1
|
Thao PN, Nishijo M, Tai PT, Nghi TN, Yokawa T, Hoa VT, Tien TV, Kien NX, Anh TH, Nishino Y, Nishijo H. Impacts of dioxin exposure on brain connectivity estimated by DTI analysis of MRI images in men residing in contaminated areas of Vietnam. Front Neurosci 2024; 18:1344653. [PMID: 38726030 PMCID: PMC11079160 DOI: 10.3389/fnins.2024.1344653] [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: 11/26/2023] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Effects of dioxin exposure on gray matter volume have been reported in previous studies, but a few studies reported effects of dioxin exposure on white matter structure. Therefore, this study was undertaken to investigate the impact of dioxin exposure on white matter microstructure in men living in the most severely dioxin-contaminated areas in Vietnam. Methods In 2019 brain MRI scans from 28 men living near Bien Hoa airbase were obtained at Dong Nai General Hospital, Vietnam, on a 3 T scanner using a conventional diffusion tensor imaging sequence. Two exposure markers were indicated by perinatal exposure estimated by assessment of maternal residency in a dioxin-contaminated area during pregnancy and by measurement of blood dioxin levels. A general linear model was used to compare fractional anisotropy (FA) values in 11 white matter tracts in both hemispheres between groups with and without perinatal dioxin exposure and groups with high and low blood dioxin levels after adjusting for covariates. Results The adjusted mean FA value in the left cingulum hippocampal part (CGH) was significantly lower in the perinatal dioxin exposure group compared with the group without perinatal dioxin exposure. The high blood TCDD group showed significantly reduced FA values in the left and right CGH and right uncinate fasciculus (UNC). Moreover, the high blood TEQ-PCDDs group showed significantly lower FA values in the left and right CGH and the left UNC. There were no significant differences in FA values between the groups with high and low TEQ-PCDFs levels or between the groups with high and low TEQ-PCDD/Fs levels. Discussion It was concluded that dioxin exposure during the perinatal period and adulthood may alter the microstructure of white matter tracts in individuals with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Pham Ngoc Thao
- Department of Functional Diagnosis, Military Hospital 103, Vietnam Military Medical University, Ha Noi, Vietnam
| | - Muneko Nishijo
- Department of Epidemiology and Public Health, Kanazawa Medical University, Ishikawa, Japan
| | - Pham The Tai
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Ha Noi, Vietnam
| | - Tran Ngoc Nghi
- Ministry of Health, Vietnamese Government, Hanoi, Vietnam
| | | | - Vu Thi Hoa
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Ha Noi, Vietnam
| | - Tran Viet Tien
- Department of Infectious and Tropical Diseases, Military Hospital 103, Vietnam Military Medical University, Ha Noi, Vietnam
| | - Nguyen Xuan Kien
- Department of Military Medical Command and Organization, Vietnam Military Medical University, Ha Noi, Vietnam
| | - Tran Hai Anh
- Department of Physiology, Vietnam Military Medical University, Ha Noi, Vietnam
| | - Yoshikazu Nishino
- Department of Epidemiology and Public Health, Kanazawa Medical University, Ishikawa, Japan
| | - Hisao Nishijo
- Department of Sport and Health Sciences, Faculty of Human Sciences, University of East Asia, Yamaguchi, Japan
| |
Collapse
|
2
|
Vu HT, Pham TN, Nishijo M, Yokawa T, Pham The T, Takiguchi T, Nishino Y, Nishijo H. Impact of dioxin exposure on brain morphometry and social anxiety in men living in the most dioxin-contaminated area in Vietnam. J Psychiatr Res 2023; 166:169-177. [PMID: 37774667 DOI: 10.1016/j.jpsychires.2023.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 06/16/2023] [Accepted: 09/13/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND Previously, we reported that the global brain volume was significantly higher in men with estimated perinatal dioxin exposure in Vietnam. In this study, we aimed to clarify which brain lobes, consisting of several gyri, contributed to the increased global brain volume. We then analyzed associations between changes in brain volume and social anxiety symptoms to investigate the role of structural changes of the brain on social anxiety following perinatal dioxin exposure. METHODS Thirty-three men living near the dioxin-contaminated Bien Hoa airbase, underwent MRI examination. The regional gray matter volumes were extracted using the SPM12 in a MATLAB environment, and compared between subgroups with and without perinatal dioxin exposure estimated from their maternal residency in Bien Hoa during pregnancy. The social anxiety questionnaire for adults (SAQ-A30) was used to assess social anxiety. RESULTS In both hemispheres, higher gray matter volume in the frontal and temporal lobes, particularly in the superior frontal gyrus, superior temporal gyrus, and temporal pole were found in men with perinatal dioxin exposure. Superior temporal gyrus volume was significantly higher in men with perinatal exposure also after adjusting for reduction of its volume associated with increasing 1,2,3,7,8-PentaCDD levels in blood. Increased volume of these gyri, which showed higher volume in men with perinatal exposure, was associated with increased SAQ-A30 scores. CONCLUSION Perinatal dioxin exposure may increase the brain volume of gyri involved in social behavior, which was associated with increased social anxiety symptoms, suggesting functional effects accompanied the morphological effects of dioxin exposure.
Collapse
Affiliation(s)
- Hoa Thi Vu
- Department of Public Health, Kanazawa Medical University, Japan; Biomedical and Pharmaceutical Research Center, Vietnam Military Medical University, Hanoi, Viet Nam
| | - Thao Ngoc Pham
- Department of Functional Diagnosis, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Viet Nam
| | - Muneko Nishijo
- Department of Public Health, Kanazawa Medical University, Japan.
| | | | - Tai Pham The
- Biomedical and Pharmaceutical Research Center, Vietnam Military Medical University, Hanoi, Viet Nam
| | | | | | | |
Collapse
|
3
|
Kimura E, Suzuki G, Uramaru N, Kakeyama M, Maekawa F. 2-Chloro-3,7,8-tribromodibenzofuran as a new environmental pollutant inducing atypical ultrasonic vocalization in infant mice. Toxicol Res (Camb) 2023; 12:999-1004. [PMID: 37915473 PMCID: PMC10615804 DOI: 10.1093/toxres/tfad069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/22/2023] [Accepted: 08/08/2023] [Indexed: 11/03/2023] Open
Abstract
Epidemiological and experimental studies indicate that maternal exposure to environmental pollutants impairs the cognitive and motor functions of offspring in humans and laboratory animals. Infant ultrasonic vocalizations (USVs), the communicative behavior of pups toward caregivers, are impaired in rodent models of neurodevelopmental disorders, suggesting a useful method to evaluate the developmental neurotoxicity of environmental pollutants. Therefore, we investigated USVs emitted by mouse pups of dams exposed to 2-chloro-3,7,8-tribromodibenzofuran (TeXDF) and 1,2,3,7,8-pentabromodibenzofuran (PeBDF), which are detected in the actual environment. The USV duration and number in the pups born to dams administered with TeXDF 40 μg/kg body weight (b.w.), but not 8 μg/kg b.w., on gestational day (GD) 12.5, were significantly lower than those in the corresponding pups on postnatal days 3-9. Conversely, there was no statistical change in the USVs emitted by the pups of dams administered with PeBDF 35 or 175 μg/kg b.w. on GD 12.5. To examine whether maternal exposure leads to behavioral impairments in adulthood, we analyzed exploratory behaviors in a novel environment using IntelliCage, a fully automated testing apparatus for group-housed mice. Neither TeXDF nor PeBDF exposure induced significant differences in offspring exploration. Considered together, our findings revealed that TeXDF induces atypical USV emission in infant mice, suggesting the importance of further studies on the risk assessment of mixed brominated/chlorinated dibenzo-p-dioxins and dibenzofurans.
