1
|
Cassera E, Ferrari E, Vignati DAL, Capucciati A. The interaction between metals and catecholamines: oxidative stress, DNA damage, and implications for human health. Brain Res Bull 2025; 226:111366. [PMID: 40306586 DOI: 10.1016/j.brainresbull.2025.111366] [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: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
The interaction between metals and catecholamines plays a pivotal role in the generation of reactive oxygen species (ROS), leading to oxidative stress and DNA damage. ROS are linked to several diseases, including neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. This review examines how essential metals (iron, copper, zinc, manganese) and a few non-essential metal(loid)s (mercury, chromium, arsenic, aluminum, cadmium, and nickel) contribute to oxidative stress in the presence of catecholamines. In the presence of metals, catecholamines can cause oxidative DNA modification, possibly resulting in cell apoptosis, by taking part in redox reactions and oxidizing to the corresponding aminochrome with simultaneous ROS production. Essential metals are vital for physiological functions, but imbalances in their homeostasis can be harmful. Furthermore, non-essential metals, commonly encountered through environmental or occupational exposure, can exhibit significant toxicity. Previous studies on catecholamine-induced oxidative stress focused on copper and iron, but this review emphasizes the need to investigate other neurotoxic metals and expand existing knowledge on the interactions between metals, catecholamines, and DNA damage. Results from such research could help prioritizing the development of new assessment methods associated with adverse outcome pathways, to reliably predict harmful effects on human health, aiding in the development of therapeutical strategies. The present work will help to shed light on the interplay of metals, catecholamines, and DNA damage in different diseases hopefully fostering new research in this still understudied topic. Future research should investigate the molecular mechanisms through which these metals affect neuronal health and contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Elena Cassera
- Department of Chemistry, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Emanuele Ferrari
- National Research Council of Italy, Water Research Institute (CNR-IRSA) Molecular Ecology Group (MEG), Largo Tonolli 50, Verbania 28922, Italy.
| | | | - Andrea Capucciati
- Department of Chemistry, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy; Fondazione Grigioni per il Morbo di Parkinson, Via Gianfranco Zuretti 35, Milano 20125, Italy
| |
Collapse
|
2
|
Fahey JW, Liu H, Batt H, Panjwani AA, Tsuji P. Sulforaphane and Brain Health: From Pathways of Action to Effects on Specific Disorders. Nutrients 2025; 17:1353. [PMID: 40284217 PMCID: PMC12030691 DOI: 10.3390/nu17081353] [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: 03/26/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
The brain accounts for about 2% of the body's weight, but it consumes about 20% of the body's energy at rest, primarily derived from ATP produced in mitochondria. The brain thus has a high mitochondrial density in its neurons because of its extensive energy demands for maintaining ion gradients, neurotransmission, and synaptic activity. The brain is also extremely susceptible to damage and dysregulation caused by inflammation (neuroinflammation) and oxidative stress. Many systemic challenges to the brain can be mitigated by the phytochemical sulforaphane (SF), which is particularly important in supporting mitochondrial function. SF or its biogenic precursor glucoraphanin, from broccoli seeds or sprouts, can confer neuroprotective and cognitive benefits via diverse physiological and biochemical mechanisms. SF is able to cross the blood-brain barrier as well as to protect it, and it mitigates the consequences of destructive neuroinflammation. It also protects against the neurotoxic effects of environmental pollutants, combats the tissue and cell damage wrought by advanced glycation end products (detoxication), and supports healthy glucose metabolism. These effects are applicable to individuals of all ages, from the developing brains in periconception and infancy, to cognitively, developmentally, and traumatically challenged brains, to those in later life as well as those who are suffering with multiple chronic conditions including Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Jed W. Fahey
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- iMIND Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute of Medicine, University of Maine, Orono, ME 04469, USA
| | - Hua Liu
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Holly Batt
- Anti-AGEs Foundation, Depew, NY 14043, USA;
| | - Anita A. Panjwani
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA;
- Center on Aging and the Life Course, Purdue University, West Lafayette, IN 47907, USA
| | - Petra Tsuji
- Department of Biological Sciences, Towson University, Towson, MD 21252, USA;
| |
Collapse
|
3
|
Dutta A, Roy R, Pandey M, Chhetry S, Phukan BC, Roy A, Bhattacharya P, Borah A. Arsenic-induced mice model of Parkinson's disease: Revealing the neurotoxicity of arsenic through mitochondrial complexes inhibition and dopaminergic neurodegeneration in the substantia nigra region of brain. Brain Res 2025; 1851:149493. [PMID: 39909295 DOI: 10.1016/j.brainres.2025.149493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/05/2024] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
The role of environmental contaminants in causing Parkinson's disease (PD) is well known, with rotenone and paraquat being the notable neurotoxins. Traces of the metalloid arsenic are frequently found in drinking water which is considered a threat to the brain's health. Pre-clinical and epidemiological studies have associated arsenic with PD whereby behavioral and neurochemical alterations were observed. However, the impact of arsenic toxicity on the dopaminergic neurons of substantia nigra (SN), the hallmark region which degenerates in PD, has not been shown yet. In the present study, administration of 20 mg/kg b.w., arsenic for 28 days caused significant loss of dopaminergic neurons and their terminals respectively in the SN and striatum regions of mice brain. Moreover, the arsenic-fed rodents exhibited depleted striatal dopamine, prolonged latency to move and correct posture, and reduced exploratory behavior and neurological severity. Further, mitochondrial complexes II and IV were found to be inhibited in the SN, cortex, striatum, and hippocampus of arsenic-fed mice. Additionally, inflammatory marker glial fibrillary acidic protein (GFAP) and neuronal nitric oxide synthase (nNOS) expressed in glial cells and neurons respectively were enhanced in the nigrostriatal pathway of arsenic-fed animals. The present study for the first time reports that arsenic causes Parkinsonism by degenerating nigrostriatal dopaminergic neurons through mitochondrial complex inhibition and inflammatory stress. The study further puts forward validatory evidence for the potential of arsenic in causing PD and the reliability of the arsenic-induced PD model for exploring the disease pathogenesis and treatment.
Collapse
Affiliation(s)
- Ankumoni Dutta
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011 Assam, India; Department of Zoology, Pandit Deendayal Upadhyaya Adarsha Mahavidyalaya (PDUAM), Behali, Biswanath 784184 Assam, India.
| | - Rubina Roy
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011 Assam, India
| | - Mritunjay Pandey
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, United States
| | - Sushila Chhetry
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011 Assam, India
| | | | - Abhideep Roy
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011 Assam, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad - 382355, Gandhinagar, Gujarat, India
| | - Anupom Borah
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011 Assam, India.
| |
Collapse
|
4
|
Castellani S, Iaconisi GN, Tripaldi F, Porcelli V, Trapani A, Messina E, Guerra L, Di Franco C, Maruccio G, Monteduro AG, Corbo F, Di Gioia S, Trapani G, Conese M. Dopamine and Citicoline-Co-Loaded Solid Lipid Nanoparticles as Multifunctional Nanomedicines for Parkinson's Disease Treatment by Intranasal Administration. Pharmaceutics 2024; 16:1048. [PMID: 39204393 PMCID: PMC11360708 DOI: 10.3390/pharmaceutics16081048] [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: 07/09/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
This work aimed to evaluate the potential of the nanosystems constituted by dopamine (DA) and the antioxidant Citicoline (CIT) co-loaded in solid lipid nanoparticles (SLNs) for intranasal administration in the treatment of Parkinson disease (PD). Such nanosystems, denoted as DA-CIT-SLNs, were designed according to the concept of multifunctional nanomedicine where multiple biological roles are combined into a single nanocarrier and prepared by the melt emulsification method employing the self-emulsifying Gelucire® 50/13 as lipid matrix. The resulting DA-CIT-SLNs were characterized regarding particle size, surface charge, encapsulation efficiency, morphology, and physical stability. Differential scanning calorimetry, FT-IR, and X ray diffraction studies were carried out to gain information on solid-state features, and in vitro release tests in simulated nasal fluid (SNF) were performed. Monitoring the particle size at two temperatures (4 °C and 37 °C), the size enlargement observed over the time at 37 °C was lower than that observed at 4 °C, even though at higher temperature, color changes occurred, indicative of possible neurotransmitter decomposition. Solid-state studies indicated a reduction in the crystallinity when DA and CIT are co-encapsulated in DA-CIT-SLNs. Interestingly, in vitro release studies in SNF indicated a sustained release of DA. Furthermore, DA-CIT SLNs displayed high cytocompatibility with both human nasal RPMI 2650 and neuronal SH-SY5Y cells. Furthermore, OxyBlot assay demonstrated considerable potential to assess the protective effect of antioxidant agents against oxidative cellular damage. Thus, such protective effect was shown by DA-CIT-SLNs, which constitute a promising formulation for PD application.
Collapse
Affiliation(s)
- Stefano Castellani
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Giorgia Natalia Iaconisi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| | - Francesca Tripaldi
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (F.T.); (F.C.); (G.T.)
| | - Vito Porcelli
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy; (V.P.); (E.M.); (L.G.)
| | - Adriana Trapani
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (F.T.); (F.C.); (G.T.)
| | - Eugenia Messina
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy; (V.P.); (E.M.); (L.G.)
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy; (V.P.); (E.M.); (L.G.)
| | | | - Giuseppe Maruccio
- Omnics Research Group, Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento and INFN Sezione di Lecce, Via per Monteroni, 73100 Lecce, Italy (A.G.M.)
- CNR-NANOTEC Institute of Nanotechnology, Via per Monteroni, 73100 Lecce, Italy
| | - Anna Grazia Monteduro
- Omnics Research Group, Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento and INFN Sezione di Lecce, Via per Monteroni, 73100 Lecce, Italy (A.G.M.)
- CNR-NANOTEC Institute of Nanotechnology, Via per Monteroni, 73100 Lecce, Italy
| | - Filomena Corbo
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (F.T.); (F.C.); (G.T.)
