1
|
Pietrzak-Wawrzyńska BA, Wnuk A, Przepiórska-Drońska K, Łach A, Kajta M. Posttreatment with PaPE-1 Protects from Aβ-Induced Neurodegeneration Through Inhibiting the Expression of Alzheimer's Disease-Related Genes and Apoptosis Process That Involves Enhanced DNA Methylation of Specific Genes. Mol Neurobiol 2024; 61:4130-4145. [PMID: 38064105 PMCID: PMC11236864 DOI: 10.1007/s12035-023-03819-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/19/2023] [Indexed: 07/11/2024]
Abstract
Targeting the non-nuclear estrogen receptor (ER) signaling has been postulated as novel therapeutic strategy for central nervous system pathologies. Recently, we showed that newly designed PaPE-1 (Pathway Preferential Estrogen-1), which selectively activates ER non-nuclear signaling pathways, elicited neuroprotection in a cellular model of Alzheimer's disease (AD) when it was applied at the same time as amyloid-β (Aβ). Since delayed treatment reflects clinical settings better than cotreatment does, current basic study proposes a novel therapeutic approach for AD that relies on a posttreatment with PaPE-1. In this study, mouse neuronal cell cultures treated with preaggregated Aβ1-42 (10 µM) showed the presence of extracellular Aβ1-42, confirming the adequacy of the AD model used. We are the first to demonstrate that a 24-h delayed posttreatment with PaPE-1 decreased the degree of Aβ-induced neurodegeneration, restored neurite outgrowth, and inhibited the expression of AD-related genes, i.e., Rbfox, Apoe, Bace2, App, and Ngrn, except for Chat, which was stimulated. In addition, PaPE-1 elicited anti-apoptotic effects by inhibiting Aβ-induced caspase activities as well as attenuating apoptotic chromatin condensation, and in these ways, PaPE-1 prevented neuronal cell death. Posttreatment with PaPE-1 also downregulated the Aβ-affected mRNA expression of apoptosis-specific factors, such as Bax, Gsk3b, Fas, and Fasl, except for Bcl2, which was upregulated by PaPE-1. In parallel, PaPE-1 decreased the protein levels of BAX, FAS, and FASL, which were elevated in response to Aβ. PaPE-1 elicited a decrease in the BAX/BCL2 ratio that corresponds to increased methylation of the Bax gene. However, the PaPE-1-evoked Bcl2 gene hypermethylation suggests other PaPE-1-dependent mechanisms to control Aβ-induced apoptosis.
Collapse
Affiliation(s)
- Bernadeta A Pietrzak-Wawrzyńska
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - Agnieszka Wnuk
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - Karolina Przepiórska-Drońska
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - Andrzej Łach
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - Małgorzata Kajta
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland.
| |
Collapse
|
2
|
Przepiórska-Drońska K, Wnuk A, Pietrzak-Wawrzyńska BA, Łach A, Biernat W, Wójtowicz AK, Kajta M. Amorfrutin B Compromises Hypoxia/Ischemia-induced Activation of Human Microglia in a PPARγ-dependent Manner: Effects on Inflammation, Proliferation Potential, and Mitochondrial Status. J Neuroimmune Pharmacol 2024; 19:34. [PMID: 38949694 PMCID: PMC11217078 DOI: 10.1007/s11481-024-10135-9] [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: 12/22/2023] [Accepted: 06/08/2024] [Indexed: 07/02/2024]
Abstract
Amorfrutin B is a selective PPARγ modulator that we demonstrated to be a promising neuroprotective compound in cellular models of stroke and perinatal asphyxia. Although neuronal mechanisms of amorfrutin B-evoked neuroprotection have been identified, none of them reflects the actions of the compound on microglia, which play a pivotal role in brain response to hypoxia/ischemia. Here, we provide evidence for amorfrutin B-induced effects on human microglia subjected to hypoxia/ischemia; the compound counteracts inflammation, and influences mitochondrial status and proliferation potential in a PPARγ-dependent manner. Post-treatment with amorfrutin B decreased the IBA1 fluorescence intensity, reduced caspase-1 activity, and downregulated IL1B/IL-1β and TNFA but not IL10/IL-10 expression, which was upregulated. Amorfrutin B also stimulated PPARγ signaling, as evidenced by increased mRNA and/or protein levels of PPARγ and PGC1α. In addition, amorfrutin B reversed the hypoxia/ischemia-evoked effects on mitochondria-related parameters, such as mitochondrial membrane potential, BCL2/BCL2 expression and metabolic activity, which were correlated with diminished proliferation potential of microglia. Interestingly, the inhibitory effect of amorfrutin B on the proliferation potential and mitochondrial function of microglia is opposite to the stimulatory effect of amorfrutin B on mouse neuronal survival, as evidenced by increased neuronal viability and reduced neurodegeneration. In summary, this study showed for the first time that amorfrutin B compromises hypoxia/ischemia-induced activation of human microglia in a PPARγ-dependent manner, which involves inhibiting inflammation, normalizing mitochondrial status, and controlling proliferation potential. These data extend the protective potential of amorfrutin B in the pharmacotherapy of hypoxic/ischemic brain injury, targeting not only neurons but also activated microglia.
Collapse
Affiliation(s)
- Karolina Przepiórska-Drońska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12, 31-343, Krakow, Poland
| | - Agnieszka Wnuk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12, 31-343, Krakow, Poland
| | - Bernadeta Angelika Pietrzak-Wawrzyńska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12, 31-343, Krakow, Poland
| | - Andrzej Łach
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12, 31-343, Krakow, Poland
| | - Weronika Biernat
- Faculty of Animal Sciences, Department of Nutrition, Animal Biotechnology and Fisheries, University of Agriculture, Adama Mickiewicza 24/28, 30-059, Krakow, Poland
| | - Anna Katarzyna Wójtowicz
- Faculty of Animal Sciences, Department of Nutrition, Animal Biotechnology and Fisheries, University of Agriculture, Adama Mickiewicza 24/28, 30-059, Krakow, Poland
| | - Małgorzata Kajta
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Neuropharmacology and Epigenetics, Smetna Street 12, 31-343, Krakow, Poland.
| |
Collapse
|
3
|
Sasaki N, Morse G, Jones L, Carpenter DO. Effects of mixtures of polychlorinated biphenyls (PCBs) and three organochlorine pesticides on cognitive function differ between older Mohawks at Akwesasne and older adults in NHANES. ENVIRONMENTAL RESEARCH 2023; 236:116861. [PMID: 37562737 DOI: 10.1016/j.envres.2023.116861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/14/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Akwesasne Mohawks has been exposed to high concentrations of polychlorinated biphenyls (PCBs) and background levels of organochlorine pesticides, hexachlorobenzene (HCB), dichlorodiphenyl dichloroethylene (DDE), and mirex. We have previously reported relative contributions to the mixture of low- and high-chlorinated PCBs, HCB, and DDE on cognitive decrements in Mohawks of various ages. OBJECTIVE This study examines differences in the mixture effects of PCB congener groups, HCB, DDE, and mirex on cognitive function in older Mohawks and less PCB-exposed older adults from the National Health and Nutrition Examination Survey (NHANES) 1999-2002 cycles. METHODS We used Bayesian kernel machine regression (BKMR) to evaluate the mixture effects of different PCB congener groups, HCB, DDE, and mirex on cognitive function in both populations. Models were adjusted for age, sex, education levels, and race/ethnicity focusing on individuals 60 years and older. RESULTS Older Mohawks had 3-fold higher mean total PCB concentrations and 1.8-fold higher mirex, but slightly lower mean DDE and HCB levels than NHANES older adults. Higher mixture concentrations were significantly associated with greater cognitive decline. In older Mohawks, low- and high-chlorinated PCBs, HCB, and DDE contributed to the cognitive score decline. In contrast, score decline in older NHANES adults were primarily from high-chlorinated PCBs and DDE with a threshold dose of approximately 2.08-2.27 ng/g and 2.02-2.40 ng/g, respectively. CONCLUSION Mixtures of PCBs and organochlorine pesticides increase the risk of cognitive decline in both older Mohawks and NHANES older adults. However, contributions to these mixture effects show significant differences. In older Mohawks, high- and low-chlorinated PCBs, DDE, and HCB are the primary contributors, while high-chlorinated PCBs and DDE are important contributors in NHANES older adults. Due to chronic heavy exposures to PCBs, older Mohawks had a significantly increased risk of cognitive decline compared to general older adults from NHANES.