Collapse
Affiliation(s)
- Eiki Kimura
- Health and Environmental Risk Research Division, National Institute for Environmental Studies, Tsukuba 305-8506, Japan
- Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
- Department of Environmental Health, University of Fukui School of Medical Sciences, Fukui 910-1193, Japan
| | - Go Suzuki
- Material Cycles Division, National Institute for Environmental Studies, Tsukuba 305-8506, Japan
| | - Naoto Uramaru
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Masaki Kakeyama
- Faculty of Human Sciences, Waseda University, Saitama 359-1192, Japan
| | - Fumihiko Maekawa
- Health and Environmental Risk Research Division, National Institute for Environmental Studies, Tsukuba 305-8506, Japan
| |
Collapse
|
4
|
Madison CA, Debler RA, Vardeleon NI, Hillbrick L, Jayaraman A, Safe S, Chapkin RS, Eitan S. Sex-dependent differences in the stress mitigating and antidepressant effects of selective aryl hydrocarbon receptor modulators. J Affect Disord 2022; 319:213-220. [PMID: 36206882 PMCID: PMC10391660 DOI: 10.1016/j.jad.2022.09.155] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Our recent study demonstrated that selective aryl hydrocarbon receptor modulators (SAhRMs), such as 1,4-dihydroxy-2-napthoic acid (DHNA) act as antidepressants in female mice. Given that some effects of certain SAhRMs are known to also be mediated via estrogen receptor signaling, this study examined whether the effects of SAhRMs on mood, emotional state, and cognition are sex-dependent. METHODS C57BL/6N mice were fed with vehicle or 20 mg/kg DHNA for three weeks prior to four weeks of unpredictable chronic mild stress (UCMS). Mice were examined for depression-like behaviors (sucrose preference, forced swim test (FST), splash test, tape groom test), emotional state (open-field test, light/dark test, marble burying, novelty-induced hypophagia, elevated-plus maze), and cognition (object location recognition, novel object recognition, Morris water maze). RESULTS In females, UCMS decreased sucrose preference and increased FST immobility time; both effects were prevented by DHNA. In males, UCMS increased FST immobility time, and increased the latency to groom in the splash test. These effects were not mitigated by DHNA. However, in males, UCMS induced an increase in novelty-induced locomotion, an increase in the time spent in the light compartment in the L/D test, and an increase in the time spent with an object in a novel location. These effects were prevented by DHNA. CONCLUSIONS Our findings indicate that DHNA has high potential to act as antidepressants in females. However, given classical interpretation, DHNA did not appear to act as an antidepressant in males. Nonetheless, our findings indicate that DHNA can mitigate stress effects and reactivity in males.
Collapse
Affiliation(s)
- Caitlin A Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Roanna A Debler
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Nathan I Vardeleon
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Lauren Hillbrick
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, College Station, 4235 TAMU, TX 77843, USA.
| |
Collapse
|
5
|
Martin NR, Patel R, Kossack ME, Tian L, Camarillo MA, Cintrón-Rivera LG, Gawdzik JC, Yue MS, Nwagugo FO, Elemans LMH, Plavicki JS. Proper modulation of AHR signaling is necessary for establishing neural connectivity and oligodendrocyte precursor cell development in the embryonic zebrafish brain. Front Mol Neurosci 2022; 15:1032302. [PMID: 36523606 PMCID: PMC9745199 DOI: 10.3389/fnmol.2022.1032302] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
2,3,7,8-tetrachlorodibenzo-[p]-dioxin (TCDD) is a persistent global pollutant that exhibits a high affinity for the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. Epidemiological studies have associated AHR agonist exposure with multiple human neuropathologies. Consistent with the human data, research studies using laboratory models have linked pollutant-induced AHR activation to disruptions in learning and memory as well as motor impairments. Our understanding of endogenous AHR functions in brain development is limited and, correspondingly, scientists are still determining which cell types and brain regions are sensitive to AHR modulation. To identify novel phenotypes resulting from pollutant-induced AHR activation and ahr2 loss of function, we utilized the optically transparent zebrafish model. Early embryonic TCDD exposure impaired embryonic brain morphogenesis, resulted in ventriculomegaly, and disrupted neural connectivity in the optic tectum, habenula, cerebellum, and olfactory bulb. Altered neural network formation was accompanied by reduced expression of synaptic vesicle 2. Loss of ahr2 function also impaired nascent network development, but did not affect gross brain or ventricular morphology. To determine whether neural AHR activation was sufficient to disrupt connectivity, we used the Gal4/UAS system to express a constitutively active AHR specifically in differentiated neurons and observed disruptions only in the cerebellum; thus, suggesting that the phenotypes resulting from global AHR activation likely involve multiple cell types. Consistent with this hypothesis, we found that TCDD exposure reduced the number of oligodendrocyte precursor cells and their derivatives. Together, our findings indicate that proper modulation of AHR signaling is necessary for the growth and maturation of the embryonic zebrafish brain.
Collapse
Affiliation(s)
- Nathan R. Martin
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Ratna Patel
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Michelle E. Kossack
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Lucy Tian
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Manuel A. Camarillo
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Layra G. Cintrón-Rivera
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Joseph C. Gawdzik
- Molecular and Environmental Toxicology Center, University of Wisconsin at Madison, Madison, WI, United States,Division of Pharmaceutical Sciences, University of Wisconsin at Madison, Madison, WI, United States
| | - Monica S. Yue
- Molecular and Environmental Toxicology Center, University of Wisconsin at Madison, Madison, WI, United States,Division of Pharmaceutical Sciences, University of Wisconsin at Madison, Madison, WI, United States
| | - Favour O. Nwagugo
- Department of Biology, University of Maryland Baltimore County, Baltimore, MD, United States
| | - Loes M. H. Elemans
- Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, Netherlands
| | - Jessica S. Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States,*Correspondence: Jessica S. Plavicki,
| |
Collapse
|
6
|
Hua J, Wang X, Zhu J, Wang Q, Zhang W, Lei L, Zhu B, Han J, Yang L, Zhou B. Decabromodiphenyl ethane induced hyperactivity in developing zebrafish at environmentally relevant concentrations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114044. [PMID: 36055044 DOI: 10.1016/j.ecoenv.2022.114044] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/11/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
Decabromodiphenyl ethane (DBDPE), a widely used novel brominated flame retardant, is gaining concerns due to rapidly increased contents in various environmental and biota samples. In the present study, zebrafish (Danio rerio) embryos were exposed to 2.91, 9.71, 29.14 and 97.12 μg/L of DBDPE until 120 h post-fertilization (hpf) to investigate the potential developmental neurotoxicity and underlying mechanisms. Chemical analysis revealed concentration-dependently increased body burdens of DBDPE in zebrafish larvae, with bioaccumulation factors (BCFs) ranging from 414 to 726. Embryonic exposure to DBDPE caused hyperactivity without affecting the development of secondary motoneuron axons and muscle fibers. However, further results implicated that DBDPE may affect the locomotor regulatory network via different mechanisms at lower and higher concentrations. On the one hand, embryonic exposure to 2.91 μg/L DBDPE transiently promoted spontaneous coiling contractions, but showed no effects on touch-response and swimming activity in zebrafish larvae. The whole-body contents of neurotransmitters were significantly decreased. Significant decreased protein abundances of α1-TUBULIN and SYN2a and molecular docking results pointed out possible interactions of DBDPE with these two proteins. However, these changes may be unconcerned with the transient hyperactivity, and the exact molecular mechanisms need further investigation. On the other hand, 29.14 and 97.12 μg/L DBDPE exposure caused longer-lasting effects in promoting spontaneous coiling contractions, and also touch-response and swimming activity. At the same time, increased ACh contents (without changes of other neurotransmitters) and ChAT activity and inhibited transcription of nAChRs were observed at higher concentrations. Molecular docking indicated direct interaction of DBDPE with ChAT. The results suggested that DBDPE induced hyperactivity at higher concentrations was probably involved with disrupted cholinergic system, with ChAT as a potential target. Given that the body burden of DBDPE in lower concentration group was comparable with those detected in wild fish, the current results may provide useful information for ecological risk assessment.
Collapse
Affiliation(s)
- Jianghuan Hua
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xiulin Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaping Zhu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Qiangwei Wang
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Wei Zhang
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resource and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Lei Lei
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Biran Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Jian Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
7
|
Alterations in Regional Brain Regional Volume Associated with Dioxin Exposure in Men Living in the Most Dioxin-Contaminated Area in Vietnam: Magnetic Resonance Imaging (MRI) Analysis Using Voxel-Based Morphometry (VBM). TOXICS 2021; 9:toxics9120353. [PMID: 34941787 PMCID: PMC8703540 DOI: 10.3390/toxics9120353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 12/25/2022]
Abstract
To clarify the influence of dioxin exposure on brain morphometry, the present study investigated associations between dioxin exposure at high levels and brain structural irregularities in 32 Vietnamese men. Two exposure markers were used: blood dioxin levels, as a marker of exposure in adulthood, and perinatal dioxin exposure, estimated by maternal residency in a dioxin-contaminated area during pregnancy. All subjects underwent brain magnetic resonance imaging (MRI) scans. We analyzed correlations between regional gray matter volumes and blood dioxin levels, and compared regional volumes between men with and without perinatal dioxin exposure using the voxel-based morphometry (VBM) tool from Statistical Parametric Mapping 12 (SPM12). Blood 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was associated with low volume of the medial temporal pole and fusiform gyrus. Toxic equivalency (TEQ)-PCDDs were correlated with low medial temporal pole volume. However, 1,2,3,4,7,8-HxCDD was associated with high middle frontal gyrus and cerebellum volume. In men with perinatal dioxin exposure, the left inferior frontal gyrus pars orbitalis volume was significantly lower than in those without perinatal exposure. These results suggest that dioxin exposure during the perinatal period and in adulthood may alter regional brain volume, which might lead to cognitive deficits and unusual social emotional behavior in Vietnamese men living in dioxin-contaminated areas.
