1
|
Anitha M, Kumar SM, Koo I, Perdew GH, Srinivasan S, Patterson AD. Modulation of Ceramide-Induced Apoptosis in Enteric Neurons by Aryl Hydrocarbon Receptor Signaling: Unveiling a New Pathway beyond ER Stress. Int J Mol Sci 2024; 25:8581. [PMID: 39201268 PMCID: PMC11354200 DOI: 10.3390/ijms25168581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a persistent organic pollutant and a potent aryl hydrocarbon receptor (AHR) ligand, causes delayed intestinal motility and affects the survival of enteric neurons. In this study, we investigated the specific signaling pathways and molecular targets involved in TCDD-induced enteric neurotoxicity. Immortalized fetal enteric neuronal (IM-FEN) cells treated with 10 nM TCDD exhibited cytotoxicity and caspase 3/7 activation, indicating apoptosis. Increased cleaved caspase-3 expression with TCDD treatment, as assessed by immunostaining in enteric neuronal cells isolated from WT mice but not in neural crest cell-specific Ahr deletion mutant mice (Wnt1Cre+/-/Ahrb(fl/fl)), emphasized the pivotal role of AHR in this process. Importantly, the apoptosis in IM-FEN cells treated with TCDD was mediated through a ceramide-dependent pathway, independent of endoplasmic reticulum stress, as evidenced by increased ceramide synthesis and the reversal of cytotoxic effects with myriocin, a potent inhibitor of ceramide biosynthesis. We identified Sptlc2 and Smpd2 as potential gene targets of AHR in ceramide regulation by a chromatin immunoprecipitation (ChIP) assay in IM-FEN cells. Additionally, TCDD downregulated phosphorylated Akt and phosphorylated Ser9-GSK-3β levels, implicating the PI3 kinase/AKT pathway in TCDD-induced neurotoxicity. Overall, this study provides important insights into the mechanisms underlying TCDD-induced enteric neurotoxicity and identifies potential targets for the development of therapeutic interventions.
Collapse
Affiliation(s)
- Mallappa Anitha
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (M.A.); (I.K.); (G.H.P.)
| | - Supriya M. Kumar
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (M.A.); (I.K.); (G.H.P.)
| | - Imhoi Koo
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (M.A.); (I.K.); (G.H.P.)
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (M.A.); (I.K.); (G.H.P.)
| | - Shanthi Srinivasan
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Atlanta VA Medical Center, Decatur, GA 30033, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (M.A.); (I.K.); (G.H.P.)
| |
Collapse
|
2
|
Murillo-González FE, García-Aguilar R, Limón-Pacheco J, Cabañas-Cortés MA, Elizondo G. 2,3,7,8-Tetrachlorodibenzo-p-dioxin and kynurenine induce Parkin expression in neuroblastoma cells through different signaling pathways mediated by the aryl hydrocarbon receptor. Toxicol Lett 2024; 394:114-127. [PMID: 38437907 DOI: 10.1016/j.toxlet.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
Parkin regulates protein degradation and mitophagy in dopaminergic neurons. Deficiencies in Parkin expression or function lead to cellular stress, cell degeneration, and the death of dopaminergic neurons, which promotes Parkinson's disease. In contrast, Parkin overexpression promotes neuronal survival. Therefore, the mechanisms of Parkin upregulation are crucial to understand. We describe here the molecular mechanism of AHR-mediated Parkin regulation in human SH-SY5Y neuroblastoma cells. Specifically, we report that the human Parkin gene (PRKN) is transcriptionally upregulated by the aryl hydrocarbon receptor (AHR) through two different selective ligand-dependent pathways. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a stress-inducing AHR ligand, indirectly promotes PRKN transcription by inducing ATF4 expression via TCDD-mediated endoplasmic reticulum (ER) stress. In contrast, kynurenine, a nontoxic AHR agonist, induces PRKN transcription by promoting AHR binding to the PRKN promoter without activating ER stress. Our results demonstrate that AHR activation may be a potential pharmacological pathway to induce human Parkin, but such a strategy must carefully consider the choice of AHR ligand to avoid neurotoxic side effects.
Collapse
Affiliation(s)
| | - Rosario García-Aguilar
- Departamento de Toxicología, CINVESTAV-IPN, Av. IPN 2508, Ciudad de México C.P. 07360, Mexico
| | - Jorge Limón-Pacheco
- Departamento de Biología Celular, CINVESTAV-IPN, Av. IPN 2508, Ciudad de México C.P. 07360, Mexico
| | | | - Guillermo Elizondo
- Departamento de Biología Celular, CINVESTAV-IPN, Av. IPN 2508, Ciudad de México C.P. 07360, Mexico.
| |
Collapse
|
3
|
Silva-Parra J, Sandu C, Felder-Schmittbuhl MP, Hernández-Kelly LC, Ortega A. Aryl Hydrocarbon Receptor in Glia Cells: A Plausible Glutamatergic Neurotransmission Orchestrator. Neurotox Res 2023; 41:103-117. [PMID: 36607593 DOI: 10.1007/s12640-022-00623-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/23/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023]
Abstract
Glutamate is the major excitatory amino acid in the vertebrate brain. Glutamatergic signaling is involved in most of the central nervous system functions. Its main components, namely receptors, ion channels, and transporters, are tightly regulated at the transcriptional, translational, and post-translational levels through a diverse array of extracellular signals, such as food, light, and neuroactive molecules. An exquisite and well-coordinated glial/neuronal bidirectional communication is required for proper excitatory amino acid signal transactions. Biochemical shuttles such as the glutamate/glutamine and the astrocyte-neuronal lactate represent the fundamental involvement of glial cells in glutamatergic transmission. In fact, the disruption of any of these coordinated biochemical intercellular cascades leads to an excitotoxic insult that underlies some aspects of most of the neurodegenerative diseases characterized thus far. In this contribution, we provide a comprehensive summary of the involvement of the Aryl hydrocarbon receptor, a ligand-dependent transcription factor in the gene expression regulation of glial glutamate transporters. These receptors might serve as potential targets for the development of novel strategies for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Janisse Silva-Parra
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Cristina Sandu
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México.
| |
Collapse
|
4
|
From Nucleus to Organs: Insights of Aryl Hydrocarbon Receptor Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms232314919. [PMID: 36499247 PMCID: PMC9738205 DOI: 10.3390/ijms232314919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a markedly established regulator of a plethora of cellular and molecular processes. Its initial role in the detoxification of xenobiotic compounds has been partially overshadowed by its involvement in homeostatic and organ physiology processes. In fact, the discovery of its ability to bind specific target regulatory sequences has allowed for the understanding of how AHR modulates such processes. Thereby, AHR presents functions in transcriptional regulation, chromatin architecture modifications and participation in different key signaling pathways. Interestingly, such fields of influence end up affecting organ and tissue homeostasis, including regenerative response both to endogenous and exogenous stimuli. Therefore, from classical spheres such as canonical transcriptional regulation in embryonic development, cell migration, differentiation or tumor progression to modern approaches in epigenetics, senescence, immune system or microbiome, this review covers all aspects derived from the balance between regulation/deregulation of AHR and its physio-pathological consequences.
