1
|
Schinz D, Schmitz‐Koep B, Zimmermann J, Brandes E, Tahedl M, Menegaux A, Dukart J, Zimmer C, Wolke D, Daamen M, Boecker H, Bartmann P, Sorg C, Hedderich DM. Indirect evidence for altered dopaminergic neurotransmission in very premature-born adults. Hum Brain Mapp 2023; 44:5125-5138. [PMID: 37608591 PMCID: PMC10502650 DOI: 10.1002/hbm.26451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 06/23/2023] [Accepted: 07/28/2023] [Indexed: 08/24/2023] Open
Abstract
While animal models indicate altered brain dopaminergic neurotransmission after premature birth, corresponding evidence in humans is scarce due to missing molecular imaging studies. To overcome this limitation, we studied dopaminergic neurotransmission changes in human prematurity indirectly by evaluating the spatial co-localization of regional alterations in blood oxygenation fluctuations with the distribution of adult dopaminergic neurotransmission. The study cohort comprised 99 very premature-born (<32 weeks of gestation and/or birth weight below 1500 g) and 107 full-term born young adults, being assessed by resting-state functional MRI (rs-fMRI) and IQ testing. Normative molecular imaging dopamine neurotransmission maps were derived from independent healthy control groups. We computed the co-localization of local (rs-fMRI) activity alterations in premature-born adults with respect to term-born individuals to different measures of dopaminergic neurotransmission. We performed selectivity analyses regarding other neuromodulatory systems and MRI measures. In addition, we tested if the strength of the co-localization is related to perinatal measures and IQ. We found selectively altered co-localization of rs-fMRI activity in the premature-born cohort with dopamine-2/3-receptor availability in premature-born adults. Alterations were specific for the dopaminergic system but not for the used MRI measure. The strength of the co-localization was negatively correlated with IQ. In line with animal studies, our findings support the notion of altered dopaminergic neurotransmission in prematurity which is associated with cognitive performance.
Collapse
Affiliation(s)
- David Schinz
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Benita Schmitz‐Koep
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Juliana Zimmermann
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Elin Brandes
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Marlene Tahedl
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Aurore Menegaux
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Juergen Dukart
- Institute of Neuroscience and MedicineBrain & Behaviour (INM‐7), Research Centre JülichJülichGermany
- Institute of Systems Neuroscience, Medical FacultyHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Claus Zimmer
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Dieter Wolke
- Department of PsychologyUniversity of WarwickCoventryUK
- Warwick Medical SchoolUniversity of WarwickCoventryUK
| | - Marcel Daamen
- Clinical Functional Imaging Group, Department of Diagnostic and Interventional RadiologyUniversity Hospital BonnBonnGermany
- Department of NeonatologyUniversity Hospital BonnBonnGermany
| | - Henning Boecker
- Clinical Functional Imaging Group, Department of Diagnostic and Interventional RadiologyUniversity Hospital BonnBonnGermany
| | - Peter Bartmann
- Department of NeonatologyUniversity Hospital BonnBonnGermany
| | - Christian Sorg
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
- Department of Psychiatry, School of MedicineTechnical University of MunichMunichGermany
| | - Dennis M. Hedderich
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| |
Collapse
|
2
|
Seib DR, Tobiansky DJ, Meitzen J, Floresco SB, Soma KK. Neurosteroids and the mesocorticolimbic system. Neurosci Biobehav Rev 2023; 153:105356. [PMID: 37567491 DOI: 10.1016/j.neubiorev.2023.105356] [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: 12/29/2022] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
The mesocorticolimbic system coordinates executive functions, such as working memory and behavioral flexibility. This circuit includes dopaminergic projections from the ventral tegmental area to the nucleus accumbens and medial prefrontal cortex. In this review, we summarize evidence that cells in multiple nodes of the mesocorticolimbic system produce neurosteroids (steroids synthesized in the nervous system) and express steroid receptors. Here, we focus on neuroandrogens (androgens synthesized in the nervous system), neuroestrogens (estrogens synthesized in the nervous system), and androgen and estrogen receptors. We also summarize how (neuro)androgens and (neuro)estrogens affect dopamine signaling in the mesocorticolimbic system and regulate executive functions. Taken together, the data suggest that steroids produced in the gonads and locally in the brain modulate higher-order cognition and executive functions.
Collapse
Affiliation(s)
- Désirée R Seib
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| | - Daniel J Tobiansky
- Department of Biology and Neuroscience Program, St. Mary's College of Maryland, St. Mary's City, MD, USA
| | - John Meitzen
- Department of Biological Sciences and Center for Human Health and the Environment, NC State University, Raleigh, NC, USA
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Kiran K Soma
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Deer LK, Su C, Thwaites NA, Davis EP, Doom JR. A framework for testing pathways from prenatal stress-responsive hormones to cardiovascular disease risk. Front Endocrinol (Lausanne) 2023; 14:1111474. [PMID: 37223037 PMCID: PMC10200937 DOI: 10.3389/fendo.2023.1111474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/10/2023] [Indexed: 05/25/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death globally, with the prevalence projected to keep rising. Risk factors for adult CVD emerge at least as early as the prenatal period. Alterations in stress-responsive hormones in the prenatal period are hypothesized to contribute to CVD in adulthood, but little is known about relations between prenatal stress-responsive hormones and early precursors of CVD, such as cardiometabolic risk and health behaviors. The current review presents a theoretical model of the relation between prenatal stress-responsive hormones and adult CVD through cardiometabolic risk markers (e.g., rapid catch-up growth, high BMI/adiposity, high blood pressure, and altered blood glucose, lipids, and metabolic hormones) and health behaviors (e.g., substance use, poor sleep, poor diet and eating behaviors, and low physical activity levels). Emerging evidence in human and non-human animal literatures suggest that altered stress-responsive hormones during gestation predict higher cardiometabolic risk and poorer health behaviors in offspring. This review additionally highlights limitations of the current literature (e.g., lack of racial/ethnic diversity, lack of examination of sex differences), and discusses future directions for this promising area of research.