Collapse
|
8
|
Buck JM, Yu L, Knopik VS, Stitzel JA. DNA methylome perturbations: an epigenetic basis for the emergingly heritable neurodevelopmental abnormalities associated with maternal smoking and maternal nicotine exposure†. Biol Reprod 2021; 105:644-666. [PMID: 34270696 PMCID: PMC8444709 DOI: 10.1093/biolre/ioab138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/29/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
Maternal smoking during pregnancy is associated with an ensemble of neurodevelopmental consequences in children and therefore constitutes a pressing public health concern. Adding to this burden, contemporary epidemiological and especially animal model research suggests that grandmaternal smoking is similarly associated with neurodevelopmental abnormalities in grandchildren, indicative of intergenerational transmission of the neurodevelopmental impacts of maternal smoking. Probing the mechanistic bases of neurodevelopmental anomalies in the children of maternal smokers and the intergenerational transmission thereof, emerging research intimates that epigenetic changes, namely DNA methylome perturbations, are key factors. Altogether, these findings warrant future research to fully elucidate the etiology of neurodevelopmental impairments in the children and grandchildren of maternal smokers and underscore the clear potential thereof to benefit public health by informing the development and implementation of preventative measures, prophylactics, and treatments. To this end, the present review aims to encapsulate the burgeoning evidence linking maternal smoking to intergenerational epigenetic inheritance of neurodevelopmental abnormalities, to identify the strengths and weaknesses thereof, and to highlight areas of emphasis for future human and animal model research therein.
Collapse
Affiliation(s)
- Jordan M Buck
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, USA
| | - Li Yu
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA
| | - Valerie S Knopik
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, USA
| |
Collapse
|
9
|
Bhuju J, Olesen KM, Muenyi CS, Patel TS, Read RW, Thompson L, Skalli O, Zheng Q, Grice EA, Sutter CH, Sutter TR. Cutaneous Effects of In Utero and Lactational Exposure of C57BL/6J Mice to 2,3,7,8-Tetrachlorodibenzo- p-dioxin. TOXICS 2021; 9:toxics9080192. [PMID: 34437510 PMCID: PMC8402454 DOI: 10.3390/toxics9080192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023]
Abstract
To determine the cutaneous effects of in utero and lactational exposure to the AHR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), pregnant C57BL/6J mice were exposed by gavage to a vehicle or 5 μg TCDD/kg body weight at embryonic day 12 and epidermal barrier formation and function were studied in their offspring from postnatal day 1 (P1) through adulthood. TCDD-exposed pups were born with acanthosis. This effect was AHR-dependent and subsided by P6 with no evidence of subsequent inflammatory dermatitis. The challenge of adult mice with MC903 showed similar inflammatory responses in control and treated animals, indicating no long-term immunosuppression to this chemical. Chloracne-like sebaceous gland hypoplasia and cyst formation were observed in TCDD-exposed P21 mice, with concomitant microbiome dysbiosis. These effects were reversed by P35. CYP1A1 and CYP1B1 expression in the skin was increased in the exposed mice until P21, then declined. Both CYP proteins co-localized with LRIG1-expressing progenitor cells at the infundibulum. CYP1B1 protein also co-localized with a second stem cell niche in the isthmus. These results indicate that this exposure to TCDD causes a chloracne-like effect without inflammation. Transient activation of the AhR, due to the shorter half-life of TCDD in mice, likely contributes to the reversibility of these effects.
Collapse
Affiliation(s)
- Jyoti Bhuju
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA
| | - Kristin M Olesen
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA
| | - Clarisse S Muenyi
- Department of Surgery, University of Tennessee Health Sciences Center, Memphis, TN 38104, USA
| | - Tejesh S Patel
- Kaplan-Amonette Department of Dermatology, University of Tennessee Health Sciences Center, Memphis, TN 38104, USA
| | - Robert W Read
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA
| | - Lauren Thompson
- Integrated Microscopy Center, University of Memphis, Memphis, TN 38152, USA
| | - Omar Skalli
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA
- Integrated Microscopy Center, University of Memphis, Memphis, TN 38152, USA
| | - Qi Zheng
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth A Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carrie Hayes Sutter
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA
- W. Harry Feinstone Center for Genomic Research, University of Memphis, Memphis, TN 38152, USA
| | - Thomas R Sutter
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA
- W. Harry Feinstone Center for Genomic Research, University of Memphis, Memphis, TN 38152, USA
| |
Collapse
|
10
|
Kimura E, Kohda M, Maekawa F, Fujii-Kuriyama Y, Tohyama C. Neurons expressing the aryl hydrocarbon receptor in the locus coeruleus and island of Calleja major are novel targets of dioxin in the mouse brain. Histochem Cell Biol 2021; 156:147-163. [PMID: 33963922 PMCID: PMC8397641 DOI: 10.1007/s00418-021-01990-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 12/14/2022]
Abstract
The aryl hydrocarbon receptor (AhR) acts as a receptor that responds to ligands, including dioxin. The AhR-ligand complex translocates from the cytoplasm into the nucleus to induce gene expression. Because dioxin exposure impairs cognitive and neurobehavioral functions, AhR-expressing neurons need to be identified for elucidation of the dioxin neurotoxicity mechanism. Immunohistochemistry was performed to detect AhR-expressing neurons in the mouse brain and confirm the specificity of the anti-AhR antibody using Ahr-/- mice. Intracellular distribution of AhR and expression level of AhR-target genes, Cyp1a1, Cyp1b1, and Ahr repressor (Ahrr), were analyzed by immunohistochemistry and quantitative RT-PCR, respectively, using mice exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The mouse brains were shown to harbor AhR in neurons of the locus coeruleus (LC) and island of Calleja major (ICjM) during developmental period in Ahr+/+ mice but not in Ahr-/- mice. A significant increase in nuclear AhR of ICjM neurons but not LC neurons was found in 14-day-old mice compared to 5- and 7-day-old mice. AhR was significantly translocated into the nucleus in LC and ICjM neurons of TCDD-exposed adult mice. Additionally, the expression levels of Cyp1a1, Cyp1b1, and Ahrr genes in the brain, a surrogate of TCDD in the tissue, were significantly increased by dioxin exposure, suggesting that dioxin-activated AhR induces gene expression in LC and ICjM neurons. This histochemical study shows the ligand-induced nuclear translocation of AhR at the single-neuron level in vivo. Thus, the neurotoxicological significance of the dioxin-activated AhR in the LC and ICjM warrants further studies.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, 305-8506, Japan. .,Research Fellow, Japan Society for the Promotion of Science (JSPS), 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan.
| | - Masanobu Kohda
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, 305-8506, Japan
| | - Fumihiko Maekawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, 305-8506, Japan
| | - Yoshiaki Fujii-Kuriyama
- Medical Research Institute, Molecular Epidemiology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, 305-8506, Japan. .,Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan.
| |
Collapse
|
11
|
Latchney SE, Majewska AK. Persistent organic pollutants at the synapse: Shared phenotypes and converging mechanisms of developmental neurotoxicity. Dev Neurobiol 2021; 81:623-652. [PMID: 33851516 DOI: 10.1002/dneu.22825] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/27/2021] [Accepted: 04/09/2021] [Indexed: 12/18/2022]
Abstract
The developing nervous system is sensitive to environmental and physiological perturbations in part due to its protracted period of prenatal and postnatal development. Epidemiological and experimental studies link developmental exposures to persistent organic pollutants (POPs) including polychlorinated biphenyls, polychlorinated dibenzo-p-dioxins, polybrominated diphenyl ethers, and benzo(a)pyrene to increased risk for neurodevelopmental disorders in children. Mechanistic studies reveal that many of the complex cellular processes that occur during sensitive periods of rapid brain development are cellular targets for developmental neurotoxicants. One area of research interest has focused on synapse formation and plasticity, processes that involve the growth and retraction of dendrites and dendritic spines. For each chemical discussed in this review, we summarize the morphological and electrophysiological data that provide evidence that developmental POP exposure produces long-lasting effects on dendritic morphology, spine formation, glutamatergic and GABAergic signaling systems, and synaptic transmission. We also discuss shared intracellular mechanisms, with a focus on calcium and thyroid hormone homeostasis, by which these chemicals act to modify synapses. We conclude our review highlighting research gaps that merit consideration when characterizing synaptic pathology elicited by chemical exposure. These gaps include low-dose and nonmonotonic dose-response effects, the temporal relationship between dendritic growth, spine formation, and synaptic activity, excitation-inhibition balance, hormonal effects, and the need for more studies in females to identify sex differences. By identifying converging pathological mechanisms elicited by POP exposure at the synapse, we can define future research directions that will advance our understanding of these chemicals on synapse structure and function.