Collapse
|
5
|
Attafi IM, Bakheet SA, Ahmad SF, Belali OM, Alanazi FE, Aljarboa SA, Al-Alallah IA, Korashy HM. Lead Nitrate Induces Inflammation and Apoptosis in Rat Lungs Through the Activation of NF-κB and AhR Signaling Pathways. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:64959-64970. [PMID: 35482242 PMCID: PMC9481511 DOI: 10.1007/s11356-022-19980-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 03/25/2022] [Indexed: 05/28/2023]
Abstract
Lead (Pb) is one of the most frequent hazardous air contaminants, where the lungs are particularly vulnerable to its toxicity. However, the Pb distribution and its impact on lung inflammation/apoptosis and particularly the involvement of nuclear factor kappa B (NF-κB) and aryl hydrocarbon receptor (AhR) signaling pathways in Pb-induced lung toxicity have not yet been fully investigated. Adult male Wistar albino rats were exposed to Pb nitrate 25, 50, and 100 mg/kg b.w. orally for 3 days. The histopathological changes of several rat organs were analyzed using hematoxylin and eosin staining. The concentrations of Pb ion in different organ tissues were quantified using inductive coupled plasma mass spectrometry, while gas chromatography-mass spectrometry was used to identify organic compounds. The changes in the mRNA and protein expression levels of inflammatory and apoptotic genes in response to Pb exposure were quantified by using RT-PCR and Western blot analyses, respectively. Treatment of rats with Pb for three consecutive days significantly increased the accumulation of Pb in lung tissues causing severe interstitial inflammation. Pb treatment also increased the percentage of lung apoptotic cells and modulated apoptotic genes (Bc2, p53, and TGF-α), inflammatory markers (IL-4, IL-10, TNF-α), and oxidative stress biomarkers (iNOS, CYP1A1, EphX) in rat lung tissues. These effects were associated with a significant increase in organic compounds, such as 3-nitrotyrosine and myeloperoxidase, and some inorganic elements, such as selenium. Importantly, the Pb-induced lung inflammation and apoptosis were associated with a proportional increase in the expression of NF-κB and AhR mRNAs and proteins. These findings clearly show that Pb induces severe inflammation and apoptosis in rat lungs and suggest that NF-κB and AhR may play a role in Pb-induced lung toxicity.
Collapse
Affiliation(s)
- Ibraheem M Attafi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Poison Control and Medical Forensic Chemistry Center, Jazan Health Affairs, Jazan, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Osamah M Belali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Aseer Central Hospital, Asser health affairs, Ministry of Health, Abha, Saudi Arabia
| | - Fawaz E Alanazi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Security Forces Hospital Program, Riyadh, Saudi Arabia
| | - Suliman A Aljarboa
- Central Laboratory, Research Center, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ibrahim A Al-Alallah
- Pathology and Clinical Laboratories Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
6
|
Berrocal M, Saez L, Mata AM. Sorcin Activates the Brain PMCA and Blocks the Inhibitory Effects of Molecular Markers of Alzheimer's Disease on the Pump Activity. Int J Mol Sci 2021; 22:ijms22116055. [PMID: 34205207 PMCID: PMC8200006 DOI: 10.3390/ijms22116055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Since dysregulation of intracellular calcium (Ca2+) levels is a common occurrence in neurodegenerative diseases, including Alzheimer’s disease (AD), the study of proteins that can correct neuronal Ca2+ dysregulation is of great interest. In previous work, we have shown that plasma membrane Ca2+-ATPase (PMCA), a high-affinity Ca2+ pump, is functionally impaired in AD and is inhibited by amyloid-β peptide (Aβ) and tau, two key components of pathological AD hallmarks. On the other hand, sorcin is a Ca2+-binding protein highly expressed in the brain, although its mechanism of action is far from being clear. Sorcin has been shown to interact with the intracellular sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA), and other modulators of intracellular Ca2+ signaling, such as the ryanodine receptor or presenilin 2, which is closely associated with AD. The present work focuses on sorcin in search of new regulators of PMCA and antagonists of Aβ and tau toxicity. Results show sorcin as an activator of PMCA, which also prevents the inhibitory effects of Aβ and tau on the pump, and counteracts the neurotoxicity of Aβ and tau by interacting with them.
Collapse
|
7
|
Jin UH, Michelhaugh SK, Polin LA, Shrestha R, Mittal S, Safe S. Omeprazole Inhibits Glioblastoma Cell Invasion and Tumor Growth. Cancers (Basel) 2020; 12:2097. [PMID: 32731514 PMCID: PMC7465678 DOI: 10.3390/cancers12082097] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Background: The aryl hydrocarbon receptor (AhR) is expressed in gliomas and the highest staining is observed in glioblastomas. A recent study showed that the AhR exhibited tumor suppressor-like activity in established and patient-derived glioblastoma cells and genomic analysis showed that this was due, in part, to suppression of CXCL12, CXCR4 and MMP9. Methods: Selective AhR modulators (SAhRMs) including AhR-active pharmaceuticals were screened for their inhibition of invasion using a spheroid invasion assay in patient-derived AhR-expressing 15-037 glioblastoma cells and in AhR-silenced 15-037 cells. Invasion, migration and cell proliferation were determined using spheroid invasion, Boyden chambers and scratch assay, and XTT metabolic assays for cell growth. Changes in gene and gene product expression were determined by real-time PCR and Western blot assays, respectively. In vivo antitumorigenic activity of omeprazole was determined in SCID mice bearing subcutaneous patient-derived 15-037 cells. Results: Results of a screening assay using patient-derived 15-037 cells (wild-type and AhR knockout) identified the AhR-active proton pump inhibitor omeprazole as an inhibitor of glioblastoma cell invasion and migration only AhR-expressing cells but not in cells where the AhR was downregulated. Omeprazole also enhanced AhR-dependent repression of the pro-invasion CXCL12, CXCR4 and MMP9 genes, and interactions and effectiveness of omeprazole plus temozolomide were response-dependent. Omeprazole (100 mg/kg/injection) inhibited and delayed tumors in SCID mice bearing patient-derived 15-037 cells injected subcutaneously. Conclusion: Our results demonstrate that omeprazole enhances AhR-dependent inhibition of glioblastoma invasion and highlights a potential new avenue for development of a novel therapeutic mechanism-based approach for treating glioblastoma.
Collapse
Affiliation(s)
- Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA;
| | - Sharon K. Michelhaugh
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA; (S.K.M.); (S.M.)
| | - Lisa A. Polin
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI 48201, USA;
| | - Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA;
| | - Sandeep Mittal
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA; (S.K.M.); (S.M.)