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| | - Giuseppe Trapani
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (F.T.); (F.C.); (G.T.)
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| |
Collapse
|
5
|
Almeer R, Alyami NM. The protective effect of apigenin against inorganic arsenic salt-induced toxicity in PC12 cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:106625-106635. [PMID: 37730986 DOI: 10.1007/s11356-023-29884-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
Poisoning by arsenic affects people worldwide, and many human illnesses and health issues, including neurotoxicity, have been linked to chronic exposure to arsenic. When exposed to arsenic, the body produces intracellular reactive oxygen species (ROS), which influence a variety of alterations in cellular activity and directly harm molecules through oxidation. Arsenic-induced lesions are improved by antioxidants with the ability to lower ROS levels. Therefore, the current research aimed to assess how well apigenin protected PC12 cells from the toxicity caused by inorganic arsenic salt (iAs). For 24 and 48 h, iAs and/or apigenin were applied to PC12 cells. Then, oxidative stress indicators like malondialdehyde (MDA), nitric oxide (NO), and ROS in addition to the enzymatic and non-enzymatic antioxidant molecules such as catalase (CAT), glutathione (GSH), and superoxide dismutase (SOD) were assessed. Moreover, after exposure to iAs, PC12 was examined for nuclear factor erythroid 2-related factor 2 (Nrf2) expression to clarify how apigenin manifests its neuroprotection. Furthermore, NF-kB p65 concentration and IL-1B, IL-6, and TNF-α mRNA expression were measured to assess neuroinflammation. Bax, caspase-3, and Bcl-2 levels were measured to investigate apigenin's potential to protect PC12 cells from iAs poisoning. The obtained results revealed that, the cell survival rate in the iAs group was significantly lower (P < 0.05), and the number of viable cells steadily increased after apigenin treatment. Furthermore, the study found that iAs decreased GSH, CAT, and SOD in the PC12 cells while increasing ROS, MDA, and NO levels. In PC12 cells, the capacity of iAs to cause oxidative stress was linked to the induction of neuroinflammation and apoptosis. Interestingly, apigenin pre-treatment of PC12 cells resulted in exceptional protection against iAs-induced neuroinflammation, oxidative stress, and apoptotic cell death. Nrf2 upregulation in PC12 cells may explain the neuroprotection effect of apigenin against iAs toxicity. In conclusion, the obtained results of the present study have clinical significance and indicate that apigenin is a promising candidate for shielding the nervous system from toxic effects caused by arsenic. These findings require further investigation using in vivo experimental models.
Collapse
Affiliation(s)
- Rafa Almeer
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, 11451, Riyadh, Saudi Arabia.
| | - Nouf M Alyami
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Rachamalla M, Salahinejad A, Khan M, Datusalia AK, Niyogi S. Chronic dietary exposure to arsenic at environmentally relevant concentrations impairs cognitive performance in adult zebrafish (Danio rerio) via oxidative stress and dopaminergic dysfunction. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 886:163771. [PMID: 37164085 DOI: 10.1016/j.scitotenv.2023.163771] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/12/2023]
Abstract
The current study was designed to evaluate the effects of chronic dietary arsenic exposure on the cognitive performance of adult zebrafish and uncover probable pathways by which arsenic mediates such neurotoxic effects. Adult zebrafish were treated with 3 different dietary arsenic concentrations (30, 60, and 100 μg/g dry weight (dw), as arsenite) in addition to control for 60 days. A latent learning paradigm, which employs a complex maze, was used to assess the cognitive performance of fish. Our results demonstrated that dietary treatment with arsenic, especially at medium (60 μg/g dw) and high (100 μg/g dw) exposure dose levels, significantly impaired the performance of fish in various latent learning tasks evaluated in the present study. Concomitant with cognitive dysfunction, chronic dietary exposure to arsenic was also found to increase arsenic accumulation and dopamine levels, and induce oxidative stress (reduced thiol redox, increased lipid peroxidation and expression of antioxidant enzyme genes) in the brain of zebrafish in a dose-dependent manner. Dopaminergic system in the brain is known to play a critical role in regulating cognitive behaviours in fish, and our observations suggested that chronic dietary treatment with medium and high arsenic doses leads to significant alterations in the expression of genes involved in dopamine signalling (dopamine receptors), synthesis (thyroxine hydroxylase) and metabolism (monoamine oxidase) in the zebrafish brain. Moreover, we also recorded significant downregulation of genes such as the brain-derived neurotrophic factor (BDNF) and ectonucleotidases (entpd2_mg, entpd2_mq, and 5'-nucleotidase), which are critical for learning and memory functions, in the zebrafish brain following chronic dietary exposure to arsenic. Overall, the present study suggests that chronic environmentally relevant dietary exposure to arsenic can impair the cognitive performance in zebrafish, essentially by inducing oxidative stress and disrupting the dopaminergic neurotransmission in the brain.
Collapse
Affiliation(s)
- Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada.
| | - Arash Salahinejad
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Maria Khan
- College of Kinesiology, University of Saskatchewan, Saskatoon, SK S7N 5B2, Canada
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, SK S7N 5B3, Canada
| |
Collapse
|
7
|
Vellingiri B, Suriyanarayanan A, Selvaraj P, Abraham KS, Pasha MY, Winster H, Gopalakrishnan AV, G S, Reddy JK, Ayyadurai N, Kumar N, Giridharan B, P S, Rao KRSS, Nachimuthu SK, Narayanasamy A, Mahalaxmi I, Venkatesan D. Role of heavy metals (copper (Cu), arsenic (As), cadmium (Cd), iron (Fe) and lithium (Li)) induced neurotoxicity. CHEMOSPHERE 2022; 301:134625. [PMID: 35439490 DOI: 10.1016/j.chemosphere.2022.134625] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/30/2022] [Accepted: 04/12/2022] [Indexed: 05/15/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative condition characterized by the dopamine (DA) neuronal loss in the substantia nigra. PD impairs motor controls symptoms such as tremor, rigidity, bradykinesia and postural imbalance gradually along with non-motor problems such as olfactory dysfunction, constipation, sleeping disorder. Though surplus of factors and mechanisms have been recognized, the precise PD etiopathogenesis is not yet implied. Reports suggest that various environmental factors play a crucial role in the causality of the PD cases. Epidemiological studies have reported that heavy metals has a role in causing defects in substantia nigra region of brain in PD. Though the reason is unknown, exposure to heavy metals is reported to be an underlying factor in PD development. Metals are classified as either essential or non-essential, and they have a role in physiological processes such protein modification, electron transport, oxygen transport, redox reactions, and cell adhesion. Excessive metal levels cause oxidative stress, protein misfolding, mitochondrial malfunction, autophagy dysregulation, and apoptosis, among other things. In this review, we check out the link between heavy metals like copper (Cu), arsenic (As), cadmium (Cd), iron (Fe), and lithium (Li) in neurodegeneration, and how it impacts the pathological conditions of PD. In conclusion, increase or decrease in heavy metals involve in regulation of neuronal functions that have an impact on neurodegeneration process. Through this review, we suggest that more research is needed in this stream to bring more novel approaches for either disease modelling or therapeutics.
Collapse
Affiliation(s)
- Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Atchaya Suriyanarayanan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Priyanka Selvaraj
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Kripa Susan Abraham
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Md Younus Pasha
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Harysh Winster
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India; Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India
| | - Singaravelu G
- Department of Education, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | | | - Niraikulam Ayyadurai
- CSIR-Central Leather Research Institute, Adyar, Chennai, 600 020, Tamil Nadu, India
| | - Nandha Kumar
- Department of Zoology, St. Joseph University, 797 115, Dimapur, Nagaland
| | - Bupesh Giridharan
- Department of Forest Science, Nagaland University, Lumami, Zunheboto, Nagaland, India
| | - Sivaprakash P
- Department of Mechanical Engineering, Dr.N.G.P. Institute of Technology, Coimbatore, 641048, Tamil Nadu, India
| | - K R S Sambasiva Rao
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796 004, Mizoram, India
| | - Senthil Kumar Nachimuthu
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796 004, Mizoram, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| | - Iyer Mahalaxmi
- Livestock Farming and Bioresource Technology, Tamil Nadu, India.
| | - Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| |
Collapse
|
8
|
De Miranda BR, Goldman SM, Miller GW, Greenamyre JT, Dorsey ER. Preventing Parkinson's Disease: An Environmental Agenda. JOURNAL OF PARKINSONS DISEASE 2021; 12:45-68. [PMID: 34719434 PMCID: PMC8842749 DOI: 10.3233/jpd-212922] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fueled by aging populations and continued environmental contamination, the global burden of Parkinson's disease (PD) is increasing. The disease, or more appropriately diseases, have multiple environmental and genetic influences but no approved disease modifying therapy. Additionally, efforts to prevent this debilitating disease have been limited. As numerous environmental contaminants (e.g., pesticides, metals, industrial chemicals) are implicated in PD, disease prevention is possible. To reduce the burden of PD, we have compiled preclinical and clinical research priorities that highlight both disease prediction and primary prevention. Though not exhaustive, the "PD prevention agenda" builds upon many years of research by our colleagues and proposes next steps through the lens of modifiable risk factors. The agenda identifies ten specific areas of further inquiry and considers the funding and policy changes that will be necessary to help prevent the world's fastest growing brain disease.
