1
|
Marques SI, Sá SI, Carmo H, Carvalho F, Silva JP. Pharmaceutical-mediated neuroimmune modulation in psychiatric/psychological adverse events. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111114. [PMID: 39111563 DOI: 10.1016/j.pnpbp.2024.111114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/21/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
The therapeutic use of many pharmaceuticals, including small molecules and biological therapies, has been associated with the onset of psychiatric and psychological adverse events (PPAEs), posing substantial concerns to patients' health and safety. These events, which encompass mood (e.g., depression, schizophrenia, suicidal ideation) and cognitive changes (e.g., learning and memory impairment, dementia) often remain undetected until advanced stages of clinical trials or pharmacovigilance, mostly because the mechanisms underlying the onset of PPAEs remain poorly understood. In recent years, the role of neuroimmune modulation (comprising an intricate interplay between various cell types and signaling pathways) in PPAEs has garnered substantial interest. Indeed, understanding these complex interactions would substantially contribute to increase the ability to predict the potential onset of PPAEs during preclinical stages of a new drug's R&D. This review provides a comprehensive summary of the most recent advances in neuroimmune modulation-related mechanisms contributing to the onset of PPAEs and their association with specific pharmaceuticals. Reported data strongly support an association between neuroimmune modulation and the onset of PPAEs. Pharmaceuticals may target specific molecular pathways and pathway elements (e.g., cholinergic and serotonergic systems), which in turn may directly or indirectly impact the inflammatory status and the homeostasis of the brain, regulating inflammation and neuronal function. Also, modulation of the peripheral immune system by pharmaceuticals that do not permeate the blood-brain barrier (e.g., monoclonal antibodies) may alter the neuroimmunomodulatory status of the brain, leading to PPAEs. In summary, this review underscores the diverse pathways through which drugs can influence brain inflammation, shedding light on potential targeted interventions.
Collapse
Affiliation(s)
- Sandra I Marques
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Susana I Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Helena Carmo
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - João P Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
2
|
Ullman MT, Clark GM, Pullman MY, Lovelett JT, Pierpont EI, Jiang X, Turkeltaub PE. The neuroanatomy of developmental language disorder: a systematic review and meta-analysis. Nat Hum Behav 2024; 8:962-975. [PMID: 38491094 DOI: 10.1038/s41562-024-01843-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/01/2024] [Indexed: 03/18/2024]
Abstract
Developmental language disorder (DLD) is a common neurodevelopmental disorder with adverse impacts that continue into adulthood. However, its neural bases remain unclear. Here we address this gap by systematically identifying and quantitatively synthesizing neuroanatomical studies of DLD using co-localization likelihood estimation, a recently developed neuroanatomical meta-analytic technique. Analyses of structural brain data (22 peer-reviewed papers, 577 participants) revealed highly consistent anomalies only in the basal ganglia (100% of participant groups in which this structure was examined, weighted by group sample sizes; 99.8% permutation-based likelihood the anomaly clustering was not due to chance). These anomalies were localized specifically to the anterior neostriatum (again 100% weighted proportion and 99.8% likelihood). As expected given the task dependence of activation, functional neuroimaging data (11 peer-reviewed papers, 414 participants) yielded less consistency, though anomalies again occurred primarily in the basal ganglia (79.0% and 95.1%). Multiple sensitivity analyses indicated that the patterns were robust. The meta-analyses elucidate the neuroanatomical signature of DLD, and implicate the basal ganglia in particular. The findings support the procedural circuit deficit hypothesis of DLD, have basic research and translational implications for the disorder, and advance our understanding of the neuroanatomy of language.
Collapse
Affiliation(s)
- Michael T Ullman
- Brain and Language Laboratory, Department of Neuroscience, Georgetown University, Washington DC, USA.
| | - Gillian M Clark
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Victoria, Australia
| | - Mariel Y Pullman
- Brain and Language Laboratory, Department of Neuroscience, Georgetown University, Washington DC, USA
- Mount Sinai Beth Israel, New York, NY, USA
| | - Jarrett T Lovelett
- Brain and Language Laboratory, Department of Neuroscience, Georgetown University, Washington DC, USA
- Department of Psychology, University of California, San Diego, La Jolla, CA, USA
| | - Elizabeth I Pierpont
- Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Xiong Jiang
- Department of Neuroscience, Georgetown University, Washington DC, USA
| | - Peter E Turkeltaub
- Center for Brain Plasticity and Recovery, Georgetown University, Washington DC, USA
- Research Division, MedStar National Rehabilitation Network, Washington DC, USA
| |
Collapse
|
3
|
McCarthy DM, Spencer TJ, Bhide PG. Preclinical Models of Attention Deficit Hyperactivity Disorder: Neurobiology, Drug Discovery, and Beyond. J Atten Disord 2024; 28:880-894. [PMID: 38084074 DOI: 10.1177/10870547231215286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
OBJECTIVE We offer an overview of ADHD research using mouse models of nicotine exposure. METHOD Nicotine exposure of C57BL/6 or Swiss Webster mice occurred during prenatal period only or during the prenatal and the pre-weaning periods. Behavioral, neuroanatomical and neurotransmitter assays were used to investigate neurobiological mechanisms of ADHD and discover candidate ADHD medications. RESULTS Our studies show that norbinaltorphimine, a selective kappa opioid receptor antagonist is a candidate novel non-stimulant ADHD treatment and that a combination of methylphenidate and naltrexone has abuse deterrent potential with therapeutic benefits for ADHD. Other studies showed transgenerational transmission of ADHD-associated behavioral traits and demonstrated that interactions between untreated ADHD and repeated mild traumatic brain injury produced behavioral traits not associated with either condition alone. CONCLUSION Preclinical models contribute to novel insights into ADHD neurobiology and are valuable tools for drug discovery and translation to benefit humans with ADHD.
Collapse
Affiliation(s)
| | - Thomas J Spencer
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pradeep G Bhide
- Florida State University College of Medicine, Tallahassee, FL, USA
| |
Collapse
|
4
|
Zhou M, Qiu W, Ohashi N, Sun L, Wronski ML, Kouyama-Suzuki E, Shirai Y, Yanagawa T, Mori T, Tabuchi K. Deep-Learning-Based Analysis Reveals a Social Behavior Deficit in Mice Exposed Prenatally to Nicotine. Cells 2024; 13:275. [PMID: 38334667 PMCID: PMC10855062 DOI: 10.3390/cells13030275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
Cigarette smoking during pregnancy is known to be associated with the incidence of attention-deficit/hyperactive disorder (ADHD). Recent developments in deep learning algorithms enable us to assess the behavioral phenotypes of animal models without cognitive bias during manual analysis. In this study, we established prenatal nicotine exposure (PNE) mice and evaluated their behavioral phenotypes using DeepLabCut and SimBA. We optimized the training parameters of DeepLabCut for pose estimation and succeeded in labeling a single-mouse or two-mouse model with high fidelity during free-moving behavior. We applied the trained network to analyze the behavior of the mice and found that PNE mice exhibited impulsivity and a lessened working memory, which are characteristics of ADHD. PNE mice also showed elevated anxiety and deficits in social interaction, reminiscent of autism spectrum disorder (ASD). We further examined PNE mice by evaluating adult neurogenesis in the hippocampus, which is a pathological hallmark of ASD, and demonstrated that newborn neurons were decreased, specifically in the ventral part of the hippocampus, which is reported to be related to emotional and social behaviors. These results support the hypothesis that PNE is a risk factor for comorbidity with ADHD and ASD in mice.
Collapse
Affiliation(s)
- Mengyun Zhou
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
| | - Wen Qiu
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
| | - Nobuhiko Ohashi
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
| | - Lihao Sun
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
| | - Marie-Louis Wronski
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Emi Kouyama-Suzuki
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
| | - Yoshinori Shirai
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
| | - Takuma Mori
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
- Department of Neuroinnovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto 390-8621, Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan; (M.Z.); (W.Q.); (N.O.); (L.S.); (M.-L.W.); (E.K.-S.); (Y.S.)
- Department of Neuroinnovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto 390-8621, Japan
| |
Collapse
|
5
|
Ugarte G, Piña R, Contreras D, Godoy F, Rubio D, Rozas C, Zeise M, Vidal R, Escobar J, Morales B. Attention Deficit-Hyperactivity Disorder (ADHD): From Abnormal Behavior to Impairment in Synaptic Plasticity. BIOLOGY 2023; 12:1241. [PMID: 37759640 PMCID: PMC10525904 DOI: 10.3390/biology12091241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023]
Abstract
Attention deficit-hyperactivity disorder (ADHD) is a neurodevelopmental disorder with high incidence in children and adolescents characterized by motor hyperactivity, impulsivity, and inattention. Magnetic resonance imaging (MRI) has revealed that neuroanatomical abnormalities such as the volume reduction in the neocortex and hippocampus are shared by several neuropsychiatric diseases such as schizophrenia, autism spectrum disorder and ADHD. Furthermore, the abnormal development and postnatal pruning of dendritic spines of neocortical neurons in schizophrenia, autism spectrum disorder and intellectual disability are well documented. Dendritic spines are dynamic structures exhibiting Hebbian and homeostatic plasticity that triggers intracellular cascades involving glutamate receptors, calcium influx and remodeling of the F-actin network. The long-term potentiation (LTP)-induced insertion of postsynaptic glutamate receptors is associated with the enlargement of spine heads and long-term depression (LTD) with spine shrinkage. Using a murine model of ADHD, a delay in dendritic spines' maturation in CA1 hippocampal neurons correlated with impaired working memory and hippocampal LTP has recently reported. The aim of this review is to summarize recent evidence that has emerged from studies focused on the neuroanatomical and genetic features found in ADHD patients as well as reports from animal models describing the molecular structure and remodeling of dendritic spines.
Collapse
Affiliation(s)
- Gonzalo Ugarte
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile; (G.U.); (D.C.); (F.G.); (D.R.); (C.R.)
| | - Ricardo Piña
- Department of Biology, Faculty of Sciences, Metropolitan University of Education Sciences, Santiago 7760197, Chile;
- Department of Human Sciences, Faculty of Human Science, Bernardo O’Higgins University, Santiago 8370854, Chile
| | - Darwin Contreras
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile; (G.U.); (D.C.); (F.G.); (D.R.); (C.R.)
| | - Felipe Godoy
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile; (G.U.); (D.C.); (F.G.); (D.R.); (C.R.)
| | - David Rubio
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile; (G.U.); (D.C.); (F.G.); (D.R.); (C.R.)
| | - Carlos Rozas
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile; (G.U.); (D.C.); (F.G.); (D.R.); (C.R.)
| | - Marc Zeise
- School of Psychology, Faculty of Humanities, University of Santiago of Chile, Santiago 9170022, Chile;
| | - Rodrigo Vidal
- Laboratory of Genomics, Molecular Ecology and Evolutionary Studies, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile;
| | - Jorge Escobar
- Institute of Chemistry, Pontifical Catholic University of Valparaíso, Valparaíso 2340000, Chile
| | - Bernardo Morales
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile; (G.U.); (D.C.); (F.G.); (D.R.); (C.R.)
| |
Collapse
|
6
|
Wells AC, Lotfipour S. Prenatal nicotine exposure during pregnancy results in adverse neurodevelopmental alterations and neurobehavioral deficits. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:11628. [PMID: 38389806 PMCID: PMC10880762 DOI: 10.3389/adar.2023.11628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/28/2023] [Indexed: 02/24/2024]
Abstract
Maternal tobacco use and nicotine exposure during pregnancy have been associated with adverse birth outcomes in infants and can lead to preventable pregnancy complications. Exposure to nicotine and other compounds in tobacco and electronic cigarettes (e-cigarettes) has been shown to increases the risk of miscarriage, prematurity, stillbirth, low birth weight, perinatal morbidity, and sudden infant death syndrome (SIDS). Additionally, recent data provided by clinical and pre-clinical research demonstrates that nicotine exposure during pregnancy may heighten the risk for adverse neurodevelopmental disorders such as Attention-Deficit Hyperactivity (ADHD), anxiety, and depression along with altering the infants underlying brain circuitry, response to neurotransmitters, and brain volume. In the United States, one in 14 women (7.2%) reported to have smoked cigarettes during their pregnancy with the global prevalence of smoking during pregnancy estimated to be 1.7%. Approximately 1.1% of women in the United States also reported to have used e-cigarettes during the last 3 months of pregnancy. Due to the large percentage of women utilizing nicotine products during pregnancy in the United States and globally, this review seeks to centralize pre-clinical and clinical studies focused on the neurobehavioral and neurodevelopmental complications associated with prenatal nicotine exposure (PNE) such as alterations to the hypothalamic-pituitary-adrenal (HPA) axis and brain regions such as the prefrontal cortex (PFC), ventral tegmental area (VTA), nucleus accumbens (NA), hippocampus, and caudate as well as changes to nAChR and cholinergic receptor signaling, long-term drug seeking behavior following PNE, and other related developmental disorders. Current literature analyzing the association between PNE and the risk for offspring developing schizophrenia, attention-deficit hyperactivity disorder (ADHD), autism spectrum disorder (ASD), anxiety, and obesity will also be discussed.