Collapse
Affiliation(s)
- LillyBelle K. Deer
- Department of Psychology, University of Denver, Denver, CO, United States
| | - Chen Su
- Department of Psychology, University of Denver, Denver, CO, United States
| | | | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, United States
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA, United States
| | - Jenalee R. Doom
- Department of Psychology, University of Denver, Denver, CO, United States
| |
Collapse
|
4
|
Dong Y, Weng J, Zhu Y, Sun D, He W, Chen Q, Cheng J, Zhu Y, Jiang Y. Transcriptomic profiling of the developing brain revealed cell-type and brain-region specificity in a mouse model of prenatal stress. BMC Genomics 2023; 24:86. [PMID: 36829105 PMCID: PMC9951484 DOI: 10.1186/s12864-023-09186-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Prenatal stress (PS) is considered as a risk factor for many mental disorders. PS-induced transcriptomic alterations may contribute to the functional dysregulation during brain development. Here, we used RNA-seq to explore changes of gene expression in the mouse fetal brain after prenatal exposure to chronic unpredictable mild stress (CUMS). RESULTS We compared the stressed brains to the controls and identified groups of significantly differentially expressed genes (DEGs). GO analysis on up-regulated DEGs revealed enrichment for the cell cycle pathways, while down-regulated DEGs were mostly enriched in the neuronal pathways related to synaptic transmission. We further performed cell-type enrichment analysis using published scRNA-seq data from the fetal mouse brain and revealed cell-type-specificity for up- and down-regulated DEGs, respectively. The up-regulated DEGs were highly enriched in the radial glia, while down-regulated DEGs were enriched in different types of neurons. Cell deconvolution analysis further showed altered cell fractions in the stressed brain, indicating accumulation of neuroblast and impaired neurogenesis. Moreover, we also observed distinct brain-region expression pattern when mapping DEGs onto the developing Allen brain atlas. The up-regulated DEGs were primarily enriched in the dorsal forebrain regions including the cortical plate and hippocampal formation. Surprisingly, down-regulated DEGs were found excluded from the cortical region, but highly expressed on various regions in the ventral forebrain, midbrain and hindbrain. CONCLUSION Taken together, we provided an unbiased data source for transcriptomic alterations of the whole fetal brain after chronic PS, and reported differential cell-type and brain-region vulnerability of the developing brain in response to environmental insults during the pregnancy.
Collapse
Affiliation(s)
- Yuhao Dong
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Jie Weng
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Yueyan Zhu
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Daijing Sun
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Wei He
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Qi Chen
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Jin Cheng
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Ying Zhu
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Yan Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
5
|
The Modification of Offspring Stress-Related Behavior and the Expression of Drd1, Drd2, and Nr3c1 by a Western-Pattern Diet in Mus Musculus. Int J Mol Sci 2022; 23:ijms23169245. [PMID: 36012509 PMCID: PMC9409213 DOI: 10.3390/ijms23169245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
The impact of early developmental experience on neurobiological pathways that may contribute to the association between diet and behavior have not yet been elucidated. The focus of the current study was to determine whether the impact of prenatal stress (PS) could be mitigated by a diet that stimulates the same neuroendocrine systems influenced by early stress, using a mouse model. Behavioral and genetic approaches were used to assess how a Western-pattern diet (WPD) interacts with PS and sex to impact the expression of anxiety-like behavior in an open-field arena, as well as the expression of the glucocorticoid receptor in the hippocampus, D1 dopamine receptors in the nucleus accumbens, and D2 dopamine receptors in the ventral tegmental area. Overall, the results demonstrated that a prenatal WPD mitigates the effects of maternal stress in dams and offspring. These results help to elucidate the relationship between pre- and post-natal nutrition, gene expression, and behaviors that lead to long-term health effects.
Collapse
|
6
|
Braun K, Bock J, Wainstock T, Matas E, Gaisler-Salomon I, Fegert J, Ziegenhain U, Segal M. Experience-induced transgenerational (re-)programming of neuronal structure and functions: Impact of stress prior and during pregnancy. Neurosci Biobehav Rev 2020; 117:281-296. [DOI: 10.1016/j.neubiorev.2017.05.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022]
|
7
|
Dunn AL, Michie PT, Hodgson DM, Harms L. Adolescent cannabinoid exposure interacts with other risk factors in schizophrenia: A review of the evidence from animal models. Neurosci Biobehav Rev 2020; 116:202-220. [PMID: 32610181 DOI: 10.1016/j.neubiorev.2020.06.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022]
Abstract
Many factors and their interaction are linked to the aetiology of schizophrenia, leading to the development of animal models of multiple risk factors and adverse exposures. Differentiating between separate and combined effects for each factor could better elucidate schizophrenia pathology, and drive development of preventative strategies for high-load risk factors. An epidemiologically valid risk factor commonly associated with schizophrenia is adolescent cannabis use. The aim of this review is to evaluate how early-life adversity from various origins, in combination with adolescent cannabinoid exposure interact, and whether these interactions confer main, synergistic or protective effects in animal models of schizophrenia-like behavioural, cognitive and morphological alterations. Patterns emerge regarding which models show consistent synergistic or protective effects, particularly those models incorporating early-life exposure to maternal deprivation and maternal immune activation, and sex-specific effects are observed. It is evident that more research needs to be conducted to better understand the risks and alterations of interacting factors, with particular interest in sex differences, to better understand the translatability of these preclinical models to humans.
Collapse
Affiliation(s)
- Ariel L Dunn
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Patricia T Michie
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Deborah M Hodgson
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Lauren Harms
- Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
8
|
María-Ríos CE, Morrow JD. Mechanisms of Shared Vulnerability to Post-traumatic Stress Disorder and Substance Use Disorders. Front Behav Neurosci 2020; 14:6. [PMID: 32082127 PMCID: PMC7006033 DOI: 10.3389/fnbeh.2020.00006] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Psychoactive substance use is a nearly universal human behavior, but a significant minority of people who use addictive substances will go on to develop an addictive disorder. Similarly, though ~90% of people experience traumatic events in their lifetime, only ~10% ever develop post-traumatic stress disorder (PTSD). Substance use disorders (SUD) and PTSD are highly comorbid, occurring in the same individual far more often than would be predicted by chance given the respective prevalence of each disorder. Some possible reasons that have been proposed for the relationship between PTSD and SUD are self-medication of anxiety with drugs or alcohol, increased exposure to traumatic events due to activities involved in acquiring illegal substances, or addictive substances altering the brain's stress response systems to make users more vulnerable to PTSD. Yet another possibility is that some people have an intrinsic vulnerability that predisposes them to both PTSD and SUD. In this review, we integrate clinical and animal data to explore these possible etiological links between SUD and PTSD, with an emphasis on interactions between dopaminergic, adrenocorticotropic, GABAergic, and glutamatergic neurobehavioral mechanisms that underlie different emotional learning styles.