Collapse
Affiliation(s)
- Nozomi Sasaki
- Institute for Health and the Environment, University at Albany, Rensselaer, NY, 12144, USA.
| | - Gayle Morse
- Institute for Health and the Environment, University at Albany, Rensselaer, NY, 12144, USA; Department of Psychology, School of Health Sciences, Russell Sage College, Troy, NY, 12180, USA
| | - Laura Jones
- Center for Biostatistics, Bassett Research Institute, Bassett Health, Cooperstown, NY, 13326, USA; Epidemiology and Biostatistics, SUNY Albany School of Public Health, Rensselaer, NY 12144, USA
| | - David O Carpenter
- Institute for Health and the Environment, University at Albany, Rensselaer, NY, 12144, USA
| |
Collapse
|
4
|
Pietrzak BA, Wnuk A, Przepiórska K, Łach A, Kajta M. Posttreatment with Ospemifene Attenuates Hypoxia- and Ischemia-Induced Apoptosis in Primary Neuronal Cells via Selective Modulation of Estrogen Receptors. Neurotox Res 2023; 41:362-379. [PMID: 37129835 PMCID: PMC10354152 DOI: 10.1007/s12640-023-00644-5] [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: 02/13/2023] [Revised: 03/17/2023] [Accepted: 04/02/2023] [Indexed: 05/03/2023]
Abstract
Stroke and perinatal asphyxia have detrimental effects on neuronal cells, causing millions of deaths worldwide each year. Since currently available therapies are insufficient, there is an urgent need for novel neuroprotective strategies to address the effects of cerebrovascular accidents. One such recent approach is based on the neuroprotective properties of estrogen receptors (ERs). However, activation of ERs by estrogens may contribute to the development of endometriosis or hormone-dependent cancers. Therefore, in this study, we utilized ospemifene, a novel selective estrogen receptor modulator (SERM) already used in dyspareunia treatment. Here, we demonstrated that posttreatment with ospemifene in primary neocortical cell cultures subjected to 18 h of hypoxia and/or ischemia followed by 6 h of reoxygenation has robust neuroprotective potential. Ospemifene partially reverses hypoxia- and ischemia-induced changes in LDH release, the degree of neurodegeneration, and metabolic activity. The mechanism of the neuroprotective actions of ospemifene involves the inhibition of apoptosis since the compound decreases caspase-3 overactivity during hypoxia and enhances mitochondrial membrane potential during ischemia. Moreover, in both models, ospemifene decreased the levels of the proapoptotic proteins BAX, FAS, FASL, and GSK3β while increasing the level of the antiapoptotic protein BCL2. Silencing of specific ERs showed that the neuroprotective actions of ospemifene are mediated mainly via ESR1 (during hypoxia and ischemia) and GPER1 (during hypoxia), which is supported by ospemifene-evoked increases in ESR1 protein levels in hypoxic and ischemic neurons. The results identify ospemifene as a promising neuroprotectant, which in the future may be used to treat injuries due to brain hypoxia/ischemia.
Collapse
Affiliation(s)
- Bernadeta A Pietrzak
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, Krakow, 31-343, Poland
| | - Agnieszka Wnuk
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, Krakow, 31-343, Poland
| | - Karolina Przepiórska
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, Krakow, 31-343, Poland
| | - Andrzej Łach
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, Krakow, 31-343, Poland
| | - Małgorzata Kajta
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, Krakow, 31-343, Poland.
| |
Collapse
|
5
|
Schaffner SL, Kobor MS. DNA methylation as a mediator of genetic and environmental influences on Parkinson's disease susceptibility: Impacts of alpha-Synuclein, physical activity, and pesticide exposure on the epigenome. Front Genet 2022; 13:971298. [PMID: 36061205 PMCID: PMC9437223 DOI: 10.3389/fgene.2022.971298] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/25/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with a complex etiology and increasing prevalence worldwide. As PD is influenced by a combination of genetic and environment/lifestyle factors in approximately 90% of cases, there is increasing interest in identification of the interindividual mechanisms underlying the development of PD as well as actionable lifestyle factors that can influence risk. This narrative review presents an outline of the genetic and environmental factors contributing to PD risk and explores the possible roles of cytosine methylation and hydroxymethylation in the etiology and/or as early-stage biomarkers of PD, with an emphasis on epigenome-wide association studies (EWAS) of PD conducted over the past decade. Specifically, we focused on variants in the SNCA gene, exposure to pesticides, and physical activity as key contributors to PD risk. Current research indicates that these factors individually impact the epigenome, particularly at the level of CpG methylation. There is also emerging evidence for interaction effects between genetic and environmental contributions to PD risk, possibly acting across multiple omics layers. We speculated that this may be one reason for the poor replicability of the results of EWAS for PD reported to date. Our goal is to provide direction for future epigenetics studies of PD to build upon existing foundations and leverage large datasets, new technologies, and relevant statistical approaches to further elucidate the etiology of this disease.
Collapse
Affiliation(s)
- Samantha L. Schaffner
- Edwin S. H. Leong Healthy Aging Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Michael S. Kobor
- Edwin S. H. Leong Healthy Aging Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Seralini GE, Jungers G. Endocrine disruptors also function as nervous disruptors and can be renamed endocrine and nervous disruptors (ENDs). Toxicol Rep 2021; 8:1538-1557. [PMID: 34430217 PMCID: PMC8365328 DOI: 10.1016/j.toxrep.2021.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 01/14/2023] Open
Abstract
Endocrine disruption (ED) and endocrine disruptors (EDs) emerged as scientific concepts in 1995, after numerous chemical pollutants were found to be responsible for reproductive dysfunction. The World Health Organization established in the United Nations Environment Programme a list of materials, plasticizers, pesticides, and various pollutants synthesized from petrochemistry that impact not only reproduction, but also hormonal functions, directly or indirectly. Cells communicate via either chemical or electrical signals transmitted within the endocrine or nervous systems. To investigate whether hormone disruptors may also interfere directly or indirectly with the development or functioning of the nervous system through either a neuroendocrine or a more general mechanism, we examined the scientific literature to ascertain the effects of EDs on the nervous system, specifically in the categories of neurotoxicity, cognition, and behaviour. To date, we demonstrated that all of the 177 EDs identified internationally by WHO are known to have an impact on the nervous system. Furthermore, the precise mechanisms underlying this neurodisruption have also been established. It was previously believed that EDs primarily function via the thyroid. However, this study presents substantial evidence that approximately 80 % of EDs operate via other mechanisms. It thus outlines a novel concept: EDs are also neurodisruptors (NDs) and can be collectively termed endocrine and nervous disruptors (ENDs). Most of ENDs are derived from petroleum residues, and their various mechanisms of action are similar to those of "spam" in electronic communications technologies. Therefore, ENDs can be considered as an instance of spam in a biological context.
Collapse
Affiliation(s)
- Gilles-Eric Seralini
- University of Caen Normandy, Network on Risks, Quality and Sustainable Development, Faculty of Sciences, Esplanade de la Paix, 14032, Caen, France
| | - Gerald Jungers
- University of Caen Normandy, Network on Risks, Quality and Sustainable Development, Faculty of Sciences, Esplanade de la Paix, 14032, Caen, France
| |
Collapse
|
7
|
Song J, Li Y, Zhao C, Zhou Q, Zhang J. Interaction of BDE-47 with nuclear receptors (NRs) based on the cytotoxicity: In vitro investigation and molecular interaction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111390. [PMID: 33049448 DOI: 10.1016/j.ecoenv.2020.111390] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/03/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are endocrine-disrupting chemicals that possess neuroendocrine and reproductive toxicity to humans and disturb thyroid hormone homeostasis, neurobehavior, and development. The most predominant congener of PBDEs in humans and other organisms is 2,2',4,4'-tetrabromodiphenyl ether (BDE-47); however, the molecular mechanisms underlying its cytotoxicity remain largely unknown. Here, we evaluated the toxic effect and underlying mechanism of nuclear receptors (NRs) induced by BDE-47 in SK-N-SH human neuroblastoma cells. The CCK-8 cell viability assay showed that the proliferation of human SK-N-SH cells exposed to BDE-47 was significantly inhibited in time- and dose-dependent manners, and flow cytometry showed that cell cycle was arrested at the S phase after BDE-47 exposure. Moreover, compared with the control group, the expression of retinoic acid receptor alpha (RXRα), pregnane X receptor (PXR), thyroid hormone receptors (TRs), and peroxisome proliferator-activated receptors (PPARs) at the mRNA and protein levels was significantly increased, as determined by quantitative PCR and western blot analysis, demonstrating that BDE-47 activated the NRs in vitro. Moreover, BDE-47 could bind to all four NRs in the affinity order of PPARγ > PXR > TRβ > RXRα under molecular dynamics. Because RXR is the promiscuous dimerization partner for a large number of NRs, ZDock was used to calculate its interaction with other three NRs. Taking the number of hydrogen bonds and ZDock scores into account, the rank of docking ability between RXRα and the NRs was PXR > TRβ > PPARγ. Further analysis of the interaction between BDE-47 and dimerized-NRs, the affinity order was RXRα > TRβ > PXR > PPARγ via Glide. The results of this study demonstrated that BDE-47 interfered the cross-talk among NRs, especially the promiscuous RXRα, which might be critical for the harmonized re-adjustment of cytotoxicity and biological regulation. Our findings provide a better understanding of the mechanisms underlying toxic effects and intermolecular interaction induced by BDE-47.