Collapse
Affiliation(s)
- Alicia C Wells
- School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Shahrdad Lotfipour
- School of Medicine, University of California, Irvine, Irvine, CA, United States
- Department of Emergency Medicine, Pharmaceutical Sciences, Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
7
|
Costa G, Pollack AE. Prenatal and postnatal drug exposure: focus on persistent central effects. Neural Regen Res 2023; 18:1697-1702. [PMID: 36751782 PMCID: PMC10154500 DOI: 10.4103/1673-5374.363190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 12/14/2022] Open
Abstract
Clinical studies indicate significant use of prescription, nonprescription and social/recreational drugs by women during pregnancy; however, limited knowledge exists about the detrimental effects that this practice may have on the developing central nervous system of the fetus. Importantly, few experimental and clinical data are available on how gestational exposure could exacerbate the effects of the same or a different drug consumed by the offspring later in life. The present review summarizes recent findings on the central toxicity elicited by several classes of drugs, administered prenatally and postnatally in experimental animals and humans, focusing on prescription and nonprescription analgesics, anti-inflammatory agents, alcohol and nicotine.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Alexia E. Pollack
- Department of Biology, University of Massachusetts-Boston, Boston, MA, USA
| |
Collapse
|
8
|
Margolis AE, Lee SH, Liu R, Goolsby L, Champagne F, Herbstman J, Beebe B. Associations between prenatal exposure to second hand smoke and infant self-regulation in a New York city longitudinal prospective birth cohort. ENVIRONMENTAL RESEARCH 2023; 227:115652. [PMID: 36894114 DOI: 10.1016/j.envres.2023.115652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/11/2023] [Accepted: 03/06/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND Prenatal exposure to active or passive maternal smoking -also referred to as second hand smoke (SHS) exposure - are associated with externalizing behaviors, hyperactivity and attention deficit hyperactivity disorder, problems which derive in part from altered self-regulation. OBJECTIVES Determine the influence of prenatal SHS on infant self-regulation using direct measures of infant behavior in 99 mothers from the Fair Start birth cohort followed at the Columbia Center for Children's Environmental Health. METHODS Self-regulation was operationalized with self-contingency, the likelihood of maintaining/changing behavior from second-to-second, measured via split-screen video recordings of mothers playing with their 4-month infants. Mother and infant facial and vocal affect, gaze-on/-off partner, and mother touch were coded on a 1 s time-base. Third trimester prenatal SHS was assessed via self-report of a smoker in the home. Weighted-lag time-series models tested conditional effects of SHS-exposure (vs. non-exposure) on infant self-contingency for eight modality-pairings (e.g., mother gaze-infant gaze). Individual-seconds time-series models and analysis of predicted values at t0 interrogated significant weighted-lag findings. Because prior findings link developmental risk factors with lowered self-contingency, we hypothesized that prenatal SHSSHS would predict lowered infant self-contingency. RESULTS Relative to non-exposed infants, those who were prenatally exposed to SHS had lower self-contingency (more variable behavior) in all eight models. Follow-up analyses showed that, given infants were likely to be in the most negative facial or vocal affect, those with prenatal SHS were more likely to make larger behavioral changes, moving into less negative or more positive affect and to alternate between gaze-on and off mother. Mothers who were exposed to SHS during pregnancy (vs. non-exposed) showed a similar, albeit less prevalent, pattern of larger changes out of negative facial affect. CONCLUSION These findings extend prior work linking prenatal SHS with youth dysregulated behavior, showing similar effects in infancy, a critically important period that sthe stage for future child development.
Collapse
Affiliation(s)
- Amy E Margolis
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Division of Child and Adolescent Psychiatry, New York State Psychiatric Institute, New York, NY 10032, USA.
| | - Sang Han Lee
- Center for Biomedical Imaging and Neuromodulation, The Nathan Kline Institute, Orangeburg, NY, 10962, USA
| | - Ran Liu
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Lindsay Goolsby
- Division of Child and Adolescent Psychiatry, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Frances Champagne
- Department of Psychology, University of Texas, Austin, TX, 78712, USA
| | - Julie Herbstman
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Beatrice Beebe
- Division of Child and Adolescent Psychiatry, New York State Psychiatric Institute, New York, NY 10032, USA
| |
Collapse
|
9
|
Switching from tobacco cigarettes in very early pregnancy: The effects of in utero e-cigarette exposure on mouse offspring neurodevelopment and behaviour. Physiol Behav 2023; 263:114118. [PMID: 36796533 DOI: 10.1016/j.physbeh.2023.114118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
INTRODUCTION Electronic cigarettes (e-cigarettes) are often perceived to be safer than smoking, which has led to some women switching to e-cigarettes during pregnancy. However, the effects of switching from smoking to e-cigarettes on both pregnancy outcomes and the foetus are largely unknown. This study aimed to investigate the effects of switching from tobacco smoking to e-cigarette use in very early pregnancy on birth outcomes, neurodevelopment and behaviour of the offspring. METHODS Female BALB/c mice were exposed to cigarette smoke for up to two weeks before being mated. Mated dams were then allocated to one of four treatment groups: (i) continued exposure to cigarette smoke (ii) exposure to e-cigarette aerosol with nicotine, (iii) or without nicotine, or (iv) medical air. Pregnant mice were exposed for 2 h per day for the duration of pregnancy. Gestational outcomes including litter size and sex ratio were assessed, in addition to early-life markers of physical- and neuro- development. At 8 weeks of age, motor coordination, anxiety, locomotion, memory and learning of the adult offspring were assessed. RESULTS Gestational outcomes and early markers of physical- and neuro- development were unaffected by in utero exposure, as well as locomotion, anxiety-like behaviour, and object recognition memory during adulthood. However, both e-cigarette groups displayed increased spatial recognition memory compared to air exposed controls. Maternal exposure to nicotine containing e-cigarette aerosol was found to increase offspring bodyweight and impair motor skill learning. CONCLUSIONS These results suggest there may be some benefits as well as negative effects of switching to e-cigarettes in early pregnancy.
Collapse
|
10
|
Sharma N, Luhach K, Golani LK, Singh B, Sharma B. Vinpocetine, a PDE1 modulator, regulates markers of cerebral health, inflammation, and oxidative stress in a rat model of prenatal alcohol-induced experimental attention deficit hyperactivity disorder. Alcohol 2022; 105:25-34. [PMID: 35995260 DOI: 10.1016/j.alcohol.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/14/2022] [Accepted: 08/15/2022] [Indexed: 01/26/2023]
Abstract
Prenatal alcohol exposure (PAE) has been shown to induce symptomatology associated with attention deficit hyperactivity disorder (ADHD) by altering neurodevelopmental trajectories. Phosphodiesterase-1 (PDE1) is expressed centrally and has been used in various experimental brain conditions. We investigated the role of vinpocetine, a PDE1 inhibitor, on behavioral phenotypes and important biochemical deficits associated with a PAE rat model of ADHD. Protein markers of cerebral health (synapsin-IIa, BDNF, and pCREB), inflammation (IL-6, IL-10, and TNF-α), and oxidative stress (TBARS, GSH, and SOD) were analyzed in three brain regions (frontal cortex, striatum, and cerebellum). Hyperactivity, inattention, and anxiety introduced in the offspring due to PAE were assayed using open-field, Y-maze, and elevated plus maze, respectively. Administration of vinpocetine (10 & 20 mg/kg, p.o. [by mouth]) to PAE rat offspring for 4 weeks resulted in improvement of the behavioral profile of the animals. Additionally, levels of protein markers such as synapsin-IIa, BDNF, pCREB, IL-10, SOD, and GSH were found to be significantly increased, with a significant reduction in markers such as TNF-α, IL-6, and TBARS in selected brain regions of vinpocetine-treated animals. Vinpocetine, a selective PDE1 inhibitor, rectified behavioral phenotypes associated with ADHD, possibly by improving cerebral function, reducing brain inflammation, and reducing brain oxidative stress. This study provides preliminary analysis and suggests that the PDE1 enzyme may be an important pharmacological tool to study ADHD as a result of PAE.
Collapse
Affiliation(s)
- Niti Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India
| | - Kanishk Luhach
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India
| | - Lalit K Golani
- Department of Chemistry & Chemical Biology, Northeastern University, Boston, MA 02115, United States
| | - Bhagwat Singh
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India
| | - Bhupesh Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India.
| |
Collapse
|
11
|
Rodefer JS, Maitland SC. Adolescent nicotine administration impacts working memory and reversal learning but not cognitive flexibility. Dev Psychobiol 2022; 64:e22343. [PMID: 36426795 DOI: 10.1002/dev.22343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/13/2022] [Accepted: 09/25/2022] [Indexed: 11/12/2022]
Abstract
There has been increased interest in early exposure to nicotine through tobacco products and vaping specifically as it relates to addiction, yet fewer studies have focused on whether behavioral effects resulting from early nicotine exposure may persist into adulthood. Our experiments tested the hypothesis that exposure to nicotine during adolescence would impair selective aspects of behavioral cognition in rodents in adulthood. Male and female adolescent rats received either nicotine (0.4 mg/kg) or vehicle injections (intraperitoneal) once daily for 10 days (PND 29-38) followed by a washout period before behavioral testing. Animals were followed in a longitudinal design and evaluated on a battery of both behavioral and cognitive tasks during adulthood (PND 90+) that included locomotor activity, working memory (novel object recognition), cognitive flexibility (attentional set-shifting task, ASST), and anxiety-like behaviors. Data suggested that subchronic exposure to nicotine during adolescence produced significant changes in working memory, in two reversal problems in the ASST, and in anxiety-related behaviors. Taken together these data may suggest that limited early exposure to nicotine may produce selective longer term impairments in cognitive and behavioral processes related to working memory and reversal learning.
Collapse
Affiliation(s)
- Joshua S Rodefer
- Department of Psychology, Mercer University, Macon, Georgia, USA.,Program of Neuroscience, Mercer University, Macon, Georgia, USA
| | - Sarah C Maitland
- Department of Psychology, Mercer University, Macon, Georgia, USA
| |
Collapse
|
12
|
Kantak KM. Rodent models of attention-deficit hyperactivity disorder: An updated framework for model validation and therapeutic drug discovery. Pharmacol Biochem Behav 2022; 216:173378. [DOI: 10.1016/j.pbb.2022.173378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 01/21/2023]
|
13
|
Jenkins S, Harker A, Gibb R. Distinct sex-dependent effects of maternal preconception nicotine and enrichment on the early development of rat offspring brain and behavior. Neurotoxicol Teratol 2022; 91:107062. [PMID: 34998861 DOI: 10.1016/j.ntt.2021.107062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/14/2021] [Accepted: 12/30/2021] [Indexed: 10/19/2022]
Abstract
Developmental nicotine exposure is harmful to offspring. Whereas much is known about the consequences of prenatal nicotine exposure, relatively little is understood about how maternal preconception nicotine impacts the next generation. Positive experiences, such as environmental enrichment/complexity, have considerable potential to improve developmental outcomes and even treat and prevent drug addiction. Therefore, the current study sought to identify how maternal exposure to moderate levels of nicotine prior to conception impacts offspring development, and if the presumably negative consequence of nicotine could be reversed by concurrent exposure to an enriched environment. We treated female Long Evans rats with nicotine in their drinking water (15 mg nicotine salt/L) for seven weeks while residing in either standard or enriched conditions. Both experiences occurred exclusively prior to mating. Nicotine exposure reduced dam fertility by ~20% (p = .06). Females reared their own litters, and offspring were tested in two assessments of early development: negative geotaxis and open field. Offspring were euthanized at weaning (P21), and their brains were processed with Golgi-Cox solution to allow quantification of dendritic spine density. Results indicate that neither maternal nicotine or enrichment had an impact on maternal care, but male offspring were impaired at negative geotaxis due to maternal nicotine, female offspring showed altered open field exploration due to maternal enrichment, and offspring of both sexes had increased spine density in OFC due to maternal enrichment. Therefore, this experiment provides novel insights into the unique, sex-dependent consequences of maternal preconception nicotine and enrichment on the early development of rat behavior and brain.
