1
|
da Cunha Menezes E, de Abreu FF, Davis JB, Maurer SV, Roshko VC, Richardson A, Dowell J, Cassella SN, Stevens HE. Effects of gestational hypothyroidism on mouse brain development: Gabaergic systems and oxidative stress. Dev Biol 2024; 515:112-120. [PMID: 39048051 PMCID: PMC11330572 DOI: 10.1016/j.ydbio.2024.07.010] [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: 10/20/2023] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Hormonal imbalance during pregnancy is a risk factor for neuropsychiatric impairment in the offspring. It has been suggested that hypothyroidism leads to dysfunction of cortical GABAergic interneurons and inhibitory system development that in turn underlies impairment of the central nervous system. Here we investigated how gestational hypothyroidism affected offspring GABAergic system development as well as redox regulation parameters, because of previous links identified between the two. Experimental Gestational Hypothyroidism (EGH) was induced in CD-1 mice with 0.02% methimazole (MMI) in drinking water from embryonic day 9 (E9) until tissue collection at embryonic day 14 (E14) or E18. We examined GABAergic cell distribution and inhibitory system development gene expression as well as redox relevant gene expression and direct measures across all embryos regardless of sex. Intrauterine restriction of maternal thyroid hormones significantly impacted both of these outcomes in brain, as well as altering redox regulation in the placenta. GAD67+ neuronal migration was reduced, accompanied by a disruption in gene expression influencing GABAergic cell migration and cortical inhibitory neural system development. EGH also altered embryonic brain gene expression of Gpx1, Nfe2l2, Cat levels in the dorsal E14 brains. Additionally, EGH resulted in elevated TBARS, Gpx1 and Nfe2l2 in the ventral E18 brains. Furthermore, EGH downregulated placental Gpx1 gene expression at E14 and increased protein oxidation at E18. These findings support the hypothesis that sufficient maternal thyroid hormone supply to the fetus influences central nervous system development, including processes of GABAergic system development and redox equilibrium.
Collapse
Affiliation(s)
- Edênia da Cunha Menezes
- Psychiatry Department, Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States; Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Fabiula Francisca de Abreu
- Departamento de Fisiologia, Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Federal de Sergipe, São Cristóvão, Brazil
| | - Jada B Davis
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Sara V Maurer
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Venezia C Roshko
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | | | - Jonathan Dowell
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Sarah N Cassella
- Neuroscience Department, Loras College, Dubuque, IA, United States
| | - Hanna E Stevens
- Psychiatry Department, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
2
|
Sánchez-Rubio M, Abarzúa-Catalán L, Del Valle A, Méndez-Ruette M, Salazar N, Sigala J, Sandoval S, Godoy MI, Luarte A, Monteiro LJ, Romero R, Choolani MA, Wyneken Ú, Illanes SE, Bátiz LF. Maternal stress during pregnancy alters circulating small extracellular vesicles and enhances their targeting to the placenta and fetus. Biol Res 2024; 57:70. [PMID: 39342314 PMCID: PMC11438166 DOI: 10.1186/s40659-024-00548-4] [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: 10/06/2023] [Accepted: 09/05/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Maternal psychological distress during pregnancy can negatively impact fetal development, resulting in long-lasting consequences for the offspring. These effects show a sex bias. The mechanisms whereby prenatal stress induces functional and/or structural changes in the placental-fetal unit remain poorly understood. Maternal circulating small extracellular vesicles (sEVs) are good candidates to act as "stress signals" in mother-to-fetus communication. Using a repetitive restraint-based rat model of prenatal stress, we examined circulating maternal sEVs under stress conditions and tested whether they could target placental-fetal tissues. RESULTS Our mild chronic maternal stress during pregnancy paradigm induced anhedonic-like behavior in pregnant dams and led to intrauterine growth restriction (IUGR), particularly in male fetuses and placentas. The concentration and cargo of maternal circulating sEVs changed under stress conditions. Specifically, there was a significant reduction in neuron-enriched proteins and a significant increase in astrocyte-enriched proteins in blood-borne sEVs from stressed dams. To study the effect of repetitive restraint stress on the biodistribution of maternal circulating sEVs in the fetoplacental unit, sEVs from pregnant dams exposed to stress or control protocol were labeled with DiR fluorescent die and injected into pregnant females previously exposed to control or stress protocol. Remarkably, maternal circulating sEVs target placental/fetal tissues and, under stress conditions, fetal tissues are more receptive to sEVs. CONCLUSION Our results suggest that maternal circulating sEVs can act as novel mediators/modulators of mother-to-fetus stress communication. Further studies are needed to identify placental/fetal cellular targets of maternal sEVs and characterize their contribution to stress-induced sex-specific placental and fetal changes.
Collapse
Affiliation(s)
- Mario Sánchez-Rubio
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
| | - Lorena Abarzúa-Catalán
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
| | - Ana Del Valle
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
| | - Maxs Méndez-Ruette
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- PhD Program in Biomedicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Natalia Salazar
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Jacinta Sigala
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Soledad Sandoval
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - María Inés Godoy
- Department of Educational Assessment, Measurement, and Registry, Universidad de Chile, Santiago, Chile
| | - Alejandro Luarte
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Lara J Monteiro
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Research Program in Biology of Reproduction, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Detroit, and Maryland, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Mahesh A Choolani
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Úrsula Wyneken
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Sebastián E Illanes
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Research Program in Biology of Reproduction, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile.