Collapse
Affiliation(s)
- Sarah E Latchney
- Department of Biology, St. Mary's College of Maryland, St. Mary's City, MD, USA.,Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Ania K Majewska
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, USA.,Center for Visual Science, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
12
|
Gileadi TE, Swamy AK, Hore Z, Horswell S, Ellegood J, Mohan C, Mizuno K, Lundebye AK, Giese KP, Stockinger B, Hogstrand C, Lerch JP, Fernandes C, Basson MA. Effects of Low-Dose Gestational TCDD Exposure on Behavior and on Hippocampal Neuron Morphology and Gene Expression in Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:57002. [PMID: 33956508 PMCID: PMC8101924 DOI: 10.1289/ehp7352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 02/19/2021] [Accepted: 03/29/2021] [Indexed: 05/03/2023]
Abstract
BACKGROUND 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a persistent and toxic environmental pollutant. Gestational exposure to TCDD has been linked to cognitive and motor deficits, and increased incidence of autism spectrum disorder (ASD) traits in children. Most animal studies of these neurodevelopmental effects involve acute TCDD exposure, which does not model typical exposure in humans. OBJECTIVES The aim of the study was to establish a dietary low-dose gestational TCDD exposure protocol and performed an initial characterization of the effects on offspring behavior, neurodevelopmental phenotypes, and gene expression. METHODS Throughout gestation, pregnant C57BL/6J mice were fed a diet containing a low dose of TCDD (9 ng TCDD/kg body weight per day) or a control diet. The offspring were tested in a battery of behavioral tests, and structural brain alterations were investigated by magnetic resonance imaging. The dendritic morphology of pyramidal neurons in the hippocampal Cornu Ammonis (CA)1 area was analyzed. RNA sequencing was performed on hippocampi of postnatal day 14 TCDD-exposed and control offspring. RESULTS TCDD-exposed females displayed subtle deficits in motor coordination and reversal learning. Volumetric difference between diet groups were observed in regions of the hippocampal formation, mammillary bodies, and cerebellum, alongside higher dendritic arborization of pyramidal neurons in the hippocampal CA1 region of TCDD-exposed females. RNA-seq analysis identified 405 differentially expressed genes in the hippocampus, enriched for genes with functions in regulation of microtubules, axon guidance, extracellular matrix, and genes regulated by SMAD3. DISCUSSION Exposure to 9 ng TCDD/kg body weight per day throughout gestation was sufficient to cause specific behavioral and structural brain phenotypes in offspring. Our data suggest that alterations in SMAD3-regulated microtubule polymerization in the developing postnatal hippocampus may lead to an abnormal morphology of neuronal dendrites that persists into adulthood. These findings show that environmental low-dose gestational exposure to TCDD can have significant, long-term impacts on brain development and function. https://doi.org/10.1289/EHP7352.
Collapse
Affiliation(s)
- Talia E. Gileadi
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Abhyuday K. Swamy
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Zoe Hore
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Stuart Horswell
- Department of Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Ontario, Canada
| | - Conor Mohan
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Keiko Mizuno
- Department of Basic and Clinical Neuroscience, King’s College London, London, UK
| | | | - K. Peter Giese
- Department of Basic and Clinical Neuroscience, King’s College London, London, UK
| | | | | | - Jason P. Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - M. Albert Basson
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| |
Collapse
|
13
|
Kimura E, Suzuki G, Uramaru N, Endo T, Maekawa F. Behavioral impairments in infant and adult mouse offspring exposed to 2,3,7,8-tetrabromodibenzofuran in utero and via lactation. ENVIRONMENT INTERNATIONAL 2020; 142:105833. [PMID: 32559560 DOI: 10.1016/j.envint.2020.105833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/19/2020] [Accepted: 05/22/2020] [Indexed: 06/11/2023]
Abstract
Polybrominated dibenzo-p-dioxins and dibenzofurans (PBDD/DFs) have been unintentionally produced and emitted from the lifecycle of products containing brominated flame retardants, such as polybrominated diphenyl ether, which is suspected to cause developmental neurotoxicity (DNT). Although it is plausible that PBDD/DFs can also induce DNT, information regarding their neurotoxic potential is currently limited. Hence, in the present study, we examined the effects of in utero and lactational exposure to brominated dibenzofurans on infant and adult offspring behavior to understand the mechanism of PBDD/DFs toxicity and detect effective behavioral endpoints in DNT assessment. We analyzed the behavior of mouse offspring born to dams administered 2,3,7,8-tetrabromodibenzofuran (2,3,7,8-TeBDF; dose of 0, 9, or 45 μg/kg) or 2,3,8-tribromodibenzofuran (2,3,8-TrBDF; dose of 0, 75.6, or 378 μg/kg) on gestational day 12.5. In mouse offspring born to dams exposed to 2,3,7,8-TeBDF, the exploratory behavior in a novel environment in adulthood and ultrasonic vocalization (USV) during infancy were significantly reduced. Additionally, AhR-target genes, such as Cyp1a1, were induced in the liver of 2,3,7,8-TeBDF-exposed offspring in a dose-dependent manner. Conversely, no significant changes in the infant and adult behaviors and expression level of AhR-target genes were observed in the 2,3,8-TrBDF-exposed offspring. These results suggest that 2,3,7,8-TeBDF can induce DNT and that the analysis of exploratory behavior in a novel environment and USV may be useful endpoints to assess DNT of dioxin-related substances.
Collapse
Affiliation(s)
- Eiki Kimura
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan; Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan
| | - Go Suzuki
- Center for Material Cycles and Waste Management Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan
| | - Naoto Uramaru
- Nihon Pharmaceutical University, 10281 Komuro Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Toshihiro Endo
- Phenovance Research & Technology, 5-4-19-302A, Kashiwanoha, Kashiwa, Chiba 277-0882, Japan
| | - Fumihiko Maekawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan.
| |
Collapse
|
14
|
Zhu X, Wu Y, Li C, Yan W, Pan J, Wang S, Zhao S. Prenatal Exposure to Gossypol Impairs Corticogenesis of Mouse. Front Neurosci 2020; 14:318. [PMID: 32317927 PMCID: PMC7146080 DOI: 10.3389/fnins.2020.00318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/18/2020] [Indexed: 12/16/2022] Open
Abstract
Gossypol is a yellow polyphenolic compounds extracted from roots, stems and seeds of cotton plants. Excessive intake of gossypol induces severe pathological signs of toxicity in livestock and wildlife. Currently, gossypol has received widespread attention for its toxic effects on the reproductive system. However, reports of the effects of gossypol during corticogenesis and the development of the mouse cerebral cortex are unavailable. In the present study, gossypol was orally administrated at a dose of 0, 20, and 50 mg/kg body weight/day to pregnant mice from embryonic day 6.5 to the time of sample collection. We used in utero electroporation and immunofluorescence to demonstrate that gossypol impaired cortical neuronal migration. Furthermore, labeling with 5-bromo-2-deoxyuridine and western blot analysis revealed that gossypol disturbed the balance between proliferation and differentiation of neural progenitors, inhibited neural progenitor cell proliferation, neuronal differentiation, and maturation. Additionally, cortical progenitor apoptotic cell death increased in the developing gossypol-treated cortex, which was associated with NF-κB and MAPK pathways. In conclusion, our findings indicate that gossypol exposure disrupted neurogenesis in the developing neocortex, suggesting the potentially harmful impact of gossypol on the cerebral cortex development of humans and livestock.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Cixia Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wenyong Yan
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jiarong Pan
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Shuzhong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
15
|
Klocke C, Lein PJ. Evidence Implicating Non-Dioxin-Like Congeners as the Key Mediators of Polychlorinated Biphenyl (PCB) Developmental Neurotoxicity. Int J Mol Sci 2020; 21:E1013. [PMID: 32033061 PMCID: PMC7037228 DOI: 10.3390/ijms21031013] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 12/15/2022] Open
Abstract
Despite being banned from production for decades, polychlorinated biphenyls (PCBs) continue to pose a significant risk to human health. This is due to not only the continued release of legacy PCBs from PCB-containing equipment and materials manufactured prior to the ban on PCB production, but also the inadvertent production of PCBs as byproducts of contemporary pigment and dye production. Evidence from human and animal studies clearly identifies developmental neurotoxicity as a primary endpoint of concern associated with PCB exposures. However, the relative role(s) of specific PCB congeners in mediating the adverse effects of PCBs on the developing nervous system, and the mechanism(s) by which PCBs disrupt typical neurodevelopment remain outstanding questions. New questions are also emerging regarding the potential developmental neurotoxicity of lower chlorinated PCBs that were not present in the legacy commercial PCB mixtures, but constitute a significant proportion of contemporary human PCB exposures. Here, we review behavioral and mechanistic data obtained from experimental models as well as recent epidemiological studies that suggest the non-dioxin-like (NDL) PCBs are primarily responsible for the developmental neurotoxicity associated with PCBs. We also discuss emerging data demonstrating the potential for non-legacy, lower chlorinated PCBs to cause adverse neurodevelopmental outcomes. Molecular targets, the relevance of PCB interactions with these targets to neurodevelopmental disorders, and critical data gaps are addressed as well.