- Carilion Clinic-Neurosurgery, Roanoke, VA 24014, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
8
|
Jin UH, Karki K, Cheng Y, Michelhaugh SK, Mittal S, Safe S. The aryl hydrocarbon receptor is a tumor suppressor-like gene in glioblastoma. J Biol Chem 2019; 294:11342-11353. [PMID: 31171720 PMCID: PMC6643041 DOI: 10.1074/jbc.ra119.008882] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) plays an important role in maintaining cellular homeostasis and also in pathophysiology. For example, the interplay between the gut microbiome and microbially derived AhR ligands protects against inflammation along the gut-brain axis. The AhR and its ligands also inhibit colon carcinogenesis, but it has been reported that the AhR and its ligand kynurenine enhance glioblastoma (GBM). In this study, using both established and patient-derived GBM cells, we re-examined the role of kynurenine and the AhR in GBM, observing that kynurenine does not modulate AhR-mediated gene expression and does not affect invasion of GBM cells. Therefore, using an array of approaches, including ChIP, quantitative real-time PCR, and cell migration assays, we primarily focused on investigating the role of the AhR in GBM at the functional molecular and genomic levels. The results of transient and stable CRISPR/Cas9-mediated AhR knockdown in GBM cells indicated that loss of AhR enhances GBM tumor growth in a mouse xenograft model, increases GBM cell invasion, and up-regulates expression of pro-invasion/pro-migration genes, as determined by ingenuity pathway analysis of RNA-Seq data. We conclude that the AhR is a tumor suppressor-like gene in GBM; future studies are required to investigate whether the AhR could be a potential drug target for treating patients with GBM who express this receptor.
Collapse
Affiliation(s)
- Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843
| | - Keshav Karki
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843
| | - Yating Cheng
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843
| | | | - Sandeep Mittal
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia 24016
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
9
|
Vorontsova JE, Cherezov RO, Kuzin BA, Simonova OB. Aryl-Hydrocarbon Receptor as a Potential Target for Anticancer Therapy. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2019. [DOI: 10.1134/s1990750819010116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
10
|
Guerrina N, Traboulsi H, Eidelman DH, Baglole CJ. The Aryl Hydrocarbon Receptor and the Maintenance of Lung Health. Int J Mol Sci 2018; 19:E3882. [PMID: 30563036 PMCID: PMC6320801 DOI: 10.3390/ijms19123882] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 01/09/2023] Open
Abstract
Much of what is known about the Aryl Hydrocarbon Receptor (AhR) centers on its ability to mediate the deleterious effects of the environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; dioxin). However, the AhR is both ubiquitously-expressed and evolutionarily-conserved, suggesting that it evolved for purposes beyond strictly mediating responses to man-made environmental toxicants. There is growing evidence that the AhR is required for the maintenance of health, as it is implicated in physiological processes such as xenobiotic metabolism, organ development and immunity. Dysregulation of AhR expression and activity is also associated with a variety of disease states, particularly those at barrier organs such as the skin, gut and lungs. The lungs are particularly vulnerable to inhaled toxicants such as cigarette smoke. However, the role of the AhR in diseases such as chronic obstructive pulmonary disease (COPD)-a respiratory illness caused predominately by cigarette smoking-and lung cancer remains largely unexplored. This review will discuss the growing body of literature that provides evidence that the AhR protects the lungs against the damaging effects of cigarette smoke.
Collapse
Affiliation(s)
- Necola Guerrina
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada.
| | - Hussein Traboulsi
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| | - David H Eidelman
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
| | - Carolyn J Baglole
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada.
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
11
|
Zhang HJ, Liu YN, Xian P, Ma J, Sun YW, Chen JS, Chen X, Tang NJ. Maternal exposure to TCDD during gestation advanced sensory-motor development, but induced impairments of spatial learning and memory in adult male rat offspring. CHEMOSPHERE 2018; 212:678-686. [PMID: 30176550 DOI: 10.1016/j.chemosphere.2018.08.118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/15/2018] [Accepted: 08/23/2018] [Indexed: 05/19/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD) is an endocrine disrupting chemical (EDC) with high persistency. Even a low amount can pass the placental barrier during gestational exposure. Exposure to TCDD exposure can impair the development of the nervous system in children, leading to impaired learning ability in later-life. But the changes in neurobehavioral developments in infancy and childhood caused by TCDD are unknown. Pregnant Sprague-Dawley rats were given a consecutive daily dose of TCDD (200 or 800 ng/day/kg) or an equivalent volume of vehicle by gavage on gestational days 8-14 (GD 8-14) as the prenatal TCDD exposure model. In the offspring, early neurobehavioral development was assessed at postnatal day 5 (PND5) and eye-opening was monitored from PND10 onwards. Adult male offspring was tested by Morris Water Maze for spatial memory and learning ability evaluation. Hippocampus Nissl's staining and astrocyte GFAP immunohistochemistry were used to evaluate the activity of astrocytes. The results of the behavioral tests showed that gestational TCDD exposure induced premature motor activity and earlier eyes-opening, but lead to serious deficits of spatial memory and learning ability in the adult male offspring. Morphology and number of neurons in the hippocampus CA1 region was not affected, while the activity of astrocytes in the same region was significantly reduced. These data indicate that perinatal TCDD exposure induced premature neurobehavioral development but impaired the spatial learning and memory in adult male rat offspring. The decreased activity of astrocytes in the hippocampus may play a role in these adverse effects.
Collapse
Affiliation(s)
- Hua-Jing Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Ya-Nan Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Ping Xian
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Jing Ma
- Tianjin Cardiovascular Diseases Institute, Tianjin Chest Hospital, Tianjin, 300350, China.
| | - Ya-Wen Sun
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Jing-Shan Chen
- Department of Technology and Science, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Xi Chen
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Nai-Jun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
12
|
Vorontsova JE, Cherezov RO, Kuzin BA, Simonova OB. [Aryl-hydrocarbon receptor as a potential target for anticancer therapy]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2018; 64:397-415. [PMID: 30378556 DOI: 10.18097/pbmc20186405397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Aryl-hydrocarbon receptor (Aryl Hydrocarbon Receptor, AHR) is a ligand-dependent transcription factor, whose functions are related to xenobiotic detoxification, response to inflammation, and maintenance of tissue homeostasis. Recent investigations suggest that AHR also plays an important role in the processes of carcinogenesis. Increased expression of AHR is observed in several types of tumors and tumor cell lines. In addition, it turned out that the composition of pharmaceutical drugs used in oncotherapy includes some ligands AHR. These facts allow us to consider an aryl-hydrocarbon receptor as a potential target for anticancer therapy, especially for the treatment of severe cancers whose treatment options are very limited or do not exist at all. In this review the examples of AHR ligands' effect on tumor cell cultures and on model mice lines with AHR-dependent response are discussed.
