemiol 2020; 4:e115. [PMID: 33336134 PMCID: PMC7727467 DOI: 10.1097/ee9.0000000000000115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/23/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Metals have been shown to have a wide range of neurologic effects across the life course, but most studies consider neurodevelopment or neurodegenerative diseases in older adults. We investigated exposure to metals during adulthood in association with subclinical neurologic endpoints, considering the metals individually and as a mixture, and potential interactions among exposures. METHODS We measured blood levels of cadmium, lead, mercury, manganese, and selenium in 1007 Gulf state residents and estimated cross-sectional associations between ranked levels of blood metals and the presence of self-reported neurologic symptoms. Single pollutant models were mutually adjusted for other metals and we used quantile g-computation to evaluate associations with exposure to the combined mixture. In stratified analyses, we assessed heterogeneity by smoking and blood selenium. RESULTS The highest quartile of cadmium was associated with a higher prevalence of central nervous system symptoms (prevalence ratio [PR] = 1.50; 95% confidence interval [CI] = 1.13, 1.99), with stronger associations among nonsmokers (PR = 1.63; 95% CI = 1.11, 2.38) and those with low selenium (PR = 2.29, 95% CI = 1.50, 3.49). Selenium also modified associations between lead and peripheral nervous system symptoms, with increased symptoms in the low selenium group at all quartiles of exposure (P-trend = 0.07). Conversely, those with the highest co-exposure to mercury and selenium had reduced neurologic symptoms (PR = 0.73, 95% CI = 0.55, 0.96). Results of the mixture analysis were consistent with single chemical results. CONCLUSIONS Cadmium exhibited the most consistent relationship with increased neurologic symptoms, though lead was an important exposure in subgroup analyses. Selenium may modify subclinical neurotoxic effects of metals at non-occupational levels in adults.
Collapse
Affiliation(s)
- Emily J. Werder
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, DHHS, Research Triangle Park, North Carolina
| | - Lawrence S. Engel
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, DHHS, Research Triangle Park, North Carolina
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | | | - Dale P. Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, DHHS, Research Triangle Park, North Carolina
| |
Collapse
|
17
|
Arruda HS, Neri-Numa IA, Kido LA, Maróstica Júnior MR, Pastore GM. Recent advances and possibilities for the use of plant phenolic compounds to manage ageing-related diseases. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
18
|
Caffeic acid phenethyl ester reversed cadmium-induced cell death in hippocampus and cortex and subsequent cognitive disorders in mice: Involvements of AMPK/SIRT1 pathway and amyloid-tau-neuroinflammation axis. Food Chem Toxicol 2020; 144:111636. [PMID: 32739455 DOI: 10.1016/j.fct.2020.111636] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 12/26/2022]
Abstract
Exposure to nonbiodegradable cadmium (Cd) causes many health problems including the damage to the nervous system. This study aimed to increase knowledge about its neurotoxic effects and the neuroprotective potential of caffeic acid phenethyl ester (CAPE, a polyphenol abundant in honeybee propolis). In mice, CAPE (10 μmol/kg/day body weight) attenuated significantly learning and memory deficits induced by CdCl2 (1.5 mg/kg/day body weight). For the CdCl2-treated mice, CAPE increased crossing number in open field test, decreased the alternation in Y-maze test, and increased the latency time and error number in step down test. CAPE also inhibited CdCl2-initiated Aβ accumulation and activation of pro-inflammatory factors and microglia in the brains. Therefore, CAPE could be a food-derived neuroprotective agent against Cd-induced neurotoxicity and neurodegenerative disorders, through attenuating neuronal apoptosis and neuroinflammation via the AMPK/SIRT1 pathway and amyloid-tau-neuroinflammation axis.
Collapse
|
19
|
Ma Y, Rivera-Ingraham G, Nommick A, Bickmeyer U, Roeder T. Copper and cadmium administration induce toxicity and oxidative stress in the marine flatworm Macrostomum lignano. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 221:105428. [PMID: 32035411 DOI: 10.1016/j.aquatox.2020.105428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/22/2020] [Accepted: 01/30/2020] [Indexed: 06/10/2023]
Abstract
The contamination of coastal regions with different toxicants, including heavy metal ions such as copper and cadmium jeopardize health and survival of organisms exposed to this habitat. To study the effects of high copper and cadmium concentrations in these marine environments, we used the flatworm Macrostomum lignano as a model. This platyhelminth lives in shallow coastal water and is exposed to high concentrations of all toxicants that accumulate in these sea floors. We could show that both, cadmium and copper show toxicity at higher concentrations, with copper being more toxic than cadmium. At concentrations below acute toxicity, a reduced long-term survival was observed for both metal ions. The effects of sublethal doses comprise reduced physical activities, an increase in ROS levels within the worms, and alterations of the mitochondrial biology. Moreover, cell death events were substantially increased in response to sublethal concentrations of both metal ions and stem cell activity was reduced following exposure to higher cadmium concentrations. Finally, the expression of several genes involved in xenobiotic metabolism was substantially altered by this intervention. Taken together, M. lignano has been identified as a suitable model for marine toxicological studies as it allows to quantify several relevant life-history traits as well as of physiological and behavioral read-outs.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Kiel University, Zoological Institute, Molecular Physiology, Olshausenstrasse 40, 24098, Kiel, Germany.