- Department of Obstetrics and Gynecology, School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica (CiiB), Facultad de Medicina, Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
| | - Luis Federico Bátiz
- Research Program in Neuroscience, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- School of Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica (CiiB), Facultad de Medicina, Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
| |
Collapse
|
3
|
Zhao GP, Cheng WL, Zhang ZH, Li YX, Li YQ, Yang FW, Wang YB. The use of amino acids and their derivates to mitigate against pesticide-induced toxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116340. [PMID: 38636261 DOI: 10.1016/j.ecoenv.2024.116340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/11/2024] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
Exposure to pesticides induces oxidative stress and deleterious effects on various tissues in non-target organisms. Numerous models investigating pesticide exposure have demonstrated metabolic disturbances such as imbalances in amino acid levels within the organism. One potentially effective strategy to mitigate pesticide toxicity involves dietary intervention by supplementing exogenous amino acids and their derivates to augment the body's antioxidant capacity and mitigate pesticide-induced oxidative harm, whose mechanism including bolstering glutathione synthesis, regulating arginine-NO metabolism, mitochondria-related oxidative stress, and the open of ion channels, as well as enhancing intestinal microecology. Enhancing glutathione synthesis through supplementation of substrates N-acetylcysteine and glycine is regarded as a potent mechanism to achieve this. Selection of appropriate amino acids or their derivates for supplementation, and determining an appropriate dosage, are of the utmost importance for effective mitigation of pesticide-induced oxidative harm. More experimentation is required that involves large population samples to validate the efficacy of dietary intervention strategies, as well as to determine the effects of amino acids and their derivates on long-term and low-dose pesticide exposure. This review provides insights to guide future research aimed at preventing and alleviating pesticide toxicity through dietary intervention of amino acids and their derivates.
Collapse
Affiliation(s)
- Guo-Ping Zhao
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China.
| | - Wei-Long Cheng
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Zhi-Hui Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Yi-Xuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; National Center of Technology Innovation for Dairy, Inner Mongolia 013757, China
| | - Ying-Qiu Li
- School of Food Science and Engineering, Qilu University of Technology, Jinan 250353, China
| | - Fang-Wei Yang
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Yan-Bo Wang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
4
|
Gao H, Tian M, Geng X, Zhao J, Song Y, Wu B, Tian X, Yang Y, Ni W, Yang H. Cyfluthrin exposure during pregnancy causes neurotoxicity in offspring-Ca 2+ overload via IP3R-GRP75-VDAC1 pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116218. [PMID: 38492481 DOI: 10.1016/j.ecoenv.2024.116218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/01/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cyfluthrin (Cy) is a widely used pyrethroid insecticide. There is growing evidence that Cy can cause damage to the nervous, reproductive, and immune systems, but there is limited evidence on the potential effects of maternal Cy exposure on offspring. A model of maternal Cy exposure was used to assess its neurobehavioral effects on young-adult offspring. We found that gestational Cy exposure affected pregnancy outcomes and fetal development, and that offspring showed impairments in anxiety as well as learning and memory, accompanied by impairments in hippocampal synaptic ultrastructure and synaptic plasticity. In addition, the IP3R-GRP75-VDAC1 apoptogenic pathway was also upregulated, and in vitro models showed that inhibition of this pathway alleviated neuronal apoptosis as well as synaptic plasticity damage. In conclusion, maternal Cy exposure during pregnancy can cause neurobehavioral abnormalities and synaptic damage in offspring, which may be related to neuronal apoptosis induced by activation of the IP3R-GRP75-VDAC1 pathway in the hippocampus of offspring. Our findings provide clues to understand the neurotoxicity mechanism of maternal Cy exposure to offspring during pregnancy.
Collapse
Affiliation(s)
- Haoxuan Gao
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Mi Tian
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Xiaozhe Geng
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Ji Zhao
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Yanan Song
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Bing Wu
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Xueyan Tian
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Yong Yang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Wensi Ni
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.
| | - Huifang Yang
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.
| |
Collapse
|
5
|
Macedo MB, Cunha EO, Reis AD, Machado MS, de Campos D, Malysz T, Dallegrave E. Morphometric Evaluation of the Recurrent Laryngeal Nerve of Wistar Rats Exposed to Pesticides. J Voice 2024; 38:264-272. [PMID: 34782225 DOI: 10.1016/j.jvoice.2021.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 09/23/2021] [Indexed: 10/19/2022]
Abstract
The literature has been shown that exposition by inhalation to chemical compounds can cause vocal disorders and dysphagia in humans, in addition to other symptoms that are manifested according to the type, concentration and duration of exposure to the substance. Cypermethrin and dichlorvos are pesticides widely used in agriculture, public health, veterinary, and home environments. Despite the scientific evidence that cypermethrin and dichlorvos can cause neurodegenerative damage and motor alterations, there are no studies evaluating the toxic effects of these pesticides on the morphology of structures responsible for vocal mobility, especially to the Recurrent Laryngeal Nerve (RLN). Considering the association between vocal disorders in humans and variations in RLN and morphometry, the aim of this study was to evaluate the possible alterations in the microstructure of RLN secondary to subchronic exposure to cypermethrin (pyrethroid) and dichlorvos (organophosphate) in Wistar rats. The experimental protocol (approved by CEUA-UFCSPA: 321/15 and 323/15) consisted of 15 male Wistar rats, allocated in 3 groups: Control (n = 5, exposed to water), Cypermethrin (n = 5, exposed to cypermethrin - 1/10 of the inhalation median lethal concentration [LC50] - 0.25 mg/L) and dichlorvos (n = 5, exposed to dichlorvos - 1/10 of the LC50 - 1.5 mg/L). Inhalation exposure was performed for 4 hours, 5 times per week, for 6 weeks. The nerves were collected, histologically processed and analyzed using morphometric parameters measured using ZEN 2.6 (Zeiss - Germany). The cypermethrin and dichlorvos groups showed significant changes (P < 0.001, ANOVA) in the g-ratio and in the thickness of the myelin sheath of the RLN when compared to the control animals, however, none of the other parameters evaluated showed statistically significant differences. These findings indicate that repeated inhalation exposure to commercial products of cypermethrin and dichlorvos is able to modify the structure of the RLN and possibly generating vocal changes and / or dysphagia.