Collapse
Affiliation(s)
| | - Jonathan D. Morrow
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
9
|
Swann AC, Lijffijt M, O'Brien B, Mathew SJ. Impulsivity and Suicidal Behavior. Curr Top Behav Neurosci 2020; 47:179-195. [PMID: 32472429 DOI: 10.1007/7854_2020_144] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Suicide is the leading cause of injury mortality in the United States and the second-leading cause of death in people aged 10-34 years. While many long-term risk factors are known, the short-term prediction of suicidal behavior remains elusive. Many characteristics of suicidal behavior cut across diagnoses, but suicide is increased in recurrent psychiatric disorders, addictive disorders, and trauma-related disorders. Suicide results from the interaction of short-term and long-term behavioral regulation. The shorter the time-course of the mechanism, the closer it is to actual suicidal behavior, and the harder it is to prevent. We will discuss the manner in which impulsivity, a major determinant of short-term suicide risk, interacts with longer-term risk factors, especially sensitization to addictive or traumatic stimuli. Impulsivity predisposes to sensitization; in turn, impulsivity is a prominent component of sensitized behavior. Impulsivity can be described as a general pattern of behavior ("trait" impulsivity), as responses that are not conformed to their context (action-impulsivity), or as inability to delay reward or to take future consequences into account (choice-impulsivity). Each of these contributes to suicidal behavior. The neural mechanisms of impulsivity and sensitization are analogous, and sensitization can produce rapidly fluctuating patterns of impulsive behavior, arousal, and anhedonia. In order to recognize and prevent suicidal behavior, it is necessary to identify factors associated with susceptibility to bouts of impulsive behavior in people at elevated long-term risk.
Collapse
Affiliation(s)
- Alan C Swann
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA. .,Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.
| | - Marijn Lijffijt
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Research Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Brittany O'Brien
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Sanjay J Mathew
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.,Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
10
|
Roles of nucleus accumbens shell and core in footshock-induced stress altering behavioral sensitization by methamphetamine in acquisition and testing: Running head: stress, nucleus accumbens, and behavioral sensitization. Behav Brain Res 2019; 380:112434. [PMID: 31846629 DOI: 10.1016/j.bbr.2019.112434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/02/2019] [Accepted: 12/13/2019] [Indexed: 12/15/2022]
Abstract
How the subregions of the nucleus accumbens (NAc) shell and core and stress are involved in behavioral sensitization induced by psychostimulants remains unclear. The present study manipulated methamphetamine (MAMPH) injections, lesions of the NAc shell or core, and footshock-treatment-induced stress to address this issue. The present data showed that during the acquisition phase, MAMPH injections, lesions of the NAc shell, and footshock treatments induced hyperactivity for the NAc shell. For the NAc core, MAMPH injections induced hyperactivity; however, lesions of the NAc core did not affect locomotor activity. Footshock treatments disrupted hyperactivity of behavioral sensitization. During the testing phase, MAMPH injections, lesions of the NAc shell, and footshock-treatment-induced stress facilitated hyperactivity for the NAc shell. For the NAc core, MAMPH injections and footshock-treatment-induced stress increased hyperactivity. However, the lesion of the NAc core did not affect locomotor activity. In conclusion, MAMPH injections and footshock-treatment-induced stress play an excitatory role for the NAc shell in acquisition and testing. For the NAc core, footshock-treatment-induced stress plays an inhibitory role in acquisition but an excitatory role in testing. The NAc core was not involved in MAMPH-induced behavioral sensitization in acquisition and testing. The NAc shell plays an inhibitory role in acquisition and testing phases. The present data might provide some insights for drug addiction. The results should be discussed further.
Collapse
|
11
|
Bagley JR, Szumlinski KK, Kippin TE. Discovery of early life stress interacting and sex-specific quantitative trait loci impacting cocaine responsiveness. Br J Pharmacol 2019; 176:4159-4172. [PMID: 30874305 DOI: 10.1111/bph.14661] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/12/2019] [Accepted: 02/18/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Addiction vulnerability involves complex gene X environment interactions leading to a pathological response to drugs. Identification of the genes involved in these interactions is an important step in understanding the underlying neurobiology and rarely have such analyses examined sex-specific influences. To dissect this interaction, we examined the impact of prenatal stress (PNS) on cocaine responsiveness in male and female mice of the BXD recombinant inbred panel. EXPERIMENTAL APPROACH BXD strains were subjected to timed mating and assigned to PNS or control groups. PNS dams were subjected to restraint stress (1-hr restraint, three times daily) starting between embryonic day (E) 11 and 14 and continued until parturition. Adult male and female, control and PNS offspring were tested for locomotor response to initial and repeated cocaine injections (sensitization) as well as cocaine-induced conditioned place preference (CPP). KEY RESULTS Strain, PNS, and sex interacted to modulate initial and sensitized cocaine-induced locomotion, as well as CPP. Moreover, a quantitative trait locus (QTL) interacting with PNS regulating initial locomotor response to cocaine (chromosome X, 37.91 to 50.95 Mb) was identified. Also PNS-independent, female-specific QTLs regulating CPP (chromosome 11, 65.50 to 81.31 Mb) and sensitized cocaine-induced locomotion (chromosome 16, 95.79 to 98.32 Mb) were identified. Publicly available mRNA expression data were utilized to identify cis-eQTL and transcript covariation with the behavioural phenotype to prioritize candidate genes; including Aifm1. CONCLUSIONS AND IMPLICATIONS These QTL encompass genes that may moderate genetic susceptibility to PNS and interact with sex to determine adult responsiveness to cocaine and addiction vulnerability. LINKED ARTICLES This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Jared R Bagley
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, California
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, California.,Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California.,Department of Molecular Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, California.,Institute for Collaborative Biotechnologies, University of California Santa Barbara, Santa Barbara, California
| | - Tod E Kippin
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, California.,Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California.,Department of Molecular Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, California.,Institute for Collaborative Biotechnologies, University of California Santa Barbara, Santa Barbara, California
| |
Collapse
|
12
|
Wikenius E, Myhre AM, Page CM, Moe V, Smith L, Heiervang ER, Undlien DE, LeBlanc M. Prenatal maternal depressive symptoms and infant DNA methylation: a longitudinal epigenome-wide study. Nord J Psychiatry 2019; 73:257-263. [PMID: 31070508 DOI: 10.1080/08039488.2019.1613446] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background: Prenatal maternal stress increases the risk of offspring developmental and psychological difficulties. The biological mechanisms behind these associations are mostly unknown. One explanation suggests that exposure of the fetus to maternal stress may influence DNA methylation. However, this hypothesis is largely based on animal studies, and human studies of candidate genes from single timepoints. Aim: The aim of this study was to investigate if prenatal maternal stress, in the form of maternal depressive symptoms, was associated with variation in genome-wide DNA methylation at two timepoints. Methods: One-hundred and eighty-four mother-child dyads were selected from a population of pregnant women in the Little-in-Norway study. The Edinburgh Postnatal Depression Scale (EPDS) measured maternal depressive symptoms. It was completed by the pregnant mothers between weeks 17 and 32 of gestation. DNA was obtained from infant saliva cells at two timepoints (age 6 weeks and 12 months). DNA methylation was measured in 274 samples from 6 weeks (n = 146) and 12 months (n = 128) using the Illumina Infinium HumanMethylation 450 BeadChip. Linear regression analyses of prenatal maternal depressive symptoms and infant methylation were performed at 6 weeks and 12 months separately, and for both timepoints together using a mixed model. Results: The analyses revealed no significant genome-wide association between maternal depressive symptoms and infant DNA methylation in the separate analyses and for both timepoints together. Conclusions: This sample of pregnant women and their infants living in Norway did not reveal associations between maternal depressive symptoms and infant DNA methylation.