Collapse
Affiliation(s)
- J E Vorontsova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - R O Cherezov
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - B A Kuzin
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - O B Simonova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
13
|
The Aryl Hydrocarbon Receptor and the Nervous System. Int J Mol Sci 2018; 19:ijms19092504. [PMID: 30149528 PMCID: PMC6163841 DOI: 10.3390/ijms19092504] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022] Open
Abstract
The aryl hydrocarbon receptor (or AhR) is a cytoplasmic receptor of pollutants. It translocates into the nucleus upon binding to its ligands, and forms a heterodimer with ARNT (AhR nuclear translocator). The heterodimer is a transcription factor, which regulates the transcription of xenobiotic metabolizing enzymes. Expressed in many cells in vertebrates, it is mostly present in neuronal cell types in invertebrates, where it regulates dendritic morphology or feeding behavior. Surprisingly, few investigations have been conducted to unravel the function of the AhR in the central or peripheral nervous systems of vertebrates. In this review, we will present how the AhR regulates neural functions in both invertebrates and vertebrates as deduced mainly from the effects of xenobiotics. We will introduce some of the molecular mechanisms triggered by the well-known AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), which impact on neuronal proliferation, differentiation, and survival. Finally, we will point out the common features found in mice that are exposed to pollutants, and in AhR knockout mice.
Collapse
|
14
|
Kolluri SK, Jin UH, Safe S. Role of the aryl hydrocarbon receptor in carcinogenesis and potential as an anti-cancer drug target. Arch Toxicol 2017; 91:2497-2513. [PMID: 28508231 PMCID: PMC6357772 DOI: 10.1007/s00204-017-1981-2] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 05/08/2017] [Indexed: 12/31/2022]
Abstract
The aryl hydrocarbon receptor (AhR) was initially identified as the receptor that binds and mediates the toxic effects induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and structurally related halogenated aromatics. Other toxic compounds including some polynuclear aromatic hydrocarbons act through the AhR; however, during the last 25 years, it has become apparent that the AhR plays an essential role in maintaining cellular homeostasis. Moreover, the scope of ligands that bind the AhR includes endogenous compounds such as multiple tryptophan metabolites, other endogenous biochemicals, pharmaceuticals and health-promoting phytochemicals including flavonoids, indole-3-carbinol and its metabolites. It has also been shown that like other receptors, the AhR is a drug target for multiple diseases including cancer, where both AhR agonists and antagonists effectively block many of the critical hallmarks of cancer in multiple tumor types. This review describes the anti-cancer activities of AhR ligands and demonstrates that it is time to separate the AhR from TCDD and exploit the potential of the AhR as a novel target for cancer chemotherapy.
Collapse
Affiliation(s)
- Siva Kumar Kolluri
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A & M University, 4466 TAMU, College Station, TX, 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A & M University, 4466 TAMU, College Station, TX, 77843, USA.
| |
Collapse
|
15
|
Yang Y, Chen S, Zhang Y, Lin X, Song Y, Xue Z, Qian H, Wang S, Wan G, Zheng X, Zhang L. Induction of autophagy by spermidine is neuroprotective via inhibition of caspase 3-mediated Beclin 1 cleavage. Cell Death Dis 2017; 8:e2738. [PMID: 28383560 PMCID: PMC5477584 DOI: 10.1038/cddis.2017.161] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/13/2017] [Accepted: 03/13/2017] [Indexed: 12/13/2022]
Abstract
Spermidine, a natural polyamine presented widely in mammalian cells, has been implicated to extend the lifespan of several model organisms by inducing autophagy. However, the effect of spermidine against neuronal damage has not yet been fully determined. In this study, neuronal cell injury was induced by treating PC12 cells and cortical neurons with 1 μM staurosporine (STS). We found that STS-induced cell injury could be efficiently attenuated by pretreatment with 1 mM spermidine. Spermidine inhibited the caspase 3 activation induced by STS. Moreover, STS incubation resulted in autophagic degradation failure, which could be attenuated by the pretreatment of spermidine. Knocking down the expression of Beclin 1 efficiently suppressed autophagosome and autolysosome accumulation, and abolished the protective effects of spermidine against STS-induced neurotoxicity. Increased Beclin 1 cleavage and partial nuclear translocation of Beclin 1 fragment was detected in STS-treated cells, which could be blocked by spermidine, pan-caspase inhibitor or caspase 3-specific inhibitor. The nuclear translocation of Beclin 1 fragment universally occurs in damaged neurons. Beclin 1 mutation at the sites of 146 and 149 prevented the intracellular re-distribution of Beclin 1 induced by STS. In addition, intraperitoneal injection of spermidine ameliorated ischemia/reperfusion-induced neuronal injury in the hippocampus and cortex of rats, possibly via blocking caspase 3 activation and consequent Beclin 1 cleavage. Our findings suggest that caspase 3-mediated Beclin 1 cleavage occurs in acute neuronal cell injury both in vitro and in vivo. The neuroprotective effect of spermidine may be related to inhibition of the caspase 3-mediated Beclin 1 cleavage and restoration of the Beclin 1-dependent autophagy.
Collapse
Affiliation(s)
- Yi Yang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, China
- Department of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Sicong Chen
- Department of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
- Clinical Research Center, The 2nd Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuqing Zhang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Xiaoxia Lin
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Yiyin Song
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Zhaoliang Xue
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
| | - Haoran Qian
- Department of General Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
| | - Shanshan Wang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Guihua Wan
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Xiaoxiang Zheng
- Department of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Lihui Zhang
- Department of Pharmacology, Hangzhou Key Laboratory of Medical Neurobiology, School of Medicine, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
16
|
AhR-dependent 2,3,7,8-tetrachlorodibenzo- p -dioxin toxicity in human neuronal cell line SHSY5Y. Neurotoxicology 2016; 56:55-63. [DOI: 10.1016/j.neuro.2016.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/04/2016] [Accepted: 07/04/2016] [Indexed: 11/21/2022]
|
17
|
Kalaiselvan I, Senthamarai M, Kasi PD. 2,3,7,8-TCDD-mediated toxicity in peripheral blood mononuclear cells is alleviated by the antioxidants present in Gelidiella acerosa: an in vitro study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:5111-5121. [PMID: 25388558 DOI: 10.1007/s11356-014-3799-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 10/29/2014] [Indexed: 06/04/2023]
Abstract
Seaweeds have been used as a source of traditional medicine worldwide for the treatment of various ailments, mainly due to their ability to quench the free radicals. The present study aims at evaluating the protective effect of methanolic extract of Gelidiella acerosa, an edible red seaweed against 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced toxicity in peripheral blood mononuclear cells (PBMC). For evaluating the protective effect of G. acerosa, PBMC were divided into four groups: vehicle control, TCDD (10 nM), TCDD + G. acerosa (300 μg/ml), and G. acerosa alone treated. Scavenging of intracellular reactive oxygen species (ROS) induced by TCDD was assessed by the dichloro-dihydro-fluorescein diacetate (DCFH-DA) method. Alterations at macromolecular level were quantified through lipid peroxidation (LPO) level, protein carbonyl content (PCC) level, and comet assay. The cellular morphology upon TCDD toxicity and G. acerosa treatment was obtained by light microscopy and histopathological studies. The chemical composition present in the methanolic extract of G. acerosa was determined by gas chromatography-mass spectrometry (GC-MS) analysis. The results reveal that 10 nM TCDD caused significant (P < 0.05) reduction in cell viability (94.10 ± 0.99), and treatment with 300 μg/ml extract increased the cell viability (99.24 ± 0.69). TCDD treatment resulted in a significant increase in the production of ROS, LPO (114 ± 0.09), and PCC (15.13 ± 1.53) compared to the control, whereas co-treatment with G. acerosa significantly (P < 0.05) mitigated the effects. Further, G. acerosa significantly (P < 0.05) prevented TCDD-induced genotoxicity and cell damage. GC-MS analysis showed the presence of n-hexadecanoic acid (retention time (RT) 13.15), cholesterol (RT 28.80), α-D-glucopyranose, 4-O-α-D-galactopyranosyl (RT 20.01), and azulene (RT 4.20). The findings suggest that G. acerosa has a strong protective ability against TCDD-induced cytotoxicity, oxidative stress, and DNA damage.