Collapse
Affiliation(s)
- Jiayi Song
- POPs Lab, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Yunxiu Li
- POPs Lab, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Chunyan Zhao
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jianqing Zhang
- POPs Lab, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China.
| |
Collapse
|
8
|
Berntsen HF, Duale N, Bjørklund CG, Rangel-Huerta OD, Dyrberg K, Hofer T, Rakkestad KE, Østby G, Halsne R, Boge G, Paulsen RE, Myhre O, Ropstad E. Effects of a human-based mixture of persistent organic pollutants on the in vivo exposed cerebellum and cerebellar neuronal cultures exposed in vitro. ENVIRONMENT INTERNATIONAL 2021; 146:106240. [PMID: 33186814 DOI: 10.1016/j.envint.2020.106240] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/25/2020] [Accepted: 10/22/2020] [Indexed: 06/11/2023]
Abstract
Exposure to persistent organic pollutants (POPs), encompassing chlorinated (Cl), brominated (Br) and perfluoroalkyl acid (PFAA) compounds is associated with adverse neurobehaviour in humans and animals, and is observed to cause adverse effects in nerve cell cultures. Most studies focus on single POPs, whereas studies on effects of complex mixtures are limited. We examined the effects of a mixture of 29 persistent compounds (Cl + Br + PFAA, named Total mixture), as well as 6 sub-mixtures on in vitro exposed rat cerebellar granule neurons (CGNs). Protein expression studies of cerebella from in vivo exposed mice offspring were also conducted. The selection of chemicals for the POP mixture was based on compounds being prominent in food, breast milk or blood from the Scandinavian human population. The Total mixture and sub-mixtures containing PFAAs caused greater toxicity in rat CGNs than the single or combined Cl/Br sub-mixtures, with significant impact on viability from 500x human blood levels. The potencies for these mixtures based on LC50 values were Br + PFAA mixture > Total mixture > Cl + PFAA mixture > PFAA mixture. These mixtures also accelerated induced lipid peroxidation. Protection by the competitive N-methyl-D-aspartate (NMDA) receptor antagonist 3-((R)-2-Carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP) indicated involvement of the NMDA receptor in PFAA and Total mixture-, but not Cl mixture-induced toxicity. Gene-expression studies in rat CGNs using a sub-toxic and marginally toxic concentration ((0.4 nM-5.5 µM) 333x and (1 nM-8.2 µM) 500x human blood levels) of the mixtures, revealed differential expression of genes involved in apoptosis, oxidative stress, neurotransmission and cerebellar development, with more genes affected at the marginally toxic concentration. The two important neurodevelopmental markers Pax6 and Grin2b were downregulated at 500x human blood levels, accompanied by decreases in PAX6 and GluN2B protein levels, in cerebellum of offspring mice from mothers exposed to the Total mixture throughout pregnancy and lactation. In rat CGNs, the glutathione peroxidase gene Prdx6 and the regulatory transmembrane glycoprotein gene Sirpa were highly upregulated at both concentrations. In conclusion, our results support that early-life exposure to mixtures of POPs can cause adverse neurodevelopmental effects.
Collapse
Affiliation(s)
- Hanne Friis Berntsen
- Department of Production Animal Clinical Sciences, NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102 Oslo, Norway; National Institute of Occupational Health, P.O. Box 5330 Majorstuen, 0304 Oslo, Norway.
| | - Nur Duale
- Section of Molecular Toxicology, Norwegian Institute of Public Health, N-0403 Oslo, Norway.
| | - Cesilie Granum Bjørklund
- Department of Production Animal Clinical Sciences, NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102 Oslo, Norway.
| | | | - Kine Dyrberg
- Department of Production Animal Clinical Sciences, NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102 Oslo, Norway.
| | - Tim Hofer
- Section of Toxicology and Risk Assessment, Norwegian Institute of Public Health, N-0403, Oslo, Norway.
| | - Kirsten Eline Rakkestad
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1072, Blindern, NO-0316 Oslo, Norway.
| | - Gunn Østby
- Department of Production Animal Clinical Sciences, NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102 Oslo, Norway.
| | - Ruth Halsne
- Department of Production Animal Clinical Sciences, NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102 Oslo, Norway.
| | - Gudrun Boge
- Department of Companion Animal Clinical Sciences, NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102 Oslo, Norway.
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1072, Blindern, NO-0316 Oslo, Norway.
| | - Oddvar Myhre
- Section of Toxicology and Risk Assessment, Norwegian Institute of Public Health, N-0403, Oslo, Norway.
| | - Erik Ropstad
- Department of Production Animal Clinical Sciences, NMBU-School of Veterinary Science, P.O. Box 369 sentrum, N-0102 Oslo, Norway.
| |
Collapse
|
9
|
Wnuk A, Rzemieniec J, Przepiórska K, Wesołowska J, Wójtowicz AK, Kajta M. Autophagy-related neurotoxicity is mediated via AHR and CAR in mouse neurons exposed to DDE. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 742:140599. [PMID: 32721735 DOI: 10.1016/j.scitotenv.2020.140599] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/25/2020] [Accepted: 06/27/2020] [Indexed: 06/11/2023]
Abstract
DDE (dichlorodiphenyldichloroethylene) is an environmental metabolite of the pesticide DDT, which is still present in the environment, and its insecticidal properties are used to fight malaria and the Zika virus disease. We showed for the first time that the neurotoxic effects of DDE involve autophagy, as demonstrated by elevated levels of Becn1, Map1lc3a/MAP1LC3A, Map1lc3b, and Nup62/NUP62 and an increase in autophagosome formation. The suggestion that the aryl hydrocarbon receptor (AHR) and the constitutive androstane receptor (CAR) are involved in the neurotoxic effect of DDE was supported by increases in the mRNA and protein expression of these receptors, as detected by qPCR, ELISA, immunofluorescence labeling and confocal microscopy. Selective antagonists of the receptors, including alpha-naphthoflavone, CH223191, and CINPA 1, inhibited p,p'-DDE- and o,p'-DDE-induced LDH release and caspase-3 activity, while specific siRNAs (Ahr and Car siRNA) reduced the levels of p,p'-DDE- and o,p'-DDE-induced autophagosome formation. Although the neurotoxic effects of DDE were isomer independent, the mechanisms of p,p'- and o,p'-DDE were isomer specific. Therefore, we identified previously unknown mechanisms of the neurotoxic actions of DDE that, in addition to inducing apoptosis, stimulate autophagy in mouse neocortical cultures and induce AHR and CAR signaling.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna street 12, 31-343 Krakow, Poland
| | - Joanna Rzemieniec
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna street 12, 31-343 Krakow, Poland
| | - Karolina Przepiórska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna street 12, 31-343 Krakow, Poland
| | - Julita Wesołowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Laboratory for In vivo and In Vitro Imaging, Smetna street 12, 31-343 Krakow, Poland
| | - Anna Katarzyna Wójtowicz
- University of Agriculture, Faculty of Animal Sciences, Department of Nutrition, Animal Biotechnology and Fisheries, Adama Mickiewicza 24/28, 30-059 Krakow, Poland
| | - Małgorzata Kajta
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna street 12, 31-343 Krakow, Poland.
| |
Collapse
|
10
|
Tao J, Bai C, Chen Y, Zhou H, Liu Y, Shi Q, Pan W, Dong H, Li L, Xu H, Tanguay R, Huang C, Dong Q. Environmental relevant concentrations of benzophenone-3 induced developmental neurotoxicity in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 721:137686. [PMID: 32169642 DOI: 10.1016/j.scitotenv.2020.137686] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/04/2020] [Accepted: 03/01/2020] [Indexed: 06/10/2023]
Abstract
Benzophenone-3 (BP3 or oxybenzone) is an organic UV filter that has been widely used in personal care products. Its frequent detection in the environment and humans as well as its structural similarity to estradiol have prompted most research focus on its endocrine effect. However, these effects are usually associated with concentrations 10-100 fold higher than its environmental relevant concentrations. Few studies explore its adverse effects at environmental relevant concentrations. In the present study, we evaluated the developmental neurotoxic (DNT) effects of low concentration BP3 exposure during a sensitive developmental window in zebrafish. Our findings revealed that BP3 exposure at 10 μg/L (0.04 μM) during 6-24 h post fertilization (hpf) led to various DNT effects such as increased spontaneous movement at 21 and 24 hpf, decreased touch response at 27 hpf, heightened hyperactivity in locomotor response at 5 day post fertilization (dpf), decreased shoaling behavior at 11 dpf and decreased mirror attacks at 12 dpf. These effects were accompanied with decreased axonal growth at 27 hpf, decreased cell proliferation and increased cell apoptosis in the head region of larval zebrafish immediately after BP3 exposure at 24 hpf, and increased expression of retinoid X receptor gene rxrgb at 5 dpf. Interestingly, rxrgb knockdown through morpholino injection largely restored most of BP3-induced DNT effects, axonal growth delay, cell proliferation and cell apoptosis, suggesting that BP3-induced DNT effects are likely mediated through the Rxrgb receptor. In considering with recent findings on the endocrine effects of BP3, we conclude that BP3 at environmental relevant concentrations has limited estrogenic effect, but is neurotoxic to developing embryos in zebrafish.