Collapse
Affiliation(s)
- Mateus Belmonte Macedo
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Laboratory of Research in Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Eduarda Oliveira Cunha
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Laboratory of Research in Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Aléxia Dos Reis
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Laboratory of Research in Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Márcia Salgado Machado
- Department of Speech Therapy and Audiology, University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Laboratory of Research in Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Deivis de Campos
- Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Department of Life Sciences, University of Santa Cruz do Sul (UNISC), Santa Cruz do Sul, RS, Brazil; Graduate Program in Health Promotion, University of Santa Cruz do Sul (UNISC), Santa Cruz do Sul, RS, Brazil
| | - Taís Malysz
- Graduate Program in Neurosciences, Basic Health Sciences Institute, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Eliane Dallegrave
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Department of Pharmacosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Laboratory of Research in Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.
| |
Collapse
|
6
|
Wolfe J, Marsit C. Pyrethroid pesticide exposure and placental effects. Mol Cell Endocrinol 2023; 578:112070. [PMID: 37722502 PMCID: PMC10591723 DOI: 10.1016/j.mce.2023.112070] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/20/2023]
Abstract
Human exposures to pyrethroid pesticides have increased in recent years following the bans and sanctions placed on other families of pesticides. Although pyrethroids are currently widely used across the United States and throughout the world, and their overt neurological toxicity classified, the extent of their toxicity through low dose and chronic exposures on humans is less well characterized, particularly when it comes to prenatal exposures, their impacts on neurodevelopment, and any role for the placenta in those effects. In this review, we assess the state of research on pyrethroid pesticide exposure and placental effects. These studies presented hormone disrupting, genotoxic, neurodevelopmental and neurobehavioral effects, among others, following prenatal pyrethroid exposures, and highlights a need for future research to assess gaps relating to effects in the human placenta and mechanisms of toxicity as well as shortcomings in the reproducibility and standardization of the methodologies presented.
Collapse
Affiliation(s)
- Joshua Wolfe
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Carmen Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
7
|
Lesseur C, Kaur K, Kelly SD, Hermetz K, Williams R, Hao K, Marsit CJ, Caudle WM, Chen J. Effects of prenatal pesticide exposure on the fetal brain and placenta transcriptomes in a rodent model. Toxicology 2023; 490:153498. [PMID: 37019170 PMCID: PMC10152924 DOI: 10.1016/j.tox.2023.153498] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023]
Abstract
Organophosphate and pyrethroid pesticides are among the most extensively used insecticides worldwide. Prenatal exposures to both classes of pesticides have been linked to a wide range of neurobehavioral deficits in the offspring. The placenta is a neuroendocrine organ and the crucial regulator of the intrauterine environment; early-life toxicant exposures could impact neurobehavior by disrupting placental processes. Female C57BL/6 J mice were exposed via oral gavage to an organophosphate, chlorpyrifos (CPF) at 5 mg/kg, a pyrethroid, deltamethrin (DM), at 3 mg/kg, or vehicle only control (CTL). Exposure began two weeks before breeding and continued every three days until euthanasia at gestational day 17. The transcriptomes of fetal brain (CTL n = 18, CPF n = 6, DM n = 8) and placenta (CTL n = 19, CPF n = 16, DM n = 12) were obtained through RNA sequencing, and resulting data was evaluated using weighted gene co-expression networks, differential expression, and pathway analyses. Fourteen brain gene co-expression modules were identified; CPF exposure disrupted the module related to ribosome and oxidative phosphorylation, whereas DM disrupted the modules related to extracellular matrix and calcium signaling. In the placenta, network analyses revealed 12 gene co-expression modules. While CPF exposure disrupted modules related to endocytosis, Notch and Mapk signaling, DM exposure dysregulated modules linked to spliceosome, lysosome and Mapk signaling pathways. Overall, in both tissues, CPF exposure impacted oxidative phosphorylation, while DM was linked to genes involved in spliceosome and cell cycle. The transcription factor Max involved in cell proliferation was overexpressed by both pesticides in both tissues. In summary, gestational exposure to two different classes of pesticide can induce similar pathway-level transcriptome changes in the placenta and the brain; further studies should investigate if these changes are linked to neurobehavioral impairments.
Collapse
Affiliation(s)
- Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY 10029, USA
| | - Kirtan Kaur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY 10029, USA
| | - Sean D Kelly
- Gangarosa Department of Environmental Health, Rollins School of Public Health Emory University, Atlanta, GA 30322, USA
| | - Karen Hermetz
- Gangarosa Department of Environmental Health, Rollins School of Public Health Emory University, Atlanta, GA 30322, USA
| | - Randy Williams
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY 10029, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health Emory University, Atlanta, GA 30322, USA
| | - W Michael Caudle
- Gangarosa Department of Environmental Health, Rollins School of Public Health Emory University, Atlanta, GA 30322, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY 10029, USA.
| |
Collapse
|
8
|
Carver AJ, Taylor RJ, Stevens HE. Mouse In Vivo Placental Targeted CRISPR Manipulation. J Vis Exp 2023:10.3791/64760. [PMID: 37125793 PMCID: PMC10664715 DOI: 10.3791/64760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
The placenta is an essential organ that regulates and maintains mammalian development in utero. The placenta is responsible for the transfer of nutrients and waste between the mother and fetus and the production and delivery of growth factors and hormones. Placental genetic manipulations in mice are critical for understanding the placenta's specific role in prenatal development. Placental-specific Cre-expressing transgenic mice have varying effectiveness, and other methods for placental gene manipulation can be useful alternatives. This paper describes a technique to directly alter placental gene expression using CRISPR gene manipulation, which can be used to modify the expression of targeted genes. Using a relatively advanced surgical approach, pregnant dams undergo a laparotomy on embryonic day 12.5 (E12.5), and a CRISPR plasmid is delivered by a glass micropipette into the individual placentas. The plasmid is immediately electroporated after each injection. After dam recovery, the placentas and embryos can continue development until assessment at a later time point. The evaluation of the placenta and offspring after the use of this technique can determine the role of time-specific placental function in development. This type of manipulation will allow for a better understanding of how placental genetics and function impact fetal growth and development in multiple disease contexts.