Collapse
Affiliation(s)
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA;
| |
Collapse
|
16
|
Loss of Tiparp Results in Aberrant Layering of the Cerebral Cortex. eNeuro 2019; 6:ENEURO.0239-19.2019. [PMID: 31704703 PMCID: PMC6883171 DOI: 10.1523/eneuro.0239-19.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/13/2019] [Accepted: 10/10/2019] [Indexed: 01/26/2023] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly-ADP-ribose polymerase (TIPARP) is an enzyme that adds a single ADP-ribose moiety to itself or other proteins. Tiparp is highly expressed in the brain; however, its function in this organ is unknown. Here, we used Tiparp–/– mice to determine Tiparp’s role in the development of the prefrontal cortex. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly-ADP-ribose polymerase (TIPARP) is an enzyme that adds a single ADP-ribose moiety to itself or other proteins. Tiparp is highly expressed in the brain; however, its function in this organ is unknown. Here, we used Tiparp–/– mice to determine Tiparp’s role in the development of the prefrontal cortex. Loss of Tiparp resulted in an aberrant organization of the mouse cortex, where the upper layers presented increased cell density in the knock-out mice compared with wild type. Tiparp loss predominantly affected the correct distribution and number of GABAergic neurons. Furthermore, neural progenitor cell proliferation was significantly reduced. Neural stem cells (NSCs) derived from Tiparp–/– mice showed a slower rate of migration. Cytoskeletal components, such as α-tubulin are key regulators of neuronal differentiation and cortical development. α-tubulin mono-ADP ribosylation (MAR) levels were reduced in Tiparp–/– cells, suggesting that Tiparp plays a role in the MAR of α-tubulin. Despite the mild phenotype presented by Tiparp–/– mice, our findings reveal an important function for Tiparp and MAR in the correct development of the cortex. Unravelling Tiparp’s role in the cortex, could pave the way to a better understanding of a wide spectrum of neurological diseases which are known to have increased expression of TIPARP.
Collapse
|
17
|
Nghiem GT, Nishijo M, Pham TN, Ito M, Pham TT, Tran AH, Nishimaru H, Nishino Y, Nishijo H. Adverse effects of maternal dioxin exposure on fetal brain development before birth assessed by neonatal electroencephalography (EEG) leading to poor neurodevelopment; a 2-year follow-up study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 667:718-729. [PMID: 30849612 DOI: 10.1016/j.scitotenv.2019.02.395] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/13/2019] [Accepted: 02/25/2019] [Indexed: 06/09/2023]
Abstract
We previously reported the adverse effects of perinatal dioxin exposure on child neurodevelopment around a former US military airbase in Vietnam. In the present study, we investigated the effects of maternal dioxin exposure on fetal brain development, which may predict neurodevelopmental outcomes in early childhood. A total of 55 newborns with mothers from dioxin-contaminated areas were recruited in the prefecture hospital in Bien Hoa, Vietnam. Dioxins in maternal breast milk collected 1 month after birth were used as a maternal exposure marker. Relative powers and coherence were computed from neonatal electroencephalogram (EEG) records during active sleep stages. Relationships between the EEG parameters and dioxin exposure markers were analyzed using linear regression and a general linear model after adjusting for gestational age, body length, and head circumference of infants at birth. Using data from 47 infants whose neurodevelopment was examined in a 2-year follow-up study, associations between EEG parameters and neurodevelopment were analyzed after adjusting for confounding factors. On the right frontal and parietal regions, relative delta powers were significantly decreased, and relative alpha and beta powers were significantly increased with increasing dioxin exposure. Increases in delta power and decreases in alpha power on the right frontal and parietal regions were associated with an increase in language scores at 2 years of age. Furthermore, intra- and inter-hemispheric coherence in theta and alpha bands were positively and inversely correlated with dioxin exposure, respectively, and increased intra-coherence in the right hemisphere was associated with lower language scores. These findings suggest that prenatal dioxin exposure affects neuronal activity and functional connectivity between brain regions, and may lead to poor language development.
Collapse
Affiliation(s)
- GiangThi Thuy Nghiem
- System Emotional Science, Graduate School of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Muneko Nishijo
- Department of Public Health, Kanazawa Medical University, Ishikawa 920-0293, Japan.
| | - Thao Ngoc Pham
- Department of Public Health, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Mika Ito
- Department of Obstetrics and Gynecology, University of Toyama, Toyama 930-0194, Japan
| | - Tai The Pham
- Biomedical and Pharmaceutical Research Center, Vietnamese Military Medical University, Hanoi, Viet Nam
| | - Anh Hai Tran
- Biomedical and Pharmaceutical Research Center, Vietnamese Military Medical University, Hanoi, Viet Nam
| | - Hiroshi Nishimaru
- System Emotional Science, Graduate School of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Yoshikazu Nishino
- Department of Public Health, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Hisao Nishijo
- System Emotional Science, Graduate School of Medicine, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
18
|
Zhang HJ, Liu YN, Xian P, Ma J, Sun YW, Chen JS, Chen X, Tang NJ. Maternal exposure to TCDD during gestation advanced sensory-motor development, but induced impairments of spatial learning and memory in adult male rat offspring. CHEMOSPHERE 2018; 212:678-686. [PMID: 30176550 DOI: 10.1016/j.chemosphere.2018.08.118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/15/2018] [Accepted: 08/23/2018] [Indexed: 05/19/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-Dioxin (TCDD) is an endocrine disrupting chemical (EDC) with high persistency. Even a low amount can pass the placental barrier during gestational exposure. Exposure to TCDD exposure can impair the development of the nervous system in children, leading to impaired learning ability in later-life. But the changes in neurobehavioral developments in infancy and childhood caused by TCDD are unknown. Pregnant Sprague-Dawley rats were given a consecutive daily dose of TCDD (200 or 800 ng/day/kg) or an equivalent volume of vehicle by gavage on gestational days 8-14 (GD 8-14) as the prenatal TCDD exposure model. In the offspring, early neurobehavioral development was assessed at postnatal day 5 (PND5) and eye-opening was monitored from PND10 onwards. Adult male offspring was tested by Morris Water Maze for spatial memory and learning ability evaluation. Hippocampus Nissl's staining and astrocyte GFAP immunohistochemistry were used to evaluate the activity of astrocytes. The results of the behavioral tests showed that gestational TCDD exposure induced premature motor activity and earlier eyes-opening, but lead to serious deficits of spatial memory and learning ability in the adult male offspring. Morphology and number of neurons in the hippocampus CA1 region was not affected, while the activity of astrocytes in the same region was significantly reduced. These data indicate that perinatal TCDD exposure induced premature neurobehavioral development but impaired the spatial learning and memory in adult male rat offspring. The decreased activity of astrocytes in the hippocampus may play a role in these adverse effects.
Collapse
Affiliation(s)
- Hua-Jing Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Ya-Nan Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Ping Xian
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Jing Ma
- Tianjin Cardiovascular Diseases Institute, Tianjin Chest Hospital, Tianjin, 300350, China.
| | - Ya-Wen Sun
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Jing-Shan Chen
- Department of Technology and Science, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Xi Chen
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| | - Nai-Jun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
19
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Nebbia CS, Oswald IP, Petersen A, Rose M, Roudot AC, Schwerdtle T, Vleminckx C, Vollmer G, Wallace H, Fürst P, Håkansson H, Halldorsson T, Lundebye AK, Pohjanvirta R, Rylander L, Smith A, van Loveren H, Waalkens-Berendsen I, Zeilmaker M, Binaglia M, Gómez Ruiz JÁ, Horváth Z, Christoph E, Ciccolallo L, Ramos Bordajandi L, Steinkellner H, Hoogenboom LR. Risk for animal and human health related to the presence of dioxins and dioxin-like PCBs in feed and food. EFSA J 2018; 16:e05333. [PMID: 32625737 PMCID: PMC7009407 DOI: 10.2903/j.efsa.2018.5333] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The European Commission asked EFSA for a scientific opinion on the risks for animal and human health related to the presence of dioxins (PCDD/Fs) and DL-PCBs in feed and food. The data from experimental animal and epidemiological studies were reviewed and it was decided to base the human risk assessment on effects observed in humans and to use animal data as supportive evidence. The critical effect was on semen quality, following pre- and postnatal exposure. The critical study showed a NOAEL of 7.0 pg WHO2005-TEQ/g fat in blood sampled at age 9 years based on PCDD/F-TEQs. No association was observed when including DL-PCB-TEQs. Using toxicokinetic modelling and taking into account the exposure from breastfeeding and a twofold higher intake during childhood, it was estimated that daily exposure in adolescents and adults should be below 0.25 pg TEQ/kg bw/day. The CONTAM Panel established a TWI of 2 pg TEQ/kg bw/week. With occurrence and consumption data from European countries, the mean and P95 intake of total TEQ by Adolescents, Adults, Elderly and Very Elderly varied between, respectively, 2.1 to 10.5, and 5.3 to 30.4 pg TEQ/kg bw/week, implying a considerable exceedance of the TWI. Toddlers and Other Children showed a higher exposure than older age groups, but this was accounted for when deriving the TWI. Exposure to PCDD/F-TEQ only was on average 2.4- and 2.7-fold lower for mean and P95 exposure than for total TEQ. PCDD/Fs and DL-PCBs are transferred to milk and eggs, and accumulate in fatty tissues and liver. Transfer rates and bioconcentration factors were identified for various species. The CONTAM Panel was not able to identify reference values in most farm and companion animals with the exception of NOAELs for mink, chicken and some fish species. The estimated exposure from feed for these species does not imply a risk.