| | - Georgina Rivera-Ingraham
- Laboratoire Environement de Petit Saut, Hydreco-Guyane. BP 823, 97310, Kourou, French Guiana, France.
| | - Aude Nommick
- Institut de Biologie de Dévelopement de Marseille, Marseille, France.
| | - Ulf Bickmeyer
- Alfred-Wegener-Institute Helmholtz Center for Polar- and Marine Research, Biosciences, Ecological Chemistry, Am Handelshafen 12, 27570, Bremerhaven, Germany.
| | - Thomas Roeder
- Kiel University, Zoological Institute, Molecular Physiology, Olshausenstrasse 40, 24098, Kiel, Germany; German Center for Lung Research (DZL, Airway Research Center North), Kiel, Germany.
| |
Collapse
|
20
|
Qu KC, Wang ZY, Tang KK, Zhu YS, Fan RF. Trehalose suppresses cadmium-activated Nrf2 signaling pathway to protect against spleen injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 181:224-230. [PMID: 31195231 DOI: 10.1016/j.ecoenv.2019.06.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/31/2019] [Accepted: 06/02/2019] [Indexed: 06/09/2023]
Abstract
Cadmium (Cd), as a kind of ubiquitous and highly toxic heavy metal pollutants, has been known to result in immunotoxicity in animals. As a multifunctional bioactivity disaccharide, trehalose (Tre) is characterized by antioxidative, antiapoptotic, and accelerating autophagy. In this study, Sprague-Dawley (SD) rats were fed with cadmium chloride (CdCl2) and/or Tre to explore the molecular mechanisms of Tre-protected against spleen injury caused by Cd exposure. Firstly, the results showed that Tre partially recovered splenic pathological changes induced by Cd exposure. Secondly, Tre dramatically declined the level of methane dicarboxylic aldehyde (MDA) and elevated the level of total antioxidant capacity (T-AOC) to weaken oxidative stress caused by Cd exposure in spleen tissue. Moreover, the results showed that Tre significantly suppressed Cd-induced the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and up-regulated the protein expression of nuclear Nrf2. Thirdly, Tre remarkably reduced the protein expression of sequestosome 1 (p62/SQSTM1) and microtubule-associated protein light chain 3II (LC-3II) to restore autophagy inhibition induced by Cd exposure. Finally, the results of TUNEL and the expression of apoptosis marker proteins showed that Tre significantly inhibited Cd-induced apoptosis in spleen tissue to exert its protective effects. In summary, the results indicated that Tre modulated Nrf2 signaling pathway, which interacted with apoptosis and autophagy to against Cd-induced spleen injury, providing potential therapeutic strategies for the prevention and treatment of Cd-related immune system diseases.
Collapse
Affiliation(s)
- Kui-Chao Qu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China
| | - Zhen-Yong Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China
| | - Kou-Kou Tang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China
| | - Yi-Song Zhu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China
| | - Rui-Feng Fan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China.
| |
Collapse
|
21
|
Qian ZM, Ke Y. Brain iron transport. Biol Rev Camb Philos Soc 2019; 94:1672-1684. [PMID: 31190441 DOI: 10.1111/brv.12521] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 12/19/2022]
Abstract
Brain iron is a crucial participant and regulator of normal physiological activity. However, excess iron is involved in the formation of free radicals, and has been associated with oxidative damage to neuronal and other brain cells. Abnormally high brain iron levels have been observed in various neurodegenerative diseases, including neurodegeneration with brain iron accumulation, Alzheimer's disease, Parkinson's disease and Huntington's disease. However, the key question of why iron levels increase in the relevant regions of the brain remains to be answered. A full understanding of the homeostatic mechanisms involved in brain iron transport and metabolism is therefore critical not only for elucidating the pathophysiological mechanisms responsible for excess iron accumulation in the brain but also for developing pharmacological interventions to disrupt the chain of pathological events occurring in these neurodegenerative diseases. Numerous studies have been conducted, but to date no effort to synthesize these studies and ideas into a systematic and coherent summary has been made, especially concerning iron transport across the luminal (apical) membrane of the capillary endothelium and the membranes of different brain cell types. Herein, we review key findings on brain iron transport, highlighting the mechanisms involved in iron transport across the luminal (apical) as well as the abluminal (basal) membrane of the blood-brain barrier, the blood-cerebrospinal fluid barrier, and iron uptake and release in neurons, oligodendrocytes, astrocytes and microglia within the brain. We offer suggestions for addressing the many important gaps in our understanding of this important topic, and provide new insights into the potential causes of abnormally increased iron levels in regions of the brain in neurodegenerative disorders.
Collapse
Affiliation(s)
- Zhong-Ming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, 226019, China.,Laboratory of Neuropharmacology, School of Pharmacy, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
22
|
Wang S, Ren X, Hu X, Zhou L, Zhang C, Zhang M. Cadmium-induced apoptosis through reactive oxygen species-mediated mitochondrial oxidative stress and the JNK signaling pathway in TM3 cells, a model of mouse Leydig cells. Toxicol Appl Pharmacol 2019; 368:37-48. [DOI: 10.1016/j.taap.2019.02.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/24/2022]
|