Collapse
Affiliation(s)
- Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama atBirmingham, Birmingham, AL, USA
| | - Samuel M Goldman
- Division of Occupational and Environmental Medicine, San Francisco VeteransAffairs Health Care System, School of Medicine, University ofCalifornia-San Francisco, San Francisco, CA, USA
| | - Gary W Miller
- Department of Environmnetal Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, Universityof Pittsburgh, Pittsburgh, PA, USA
| | - E Ray Dorsey
- Center for Health+Technology and Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
9
|
Thakur M, Rachamalla M, Niyogi S, Datusalia AK, Flora SJS. Molecular Mechanism of Arsenic-Induced Neurotoxicity including Neuronal Dysfunctions. Int J Mol Sci 2021; 22:10077. [PMID: 34576240 PMCID: PMC8471829 DOI: 10.3390/ijms221810077] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Arsenic is a key environmental toxicant having significant impacts on human health. Millions of people in developing countries such as Bangladesh, Mexico, Taiwan, and India are affected by arsenic contamination through groundwater. Environmental contamination of arsenic leads to leads to various types of cancers, coronary and neurological ailments in human. There are several sources of arsenic exposure such as drinking water, diet, wood preservatives, smoking, air and cosmetics, while, drinking water is the most explored route. Inorganic arsenic exhibits higher levels of toxicity compared its organic forms. Exposure to inorganic arsenic is known to cause major neurological effects such as cytotoxicity, chromosomal aberration, damage to cellular DNA and genotoxicity. On the other hand, long-term exposure to arsenic may cause neurobehavioral effects in the juvenile stage, which may have detrimental effects in the later stages of life. Thus, it is important to understand the toxicology and underlying molecular mechanism of arsenic which will help to mitigate its detrimental effects. The present review focuses on the epidemiology, and the toxic mechanisms responsible for arsenic induced neurobehavioral diseases, including strategies for its management from water, community and household premises. The review also provides a critical analysis of epigenetic and transgenerational modifications, mitochondrial oxidative stress, molecular mechanisms of arsenic-induced oxidative stress, and neuronal dysfunction.
Collapse
Affiliation(s)
- Manisha Thakur
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (M.R.); (S.N.)
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; (M.R.); (S.N.)
- Toxicology Centre, Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| | - Swaran Jeet Singh Flora
- Department of Pharmacology and Toxicology, Transit Campus, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India; (M.T.); (A.K.D.)
| |
Collapse
|
10
|
Kaur I, Behl T, Aleya L, Rahman MH, Kumar A, Arora S, Akter R. Role of metallic pollutants in neurodegeneration: effects of aluminum, lead, mercury, and arsenic in mediating brain impairment events and autism spectrum disorder. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:8989-9001. [PMID: 33447979 DOI: 10.1007/s11356-020-12255-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 12/27/2020] [Indexed: 04/16/2023]
Abstract
Autism spectrum disorder (ASD) is a developmental disorder of the brain characterized by shortfall in the social portfolio of an individual and abbreviated interactive and communication aspects rendering stereotypical behavior and pitfalls in a child's memory, thinking, and learning capabilities. The incidence of ASD has accelerated since the past decade, portraying environment as one of the primary assets, comprising of metallic components aiming to curb the neurodevelopmental pathways in an individual. Many regulations like Clean Air Act and critical steps taken by countries all over the globe, like Sweden and the USA, have rendered the necessity to study the effects of environmental metallic components on ASD progression. The review focuses on the primary metallic components present in the environment (aluminum, lead, mercury, and arsenic), responsible for accelerating ASD symptoms by a set of general mechanisms like oxidative stress reduction, glycolysis suppression, microglial activation, and metalloprotein disruption, resulting in apoptotic signaling, neurotoxic effects, and neuroinflammatory responses. The effect of these metals can be retarded by certain protective strategies like chelation, dietary correction, certain agents (curcumin, mangiferin, selenium), and detoxification enhancement, which can necessarily halt the neurodegenerative effects.
Collapse
Affiliation(s)
- Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Paris, France
| | - Md Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Seoul, South Korea
- Department of Pharmacy, Southeast University, Banani, Dhaka, Bangladesh
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Seoul, South Korea
| |
Collapse
|
11
|
Schepici G, Bramanti P, Mazzon E. Efficacy of Sulforaphane in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21228637. [PMID: 33207780 PMCID: PMC7698208 DOI: 10.3390/ijms21228637] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/10/2020] [Accepted: 11/14/2020] [Indexed: 12/14/2022] Open
Abstract
Sulforaphane (SFN) is a phytocompound belonging to the isothiocyanate family. Although it was also found in seeds and mature plants, SFN is mainly present in sprouts of many cruciferous vegetables, including cabbage, broccoli, cauliflower, and Brussels sprouts. SFN is produced by the conversion of glucoraphanin through the enzyme myrosinase, which leads to the formation of this isothiocyanate. SFN is especially characterized by antioxidant, anti-inflammatory, and anti-apoptotic properties, and for this reason, it aroused the interest of researchers. The aim of this review is to summarize the experimental studies present on Pubmed that report the efficacy of SFN in the treatment of neurodegenerative disease, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple sclerosis (MS). Therefore, thanks to its beneficial effects, SFN could be useful as a supplement to counteracting neurodegenerative diseases.
Collapse
|
12
|
Khodadadi H, Jahromi GP, Zaeinalifard G, Fasihi-Ramandi M, Esmaeili M, Shahriary A. Neuroprotective and Antiapoptotic Effects of Allopregnanolone and Curcumin on Arsenic-Induced Toxicity in SH-SY5Y Dopaminergic Human Neuroblastoma Cells. NEUROPHYSIOLOGY+ 2020. [DOI: 10.1007/s11062-020-09861-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
13
|
Arsenic Toxicity: Molecular Targets and Therapeutic Agents. Biomolecules 2020; 10:biom10020235. [PMID: 32033229 PMCID: PMC7072575 DOI: 10.3390/biom10020235] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
High arsenic (As) levels in food and drinking water, or under some occupational conditions, can precipitate chronic toxicity and in some cases cancer. Millions of people are exposed to unacceptable amounts of As through drinking water and food. Highly exposed individuals may develop acute, subacute, or chronic signs of poisoning, characterized by skin lesions, cardiovascular symptoms, and in some cases, multi-organ failure. Inorganic arsenite(III) and organic arsenicals with the general formula R-As2+ are bound tightly to thiol groups, particularly to vicinal dithiols such as dihydrolipoic acid (DHLA), which together with some seleno-enzymes constitute vulnerable targets for the toxic action of As. In addition, R-As2+-compounds have even higher affinity to selenol groups, e.g., in thioredoxin reductase that also possesses a thiol group vicinal to the selenol. Inhibition of this and other ROS scavenging seleno-enzymes explain the oxidative stress associated with arsenic poisoning. The development of chelating agents, such as the dithiols BAL (dimercaptopropanol), DMPS (dimercapto-propanesulfonate) and DMSA (dimercaptosuccinic acid), took advantage of the fact that As had high affinity towards vicinal dithiols. Primary prevention by reducing exposure of the millions of people exposed to unacceptable As levels should be the prioritized strategy. However, in acute and subacute and even some cases with chronic As poisonings chelation treatment with therapeutic dithiols, in particular DMPS appears promising as regards alleviation of symptoms. In acute cases, initial treatment with BAL combined with DMPS should be considered.
Collapse
|
14
|
Garza-Lombó C, Pappa A, Panayiotidis MI, Gonsebatt ME, Franco R. Arsenic-induced neurotoxicity: a mechanistic appraisal. J Biol Inorg Chem 2019; 24:1305-1316. [PMID: 31748979 DOI: 10.1007/s00775-019-01740-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022]
Abstract
Arsenic is a metalloid found in groundwater as a byproduct of soil/rock erosion and industrial and agricultural processes. This xenobiotic elicits its toxicity through different mechanisms, and it has been identified as a toxicant that affects virtually every organ or tissue in the body. In the central nervous system, exposure to arsenic can induce cognitive dysfunction. Furthermore, iAs has been linked to several neurological disorders, including neurodevelopmental alterations, and is considered a risk factor for neurodegenerative disorders. However, the exact mechanisms involved are still unclear. In this review, we aim to appraise the neurotoxic effects of arsenic and the molecular mechanisms involved. First, we discuss the epidemiological studies reporting on the effects of arsenic in intellectual and cognitive function during development as well as studies showing the correlation between arsenic exposure and altered cognition and mental health in adults. The neurotoxic effects of arsenic and the potential mechanisms associated with neurodegeneration are also reviewed including data from experimental models supporting epidemiological evidence of arsenic as a neurotoxicant. Next, we focused on recent literature regarding arsenic metabolism and the molecular mechanisms that begin to explain how arsenic damages the central nervous system including, oxidative stress, energy failure and mitochondrial dysfunction, epigenetics, alterations in neurotransmitter homeostasis and synaptic transmission, cell death pathways, and inflammation. Outlining the specific mechanisms by which arsenic alters the cell function is key to understand the neurotoxic effects that convey cognitive dysfunction, neurodevelopmental alterations, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.,Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - María E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA. .,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
15
|
Klomparens EA, Ding Y. The neuroprotective mechanisms and effects of sulforaphane. Brain Circ 2019; 5:74-83. [PMID: 31334360 PMCID: PMC6611193 DOI: 10.4103/bc.bc_7_19] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/12/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022] Open
Abstract
Sulforaphane (SFN) is a phytochemical found in cruciferous vegetables. It has been shown to have many protective effects against many diseases, including multiple types of cancer. SFN is a potent activator of the nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant response element (ARE) genetic pathway. Upregulation of Nrf2-ARE increases the availability of multiple antioxidants. A substantial amount of preclinical research regarding the ability of SFN to protect the nervous system from many diseases and toxins has been done, but only a few small human trials have been completed. Preclinical data suggest that SFN protects the nervous system through multiple mechanisms and may help reduce the risk of many diseases and reduce the burden of symptoms in existing conditions. This review focuses on the literature regarding the protective effects of SFN on the nervous system. A discussion of neuroprotective mechanisms is followed by a discussion of the protective effects elicited by SFN administration in a multitude of neurological diseases and toxin exposures. SFN is a promising neuroprotective phytochemical which needs further human trials to evaluate its efficacy in preventing and decreasing the burden of many neurological diseases.