Collapse
Affiliation(s)
- Serena Jenkins
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Dr W, Lethbridge, AB T1K 3M4, Canada.
| | - Allonna Harker
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Dr W, Lethbridge, AB T1K 3M4, Canada.
| | - Robbin Gibb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Dr W, Lethbridge, AB T1K 3M4, Canada.
| |
Collapse
|
14
|
Stanford SC. Animal Models of ADHD? Curr Top Behav Neurosci 2022; 57:363-393. [PMID: 35604570 DOI: 10.1007/7854_2022_342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
To describe animals that express abnormal behaviors as a model of Attention-Deficit Hyperactivity Disorder (ADHD) implies that the abnormalities are analogous to those expressed by ADHD patients. The diagnostic features of ADHD comprise inattentiveness, impulsivity, and hyperactivity and so these behaviors are fundamental for validation of any animal model of this disorder. Several experimental interventions such as neurotoxic lesion of neonatal rats with 6-hydroxydopamine (6-OHDA), genetic alterations, or selective inbreeding of rodents have produced animals that express each of these impairments to some extent. This article appraises the validity of claims that these procedures have produced a model of ADHD, which is essential if they are to be used to investigate the underlying cause(s) of ADHD and its abnormal neurobiology.
Collapse
Affiliation(s)
- S Clare Stanford
- Department of Neuroscience Physiology and Pharmacology, University College London, London, UK.
| |
Collapse
|
15
|
Alaee E, Moazen P, Pattij T, Semnanian S, Azizi H. Prenatal exposure to morphine impairs attention and impulsivity in adult rats. Psychopharmacology (Berl) 2021; 238:2729-2741. [PMID: 34405254 DOI: 10.1007/s00213-021-05888-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/27/2021] [Indexed: 10/20/2022]
Abstract
RATIONALE An alarming number of neonates born with prenatal exposure to morphine has resulted from the opioid epidemic; however, the long-term effects of prenatal opioid exposure on offspring behavior remain relatively unknown. In this study, we evaluated whether prenatal exposure to the mu opioid receptor agonist, morphine, has enduring effects on cognitive functions in adult life. METHODS On embryonic days 11-18 (E11-E18), female pregnant rats were injected subcutaneously with either morphine or saline twice daily. Adult male offspring that was prenatally exposed to saline or morphine was trained in the 5-choice serial reaction time test (5-CSRTT) to test their cognitive abilities under baseline conditions. Next, these rats were treated with saline (1 ml/kg), naloxone (1 mg/kg), and acute morphine (1, 3, 5 mg/kg), subcutaneously, once daily and following drug challenges rats were tested in the 5-CSRTT. Meanwhile, behavioral performance on training days between opioid drug challenges were analyzed to monitor possible drug-induced shifts in baseline performance. As a final experiment in order to investigate subchronic exposure to morphine, rats were injected with 5 mg/kg morphine for 5 days and then naloxone in the last day of the experiment (day 6). RESULTS Firstly, during acquisition of a stable baseline in the training phase, rats prenatally exposed to morphine showed delayed learning of the task demands. Furthermore, under baseline responding the rats prenatally exposed to morphine showed declined inhibitory control demonstrated by increased impulsive and compulsive-like responding compared to rats prenatally exposed to saline. Moreover, acute and subchronic morphine challenges in the rats prenatally exposed to morphine caused a deficit in visuospatial attention in comparison with saline treatment as well as the rats prenatally exposed to saline. These effects were abolished by naloxone. CONCLUSION The current findings indicate a direct causal effect of prenatal morphine exposure on inhibitory control and task learning later in life, as well as deficits in attention following morphine exposure in adulthood.
Collapse
Affiliation(s)
- Elham Alaee
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Parisa Moazen
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Tommy Pattij
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, The Netherlands
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran. .,Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
16
|
Smith BL. Improving translational relevance: The need for combined exposure models for studying prenatal adversity. Brain Behav Immun Health 2021; 16:100294. [PMID: 34589787 PMCID: PMC8474200 DOI: 10.1016/j.bbih.2021.100294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Prenatal environmental adversity is a risk factor for neurodevelopmental disorders (NDDs), with the neuroimmune environment proposed to play a role in this risk. Adverse maternal exposures are associated with cognitive consequences in the offspring that are characteristics of NDDs and simultaneous neuroimmune changes that may underlie NDD risk. In both animal models and human studies the association between prenatal environmental exposure and NDD risk has been shown to be complex. Maternal overnutrition/obesity and opioid use are two different examples of complex exposure epidemics, each with their own unique comorbidities. This review will examine maternal obesity and maternal opioid use separately, illustrating the pervasive comorbidities with each exposure to argue a need for animal models of compound prenatal exposures. Many of these comorbidities can impact neuroimmune function, warranting systematic investigation of combined exposures to begin to understand this complexity. While traditional approaches in animal models have focused on modeling a single prenatal exposure or second exposure later in life, a translational approach would begin to incorporate the most prevalent co-occurring prenatal exposures. Long term follow-up in humans is extremely challenging, so animal models can provide timely insight into neurodevelopmental consequences of complex prenatal exposures. Animal models that represent this translational context of comorbid exposures behind maternal obesity or comorbid exposures behind maternal opioid use may reveal potential synergistic neuroimmune interactions that contribute to cognitive consequences and NDD risk. Finally, translational co-exposure models can identify concerning exposure combinations to guide treatment in complex cases, and identify high risk children starting in the prenatal period where early interventions improve prognosis.
Collapse
Affiliation(s)
- Brittany L. Smith
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
17
|
Fisher ML, Pauly JR, Froeliger B, Turner JR. Translational Research in Nicotine Addiction. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a039776. [PMID: 32513669 DOI: 10.1101/cshperspect.a039776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
While commendable strides have been made in reducing smoking initiation and improving smoking cessation rates, current available smoking cessation treatment options are still only mildly efficacious and show substantial interindividual variability in their therapeutic responses. Therefore, the primary goal of preclinical research has been to further the understanding of the neural substrates and genetic influences involved in nicotine's effects and reassess potential drug targets. Pronounced advances have been made by investing in new translational approaches and placing more emphasis on bridging the gap between human and rodent models of dependence. Functional neuroimaging studies have identified key brain structures involved with nicotine-dependence phenotypes such as craving, impulsivity, withdrawal symptoms, and smoking cessation outcomes. Following up with these findings, rodent-modeling techniques have made it possible to dissect the neural circuits involved in these motivated behaviors and ascertain mechanisms underlying nicotine's interactive effects on brain structure and function. Likewise, translational studies investigating single-nucleotide polymorphisms (SNPs) within the cholinergic, dopaminergic, and opioid systems have found high levels of involvement of these neurotransmitter systems in regulating the reinforcing aspects of nicotine in both humans and mouse models. These findings and coordinated efforts between human and rodent studies pave the way for future work determining gene by drug interactions and tailoring treatment options to each individual smoker.
Collapse
Affiliation(s)
- Miranda L Fisher
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536-0596, USA
| | - James R Pauly
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536-0596, USA
| | - Brett Froeliger
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Jill R Turner
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536-0596, USA
| |
Collapse
|
18
|
Vasiliev G, Chadaeva I, Rasskazov D, Ponomarenko P, Sharypova E, Drachkova I, Bogomolov A, Savinkova L, Ponomarenko M, Kolchanov N, Osadchuk A, Oshchepkov D, Osadchuk L. A Bioinformatics Model of Human Diseases on the Basis of Differentially Expressed Genes (of Domestic Versus Wild Animals) That Are Orthologs of Human Genes Associated with Reproductive-Potential Changes. Int J Mol Sci 2021; 22:2346. [PMID: 33652917 PMCID: PMC7956675 DOI: 10.3390/ijms22052346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 02/13/2021] [Accepted: 02/23/2021] [Indexed: 12/18/2022] Open
Abstract
Earlier, after our bioinformatic analysis of single-nucleotide polymorphisms of TATA-binding protein-binding sites within gene promoters on the human Y chromosome, we suggested that human reproductive potential diminishes during self-domestication. Here, we implemented bioinformatics models of human diseases using animal in vivo genome-wide RNA-Seq data to compare the effect of co-directed changes in the expression of orthologous genes on human reproductive potential and during the divergence of domestic and wild animals from their nearest common ancestor (NCA). For example, serotonin receptor 3A (HTR3A) deficiency contributes to sudden death in pregnancy, consistently with Htr3a underexpression in guinea pigs (Cavia porcellus) during their divergence from their NCA with cavy (C. aperea). Overall, 25 and three differentially expressed genes (hereinafter, DEGs) in domestic animals versus 11 and 17 DEGs in wild animals show the direction consistent with human orthologous gene-markers of reduced and increased reproductive potential. This indicates a reliable association between DEGs in domestic animals and human orthologous genes reducing reproductive potential (Pearson's χ2 test p < 0.001, Fisher's exact test p < 0.05, binomial distribution p < 0.0001), whereas DEGs in wild animals uniformly match human orthologous genes decreasing and increasing human reproductive potential (p > 0.1; binomial distribution), thus enforcing the norm (wild type).
Collapse
Affiliation(s)
- Gennady Vasiliev
- Novosibirsk State University, 630090 Novosibirsk, Russia;
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Irina Chadaeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Dmitry Rasskazov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Petr Ponomarenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Ekaterina Sharypova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Irina Drachkova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Anton Bogomolov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Ludmila Savinkova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Mikhail Ponomarenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Nikolay Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Alexander Osadchuk
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Dmitry Oshchepkov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| | - Ludmila Osadchuk
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (I.C.); (D.R.); (P.P.); (E.S.); (I.D.); (A.B.); (L.S.); (N.K.); (A.O.); (D.O.); (L.O.)
| |
Collapse
|
19
|
Vorhees CV, Williams MT, Hawkey AB, Levin ED. Translating Neurobehavioral Toxicity Across Species From Zebrafish to Rats to Humans: Implications for Risk Assessment. FRONTIERS IN TOXICOLOGY 2021; 3:629229. [PMID: 35295117 PMCID: PMC8915800 DOI: 10.3389/ftox.2021.629229] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
There is a spectrum of approaches to neurotoxicological science from high-throughput in vitro cell-based assays, through a variety of experimental animal models to human epidemiological and clinical studies. Each level of analysis has its own advantages and limitations. Experimental animal models give essential information for neurobehavioral toxicology, providing cause-and-effect information regarding risks of neurobehavioral dysfunction caused by toxicant exposure. Human epidemiological and clinical studies give the closest information to characterizing human risk, but without randomized treatment of subjects to different toxicant doses can only give information about association between toxicant exposure and neurobehavioral impairment. In vitro methods give much needed high throughput for many chemicals and mixtures but cannot provide information about toxicant impacts on behavioral function. Crucial to the utility of experimental animal model studies is cross-species translation. This is vital for both risk assessment and mechanistic determination. Interspecies extrapolation is important to characterize from experimental animal models to humans and between different experimental animal models. This article reviews the literature concerning extrapolation of neurobehavioral toxicology from established rat models to humans and from zebrafish a newer experimental model to rats. The functions covered include locomotor activity, emotion, and cognition and the neurotoxicants covered include pesticides, metals, drugs of abuse, flame retardants and polycyclic aromatic hydrocarbons. With more complete understanding of the strengths and limitations of interspecies translation, we can better use animal models to protect humans from neurobehavioral toxicity.