Collapse
Affiliation(s)
- Annemarie J Carver
- Interdisciplinary Graduate Program in Genetics, University of Iowa; Department of Psychiatry, Carver College of Medicine, University of Iowa; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa
| | - Robert J Taylor
- Department of Psychiatry, Carver College of Medicine, University of Iowa; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa
| | - Hanna E Stevens
- Interdisciplinary Graduate Program in Genetics, University of Iowa; Department of Psychiatry, Carver College of Medicine, University of Iowa; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa; Hawk-IDDRC, University of Iowa;
| |
Collapse
|
9
|
Curtis MA, Dhamsania RK, Branco RC, Guo JD, Creeden J, Neifer KL, Black CA, Winokur EJ, Andari E, Dias BG, Liu RC, Gourley SL, Miller GW, Burkett JP. Developmental pyrethroid exposure causes a neurodevelopmental disorder phenotype in mice. PNAS NEXUS 2023; 2:pgad085. [PMID: 37113978 PMCID: PMC10129348 DOI: 10.1093/pnasnexus/pgad085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 03/06/2023] [Indexed: 04/29/2023]
Abstract
Neurodevelopmental disorders (NDDs) are a widespread and growing public health challenge, affecting as many as 17% of children in the United States. Recent epidemiological studies have implicated ambient exposure to pyrethroid pesticides during pregnancy in the risk for NDDs in the unborn child. Using a litter-based, independent discovery-replication cohort design, we exposed mouse dams orally during pregnancy and lactation to the Environmental Protection Agency's reference pyrethroid, deltamethrin, at 3 mg/kg, a concentration well below the benchmark dose used for regulatory guidance. The resulting offspring were tested using behavioral and molecular methods targeting behavioral phenotypes relevant to autism and NDD, as well as changes to the striatal dopamine system. Low-dose developmental exposure to the pyrethroid deltamethrin (DPE) decreased pup vocalizations, increased repetitive behaviors, and impaired both fear conditioning and operant conditioning. Compared with control mice, DPE mice had greater total striatal dopamine, dopamine metabolites, and stimulated dopamine release, but no difference in vesicular dopamine capacity or protein markers of dopamine vesicles. Dopamine transporter protein levels were increased in DPE mice, but not temporal dopamine reuptake. Striatal medium spiny neurons showed changes in electrophysiological properties consistent with a compensatory decrease in neuronal excitability. Combined with previous findings, these results implicate DPE as a direct cause of an NDD-relevant behavioral phenotype and striatal dopamine dysfunction in mice and implicate the cytosolic compartment as the location of excess striatal dopamine.
Collapse
Affiliation(s)
- Melissa A Curtis
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, USA
| | - Rohan K Dhamsania
- College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA 19131, USA
| | - Rachel C Branco
- Laney Graduate School, Emory University, Atlanta, GA 30322, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Ji-Dong Guo
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Justin Creeden
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Kari L Neifer
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, USA
| | - Carlie A Black
- Laney Graduate School, Emory University, Atlanta, GA 30322, USA
- Schiemer School of Psychology and Biblical Counseling, Truett McConnell University, Cleveland, GA 30528, USA
| | - Emily J Winokur
- College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
- Department of Cognitive Science, University of California San Diego, La Jolla, CA 92093, USA
| | - Elissar Andari
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Brian G Dias
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA 90089, USA
- Division of Endocrinology, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA 90027, USA
| | - Robert C Liu
- Department of Biology, Emory University, Atlanta, GA 30322, USA
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA
| | - Shannon L Gourley
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory National Primate Research Center, Atlanta, GA 30329, USA
| | - Gary W Miller
- Department of Environmental Health, Emory Rollins School of Public Health, Atlanta, GA 30322, USA
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
10
|
Dong Y, Weng J, Zhu Y, Sun D, He W, Chen Q, Cheng J, Zhu Y, Jiang Y. Transcriptomic profiling of the developing brain revealed cell-type and brain-region specificity in a mouse model of prenatal stress. BMC Genomics 2023; 24:86. [PMID: 36829105 PMCID: PMC9951484 DOI: 10.1186/s12864-023-09186-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Prenatal stress (PS) is considered as a risk factor for many mental disorders. PS-induced transcriptomic alterations may contribute to the functional dysregulation during brain development. Here, we used RNA-seq to explore changes of gene expression in the mouse fetal brain after prenatal exposure to chronic unpredictable mild stress (CUMS). RESULTS We compared the stressed brains to the controls and identified groups of significantly differentially expressed genes (DEGs). GO analysis on up-regulated DEGs revealed enrichment for the cell cycle pathways, while down-regulated DEGs were mostly enriched in the neuronal pathways related to synaptic transmission. We further performed cell-type enrichment analysis using published scRNA-seq data from the fetal mouse brain and revealed cell-type-specificity for up- and down-regulated DEGs, respectively. The up-regulated DEGs were highly enriched in the radial glia, while down-regulated DEGs were enriched in different types of neurons. Cell deconvolution analysis further showed altered cell fractions in the stressed brain, indicating accumulation of neuroblast and impaired neurogenesis. Moreover, we also observed distinct brain-region expression pattern when mapping DEGs onto the developing Allen brain atlas. The up-regulated DEGs were primarily enriched in the dorsal forebrain regions including the cortical plate and hippocampal formation. Surprisingly, down-regulated DEGs were found excluded from the cortical region, but highly expressed on various regions in the ventral forebrain, midbrain and hindbrain. CONCLUSION Taken together, we provided an unbiased data source for transcriptomic alterations of the whole fetal brain after chronic PS, and reported differential cell-type and brain-region vulnerability of the developing brain in response to environmental insults during the pregnancy.
Collapse
Affiliation(s)
- Yuhao Dong
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Jie Weng
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Yueyan Zhu
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Daijing Sun
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Wei He
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Qi Chen
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Jin Cheng
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Ying Zhu
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Yan Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
11
|
Elser BA, Simonsen D, Lehmler HJ, Stevens HE. Maternal and fetal tissue distribution of α-cypermethrin and permethrin in pregnant CD-1 mice. ENVIRONMENTAL ADVANCES 2022; 8:100239. [PMID: 36059860 PMCID: PMC9435064 DOI: 10.1016/j.envadv.2022.100239] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Pyrethroid insecticides are widely used throughout agriculture and household products. Recent studies suggest that prenatal exposure to these insecticides may adversely affect fetal development; however, little is known about the distribution of these chemicals in pregnant animals. The present study aimed to address this gap in knowledge by investigating the distribution of two commonly used pyrethroid insecticides, permethrin and α-cypermethrin, in maternal and fetal tissues of pregnant CD-1 mice. Dams were dosed from gestational days 6 to 16 via oral gavage with permethrin (1.5, 15, and 50 mg/kg), α-cypermethrin (0.3, 3, and 10 mg/kg), or corn oil vehicle. Pyrethroid levels were determined in gestational day 16 tissues collected 90 min after the final dose was administered. Across maternal tissues, levels of both pyrethroids were the highest in maternal ovaries, followed by liver and brain, respectively. In addition, levels of both pyrethroids in maternal tissues and placenta were significantly higher than those in the fetal body and amniotic fluid, suggesting that these compounds may exhibit low transfer across the mouse placenta. While additional toxicokinetic studies are needed to verify the time course of pyrethroids in the fetal compartment, these findings support investigation into indirect modes of action relevant to the effects of pyrethroids on mammalian fetal development.