Collapse
Affiliation(s)
- Junyan Tao
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chenglian Bai
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yuanhong Chen
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Huanhuan Zhou
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yahui Liu
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Qingyu Shi
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Wenhao Pan
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Haojia Dong
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Luyi Li
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Hui Xu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Robyn Tanguay
- Sinnhuber Aquatic Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States of America
| | - Changjiang Huang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China.
| | - Qiaoxiang Dong
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China; The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
11
|
The neuroprotective action of 3,3'-diindolylmethane against ischemia involves an inhibition of apoptosis and autophagy that depends on HDAC and AhR/CYP1A1 but not ERα/CYP19A1 signaling. Apoptosis 2020; 24:435-452. [PMID: 30778709 PMCID: PMC6522467 DOI: 10.1007/s10495-019-01522-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There are no studies examining the effects of 3,3′-diindolylmethane (DIM) in neuronal cells subjected to ischemia. Little is also known about the roles of apoptosis and autophagy as well as AhR and ERα signaling and HDACs in DIM action. We demonstrated for the first time the strong neuroprotective capacity of DIM in mouse primary hippocampal cell cultures exposed to ischemia at early and later stages of neuronal development. The protective effects of DIM were mediated via inhibition of ischemia-induced apoptosis and autophagy that was accompanied by a decrease in AhR/CYP1A1 signaling and an increase in HDAC activity. DIM decreased the levels of pro-apoptotic factors, i.e., Fas, Caspase-3, and p38 mitogen-activated protein kinase (MAPK). DIM also reduced the protein levels of autophagy-related Beclin-1 (BECN1) and microtubule-associated proteins 1A/1B light chain (LC3), partially reversed the ischemia-induced decrease in Nucleoporin 62 (NUP62) and inhibited autophagosome formation. In addition, DIM completely reversed the ischemia-induced decrease in histone deacetylase (HDAC) activity in hippocampal neurons. Although DIM inhibited AhR/CYP1A1 signaling, it did not influence the protein expression levels of ERα and ERα-regulated CYP19A1 which are known to be controlled by AhR. This study demonstrated for the first time, that the neuroprotective action of 3,3′-diindolylmethane against ischemia involves an inhibition of apoptosis and autophagy and depends on AhR/CYP1A1 signaling and HDAC activity, thus creating the possibility of developing new therapeutic strategies that target neuronal degeneration at specific molecular levels.
Collapse
|
12
|
Duparc S, Chalon S, Miller S, Richardson N, Toovey S. Neurological and psychiatric safety of tafenoquine in Plasmodium vivax relapse prevention: a review. Malar J 2020; 19:111. [PMID: 32169086 PMCID: PMC7071640 DOI: 10.1186/s12936-020-03184-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/06/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Tafenoquine is an 8-aminoquinoline anti-malarial drug recently approved as a single-dose (300 mg) therapy for Plasmodium vivax relapse prevention, when co-administered with 3-days of chloroquine or other blood schizonticide. Tafenoquine 200 mg weekly after a loading dose is also approved as travellers' prophylaxis. The development of tafenoquine has been conducted over many years, using various dosing regimens in diverse populations. METHODS This review brings together all the preclinical and clinical data concerning tafenoquine central nervous system safety. Data were assembled from published sources. The risk of neuropsychiatric adverse events (NPAEs) with single-dose tafenoquine (300 mg) in combination with chloroquine to achieve P. vivax relapse prevention is particularly examined. RESULTS There was no evidence of neurotoxicity with tafenoquine in preclinical animal models. In clinical studies in P. vivax relapse prevention, nervous system adverse events, mainly headache and dizziness, occurred in 11.4% (36/317) of patients with tafenoquine (300 mg)/chloroquine versus 10.2% (19/187) with placebo/chloroquine; and in 15.5% (75/483) of patients with tafenoquine/chloroquine versus 13.3% (35/264) with primaquine (15 mg/day for 14 days)/chloroquine. Psychiatric adverse events, mainly insomnia, occurred in 3.8% (12/317) of patients with tafenoquine/chloroquine versus 2.7% (5/187) with placebo/chloroquine; and in 2.9% (14/483) of patients with tafenoquine/chloroquine versus 3.4% (9/264) for primaquine/chloroquine. There were no serious or severe NPAEs observed with tafenoquine (300 mg)/chloroquine in these studies. CONCLUSIONS The risk:benefit of single-dose tafenoquine/chloroquine in P. vivax relapse prevention is favourable in the presence of malaria, with a low risk of NPAEs, similar to that seen with chloroquine alone or primaquine/chloroquine.
Collapse
Affiliation(s)
- Stephan Duparc
- Medicines for Malaria Venture, Route de Pré-Bois 20, 1215, Geneva 15, Switzerland.
| | - Stephan Chalon
- Medicines for Malaria Venture, Route de Pré-Bois 20, 1215, Geneva 15, Switzerland
| | | | | | - Stephen Toovey
- Medicines for Malaria Venture, Route de Pré-Bois 20, 1215, Geneva 15, Switzerland.,Pegasus Research, London, UK
| |
Collapse
|
13
|
Morales-Prieto N, López de Lerma N, Pacheco IL, Huertas-Abril PV, Pérez J, Peinado R, Abril N. Protective effect of Pedro-Ximénez must against p,p'-DDE-induced liver damages in aged Mus spretus mice. Food Chem Toxicol 2020; 136:110984. [PMID: 31765701 DOI: 10.1016/j.fct.2019.110984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/14/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
|
14
|
Kajta M, Rzemieniec J, Wnuk A, Lasoń W. Triclocarban impairs autophagy in neuronal cells and disrupts estrogen receptor signaling via hypermethylation of specific genes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 701:134818. [PMID: 31706213 DOI: 10.1016/j.scitotenv.2019.134818] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 05/20/2023]
Abstract
Although an increasing body of evidence suggests that triclocarban, a phenyl ether classified as a contaminant of emerging concern, presents a risk to development, there is limited data available on the potential interplay of triclocarban with the developing mammalian nervous system. This study was aimed to investigate the impact of environmentally pervasive chemical triclocarban on autophagy and estrogen receptor-mediated signaling pathways in mouse neurons. The study showed that triclocarban impaired autophagy and disrupted estrogen receptor signaling in mouse embryonic neurons in primary culture. Triclocarban used at environmentally relevant concentrations inhibited the mRNA and protein expression of ESR1 and GPER1 but not ESR2. The triclocarban-induced decrease in the expression of estrogen receptors was supported by the colocalization of the receptors in mouse neurons and corresponded to hypermethylation of the Esr1 and Gper1 genes. Selective antagonists increased the effects of triclocarban, which suggests that the neurotoxic effects of triclocarban, in addition to decreasing estrogen receptor expression, are mediated via inhibition of the neuroprotective capacity of the receptors. Furthermore, Becn1 and Atg7 siRNAs potentiated the caspase-3-dependent effect of triclocarban, which points to triclocarban-induced impairment of autophagy. Indeed, triclocarban dysregulated the expression of autophagy-related genes, and caused a time-dependent inhibition of the mRNA expression of Becn1, Map1lc3a, Map1lc3b, Nup62, and Atg7, which was correlated with a decrease in the protein levels of MAP1LC3B, BECN1 and autophagosomes, but not NUP62 protein level which was increased. Intriguingly, the Esr1 and Gper1 siRNAs did not affect the level of autophagosomes, suggesting that the triclocarban-induced impairment of autophagy is independent of the triclocarban-induced disruption of estrogen receptor signaling in mammalian neurons. Because our data provided evidence that triclocarban has the capacity to impair autophagy and disrupt estrogen receptor signaling in brain neurons at an early developmental stage, we postulate to categorize the compound as a neurodevelopmental risk factor.
Collapse
Affiliation(s)
- M Kajta
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland.
| | - J Rzemieniec
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland
| | - A Wnuk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland
| | - W Lasoń
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland
| |
Collapse
|
15
|
Rodríguez-Moro G, Abril N, Jara-Biedma R, Ramírez-Acosta S, Gómez-Ariza JL, García-Barrera T. Metabolic Impairments Caused by a "Chemical Cocktail" of DDE and Selenium in Mice Using Direct Infusion Triple Quadrupole Time-of-Flight and Gas Chromatography-Mass Spectrometry. Chem Res Toxicol 2019; 32:1940-1954. [PMID: 31532635 DOI: 10.1021/acs.chemrestox.9b00102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Among organic contaminants, pesticides are one of the most important groups of chemicals due to their persistent character and toxicity. However, the biological systems are exposed to a complex environment in which the contaminants can interact in a synergistic/antagonistic fashion, and for this reason, the study of "chemical cocktails" is of great interest to fully understand the final biological effect. In this way, selenium is known for its antagonistic action against several toxicants. In this paper, metabolic impairments caused by the joint exposure of p,p'-dichloro diphenyl trichloroethane (DDE) and selenium (Se) have been issued for the first time. A metabolomic workflow was applied to mice fed DDE and DDE with Se diet, on the basis of the complementary use of two organic mass spectrometric techniques, combining direct infusion mass spectrometry (DI-ESI-QqQ-TOF MS) and gas chromatography-mass spectrometry (GC-MS). The results show a good classification between the studied groups caused by about 70 altered metabolites in the liver, kidney, or brain, including the pathways of energy metabolism, degradation of phospholipidic membrane, β-oxidation, and oxidative stress, which confirm the potential of combined metabolomic platforms in environmental studies.