Collapse
Affiliation(s)
- Eric A Klomparens
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
- John D. Dingell VA Medical Center, Detroit, MI, USA
| |
Collapse
|
16
|
Impedance Study of Dopamine Effects after Application on 2D and 3D Neuroblastoma Cell Cultures Developed on a 3D-Printed Well. CHEMOSENSORS 2019. [DOI: 10.3390/chemosensors7010006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this work, the assessment of the interactions of a bioactive substance applied to immobilized cells in either a two-dimensional (2D) or three-dimensional (3D) arrangement mimicking in vivo tissue conditions is presented. In particular, dopamine (DA) was selected as a stimulant for the implementation of an impedance analysis with a specific type of neural cells (murine neuroblastoma). The aim of this study was the extraction of calibration curves at various frequencies with different known dopamine concentrations for the description of the behavior of dopamine applied to 2D and 3D cell cultures. The results present the evaluation of the mean impedance value for each immobilization technique in each frequency. The differential responses showed the importance of the impedance when frequency is applied in both 2D and 3D immobilization cases. More specifically, in 2D immobilization matrix impedance shows higher values in comparison with the 3D cell culture. Additionally, in the 3D case, the impedance decreases with increasing concentration, while in the 2D case, an opposite behavior was observed.
Collapse
|
17
|
Bjørklund G, Skalny AV, Rahman MM, Dadar M, Yassa HA, Aaseth J, Chirumbolo S, Skalnaya MG, Tinkov AA. Toxic metal(loid)-based pollutants and their possible role in autism spectrum disorder. ENVIRONMENTAL RESEARCH 2018; 166:234-250. [PMID: 29902778 DOI: 10.1016/j.envres.2018.05.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 06/08/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social interaction, verbal and non-verbal communication, and stereotypic behaviors. Many studies support a significant relationship between many different environmental factors in ASD etiology. These factors include increased daily exposure to various toxic metal-based environmental pollutants, which represent a cause for concern in public health. This article reviews the most relevant toxic metals, commonly found, environmental pollutants, i.e., lead (Pb), mercury (Hg), aluminum (Al), and the metalloid arsenic (As). Additionally, it discusses how pollutants can be a possible pathogenetic cause of ASD through various mechanisms including neuroinflammation in different regions of the brain, fundamentally occurring through elevation of the proinflammatory profile of cytokines and aberrant expression of nuclear factor kappa B (NF-κB). Due to the worldwide increase in toxic environmental pollution, studies on the role of pollutants in neurodevelopmental disorders, including direct effects on the developing brain and the subjects' genetic susceptibility and polymorphism, are of utmost importance to achieve the best therapeutic approach and preventive strategies.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| | - Anatoly V Skalny
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; All-Russian Research Institute of Medicinal and Aromatic Plants, Moscow, Russia
| | - Md Mostafizur Rahman
- Department of Environmental Sciences, Jahangirnagar University, Dhaka, Bangladesh; Graduate School of Environmental Science, Hokkaido University, Japan
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Heba A Yassa
- Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Jan Aaseth
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, Elverum, Norway; Department of Research, Innlandet Hospital Trust, Brumunddal, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | | | - Alexey A Tinkov
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| |
Collapse
|
18
|
Almeida AS, Soares NL, Sequeira CO, Pereira SA, Sonnewald U, Vieira HLA. Improvement of neuronal differentiation by carbon monoxide: Role of pentose phosphate pathway. Redox Biol 2018; 17:338-347. [PMID: 29793167 PMCID: PMC6007049 DOI: 10.1016/j.redox.2018.05.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/24/2018] [Accepted: 05/10/2018] [Indexed: 12/13/2022] Open
Abstract
Over the last decades, the silent-killer carbon monoxide (CO) has been shown to also be an endogenous cytoprotective molecule able to inhibit cell death and modulate mitochondrial metabolism. Neuronal metabolism is mostly oxidative and neurons also use glucose for maintaining their anti-oxidant status by generation of reduced glutathione (GSH) via the pentose-phosphate pathway (PPP). It is established that neuronal differentiation depends on reactive oxygen species (ROS) generation and signalling, however there is a lack of information about modulation of the PPP during adult neurogenesis. Thus, the main goal of this study was to unravel the role of CO on cell metabolism during neuronal differentiation, particularly by targeting PPP flux and GSH levels as anti-oxidant system. A human neuroblastoma SH-S5Y5 cell line was used, which differentiates into post-mitotic neurons by treatment with retinoic acid (RA), supplemented or not with CO-releasing molecule-A1 (CORM-A1). SH-SY5Y cell differentiation supplemented with CORM-A1 prompted an increase in neuronal yield production. It did, however, not alter glycolytic metabolism, but increased the PPP. In fact, CORM-A1 treatment stimulated (i) mRNA expression of 6-phosphogluconate dehydrogenase (PGDH) and transketolase (TKT), which are enzymes for oxidative and non-oxidative phases of the PPP, respectively and (ii) protein expression and activity of glucose 6-phosphate dehydrogenase (G6PD) the rate-limiting enzyme of the PPP. Likewise, whenever G6PD was knocked-down CO-induced improvement on neuronal differentiation was reverted, while pharmacological inhibition of GSH synthesis did not change CO's effect on the improvement of neuronal differentiation. Both results indicate the key role of PPP in CO-modulation of neuronal differentiation. Furthermore, at the end of SH-SY5Y neuronal differentiation process, CORM-A1 supplementation increased the ratio of reduced and oxidized glutathione (GSH/GSSG) without alteration of GSH metabolism. These data corroborate with PPP stimulation. In conclusion, CO improves neuronal differentiation of SH-S5Y5 cells by stimulating the PPP and modulating the GSH system.
Collapse
Affiliation(s)
- Ana S Almeida
- CEDOC, Faculdade de Ciência Médicas/NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; Instituto de Tecnologia Química e Biológica (ITQB), Universidade Nova de Lisboa, Apartado 127, 2781-901 Oeiras, Portugal; Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal
| | - Nuno L Soares
- CEDOC, Faculdade de Ciência Médicas/NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Catarina O Sequeira
- CEDOC, Faculdade de Ciência Médicas/NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Sofia A Pereira
- CEDOC, Faculdade de Ciência Médicas/NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | | | - Helena L A Vieira
- CEDOC, Faculdade de Ciência Médicas/NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal.
| |
Collapse
|
19
|
Srivastava P, Dhuriya YK, Kumar V, Srivastava A, Gupta R, Shukla RK, Yadav RS, Dwivedi HN, Pant AB, Khanna VK. PI3K/Akt/GSK3β induced CREB activation ameliorates arsenic mediated alterations in NMDA receptors and associated signaling in rat hippocampus: Neuroprotective role of curcumin. Neurotoxicology 2018; 67:190-205. [PMID: 29723552 DOI: 10.1016/j.neuro.2018.04.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 04/26/2018] [Accepted: 04/29/2018] [Indexed: 12/20/2022]
Abstract
Protective efficacy of curcumin in arsenic induced NMDA receptor dysfunctions and PI3K/Akt/ GSK3β signalling in hippocampus has been investigated in vivo and in vitro. Exposure to sodium arsenite (in vivo - 20 mg/kg, body weight p.o. for 28 days; in vitro - 10 μM for 24 h) and curcumin (in vivo - 100 mg/kg body weight p.o. for 28 days; in vitro - 20 μM for 24 h) was carried out alone or simultaneously. Treatment with curcumin ameliorated sodium arsenite induced alterations in the levels of NMDA receptors, its receptor subunits and synaptic proteins - pCaMKIIα, PSD-95 and SynGAP both in vivo and in vitro. Decreased levels of BDNF, pAkt, pERK1/2, pGSK3β and pCREB on sodium arsenite exposure were also protected by curcumin. Curcumin was found to decrease sodium arsenite induced changes in hippocampus by modulating PI3K/Akt/GSK3β neuronal survival pathway, known to regulate various cellular events. Treatment of hippocampal cultures with pharmacological inhibitors for ERK1/2, GSK3β and Akt individually inhibited levels of CREB and proteins associated with PI3K/Akt/GSK3β pathway. Simultaneous treatment with curcumin was found to improve sodium arsenite induced learning and memory deficits in rats assessed by water maze and Y-maze. The results provide evidence that curcumin exercises its neuroprotective effect involving PI3K/Akt pathway which may affect NMDA receptors and downstream signalling through TrKβ and BDNF in arsenic induced cognitive deficits in hippocampus.
Collapse
Affiliation(s)
- Pranay Srivastava
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India; School of Pharmacy, Babu Banarsi Das University, Faizabad Road, Lucknow, 226 028, UP, India
| | - Yogesh K Dhuriya
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India
| | - Vivek Kumar
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, Brazil
| | - Akriti Srivastava
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India
| | - Richa Gupta
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India; School of Pharmacy, Babu Banarsi Das University, Faizabad Road, Lucknow, 226 028, UP, India
| | - Rajendra K Shukla
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India
| | - Rajesh S Yadav
- Department of Criminology and Forensic Science, Dr. Harisingh Gour Central University, Sagar, 470003, MP, India
| | - Hari N Dwivedi
- School of Pharmacy, Babu Banarsi Das University, Faizabad Road, Lucknow, 226 028, UP, India
| | - Aditya B Pant
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India.
| | - Vinay K Khanna
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India.