Collapse
Affiliation(s)
- Benjamin A. Elser
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, 1330 Pappajohn Biomedical Discovery Building, Iowa City, IA 52242, USA
| | - Derek Simonsen
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, IA, USA
- Department of Occupational and Environmental Health, The University of Iowa, Iowa City, IA 52242, USA
| | - Hans-Joachim Lehmler
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, IA, USA
- Department of Occupational and Environmental Health, The University of Iowa, Iowa City, IA 52242, USA
| | - Hanna E. Stevens
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, 1330 Pappajohn Biomedical Discovery Building, Iowa City, IA 52242, USA
- Corresponding author at: Department of Psychiatry, Carver College of Medicine, The University of Iowa, 1330 Pappajohn Biomedical Discovery Building, Iowa City, IA 52242, USA. (H.E. Stevens)
| |
Collapse
|
12
|
Wang Q, Wang XX, Xie JF, Yao TT, Xu LL, Wang LS, Yu Y, Xu LC. Cypermethrin inhibits proliferation of Sertoli cells through AR involving DAB2IP/PI3K/AKT signaling pathway in vitro. Toxicol Res (Camb) 2022; 11:583-591. [DOI: 10.1093/toxres/tfac037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/27/2022] [Accepted: 05/09/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Cypermethrin (CP) exhibits anti-androgenic effects through antagonism on androgen receptor (AR) activation. This study was to identify whether AR-mediated disabled 2 interacting protein (DAB2IP)/phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway was involved in CP-induced mouse Sertoli cells (TM4) proliferation disorder. Real-Time Cell Analysis-iCELLigence system was to measure cell proliferation. Bioinformatic analyses were performed to identify AR-regulated genes. Quantitative Real-Time PCR and western blot were to detect the genes and proteins levels in AR-mediated DAB2IP/PI3K/AKT pathway. Results showed CP suppressed TM4 proliferation and the expression of AR. Activation of AR restored the inhibition efficacy of CP on TM4 proliferation. AR regulated DAB2IP expression and phosphorylation levels of PI3K and AKT in CP-exposed TM4 cells. In addition, knockdown of DAB2IP alleviated the inhibition efficacy of CP on cell proliferation and phosphorylation of PI3K and AKT. Taken together, AR was a modulator in CP-induced inhibition of Sertoli cells proliferation by negatively regulating DAB2IP/PI3K/AKT signaling pathway. The study may provide a new insight for the mechanisms of male reproductive toxicity induced by CP.
Collapse
Affiliation(s)
- Qi Wang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University , 209 Tong-Shan Road, Xuzhou, Jiangsu 221004 , China
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University , Xuzhou, Jiangsu 221004 , China
| | - Xu-Xu Wang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University , 209 Tong-Shan Road, Xuzhou, Jiangsu 221004 , China
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University , Xuzhou, Jiangsu 221004 , China
| | - Jia-Fei Xie
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University , 209 Tong-Shan Road, Xuzhou, Jiangsu 221004 , China
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University , Xuzhou, Jiangsu 221004 , China
| | - Ting-Ting Yao
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University , 209 Tong-Shan Road, Xuzhou, Jiangsu 221004 , China
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University , Xuzhou, Jiangsu 221004 , China
| | - Lin-Lin Xu
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University , 209 Tong-Shan Road, Xuzhou, Jiangsu 221004 , China
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University , Xuzhou, Jiangsu 221004 , China
| | - Lu-Shan Wang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University , 209 Tong-Shan Road, Xuzhou, Jiangsu 221004 , China
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University , Xuzhou, Jiangsu 221004 , China
| | - Yue Yu
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University , 209 Tong-Shan Road, Xuzhou, Jiangsu 221004 , China
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University , Xuzhou, Jiangsu 221004 , China
| | - Li-Chun Xu
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University , 209 Tong-Shan Road, Xuzhou, Jiangsu 221004 , China
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University , Xuzhou, Jiangsu 221004 , China
| |
Collapse
|
13
|
Elser BA, Hing B, Stevens HE. A narrative review of converging evidence addressing developmental toxicity of pyrethroid insecticides. Crit Rev Toxicol 2022; 52:371-388. [PMID: 36345971 PMCID: PMC9930199 DOI: 10.1080/10408444.2022.2122769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/11/2022]
Abstract
Pyrethroid insecticides are broadly used in agriculture and household products throughout the world. Exposure to this class of insecticides is widespread, and while generally believed to be safe for use, there is increasing concern regarding their effects on neurodevelopment. Due to the critical roles that molecular targets of pyrethroids play in the regulation of neurodevelopment, particular focus has been placed on evaluating the effects of in utero and childhood pyrethroid exposure on child cognition and behavior. As such, this narrative review synthesizes an assessment of converging study types; we review reports of neonatal pyrethroid levels together with current epidemiological literature that convergently address the risk for developmental toxicity linked to exposure to pyrethroid insecticides. We first address studies that assess the degree of direct fetal exposure to pyrethroids in utero through measurements in cord blood, meconium, and amniotic fluid. We then focus on the links between prenatal exposure to these insecticides and child neurodevelopment, fetal growth, and other adverse birth outcomes. Furthermore, we assess the effects of postnatal exposure on child neurodevelopment through a review of the data on pediatric exposures and child cognitive and behavioral outcomes. Study quality was evaluated individually, and the weight of evidence was assessed broadly to characterize these effects. Overall, while definitive conclusions cannot be reached from the currently available literature, the available data suggest that the potential links between pyrethroid exposure and child neurodevelopmental effects deserve further investigation.