Collapse
Affiliation(s)
- Charles V. Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States
| | - Michael T. Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States
| | - Andrew B. Hawkey
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| | - Edward D. Levin
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
20
|
Zhang L, McCarthy DM, Eskow Jaunarajs KL, Biederman J, Spencer TJ, Bhide PG. Frontal Cortical Monoamine Release, Attention, and Working Memory in a Perinatal Nicotine Exposure Mouse Model Following Kappa Opioid Receptor Antagonism. Cereb Cortex 2021; 31:483-496. [PMID: 32869057 DOI: 10.1093/cercor/bhaa238] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 02/27/2024] Open
Abstract
Perinatal nicotine exposure (PNE) produces frontal cortical hypo-dopaminergic state and attention and working memory deficits consistent with neurodevelopmental disorders such as attention deficit hyperactivity disorder (ADHD). Methylphenidate alleviates ADHD symptoms by increasing extracellular dopamine and noradrenaline. Kappa opioid receptor (KOR) antagonism may be another mechanism to achieve the same results because KOR activation inhibits frontal cortical dopamine release. We administered the selective KOR antagonist norbinaltorphimine (norBNI) (20 mg/kg; intraperitoneal) or methylphenidate (0.75 mg/kg; intraperitoneal) to PNE mouse model and examined frontal cortical monoamine release, attention, and working memory. Both compounds increased dopamine and noradrenaline release but neither influenced serotonin release. Both compounds improved object-based attention and working memory in the PNE group, with norBNI's effects evident at 2.5 h and 5.5 h but absent at 24 h. Methylphenidate's effects were evident at 0.5 h but not at 2.5 h. norBNI's effects temporally coincided with frontal cortical c-Jun N-terminal kinase phosphorylation. norBNI did not alter tissue dopamine content in the nucleus accumbens, offering preliminary support for lack of reinforcement.
Collapse
Affiliation(s)
- Lin Zhang
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Deirdre M McCarthy
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | | | - Joseph Biederman
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Thomas J Spencer
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pradeep G Bhide
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
21
|
Eliasen JN, Krall J, Frølund B, Kohlmeier KA. Sex-specific alterations in GABA receptor-mediated responses in laterodorsal tegmentum are associated with prenatal exposure to nicotine. Dev Neurobiol 2020; 80:178-199. [PMID: 32628361 DOI: 10.1002/dneu.22772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Smoking during pregnancy is associated with deleterious physiological and cognitive effects on the offspring, which are likely due to nicotine-induced alteration in the development of neurotransmitter systems. Prenatal nicotine exposure (PNE) in rodents is associated with changes in behaviors controlled in part by the pontine laterodorsal tegmentum (LDT), and LDT excitatory signaling is altered in a sex and age-dependent manner by PNE. As effects on GABAergic LDT signaling are unknown, we used calcium imaging to evaluate GABAA receptor- (GABAA R as well as GABAA -ρ R) and GABAB receptor (GABAB R)-mediated calcium responses in LDT brain slices from female and male PNE mice in two different age groups. Overall, in older PNE females, changes in calcium induced by stimulation of GABAA R and GABAB R, including GABAA -ρ R were shifted toward calcium rises. In both young and old males, PNE was associated with alterations in calcium mediated by all three receptors; however, the GABAA R was the most affected. These results show for the first time that PNE is associated with alterations in GABAergic transmission in the LDT in a sex- and age-dependent manner, and these data are the first to show PNE-associated alterations in functionality of GABA receptors in any nucleus. PNE-associated alterations in LDT GABAergic transmission within the LDT would be expected to alter output to target regions and could play a role in LDT-implicated, negative behavioral outcomes following gestational exposure to smoking. Accordingly, our data provide further supportive evidence of the importance of eliminating the consumption of nicotine during pregnancy.
Collapse
Affiliation(s)
- Jannik Nicklas Eliasen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Krall
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Frølund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Polli FS, Scharff MB, Ipsen TH, Aznar S, Kohlmeier KA, Andreasen JT. Prenatal nicotine exposure in mice induces sex-dependent anxiety-like behavior, cognitive deficits, hyperactivity, and changes in the expression of glutamate receptor associated-genes in the prefrontal cortex. Pharmacol Biochem Behav 2020; 195:172951. [PMID: 32439454 DOI: 10.1016/j.pbb.2020.172951] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/29/2022]
Abstract
In rodents, prenatal nicotine exposure (PNE) has been associated with increased risk for development of cognitive and emotional disturbances, but the findings are somewhat conflicting. Lack of behavioral alterations following PNE could be due to the variety of methods available for nicotine delivery, exposure time and species used, with inbred strains being mostly employed. Such differences suggest the need to investigate the behavioral phenotype in each PNE model available if we are to find models with enhanced translational value. In this study, we assessed sex-dependent effects of PNE on ADHD-related behaviors and on the levels of mRNA coding for glutamate receptor subunits within the prefrontal cortex in the outbred NMRI mice exposed to nicotine via maternal drinking water during gestation. Cotinine levels were assessed in newborn pups. Behaviors related to anxiety, compulsivity, working memory, and locomotion were evaluated in both sexes of young adult offspring using the elevated zero maze, marble burying, spontaneous alternation behavior, and locomotor activity tests. Expression of mRNA coding for different glutamate receptors subunits within the prefrontal cortex (PFC) was measured using RT-qPCR. Cotinine levels in the serum of newborns confirmed fetal nicotine exposure. Both male and female offspring showed ADHD-like behaviors, such as deficit in the SAB test and hyperactivity. In addition, PNE male mice displayed anxiety- and compulsive-like behaviors, effects that were absent in female offspring. Finally, PNE reduced the mRNA expression of GluN1-, GluN2B-, and mGluR2-related genes within the PFC of male offspring, whereas it reduced the expression of mRNA coding for GluA2 subunit in female mice. PNE in NMRI mice induced sex-dependent behavioral changes, which parallels clinical findings following maternal cigarette smoke exposure. Alterations detected in PFC mRNA glutamate receptor proteins could contribute to the abnormal behavioral responses observed, but other signaling pathways or brain regions are likely involved in the behavioral susceptibility of PNE individuals.
Collapse
Affiliation(s)
- Filip S Polli
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Malthe B Scharff
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Hospital, Copenhagen 2400, Denmark
| | - Theis H Ipsen
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Hospital, Copenhagen 2400, Denmark
| | - Susana Aznar
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Hospital, Copenhagen 2400, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jesper T Andreasen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark.
| |
Collapse
|
23
|
Cellular and Molecular Changes in Hippocampal Glutamate Signaling and Alterations in Learning, Attention, and Impulsivity Following Prenatal Nicotine Exposure. Mol Neurobiol 2020; 57:2002-2020. [PMID: 31916029 DOI: 10.1007/s12035-019-01854-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/11/2019] [Indexed: 12/18/2022]
Abstract
Over 70 million European pregnant women are smokers during their child-bearing years. Consumption of tobacco-containing products during pregnancy is associated with several negative behavioral outcomes for the offspring, including a higher susceptibility for the development of attention-deficit/hyperactive disorder (ADHD). In efforts to minimize fetal exposure to tobacco smoke, many women around the world switch to nicotine replacement therapies (NRTs) during the gestational period; however, prenatal nicotine exposure (PNE) in any form has been associated with alterations in cognitive processes, including learning, memory, and attention. These processes are controlled by glutamatergic signaling of hippocampal pyramidal neurons within the CA1 region, suggesting actions of nicotine on glutamatergic transmission in this region if present prenatally. Accordingly, we aimed to investigate hippocampal glutamatergic function following PNE treatment in NMRI mice employing molecular, cellular electrophysiology, and pharmacological approaches, as well as to evaluate cognition in the rodent continuous performance task (rCPT), a recently developed mouse task allowing assessment of learning, attention, and impulsivity. PNE induced increases in the expression levels of mRNA coding for different glutamate receptors and subunits within the hippocampus. Functional alterations in AMPA and NMDA receptors on CA1 pyramidal neurons of PNE mice were suggestive of higher GluA2-lacking and lower GluN2A-containing receptors, respectively. Finally, PNE was associated with reduced learning, attention, and enhanced impulsivity in the rCPT. Alterations in glutamatergic functioning in CA1 neurons parallel changes seen in the spontaneously hypertensive rat ADHD model and likely contribute to the lower cognitive performance in the rCPT.
Collapse
|
24
|
Polli FS, Kohlmeier KA. Alterations in NMDAR-mediated signaling within the laterodorsal tegmental nucleus are associated with prenatal nicotine exposure. Neuropharmacology 2019; 158:107744. [DOI: 10.1016/j.neuropharm.2019.107744] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/23/2019] [Accepted: 08/18/2019] [Indexed: 12/18/2022]
|
25
|
Polli FS, Kohlmeier KA. Prenatal Nicotine Exposure in Rodents: Why Are There So Many Variations in Behavioral Outcomes? Nicotine Tob Res 2019; 22:1694-1710. [DOI: 10.1093/ntr/ntz196] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/05/2019] [Indexed: 01/01/2023]
Abstract
Abstract
Introduction
The World Health Organization (WHO) reported that smoking cessation rates among women have stagnated in the past decade and estimates that hundreds of millions of women will be smokers in the next decade. Social, environmental, and biological conditions render women more susceptible to nicotine addiction, imposing additional challenges to quit smoking during gestation, which is likely why more than 8% of pregnancies in Europe are associated with smoking. In epidemiological investigations, individuals born from gestational exposure to smoking exhibit a higher risk of development of attention-deficit/hyperactive disorder (ADHD) and liability to drug dependence. Among other teratogenic compounds present in tobacco smoke, nicotine actions during neuronal development could contribute to the observed outcomes as nicotine misleads signaling among progenitor cells during brain development. Several experimental approaches have been developed to address the consequences of prenatal nicotine exposure (PNE) to the brain and behavior but, after four decades of studies, inconsistent data have been reported and the lack of consensus in the field has compromised the hypothesis that gestational nicotine exposure participates in cognitive and emotional behavioral deficits.
Aims
In this review, we discuss the most commonly used PNE models with focus on their advantages and disadvantages, their relative validity, and how the different technical approaches could play a role in the disparate outcomes.
Results
We propose methodological considerations, which could improve the translational significance of the PNE models.
Conclusions
Such alterations might be helpful in reconciling experimental findings, as well as leading to development of treatment targets for maladaptive behaviors in those prenatally exposed.
Implications
In this article, we have reviewed the advantages and disadvantages of different variables of the commonly used experimental models of PNE. We discuss how variations in the nicotine administration methods, the timing of nicotine exposure, nicotine doses, and species employed could contribute to the disparate findings in outcomes for PNE offspring, both in behavior and neuronal changes. In addition, recent findings suggest consideration of epigenetic effects extending across generations. Finally, we have suggested improvements in the available PNE models that could contribute to the enhancement of their validity, which could assist in the reconciliation of experimental findings.
Collapse
Affiliation(s)
- Filip Souza Polli
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristi Anne Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Hawkey A, Junaid S, Yao L, Spiera Z, White H, Cauley M, Levin ED. Gestational exposure to nicotine and/or benzo[a]pyrene causes long-lasting neurobehavioral consequences. Birth Defects Res 2019; 111:1248-1258. [PMID: 31368242 DOI: 10.1002/bdr2.1568] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 07/02/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022]
Abstract
Tobacco smoke is a complex mixture that includes thousands of compounds. Previously, we have found that gestational exposure to the complex mixture of tobacco smoke extract caused long-term neurobehavioral impairments. In this study, we examined the interaction of two of the most biologically active, nicotine and benzo[a]pyrene (BaP). Developmental effects were determined in Sprague-Dawley rats prenatally exposed to low doses of BaP and nicotine (0.03 mg/kg/day of BaP and 2 mg/kg/day of nicotine) via maternal osmotic minipumps throughout gestation. Behavioral function was assessed in the offspring via a battery of tests through adolescence into adulthood. There were sex-selective effects in four of the behavioral tests. In the elevated plus maze, there was a significant interaction of BaP and sex, where BaP-treated males showed a trend for increased activity. In the novelty suppressed feeding test, there were significant sex selective effects in males such that the normal sex difference in the behavior in this test was eliminated. Male offspring with prenatal exposure to either nicotine or BaP showed significant reductions in fear response. In the Figure-8 locomotor activity test, BAP-exposed male offspring were significantly hyperactive. This also eliminated the sex difference typically seen in this test. This effect persisted into adulthood. In the attention task, males exposed to nicotine during gestation showed a significant percent hit impairment. BaP reversed this effect. No significant effects were seen with percent correct rejection. These data show that both nicotine and BaP cause persisting sex-selective behavioral effects that persist into adulthood.