| |
Collapse
|
20
|
Stahl K, Rahmani S, Prydz A, Skauli N, MacAulay N, Mylonakou MN, Torp R, Skare Ø, Berg T, Leergaard TB, Paulsen RE, Ottersen OP, Amiry-Moghaddam M. Targeted deletion of the aquaglyceroporin AQP9 is protective in a mouse model of Parkinson's disease. PLoS One 2018; 13:e0194896. [PMID: 29566083 PMCID: PMC5864064 DOI: 10.1371/journal.pone.0194896] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
More than 90% of the cases of Parkinson’s disease have unknown etiology. Gradual loss of dopaminergic neurons of substantia nigra is the main cause of morbidity in this disease. External factors such as environmental toxins are believed to play a role in the cell loss, although the cause of the selective vulnerability of dopaminergic neurons remains unknown. We have previously shown that aquaglyceroporin AQP9 is expressed in dopaminergic neurons and astrocytes of rodent brain. AQP9 is permeable to a broad spectrum of substrates including purines, pyrimidines, and lactate, in addition to water and glycerol. Here we test our hypothesis that AQP9 serves as an influx route for exogenous toxins and, hence, may contribute to the selective vulnerability of nigral dopaminergic (tyrosine hydroxylase-positive) neurons. Using Xenopus oocytes injected with Aqp9 cRNA, we show that AQP9 is permeable to the parkinsonogenic toxin 1-methyl-4-phenylpyridinium (MPP+). Stable expression of AQP9 in HEK cells increases their vulnerability to MPP+ and to arsenite—another parkinsonogenic toxin. Conversely, targeted deletion of Aqp9 in mice protects nigral dopaminergic neurons against MPP+ toxicity. A protective effect of Aqp9 deletion was demonstrated in organotypic slice cultures of mouse midbrain exposed to MPP+in vitro and in mice subjected to intrastriatal injections of MPP+in vivo. Seven days after intrastriatal MPP+ injections, the population of tyrosine hydroxylase-positive cells in substantia nigra is reduced by 48% in Aqp9 knockout mice compared with 67% in WT littermates. Our results show that AQP9 –selectively expressed in catecholaminergic neurons—is permeable to MPP+ and suggest that this aquaglyceroporin contributes to the selective vulnerability of nigral dopaminergic neurons by providing an entry route for parkinsonogenic toxins. To our knowledge this is the first evidence implicating a toxin permeable membrane channel in the pathophysiology of Parkinson’s disease.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacokinetics
- Animals
- Aquaporins/genetics
- Disease Models, Animal
- Dopaminergic Neurons/drug effects
- Dopaminergic Neurons/metabolism
- Female
- Gene Deletion
- HEK293 Cells
- Humans
- MPTP Poisoning/genetics
- MPTP Poisoning/metabolism
- MPTP Poisoning/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mutagenesis, Site-Directed
- Neuroprotection/genetics
- Neuroprotective Agents/metabolism
- Parkinson Disease/genetics
- Parkinson Disease/pathology
- Parkinson Disease, Secondary/chemically induced
- Parkinson Disease, Secondary/genetics
- Parkinson Disease, Secondary/metabolism
- Parkinson Disease, Secondary/pathology
- Substantia Nigra/drug effects
- Substantia Nigra/metabolism
- Xenopus laevis
Collapse
Affiliation(s)
- Katja Stahl
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Soulmaz Rahmani
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Agnete Prydz
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nadia Skauli
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nanna MacAulay
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria N. Mylonakou
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, Norway Biotechnology Centre, University of Oslo, Oslo, Norway
| | - Reidun Torp
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Øivind Skare
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
| | - Torill Berg
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Trygve B. Leergaard
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ragnhild E. Paulsen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Ole P. Ottersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Karolinska Institutet, Stockholm, Sweden
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- * E-mail:
| |
Collapse
|
21
|
Wu Y, Jiang X, Yang K, Xia Y, Cheng S, Tang Q, Bai L, Qiu J, Chen C. Inhibition of α-Synuclein contributes to the ameliorative effects of dietary flavonoids luteolin on arsenite-induced apoptotic cell death in the dopaminergic PC12 cells. Toxicol Mech Methods 2017; 27:598-608. [DOI: 10.1080/15376516.2017.1339155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yi Wu
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Jiulongpo Municipal Center for Disease Control and Prevention, Chongqing, People’s Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Kai Yang
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Shuqun Cheng
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Qianghu Tang
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - LuLu Bai
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jingfu Qiu
- Department of Hygiene Inspection and Quarantine, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
22
|
Preciados M, Yoo C, Roy D. Estrogenic Endocrine Disrupting Chemicals Influencing NRF1 Regulated Gene Networks in the Development of Complex Human Brain Diseases. Int J Mol Sci 2016; 17:E2086. [PMID: 27983596 PMCID: PMC5187886 DOI: 10.3390/ijms17122086] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/21/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
During the development of an individual from a single cell to prenatal stages to adolescence to adulthood and through the complete life span, humans are exposed to countless environmental and stochastic factors, including estrogenic endocrine disrupting chemicals. Brain cells and neural circuits are likely to be influenced by estrogenic endocrine disruptors (EEDs) because they strongly dependent on estrogens. In this review, we discuss both environmental, epidemiological, and experimental evidence on brain health with exposure to oral contraceptives, hormonal therapy, and EEDs such as bisphenol-A (BPA), polychlorinated biphenyls (PCBs), phthalates, and metalloestrogens, such as, arsenic, cadmium, and manganese. Also we discuss the brain health effects associated from exposure to EEDs including the promotion of neurodegeneration, protection against neurodegeneration, and involvement in various neurological deficits; changes in rearing behavior, locomotion, anxiety, learning difficulties, memory issues, and neuronal abnormalities. The effects of EEDs on the brain are varied during the entire life span and far-reaching with many different mechanisms. To understand endocrine disrupting chemicals mechanisms, we use bioinformatics, molecular, and epidemiologic approaches. Through those approaches, we learn how the effects of EEDs on the brain go beyond known mechanism to disrupt the circulatory and neural estrogen function and estrogen-mediated signaling. Effects on EEDs-modified estrogen and nuclear respiratory factor 1 (NRF1) signaling genes with exposure to natural estrogen, pharmacological estrogen-ethinyl estradiol, PCBs, phthalates, BPA, and metalloestrogens are presented here. Bioinformatics analysis of gene-EEDs interactions and brain disease associations identified hundreds of genes that were altered by exposure to estrogen, phthalate, PCBs, BPA or metalloestrogens. Many genes modified by EEDs are common targets of both 17 β-estradiol (E2) and NRF1. Some of these genes are involved with brain diseases, such as Alzheimer's Disease (AD), Parkinson's Disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, Autism Spectrum Disorder, and Brain Neoplasms. For example, the search of enriched pathways showed that top ten E2 interacting genes in AD-APOE, APP, ATP5A1, CALM1, CASP3, GSK3B, IL1B, MAPT, PSEN2 and TNF-underlie the enrichment of the Kyoto Encyclopedia of Genes and Genomes (KEGG) AD pathway. With AD, the six E2-responsive genes are NRF1 target genes: APBB2, DPYSL2, EIF2S1, ENO1, MAPT, and PAXIP1. These genes are also responsive to the following EEDs: ethinyl estradiol (APBB2, DPYSL2, EIF2S1, ENO1, MAPT, and PAXIP1), BPA (APBB2, EIF2S1, ENO1, MAPT, and PAXIP1), dibutyl phthalate (DPYSL2, EIF2S1, and ENO1), diethylhexyl phthalate (DPYSL2 and MAPT). To validate findings from Comparative Toxicogenomics Database (CTD) curated data, we used Bayesian network (BN) analysis on microarray data of AD patients. We observed that both gender and NRF1 were associated with AD. The female NRF1 gene network is completely different from male human AD patients. AD-associated NRF1 target genes-APLP1, APP, GRIN1, GRIN2B, MAPT, PSEN2, PEN2, and IDE-are also regulated by E2. NRF1 regulates targets genes with diverse functions, including cell growth, apoptosis/autophagy, mitochondrial biogenesis, genomic instability, neurogenesis, neuroplasticity, synaptogenesis, and senescence. By activating or repressing the genes involved in cell proliferation, growth suppression, DNA damage/repair, apoptosis/autophagy, angiogenesis, estrogen signaling, neurogenesis, synaptogenesis, and senescence, and inducing a wide range of DNA damage, genomic instability and DNA methylation and transcriptional repression, NRF1 may act as a major regulator of EEDs-induced brain health deficits. In summary, estrogenic endocrine disrupting chemicals-modified genes in brain health deficits are part of both estrogen and NRF1 signaling pathways. Our findings suggest that in addition to estrogen signaling, EEDs influencing NRF1 regulated communities of genes across genomic and epigenomic multiple networks may contribute in the development of complex chronic human brain health disorders.
Collapse
Affiliation(s)
- Mark Preciados
- Department of Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA.
| | - Changwon Yoo
- Department of Biostatistics, Florida International University, Miami, FL 33199, USA.
| | - Deodutta Roy
- Department of Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
23
|
Coherent and Contradictory Facts, Feats and Fictions Associated with Metal Accumulation in Parkinson's Disease: Epicenter or Outcome, Yet a Demigod Question. Mol Neurobiol 2016; 54:4738-4755. [PMID: 27480264 DOI: 10.1007/s12035-016-0016-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/12/2016] [Indexed: 01/30/2023]
Abstract
Unwarranted exposure due to liberal use of metals for maintaining the lavish life and to achieve the food demand for escalating population along with an incredible boost in the average human life span owing to orchestrated progress in rejuvenation therapy have gradually increased the occurrence of Parkinson's disease (PD). Etiology is albeit elusive; association of PD with metal accumulation has never been overlooked due to noteworthy similitude between metal-exposure symptoms and a few cardinal features of disease. Even though metals are entailed in the vital functions, a hysterical shift, primarily augmentation, escorts the stern nigrostriatal dopaminergic neurodegeneration. An increase in the passage of metals through the blood brain barrier and impaired metabolic activity and elimination system could lead to metal accumulation in the brain, which eventually makes dopaminergic neurons quite susceptible. In the present article, an update on implication of metal accumulation in PD/Parkinsonism has been provided. Moreover, encouraging and paradoxical facts and fictions associated with metal accumulation in PD/Parkinsonism have also been compiled. Systematic literature survey of PD is performed to describe updated information if metal accumulation is an epicenter or merely an outcome. Finally, a perspective on the association of metal accumulation with pesticide-induced Parkinsonism has been explained to unveil the likely impact of the former in the latter.