Collapse
Affiliation(s)
| | - Nieves Abril
- Department of Biochemistry and Molecular Biology, International Agrofood Campus of Excellence International ceiA3 , University of Córdoba , Campus de Rabanales, Edificio Severo Ochoa , E-14071 Córdoba , Spain
| | | | | | | | | |
Collapse
|
16
|
Ibañez HC, Melanda VS, Gerber VKQ, Licht OAB, Ibañez MVC, Aguiar Júnior TR, Mello RG, Komechen H, Andrade DP, Picharski GL, Figueiredo DPG, Pianovski MAD, Figueiredo MMO, Custódio G, Parise IZS, Castilho LM, Paraizo MM, Edinger C, Fiori CMCM, Pedrini H, Kiesel Filho N, Fabro ALMR, Fachin RD, Ogradowski KRP, Parise GA, Saldiva PHN, Legal EF, Rosati R, Rodriguez-Galindo C, Ribeiro RC, Zambetti GP, Lalli E, Figueiredo BC. Spatial trends in congenital malformations and stream water chemistry in Southern Brazil. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 650:1278-1291. [PMID: 30308815 DOI: 10.1016/j.scitotenv.2018.09.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/17/2018] [Accepted: 09/04/2018] [Indexed: 06/08/2023]
Abstract
The incidence of variable congenital malformation (CM) among 399 municipalities in the state of Paraná, southern Brazil, suggests the etiological role of environmental factors. This study examined a) environmental concentrations of chlorine anions (Cl-) associated with organochlorines (OCs) and b) associations between these chemicals and agricultural output with CMs using a geographical information system. In one of the three years during the sampling period (2008, 2009 or 2010) Cl-, dichlorodiphenyltrichloroethane (p,p'-DDT), dichlorodiphenyldichloroethylene (p,p'-DDE), dichlorodiphenyldichloroethane (p,p'-DDD), and endosulfan levels were measured in 465 (465/736, 63%) catchment basins. Agricultural outputs for crops during 2006-2010 were also evaluated (t/km2). Further, CM kernel density for the 399 municipalities in Paraná during 2007-2014 was investigated. Cl- levels increased significantly in one of the three years (2008, 2009 or 2010) in western catchment basins, compared to 1996 (p < 0.0001). The municipalities were divided according to the obtained Cl- levels, where sub-region C2 (central-southern) < 1.8 mg/L ≤ sub-regions C1 (northern-western) and C3 (eastern-southern). We identified 8756 cases of CMs among 1,221,287 newborns (NB) in all sub-regions. C1 had higher DDT-DDE-DDD (p,p'-DDT + p,p'-DDE + p,p'-DDD) concentrations, agricultural output, and CM kernel density. C2 and C3 had minor agricultural outputs (per square kilometer) and CM densities. A 2.96 mg/L increase in Cl- between sub-regions C1 and C2 was co-localized with a 45% increase in CM density (spatial relative risk = 1.45, CI 95%: 1.36-1.55). C1 had the highest log likelihood ratios (p = 0.001) identified via SaTScan clustering analyses. Organochlorines and other toxic chlorinated chemicals may contribute to CMs in humans, and these chemicals are ultimately transformed and release Cl- in rivers. Higher Cl- levels were correlated significantly with higher agricultural productivity, DDT-DDE-DDD levels, and CMs in some parts of the northern and western sub-regions (C1).
Collapse
Affiliation(s)
- Humberto C Ibañez
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | - Viviane S Melanda
- Departamento de Vigilância Epidemiológica, Secretaria do Estado da Saúde do Paraná, Curitiba, PR, Brazil
| | - Viviane K Q Gerber
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | - Otavio A B Licht
- Instituto de Terras Cartografia e Geologia, Curitiba, PR, Brazil
| | | | - Terêncio R Aguiar Júnior
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Rosiane G Mello
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | - Heloisa Komechen
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Diancarlos P Andrade
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | - Gledson L Picharski
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | - Damasio P G Figueiredo
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Mara A D Pianovski
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Universidade Federal do Paraná, Curitiba, PR, Brazil; Erasto Gaertner Hospital, Curitiba, PR, Brazil
| | - Mirna M O Figueiredo
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Gislaine Custódio
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Ivy Z S Parise
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | | | - Mariana M Paraizo
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | - Chloe Edinger
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil
| | - Carmem M C M Fiori
- União Oeste Paranaense de Estudos e Combate ao Câncer - UOPECCAN, Cascavel, PR, Brazil; Universidade Estadual do Oeste do Paraná, Cascavel, PR, Brazil
| | - Hélio Pedrini
- Instituto de Computação, Universidade de Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Ana Luiza M R Fabro
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Hospital Pequeno Príncipe, Curitiba, PR, Brazil
| | - Rayssa D Fachin
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil
| | - Karin R P Ogradowski
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | - Guilherme A Parise
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Paulo H N Saldiva
- Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Roberto Rosati
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil
| | - Carlos Rodriguez-Galindo
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Raul C Ribeiro
- Faculdades Pequeno Principe, Curitiba, PR, Brazil; Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gerard P Zambetti
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, 660 route des Lucioles, Sophia Antipolis, Valbonne, France
| | - Bonald C Figueiredo
- Pelé Pequeno Príncipe Research Institute, Curitiba, PR, Brazil; Faculdades Pequeno Principe, Curitiba, PR, Brazil; Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Universidade Federal do Paraná, Curitiba, PR, Brazil; Departamento de Saúde Coletiva, Universidade Federal do Paraná, Curitiba, PR, Brazil.
| |
Collapse
|
17
|
Prenatal Exposure to Benzophenone-3 Impairs Autophagy, Disrupts RXRs/PPARγ Signaling, and Alters Epigenetic and Post-Translational Statuses in Brain Neurons. Mol Neurobiol 2018; 56:4820-4837. [PMID: 30402708 PMCID: PMC6647400 DOI: 10.1007/s12035-018-1401-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 10/17/2018] [Indexed: 02/07/2023]
Abstract
The UV absorber benzophenone-3 (BP-3) is the most extensively used chemical substance in various personal care products. Despite that BP-3 exposure is widespread, knowledge about the impact of BP-3 on the brain development is negligible. The present study aimed to explore the mechanisms of prenatal exposure to BP-3 in neuronal cells, with particular emphasis on autophagy and nuclear receptors signaling as well as the epigenetic and post-translational modifications occurring in response to BP-3. To observe the impact of prenatal exposure to BP-3, we administered BP-3 to pregnant mice, and next, we isolated brain tissue from pretreated embryos for primary cell neocortical culture. Our study revealed that prenatal exposure to BP-3 (used in environmentally relevant doses) impairs autophagy in terms of BECLIN-1, MAP1LC3B, autophagosomes, and autophagy-related factors; disrupts the levels of retinoid X receptors (RXRs) and peroxisome proliferator-activated receptor gamma (PPARγ); alters epigenetic status (i.e., attenuates HDAC and sirtuin activities); inhibits post-translational modifications in terms of global sumoylation; and dysregulates expression of neurogenesis- and neurotransmitter-related genes as well as miRNAs involved in pathologies of the nervous system. Our study also showed that BP-3 has good permeability through the BBB. We strongly suggest that BP-3-evoked effects may substantiate a fetal basis of the adult onset of neurological diseases, particularly schizophrenia and Alzheimer’s disease.
Collapse
|
18
|
Wnuk A, Rzemieniec J, Litwa E, Lasoń W, Kajta M. Prenatal exposure to benzophenone-3 (BP-3) induces apoptosis, disrupts estrogen receptor expression and alters the epigenetic status of mouse neurons. J Steroid Biochem Mol Biol 2018; 182:106-118. [PMID: 29704544 DOI: 10.1016/j.jsbmb.2018.04.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 12/31/2022]
Abstract
Current evidence indicates that benzophenone-3 (BP-3) can pass through the placental and blood-brain barriers and thus can likely affect infant neurodevelopment. Despite widespread exposure, data showing the effects of BP-3 on the developing nervous system are scarce. This study revealed for the first time that prenatal exposure to BP-3 led to apoptosis and neurotoxicity, altered the levels of estrogen receptors (ERs) and changed the epigenetic status of mouse neurons. In the present study, subcutaneous injections of pregnant mice with BP-3 at 50 mg/kg, which is an environmentally relevant dose, evoked activation of caspase-3 and lactate dehydrogenase (LDH) release as well as substantial loss of mitochondrial membrane potential in neocortical cells of their embryonic offspring. Apoptosis-focused microarray analysis of neocortical cells revealed up-regulation of 22 genes involved in apoptotic cell death. This effect was supported by increased BAX and CASP3 mRNA and protein levels, as evidenced by qPCR, ELISAs and western blots. BP-3-induced apoptosis and neurotoxicity were accompanied by decreases in the mRNA and protein expression levels of ESR1 and ESR2 (also known as ERα and ERβ), with a simultaneous increase in GPER1 (also known as GPR30) expression. In addition to the demonstration that treatment of pregnant mice with BP-3 induced apoptosis, caused neurotoxicity and altered ERs expression levels in neocortical cells of their embryonic offspring, we showed that prenatal administration of BP-3 inhibited global DNA methylation as well as reduced DNMTs activity. BP-3 also caused specific hypomethylation of the genes Gper1 and Bax, an effect that was accompanied by increased mRNA and protein expression levels. In addition, BP-3 caused hypermethylation of the genes Esr1, Esr2 and Bcl2, which could explain the reduced mRNA and protein levels of the estrogen receptors. This study demonstrated for the first time that prenatal exposure to BP-3 caused severe neuronal apoptosis that was accompanied by impaired ESR1/ESR2 expression, enhanced GPER1 expression, global DNA hypomethylation and altered methylation statuses of apoptosis-related and ERs genes. We suggest that the effects of BP-3 in embryonic neurons may be the fetal basis of the adult onset of nervous system disease.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Joanna Rzemieniec
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Ewa Litwa
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Władysław Lasoń
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Małgorzata Kajta
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland.
| |
Collapse
|
19
|
Watanabe M, Kakuta H. Retinoid X Receptor Antagonists. Int J Mol Sci 2018; 19:ijms19082354. [PMID: 30103423 PMCID: PMC6121510 DOI: 10.3390/ijms19082354] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022] Open
Abstract
Retinoid X receptor (RXR) antagonists are not only useful as chemical tools for biological research, but are also candidate drugs for the treatment of various diseases, including diabetes and allergies, although no RXR antagonist has yet been approved for clinical use. In this review, we present a brief overview of RXR structure, function, and target genes, and describe currently available RXR antagonists, their structural classification, and their evaluation, focusing on the latest research.