Collapse
|
20
|
Sethi S, Keil KP, Lein PJ. 3,3'-Dichlorobiphenyl (PCB 11) promotes dendritic arborization in primary rat cortical neurons via a CREB-dependent mechanism. Arch Toxicol 2018; 92:3337-3345. [PMID: 30225637 PMCID: PMC6196112 DOI: 10.1007/s00204-018-2307-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023]
Abstract
PCB 11 (3,3'-dichlorobiphenyl), a contemporary congener produced as a byproduct of current pigment production processes, has recently emerged as a prevalent worldwide pollutant. We recently demonstrated that exposure to PCB 11 increases dendritic arborization in vitro, but the mechanism(s) mediating this effect remain unknown. To address this data gap, primary cortical neuron-glia co-cultures derived from neonatal Sprague-Dawley rats were exposed for 48 h to either vehicle (0.1% DMSO) or PCB 11 at concentrations ranging from 1 fM to 1 nM in the absence or presence of pharmacologic antagonists of established molecular targets of higher chlorinated PCBs. Reporter cell lines were used to test activity of PCB 11 at the aryl hydrocarbon receptor (AhR) and thyroid hormone receptor (THR). PCB 11 lacked activity at the AhR and THR, and antagonism of these receptors had no effect on the dendrite-promoting activity of PCB 11. Pharmacologic antagonism of various calcium channels or treatment with antioxidants also did not alter PCB 11-induced dendritic arborization. In contrast, pharmacologic blockade or shRNA knockdown of cAMP response element-binding protein (CREB) significantly decreased dendritic growth in PCB 11-exposed cultures, suggesting PCB 11 promotes dendritic growth via CREB-mediated mechanisms. Since CREB signaling is crucial for normal neurodevelopment, and perturbations of CREB signaling have been associated with neurodevelopmental disorders, our findings suggest that this contemporary pollutant poses a threat to the developing brain, particularly in individuals with heritable mutations that promote CREB signaling.
Collapse
Affiliation(s)
- Sunjay Sethi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Kimberly P Keil
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California-Davis, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA.
| |
Collapse
|
21
|
The Aryl Hydrocarbon Receptor and the Nervous System. Int J Mol Sci 2018; 19:ijms19092504. [PMID: 30149528 PMCID: PMC6163841 DOI: 10.3390/ijms19092504] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022] Open
Abstract
The aryl hydrocarbon receptor (or AhR) is a cytoplasmic receptor of pollutants. It translocates into the nucleus upon binding to its ligands, and forms a heterodimer with ARNT (AhR nuclear translocator). The heterodimer is a transcription factor, which regulates the transcription of xenobiotic metabolizing enzymes. Expressed in many cells in vertebrates, it is mostly present in neuronal cell types in invertebrates, where it regulates dendritic morphology or feeding behavior. Surprisingly, few investigations have been conducted to unravel the function of the AhR in the central or peripheral nervous systems of vertebrates. In this review, we will present how the AhR regulates neural functions in both invertebrates and vertebrates as deduced mainly from the effects of xenobiotics. We will introduce some of the molecular mechanisms triggered by the well-known AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), which impact on neuronal proliferation, differentiation, and survival. Finally, we will point out the common features found in mice that are exposed to pollutants, and in AhR knockout mice.
Collapse
|
22
|
Garcia GR, Bugel SM, Truong L, Spagnoli S, Tanguay RL. AHR2 required for normal behavioral responses and proper development of the skeletal and reproductive systems in zebrafish. PLoS One 2018; 13:e0193484. [PMID: 29494622 PMCID: PMC5832240 DOI: 10.1371/journal.pone.0193484] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/12/2018] [Indexed: 01/24/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a conserved ligand-activated transcription factor required for proper vertebrate development and homeostasis. The inappropriate activation of AHR by ubiquitous pollutants can lead to adverse effects on wildlife and human health. The zebrafish is a powerful model system that provides a vertebrate data stream that anchors hypothesis at the genetic and cellular levels to observations at the morphological and behavioral level, in a high-throughput format. In order to investigate the endogenous functions of AHR, we generated an AHR2 (homolog of human AHR)-null zebrafish line (ahr2osu1) using the clustered, regulatory interspaced, short palindromic repeats (CRISPR)-Cas9 precision genome editing method. In zebrafish, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) mediated toxicity requires AHR2. The AHR2-null line was resistant to TCDD-induced toxicity, indicating the line can be used to investigate the biological and toxicological functions of AHR2. The AHR2-null zebrafish exhibited decreased survival and fecundity compared to the wild type line. At 36 weeks, histological evaluations of the AHR2-null ovaries revealed a reduction of mature follicles when compared to wild type ovaries, suggesting AHR2 regulates follicle growth in zebrafish. AHR2-null adults had malformed cranial skeletal bones and severely damaged fins. Our data suggests AHR2 regulates some aspect(s) of neuromuscular and/or sensory system development, with impaired behavioral responses observed in larval and adult AHR2-null zebrafish. This study increases our understanding of the endogenous functions of AHR, which may help foster a better understanding of the target organs and molecular mechanisms involved in AHR-mediated toxicities.
Collapse
Affiliation(s)
- Gloria R. Garcia
- Department of Environmental & Molecular Toxicology, Environmental Health Sciences Center, Sinnhuber Aquatic Research Laboratory, Oregon State University, Corvallis, OR, United States of America
| | - Sean M. Bugel
- Department of Environmental & Molecular Toxicology, Environmental Health Sciences Center, Sinnhuber Aquatic Research Laboratory, Oregon State University, Corvallis, OR, United States of America
| | - Lisa Truong
- Department of Environmental & Molecular Toxicology, Environmental Health Sciences Center, Sinnhuber Aquatic Research Laboratory, Oregon State University, Corvallis, OR, United States of America
| | - Sean Spagnoli
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States of America
| | - Robert L. Tanguay
- Department of Environmental & Molecular Toxicology, Environmental Health Sciences Center, Sinnhuber Aquatic Research Laboratory, Oregon State University, Corvallis, OR, United States of America
| |
Collapse
|
23
|
Kimura E, Tohyama C. Vocalization as a novel endpoint of atypical attachment behavior in 2,3,7,8-tetrachlorodibenzo-p-dioxin-exposed infant mice. Arch Toxicol 2018; 92:1741-1749. [PMID: 29445839 DOI: 10.1007/s00204-018-2176-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 02/08/2018] [Indexed: 01/24/2023]
Abstract
Mammalian attachment behaviors, such as crying, are essential for infant survival by receiving food, protection, and warmth from caregivers. Ultrasonic vocalization (USV) of infant rodents functions to promote maternal proximity. Impaired USV emission has been reported in mouse models of autism spectrum disorder, suggesting that USV is associated with higher brain function. In utero and lactational dioxin exposure is known to induce higher brain function abnormalities in adulthood; however, whether perinatal dioxin exposure affects behavior during infancy is unclear. Therefore, we studied the impact of dioxin exposure on USV emission in infant mice born to dams treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 0.6 or 3.0 µg/kg) on gestational day 12.5. On postnatal days 3-9, USVs of the offspring were recorded for 1 min using a microphone in a sound-attenuated chamber. The total USV and mean call durations in infant mice exposed to 3.0 µg/kg, but not 0.6 µg/kg, were shorter than those in the control mice. In addition, the percentages of complicated call types (i.e., chevron and wave) in mice exposed to 3.0 µg/kg were decreased. Dioxin-induced gene expression changes occurred in the brains of mice exposed to 3.0 µg/kg; however, body weight, motor activity, and vocal fold structure were not significantly affected. These results suggest that infant USV is a useful behavioral endpoint in developmental neurotoxicity assessment that may be used to evaluate effects of chemical exposure on the infant-caregiver interaction.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
24
|
van Dongen J, Bonder MJ, Dekkers KF, Nivard MG, van Iterson M, Willemsen G, Beekman M, van der Spek A, van Meurs JBJ, Franke L, Heijmans BT, van Duijn CM, Slagboom PE, Boomsma DI. DNA methylation signatures of educational attainment. NPJ SCIENCE OF LEARNING 2018; 3:7. [PMID: 30631468 PMCID: PMC6220239 DOI: 10.1038/s41539-018-0020-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/18/2017] [Accepted: 02/09/2018] [Indexed: 05/09/2023]
Abstract
Educational attainment is a key behavioural measure in studies of cognitive and physical health, and socioeconomic status. We measured DNA methylation at 410,746 CpGs (N = 4152) and identified 58 CpGs associated with educational attainment at loci characterized by pleiotropic functions shared with neuronal, immune and developmental processes. Associations overlapped with those for smoking behaviour, but remained after accounting for smoking at many CpGs: Effect sizes were on average 28% smaller and genome-wide significant at 11 CpGs after adjusting for smoking and were 62% smaller in never smokers. We examined sources and biological implications of education-related methylation differences, demonstrating correlations with maternal prenatal folate, smoking and air pollution signatures, and associations with gene expression in cis, dynamic methylation in foetal brain, and correlations between blood and brain. Our findings show that the methylome of lower-educated people resembles that of smokers beyond effects of their own smoking behaviour and shows traces of various other exposures.