Collapse
|
24
|
Cholanians AB, Phan AV, Ditzel EJ, Camenisch TD, Lau SS, Monks TJ. From the Cover: Arsenic Induces Accumulation of α-Synuclein: Implications for Synucleinopathies and Neurodegeneration. Toxicol Sci 2016; 153:271-81. [PMID: 27413109 DOI: 10.1093/toxsci/kfw117] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Synucleinopathies, including Parkinson's disease (PD), are neurodegenerative diseases characterized by accumulation of α-synuclein (SYN), a small neuronal protein with prion like properties that plays a central role in PD pathogenesis. SYN can misfold and generate toxic oligomers/aggregates, which can be cytotoxic. Environmental arsenic (As)-containing pesticide use correlates with increased incidence of PD. Moreover, because As exposure can lead to inhibition of autophagic flux we hypothesize that As can facilitate the accumulation of toxic SYN oligomers/aggregates and subsequent increases in markers of autophagy. We therefore examined the role of As in the oligomerization of SYN, and the consequences thereof. Chronic exposure of SH-SY5Y cells overexpressing SYN to As caused a dose-dependent oligomerization of SYN, with concomitant increases in protein ubiquitination and expression of other stress markers (protein glutathione binding, γ-GCS, light chain 3 (LC3)-I/II, P62, and NAD(P)H dehydrogenase quinone 1), indicative of an increased proteotoxic stress. Immunocytochemical analyses revealed an accumulation of SYN, and it's colocalization with LC3, a major autophagic protein. Mice exposed to As (100 ppb) for 1 month, exhibited elevated SYN accumulation in the cortex and striatum, and elevations in protein ubiquitination and LC3-I and II levels. However, tyrosine hydroxylase (TH), an indicator of dopaminergic cell density, was upregulated in the As exposed animals. Because SYN can inhibit TH function, and As can decrease monoamine levels, As exposure possibly leads to compensatory mechanisms leading to an increase in TH expression. Our findings suggest that susceptible individuals may be at higher risk of developing synucleinopathies and/or neurodegeneration due to environmental As exposure.
Collapse
Affiliation(s)
- Aram B Cholanians
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Arizona 85721
| | - Andy V Phan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Arizona 85721
| | - Eric J Ditzel
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Arizona 85721
| | - Todd D Camenisch
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Arizona 85721
| | - Serrine S Lau
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Arizona 85721
| | - Terrence J Monks
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Arizona 85721
| |
Collapse
|
25
|
Andrade VL, Mateus ML, Batoréu MC, Aschner M, Marreilha dos Santos AP. Lead, Arsenic, and Manganese Metal Mixture Exposures: Focus on Biomarkers of Effect. Biol Trace Elem Res 2015; 166:13-23. [PMID: 25693681 PMCID: PMC4470849 DOI: 10.1007/s12011-015-0267-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 02/04/2015] [Indexed: 10/24/2022]
Abstract
The increasing exposure of human populations to excessive levels of metals continues to represent a matter of public health concern. Several biomarkers have been studied and proposed for the detection of adverse health effects induced by lead (Pb), arsenic (As), and manganese (Mn); however, these studies have relied on exposures to each single metal, which fails to replicate real-life exposure scenarios. These three metals are commonly detected in different environmental, occupational, and food contexts and they share common neurotoxic effects, which are progressive and once clinically apparent may be irreversible. Thus, chronic exposure to low levels of a mixture of these metals may represent an additive risk of toxicity. Building upon their shared mechanisms of toxicity, such as oxidative stress, interference with neurotransmitters, and effects on the hematopoietic system, we address putative biomarkers, which may assist in assessing the onset of neurological diseases associated with exposure to this metal mixture.
Collapse
Affiliation(s)
- VL Andrade
- Instituto de Investigação do Medicamento, iMed.UL, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - ML Mateus
- Instituto de Investigação do Medicamento, iMed.UL, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - MC Batoréu
- Instituto de Investigação do Medicamento, iMed.UL, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - M Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 10461 NY, USA
| | - AP Marreilha dos Santos
- Instituto de Investigação do Medicamento, iMed.UL, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Corresponding author – , Tel – 351217946400, Fax - 351217946470
| |
Collapse
|
26
|
Gumilar F, Lencinas I, Bras C, Giannuzzi L, Minetti A. Locomotor activity and sensory – motor developmental alterations in rat offspring exposed to arsenic prenatally and via lactation. Neurotoxicol Teratol 2015; 49:1-9. [DOI: 10.1016/j.ntt.2015.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 02/12/2015] [Accepted: 02/18/2015] [Indexed: 12/18/2022]
|
27
|
Lead Intoxication Synergies of the Ethanol-Induced Toxic Responses in Neuronal Cells--PC12. Mol Neurobiol 2014; 52:1504-1520. [PMID: 25367877 DOI: 10.1007/s12035-014-8928-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 10/07/2014] [Indexed: 01/05/2023]
Abstract
Lead (Pb)-induced neurodegeneration and its link with widespread neurobehavioral changes are well documented. Experimental evidences suggest that ethanol could enhance the absorption of metals in the body, and alcohol consumption may increase the susceptibility to metal intoxication in the brain. However, the underlying mechanism of ethanol action in affecting metal toxicity in brain cells is poorly understood. Thus, an attempt was made to investigate the modulatory effect of ethanol on Pb intoxication in PC12 cells, a rat pheochromocytoma. Cells were co-exposed to biological safe doses of Pb (10 μM) and ethanol (200 mM), and data were compared to the response of cells which received independent exposure to these chemicals at similar doses. Ethanol (200 mM) exposure significantly aggravated the Pb-induced alterations in the end points associated with oxidative stress and apoptosis. The finding confirms the involvement of reactive oxygen species (ROS)-mediated oxidative stress, and impairment of mitochondrial membrane potential, which subsequently facilitate the translocation of triggering proteins between cytoplasm and mitochondria. We further confirmed the apoptotic changes due to induction of mitochondria-mediated caspase cascade. These cellular changes were found to recover significantly, if the cells are exposed to N-acetyl cysteine (NAC), a known antioxidant. Our data suggest that ethanol may potentiate Pb-induced cellular damage in brain cells, but such damaging effects could be recovered by inhibition of ROS generation. These results open up further possibilities for the design of new therapeutics based on antioxidants to prevent neurodegeneration and associated health problems.
Collapse
|
28
|
Vasconcellos AP, Colello S, Kyle ME, Shin JJ. Societal-level Risk Factors Associated with Pediatric Hearing Loss: A Systematic Review. Otolaryngol Head Neck Surg 2014; 151:29-41. [PMID: 24671458 DOI: 10.1177/0194599814526561] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/12/2014] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To determine if the current body of evidence describes specific threshold values of concern for modifiable societal-level risk factors for pediatric hearing loss, with the overarching goal of providing actionable guidance for the prevention and screening of audiological deficits in children. DATA SOURCES Three related systematic reviews were performed. Computerized PubMed, Embase, and Cochrane Library searches were performed from inception through October 2013 and were supplemented with manual searches. REVIEW METHODS Inclusion/exclusion criteria were designed to determine specific threshold values of societal-level risk factors on hearing loss in the pediatric population. Searches and data extraction were performed by independent reviewers. RESULTS There were 20 criterion-meeting studies with 29,128 participants. Infants less than 2 standard deviations below standardized weight, length, or body mass index were at increased risk. Specific nutritional deficiencies related to iodine and thiamine may also increase risk, although data are limited and threshold values of concern have not been quantified. Blood lead levels above 10 µg/dL were significantly associated with pediatric sensorineural loss, and mixed findings were noted for other heavy metals. Hearing loss was also more prevalent among children of socioeconomically disadvantaged families, as measured by a poverty income ratio less than 0.3 to 1, higher deprivation category status, and head of household employment as a manual laborer. CONCLUSIONS Increasing our understanding of specific thresholds of risk associated with causative factors forms the foundation for preventive and targeted screening programs as well as future research endeavors.
Collapse
|
29
|
Barbosa DJ, Capela JP, Silva R, Vilas-Boas V, Ferreira LM, Branco PS, Fernandes E, Bastos MDL, Carvalho F. The mixture of "ecstasy" and its metabolites is toxic to human SH-SY5Y differentiated cells at in vivo relevant concentrations. Arch Toxicol 2014; 88:455-473. [PMID: 24101030 DOI: 10.1007/s00204-013-1120-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 08/22/2013] [Indexed: 12/21/2022]
Abstract
The neurotoxicity of "ecstasy" (3,4-methylenedioxymethamphetamine, MDMA) is thought to involve hepatic metabolism, though its real contribution is not completely understood. Most in vitro neurotoxicity studies concern isolated exposures of MDMA or its metabolites, at high concentrations, not considering their mixture, as expected in vivo. Therefore, our postulate is that combined deleterious effects of MDMA and its metabolites, at low micromolar concentrations that may be attained into the brain, may elicit neurotoxicity. Using human SH-SY5Y differentiated cells as dopaminergic neuronal model, we studied the neurotoxicity of MDMA and its MDMA metabolites α-methyldopamine and N-methyl-α-methyldopamine and their correspondent glutathione and N-acetylcysteine monoconjugates, under isolated exposure and as a mixture, at normothermic or hyperthermic conditions. The results showed that the mixture of MDMA and its metabolites was toxic to SH-SY5Y differentiated cells, an effect potentiated by hyperthermia and prevented by N-acetylcysteine. As a mixture, MDMA and its metabolites presented a different toxicity profile, compared to each compound alone, even at equimolar concentrations. Caspase 3 activation, increased reactive oxygen species production, and intracellular Ca(2+) raises were implicated in the toxic effect. The mixture increased intracellular glutathione levels by increasing its de novo synthesis. In conclusion, this study demonstrated, for the first time, that the mixture of MDMA and its metabolites, at low micromolar concentrations, which represents a more realistic approach of the in vivo scenario, elicited toxicity to human SH-SY5Y differentiated cells, thus constituting a new insight into the context of MDMA-related neurotoxicity.