Collapse
Affiliation(s)
- Ilavarasi Kalaiselvan
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India
| | | | - Pandima Devi Kasi
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India.
| |
Collapse
|
18
|
Hecht E, Zago M, Sarill M, Rico de Souza A, Gomez A, Matthews J, Hamid Q, Eidelman DH, Baglole CJ. Aryl hydrocarbon receptor-dependent regulation of miR-196a expression controls lung fibroblast apoptosis but not proliferation. Toxicol Appl Pharmacol 2014; 280:511-25. [PMID: 25178717 DOI: 10.1016/j.taap.2014.08.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 12/29/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor implicated in the regulation of apoptosis and proliferation. Although activation of the AhR by xenobiotics such as dioxin inhibits the cell cycle and control apoptosis, paradoxically, AhR expression also promotes cell proliferation and survival independent of exogenous ligands. The microRNA (miRNA) miR-196a has also emerged as a regulator of proliferation and apoptosis but a relationship between the AhR and miR-196a is not known. Therefore, we hypothesized that AhR-dependent regulation of endogenous miR-196a expression would promote cell survival and proliferation. Utilizing lung fibroblasts from AhR deficient (AhR(-/-)) and wild-type (AhR(+/+)) mice, we show that there is ligand-independent regulation of miRNA, including low miR-196a in AhR(-/-) cells. Validation by qRT-PCR revealed a significant decrease in basal expression of miR-196a in AhR(-/-) compared to AhR(+/+) cells. Exposure to AhR agonists benzo[a]pyrene (B[a]P) and FICZ as well as AhR antagonist CH-223191 decreased miR-196a expression in AhR(+/+) fibroblasts concomitant with decreased AhR protein levels. There was increased proliferation only in AhR(+/+) lung fibroblasts in response to serum, corresponding to a decrease in p27(KIP1) protein, a cyclin-dependent kinase inhibitor. Increasing the cellular levels of miR-196a had no effect on proliferation or expression of p27(KIP1) in AhR(-/-) fibroblasts but attenuated cigarette smoke-induced apoptosis. This study provides the first evidence that AhR expression is essential for the physiological regulation of cellular miRNA levels- including miR-196a. Future experiments designed to elucidate the functional relationship between the AhR and miR-196a may delineate additional novel ligand-independent roles for the AhR.
Collapse
Affiliation(s)
- Emelia Hecht
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Michela Zago
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Miles Sarill
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Angela Rico de Souza
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Alvin Gomez
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Jason Matthews
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Qutayba Hamid
- Department of Medicine, McGill University, Montreal, Quebec, Canada; Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - David H Eidelman
- Department of Medicine, McGill University, Montreal, Quebec, Canada; Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Carolyn J Baglole
- Department of Medicine, McGill University, Montreal, Quebec, Canada; Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
19
|
Jiang J, Duan Z, Nie X, Xi H, Li A, Guo A, Wu Q, Jiang S, Zhao J, Chen G. Activation of neuronal nitric oxide synthase (nNOS) signaling pathway in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced neurotoxicity. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:119-130. [PMID: 24930124 DOI: 10.1016/j.etap.2014.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 05/07/2014] [Accepted: 05/09/2014] [Indexed: 06/03/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been reported to cause alterations in cognitive and motor behavior during both development and adulthood. In this study, the neuronal nitric oxide synthase (nNOS) signaling pathway was investigated in differentiated pheochromocytoma (PC12) cells to better understand the mechanisms of TCDD-induced neurotoxicity. TCDD exposure induced a time- and dose-dependent increase in nNOS expression. High levels of nitric oxide (NO) production by nNOS activation induced mitochondrial cytochrome c (Cyt-c) release and down-regulation of Bcl-2. Additionally, TCDD increased the expression of active caspase-3 and significantly led to apoptosis in PC12 cells. However, these effects above could be effectively inhibited by the addition of 7-nitroindazole (7-NI), a highly selective nNOS inhibitor. Moreover, in the brain cortex of Sprague-Dawley (SD) rats, nNOS was also found to have certain relationship with TCDD-induced neuronal apoptosis. Together, our findings establish a role for nNOS as an enhancer of TCDD-induced apoptosis in PC12 cells.
Collapse
Affiliation(s)
- Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Zhiqing Duan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Xiaoke Nie
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Hanqing Xi
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Aihong Li
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Aisong Guo
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Qiyun Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Shengyang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Jianya Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Gang Chen
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China.
| |
Collapse
|
20
|
Duan Z, Zhao J, Fan X, Tang C, Liang L, Nie X, Liu J, Wu Q, Xu G. The PERK-eIF2α signaling pathway is involved in TCDD-induced ER stress in PC12 cells. Neurotoxicology 2014; 44:149-59. [PMID: 24932542 DOI: 10.1016/j.neuro.2014.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 06/04/2014] [Accepted: 06/07/2014] [Indexed: 11/19/2022]
Abstract
Studies have shown that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces apoptotic cell death in neuronal cells. However, whether this is the result of endoplasmic reticulum (ER) stress-mediated apoptosis remains unknown. In this study, we determined whether ER stress plays a role in the TCDD-induced apoptosis of pheochromocytoma (PC12) cells and primary neurons. PC12 cells were exposed to different TCDD concentrations (1, 10, 100, 200, or 500nM) for varying lengths of time (1, 3, 6, 12, or 24h). TCDD concentrations much higher than 10nM (100, 200, or 500nM) markedly increased glucose-regulated protein (GRP78) and C/EBP homologous protein (CHOP) levels, which are hallmarks of ER stress. We also evaluated the effects of TCDD on ER morphology in PC12 cells and primary neurons that were treated with different TCDD concentrations (1, 10, 50, or 200nM) for 24h. Ultrastructural ER alterations were observed with transmission electron microscopy in PC12 cells and primary neurons treated with high concentrations of TCDD. Furthermore, TCDD-induced ER stress significantly promoted the activation of the PKR-like ER kinase (PERK), a sensor for the unfolded protein response (UPR), and its downstream target eukaryotic translation initiation factor 2 α (eIF2α); in contrast, TCDD did not appear to affect inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6 (ATF6), two other UPR sensors. Importantly, TCDD significantly inhibited eIF2α phosphorylation and triggered apoptosis in PC12 cells after 6-24h of treatment. Salubrinal, which activates the PERK-eIF2α pathway, significantly enhanced eIF2α phosphorylation in PC12 cells and attenuated the TCDD-induced cell death. In contrast, knocking down eIF2α using small interfering RNA markedly enhanced TCDD-induced cell death. Together, these results indicate that the PERK-eIF2α pathway plays an important role in TCDD-induced ER stress and apoptosis in PC12 cells.