Collapse
Affiliation(s)
- Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marc Jan Bonder
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Koen F. Dekkers
- Molecular Epidemiology section, Leiden University Medical Center, Leiden, The Netherlands
| | - Michel G. Nivard
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maarten van Iterson
- Molecular Epidemiology section, Leiden University Medical Center, Leiden, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marian Beekman
- Molecular Epidemiology section, Leiden University Medical Center, Leiden, The Netherlands
| | - Ashley van der Spek
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Bastiaan T. Heijmans
- Molecular Epidemiology section, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelia M. van Duijn
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus Medical Center, Rotterdam, The Netherlands
| | - P. Eline Slagboom
- Molecular Epidemiology section, Leiden University Medical Center, Leiden, The Netherlands
| | - Dorret I. Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
25
|
Aluru N, Karchner SI, Glazer L. Early Life Exposure to Low Levels of AHR Agonist PCB126 (3,3',4,4',5-Pentachlorobiphenyl) Reprograms Gene Expression in Adult Brain. Toxicol Sci 2017; 160:386-397. [PMID: 28973690 PMCID: PMC5837202 DOI: 10.1093/toxsci/kfx192] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Early life exposure to environmental chemicals can have long-term consequences that are not always apparent until later in life. We recently demonstrated that developmental exposure of zebrafish to low, nonembryotoxic levels of 3,3',4,4',5-pentachlorobiphenyl (PCB126) did not affect larval behavior, but caused changes in adult behavior. The objective of this study was to investigate the underlying molecular basis for adult behavioral phenotypes resulting from early life exposure to PCB126. We exposed zebrafish embryos to PCB126 during early development and measured transcriptional profiles in whole embryos, larvae and adult male brains using RNA-sequencing. Early life exposure to 0.3 nM PCB126 induced cyp1a transcript levels in 2-dpf embryos, but not in 5-dpf larvae, suggesting transient activation of aryl hydrocarbon receptor with this treatment. No significant induction of cyp1a was observed in the brains of adults exposed as embryos to PCB126. However, a total of 2209 and 1628 genes were differentially expressed in 0.3 and 1.2 nM PCB126-exposed groups, respectively. KEGG pathway analyses of upregulated genes in the brain suggest enrichment of calcium signaling, MAPK and notch signaling, and lysine degradation pathways. Calcium is an important signaling molecule in the brain and altered calcium homeostasis could affect neurobehavior. The downregulated genes in the brain were enriched with oxidative phosphorylation and various metabolic pathways, suggesting that the metabolic capacity of the brain is impaired. Overall, our results suggest that PCB exposure during sensitive periods of early development alters normal development of the brain by reprogramming gene expression patterns, which may result in alterations in adult behavior.
Collapse
Affiliation(s)
- Neelakanteswar Aluru
- Biology Department, Woods Hole Oceanographic Institution and Woods Hole Center for Oceans and Human Health, Woods Hole, Massachusetts 02543
| | - Sibel I Karchner
- Biology Department, Woods Hole Oceanographic Institution and Woods Hole Center for Oceans and Human Health, Woods Hole, Massachusetts 02543
| | - Lilah Glazer
- Biology Department, Woods Hole Oceanographic Institution and Woods Hole Center for Oceans and Human Health, Woods Hole, Massachusetts 02543
| |
Collapse
|
26
|
Kimura E, Kubo KI, Endo T, Ling W, Nakajima K, Kakeyama M, Tohyama C. Impaired dendritic growth and positioning of cortical pyramidal neurons by activation of aryl hydrocarbon receptor signaling in the developing mouse. PLoS One 2017; 12:e0183497. [PMID: 28820910 PMCID: PMC5562321 DOI: 10.1371/journal.pone.0183497] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/05/2017] [Indexed: 11/24/2022] Open
Abstract
The basic helix-loop-helix (bHLH) transcription factors exert multiple functions in mammalian cerebral cortex development. The aryl hydrocarbon receptor (AhR), a member of the bHLH-Per-Arnt-Sim subfamily, is a ligand-activated transcription factor reported to regulate nervous system development in both invertebrates and vertebrates, but the functions that AhR signaling pathway may have for mammalian cerebral cortex development remains elusive. Although the endogenous ligand involved in brain developmental process has not been identified, the environmental pollutant dioxin potently binds AhR and induces abnormalities in higher brain function of laboratory animals. Thus, we studied how activation of AhR signaling influences cortical development in mice. To this end, we produced mice expressing either constitutively active-AhR (CA-AhR), which has the capacity for ligand-independent activation of downstream genes, or AhR, which requires its ligands for activation. In brief, CA-AhR-expressing plasmid and AhR-expressing plasmid were each transfected into neural stems cells in the developing cerebrum by in utero electroporation on embryonic day 14.5. On postnatal day 14, mice transfected in utero with CA-AhR, but not those transfected with AhR, exhibited drastically reduced dendritic arborization of layer II/III pyramidal neurons and impaired neuronal positioning in the developing somatosensory cortex. The effects of CA-AhR were observed for dendrite development but not for the commissural fiber projection, suggesting a preferential influence on dendrites. The present results indicate that over-activation of AhR perturbs neuronal migration and morphological development in mammalian cortex, supporting previous observations of impaired dendritic structure, cortical dysgenesis, and behavioral abnormalities following perinatal dioxin exposure.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Ken-ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Toshihiro Endo
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wenting Ling
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Kakeyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Neuroscience and Preventive Medicine, Faculty of Human Sciences, Waseda University, Tokorozawa, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- * E-mail:
| |
Collapse
|
27
|
Tohyama C. Developmental neurotoxicity test guidelines: problems and perspectives. J Toxicol Sci 2017; 41:SP69-SP79. [PMID: 28250285 DOI: 10.2131/jts.41.sp69] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Epidemiologic evidence has demonstrated associations between early life exposure to industrial chemicals and the occurrence of disease states, including cognitive and behavioral abnormalities, in children. The developing brain in the fetal and infantile periods is extremely vulnerable to chemicals because the blood-brain barrier is not completely formed during these periods. The Organisation for Economic Co-operation and Development (OECD) developmental neurotoxicity (DNT) test guideline, TG426, updated in 2007, comprises in vivo behavioral observational tests and other tests intended to assess DNT induced by exposure to industrial chemicals. These chemicals may enter the market without having been subjected to DNT testing, as DNT test data is not mandated by law at the time of chemical registration. In addition, proprietary rights have led to problems concerning the non-disclosure of industrial chemical toxicity test data, including DNT test data. To overcome the disadvantages of high-cost and low time efficiency of in vivo DNT tests, in vitro or in silico tests are the proposed alternatives, but it is unlikely that the results of such tests would reflect changes in higher brain functions. Accordingly, the current DNT test guidelines need to be revised to avoid overlooking or neglecting the occurrence of DNT induced by exposure to low doses of chemicals. This review also proposes the introduction of novel in vivo DNT testing methods in light of a cost-performance analysis.