Collapse
Affiliation(s)
- Daniel José Barbosa
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Laboratory, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal,
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chen ML, Lin CH, Lee MJ, Wu RM. BST1 rs11724635 interacts with environmental factors to increase the risk of Parkinson's disease in a Taiwanese population. Parkinsonism Relat Disord 2013; 20:280-3. [PMID: 24342025 DOI: 10.1016/j.parkreldis.2013.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 11/11/2013] [Accepted: 11/12/2013] [Indexed: 10/26/2022]
Abstract
A recently published genome-wide association study in Caucasian and Asian populations showed a significant association between the bone marrow stromal cell antigen 1 (BST1) SNP rs11724635 and increased risk for Parkinson's disease (PD). To investigate whether BST1 rs11724635 increases the risk of PD, either by itself or in combination with environmental factors, we performed an association analysis of BST1 rs11724635 in a large cohort of Taiwanese patients with PD and age matched controls. The study used TaqMan genotyping, logistic regression, and haplotype methods. The genotype distribution of rs11724635 in PD patients (N = 468; p = 0.50) and control subjects (N = 487; p = 0.44) was consistent with Hardy-Weinberg equilibrium. Compared with the AA genotype, the frequency of both CA and CC genotypes was not significantly different between the patient and control groups. The adjusted odd ratios (ORs) for CA and CC were not statistically significant (CA: OR = 0.962, 95% CI = 0.643-1.439, p = 0.850; CC: OR = 0.992, 95% CI = 0.654-1.503, p = 0.969). Of note, ever use of well water before the onset of PD symptoms had an impact on the occurrence of PD through interactions with BST1 rs11724635 AC (OR = 1.453, p = 0.024) and CC (OR = 1.623, p = 0.008). Our results show that the BST1 rs11724635 polymorphism alone is not associated with the development of PD, but it can interact with well water drinking to increase the risk of PD in this Taiwanese population.
Collapse
Affiliation(s)
- Meng-Ling Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan; Institute of Zoology, National Taiwan University, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ruey-Meei Wu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
31
|
Ferreira PS, Nogueira TB, Costa VM, Branco PS, Ferreira LM, Fernandes E, Bastos ML, Meisel A, Carvalho F, Capela JP. Neurotoxicity of "ecstasy" and its metabolites in human dopaminergic differentiated SH-SY5Y cells. Toxicol Lett 2013; 216:159-170. [PMID: 23194825 DOI: 10.1016/j.toxlet.2012.11.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Revised: 11/17/2012] [Accepted: 11/19/2012] [Indexed: 11/16/2022]
Abstract
"Ecstasy" (3,4-methylenedioxymethamphetamine or MDMA) is a widely abused recreational drug, reported to produce neurotoxic effects, both in laboratory animals and in humans. MDMA metabolites can be major contributors for MDMA neurotoxicity. This work studied the neurotoxicity of MDMA and its catechol metabolites, α-methyldopamine (α-MeDA) and N-methyl-α-methyldopamine (N-Me-α-MeDA) in human dopaminergic SH-SY5Y cells differentiated with retinoic acid and 12-O-tetradecanoyl-phorbol-13-acetate. Differentiation led to SH-SY5Y neurons with higher ability to accumulate dopamine and higher resistance towards dopamine neurotoxicity. MDMA catechol metabolites were neurotoxic to SH-SY5Y neurons, leading to caspase 3-independent cell death in a concentration- and time-dependent manner. MDMA did not show a concentration- and time-dependent death. Pre-treatment with the antioxidant and glutathione precursor, N-acetylcysteine (NAC), resulted in strong protection against the MDMA metabolites' neurotoxicity. Neither the superoxide radical scavenger, tiron, nor the inhibitor of the dopamine (DA) transporter, GBR 12909, prevented the metabolites' toxicity. Cells exposed to α-MeDA showed an increase in intracellular glutathione (GSH) levels, which, at the 48 h time-point, was not dependent in the activity increase of γ-glutamylcysteine synthetase (γ-GCS), revealing a possible transient effect. Importantly, pre-treatment with buthionine sulfoximine (BSO), an inhibitor of γ-GCS, prevented α-MeDA induced increase in GSH levels, but did not augment this metabolite cytotoxicity. Even so, BSO pre-treatment abolished NAC protective effects against α-MeDA neurotoxicity, which were, at least partially, due to GSH de novo synthesis. Inversely, pre-treatment of cells with BSO augmented N-Me-α-MeDA-induced neurotoxicity, but only slightly affected NAC neuroprotection. In conclusion, MDMA catechol metabolites promote differential toxic effects to differentiated dopaminergic human SH-SY5Y cells.
Collapse
Affiliation(s)
- Patrícia Silva Ferreira
- REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Aung KH, Kurihara R, Nakashima S, Maekawa F, Nohara K, Kobayashi T, Tsukahara S. Inhibition of neurite outgrowth and alteration of cytoskeletal gene expression by sodium arsenite. Neurotoxicology 2013; 34:226-35. [DOI: 10.1016/j.neuro.2012.09.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 09/07/2012] [Accepted: 09/17/2012] [Indexed: 10/27/2022]
|
33
|
Z-Ligustilide Potentiates the Cytotoxicity of Dopamine in Rat Dopaminergic PC12 Cells. Neurotox Res 2012; 22:345-54. [DOI: 10.1007/s12640-012-9319-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 03/12/2012] [Accepted: 03/14/2012] [Indexed: 01/01/2023]
|
34
|
Fimognari C, Turrini E, Ferruzzi L, Lenzi M, Hrelia P. Natural isothiocyanates: genotoxic potential versus chemoprevention. Mutat Res 2011; 750:107-131. [PMID: 22178957 DOI: 10.1016/j.mrrev.2011.12.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 12/01/2011] [Accepted: 12/02/2011] [Indexed: 12/12/2022]
Abstract
Isothiocyanates, occurring in many dietary cruciferous vegetables, show interesting chemopreventive activities against several chronic-degenerative diseases, including cancer, cardiovascular diseases, neurodegeneration, diabetes. The electrophilic carbon residue in the isothiocyanate moiety reacts with biological nucleophiles and modification of proteins is recognized as a key mechanism underlying the biological activity of isothiocyanates. The nuclear factor-erythroid-2-related factor 2 system, which orchestrates the expression of a wide array of antioxidant genes, plays a role in the protective effect of isothiocyanates against almost all the pathological conditions reported above. Recent emerging findings suggest a further common mechanism. Chronic inflammation plays a central role in many human diseases and isothiocyanates inhibit the activity of many inflammation components, suppress cyclooxygenase 2, and irreversibly inactivate the macrophage migration inhibitory factor. Due to their electrophilic reactivity, some isothiocyanates are able to form adducts with DNA and induce gene mutations and chromosomal aberrations. DNA damage has been demonstrated to be involved in the pathogenesis of various chronic-degenerative diseases of epidemiological relevance. Thus, the genotoxicity of the isothiocyanates should be carefully considered. In addition, the dose-response relationship for genotoxic compounds does not suggest evidence of a threshold. Thus, chemicals that are genotoxic pose a greater potential risk to humans than non-genotoxic compounds. Dietary consumption levels of isothiocyanates appear to be several orders of magnitude lower than the doses used in the genotoxicity studies and thus it is highly unlikely that such toxicities would occur in humans. However, the beneficial properties of isothiocyanates stimulated an increase of dietary supplements and functional foods with highly enriched isothiocyanate concentrations on the market. Whether such concentrations may exert a potential health risk cannot be excluded with certainty and an accurate evaluation of the toxicological profile of isothiocyanates should be prompted before any major increase in their consumption be recommended or their clinical use suggested.
Collapse
Affiliation(s)
- Carmela Fimognari
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Eleonora Turrini
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Lorenzo Ferruzzi
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Monia Lenzi
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
35
|
Posser T, de Paula MT, Franco JL, Leal RB, da Rocha JBT. Diphenyl diselenide induces apoptotic cell death and modulates ERK1/2 phosphorylation in human neuroblastoma SH-SY5Y cells. Arch Toxicol 2010; 85:645-51. [PMID: 20924558 DOI: 10.1007/s00204-010-0602-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 09/22/2010] [Indexed: 01/06/2023]
Abstract
Diphenyl diselenide (PhSe)(2) is a synthetic organoselenium compound displaying glutathione peroxidase-like activity. Protective and antioxidant potential of (PhSe)(2) have been extensively investigated in in vivo and in vitro studies. In spite of this, there is a lack of studies addressed to the investigation of potential cytotoxic effect and signaling pathways modulated by this compound. Herein, we aimed to analyze the effects of 24-h treatment with (PhSe)(2) on cell viability and a possible modulation of signaling pathways in human neuroblastoma cell line SH-SY5Y. For this purpose, cells were incubated with (PhSe)(2) (0.3-30 μM) for 24 h and cell viability, apoptotic cell death and modulation of MAPKs (ERK1/2 and p38(MAPK)), and PKC substrates phosphorylation was determined. (PhSe)(2) treatment significantly decreased cell viability and increased the number of apoptotic cells with induction of PARP cleavage. An increase in ERK1/2 phosphorylation was observed at (PhSe)(2) 3 μM. In contrast, higher concentrations of the chalcogenide inhibited ERK1/2, p38(MAPK) and PKC substrate phosphorylation. Pre-treatment with ERK1/2 inhibitor, U0126, increased cell susceptibility to (PhSe)(2). Together, these data indicate a cytotoxic potential of (PhSe)(2) in a neuronal cell line, which appears to be mediated by the ERK1/2 pathway.