Collapse
Affiliation(s)
- Zhiqing Duan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Jianya Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Xikang Fan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Cuiying Tang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Lingwei Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Xiaoke Nie
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China; Xinglin College, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Jiao Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | - Qiyun Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China.
| | - Guangfei Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China.
| |
Collapse
|
21
|
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces expression of p27kip1 and FoxO3a in female rat cerebral cortex and PC12 cells. Toxicol Lett 2014; 226:294-302. [DOI: 10.1016/j.toxlet.2014.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/15/2014] [Accepted: 02/22/2014] [Indexed: 12/17/2022]
|
22
|
Wan C, Liu J, Nie X, Zhao J, Zhou S, Duan Z, Tang C, Liang L, Xu G. 2, 3, 7, 8-Tetrachlorodibenzo-P-dioxin (TCDD) induces premature senescence in human and rodent neuronal cells via ROS-dependent mechanisms. PLoS One 2014; 9:e89811. [PMID: 24587053 PMCID: PMC3933666 DOI: 10.1371/journal.pone.0089811] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 01/27/2014] [Indexed: 11/30/2022] Open
Abstract
The widespread environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a potent toxicant that causes significant neurotoxicity. However, the biological events that participate in this process remain largely elusive. In the present study, we demonstrated that TCDD exposure triggered apparent premature senescence in rat pheochromocytoma (PC12) and human neuroblastoma SH-SY5Y cells. Senescence-associated β-galactosidase (SA-β-Gal) assay revealed that TCDD induced senescence in PC12 neuronal cells at doses as low as 10 nM. TCDD led to F-actin reorganization and the appearance of an alternative senescence marker, γ-H2AX foci, both of which are important features of cellular senescence. In addition, TCDD exposure altered the expression of senescence marker proteins, such as p16, p21 and p-Rb, in both dose- and time-dependent manners. Furthermore, we demonstrated that TCDD promotes mitochondrial dysfunction and the accumulation of cellular reactive oxygen species (ROS) in PC12 cells, leading to the activation of signaling pathways that are involved in ROS metabolism and senescence. TCDD-induced ROS generation promoted significant oxidative DNA damage and lipid peroxidation. Notably, treatment with the ROS scavenger N-acetylcysteine (NAC) markedly attenuated TCDD-induced ROS production, cellular oxidative damage and neuronal senescence. Moreover, we found that TCDD induced a similar ROS-mediated senescence response in human neuroblastoma SH-SY5Y cells. In sum, these results demonstrate for the first time that TCDD induces premature senescence in neuronal cells by promoting intracellular ROS production, supporting the idea that accelerating the onset of neuronal senescence may be an important mechanism underlying TCDD-induced neurotoxic effects.
Collapse
Affiliation(s)
- Chunhua Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jiao Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Xiaoke Nie
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jianya Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Songlin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Zhiqing Duan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Cuiying Tang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Lingwei Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Guangfei Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
- * E-mail: .
| |
Collapse
|
23
|
2,3,7,8-Tetrachlorodibenzo-p-dioxin induces apoptosis by disruption of intracellular calcium homeostasis in human neuronal cell line SHSY5Y. Apoptosis 2014; 17:1170-81. [PMID: 22986482 DOI: 10.1007/s10495-012-0760-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The persistent xenobiotic agent 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces neurotoxic effects that alters neurodevelopment and behavior both during development and adulthood. There are many ongoing efforts to determine the molecular mechanisms of TCDD-mediated neurotoxicity, the signaling pathways involved and its molecular targets in neurons. In this work, we have used SHSY5Y human neuroblastoma cells to characterize the TCDD-induced toxicity. TCDD produces a loss of viability linked to an increased caspase-3 activity, PARP-1 fragmentation, DNA laddering, nuclear fragmentation and hypodiploid (apoptotic) DNA content, in a similar way than staurosporine, a prototypical molecule of apoptosis induction. In addition, TCDD produces a decrease of mitochondrial membrane potential and an increase of intracellular calcium concentration (P < 0.05). Finally, based on the high lipophilic properties of the dioxin, we test the TCDD effect on the membrane integrity using sarcoplasmic reticulum vesicles as a model. TCDD produces calcium efflux through the membrane and an anisotropy decrease (P < 0.05) that reflects an increase in membrane fluidity. Altogether these results support the hypothesis that TCDD toxicity in SHSY5Y neuroblastoma cells provokes the disruption of calcium homeostasis, probably affecting membrane structural integrity, leading to an apoptotic process.
Collapse
|
24
|
Xu G, Li Y, Yoshimoto K, Wu Q, Chen G, Iwata T, Mizusawa N, Wan C, Nie X. 2,3,7,8-Tetrachlorodibenzo-p-dioxin stimulates proliferation of HAPI microglia by affecting the Akt/GSK-3β/cyclin D1 signaling pathway. Toxicol Lett 2013; 224:362-70. [PMID: 24231000 DOI: 10.1016/j.toxlet.2013.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/28/2013] [Accepted: 11/03/2013] [Indexed: 12/19/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is an environmental toxin that induces apoptosis of neurons and a pro-inflammatory response in microglial cells. First, we found that TCDD induced proliferation of HAPI microglial cells in a dose- and time-dependent manner. Flow cytometry analysis showed that this proliferation by TCDD was due to mainly enhancing the G1 to S phase transition. Next, it was found that TCDD treatment led to up-regulation of cyclin D1, which induces cell cycle progression from G1 to S phase, in a time-dependent manner. As for molecular mechanism, we revealed that TCDD was capable of inducing Akt phosphorylation and activation, resulting in phosphorylation and inactivation of glycogen synthase kinase-3β (GSK-3β). Inactivated GSK-3β attenuated proteasomal degradation of cyclin D1 by reducing Thr(286)-phosphorylated cyclin D1 levels. Moreover, inactivated GSK-3β increased cyclin D1 gene transcription by increasing its transcription factor β-catenin in the nucleus. Further, blockage of phosphoinositide 3-kinase/Akt kinase with their specific inhibitors, LY294002 and Akt 1/2 kinase inhibitor, significantly reduced TCDD-enhanced proliferation of HAPI microglial cells. In conclusion, TCDD stimulates proliferation of HAPI microglial cells by affecting the Akt/GSK-3β/cyclin D1 signaling pathway.