Collapse
Affiliation(s)
- Masaki Watanabe
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|
20
|
Kajta M, Wnuk A, Rzemieniec J, Lason W, Mackowiak M, Chwastek E, Staniszewska M, Nehring I, Wojtowicz AK. Triclocarban Disrupts the Epigenetic Status of Neuronal Cells and Induces AHR/CAR-Mediated Apoptosis. Mol Neurobiol 2018; 56:3113-3131. [PMID: 30097849 PMCID: PMC6476872 DOI: 10.1007/s12035-018-1285-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
Triclocarban is a phenyl ether that has recently been classified as a contaminant of emerging concern. Evidence shows that triclocarban is present in human tissues, but little is known about the impact of triclocarban on the nervous system, particularly at early developmental stages. This study demonstrated that triclocarban that was used at environmentally relevant concentrations induced apoptosis in mouse embryonic neurons, inhibited sumoylation, and changed the epigenetic status, as evidenced by impaired activities of HDAC, sirtuins, and DNMT, global DNA hypomethylation, and alterations of methylation levels of bax, bcl2, Ahr, and Car genes. The use of selective antagonists and specific siRNAs, which was followed by the co-localization of aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR) in mouse neurons, points to the involvement of AHR and CAR in triclocarban-induced neurotoxicity. A 24-h treatment with triclocarban enhanced protein levels of the receptors which was paralleled by Car hypomethylation and Ahr hypermethylation. Car hypomethylation is in line with global DNA hypomethylation and explains the increased mRNA and protein levels of CAR in response to triclocarban. Ahr hypermethylation could reflect reduced Ahr mRNA expression and corresponds to lowered protein levels after 3- and 6-h exposures to triclocarban that is likely related to proteasomal degradation of activated AHR. We hypothesize that the triclocarban-induced apoptosis in mouse neurons and the disruption of epigenetic status involve both AHR- and CAR-mediated effects, which may substantiate a fetal basis of the adult onset of neurological diseases; however, the expression of the receptors is regulated in different ways.
Collapse
Affiliation(s)
- M Kajta
- Institute of Pharmacology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland.
| | - A Wnuk
- Institute of Pharmacology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - J Rzemieniec
- Institute of Pharmacology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - W Lason
- Institute of Pharmacology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - M Mackowiak
- Institute of Pharmacology, Department of Pharmacology, Laboratory of Brain Biostructure, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - E Chwastek
- Department of Cell Biology and Imaging, Confocal Microscopy Laboratory, Institute of Zoology, Jagiellonian University, Gronostajowa Street 9, 30-387, Krakow, Poland
| | - M Staniszewska
- Institute of Oceanography, University of Gdansk, Al. Marszałka Piłsudskiego 46, 81-378, Gdynia, Poland
| | - I Nehring
- Institute of Oceanography, University of Gdansk, Al. Marszałka Piłsudskiego 46, 81-378, Gdynia, Poland
| | - A K Wojtowicz
- Department of Animal Biotechnology, Faculty of Animal Sciences, University of Agriculture, Redzina Street 1B, 30-248, Krakow, Poland
| |
Collapse
|
21
|
Morales-Prieto N, Ruiz-Laguna J, Sheehan D, Abril N. Transcriptome signatures of p,p´-DDE-induced liver damage in Mus spretus mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 238:150-167. [PMID: 29554563 DOI: 10.1016/j.envpol.2018.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/19/2018] [Accepted: 03/04/2018] [Indexed: 06/08/2023]
Abstract
The use of DDT (1,1,1-trichloro-2,2-bis(p-chlorophenyl) ethane) in some countries, although regulated, is contributing to an increased worldwide risk of exposure to this organochlorine pesticide or its derivative p,p'-DDE [1,1-dichloro-2,2-bis(p-chlorophenyl) ethylene]. Many studies have associated p,p'-DDE exposure to type 2 diabetes, obesity and alterations of the reproductive system, but their molecular mechanisms of toxicity remain poorly understood. We have addressed this issue by using commercial microarrays based on probes for the entire Mus musculus genome to determine the hepatic transcriptional signatures of p,p'-DDE in the phylogenetically close mouse species Mus spretus. High-stringency hybridization conditions and analysis assured reliable results, which were also verified, in part, by qRT-PCR, immunoblotting and/or enzymatic activity. Our data linked 198 deregulated genes to mitochondrial dysfunction and perturbations of central signaling pathways (kinases, lipids, and retinoic acid) leading to enhanced lipogenesis and aerobic glycolysis, inflammation, cell proliferation and testosterone catabolism and excretion. Alterations of transcript levels of genes encoding enzymes involved in testosterone catabolism and excretion would explain the relationships established between p,p´-DDE exposure and reproductive disorders, obesity and diabetes. Further studies will help to fully understand the molecular basis of p,p´-DDE molecular toxicity in liver and reproductive organs, to identify effective exposure biomarkers and perhaps to design efficient p,p'-DDE exposure counteractive strategies.
Collapse
Affiliation(s)
- Noelia Morales-Prieto
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - Julia Ruiz-Laguna
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - David Sheehan
- College of Arts and Science, Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain.
| |
Collapse
|
22
|
Leung YK, Ouyang B, Niu L, Xie C, Ying J, Medvedovic M, Chen A, Weihe P, Valvi D, Grandjean P, Ho SM. Identification of sex-specific DNA methylation changes driven by specific chemicals in cord blood in a Faroese birth cohort. Epigenetics 2018; 13:290-300. [PMID: 29560787 DOI: 10.1080/15592294.2018.1445901] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Faroe islanders consume marine foods contaminated with methylmercury (MeHg), polychlorinated biphenyls (PCBs), and other toxicants associated with chronic disease risks. Differential DNA methylation at specific CpG sites in cord blood may serve as a surrogate biomarker of health impacts from chemical exposures. We aimed to identify key environmental chemicals in cord blood associated with DNA methylation changes in a population with elevated exposure to chemical mixtures. We studied 72 participants of a Faroese birth cohort recruited between 1986 and 1987 and followed until adulthood. The cord blood DNA methylome was profiled using Infinium HumanMethylation450 BeadChips. We determined the associations of CpG site changes with concentrations of MeHg, major PCBs, other organochlorine compounds [hexachlorobenzene (HCB), p,p'-dichlorodiphenyldichloroethylene (p,p'-DDE) and p,p'-dichlorodiphenyltrichloroethane], and perfluoroalkyl substances. In a combined sex analysis, among the 16 chemicals studied, PCB congener 105 (CB-105) exposure was associated with the majority of differentially methylated CpG sites (214 out of a total of 250). In female-only analysis, only 73 CB-105 associated CpG sites were detected, 44 of which were mapped to genes in the ELAV1-associated cancer network. In males-only, methylation changes were seen for perfluorooctane sulfonate, HCB, and p,p'-DDE in 10,598, 1,238, and 1,473 CpG sites, respectively, 15% of which were enriched in cytobands of the X-chromosome associated with neurological disorders. In this multiple-pollutant and genome-wide study, we identified key epigenetic toxicants. The significant enrichment of specific X-chromosome sites in males implies potential sex-specific epigenome responses to prenatal chemical exposures.