Collapse
Affiliation(s)
- Benjamin A Elser
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Benjamin Hing
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Hanna E Stevens
- Interdisciplinary Graduate Program in Human Toxicology, Graduate College, The University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
14
|
Davis JLB, O’Connor M, Erlbacher H, Schlichte SL, Stevens HE. The Impact of Maternal Antioxidants on Prenatal Stress Effects on Offspring Neurobiology and Behavior. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2022; 95:87-104. [PMID: 35370489 PMCID: PMC8961714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prenatal stress is a neuropsychiatric risk factor, and effects may be mediated by prenatal oxidative stress. Cell types in the brain sensitive to oxidative stress-cortical microglia and cortical and hippocampal interneurons-may be altered by oxidative stress generated during prenatal stress and may be neurobiological substrates for altered behavior. Our objective was to determine the critical nature of oxidative stress in prenatal stress effects by manipulating prenatal antioxidants. CD1 mouse dams underwent restraint embryonic day 12 to 18 three times daily or no stress and received intraperitoneal injections before each stress period of vehicle, N-acetylcysteine (200 mg/kg daily), or astaxanthin (30 mg/kg before first daily stress, 10 mg/kg before second/third stresses). Adult male and female offspring behavior, microglia, and interneurons were assessed. Results supported the hypothesis that prenatal stress-induced oxidative stress affects microglia; microglia ramification increased after prenatal stress, and both antioxidants prevented these effects. In addition, N-acetylcysteine or astaxanthin was effective in preventing distinct male and female interneuron changes; decreased female medial frontal cortical parvalbumin interneurons was prevented by either antioxidant; increased male medial frontal cortical parvalbumin interneurons was prevented by N-acetylcysteine and decreased male hippocampal GAD67GFP+ cells prevented by astaxanthin. Prenatal stress-induced increased anxiety-like behavior and decreased sociability were not prevented by prenatal antioxidants. Sensorimotor gating deficits in males was partially prevented by prenatal astaxanthin. This study demonstrates the importance of oxidative stress for persistent impacts on offspring cortical microglia and interneurons, but did not link these changes with anxiety-like, social, and sensorimotor gating behaviors.
Collapse
Affiliation(s)
- Jada L-B Davis
- Department of Psychiatry, University of Iowa, Iowa
City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience,
University of Iowa, Iowa City, IA, USA
| | - Mara O’Connor
- Department of Psychiatry, University of Iowa, Iowa
City, IA, USA
| | - Hannah Erlbacher
- Department of Psychiatry, University of Iowa, Iowa
City, IA, USA
| | | | - Hanna E. Stevens
- Department of Psychiatry, University of Iowa, Iowa
City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience,
University of Iowa, Iowa City, IA, USA
| |
Collapse
|
15
|
Pinto-Ribeiro L, Silva C, Andrade N, Martel F. α-tocopherol prevents oxidative stress-induced proliferative dysfunction in first-trimester human placental (HTR-8/SVneo) cells. Reprod Biol 2022; 22:100602. [PMID: 35016050 DOI: 10.1016/j.repbio.2022.100602] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/09/2021] [Accepted: 01/02/2022] [Indexed: 02/06/2023]
Abstract
Extravillous trophoblasts (EVTs) are the main participants in the process of placentation, an early process critical for placental growth and function involving an adequate invasion and complete remodelling of the maternal spiral arteries during early pregnancy. An increase in oxidative stress during pregnancy is associated with the onset and progression of several pregnancy disorders, including preeclampsia and gestational diabetes mellitus and it also occurs due to exposure of pregnant women to some xenobiotics (eg. alcohol). This study aimed to investigate how oxidative stress affects EVTs, and the ability of several distinct antioxidant agents to prevent these changes. For this, we exposed HTR8/SVneo cells to tert-butylhydroperoxide (0.5 μM; 24 h), which was able to increase lipid peroxidation and protein carbonyl levels. Under these conditions, there was a decrease in proliferation rates, culture growth, migratory and angiogenic capacities and an increase in the apoptosis rates. The antiproliferative effect of TBH was supressed by simultaneous treatment of the cells with α-tocopherol, but other antioxidants (vitamin C, allopurinol, apocynin, N-acetylcysteine, quercetin and resveratrol) were ineffective. α-tocopherol was also able to abolish the effect of TBH on lipid peroxidation and protein carbonyl levels. Overall, our results show that oxidative stress interferes with EVT characteristics essential for the placentation process, which may contribute to the association between oxidative stress and pregnancy disorders. Our results also show that the nature of the in vitro model of oxidative stress-induction is an important determinant of the cellular consequences of oxidative stress and, therefore, of the efficacy of antioxidants.
Collapse
Affiliation(s)
- Lígia Pinto-Ribeiro
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Cláudia Silva
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.
| |
Collapse
|
16
|
Li J, Bi H. The effect and mechanism of cypermethrin-induced hippocampal neurotoxicity as determined by network pharmacology analysis and experimental validation. Bioengineered 2021; 12:9279-9289. [PMID: 34714723 PMCID: PMC8810029 DOI: 10.1080/21655979.2021.2000106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Cypermethrin (CMN) is a widely used artificial synthetic pesticide that causes neurotoxicity in the hippocampus. However, the underlying toxicological targets and mechanisms remain unclear. In this study, network pharmacology analysis and in vitro models were integrated to investigate the effect and mechanism of CMN-induced hippocampal neurotoxicity. A total of 88 targets of CMN-induced hippocampal neurotoxicity were predicted. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes enrichment (KEGG) analyses suggested that these targets were related to multiple GO terms and signaling pathways. To further investigate underlying mechanism, the top 10 hub targets (Akt1, Tnf, Ptgs2, Casp3, Igf1, Sirt1, Jun, Cat, Il10, and Bcl2l1) were screened. Furthermore, cell viability and lactate dehydrogenase (LDH) assays demonstrated that CMN was toxic to HT22 cells in a time- and dose-dependent manner. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining revealed that treatment with CMN increased the proportion of apoptotic cells. In addition, the real-time quantitative polymerase chain reaction (RT-qPCR) results indicated that CMN altered the mRNA expression levels of most of the hub targets, with the exceptions of Igf1 and Jun. The results demonstrated that multiple targets and signaling pathways were involved in CMN-induced hippocampal neurotoxicity. These findings provided reference values for subsequent studies of the toxicological mechanism of CMN.