Collapse
Affiliation(s)
- Andrew Hawkey
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Shaqif Junaid
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Leah Yao
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Zachary Spiera
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Hannah White
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Marty Cauley
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Edward D Levin
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
27
|
Yates JR. Examining the neurochemical underpinnings of animal models of risky choice: Methodological and analytic considerations. Exp Clin Psychopharmacol 2019; 27:178-201. [PMID: 30570275 PMCID: PMC6467223 DOI: 10.1037/pha0000239] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Because risky choice is associated with several psychiatric conditions, recent research has focused on examining the underlying neurochemical processes that control risk-based decision-making. Not surprisingly, several tasks have been developed to study the neural mechanisms involved in risky choice. The current review will briefly discuss the major tasks used to measure risky choice and will summarize the contribution of several major neurotransmitter systems to this behavior. To date, the most common measures of risky choice are the probability discounting task, the risky decision task, and the rat gambling task. Across these three tasks, the contribution of the dopaminergic system has been most studied, although the effects of serotonergic, adrenergic, cholinergic, and glutamatergic ligands will be discussed. Drug effects across these tasks have been inconsistent, which makes determining the precise role of neurotransmitter systems in risky choice somewhat difficult. Furthermore, procedural differences can modulate drug effects in these procedures, and the way data are analyzed can alter the interpretations one makes concerning pharmacological manipulations. By taking these methodological/analytic considerations into account, we may better elucidate the neurochemistry of risky decision-making. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
|
28
|
McCarthy DM, Morgan TJ, Lowe SE, Williamson MJ, Spencer TJ, Biederman J, Bhide PG. Nicotine exposure of male mice produces behavioral impairment in multiple generations of descendants. PLoS Biol 2018; 16:e2006497. [PMID: 30325916 PMCID: PMC6191076 DOI: 10.1371/journal.pbio.2006497] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/13/2018] [Indexed: 12/27/2022] Open
Abstract
Use of tobacco products is injurious to health in men and women. However, tobacco use by pregnant women receives greater scrutiny because it can also compromise the health of future generations. More men smoke cigarettes than women. Yet the impact of nicotine use by men upon their descendants has not been as widely scrutinized. We exposed male C57BL/6 mice to nicotine (200 μg/mL in drinking water) for 12 wk and bred the mice with drug-naïve females to produce the F1 generation. Male and female F1 mice were bred with drug-naïve partners to produce the F2 generation. We analyzed spontaneous locomotor activity, working memory, attention, and reversal learning in male and female F1 and F2 mice. Both male and female F1 mice derived from the nicotine-exposed males showed significant increases in spontaneous locomotor activity and significant deficits in reversal learning. The male F1 mice also showed significant deficits in attention, brain monoamine content, and dopamine receptor mRNA expression. Examination of the F2 generation showed that male F2 mice derived from paternally nicotine-exposed female F1 mice had significant deficits in reversal learning. Analysis of epigenetic changes in the spermatozoa of the nicotine-exposed male founders (F0) showed significant changes in global DNA methylation and DNA methylation at promoter regions of the dopamine D2 receptor gene. Our findings show that nicotine exposure of male mice produces behavioral changes in multiple generations of descendants. Nicotine-induced changes in spermatozoal DNA methylation are a plausible mechanism for the transgenerational transmission of the phenotypes. These findings underscore the need to enlarge the current focus of research and public policy targeting nicotine exposure of pregnant mothers by a more equitable focus on nicotine exposure of the mother and the father.
Collapse
Affiliation(s)
- Deirdre M. McCarthy
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| | - Thomas J. Morgan
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| | - Sarah E. Lowe
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| | - Matthew J. Williamson
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| | - Thomas J. Spencer
- Pediatric Psychopharmacology, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joseph Biederman
- Pediatric Psychopharmacology, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Pradeep G. Bhide
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| |
Collapse
|
29
|
Micalizzi L, Knopik VS. Maternal smoking during pregnancy and offspring executive function: What do we know and what are the next steps? Dev Psychopathol 2018; 30:1333-1354. [PMID: 29144227 PMCID: PMC6028309 DOI: 10.1017/s0954579417001687] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Children exposed to maternal smoking during pregnancy (MSDP) exhibit difficulties in executive function (EF) from infancy through adolescence. Due to the developmental significance of EF as a predictor of adaptive functioning throughout the life span, the MSDP-EF relation has clear public health implications. In this paper, we provide a comprehensive review of the literature on the relationship between MSDP and offspring EF across development; consider brain-based assessments, animal models, and genetically informed studies in an effort to elucidate plausible pathways of effects; discuss implications for prevention and intervention; and make calls to action for future research.
Collapse
Affiliation(s)
- Lauren Micalizzi
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital
- Department of Psychiatry and Human Behavior, The Warren Alpert School of Medicine, Brown University
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University
| | - Valerie S. Knopik
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital
- Department of Psychiatry and Human Behavior, The Warren Alpert School of Medicine, Brown University
- Department of Human Development and Family Studies, Purdue University
| |
Collapse
|
30
|
Brockett AT, Pribut HJ, Vázquez D, Roesch MR. The impact of drugs of abuse on executive function: characterizing long-term changes in neural correlates following chronic drug exposure and withdrawal in rats. Learn Mem 2018; 25:461-473. [PMID: 30115768 PMCID: PMC6097763 DOI: 10.1101/lm.047001.117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022]
Abstract
Addiction has long been characterized by diminished executive function, control, and impulsivity management. In particular, these deficits often manifest themselves as impairments in reversal learning, delay discounting, and response inhibition. Understanding the neurobiological substrates of these behavioral deficits is of paramount importance to our understanding of addiction. Within the cycle of addiction, periods during and after withdrawal represent a particularly difficult point of intervention in that the negative physical symptoms associated with drug removal and drug craving increase the likelihood that the patient will relapse and return to drug use in order to abate these symptoms. Moreover, it is often during this time that drug induced deficits in executive function hinder the ability of the patient to refrain from drug use. Thus, it is necessary to understand the physiological and behavioral changes associated with withdrawal and drug craving-largely manifesting as deficits in executive control-to develop more effective treatment strategies. In this review, we address the long-term impact that drugs of abuse have on the behavioral and neural correlates that give rise to executive control as measured by reversal learning, delay discounting, and stop-signal tasks, focusing particularly on our work using rats as a model system.
Collapse
Affiliation(s)
- Adam T Brockett
- Department of Psychology, University of Maryland, College Park, Maryland 20742, USA
- Program in Neuroscience and Cognitive Science, University of Maryland, College Park, Maryland 20742, USA
| | - Heather J Pribut
- Department of Psychology, University of Maryland, College Park, Maryland 20742, USA
- Program in Neuroscience and Cognitive Science, University of Maryland, College Park, Maryland 20742, USA
| | - Daniela Vázquez
- Department of Psychology, University of Maryland, College Park, Maryland 20742, USA
- Program in Neuroscience and Cognitive Science, University of Maryland, College Park, Maryland 20742, USA
| | - Matthew R Roesch
- Department of Psychology, University of Maryland, College Park, Maryland 20742, USA
- Program in Neuroscience and Cognitive Science, University of Maryland, College Park, Maryland 20742, USA
| |
Collapse
|
31
|
Bassey RB, Gondré-Lewis MC. Combined early life stressors: Prenatal nicotine and maternal deprivation interact to influence affective and drug seeking behavioral phenotypes in rats. Behav Brain Res 2018; 359:814-822. [PMID: 30055209 DOI: 10.1016/j.bbr.2018.07.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022]
Abstract
Early life stress (ELS) increases the risk for later cognitive and emotional dysfunction, and has been implicated in the etiology of multiple psychiatric disorders. We hypothesize that combined insults during gestation and infancy, critical periods of neural development, could exacerbate neuropsychiatric outcomes in later life. Thus, we investigated the effects of maternal deprivation (MD) stress alone or combined with prenatal nicotine exposure (PNE) on negative affective states, ethanol drinking, and development of mesolimbic loci that regulate depression and drug dependence. On the elevated plus maze (EPM), MD rats exhibited ∼50% increase in risk-taking behavior/decreased anxiety when compared to control, but the combined MD + PNE did not affect this specific behavior. In the open field test, however, both MD and MD + PNE groups showed 2-fold greater locomotor activity. Furthermore, whereas MD showed greater latency to fall at 40 RPM on the rotarod compared to control, the MD + PNE animals' latency to fall was significantly greater at all RPMs tested, with an approximate 15% enhancement in motor coordination overall compared to control and MD. Analyses of depressive symptomatology with the forced swim test (FST) yielded 2- and 3-fold higher immobility times in MD and MD + PNE respectively. When tested in an operant drinking paradigm to quantify the effect of treatment on 10%v/v ethanol drinking, the MD and MD + PNE groups showed heightened ethanol consumption by ∼3- and 2-fold respectively. However, the experience of PNE reduced ethanol consumption in adults relative to MD alone. To test the stressors' impact on neurons in the amygdala and ventral tegmental area (VTA), mesolimbic anatomical regions associated with mood and reward, unbiased stereological measurements were performed and revealed ∼15% increase in number and density of neurons in the amygdala for both MD and MD + PNE, and ∼13% reduction in dopaminergic-like neurons in the VTA compared to control. We report here that multiple early stressors including prenatal nicotine and MD can modulate the neuroanatomy of the amygdala and VTA. These early life stressors can interact to influence the development of depressive-like and addictive behaviors.
Collapse
Affiliation(s)
- Rosemary B Bassey
- Department of Anatomy, Howard University, College of Medicine, Washington D.C. 20059, USA; Department of Psychiatry and Behavioral Sciences, Howard University, College of Medicine, Washington D.C. 20059, USA
| | - Marjorie C Gondré-Lewis
- Department of Anatomy, Howard University, College of Medicine, Washington D.C. 20059, USA; Department of Psychiatry and Behavioral Sciences, Howard University, College of Medicine, Washington D.C. 20059, USA.
| |
Collapse
|
32
|
Morris M, Shaw A, Lambert M, Perry HH, Lowenstein E, Valenzuela D, Velazquez-Ulloa NA. Developmental nicotine exposure affects larval brain size and the adult dopaminergic system of Drosophila melanogaster. BMC DEVELOPMENTAL BIOLOGY 2018; 18:13. [PMID: 29898654 PMCID: PMC6001141 DOI: 10.1186/s12861-018-0172-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 05/21/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND Pregnant women may be exposed to nicotine if they smoke or use tobacco products, nicotine replacement therapy, or via e-cigarettes. Prenatal nicotine exposure has been shown to have deleterious effects on the nervous system in mammals including changes in brain size and in the dopaminergic system. The genetic and molecular mechanisms for these changes are not well understood. A Drosophila melanogaster model for these effects of nicotine exposure could contribute to faster identification of genes and molecular pathways underlying these effects. The purpose of this study was to determine if developmental nicotine exposure affects the nervous system of Drosophila melanogaster, focusing on changes to brain size and the dopaminergic system at two developmental stages. RESULTS We reared flies on control or nicotine food from egg to 3rd instar larvae or from egg to adult and determined effectiveness of the nicotine treatment. We used immunohistochemistry to visualize the whole brain and dopaminergic neurons, using tyrosine hydroxylase as the marker. We measured brain area, tyrosine hydroxylase fluorescence, and counted the number of dopaminergic neurons in brain clusters. We detected an increase in larval brain hemisphere area, a decrease in tyrosine hydroxylase fluorescence in adult central brains, and a decrease in the number of neurons in the PPM3 adult dopaminergic cluster. We tested involvement of Dα7, one of the nicotinic acetylcholine receptor subunits, and found it was involved in eclosion, as previously described, but not involved in brain size. CONCLUSIONS We conclude that developmental nicotine exposure in Drosophila melanogaster affects brain size and the dopaminergic system. Prenatal nicotine exposure in mammals has also been shown to have effects on brain size and in the dopaminergic system. This study further establishes Drosophila melanogaster as model organism to study the effects of developmental nicotine exposure. The genetic and molecular tools available for Drosophila research will allow elucidation of the mechanisms underlying the effects of nicotine exposure during development.