Collapse
Affiliation(s)
- Guangfei Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226001, Jiangsu, People's Republic of China; Department of Medical Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8504, Japan.
| | - Yuanye Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226001, Jiangsu, People's Republic of China; Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226001, Jiangsu, People's Republic of China
| | - Katsuhiko Yoshimoto
- Department of Medical Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8504, Japan
| | - Qiyun Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226001, Jiangsu, People's Republic of China
| | - Gang Chen
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226001, Jiangsu, People's Republic of China
| | - Takeo Iwata
- Department of Medical Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8504, Japan
| | - Noriko Mizusawa
- Department of Medical Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8504, Japan
| | - Chunhua Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226001, Jiangsu, People's Republic of China
| | - Xiaoke Nie
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226001, Jiangsu, People's Republic of China
| |
Collapse
|
25
|
Li Y, Chen G, Zhao J, Nie X, Wan C, Liu J, Duan Z, Xu G. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces microglial nitric oxide production and subsequent rat primary cortical neuron apoptosis through p38/JNK MAPK pathway. Toxicology 2013; 312:132-41. [PMID: 23969120 DOI: 10.1016/j.tox.2013.08.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/09/2013] [Accepted: 08/10/2013] [Indexed: 12/22/2022]
Abstract
It has been widely accepted that microglia, which are the innate immune cells in the brain, upon activation can cause neuronal damage. In the present study, we investigated the role of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in regulating microglial nitric oxide production and its role in causing neuronal damage. The study revealed that TCDD stimulates the expression of inducible nitric oxide synthase (iNOS) as well as the production of nitric oxide (NO) in a dose- and time-dependent manner. Further, a rapid activation of p38 and JNK MAPKs was found in HAPI microglia following TCDD treatment. Blockage of p38 and JNK kinases with their specific inhibitors, SB202190 and SP600125, significantly reduced TCDD-induced iNOS expression and NO production. In addition, it was demonstrated through treating rat primary cortical neurons with media conditioned with TCDD treated microglia that microglial iNOS activation mediates neuronal apoptosis. Lastly, it was also found that p38 and JNK MAPK inhibitors could attenuate the apoptosis of rat cortical neurons upon exposure to medium conditioned by TCDD-treated HAPI microglial cells. Based on these observations, we highlight that the p38/JNK MAPK pathways play an important role in TCDD-induced iNOS activation in rat HAPI microglia and in the subsequent induction of apoptosis in primary cortical neurons.
Collapse
Affiliation(s)
- Yuanye Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China; Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226001 Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
2,3,7,8-TCDD induces neurotoxicity and neuronal apoptosis in the rat brain cortex and PC12 cell line through the down-regulation of the Wnt/β-catenin signaling pathway. Neurotoxicology 2013; 37:63-73. [DOI: 10.1016/j.neuro.2013.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 04/12/2013] [Accepted: 04/16/2013] [Indexed: 02/02/2023]
|
27
|
Xie HQ, Xu HM, Fu HL, Hu Q, Tian WJ, Pei XH, Zhao B. AhR-mediated effects of dioxin on neuronal acetylcholinesterase expression in vitro. ENVIRONMENTAL HEALTH PERSPECTIVES 2013; 121:613-8. [PMID: 23426015 PMCID: PMC3673198 DOI: 10.1289/ehp.1206066] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 02/19/2013] [Indexed: 05/18/2023]
Abstract
BACKGROUND Deficits in cognitive functioning have been reported in humans exposed to dioxins and dioxin-like compounds. Evidence suggests that dioxins induce cholinergic dysfunction mediated by hypothyroidism. However, little is known about direct effects of dioxins on the cholinergic system. OBJECTIVES We investigated the action of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on acetylcholinesterase (AChE), a key enzyme in cholinergic neurotransmission. METHODS We used SK-N-SH human-derived neuronal cells to evaluate the effect of dioxin exposure on AChE. RESULTS We consistently found a significant decrease in enzymatic activity of AChE in cultured neurons treated with TCDD. We also found that, unlike organophosphate pesticides that directly act on the catalytic center of AChE, the suppressive effect of dioxin was through transcriptional regulation. The addition of CH223191, an inhibitor of the aryl hydrocarbon receptor (AhR)-dependent pathway, counteracted the TCDD-induced suppression of AChE, suggesting involvement of the AhR-dependent pathway. The existence of putative dioxin-responsive element (DRE) consensus sequences in the human ACHE promoter region further supported this hypothesis. Consistent with the absence of DRE elements in mouse or rat ACHE promoter regions, suppression of AChE by TCDD did not occur in rat neuronal cells, indicating a potential species-specific effect. CONCLUSIONS In SK-N-SH cells, dioxin suppressed the activity of neuronal AChE via AhR-mediated transcriptional down-regulation. This is the first study to report direct interference by dioxin with the cholinergic neurotransmission system.
Collapse
Affiliation(s)
- Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Xu G, Duan Z, Chen G, Nie X, Liu J, Zhang Y, Li Y, Wan C, Jiang J. Role of mitogen-activated protein kinase cascades in 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced apoptosis in neuronal pheochromocytoma cells. Hum Exp Toxicol 2013; 32:1278-91. [PMID: 23584357 DOI: 10.1177/0960327113482595] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Mitogen-activated protein kinases (MAPKs) are involved in neuronal death caused by many cytotoxins. Conventional MAPKs consist of three family members: extracellular signal-regulated kinase-1/2 (ERK1/2), c-Jun N-terminal kinase (JNK) and p38. It has been originally shown that ERK1/2 is important for cell survival, whereas JNK and p38 are deemed stress responsive and thus involved in apoptosis. However, information describing the role of MAPKs in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced neurotoxicity is insufficient. The aim of this study was to identify the role of MAPK cascades in TCDD-induced neurotoxicity using differentiated pheochromocytoma (PC12) cells as a model for neuronal cells. Cell viability assay, terminal deoxynucleotidyl transferase dUTP nick-end labeling assay and flow cytometry analysis showed that TCDD attenuated cell viability with a dose- and time-dependent manner and significantly induced apoptosis in primary cortical neurons and PC12 cells. Western blot analysis indicated that TCDD markedly activated the expression of ERK1/2, JNK and p38 in TCDD-treated PC12 cells. Furthermore, PD98059 (ERK1/2 inhibitor), SP600125 (JNK inhibitor) and SB202190 (p38 inhibitor) notably blocked the effect of TCDD on cell apoptosis. Based on the findings above, it is concluded that the activation of MAPK signaling pathways may be associated with TCDD-mediated neuronal apoptosis.