Collapse
Affiliation(s)
- Jianan Li
- KeyLaboratory of Environment and Health, College of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Haoran Bi
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
17
|
Schroeder R, Sridharan P, Nguyen L, Loren A, Williams NS, Kettimuthu KP, Cintrón-Pérez CJ, Vázquez-Rosa E, Pieper AA, Stevens HE. Maternal P7C3-A20 Treatment Protects Offspring from Neuropsychiatric Sequelae of Prenatal Stress. Antioxid Redox Signal 2021; 35:511-530. [PMID: 33501899 PMCID: PMC8388250 DOI: 10.1089/ars.2020.8227] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aims: Impaired embryonic cortical interneuron development from prenatal stress is linked to adult neuropsychiatric impairment, stemming in part from excessive generation of reactive oxygen species in the developing embryo. Unfortunately, there are no preventive medicines that mitigate the risk of prenatal stress to the embryo, as the underlying pathophysiologic mechanisms are poorly understood. Our goal was to interrogate the molecular basis of prenatal stress-mediated damage to the embryonic brain to identify a neuroprotective strategy. Results: Chronic prenatal stress in mice dysregulated nicotinamide adenine dinucleotide (NAD+) synthesis enzymes and cortical interneuron development in the embryonic brain, leading to axonal degeneration in the hippocampus, cognitive deficits, and depression-like behavior in adulthood. Offspring were protected from these deleterious effects by concurrent maternal administration of the NAD+-modulating agent P7C3-A20, which crossed the placenta to access the embryonic brain. Prenatal stress also produced axonal degeneration in the adult corpus callosum, which was not prevented by maternal P7C3-A20. Innovation: Prenatal stress dysregulates gene expression of NAD+-synthesis machinery and GABAergic interneuron development in the embryonic brain, which is associated with adult cognitive impairment and depression-like behavior. We establish a maternally directed treatment that protects offspring from these effects of prenatal stress. Conclusion: NAD+-synthesis machinery and GABAergic interneuron development are critical to proper embryonic brain development underlying postnatal neuropsychiatric functioning, and these systems are highly susceptible to prenatal stress. Pharmacologic stabilization of NAD+ in the stressed embryonic brain may provide a neuroprotective strategy that preserves normal embryonic development and protects offspring from neuropsychiatric impairment. Antioxid. Redox Signal. 35, 511-530.
Collapse
Affiliation(s)
- Rachel Schroeder
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA
| | - Preethy Sridharan
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.,Department of Psychiatry and Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience, Case Western Reserve University, Cleveland, Ohio, USA
| | - Lynn Nguyen
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Alexandra Loren
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kavitha P Kettimuthu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Coral J Cintrón-Pérez
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.,Department of Psychiatry and Case Western Reserve University, Cleveland, Ohio, USA
| | - Edwin Vázquez-Rosa
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.,Department of Psychiatry and Case Western Reserve University, Cleveland, Ohio, USA
| | - Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.,Department of Psychiatry and Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience, Case Western Reserve University, Cleveland, Ohio, USA.,Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Cleveland, Ohio, USA.,Weill Cornell Autism Research Program, Weill Cornell Medicine of Cornell University, New York, New York, USA
| | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
18
|
Yadav A, Tandon A, Seth B, Goyal S, Singh SJ, Tiwari SK, Agarwal S, Nair S, Chaturvedi RK. Cypermethrin Impairs Hippocampal Neurogenesis and Cognitive Functions by Altering Neural Fate Decisions in the Rat Brain. Mol Neurobiol 2021; 58:263-280. [PMID: 32920670 DOI: 10.1007/s12035-020-02108-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 08/28/2020] [Indexed: 12/31/2022]
Abstract
Neurogenesis is a developmental process that involves fine-tuned coordination between self-renewal, proliferation, and differentiation of neural stem cells (NSCs) into neurons. However, early-life assault with environmental toxicants interferes with the regular function of genes, proteins, and other molecules that build brain architecture resulting in attenuated neurogenesis. Cypermethrin is a class II synthetic pyrethroid pesticide extensively used in agriculture, veterinary, and residential applications due to its low mammalian toxicity, high bio-efficacy, and enhanced stability. Despite reports on cypermethrin-mediated behavioral and biochemical alterations, till now, no study implicates whether cypermethrin exposure has any effect on neurogenesis. Therefore, the present study was undertaken to comprehend the effects of cypermethrin treatment on embryonic and adult neurogenesis. We found that cypermethrin exposure led to a considerable decrease in the BrdU/Sox-2+, BrdU/Dcx+, and BrdU/NeuN+ co-labeled cells indicating that cypermethrin treatment decreases NSC proliferation and generation of mature and functional neurons. On the contrary, the generation of BrdU/S100β+ glial cells was increased resulting in neurogliogenesis imbalance in the hippocampus. Further, cypermethrin treatment also led to an increased number of BrdU/cleaved caspase-3+ and Fluoro-Jade B+ cells suggesting an induction of apoptosis in NSCs and increased degeneration of neurons in the hippocampus. Overall, these results explicate that cypermethrin exposure not only reduces the NSC pool but also disturbs the neuron-astrocyte ratio and potentiates neurodegeneration in the hippocampus, leading to cognitive dysfunctions in rats.
Collapse
Affiliation(s)
- Anuradha Yadav
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ankit Tandon
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Department of Biochemistry, School of Dental Sciences, Babu Banarasi Das University, BBD City, Faizabad Road, Lucknow, Uttar Pradesh, 226028, India
| | - Brashket Seth
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shweta Goyal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sangh Jyoti Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shashi Kant Tiwari
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- University of California San Diego, La Jolla, CA, 92093, USA
| | - Swati Agarwal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Saumya Nair
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31 Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
19
|
Al-Gholam MA, Issa NM. Effect of cypermethrin on the postnatal development of the medulla oblongata and the possible protective role of melatonin in albino rats. Anat Cell Biol 2020; 53:460-470. [PMID: 33361544 PMCID: PMC7769100 DOI: 10.5115/acb.20.193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 11/27/2022] Open
Abstract
Previous studies have shown that cypermethrin (CYP), a broad spectrum pesticide has a teratogenic effect on rat offspring born to an exposed dam with no information on its effect on the development of the brain. To the best of our knowledge, this research is the first attempt to study the postnatal development medulla oblongata of rat offspring exposed to CYP during the perinatal period and the possible neuroprotective role of melatonin. The offspring of treated female rats were organized into control, melatonin (1 mg/kg/day orally); CYP (12 mg/kg/day orally); and CYP/melatonin groups. The mothers received treatments from day 6 of gestation until day 21 after birth. At Postnatal days 7 and 21, the animals were sacrificed and their medulla oblongata was removed and subjected to histological, immunohistochemical, and electron microscopic studies. CYP induced neuronal degeneration by chromatolysis and pyknosis. Nuclear changes, cytoplasmic vacuolation, damage mitochondria, and breakdown of RER were also detected. Reduction of microtubule-associated protein-2 (MAP-2), myelin basic protein (MBP), and oligodendrocyte transcription factor expressions and increment of glial fibrillary acidic protein expression in the medulla oblongata of the developing rats were observed. On the other hand, melatonin led to an obvious improvement of the injured medulla oblongata tissues and ameliorating the damaging effects of CYP. In conclusion, melatonin has protected rats against CYP-induced histopathological and immunohistochemical changes. This may be due to the protection of MAP-2, conservation of MBP, an increment of oligodendrocytes, and alleviation of astrogliosis.