Collapse
Affiliation(s)
- Ellen Wikenius
- a Faculty of Medicine , Institute of Clinical Medicine, University of Oslo , Oslo , Norway.,b Child & Adolescent Mental Health Research Unit , Oslo University Hospital , Oslo , Norway
| | - Anne Margrethe Myhre
- a Faculty of Medicine , Institute of Clinical Medicine, University of Oslo , Oslo , Norway.,b Child & Adolescent Mental Health Research Unit , Oslo University Hospital , Oslo , Norway
| | - Christian Magnus Page
- c Oslo Centre for Biostatistics and Epidemiology , Oslo University Hospital , Oslo , Norway.,d Centre for Fertility and Health , Norwegian Institute of Public Health , Oslo , Norway
| | - Vibeke Moe
- e The Department of Psychology, Faculty of Social Sciences , University of Oslo , Oslo , Norway.,f Center for Child and Adolescent Mental Health , Eastern and Southern Norway (RBUP) , Oslo , Norway
| | - Lars Smith
- e The Department of Psychology, Faculty of Social Sciences , University of Oslo , Oslo , Norway
| | - Einar Røshol Heiervang
- a Faculty of Medicine , Institute of Clinical Medicine, University of Oslo , Oslo , Norway.,b Child & Adolescent Mental Health Research Unit , Oslo University Hospital , Oslo , Norway
| | - Dag Erik Undlien
- a Faculty of Medicine , Institute of Clinical Medicine, University of Oslo , Oslo , Norway.,g Department of Medical Genetics , Oslo University Hospital , Oslo , Norway
| | - Marissa LeBlanc
- c Oslo Centre for Biostatistics and Epidemiology , Oslo University Hospital , Oslo , Norway
| |
Collapse
|
13
|
|
14
|
Park G, Jung YS, Park MK, Yang CH, Kim YU. Melatonin inhibits attention-deficit/hyperactivity disorder caused by atopic dermatitis-induced psychological stress in an NC/Nga atopic-like mouse model. Sci Rep 2018; 8:14981. [PMID: 30297827 PMCID: PMC6175954 DOI: 10.1038/s41598-018-33317-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/27/2018] [Indexed: 12/28/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease with the hallmark characteristics of pruritus, psychological stress, and sleep disturbance, all possibly associated with an increased risk of attention-deficit/hyperactivity disorder (ADHD). However, the etiology of the possible association between AD and ADHD is still not well understood. 2,4-dinitrochlorobenzene or corticosterone was used to evaluate the atopic symptom and its psychologic stress in the atopic mice model. Melatonin, corticotropin-releasing hormone, corticotropin-releasing hormone receptor, urocortin, proopiomelanocortin, adrenocorticotropic hormone, corticosterone, cAMP, cAMP response element-binding protein, dopamine and noradrenaline were analyzed spectrophotometrically, and the expression of dopamine beta-hydroxylase and tyrosine hydroxylase were measured by Western blotting or immunohistochemistry. AD-related psychological stress caused an increase in the levels of dopamine beta-hydroxylase and tyrosine hydroxylase, degradation of melatonin, hyper-activity of the hypothalamic-pituitary-adrenal axis, and dysregulation of dopamine and noradrenaline levels (ADHD phenomena) in the locus coeruleus, prefrontal cortex, and striatum of the AD mouse brain. Notably, melatonin administration inhibited the development of ADHD phenomena and their-related response in the mouse model. This study demonstrated that AD-related psychological stress increased catecholamine dysfunction and accelerated the development of psychiatric comorbidities, such as ADHD.
Collapse
MESH Headings
- Animals
- Attention Deficit Disorder with Hyperactivity/chemically induced
- Attention Deficit Disorder with Hyperactivity/drug therapy
- Attention Deficit Disorder with Hyperactivity/metabolism
- Attention Deficit Disorder with Hyperactivity/pathology
- Brain/metabolism
- Brain/pathology
- Cell Line, Transformed
- Dermatitis, Atopic/chemically induced
- Dermatitis, Atopic/drug therapy
- Dermatitis, Atopic/metabolism
- Dermatitis, Atopic/pathology
- Disease Models, Animal
- Humans
- Melatonin/pharmacology
- Mice
- Stress, Psychological/chemically induced
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
- Stress, Psychological/pathology
Collapse
Affiliation(s)
- Gunhyuk Park
- The K-herb Research Center, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea.
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Moon-Ki Park
- Department of Pharmaceutical Engineering, College of Biomedical Science, Daegu Haany University, 290 Yugok-dong, Gyeongsan-si, Gyeongsangbuk-do, 38610, Republic of Korea
| | - Chae Ha Yang
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea
| | - Yong-Ung Kim
- Department of Pharmaceutical Engineering, College of Biomedical Science, Daegu Haany University, 290 Yugok-dong, Gyeongsan-si, Gyeongsangbuk-do, 38610, Republic of Korea.
| |
Collapse
|
15
|
Horn SR, Roos LE, Berkman ET, Fisher PA. Neuroendocrine and immune pathways from pre- and perinatal stress to substance abuse. Neurobiol Stress 2018; 9:140-150. [PMID: 30450380 PMCID: PMC6236513 DOI: 10.1016/j.ynstr.2018.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/06/2018] [Accepted: 09/12/2018] [Indexed: 12/26/2022] Open
Abstract
Early life adversity is a documented risk factor for substance abuse and addiction. The pre- and perinatal period (i.e., from implantation, through pregnancy, to 6 months of age) is a critical period marked by high biological plasticity and vulnerability, making perinatal stress a particularly robust form of adversity. The neuroendocrine and immune systems are key mechanisms implicated in the transmission of addiction risk. We review animal and human studies that provide preliminary evidence for links between perinatal stress, neuroendocrine and immune dysregulation, and risk for substance abuse and addiction. A translational neuroscience perspective is employed to elucidate pre- and perinatally-induced biological mechanisms linked to addiction and discuss implications for prevention and intervention efforts. Significant evidence supports associations between pre- and perinatal stress and dysregulation of the hypothalamic-pituitary-adrenal axis and immune systems as well as links between neuroendocrine/immune functioning and addiction risk. More work is needed to explicitly examine the interplay between pre- and perinatal stress and neuroendocrine/immune disruptions that together heighten substance abuse risk. Future work is needed to fully understand how pre- and perinatal stress induces biological alterations to predispose individuals to higher risk for addiction. Such knowledge will strengthen theoretically-driven and empirically-supported prevention efforts for substance abuse and addiction.