Collapse
Affiliation(s)
- Melanie Morris
- School of Medicine, University of Washington, Seattle, USA
| | - Ariel Shaw
- Biochemistry, Cell and Molecular Biology Program, Lewis & Clark College, Portland, USA
| | | | | | - Eve Lowenstein
- Biology Department, Lewis & Clark College, Portland, USA
| | | | | |
Collapse
|
33
|
Zhang L, Spencer TJ, Biederman J, Bhide PG. Attention and working memory deficits in a perinatal nicotine exposure mouse model. PLoS One 2018; 13:e0198064. [PMID: 29795664 PMCID: PMC5967717 DOI: 10.1371/journal.pone.0198064] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cigarette smoking by pregnant women is associated with a significant increase in the risk for cognitive disorders in their children. Preclinical models confirm this risk by showing that exposure of the developing brain to nicotine produces adverse behavioral outcomes. Here we describe behavioral phenotypes resulting from perinatal nicotine exposure in a mouse model, and discuss our findings in the context of findings from previously published studies using preclinical models of developmental nicotine exposure. METHODOLOGY/PRINCIPAL FINDINGS Female C57Bl/6 mice received drinking water containing nicotine (100μg/ml) + saccharin (2%) starting 3 weeks prior to breeding and continuing throughout pregnancy, and until 3 weeks postpartum. Over the same period, female mice in two control groups received drinking water containing saccharin (2%) or plain drinking water. Offspring from each group were weaned at 3-weeks of age and subjected to behavioral analyses at 3 months of age. We examined spontaneous locomotor activity, anxiety-like behavior, spatial working memory, object based attention, recognition memory and impulsive-like behavior. We found significant deficits in attention and working memory only in male mice, and no significant changes in the other behavioral phenotypes in male or female mice. Exposure to saccharin alone did not produce significant changes in either sex. CONCLUSION/SIGNIFICANCE The perinatal nicotine exposure produced significant deficits in attention and working memory in a sex-dependent manner in that the male but not female offspring displayed these behaviors. These behavioral phenotypes are associated with attention deficit hyperactivity disorder (ADHD) and have been reported in other studies that used pre- or perinatal nicotine exposure. Therefore, we suggest that preclinical models of developmental nicotine exposure could be useful tools for modeling ADHD and related disorders.
Collapse
Affiliation(s)
- Lin Zhang
- Center for Brain Repair, Biomedical Sciences, Florida State University College of Medicine, Tallahassee, United States of America
| | - Thomas J. Spencer
- Pediatric Psychopharmacology, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, United States of America
| | - Joseph Biederman
- Pediatric Psychopharmacology, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, United States of America
| | - Pradeep G. Bhide
- Center for Brain Repair, Biomedical Sciences, Florida State University College of Medicine, Tallahassee, United States of America
- * E-mail:
| |
Collapse
|
34
|
Polli FS, Kohlmeier KA. Prenatal nicotine exposure alters postsynaptic AMPA receptors and glutamate neurotransmission within the laterodorsal tegmentum (LDT) of juvenile mice. Neuropharmacology 2018; 137:71-85. [PMID: 29751228 DOI: 10.1016/j.neuropharm.2018.04.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/20/2018] [Accepted: 04/22/2018] [Indexed: 02/06/2023]
Abstract
Despite dissemination of information regarding the harm on fetal development of smoking while pregnant, the number of pregnancies associated with nicotine exposure appears to have stagnated. Presence of nicotine during neural formulation is associated with a higher susceptibility of drug dependence, suggesting an altered development of neurons in circuits involved in saliency and motivation. The laterodorsal tegmental nucleus (LDT) plays a role in coding stimuli valence via afferents to mesolimbic nuclei. Accordingly, alterations in development of neural mechanisms in the LDT could be involved in vulnerability to drug dependency. Therefore, we examined the effect of prenatal nicotine exposure (PNE) on glutamatergic functioning of LDT neurons in mouse brain slices using whole-cell, patch clamp concurrent with fluorescence-based calcium imaging. PNE was associated with larger amplitudes of AMPA-induced currents, and greater AMPA-mediated rises in intracellular calcium. AMPA/NMDA ratios and the AMPA-current rectification index were lower and higher, respectively, consistent with changes in the functionality of AMPA receptors in the PNE, which was substantiated by a greater inhibition of evoked and spontaneous glutamatergic synaptic events by a selective inhibitor of GluA2-lacking AMPA receptors. Paired pulse ratios showed a decreased probability of glutamate release from presynaptic inputs, and fluorescent imaging indicated a decreased action potential-dependent calcium increase associated with PNE. When taken together, our data suggest that PNE alters LDT glutamatergic functioning, which could alter output to mesolimbic targets. Such an alteration could play a role in altered coding of relevancy of drug stimuli that could enhance risk for development of drug dependency.
Collapse
Affiliation(s)
- Filip S Polli
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen 2100, Denmark.
| |
Collapse
|
35
|
Wang Y, Wang L, Zhu Y, Qin J. Human brain organoid-on-a-chip to model prenatal nicotine exposure. LAB ON A CHIP 2018; 18:851-860. [PMID: 29437173 DOI: 10.1039/c7lc01084b] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Nicotine has been recognized to trigger various neuronal disabilities in the fetal brain and long-lasting behavioral deficits in offspring. However, further understanding of fetal brain development under nicotine exposure is challenging due to the limitations of existing animal models. Here, we create a new brain organoid-on-a-chip system derived from human induced pluripotent stem cells (hiPSCs) that allows us to model neurodevelopmental disorders under prenatal nicotine exposure (PNE) at early stages. The brain organoid-on-a-chip system facilitates 3D culture, in situ neural differentiation, and self-organization of brain organoids under continuous perfused cultures in a controlled manner. The generated brain organoids displayed well-defined neural differentiation, regionalization, and cortical organization, which recapitulates the key features of the early stages of human brain development. The brain organoids exposed to nicotine exhibited premature neuronal differentiation with enhanced expression of the neuron marker TUJ1. Brain regionalization and cortical development were disrupted in the nicotine-treated organoids identified by the expressions of forebrain (PAX6 and FOXG1), hindbrain (PAX2 and KROX20) and cortical neural layer (preplate TBR1 and deep-layer CTIP2) markers. Moreover, the neurite outgrowth showed abnormal neuronal differentiation and migration in nicotine-treated brain organoids. These results suggest that nicotine exposure elicits impaired neurogenesis in early fetal brain development during gestation. The established brain organoid-on-a-chip system provides a promising platform to model neurodevelopmental disorders under environmental exposure, which can be extended for applications in brain disease studies and drug testing.
Collapse
Affiliation(s)
- Yaqing Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China.
| | | | | | | |
Collapse
|
36
|
Yates JR. Dissecting drug effects in preclinical models of impulsive choice: emphasis on glutamatergic compounds. Psychopharmacology (Berl) 2018; 235:607-626. [PMID: 29305628 PMCID: PMC5823766 DOI: 10.1007/s00213-017-4825-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/27/2017] [Indexed: 01/10/2023]
Abstract
RATIONALE Impulsive choice is often measured with delay discounting paradigms. Because there are multiple discounting procedures, as well as different statistical analyses that can be applied to data generated from these paradigms, there are some inconsistencies in the literature regarding drug effects on impulsive choice. OBJECTIVES The goal of the current paper is to review the methodological and analytic approaches used to measure discounting and to discuss how these differences can account for differential drug effects observed across studies. RESULTS Because some procedures/analyses use a single data point as the dependent variable, changes in this value following pharmacological treatment may be interpreted as alterations in sensitivity to delayed reinforcement, but when other procedures/analyses are used, no changes in behavior are observed. Even when multiple data points are included, some studies show that the statistical analysis (e.g., ANOVA on raw proportion of responses vs. using hyperbolic/exponential functions) can lead to different interpretations. Finally, procedural differences (e.g., delay presentation order, signaling the delay to reinforcement, etc.) in the same discounting paradigm can alter how drugs affect sensitivity to delayed reinforcement. CONCLUSIONS Future studies should utilize paradigms that allow one to observe alterations in responding at each delay (e.g., concurrent-chains schedules). Concerning statistical analyses, using parameter estimates derived from nonlinear functions or incorporating the generalized matching law can allow one to determine if drugs affect sensitivity to delayed reinforcement or impair discrimination of the large and small magnitude reinforcers. Using these approaches can help further our understanding of the neurochemical underpinnings of delay discounting.
Collapse
Affiliation(s)
- Justin R Yates
- Department of Psychological Science, Northern Kentucky University, 1 Nunn Drive, Highland Heights, KY, 41099, USA.
| |
Collapse
|
37
|
Alkam T, Mamiya T, Kimura N, Yoshida A, Kihara D, Tsunoda Y, Aoyama Y, Hiramatsu M, Kim HC, Nabeshima T. Prenatal nicotine exposure decreases the release of dopamine in the medial frontal cortex and induces atomoxetine-responsive neurobehavioral deficits in mice. Psychopharmacology (Berl) 2017; 234:1853-1869. [PMID: 28332006 DOI: 10.1007/s00213-017-4591-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/05/2017] [Indexed: 02/06/2023]
Abstract
Increased risk of attention-deficit/hyperactivity disorder (AD/HD) is partly associated with the early developmental exposure to nicotine in tobacco smoke. Emerging reports link tobacco smoke exposure or prenatal nicotine exposure (PNE) with AD/HD-like behaviors in rodent models. We have previously reported that PNE induces cognitive behavioral deficits in offspring and decreases the contents of dopamine (DA) and its turnover in the prefrontal cortex (PFC) of offspring It is well known that the dysfunction of DAergic system in the brain is one of the core factors in the pathophysiology of AD/HD. Therefore, we examined whether the effects of PNE on the DAergic system underlie the AD/HD-related behavioral changes in mouse offspring. PNE reduced the release of DA in the medial PFC (mPFC) in mouse offspring. PNE reduced the number of tyrosine hydroxylase (TH)-positive varicosities in the mPFC and in the core as well as the shell of nucleus accumbens, but not in the striatum. PNE also induced behavioral deficits in cliff avoidance, object-based attention, and sensorimotor gating in offspring. These behavioral deficits were attenuated by acute treatment with atomoxetine (3 mg/kg, s.c.) or partially attenuated by acute treatment with MPH (1 mg/kg, s.c.). Taken together, our findings support the notion that PNE induces neurobehavioral abnormalities in mouse offspring by disrupting the DAergic system and improve our understanding about the incidence of AD/HD in children whose mothers were exposed to nicotine during their pregnancy.
Collapse
Affiliation(s)
- Tursun Alkam
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Takayoshi Mamiya
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Nami Kimura
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Aya Yoshida
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Daisuke Kihara
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Yuki Tsunoda
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Yuki Aoyama
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Masayuki Hiramatsu
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, South Korea
| | - Toshitaka Nabeshima
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan.
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan.
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.
- Aino University, Ibaraki, Japan.
| |
Collapse
|
38
|
Neurodevelopmental Disorders and Environmental Toxicants: Epigenetics as an Underlying Mechanism. Int J Genomics 2017; 2017:7526592. [PMID: 28567415 PMCID: PMC5439185 DOI: 10.1155/2017/7526592] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/02/2017] [Indexed: 01/07/2023] Open
Abstract
The increasing prevalence of neurodevelopmental disorders, especially autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD), calls for more research into the identification of etiologic and risk factors. The Developmental Origin of Health and Disease (DOHaD) hypothesizes that the environment during fetal and childhood development affects the risk for many chronic diseases in later stages of life, including neurodevelopmental disorders. Epigenetics, a term describing mechanisms that cause changes in the chromosome state without affecting DNA sequences, is suggested to be the underlying mechanism, according to the DOHaD hypothesis. Moreover, many neurodevelopmental disorders are also related to epigenetic abnormalities. Experimental and epidemiological studies suggest that exposure to prenatal environmental toxicants is associated with neurodevelopmental disorders. In addition, there is also evidence that environmental toxicants can result in epigenetic alterations, notably DNA methylation. In this review, we first focus on the relationship between neurodevelopmental disorders and environmental toxicants, in particular maternal smoking, plastic-derived chemicals (bisphenol A and phthalates), persistent organic pollutants, and heavy metals. We then review studies showing the epigenetic effects of those environmental factors in humans that may affect normal neurodevelopment.