Collapse
Affiliation(s)
- Marwa A Al-Gholam
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| | - Noha M Issa
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| |
Collapse
|
20
|
Lehmler HJ, Simonsen D, Liu B, Bao W. Environmental exposure to pyrethroid pesticides in a nationally representative sample of U.S. adults and children: The National Health and Nutrition Examination Survey 2007-2012. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 267:115489. [PMID: 33254662 PMCID: PMC7708675 DOI: 10.1016/j.envpol.2020.115489] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 05/14/2023]
Abstract
Pyrethroids are an important class of insecticides, and thousands of tons of these compounds are used in the United States every year. This study characterized exposures to pyrethroids and assessed demographic, socioeconomic, and lifestyle factors that modulate pyrethroid exposure using data from the National Health and Nutrition Examination Survey (NHANES) 2007-2012, a nationally representative survey of the non-institutionalized population of the United States. Urinary levels of commonly used biomarkers of pyrethroid exposure, including 3-phenoxybenzoic acid (3-PBA), 4-fluoro-3-phenoxybenzoic acid (F-PBA), and cis-dibromovinyl-dimethylcyclopropane carboxylic acid (DBCA), were determined by liquid chromatography-tandem mass spectrometry. The detection rate of 3-PBA, a nonspecific metabolite of several pyrethroids, was 78.1% in adults (N = 5233) and 79.3% in children (N = 2295). The detection rates of all other pyrethroid metabolites were <10%. The median urinary level of 3-PBA in adults was 0.47 μg/L (interquartile range, 0.14-1.22 μg/L). For children, the median urinary level was 0.49 μg/L (interquartile range, 0.17-1.29 μg/L). Age, gender, family income-to-poverty ratio (PIR), levels of physical activity, alcohol intake, and body mass index were associated with 3-PBA levels in adults. In children, age, gender, race/ethnicity, and PIR were associated with 3-PBA levels. 3-PBA levels also differed significantly across NHANES cycles, with higher levels observed in NHANES 2011-2012. Geometric mean 3-PBA levels in U.S. adults were 0.41 μg/L in NHANES 2007-2008, 0.41 μg/L in NHANES 2009-2010, and 0.66 μg/L in NHANES 2011-2012. In U.S. children, geometric mean 3-PBA levels were 0.40 μg/L in NHANES 2007-2008, 0.46 μg/L in NHANES 2009-2010, and 0.70 μg/L in NHANES 2011-2012. These results demonstrate that pyrethroid exposures remain a current environmental health concern and lay the foundation for further preclinical and epidemiological studies assessing human health risks associated with pyrethroids.
Collapse
Affiliation(s)
- Hans-Joachim Lehmler
- Department of Occupational & Environmental Health, College of Public Health, University of Iowa, Iowa City, IA, United States; Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, United States.
| | - Derek Simonsen
- Department of Occupational & Environmental Health, College of Public Health, University of Iowa, Iowa City, IA, United States; Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, United States
| | - Buyun Liu
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, United States
| | - Wei Bao
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
21
|
Hassouna I. Transplacental neurotoxicity of cypermethrin induced astrogliosis, microgliosis and depletion of let-7 miRNAs expression in the developing rat cerebral cortex. Toxicol Rep 2020; 7:1608-1615. [PMID: 33312879 PMCID: PMC7721691 DOI: 10.1016/j.toxrep.2020.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/08/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Transplacental neurotoxicity of the pyrethroid insecticide, cypermethrin DNA alterations and immunohistochemical staining of astrocytes and microglia Cypermethrin induces astrogliosis and microgliosis in cerebral cortex MicroRNAs let7a, b, and c deplete in cerebral cortex of rat pups at postanal days
The use of type II pyrethroids, cypermethrin is becoming a growing concern among environmental research centers. While most studies have attempted to cover the areas of DNA damage and microglia activation following exposure to cypermethin in the adult or postnatal life, less is known about the exact degree of neurotoxicity that results from exposure to transplacental sublethal doses of cypermethrin. To study the transplacental neurotoxicity of cypermethrin, pregnant rats were orally administered 10 % of LD50 (25 mg/kg body weight) cypermethrin, one dose daily for one week during the gestational days 15–21. The pups were investigated at postnatal day7, 14 and 21 after birth. In brain, DNA alterations were detected, astrocytes and microglia quantification were performed and some let7 family member miRNAs are estimated. The results show a gain of three major bands in the range of 350bp to 2100bp with high intensities in cortex exposed to cypermethrin compared with similar pattern indicating unaffected genomic regions in thalamus and hypothalamus at 21days. Moreover, increases in the percentage of GFAP positive astrocytes and IBA1 positive microglia indicate astrogliosis and microgliosis respectively due to cypermethrin treatment in cerebral cortex. For the first time, drastically reduced expression of let7a, b and c members are also associated with gliosis and DNA alterations, which are detected in cerebral cortex, following transplacental neurotoxicity of cypermethrin. Taking together, these results suggest that cypermethrin neurotoxicity may be mediated partly through let7 miRNAs.
Collapse
Affiliation(s)
- Imam Hassouna
- Physiology Unit, Zoology Department, Faculty of Science, Menoufia University, Shebin Elkom, Egypt
| |
Collapse
|