Collapse
Affiliation(s)
- Sarah R Horn
- University of Oregon, Department of Psychology, 1227 University of Oregon, Eugene, OR, 97402, USA
| | - Leslie E Roos
- University of Oregon, Department of Psychology, 1227 University of Oregon, Eugene, OR, 97402, USA
| | - Elliot T Berkman
- University of Oregon, Department of Psychology, 1227 University of Oregon, Eugene, OR, 97402, USA
| | - Philip A Fisher
- University of Oregon, Department of Psychology, 1227 University of Oregon, Eugene, OR, 97402, USA
| |
Collapse
|
16
|
Frasch MG, Lobmaier SM, Stampalija T, Desplats P, Pallarés ME, Pastor V, Brocco MA, Wu HT, Schulkin J, Herry CL, Seely AJE, Metz GAS, Louzoun Y, Antonelli MC. Non-invasive biomarkers of fetal brain development reflecting prenatal stress: An integrative multi-scale multi-species perspective on data collection and analysis. Neurosci Biobehav Rev 2018; 117:165-183. [PMID: 29859198 DOI: 10.1016/j.neubiorev.2018.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/09/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023]
Abstract
Prenatal stress (PS) impacts early postnatal behavioural and cognitive development. This process of 'fetal programming' is mediated by the effects of the prenatal experience on the developing hypothalamic-pituitary-adrenal (HPA) axis and autonomic nervous system (ANS). We derive a multi-scale multi-species approach to devising preclinical and clinical studies to identify early non-invasively available pre- and postnatal biomarkers of PS. The multiple scales include brain epigenome, metabolome, microbiome and the ANS activity gauged via an array of advanced non-invasively obtainable properties of fetal heart rate fluctuations. The proposed framework has the potential to reveal mechanistic links between maternal stress during pregnancy and changes across these physiological scales. Such biomarkers may hence be useful as early and non-invasive predictors of neurodevelopmental trajectories influenced by the PS as well as follow-up indicators of success of therapeutic interventions to correct such altered neurodevelopmental trajectories. PS studies must be conducted on multiple scales derived from concerted observations in multiple animal models and human cohorts performed in an interactive and iterative manner and deploying machine learning for data synthesis, identification and validation of the best non-invasive detection and follow-up biomarkers, a prerequisite for designing effective therapeutic interventions.
Collapse
Affiliation(s)
- Martin G Frasch
- Department of Obstetrics and Gynecology, University of Washington, Seattle, USA.
| | - Silvia M Lobmaier
- Frauenklinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tamara Stampalija
- Unit of Fetal Medicine and Prenatal Diagnosis, Institute for Mother and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Paula Desplats
- University of California, Departments of Neurosciences and Pathology, San Diego, USA
| | - María Eugenia Pallarés
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Verónica Pastor
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Marcela A Brocco
- Instituto de Investigaciones Biotecnológicas - Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín - Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), San Martín, Buenos Aires, Argentina
| | - Hau-Tieng Wu
- Department of Mathematics and Department of Statistical Science, Duke University, Durham, NC, USA; Mathematics Division, National Center for Theoretical Sciences, Taipei, Taiwan
| | - Jay Schulkin
- Department of Obstetrics and Gynecology, University of Washington, Seattle, USA
| | | | | | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Yoram Louzoun
- Bar-Ilan University, Department of Applied Mathematics, Israel
| | - Marta C Antonelli
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Argentina
| |
Collapse
|
17
|
Thomas MB, Becker JB. Sex differences in prenatal stress effects on cocaine pursuit in rats. Physiol Behav 2017; 203:3-9. [PMID: 29055747 DOI: 10.1016/j.physbeh.2017.10.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/02/2017] [Accepted: 10/17/2017] [Indexed: 11/29/2022]
Abstract
Disruption of early-life ontogeny has severe and persistent consequences for the health of the developing organism. Both clinical and preclinical findings indicate that such interference can be caused by maternal stress during the gestation period (prenatal stress [PS]). In rats, PS facilitates the rewarding and neurochemical-stimulating effects of drugs, suggesting that PS may represent a risk factor for drug abuse in humans. Very little, however, is known about its effects in females, even though sex differences in drug susceptibility have been well documented in no PS (NPS) controls. Thus, we tested for independent effects and interactions between maternal restraint stress during the last week of gestation and sex on drug use with an extended regimen of drug self-administration. Male and female rats were provided daily access to a large but controlled amount of cocaine for seven weeks. Drug pursuit during the final week was used to indicate susceptibility to developing an addiction-like phenotype, based on reports that drug use becomes increasingly compulsive-like after weeks of testing. Overall, females satisfied more addiction-like criteria than males, and the same was true for PS rats when compared to NPS controls. In addition, sex and PS interacted to disproportionately promote drug pursuit of females with a history of PS. These results indicate that sex differences in drug susceptibility persist with continued drug exposure, and that PS widens this difference by more severely affecting females. In all, PS may be a risk factor for drug addiction in humans, and to a greater extent in women vs. men.