Collapse
Affiliation(s)
- Yuet-Kin Leung
- a Division of Environmental Genetics and Molecular Toxicology.,e Center of Environmental Genetics.,f Cincinnati Cancer Center , University of Cincinnati Medical Center , Cincinnati , USA
| | - Bin Ouyang
- a Division of Environmental Genetics and Molecular Toxicology.,e Center of Environmental Genetics
| | - Liang Niu
- b Biostatistics & Bioinformatics.,e Center of Environmental Genetics
| | - Changchun Xie
- b Biostatistics & Bioinformatics.,e Center of Environmental Genetics
| | - Jun Ying
- b Biostatistics & Bioinformatics.,c Public Health Science and
| | - Mario Medvedovic
- b Biostatistics & Bioinformatics.,e Center of Environmental Genetics.,f Cincinnati Cancer Center , University of Cincinnati Medical Center , Cincinnati , USA
| | - Aimin Chen
- d Epidemiology Department of Environmental Health.,e Center of Environmental Genetics
| | - Pal Weihe
- h Department of Occupational Medicine and Public Health , Faroese Hospital System , Torshavn , Faroe Islands
| | - Damaskini Valvi
- i Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , USA
| | - Philippe Grandjean
- i Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , USA.,j Department of Environmental Medicine , University of Southern Denmark , Odense , Denmark
| | - Shuk-Mei Ho
- a Division of Environmental Genetics and Molecular Toxicology.,e Center of Environmental Genetics.,f Cincinnati Cancer Center , University of Cincinnati Medical Center , Cincinnati , USA.,g Cincinnati Veteran Affairs Medical Center , Cincinnati , USA
| |
Collapse
|
23
|
Silencing carboxylesterase 1 in human THP-1 macrophages perturbs genes regulated by PPARγ/RXR and RAR/RXR: down-regulation of CYP27A1-LXRα signaling. Biochem J 2018; 475:621-642. [PMID: 29321244 DOI: 10.1042/bcj20180008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/04/2018] [Accepted: 01/09/2018] [Indexed: 02/07/2023]
Abstract
Macrophage foam cells store excess cholesterol as cholesteryl esters, which need to be hydrolyzed for cholesterol efflux. We recently reported that silencing expression of carboxylesterase 1 (CES1) in human THP-1 macrophages [CES1KD (THP-1 cells with CES1 expression knocked down) macrophages] reduced cholesterol uptake and decreased expression of CD36 and scavenger receptor-A in cells loaded with acetylated low-density lipoprotein (acLDL). Here, we report that CES1KD macrophages exhibit reduced transcription of cytochrome P45027A1 (CYP27A1) in nonloaded and acLDL-loaded cells. Moreover, levels of CYP27A1 protein and its enzymatic product, 27-hydroxycholesterol, were markedly reduced in CES1KD macrophages. Transcription of LXRα (liver X receptor α) and ABCA1 (ATP-binding cassette transporter A1) was also decreased in acLDL-loaded CES1KD macrophages, suggesting reduced signaling through PPARγ-CYP27A1-LXRα. Consistent with this, treatment of CES1KD macrophages with agonists for PPARγ, RAR, and/or RAR/RXR partially restored transcription of CYP27A1 and LXRα, and repaired cholesterol influx. Conversely, treatment of control macrophages with antagonists for PPARγ and/or RXR decreased transcription of CYP27A1 and LXRα Pharmacologic inhibition of CES1 in both wild-type THP-1 cells and primary human macrophages also decreased CYP27A1 transcription. CES1 silencing did not affect transcript levels of PPARγ and RXR in acLDL-loaded macrophages, whereas it did reduce the catabolism of the endocannabinoid 2-arachidonoylglycerol. Finally, the gene expression profile of CES1KD macrophages was similar to that of PPARγ knockdown cells following acLDL exposures, further suggesting a mechanistic link between CES1 and PPARγ. These results are consistent with a model in which abrogation of CES1 function attenuates the CYP27A1-LXRα-ABCA1 signaling axis by depleting endogenous ligands for the nuclear receptors PPARγ, RAR, and/or RXR that regulate cholesterol homeostasis.
Collapse
|
24
|
Rzemieniec J, Litwa E, Wnuk A, Lason W, Kajta M. Bazedoxifene and raloxifene protect neocortical neurons undergoing hypoxia via targeting ERα and PPAR-γ. Mol Cell Endocrinol 2018; 461:64-78. [PMID: 28859903 DOI: 10.1016/j.mce.2017.08.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/04/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022]
Abstract
Selective estrogen receptor modulators (SERMs) such as bazedoxifene and raloxifene are recognized to mainly act via estrogen receptors (ERs), but there is no study examining the involvement of PPAR-γ in their actions, especially in neurons undergoing hypoxia. Little is also known about age-dependent actions of the SERMs on neuronal tissue challenged with hypoxia. In this study, bazedoxifene and raloxifene protected neocortical cells against hypoxia at early and later developmental stages. Both SERMs evoked caspase-3-independent neuroprotection and increased protein levels of ERα (66 and 46 kDa isoforms) and PPAR-γ. In addition, bazedoxifene enhanced expression of ERα-regulated Cyp19a1 mRNA. Using double siRNA silencing, for the first time we demonstrated a key role of ERα and PPAR-γ in the neuroprotective action of the SERMs in neocortical neurons undergoing hypoxia. This study provides prospects for the development of a new therapeutic strategies against hypoxic brain injury that selectively target ERα and/or PPAR-γ.
Collapse
Affiliation(s)
- J Rzemieniec
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - E Litwa
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - A Wnuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - W Lason
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - M Kajta
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland.
| |
Collapse
|
25
|
Wnuk A, Kajta M. Steroid and Xenobiotic Receptor Signalling in Apoptosis and Autophagy of the Nervous System. Int J Mol Sci 2017; 18:ijms18112394. [PMID: 29137141 PMCID: PMC5713362 DOI: 10.3390/ijms18112394] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022] Open
Abstract
Apoptosis and autophagy are involved in neural development and in the response of the nervous system to a variety of insults. Apoptosis is responsible for cell elimination, whereas autophagy can eliminate the cells or keep them alive, even in conditions lacking trophic factors. Therefore, both processes may function synergistically or antagonistically. Steroid and xenobiotic receptors are regulators of apoptosis and autophagy; however, their actions in various pathologies are complex. In general, the estrogen (ER), progesterone (PR), and mineralocorticoid (MR) receptors mediate anti-apoptotic signalling, whereas the androgen (AR) and glucocorticoid (GR) receptors participate in pro-apoptotic pathways. ER-mediated neuroprotection is attributed to estrogen and selective ER modulators in apoptosis- and autophagy-related neurodegenerative diseases, such as Alzheimer’s and Parkinson’s diseases, stroke, multiple sclerosis, and retinopathies. PR activation appeared particularly effective in treating traumatic brain and spinal cord injuries and ischemic stroke. Except for in the retina, activated GR is engaged in neuronal cell death, whereas MR signalling appeared to be associated with neuroprotection. In addition to steroid receptors, the aryl hydrocarbon receptor (AHR) mediates the induction and propagation of apoptosis, whereas the peroxisome proliferator-activated receptors (PPARs) inhibit this programmed cell death. Most of the retinoid X receptor-related xenobiotic receptors stimulate apoptotic processes that accompany neural pathologies. Among the possible therapeutic strategies based on targeting apoptosis via steroid and xenobiotic receptors, the most promising are the selective modulators of the ER, AR, AHR, PPARγ agonists, flavonoids, and miRNAs. The prospective therapies to overcome neuronal cell death by targeting autophagy via steroid and xenobiotic receptors are much less recognized.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland.
| | - Małgorzata Kajta
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland.
| |
Collapse
|
26
|
Morales-Prieto N, Abril N. REDOX proteomics reveals energy metabolism alterations in the liver of M. spretus mice exposed to p, p'-DDE. CHEMOSPHERE 2017; 186:848-863. [PMID: 28826133 DOI: 10.1016/j.chemosphere.2017.08.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/27/2017] [Accepted: 08/11/2017] [Indexed: 06/07/2023]
Abstract
The toxicity induced by the pesticide 2,2-bis(p-chlorophenyl)-1,1,1,-trichloroethane (DDT) and its derivative 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (p,p'-DDE) has been associated with mitochondrial dysfunction, uncoupling of oxidative phosphorylation and respiratory chain electron transport, intracellular ion imbalance, generation of reactive oxygen species and impairment of the antioxidant defense system. A disruption in the cellular redox status causes protein Cys-based regulatory shifts that influence the activity of many proteins and trigger signal transduction alterations. Here, we analyzed the ability of p,p'-DDE to alter the activities of hepatic antioxidants and glycolytic enzymes to investigate the oxidative stress generation in the liver of p,p'-DDE-fed M. spretus mice. We also determined the consequences of the treatment on the redox status in the thiol Cys groups. The data indicate that the liver of p,p'-DDE exposed mice lacks certain protective enzymes, and p,p'-DDE caused a metabolic reprogramming that increased the glycolytic rate and disturbed the metabolism of lipids. Our results suggested that the overall metabolism of the liver was altered because important signaling pathways are controlled by p,p'-DDE-deregulated proteins. The histological data support the proposed metabolic consequences of the p,p'-DDE exposure.
Collapse
Affiliation(s)
- Noelia Morales-Prieto
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, España, Spain
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, España, Spain.
| |
Collapse
|
27
|
Wnuk A, Rzemieniec J, Lasoń W, Krzeptowski W, Kajta M. Benzophenone-3 Impairs Autophagy, Alters Epigenetic Status, and Disrupts Retinoid X Receptor Signaling in Apoptotic Neuronal Cells. Mol Neurobiol 2017; 55:5059-5074. [PMID: 28815487 PMCID: PMC5948252 DOI: 10.1007/s12035-017-0704-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/01/2017] [Indexed: 12/16/2022]
Abstract
Benzophenone-3 (BP-3) is the most widely used compound among UV filters for the prevention of photodegradation. Population studies have demonstrated that it penetrates through the skin and crosses the blood-brain barrier. However, little is known about the impact of BP-3 on the nervous system and its possible adverse effects on the developing brain. We demonstrated that the neurotoxic effects of BP-3 were accompanied by the induction of apoptosis, as evidenced by apoptosis-related caspase-3 activation and apoptotic body formation as well as the inhibition of autophagy, as determined by the downregulation of autophagy-related genes, decreased autophagosome formation, and reduced LC3B-to-LC3A ratio. In this study, we showed for the first time that the BP-3-induced apoptosis of neuronal cells is mediated via the stimulation of RXRα signaling and the attenuation of RXRβ/RXRγ signaling, as demonstrated using selective antagonist and specific siRNAs as well as by measuring the mRNA and protein expression levels of the receptors. This study also demonstrated that environmentally relevant concentrations of BP-3 were able to inhibit autophagy and disrupt the epigenetic status of neuronal cells, as evidenced by the inhibition of global DNA methylation as well as the reduction of histone deacetylases and histone acetyl transferases activity, which may increase the risks of neurodevelopmental abnormalities and/or neural degenerations.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - Joanna Rzemieniec
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - Władysław Lasoń
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - Wojciech Krzeptowski
- Department of Cell Biology and Imaging, Institute of Zoology, Jagiellonian University, Gronostajowa Street 9, 30-387, Krakow, Poland
| | - Małgorzata Kajta
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland.