Collapse
Affiliation(s)
- G Xu
- 1Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Iida M, Kim EY, Murakami Y, Shima Y, Iwata H. Toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on the peripheral nervous system of developing red seabream (Pagrus major). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2013; 128-129:193-202. [PMID: 23314332 DOI: 10.1016/j.aquatox.2012.12.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 12/07/2012] [Accepted: 12/11/2012] [Indexed: 06/01/2023]
Abstract
We investigated 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced effects on the morphology of peripheral nervous system (PNS) in the developing red seabream (Pagrus major) embryos. The embryos at 10h post-fertilization (hpf) were treated with 0, 0.1, 0.4 or 1.7 μg/L of TCDD in seawater for 80 min. The morphology of PNS was microscopically observed with florescence staining using an anti-acetylated tubulin antibody at 48, 78, 120 and 136 hpf. Axon length of facial nerve (VII) was found to be shortened by TCDD exposure. Axon guidance in the glossopharyngeal nerve (IX) and vagus nerve (X) was altered at 120 and 136 hpf in a TCDD dose-dependent manner. Lowest observable effect level of TCDD (0.1 μg/L) that induced the morphological alteration of PNS was lower than those of other endpoints on morphological deformities so far reported. Given that the growth cone at the tip of growing nerve axons advances under the influence of its surrounding tissues, we hypothesized that TCDD exposure would affect (1) the nerve cell proliferation/differentiation, (2) the structure of muscle as an axon target and (3) the nerve guidance factor in the embryos. By the immunostaining of embryos with an antibody against the neuronal specific RNA-binding protein, HuD, and an antibody against the sarcomeric myosin, no morphological effects were observed on the neural proliferation/differentiation and the structure of facial muscles of TCDD-treated embryos. In contrast, whole mount in situ hybridization of semaphorin 3A (Sema3A), a secretory axon repulsion factor, revealed the altered expression pattern of its transcripts in TCDD-treated embryos. Our findings suggest that TCDD treatment affects the projection of PNS in the developing red seabream embryos through the effects on the axonal growth cone guidance molecule such as Sema3A, but not on the neuronal differentiation/proliferation and axon target. The PNS in developing embryos may be one of the most sensitive biomarkers to the exposure of dioxin-like compounds.
Collapse
Affiliation(s)
- Midori Iida
- Center for Marine Environmental Studies (CMES), Ehime University, Bunkyo-cho 2-5, Matsuyama, Japan
| | | | | | | | | |
Collapse
|
30
|
Sánchez-Martín FJ, Fernández-Salguero PM, Merino JM. Aryl hydrocarbon receptor-dependent induction of apoptosis by 2,3,7,8-tetrachlorodibenzo-p-dioxin in cerebellar granule cells from mouse. J Neurochem 2011; 118:153-62. [PMID: 21534955 DOI: 10.1111/j.1471-4159.2011.07291.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a prototypical environmental contaminant with neurotoxic properties that alters neurodevelopment and behavior. TCDD is a ligand of the aryl hydrocarbon receptor (AhR), which is a key signaling molecule to fully understand the toxic and carcinogenic properties of dioxin. Much effort is underway to unravel the molecular mechanisms and the signaling pathways involved in TCDD-induced neurotoxicity, and to define its molecular targets in neurons. We have used cerebellar granule cells (CGC) from wild-type (AhR+/+) and AhR-null (AhR-/-) mice to characterize the cell death that takes place in neurons after TCDD toxicity. TCDD induced cell death in CGC cultures from wild-type mice with an EC(50) of 127±21 nM. On the contrary, when CGC neurons from AhR-null mice were treated with TCDD no significant cell death was observed. The role of AhR in TCDD-induced death was further assessed by using the antagonists resveratrol and α-naphtoflavone, which readily protected against TCDD toxicity in AhR+/+ CGC cultures. AhR+/+ CGC cultures treated with TCDD showed nuclear fragmentation, DNA laddering, and increased caspase 3 activity, similarly to what was found by the use of staurosporine, a well-established inducer of apoptosis. Finally, the AhR pathway was active in CGC because TCDD could induce the expression of the target gene cytochrome P450 1A2 in AhR+/+ CGC cultures. All together these results support the hypothesis that TCDD toxicity in CGC neurons involves the AhR and that it takes place mainly through an apoptotic process. AhR could be then considered a novel target in neurotoxicity and neurodegeneration whose down-modulation could block certain xenobiotic-related adverse effects in CNS.
Collapse
Affiliation(s)
- Francisco J Sánchez-Martín
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | | | | |
Collapse
|
31
|
Lin S, Yang Z, Zhang X, Bian Z, Cai Z. Hippocampal metabolomics reveals 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicity associated with ageing in Sprague-Dawley rats. Talanta 2011; 85:1007-12. [PMID: 21726731 DOI: 10.1016/j.talanta.2011.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 05/01/2011] [Accepted: 05/05/2011] [Indexed: 10/18/2022]
Abstract
Metabolomics, the exponentially developing technique, could provide a systemic mapping in toxicology by directly measuring small molecular metabolites. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) was found to be neurotoxic in mammalian animals. In this study, we employed liquid chromatography/quadrupole time-of-flight mass spectrometry for non-targeted analysis of metabolic profiling in hippocampal sample sets of the rats exposed to TCDD. Hippocampal metabolome from different ages of the healthy rats (4-week, 12-week and 20-week) was also deciphered. The relationship between the two tested cases was unlocked to delineate TCDD toxicity associated with ageing. Tandem mass spectrometry fragmentation in conjunction with metabolic database searching and compared to authentic standards was utilized for metabolite identification. As a consequence, the reduced levels of phenylalanine and leucine/isoleucine as well as the up-regulation of inosine and hypoxanthine were highlighted for understanding of TCDD toxicity related to age in rats and the trajectory was depicted by principal components analysis.
Collapse
Affiliation(s)
- Shuhai Lin
- Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | | | | | | | | |
Collapse
|
32
|
Chopra M, Schrenk D. Dioxin toxicity, aryl hydrocarbon receptor signaling, and apoptosis-persistent pollutants affect programmed cell death. Crit Rev Toxicol 2011; 41:292-320. [PMID: 21323611 DOI: 10.3109/10408444.2010.524635] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exogenous ligands of the aryl hydrocarbon receptor (AhR) such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related substances are highly toxic pollutants ubiquitously present in the environment. They cause a variety of toxic effects to different organs and tissues. Among other effects, TCDD exposure to laboratory animals leads to thymus atrophy and immunosuppression on the one hand, and to tumor formation on the other. Apoptosis appears to be involved in both these toxic effects: AhR activation by TCDD was discussed to induce apoptosis of immune cells, leading to the depletion of thymocytes and ultimately immunosuppression. This mechanism could help to explain the highly immunotoxic actions of TCDD but it is nevertheless under debate whether this is the mode of action for immunosuppression by this class of chemical substances. In other cell types, especially liver cells, TCDD inhibits apoptosis induced by genotoxic treatment. In initiation-promotion studies, TCDD was shown to be a potent liver tumor promoter. Among other theories it was hypothesized that TCDD acts as a tumor promoter by preventing initiated cells from undergoing apoptosis. The exact mechanisms of apoptosis inhibition by TCDD are not fully understood, but both in vivo and in vitro studies consistently showed an involvement of the tumor suppressor p53 in this effect. Various strings of evidence have been established linking apoptosis to the detrimental effects of exogenous activation of the AhR. Within this article, studies elucidating the effects of TCDD and related substances on apoptosis signaling, be it inducing or repressing, is to be reviewed.
Collapse
Affiliation(s)
- Martin Chopra
- Institute of Food Chemistry and Toxicology, University of Kaiserslautern, Kaiserslautern, Germany
| | | |
Collapse
|