Collapse
Affiliation(s)
- Mark B Thomas
- Psychology Department, University of Michigan, Ann Arbor, MI, United States; Sciformix, 1500 West Park Drive, Westborough, MA, United States
| | - Jill B Becker
- Psychology Department, University of Michigan, Ann Arbor, MI, United States; Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States; Neuroscience Program, Psychiatry Department, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
18
|
Oliveira C, Scarabelot VL, Vercelino R, Silveira NP, Adachi LN, Regner GG, Silva LS, Macedo IC, Souza A, Caumo W, Torres IL. Morphine exposure and maternal deprivation during the early postnatal period alter neuromotor development and nerve growth factor levels. Int J Dev Neurosci 2017; 63:8-15. [DOI: 10.1016/j.ijdevneu.2017.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 09/02/2017] [Accepted: 09/03/2017] [Indexed: 12/17/2022] Open
Affiliation(s)
- Carla Oliveira
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduaçăo em Medicina: Ciências MédicasFaculdade de MedicinaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Vanessa L. Scarabelot
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Rafael Vercelino
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Centro Universitário FADERGSPorto AlegreRSBrazil
- Health and Wellness School Laureate International Universities
| | - Natalia P. Silveira
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Lauren N.S. Adachi
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduaçăo em Medicina: Ciências MédicasFaculdade de MedicinaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Gabriela G. Regner
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Lisiane S. Silva
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Isabel Cristina Macedo
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Universidade Federal do PampaAvenida Antônio Trilha, 184797300‐000São GabrielRSBrazil
| | - Andressa Souza
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Wolnei Caumo
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduaçăo em Medicina: Ciências MédicasFaculdade de MedicinaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Iraci L.S. Torres
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduaçăo em Medicina: Ciências MédicasFaculdade de MedicinaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| |
Collapse
|
19
|
Nazeri M, Ebrahimi A, Aghaei I, Ghotbi Ravandi S, Shabani M. Psychological stress has a higher rate of developing addictive behaviors compared to physical stress in rat offspring. EXCLI JOURNAL 2017; 16:903-913. [PMID: 28900372 PMCID: PMC5579401 DOI: 10.17179/excli2016-685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 06/09/2017] [Indexed: 01/01/2023]
Abstract
Prenatal stress could have great influence on development of offspring and might alter cognitive function and other physiological processes of children. The current study was conducted to study the effect of physical or psychological prenatal stress on addictive and anxiety-like behavior of male and female offspring during their adolescence period (postnatal day (PND) 40). Adult female rats were exposed to physical (swimming) or psychological (observing another female rat swimming) stress from day six of gestation for 10 days. Male and female offspring were assayed for anxiety-like behavior, motor and balance function and morphine conditioned place preference using the open field, elevated plus maze (EPM), rotarod and wire grip assay and conditioned place preference. Offspring in both physical and psychological prenatal stress groups demonstrated significant increase in anxiety-like behavior in EPM paradigm, but no alterations were observed in motor and balance function of animals. Offspring in the psychological prenatal stress group had an increased preference for morphine in comparison to control and physical prenatal stress groups. Results of the current study demonstrated that animals exposed to psychological stress during fetal development are at a higher risk of developing addictive behaviors. Further research might elucidate the exact mechanisms involved to provide better preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Masoud Nazeri
- Department of Oral Medicine and Chronic Headache and Facial Pain Clinic, School of Dentistry, Kerman, IranUniversity of Medical Sciences, Kerman, Iran
| | - Arezoo Ebrahimi
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Iraj Aghaei
- Social Determinants of Health Research Center, Gilan University of Medical Sciences, Rasht, Iran
| | - Samaneh Ghotbi Ravandi
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
20
|
Fujáková-Lipski M, Kaping D, Šírová J, Horáček J, Páleníček T, Zach P, Klaschka J, Kačer P, Syslová K, Vrajová M, Bubenikova-Valešová V, Beste C, Šlamberová R. Trans-generational neurochemical modulation of methamphetamine in the adult brain of the Wistar rat. Arch Toxicol 2017; 91:3373-3384. [PMID: 28477265 DOI: 10.1007/s00204-017-1969-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 03/31/2017] [Indexed: 11/25/2022]
Abstract
Chronic methamphetamine (METH) abuse has been shown to elicit strong neurotoxic effects. Yet, with an increasing number of children born to METH abusing mothers maturing into adulthood, one important question is how far do the neurotoxic effects of METH alter various neurotransmitter systems in the adult METH-exposed offspring. The purpose of this study was to investigate long-term trans-generational neurochemical changes, following prenatal METH exposure, in the adult Wistar rat brain. METH or saline (SAL-control animals) was administered to pregnant dams throughout the entire gestation period (G0-G22). At postnatal day 90, dopamine, serotonin, glutamate and GABA were measured in the adult brain before (baseline) and after a METH re-administration using in vivo microdialysis and liquid chromatography/mass spectrometry. The results show that METH-exposure increased basal levels of monoamines and glutamate, but decreased GABA levels in all measured brain regions. Acute challenge with METH injection in the METH-exposed group induced a lower increase in the monoamine system relative to the increase in the GABAergic and glutamatergic system. The data show that prenatal METH exposure has strong effects on the monoaminergic, GABAergic and glutamatergic system even when exposure to METH was limited to the prenatal phase. Toxicological effects of METH have therefore longer lasting effects as currently considered and seem to affect the excitatory-inhibitory balance in the brain having strong implications for cognitive and behavioral functioning.
Collapse
Affiliation(s)
- Michaela Fujáková-Lipski
- National Institute of Mental Health, Klecany, Czech Republic.
- Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Daniel Kaping
- National Institute of Mental Health, Klecany, Czech Republic.
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Jana Šírová
- National Institute of Mental Health, Klecany, Czech Republic
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiří Horáček
- National Institute of Mental Health, Klecany, Czech Republic
| | - Tomáš Páleníček
- National Institute of Mental Health, Klecany, Czech Republic
| | - Petr Zach
- Department of Anatomy, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Klaschka
- Department of Medical Informatics and Biostatistics, Institute of Computer Science, The Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Kačer
- National Institute of Mental Health, Klecany, Czech Republic
| | - Kamila Syslová
- Department of Organic Technology, University of Chemistry and Technology, Prague, Czech Republic
| | - Monika Vrajová
- National Institute of Mental Health, Klecany, Czech Republic
| | | | - Christian Beste
- National Institute of Mental Health, Klecany, Czech Republic
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Romana Šlamberová
- Department of Normal, Pathological and Clinical Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
21
|
Enduring, Sexually Dimorphic Impact of In Utero Exposure to Elevated Levels of Glucocorticoids on Midbrain Dopaminergic Populations. Brain Sci 2016; 7:brainsci7010005. [PMID: 28042822 PMCID: PMC5297294 DOI: 10.3390/brainsci7010005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/14/2016] [Accepted: 12/16/2016] [Indexed: 11/17/2022] Open
Abstract
Glucocorticoid hormones (GCs) released from the fetal/maternal glands during late gestation are required for normal development of mammalian organs and tissues. Accordingly, synthetic glucocorticoids have proven to be invaluable in perinatal medicine where they are widely used to accelerate fetal lung maturation when there is risk of pre-term birth and to promote infant survival. However, clinical and pre-clinical studies have demonstrated that inappropriate exposure of the developing brain to elevated levels of GCs, either as a result of clinical over-use or after stress-induced activation of the fetal/maternal adrenal cortex, is linked with significant effects on brain structure, neurological function and behaviour in later life. In order to understand the underlying neural processes, particular interest has focused on the midbrain dopaminergic systems, which are critical regulators of normal adaptive behaviours, cognitive and sensorimotor functions. Specifically, using a rodent model of GC exposure in late gestation (approximating human brain development at late second/early third trimester), we demonstrated enduring effects on the shape and volume of the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) (origins of the mesocorticolimbic and nigrostriatal dopaminergic pathways) on the topographical organisation and size of the dopaminergic neuronal populations and astrocytes within these nuclei and on target innervation density and neurochemical markers of dopaminergic transmission (receptors, transporters, basal and amphetamine-stimulated dopamine release at striatal and prefrontal cortical sites) that impact on the adult brain. The effects of antenatal GC treatment (AGT) were both profound and sexually-dimorphic, not only in terms of quantitative change but also qualitatively, with several parameters affected in the opposite direction in males and females. Although such substantial neurobiological changes might presage marked behavioural effects, in utero GC exposure had only a modest or no effect, depending on sex, on a range of conditioned and unconditioned behaviours known to depend on midbrain dopaminergic transmission. Collectively, these findings suggest that apparent behavioural normality in certain tests, but not others, arises from AGT-induced adaptations or compensatory mechanisms within the midbrain dopaminergic systems, which preserve some, but not all functions. Furthermore, the capacities for molecular adaptations to early environmental challenge are different, even opponent, in males and females, which may account for their differential resilience or failure to perform adequately in behavioural tests. Behavioural "normality" is thus achieved by the midbrain dopaminergic network operating outside its normal limits (in a state of allostasis), rendering it at greater risk to malfunction when challenged in later life. Sex-specific neurobiological programming of midbrain dopaminergic systems may, therefore, have psychopathological relevance for the sex bias commonly found in brain disorders associated with these systems, and which have a neurodevelopmental component, including schizophrenia, ADHD (attention/deficit hyperactivity disorders), autism, depression and substance abuse.