Collapse
|
39
|
Zhu J, Fan F, McCarthy DM, Zhang L, Cannon EN, Spencer TJ, Biederman J, Bhide PG. A prenatal nicotine exposure mouse model of methylphenidate responsive ADHD‐associated cognitive phenotypes. Int J Dev Neurosci 2017; 58:26-34. [DOI: 10.1016/j.ijdevneu.2017.01.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/21/2017] [Accepted: 01/27/2017] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jinmin Zhu
- Center for Brain Repair and The Department of Biomedical SciencesFlorida State University College of MedicineTallahasseeFL32306United States
| | - Fangfang Fan
- Center for Brain Repair and The Department of Biomedical SciencesFlorida State University College of MedicineTallahasseeFL32306United States
| | - Deirdre M. McCarthy
- Center for Brain Repair and The Department of Biomedical SciencesFlorida State University College of MedicineTallahasseeFL32306United States
| | - Lin Zhang
- Center for Brain Repair and The Department of Biomedical SciencesFlorida State University College of MedicineTallahasseeFL32306United States
| | - Elisa N. Cannon
- Center for Brain Repair and The Department of Biomedical SciencesFlorida State University College of MedicineTallahasseeFL32306United States
| | - Thomas J. Spencer
- Pediatric Psychopharmacology, Department of PsychiatryMassachusetts General Hospital, Harvard Medical SchoolBostonMA02114United States
| | - Joseph Biederman
- Pediatric Psychopharmacology, Department of PsychiatryMassachusetts General Hospital, Harvard Medical SchoolBostonMA02114United States
| | - Pradeep G. Bhide
- Center for Brain Repair and The Department of Biomedical SciencesFlorida State University College of MedicineTallahasseeFL32306United States
| |
Collapse
|
40
|
England LJ, Aagaard K, Bloch M, Conway K, Cosgrove K, Grana R, Gould TJ, Hatsukami D, Jensen F, Kandel D, Lanphear B, Leslie F, Pauly JR, Neiderhiser J, Rubinstein M, Slotkin TA, Spindel E, Stroud L, Wakschlag L. Developmental toxicity of nicotine: A transdisciplinary synthesis and implications for emerging tobacco products. Neurosci Biobehav Rev 2017; 72:176-189. [PMID: 27890689 PMCID: PMC5965681 DOI: 10.1016/j.neubiorev.2016.11.013] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/18/2016] [Accepted: 11/19/2016] [Indexed: 12/24/2022]
Abstract
While the health risks associated with adult cigarette smoking have been well described, effects of nicotine exposure during periods of developmental vulnerability are often overlooked. Using MEDLINE and PubMed literature searches, books, reports and expert opinion, a transdisciplinary group of scientists reviewed human and animal research on the health effects of exposure to nicotine during pregnancy and adolescence. A synthesis of this research supports that nicotine contributes critically to adverse effects of gestational tobacco exposure, including reduced pulmonary function, auditory processing defects, impaired infant cardiorespiratory function, and may contribute to cognitive and behavioral deficits in later life. Nicotine exposure during adolescence is associated with deficits in working memory, attention, and auditory processing, as well as increased impulsivity and anxiety. Finally, recent animal studies suggest that nicotine has a priming effect that increases addiction liability for other drugs. The evidence that nicotine adversely affects fetal and adolescent development is sufficient to warrant public health measures to protect pregnant women, children, and adolescents from nicotine exposure.
Collapse
Affiliation(s)
- Lucinda J England
- Office on Smoking and Health, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Kjersti Aagaard
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Michele Bloch
- Division of Cancer Control and Population Science, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Kevin Conway
- Division of Epidemiology, Services and Prevention Research, National Institute on Drug Abuse, National Institutes of Health, Rockville, MD, USA
| | - Kelly Cosgrove
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Rachel Grana
- Division of Cancer Control and Population Science, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Pennsylvania State University, PA, USA
| | | | - Frances Jensen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Denise Kandel
- Department of Psychiatry and Mailman School of Public Health, Columbia University, New York State Psychiatric Institute, New York, NY, USA
| | | | - Frances Leslie
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA, USA
| | - James R Pauly
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Jenae Neiderhiser
- Department of Psychology, Pennsylvania State University, University Park, PA, USA
| | - Mark Rubinstein
- Department of Pediatrics, School of Medicine, University of California, San Francisco, CA, USA
| | - Theodore A Slotkin
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Eliot Spindel
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Laura Stroud
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
| | - Lauren Wakschlag
- Department of Medical Social Sciences Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
41
|
Kanlikilicer P, Zhang D, Dragomir A, Akay YM, Akay M. Gene expression profiling of midbrain dopamine neurons upon gestational nicotine exposure. Med Biol Eng Comput 2016; 55:467-482. [PMID: 27255453 DOI: 10.1007/s11517-016-1531-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/18/2016] [Indexed: 12/11/2022]
Abstract
Maternal smoking during pregnancy is associated with low birth weight, increased risk of stillbirth, conduct disorder, attention-deficit/hyperactivity disorder and neurocognitive deficits. Ventral tegmental area dopamine (DA) neurons in the mesocorticolimbic pathway were suggested to play a critical role in these pathological mechanisms induced by nicotine. Nicotine-mediated changes in genetic expression during pregnancy are of great interest for current researchers. We used patch clamp methods to identify and harvest DA and non-DA neurons separately and assayed them using oligonucleotide arrays to elucidate the alterations in gene expressions in these cells upon gestational nicotine exposure. Microarray analysis identified a set of 135 genes as significantly differentially expressed between DA and non-DA neurons. Some of the genes were found to be related to neurological disease pathways, such as Alzheimer's disease, Parkinson's disease and Huntington's disease. Significantly up-/down-regulated genes found in DA neurons were mostly related to G-protein-coupled protein receptor signaling and developmental processes. These alterations in gene expressions may explain, partially at least, the possible pathological mechanisms for the diseases induced by maternal smoking.
Collapse
Affiliation(s)
- Pınar Kanlikilicer
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA
| | - Die Zhang
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA
| | - Andrei Dragomir
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, SERC Building, 3605 Cullen Blvd, Houston, TX, 77204, USA.
| |
Collapse
|
42
|
Nespoli E, Rizzo F, Boeckers TM, Hengerer B, Ludolph AG. Addressing the Complexity of Tourette's Syndrome through the Use of Animal Models. Front Neurosci 2016; 10:133. [PMID: 27092043 PMCID: PMC4824761 DOI: 10.3389/fnins.2016.00133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/16/2016] [Indexed: 01/06/2023] Open
Abstract
Tourette's syndrome (TS) is a neurodevelopmental disorder characterized by fluctuating motor and vocal tics, usually preceded by sensory premonitions, called premonitory urges. Besides tics, the vast majority—up to 90%—of TS patients suffer from psychiatric comorbidities, mainly attention deficit/hyperactivity disorder (ADHD) and obsessive-compulsive disorder (OCD). The etiology of TS remains elusive. Genetics is believed to play an important role, but it is clear that other factors contribute to TS, possibly altering brain functioning and architecture during a sensitive phase of neural development. Clinical brain imaging and genetic studies have contributed to elucidate TS pathophysiology and disease mechanisms; however, TS disease etiology still is poorly understood. Findings from genetic studies led to the development of genetic animal models, but they poorly reflect the pathophysiology of TS. Addressing the role of neurotransmission, brain regions, and brain circuits in TS disease pathomechanisms is another focus area for preclinical TS model development. We are now in an interesting moment in time when numerous innovative animal models are continuously brought to the attention of the public. Due to the diverse and largely unknown etiology of TS, there is no single preclinical model featuring all different aspects of TS symptomatology. TS has been dissected into its key symptomst hat have been investigated separately, in line with the Research Domain Criteria concept. The different rationales used to develop the respective animal models are critically reviewed, to discuss the potential of the contribution of animal models to elucidate TS disease mechanisms.
Collapse
Affiliation(s)
- Ester Nespoli
- Competence in Neuro Spine Department, Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der Riss, Germany; Department of Child and Adolescence Psychiatry/Psychotherapy, University of UlmUlm, Germany
| | - Francesca Rizzo
- Department of Child and Adolescence Psychiatry/Psychotherapy, University of UlmUlm, Germany; Institute of Anatomy and Cell Biology, University of UlmUlm, Germany
| | - Tobias M Boeckers
- Institute of Anatomy and Cell Biology, University of Ulm Ulm, Germany
| | - Bastian Hengerer
- Competence in Neuro Spine Department, Boehringer Ingelheim Pharma GmbH & Co. KG Biberach an der Riss, Germany
| | - Andrea G Ludolph
- Department of Child and Adolescence Psychiatry/Psychotherapy, University of Ulm Ulm, Germany
| |
Collapse
|
43
|
Hamdi Y, Madfai H, Belhareth R, Mokni M, Masmoudi-Kouki O, Amri M. Prenatal exposure to cigarette smoke enhances oxidative stress in astrocytes of neonatal rat. Toxicol Mech Methods 2016; 26:231-7. [PMID: 26998663 DOI: 10.3109/15376516.2016.1156205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Oxidative stress is involved in the pathogenesis of smoking-related disease. Protection of astrocytes from oxidative insult appears essential to maintain brain function. In this study, we have investigated the effect of gestational cigarette exposure on astrocyte survival. Pregnant female were randomly allocated to the control group or to the cigarette smoke group in which they were placed in an exposure chamber and inhale three cigarettes smoke twice a day for a period of 20 days. The control group was kept in the exposure chamber for the same duration, but without exposure to cigarette smoke. Newborn rats from both groups were weighed 24 h after birth and then cerebral hemispheres were collected for astrocyte culture. Incubation of astrocytes isolated from animals exposed to cigarette smoke with 300 μM H2O2 for 1 h induced a significant decrease of the proportion of surviving cells compared to cells isolated form control animals. We have observed that H2O2-treated astroglial cells derived from cigarette smoke exposure showed more reduced superoxide dismutase and catalase activities than H2O2-treated astroglial cells from control animals. In conclusion, this study indicates that astroglial cells derived from newborn rats exposed in utero to cigarette smoke are more vulnerable to oxidative assault than cultured astrocytes obtained from control animals. These results point out the existence of excitotoxic lesions in newborn exposed in utero to cigarette smoke and suggest that despite their high antioxidative activities, astrocytes cannot survive and protect neurons under massive oxidative stress.
Collapse
Affiliation(s)
- Yosra Hamdi
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Hayfa Madfai
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Rym Belhareth
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Meherzia Mokni
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Olfa Masmoudi-Kouki
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| | - Mohamed Amri
- a Laboratory of Functional Neurophysiology and Pathology, Research Unit UR11ES09, Department of Biological Sciences , Faculty of Science of Tunis, University Tunis El Manar , Tunis , Tunisia
| |
Collapse
|
44
|
Hall BJ, Cauley M, Burke DA, Kiany A, Slotkin TA, Levin ED. Cognitive and Behavioral Impairments Evoked by Low-Level Exposure to Tobacco Smoke Components: Comparison with Nicotine Alone. Toxicol Sci 2016; 151:236-44. [PMID: 26919958 DOI: 10.1093/toxsci/kfw042] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Active maternal smoking has adverse effects on neurobehavioral development of the offspring, with nicotine (Nic) providing much of the underlying causative mechanism. To determine whether the lower exposures caused by second-hand smoke are deleterious, we administered tobacco smoke extract (TSE) to pregnant rats starting preconception and continued through the second postnatal week, corresponding to all 3 trimesters of fetal brain development. Dosing was adjusted to produce maternal plasma Nic concentrations encountered with second-hand smoke, an order of magnitude below those seen in active smokers. We then compared TSE effects to those of an equivalent dose of Nic alone, and to a 10-fold higher Nic dose. Gestational exposure to TSE and Nic significantly disrupted cognitive and behavioral function in behavioral tests given during adolescence and adulthood (postnatal weeks 4-40), producing hyperactivity, working memory deficits, and impairments in emotional processing, even at the low exposure levels corresponding to second-hand smoke. Although TSE effects were highly correlated with those of Nic, the effects of TSE were much larger than could be attributed to just the Nic in the mixture. Indeed, TSE effects more closely resembled those of the 10-fold higher Nic levels, but still exceeded their magnitude. In combination with our earlier findings, this study thus completes the chain of causation to prove that second-hand smoke exposure causes neurodevelopmental deficits, originating in disruption of neurodifferentiation, leading to miswiring of neuronal circuits, and as shown here, culminating in behavioral dysfunction. As low level exposure to Nic alone produced neurobehavioral teratology, 'harm reduction' Nic products do not abolish the potential for neurodevelopmental damage.
Collapse
Affiliation(s)
| | - Marty Cauley
- *Department of Psychiatry and Behavioral Sciences
| | | | - Abtin Kiany
- *Department of Psychiatry and Behavioral Sciences
| | - Theodore A Slotkin
- *Department of Psychiatry and Behavioral Sciences Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710
| | - Edward D Levin
- *Department of Psychiatry and Behavioral Sciences Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
45
|
Hadas I, Gal R, Bokovza L, Meiran N, Feifel D, Zangen A. Exposure to salient, dynamic sensory stimuli during development increases distractibility in adulthood. Sci Rep 2016; 6:21129. [PMID: 26882890 PMCID: PMC4756326 DOI: 10.1038/srep21129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/18/2016] [Indexed: 11/23/2022] Open
Abstract
It has been suggested that excessive exposure of children to the dynamic and highly salient audio-visual stimuli conveyed by electronic media may induce attention-related deficits in adulthood. This study was designed to evaluate this hypothesis in a controlled animal model setup. Building on their natural responsiveness to odors, we exposed juvenile rats for 1 h daily to a dynamic series of interchanging, highly salient odors, while controls were exposed to a non-changing mixture of these odors. Upon reaching adulthood, we tested the attentional capacity of the rats and measured their brain-derived neurotrophic factor (BDNF) levels as a proxy of neuronal plasticity. As compared with controls, rats exposed to the dynamic stimulation showed no attentional deficits under baseline task conditions, but their performance was dramatically impaired when an auditory distractor was introduced in the task. In addition, BDNF levels in the dorsal striatum of these rats were significantly increased relative to controls. These findings provide first empirical evidence that a continuous exposure to dynamic, highly salient stimuli has long-term effects on attentional functions later in life, and that these effects may have neural correlates in the dorsal striatum.