| |
Collapse
|
28
|
Miranda-Morales E, Meier K, Sandoval-Carrillo A, Salas-Pacheco J, Vázquez-Cárdenas P, Arias-Carrión O. Implications of DNA Methylation in Parkinson's Disease. Front Mol Neurosci 2017; 10:225. [PMID: 28769760 PMCID: PMC5513956 DOI: 10.3389/fnmol.2017.00225] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/03/2017] [Indexed: 12/13/2022] Open
Abstract
It has been 200 years since Parkinson’s disease (PD) was first described, yet many aspects of its etiopathogenesis remain unclear. PD is a progressive and complex neurodegenerative disorder caused by genetic and environmental factors including aging, nutrition, pesticides and exposure to heavy metals. DNA methylation may be altered in response to some of these factors; therefore, it is proposed that epigenetic mechanisms, particularly DNA methylation, can have a fundamental role in gene–environment interactions that are related with PD. Epigenetic changes in PD-associated genes are now widely studied in different populations, to discover the mechanisms that contribute to disease development and identify novel biomarkers for early diagnosis and future pharmacological treatment. While initial studies sought to find associations between promoter DNA methylation and the regulation of associated genes in PD brain tissue, more recent studies have described concordant DNA methylation patterns between blood and brain tissue DNA. These data justify the use of peripheral blood samples instead of brain tissue for epigenetic studies. Here, we summarize the current data about DNA methylation changes in PD and discuss the potential of DNA methylation as a potential biomarker for PD. Additionally, we discuss environmental and nutritional factors that have been implicated in DNA methylation. Although the search for significant DNA methylation changes and gene expression analyses of PD-associated genes have yielded inconsistent and contradictory results, epigenetic modifications remain under investigation for their potential to reveal the link between environmental risk factors and the development of PD.
Collapse
Affiliation(s)
- Ernesto Miranda-Morales
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea GonzálezMexico City, Mexico.,Instituto de Investigación Científica, Universidad Juárez del Estado de DurangoDurango, Mexico
| | - Karin Meier
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de MéxicoMexico City, Mexico
| | - Ada Sandoval-Carrillo
- Instituto de Investigación Científica, Universidad Juárez del Estado de DurangoDurango, Mexico
| | - José Salas-Pacheco
- Instituto de Investigación Científica, Universidad Juárez del Estado de DurangoDurango, Mexico
| | | | - Oscar Arias-Carrión
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea GonzálezMexico City, Mexico
| |
Collapse
|
29
|
Kajta M, Wnuk A, Rzemieniec J, Litwa E, Lason W, Zelek-Molik A, Nalepa I, Rogóż Z, Grochowalski A, Wojtowicz AK. Depressive-like effect of prenatal exposure to DDT involves global DNA hypomethylation and impairment of GPER1/ESR1 protein levels but not ESR2 and AHR/ARNT signaling. J Steroid Biochem Mol Biol 2017; 171:94-109. [PMID: 28263910 DOI: 10.1016/j.jsbmb.2017.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 01/01/2023]
Abstract
Several lines of evidence suggest that exposures to Endocrine Disrupting Chemicals (EDCs) such as pesticides increase the risks of neuropsychiatric disorders. Despite extended residual persistence of dichlorodiphenyltrichloroethane (DDT) in the environment, the mechanisms of perinatal actions of DDT that could account for adult-onset of depression are largely unknown. This study demonstrated the isomer-specific induction of depressive-like behavior and impairment of Htr1a/serotonin signaling in one-month-old mice that were prenatally exposed to DDT. The effects were reversed by the antidepressant citalopram as evidenced in the forced swimming (FST) and tail suspension (TST) tests in the male and female mice. Prenatally administered DDT accumulated in mouse brain as determined with gas chromatography and tandem mass spectrometry, led to global DNA hypomethylation, and altered the levels of methylated DNA in specific genes. The induction of depressive-like behavior and impairment of Htr1a/serotonin signaling were accompanied by p,p'-DDT-specific decrease in the levels of estrogen receptors i.e. ESR1 and/or GPER1 depending on sex. In contrast, o,p'-DDT did not induce depressive-like effects and exhibited quite distinct pattern of biochemical alterations that was related to aryl hydrocarbon receptor (AHR), its nuclear translocator ARNT, and ESR2. Exposure to o,p'-DDT increased AHR expression in male and female brains, and reduced expression levels of ARNT and ESR2 in the female brains. The evolution of p,p'-DDT-induced depressive-like behavior was preceded by attenuation of Htr1a and Gper1/GPER1 expression as observed in the 7-day-old mouse pups. Because p,p'-DDT caused sex- and age-independent attenuation of GPER1, we suggest that impairment of GPER1 signaling plays a key role in the propagation of DDT-induced depressive-like symptoms.
Collapse
Affiliation(s)
- Malgorzata Kajta
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343 Krakow, Poland.
| | - Agnieszka Wnuk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343 Krakow, Poland
| | - Joanna Rzemieniec
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343 Krakow, Poland
| | - Ewa Litwa
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343 Krakow, Poland
| | - Wladyslaw Lason
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343 Krakow, Poland
| | - Agnieszka Zelek-Molik
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343 Krakow, Poland
| | - Irena Nalepa
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343 Krakow, Poland
| | - Zofia Rogóż
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343 Krakow, Poland
| | - Adam Grochowalski
- Department of Analytical Chemistry, Krakow University of Technology, Warszawska Street 24, 31-155 Krakow, Poland
| | - Anna K Wojtowicz
- Department of Animal Biotechnology, Faculty of Animal Sciences, University of Agriculture, Redzina Street 1B, 30-248 Krakow, Poland
| |
Collapse
|
30
|
Wnuk A, Rzemieniec J, Lasoń W, Krzeptowski W, Kajta M. Apoptosis Induced by the UV Filter Benzophenone-3 in Mouse Neuronal Cells Is Mediated via Attenuation of Erα/Pparγ and Stimulation of Erβ/Gpr30 Signaling. Mol Neurobiol 2017; 55:2362-2383. [PMID: 28357806 PMCID: PMC5840254 DOI: 10.1007/s12035-017-0480-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 03/03/2017] [Indexed: 12/22/2022]
Abstract
Although benzophenone-3 (BP-3) has frequently been reported to play a role in endocrine disruption, there is insufficient data regarding the impact of BP-3 on the nervous system, including its possible adverse effects on the developing brain. Our study demonstrated that BP-3 caused neurotoxicity and activated apoptosis via an intrinsic pathway involving the loss of mitochondrial membrane potential and the activation of caspases-9 and -3 and kinases p38/MAPK and Gsk3β. These biochemical alterations were accompanied by ROS production, increased apoptotic body formation and impaired cell survival, and by an upregulation of the genes involved in apoptosis. The BP-3-induced effects were tissue-specific and age-dependent with the most pronounced effects observed in neocortical cells at 7 days in vitro. BP-3 changed the messenger RNA (mRNA) expression levels of Erα, Erβ, Gpr30, and Pparγ in a time-dependent manner. At 3 h of exposure, BP-3 downregulated estrogen receptor mRNAs but upregulated Pparγ mRNA. After prolonged exposures, BP-3 downregulated the receptor mRNAs except for Erβ mRNA that was upregulated. The BP-3-induced patterns of mRNA expression measured at 6 and 24 h of exposure reflected alterations in the protein levels of the receptors and paralleled their immunofluorescent labeling. Erα and Pparγ agonists diminished, but Erβ and Gpr30 agonists stimulated the BP-3-induced apoptotic and neurotoxic effects. Receptor antagonists caused the opposite effects, except for ICI 182,780. This is in line with a substantial reduction in the effects of BP-3 in cells with siRNA-silenced Erβ/Gpr30 and the maintenance of BP-3 effects in Erα- and Pparγ siRNA-transfected cells. We showed for the first time that BP-3-affected mRNA and protein expression levels of Erα, Erβ, Gpr30, and Pparγ, paralleled BP-3-induced apoptosis and neurotoxicity. Therefore, we suggest that BP-3-evoked apoptosis of neuronal cells is mediated via attenuation of Erα/Pparγ and stimulation of Erβ/Gpr30 signaling.
Collapse
Affiliation(s)
- A Wnuk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - J Rzemieniec
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - W Lasoń
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland
| | - W Krzeptowski
- Department of Cell Biology and Imaging, Institute of Zoology, Jagiellonian University, Gronostajowa Street 9, 30-387, Krakow, Poland
| | - M Kajta
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna Street 12, 31-343, Krakow, Poland.
| |
Collapse
|