Collapse
|
22
|
Yates NJ, Robertson D, Rodger J, Martin-Iverson MT. Effects of Neonatal Dexamethasone Exposure on Adult Neuropsychiatric Traits in Rats. PLoS One 2016; 11:e0167220. [PMID: 27936175 PMCID: PMC5147874 DOI: 10.1371/journal.pone.0167220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/10/2016] [Indexed: 11/18/2022] Open
Abstract
The effects of early life stress in utero or in neonates has long-term consequences on hypothalamic-pituitary-adrenal (HPA) stress axis function and neurodevelopment. These effects extend into adulthood and may underpin a variety of mental illnesses and be related to various developmental and cognitive changes. We examined the potential role of neonatal HPA axis activation on adult psychopathology and dopamine sensitivity in the mature rat using neonatal exposure to the synthetic glucocorticoid receptor agonist and stress hormone, dexamethasone. We utilized a comprehensive battery of assessments for behaviour, brain function and gene expression to determine if elevated early life HPA activation is associated with adult-onset neuropsychiatric traits. Dexamethasone exposure increased startle reactivity under all conditions tested, but decreased sensitivity of sensorimotor gating to dopaminergic disruption–contrasting with what is observed in several neuropsychiatric diseases. Under certain conditions there also appeared to be mild long-term changes in stress and anxiety-related behaviours with neonatal dexamethasone exposure. Electrophysiology revealed that there were no consistent neuropsychiatric abnormalities in auditory processing or resting state brain function with dexamethasone exposure. However, neonatal dexamethasone altered auditory cortex glucocorticoid activation, and auditory cortex synchronization. Our results indicate that neonatal HPA axis activation by dexamethasone alters several aspects of adult brain function and behaviour and may induce long-term changes in emotional stress-reactivity. However, neonatal dexamethasone exposure is not specifically related to any particular neuropsychiatric disease.
Collapse
Affiliation(s)
- Nathanael J. Yates
- School of Animal Biology, Faculty of Science, The University of Western Australia, Crawley, Western Australia, Australia
- * E-mail:
| | - Donald Robertson
- School of Anatomy, Physiology, and Human Biology, Faculty of Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Jennifer Rodger
- School of Animal Biology, Faculty of Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Mathew T. Martin-Iverson
- School of Medicine and Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
23
|
Pastor V, Antonelli MC, Pallarés ME. Unravelling the Link Between Prenatal Stress, Dopamine and Substance Use Disorder. Neurotox Res 2016; 31:169-186. [DOI: 10.1007/s12640-016-9674-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 12/21/2022]
|
24
|
Trifilieff P, Ducrocq F, van der Veldt S, Martinez D. Blunted Dopamine Transmission in Addiction: Potential Mechanisms and Implications for Behavior. Semin Nucl Med 2016; 47:64-74. [PMID: 27987559 DOI: 10.1053/j.semnuclmed.2016.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Positron emission tomography (PET) imaging consistently shows blunted striatal dopamine release and decreased dopamine D2 receptor availability in addiction. Here, we review the preclinical and clinical studies indicating that this neurobiological phenotype is likely to be both a consequence of chronic drug consumption and a vulnerability factor in the development of addiction. We propose that, behaviorally, blunted striatal dopamine transmission could reflect the increased impulsivity and altered cost/benefit computations that are associated with addiction. The factors that influence blunted striatal dopamine transmission in addiction are unknown. Herein, we give an overview of various factors, genetic, environmental, and social, that are known to affect dopamine transmission and that have been associated with the vulnerability to develop addiction. Altogether, these data suggest that blunted dopamine transmission and decreased D2 receptor availability are biomarkers both for the development of addiction and resistance to treatment. These findings support the view that blunted dopamine reflects impulsive behavior and deficits in motivation, which lead to the escalation of drug use.
Collapse
Affiliation(s)
- Pierre Trifilieff
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, University of Bordeaux, Bordeaux, France.
| | - Fabien Ducrocq
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, University of Bordeaux, Bordeaux, France
| | - Suzanne van der Veldt
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, University of Bordeaux, Bordeaux, France; Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Diana Martinez
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical College, New York, NY.
| |
Collapse
|
25
|
Peleg-Raibstein D, Sarker G, Litwan K, Krämer SD, Ametamey SM, Schibli R, Wolfrum C. Enhanced sensitivity to drugs of abuse and palatable foods following maternal overnutrition. Transl Psychiatry 2016; 6:e911. [PMID: 27701408 PMCID: PMC5315546 DOI: 10.1038/tp.2016.176] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 12/13/2022] Open
Abstract
Epidemiological studies have shown an association between maternal overnutrition and increased risk of the progeny for the development of obesity as well as psychiatric disorders. Animal studies have shown results regarding maternal high-fat diet (HFD) and a greater risk of the offspring to develop obesity. However, it still remains unknown whether maternal HFD can program the central reward system in such a way that it will imprint long-term changes that will predispose the offspring to addictive-like behaviors that may lead to obesity. We exposed female dams to either laboratory chow or HFD for a period of 9 weeks: 3 weeks before conception, during gestation and lactation. Offspring born to either control or HFD-exposed dams were examined in behavioral, neurochemical, neuroanatomical, metabolic and positron emission tomography (PET) scan tests. Our results demonstrate that HFD offspring compared with controls consume more alcohol, exhibit increased sensitivity to amphetamine and show greater conditioned place preference to cocaine. In addition, maternal HFD leads to increased preference to sucrose as well as to HFD while leaving the general feeding behavior intact. The hedonic behavioral alterations are accompanied by reduction of striatal dopamine and by increased dopamine 2 receptors in the same brain region as evaluated by post-mortem neurochemical, immunohistochemical as well as PET analyses. Taken together, our data suggest that maternal overnutrition predisposes the offspring to develop hedonic-like behaviors to both drugs of abuse as well as palatable foods and that these types of behaviors may share common neuronal underlying mechanisms that can lead to obesity.