Collapse
Affiliation(s)
- Chiharu Tohyama
- Health, Environment, Science and Technology International Consulting (HESTIC)
| |
Collapse
|
28
|
Kimura E, Kubo KI, Endo T, Nakajima K, Kakeyama M, Tohyama C. Excessive activation of AhR signaling disrupts neuronal migration in the hippocampal CA1 region in the developing mouse. J Toxicol Sci 2017; 42:25-30. [PMID: 28070106 DOI: 10.2131/jts.42.25] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The aryl hydrocarbon receptor (AhR) avidly binds dioxin, a ubiquitous environmental contaminant. Disruption of downstream AhR signaling has been reported to alter neuronal development, and rodent offspring exposed to dioxin during gestation and lactation showed abnormalities in learning and memory, emotion, and social behavior. However, the mechanism behind the disrupted AhR signaling and developmental neurotoxicity induced by xenobiotic ligands remains elusive. Therefore, we studied how excessive AhR activation affects neuronal migration in the hippocampal CA1 region of the developing mouse brain. We transfected constitutively active (CA)-AhR, AhR, or control vector plasmids into neurons via in utero electroporation on gestational day 14 and analyzed neuronal positioning in the hippocampal CA1 region of offspring on postnatal day 14. CA-AhR transfection affected neuronal positioning, whereas no change was observed in AhR-transfected or control hippocampus. These results suggest that constitutively activated AhR signaling disrupts neuronal migration during hippocampal development. Further studies are needed to investigate whether such developmental disruption in the hippocampus leads to the abnormal cognition and behavior of rodent offspring upon maternal exposure to AhR xenobiotic ligands.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo
| | | | | | | | | | | |
Collapse
|
29
|
Hahn ME, Karchner SI, Merson RR. Diversity as Opportunity: Insights from 600 Million Years of AHR Evolution. CURRENT OPINION IN TOXICOLOGY 2017; 2:58-71. [PMID: 28286876 DOI: 10.1016/j.cotox.2017.02.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The aryl hydrocarbon receptor (AHR) was for many years of interest only to pharmacologists and toxicologists. However, this protein has fundamental roles in biology that are being revealed through studies in diverse animal species. The AHR is an ancient protein. AHR homologs exist in most major groups of modern bilaterian animals, including deuterostomes (chordates, hemichordates, echinoderms) and the two major clades of protostome invertebrates [ecdysozoans (e.g. arthropods and nematodes) and lophotrochozoans (e.g. molluscs and annelids)]. AHR homologs also have been identified in cnidarians such as the sea anemone Nematostella and in the genome of Trichoplax, a placozoan. Bilaterians, cnidarians, and placozoans form the clade Eumetazoa, whose last common ancestor lived approximately 600 million years ago (MYA). The presence of AHR homologs in modern representatives of all these groups indicates that the original eumetazoan animal possessed an AHR homolog. Studies in invertebrates and vertebrates reveal parallel functions of AHR in the development and function of sensory neural systems, suggesting that these may be ancestral roles. Vertebrate animals are characterized by the expansion and diversification of AHRs, via gene and genome duplications, from the ancestral protoAHR into at least five classes of AHR-like proteins: AHR, AHR1, AHR2, AHR3, and AHRR. The evolution of multiple AHRs in vertebrates coincided with the acquisition of high-affinity binding of halogenated and polynuclear aromatic hydrocarbons and the emergence of adaptive functions involving regulation of xenobiotic-metabolizing enzymes and roles in adaptive immunity. The existence of multiple AHRs may have facilitated subfunction partitioning and specialization of specific AHR types in some taxa. Additional research in diverse model and non-model species will continue to enrich our understanding of AHR and its pleiotropic roles in biology and toxicology.
Collapse
Affiliation(s)
- Mark E Hahn
- Biology Department, Woods Hole Oceanographic Institution, MS-32, Woods Hole, MA 02543, USA
| | - Sibel I Karchner
- Biology Department, Woods Hole Oceanographic Institution, MS-32, Woods Hole, MA 02543, USA
| | - Rebeka R Merson
- Biology Department, Rhode Island College, 600 Mt. Pleasant Avenue, 251 Fogarty Life Sciences, Providence, RI 02908
| |
Collapse
|
30
|
Kimura E, Tohyama C. Embryonic and Postnatal Expression of Aryl Hydrocarbon Receptor mRNA in Mouse Brain. Front Neuroanat 2017; 11:4. [PMID: 28223923 PMCID: PMC5293765 DOI: 10.3389/fnana.2017.00004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 01/17/2017] [Indexed: 12/21/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR), a member of the basic helix-loop-helix-Per-Arnt-Sim transcription factor family, plays a critical role in the developing nervous system of invertebrates and vertebrates. Dioxin, a ubiquitous environmental pollutant, avidly binds to this receptor, and maternal exposure to dioxin has been shown to impair higher brain functions and dendritic morphogenesis, possibly via an AhR-dependent mechanism. However, there is little information on AhR expression in the developing mammalian brain. To address this issue, the present study analyzed AhR mRNA expression in the brains of embryonic, juvenile, and adult mice by reverse transcription (RT)-PCR and in situ hybridization. In early brain development (embryonic day 12.5), AhR transcript was detected in the innermost cortical layer. The mRNA was also expressed in the hippocampus, cerebral cortex, cerebellum, olfactory bulb, and rostral migratory stream on embryonic day 18.5, postnatal days 3, 7, and 14, and in 12-week-old (adult) mice. Hippocampal expression was abundant in the CA1 and CA3 pyramidal and dentate gyrus granule cell layers, where expression level of AhR mRNA in 12-week old is higher than that in 7-day old. These results reveal temporal and spatial patterns of AhR mRNA expression in the mouse brain, providing the information that may contribute to the elucidation of the physiologic and toxicologic significance of AhR in the developing brain.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of TokyoTokyo, Japan; Environmental Biology Laboratory, Faculty of Medicine, University of TsukubaTsukuba, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of TokyoTokyo, Japan; Environmental Biology Laboratory, Faculty of Medicine, University of TsukubaTsukuba, Japan
| |
Collapse
|
31
|
AhR signaling activation disrupts migration and dendritic growth of olfactory interneurons in the developing mouse. Sci Rep 2016; 6:26386. [PMID: 27197834 PMCID: PMC4873754 DOI: 10.1038/srep26386] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/29/2016] [Indexed: 12/21/2022] Open
Abstract
Perinatal exposure to a low level of dioxin, a ubiquitous environmental pollutant, has been shown to induce abnormalities in learning and memory, emotion, and sociality in laboratory animals later in adulthood. However, how aryl hydrocarbon receptor (AhR) signaling activation disrupts the higher brain function remains unclear. Therefore, we studied the possible effects of excessive activation of AhR signaling on neurodevelopmental processes, such as cellular migration and neurite growth, in mice. To this end, we transfected a constitutively active-AhR plasmid into stem cells in the lateral ventricle by in vivo electroporation on postnatal day 1. Transfection was found to induce tangential migration delay and morphological abnormalities in neuronal precursors in the rostral migratory stream at 6 days post-electroporation (dpe) as well as disrupt radial migration in the olfactory bulb and apical and basal dendritic growth of the olfactory interneurons in the granule cell layer at 13 and 20 dpe. These results suggest that the retarded development of interneurons by the excessive AhR signaling may at least in part explain the dioxin-induced abnormal behavioral alterations previously reported in laboratory animals.
Collapse
|
32
|
Kimura E, Endo T, Yoshioka W, Ding Y, Ujita W, Kakeyama M, Tohyama C. In utero and lactational dioxin exposure induces Sema3b and Sema3g gene expression in the developing mouse brain. Biochem Biophys Res Commun 2016; 476:108-13. [PMID: 27178212 DOI: 10.1016/j.bbrc.2016.05.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 05/09/2016] [Indexed: 12/17/2022]
Abstract
In the developing mammalian brain, neural network formation is regulated by complex signaling cascades. In utero and lactational dioxin exposure is known to induce higher brain function abnormalities and dendritic growth disruption in rodents. However, it is unclear whether perinatal dioxin exposure affects the expression of genes involved in neural network formation. Therefore, we investigated changes in gene expression in the brain regions of developing mice born to dams administered 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; dose: 0, 0.6, or 3.0 μg/kg) on gestational day 12.5. Quantitative RT-PCR showed that TCDD exposure induced Ahrr expression in the cerebral cortex, hippocampus, and olfactory bulb of 3-day-old mice. Gene microarray analysis indicated that the mRNA expression levels of Sema3b and Sema3g, which encode proteins that are known to control axonal projections, were elevated in the olfactory bulb of TCDD-exposed mice, and the induction of these genes was observed during a 2-week postnatal period. Increased Sema3g expression was also observed in the brain but not in the kidney, liver, lung, and spleen of TCDD-exposed neonatal mice. These results indicate that the Sema3b and Sema3g genes are sensitive to brain-specific induction by dioxin exposure, which may disrupt neural network formation in the mammalian nervous system, thereby leading to abnormal higher brain function in adulthood.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan; Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Toshihiro Endo
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan
| | - Wataru Yoshioka
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yunjie Ding
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan
| | - Waka Ujita
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan
| | - Masaki Kakeyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan; Laboratory for Systems Neuroscience and Preventive Medicine, Faculty of Human Sciences, Waseda University, Tokorozawa, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Japan; Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|