Collapse
Affiliation(s)
- Thaís Posser
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, 97105-900, Brazil.
| | | | | | | | | |
Collapse
|
36
|
Rodríguez VM, Limón-Pacheco JH, Carrizales L, Mendoza-Trejo MS, Giordano M. Chronic exposure to low levels of inorganic arsenic causes alterations in locomotor activity and in the expression of dopaminergic and antioxidant systems in the albino rat. Neurotoxicol Teratol 2010; 32:640-7. [PMID: 20699118 DOI: 10.1016/j.ntt.2010.07.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 07/30/2010] [Accepted: 07/30/2010] [Indexed: 12/21/2022]
Abstract
Several studies have associated chronic arsenicism with decreases in IQ and sensory and motor alterations in humans. Likewise, studies of rodents exposed to inorganic arsenic ((i)As) have found changes in locomotor activity, brain neurochemistry, behavioral tasks, oxidative stress, and in sensory and motor nerves. In the current study, male Sprague-Dawley rats were exposed to environmentally relevant doses of (i)As (0.05, 0.5 mg (i)As/L) and to a high dose (50 mg (i)As/L) in drinking water for one year. Hypoactivity and increases in the striatal dopamine content were found in the group treated with 50 mg (i)As/L. Exposure to 0.5 and 50 mg (i)As/L increased the total brain content of As. Furthermore, (i)As exposure produced a dose-dependent up-regulation of mRNA for Mn-SOD and Trx-1 and a down-regulation of DAR-D₂ mRNA levels in the nucleus accumbens. DAR-D₁ and Nrf2 mRNA expression were down-regulated in nucleus accumbens in the group exposed to 50 mg (i)As/L. Trx-1 mRNA levels were up-regulated in the cortex in an (i)As dose-dependent manner, while DAR-D₁ mRNA expression was increased in striatum in the 0.5 mg (i)As/L group. These results show that chronic exposure to low levels of arsenic causes subtle but region-specific changes in the nervous system, especially in antioxidant systems and dopaminergic elements. These changes became behaviorally evident only in the group exposed to 50 mg (i)As/L.
Collapse
Affiliation(s)
- Verónica Mireya Rodríguez
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro 76230, México.
| | | | | | | | | |
Collapse
|
37
|
Stack C, Ho D, Wille E, Calingasan NY, Williams C, Liby K, Sporn M, Dumont M, Beal MF. Triterpenoids CDDO-ethyl amide and CDDO-trifluoroethyl amide improve the behavioral phenotype and brain pathology in a transgenic mouse model of Huntington's disease. Free Radic Biol Med 2010; 49:147-58. [PMID: 20338236 PMCID: PMC2916021 DOI: 10.1016/j.freeradbiomed.2010.03.017] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 03/12/2010] [Accepted: 03/16/2010] [Indexed: 12/20/2022]
Abstract
Oxidative stress is a prominent feature of Huntington's disease (HD) due to mitochondrial dysfunction and the ensuing overproduction of reactive oxygen species (ROS). This phenomenon ultimately contributes to cognitive and motor impairment, as well as brain pathology, especially in the striatum. Targeting the transcription of the endogenous antioxidant machinery could be a promising therapeutic approach. The NF-E2-related factor-2 (Nrf2)/antioxidant response element (ARE) signaling pathway is an important pathway involved in antioxidant and anti-inflammatory responses. Synthetic triterpenoids, which are derived from 2-Cyano-3,12-Dioxooleana-1,9-Dien-28-Oic acid (CDDO) activate the Nrf2/ARE pathway and reduce oxidative stress in animal models of neurodegenerative diseases. We investigated the effects of CDDO-ethyl amide (CDDO-EA) and CDDO-trifluoroethyl amide (CDDO-TFEA) in N171-82Q mice, a transgenic mouse model of HD. CDDO-EA or CDDO-TFEA were administered in the diet at various concentrations, starting at 30days of age. CDDO-EA and CDDO-TFEA upregulated Nrf2/ARE induced genes in the brain and peripheral tissues, reduced oxidative stress, improved motor impairment and increased longevity. They also rescued striatal atrophy in the brain and vacuolation in the brown adipose tissue. Therefore compounds targeting the Nrf2/ARE pathway show great promise for the treatment of HD.
Collapse
Affiliation(s)
- Cliona Stack
- Department of Neurology and Neuroscience, Weill Cornell Medical College, 525 East 68 Street, New York, New York, 10065, USA
| | - Daniel Ho
- Department of Neurology and Neuroscience, Weill Cornell Medical College, 525 East 68 Street, New York, New York, 10065, USA
| | - Elizabeth Wille
- Department of Neurology and Neuroscience, Weill Cornell Medical College, 525 East 68 Street, New York, New York, 10065, USA
| | - Noel Y. Calingasan
- Department of Neurology and Neuroscience, Weill Cornell Medical College, 525 East 68 Street, New York, New York, 10065, USA
| | - Charlotte Williams
- Department of Pharmacology and Toxicology, Dartmouth Medical School, 7650 Ransem, Hanover, New Hampshire, 03755, USA
| | - Karen Liby
- Department of Pharmacology and Toxicology, Dartmouth Medical School, 7650 Ransem, Hanover, New Hampshire, 03755, USA
| | - Michael Sporn
- Department of Pharmacology and Toxicology, Dartmouth Medical School, 7650 Ransem, Hanover, New Hampshire, 03755, USA
| | - Magali Dumont
- Department of Neurology and Neuroscience, Weill Cornell Medical College, 525 East 68 Street, New York, New York, 10065, USA
| | - M. Flint Beal
- Department of Neurology and Neuroscience, Weill Cornell Medical College, 525 East 68 Street, New York, New York, 10065, USA
| |
Collapse
|
38
|
Aquaglyceroporin 9 in brain pathologies. Neuroscience 2010; 168:1047-57. [DOI: 10.1016/j.neuroscience.2009.10.030] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 10/02/2009] [Accepted: 10/13/2009] [Indexed: 12/21/2022]
|
39
|
Yadav S, Shi Y, Wang F, Wang H. Arsenite induces apoptosis in human mesenchymal stem cells by altering Bcl-2 family proteins and by activating intrinsic pathway. Toxicol Appl Pharmacol 2010; 244:263-72. [PMID: 20083129 DOI: 10.1016/j.taap.2010.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 01/04/2010] [Accepted: 01/05/2010] [Indexed: 01/23/2023]
Abstract
PURPOSE Environmental exposure to arsenic is an important public health issue. The effects of arsenic on different tissues and organs have been intensively studied. However, the effects of arsenic on bone marrow mesenchymal stem cells (MSCs) have not been reported. This study is designed to investigate the cell death process caused by arsenite and its related underlying mechanisms on MSCs. The rationale is that absorbed arsenic in the blood circulation can reach to the bone marrow and may affect the cell survival of MSCs. METHODS MSCs of passage 1 were purchased from Tulane University, grown till 70% confluency level and plated according to the experimental requirements followed by treatment with arsenite at various concentrations and time points. Arsenite (iAs(III)) induced cytotoxic effects were confirmed by cell viability and cell cycle analysis. For the presence of canonic apoptosis markers; DNA damage, exposure of intramembrane phosphotidylserine, protein and m-RNA expression levels were analyzed. RESULTS iAs(III) induced growth inhibition, G2-M arrest and apoptotic cell death in MSCs, the apoptosis induced by iAs(III) in the cultured MSCs was, via altering Bcl-2 family proteins and by involving intrinsic pathway. CONCLUSION iAs(III) can induce apoptosis in bone marrow-derived MSCs via Bcl-2 family proteins, regulating intrinsic apoptotic pathway. Due to the multipotency of MSC, acting as progenitor cells for a variety of connective tissues including bone, adipose, cartilage and muscle, these effects of arsenic may be important in assessing the health risk of the arsenic compounds and understanding the mechanisms of arsenic-induced harmful effects.
Collapse
Affiliation(s)
- Santosh Yadav
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|
40
|
Mylonakou MN, Petersen PH, Rinvik E, Rojek A, Valdimarsdottir E, Zelenin S, Zeuthen T, Nielsen S, Ottersen OP, Amiry-Moghaddam M. Analysis of mice with targeted deletion of AQP9 gene provides conclusive evidence for expression of AQP9 in neurons. J Neurosci Res 2009; 87:1310-22. [PMID: 19115411 DOI: 10.1002/jnr.21952] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AQP9 is an aquaglyceroporin that serves important functions in peripheral organs, including the liver. Reflecting the lack of AQP9 knockout mice, uncertainties still prevail regarding the localization and roles of AQP9 in the central nervous system. Here we present a comprehensive analysis of AQP9 gene expression in brain, based on a quantitative and multipronged approach that includes the use of animals with targeted deletion of the AQP9 gene. We show by real-time PCR that AQP9 mRNA concentration in rat and mouse brain is approximately 3% and approximately 0.5%, respectively, of that in rat and mouse liver, the organ with the highest level of AQP9. By blue native gel analysis it could be demonstrated that the brain contains tetrameric AQP9, corresponding to the functional form of AQP9. The band corresponding to the AQP9 tetramer was absent in AQP9 knockout brain and liver. Immunocytochemistry and in situ hybridization analyses with AQP9 knockout controls show that subpopulations of nigral neurons express AQP9 both at the mRNA and at the protein levels and that populations of cortical cells (including hilar neurons in the hippocampus) contain AQP9 mRNA but no detectable AQP9 immunosignal. The present data provide conclusive evidence for the presence of tetrameric AQP9 in brain and for the expression of AQP9 in neurons.
Collapse
Affiliation(s)
- Maria N Mylonakou
- Centre for Molecular Biology and Neuroscience, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
c-Jun N-terminal kinase mediates lactacystin-induced dopamine neuron degeneration. J Neuropathol Exp Neurol 2008; 67:933-44. [PMID: 18800014 DOI: 10.1097/nen.0b013e318186de64] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Parkinson disease is characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta. It has been proposed that dysfunction of the ubiquitin proteasome system plays an important role in the pathogenesis of Parkinson disease, but the mechanisms underlying ubiquitin proteasome system-related neuron degeneration are unknown. Here, we demonstrate that the proteasome inhibitor lactacystin induces phosphorylation of c-Jun N-terminal kinase (JNK) and c-Jun, the release of cytochrome c, activation of both caspase-9 and caspase-3, and sequential apoptosis of dopaminergic neurons in vitro. Most of these effects can be attenuated by the JNK inhibitor SP600125. Furthermore, infusion of lactacystin in rats in vivo also leads to phosphorylation of JNK before nigral neuron loss; chronic administration of SP600125 also blocks this loss. These results indicate that JNK is involved in proteasome inhibition-induced dopaminergic neuron degeneration through caspase-3-mediated apoptotic pathways, suggesting that this kinase may be a therapeutic target for the prevention of substantia nigra pars compacta degeneration in Parkinson disease patients.
Collapse
|