Collapse
Affiliation(s)
- Itay Hadas
- Department of Life Sciences and the Zlotowski Centre for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ram Gal
- Department of Life Sciences and the Zlotowski Centre for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lihi Bokovza
- Department of Life Sciences and the Zlotowski Centre for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nachshon Meiran
- Department of Psychology and the Zlotowski Centre for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - David Feifel
- Department of Psychiatry and Neurosciences Program, University of California, San Diego, USA
| | - Abraham Zangen
- Department of Life Sciences and the Zlotowski Centre for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
46
|
Bryden DW, Burton AC, Barnett BR, Cohen VJ, Hearn TN, Jones EA, Kariyil RJ, Kunin A, In Kwak S, Lee J, Lubinski BL, Rao GK, Zhan A, Roesch MR. Prenatal Nicotine Exposure Impairs Executive Control Signals in Medial Prefrontal Cortex. Neuropsychopharmacology 2016; 41:716-25. [PMID: 26189451 PMCID: PMC4707818 DOI: 10.1038/npp.2015.197] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/26/2015] [Accepted: 05/27/2015] [Indexed: 01/06/2023]
Abstract
Prenatal nicotine exposure (PNE) is linked to numerous psychiatric disorders including attention deficit hyperactivity disorder (ADHD). Current literature suggests that core deficits observed in ADHD reflect abnormal inhibitory control governed by the prefrontal cortex. Yet, it is unclear how neural activity in the medial prefrontal cortex (mPFC) is modulated during tasks that assess response inhibition or if these neural correlates, along with behavior, are affected by PNE. To address this issue, we recorded from single mPFC neurons in control and PNE rats as they performed a stop-signal task. We found that PNE rats were faster for all trial-types, made more premature responses, and were less likely to inhibit behavior on 'STOP' trials during which rats had to inhibit an already initiated response. Activity in mPFC was modulated by response direction and was positively correlated with accuracy and movement time in control but not PNE rats. Although the number of single neurons correlated with response direction was significantly reduced by PNE, neural activity observed on general STOP trials was largely unaffected. However, dramatic behavioral deficits on STOP trials immediately following non-conflicting (GO) trials in the PNE group appear to be mediated by the loss of conflict monitoring signals in mPFC. We conclude that prenatal nicotine exposure makes rats impulsive and disrupts firing of mPFC neurons that carry signals related to response direction and conflict monitoring.
Collapse
Affiliation(s)
- Daniel W Bryden
- Department of Psychology, University of Maryland, College Park, MD, USA,Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA,Department of Psychology and Program in Neuroscience and Cognitive Science, University of Maryland, Bio-psyc Building, College Park, MD 20742, USA, Tel: 301 405 2274, Fax: 301 314 9566, E-mail: or
| | - Amanda C Burton
- Department of Psychology, University of Maryland, College Park, MD, USA,Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA
| | - Brian R Barnett
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Valerie J Cohen
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Taylor N Hearn
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Emily A Jones
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Reshma J Kariyil
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Alice Kunin
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Sae In Kwak
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Jessica Lee
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Brooke L Lubinski
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Gautam K Rao
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Ashley Zhan
- Gemstone Honors Program, University of Maryland, College Park, MD, USA
| | - Matthew R Roesch
- Department of Psychology, University of Maryland, College Park, MD, USA,Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA,Department of Psychology and Program in Neuroscience and Cognitive Science, University of Maryland, Bio-psyc Building, College Park, MD 20742, USA, Tel: 301 405 2274, Fax: 301 314 9566, E-mail: or
| |
Collapse
|
47
|
Sheets LP, Li AA, Minnema DJ, Collier RH, Creek MR, Peffer RC. A critical review of neonicotinoid insecticides for developmental neurotoxicity. Crit Rev Toxicol 2016; 46:153-90. [PMID: 26513508 PMCID: PMC4732412 DOI: 10.3109/10408444.2015.1090948] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 01/21/2023]
Abstract
A comprehensive review of published and previously unpublished studies was performed to evaluate the neonicotinoid insecticides for evidence of developmental neurotoxicity (DNT). These insecticides have favorable safety profiles, due to their preferential affinity for nicotinic receptor (nAChR) subtypes in insects, poor penetration of the mammalian blood-brain barrier, and low application rates. Nevertheless, examination of this issue is warranted, due to their insecticidal mode of action and potential exposure with agricultural and residential uses. This review identified in vitro, in vivo, and epidemiology studies in the literature and studies performed in rats in accordance with GLP standards and EPA guidelines with imidacloprid, acetamiprid, thiacloprid, clothianidin, thiamethoxam, and dinotefuran, which are all the neonicotinoids currently registered in major markets. For the guideline-based studies, treatment was administered via the diet or gavage to primiparous female rats at three dose levels, plus a vehicle control (≥20/dose level), from gestation day 0 or 6 to lactation day 21. F1 males and females were evaluated using measures of motor activity, acoustic startle response, cognition, brain morphometry, and neuropathology. The principal effects in F1 animals were associated with decreased body weight (delayed sexual maturation, decreased brain weight, and morphometric measurements) and acute toxicity (decreased activity during exposure) at high doses, without neuropathology or impaired cognition. No common effects were identified among the neonicotinoids that were consistent with DNT or the neurodevelopmental effects associated with nicotine. Findings at high doses were associated with evidence of systemic toxicity, which indicates that these insecticides do not selectively affect the developing nervous system.
Collapse
Affiliation(s)
| | - Abby A. Li
- Exponent Health Sciences,
San Francisco,
CA,
USA
| | | | | | | | | |
Collapse
|
48
|
Besson M, Forget B. Cognitive Dysfunction, Affective States, and Vulnerability to Nicotine Addiction: A Multifactorial Perspective. Front Psychiatry 2016; 7:160. [PMID: 27708591 PMCID: PMC5030478 DOI: 10.3389/fpsyt.2016.00160] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 09/06/2016] [Indexed: 11/17/2022] Open
Abstract
Although smoking prevalence has declined in recent years, certain subpopulations continue to smoke at disproportionately high rates and show resistance to cessation treatments. Individuals showing cognitive and affective impairments, including emotional distress and deficits in attention, memory, and inhibitory control, particularly in the context of psychiatric conditions, such as attention-deficit hyperactivity disorder, schizophrenia, and mood disorders, are at higher risk for tobacco addiction. Nicotine has been shown to improve cognitive and emotional processing in some conditions, including during tobacco abstinence. Self-medication of cognitive deficits or negative affect has been proposed to underlie high rates of tobacco smoking among people with psychiatric disorders. However, pre-existing cognitive and mood disorders may also influence the development and maintenance of nicotine dependence, by biasing nicotine-induced alterations in information processing and associative learning, decision-making, and inhibitory control. Here, we discuss the potential forms of contribution of cognitive and affective deficits to nicotine addiction-related processes, by reviewing major clinical and preclinical studies investigating either the procognitive and therapeutic action of nicotine or the putative primary role of cognitive and emotional impairments in addiction-like features.
Collapse
Affiliation(s)
- Morgane Besson
- Unité de Neurobiologie Intégrative des Systèmes Cholinergiques, Department of Neuroscience, CNRS UMR 3571, Institut Pasteur , Paris , France
| | - Benoît Forget
- Unité de Neurobiologie Intégrative des Systèmes Cholinergiques, Department of Neuroscience, CNRS UMR 3571, Institut Pasteur , Paris , France
| |
Collapse
|
49
|
Aoyama Y, Toriumi K, Mouri A, Hattori T, Ueda E, Shimato A, Sakakibara N, Soh Y, Mamiya T, Nagai T, Kim HC, Hiramatsu M, Nabeshima T, Yamada K. Prenatal Nicotine Exposure Impairs the Proliferation of Neuronal Progenitors, Leading to Fewer Glutamatergic Neurons in the Medial Prefrontal Cortex. Neuropsychopharmacology 2016; 41:578-89. [PMID: 26105135 PMCID: PMC5130133 DOI: 10.1038/npp.2015.186] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/22/2015] [Accepted: 06/13/2015] [Indexed: 12/20/2022]
Abstract
Cigarette smoking during pregnancy is associated with various disabilities in the offspring such as attention deficit/hyperactivity disorder, learning disabilities, and persistent anxiety. We have reported that nicotine exposure in female mice during pregnancy, in particular from embryonic day 14 (E14) to postnatal day 0 (P0), induces long-lasting behavioral deficits in offspring. However, the mechanism by which prenatal nicotine exposure (PNE) affects neurodevelopment, resulting in behavioral deficits, has remained unclear. Here, we report that PNE disrupted the proliferation of neuronal progenitors, leading to a decrease in the progenitor pool in the ventricular and subventricular zones. In addition, using a cumulative 5-bromo-2'-deoxyuridine labeling assay, we evaluated the rate of cell cycle progression causing the impairment of neuronal progenitor proliferation, and uncovered anomalous cell cycle kinetics in mice with PNE. Accordingly, the density of glutamatergic neurons in the medial prefrontal cortex (medial PFC) was reduced, implying glutamatergic dysregulation. Mice with PNE exhibited behavioral impairments in attentional function and behavioral flexibility in adulthood, and the deficits were ameliorated by microinjection of D-cycloserine into the PFC. Collectively, our findings suggest that PNE affects the proliferation and maturation of progenitor cells to glutamatergic neuron during neurodevelopment in the medial PFC, which may be associated with cognitive deficits in the offspring.
Collapse
Affiliation(s)
- Yuki Aoyama
- Department of Neuropsychopharmacology and Hospital Pharmacy, Graduate School of Medicine, Nagoya University, Nagoya, Japan,Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Kazuya Toriumi
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Akihiro Mouri
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan,Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Tomoya Hattori
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Eriko Ueda
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Akane Shimato
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Nami Sakakibara
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Yuka Soh
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Takayoshi Mamiya
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan,Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hyoung-Chun Kim
- Department of Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, South Korea
| | - Masayuki Hiramatsu
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan,Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Toshitaka Nabeshima
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan,Nabeshima Laboratory, Faculty of Pharmacy, Meijo University, Nagoya, Japan,Nabeshima Laboratory, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tenpaku-ku, Nagoya 468-8503, Japan, Tel: +81 52 839 2756, Fax: +81 52 839 2756, E-mail:
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Graduate School of Medicine, Nagoya University, Nagoya, Japan,Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan,Department of Neuropsychopharmacology and Hospital Pharmacy, Graduate School of Medicine, Nagoya University, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan, Tel: +81 52 744 2674, Fax: +81 52 744 2979, E-mail:
| |
Collapse
|
50
|
Kutlu MG, Parikh V, Gould TJ. Nicotine Addiction and Psychiatric Disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 124:171-208. [PMID: 26472530 DOI: 10.1016/bs.irn.2015.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Even though smoking rates have long been on the decline, nicotine addiction still affects 20% of the US population today. Moreover, nicotine dependence shows high comorbidity with many mental illnesses including, but are not limited to, attention deficit hyperactivity disorder, anxiety disorders, and depression. The reason for the high rates of smoking in patients with mental illnesses may relate to attempts to self-medicate with nicotine. While nicotine may alleviate the symptoms of mental disorders, nicotine abstinence has been shown to worsen the symptoms of these disorders. In this chapter, we review the studies from animal and human research examining the bidirectional relationship between nicotine and attention deficit hyperactivity disorder, anxiety disorders, and depression as well as studies examining the roles of specific subunits of nicotinic acetylcholine receptors (nAChRs) in the interaction between nicotine and these mental illnesses. The results of these studies suggest that activation, desensitization, and upregulation of nAChRs modulate the effects of nicotine on mental illnesses.
Collapse
Affiliation(s)
| | - Vinay Parikh
- Temple University, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|