Collapse
Affiliation(s)
- D Peleg-Raibstein
- Department of Health Science and Technology, Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland,Department of Health Science and Technology, Laboratory of Translational Nutrition Biology, ETH Zurich, Schorenstrasse 16, Schwerzenbach 8603 Switzerland. E-mail:
| | - G Sarker
- Department of Health Science and Technology, Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - K Litwan
- Department of Health Science and Technology, Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - S D Krämer
- Center for Radiopharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - S M Ametamey
- Center for Radiopharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - R Schibli
- Center for Radiopharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - C Wolfrum
- Department of Health Science and Technology, Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| |
Collapse
|
26
|
Lakehayli S, Said N, El Khachibi M, El Ouahli M, Nadifi S, Hakkou F, Tazi A. Prenatal stress alters diazepam withdrawal syndrome and 5HT1A receptor expression in the raphe nuclei of adult rats. Neuroscience 2016; 330:50-6. [DOI: 10.1016/j.neuroscience.2016.05.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 05/17/2016] [Accepted: 05/17/2016] [Indexed: 10/21/2022]
|
27
|
Long-term consequences of prenatal stress and neurotoxicants exposure on neurodevelopment. Prog Neurobiol 2016; 155:21-35. [PMID: 27236051 DOI: 10.1016/j.pneurobio.2016.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 02/19/2016] [Accepted: 05/24/2016] [Indexed: 11/20/2022]
Abstract
There is a large consensus that the prenatal environment determines the susceptibility to pathological conditions later in life. The hypothesis most widely accepted is that exposure to insults inducing adverse conditions in-utero may have negative effects on the development of target organs, disrupting homeostasis and increasing the risk of diseases at adulthood. Several models have been proposed to investigate the fetal origins of adult diseases, but although these approaches hold true for almost all diseases, particular attention has been focused on disorders related to the central nervous system, since the brain is particularly sensitive to alterations of the microenvironment during early development. Neurobiological disorders can be broadly divided into developmental, neurodegenerative and neuropsychiatric disorders. Even though most of these diseases share genetic risk factors, the onset of the disorders cannot be explained solely by inheritance. Therefore, current understanding presumes that the interactions of environmental input, may lead to different disorders. Among the insults that can play a direct or indirect role in the development of neurobiological disorders are stress, infections, drug abuse, and environmental contaminants. Our laboratories have been involved in the study of the neurobiological impact of gestational stress on the offspring (Dr. Antonelli's lab) and on the effect of gestational exposure to toxicants, mainly methyl mercury (MeHg) and perfluorinated compounds (PFCs) (Dr. Ceccatelli's lab). In this focused review, we will review the specialized literature but we will concentrate mostly on our own work on the long term neurodevelopmental consequences of gestational exposure to stress and neurotoxicants.
Collapse
|
28
|
Neuroscience of resilience and vulnerability for addiction medicine. PROGRESS IN BRAIN RESEARCH 2016; 223:3-18. [DOI: 10.1016/bs.pbr.2015.09.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Social interaction reward decreases p38 activation in the nucleus accumbens shell of rats. Neuropharmacology 2015; 99:510-6. [PMID: 26300300 PMCID: PMC5056637 DOI: 10.1016/j.neuropharm.2015.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 01/23/2023]
Abstract
We have previously shown that animals acquired robust conditioned place preference (CPP) to either social interaction alone or cocaine alone. Recently it has been reported that drugs of abuse abnormally activated p38, a member of mitogen-activated protein kinase family, in the nucleus accumbens. In this study, we aimed to investigate the expression of the activated form of p38 (pp38) in the nucleus accumbens shell and core of rats expressing either cocaine CPP or social interaction CPP 1 h, 2 h and 24 h after the CPP test. We hypothesized that cocaine CPP will increase pp38 in the nucleus accumbens shell/core as compared to social interaction CPP. Surprisingly, we found that 24 h after social interaction CPP, pp38 neuronal levels were decreased in the nucleus accumbens shell to the level of naïve rats. Control saline rats that received saline in both compartments of the CPP apparatus and cocaine CPP rats showed similar enhanced p38 activation as compared to naïve and social interaction CPP rats. We also found that the percentage of neurons expressing dopaminergic receptor D2R and pp38 was also decreased in the shell of the nucleus accumbens of social interaction CPP rats as compared to controls. Given the emerging role of p38 in stress/anxiety behaviors, these results suggest that (1) social interaction reward has anti-stress effects; (2) cocaine conditioning per se does not affect p38 activation and that (3) marginal stress is sufficient to induce p38 activation in the shell of the nucleus accumbens.
Collapse
|
30
|
Said N, Lakehayli S, El Khachibi M, El Ouahli M, Nadifi S, Hakkou F, Tazi A. Prenatal stress induces vulnerability to nicotine addiction and alters D2 receptors' expression in the nucleus accumbens in adult rats. Neuroscience 2015; 304:279-85. [PMID: 26192093 DOI: 10.1016/j.neuroscience.2015.07.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/22/2015] [Accepted: 07/10/2015] [Indexed: 01/15/2023]
Abstract
Prenatal stress (PS) can induce several long-lasting behavioral and molecular abnormalities in rats. It can also be considered as a risk factor for many psychiatric diseases like schizophrenia, depression or PTSD and predispose to addiction. In this study, we investigated the effect of prenatal stress on the reinforcing properties of nicotine in the CPP paradigm. Then, we examined the mRNA expression of the D2 dopaminergic receptors using the quantitative real-time PCR technique in the nucleus accumbens (NAcc). We found that prenatally stressed rats exhibited a greater place preference for the nicotine-paired compartment than the control rats. Moreover, we observed an overexpression of the DRD2 gene in adult offspring stressed in utero and a downregulation in the PS NIC group (PS rats treated with nicotine) compared with their control counterparts (C NIC). These data suggest that maternal stress can permanently alter the offspring's addictive behavior and D2 receptors' expression.
Collapse
Affiliation(s)
- N Said
- Laboratory of Pharmacology, Faculty of Medicine and Pharmacy of Casablanca, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco.
| | - S Lakehayli
- Laboratory of Pharmacology, Faculty of Medicine and Pharmacy of Casablanca, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco
| | - M El Khachibi
- Genetics and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy of Casablanca, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco
| | - M El Ouahli
- Sultan Moulay Slimane University, Faculty of Sciences & Techniques Beni-Mellal, Life Sciences, Morocco
| | - S Nadifi
- Genetics and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy of Casablanca, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco
| | - F Hakkou
- Laboratory of Pharmacology, Faculty of Medicine and Pharmacy of Casablanca, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco
| | - A Tazi
- Laboratory of Pharmacology, Faculty of Medicine and Pharmacy of Casablanca, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco
| |
Collapse
|
31
|
Lakehayli S, Said N, Khachibi ME, Ouahli ME, Nadifi S, Hakkou F, Tazi A. Long-term effects of prenatal stress and diazepam on D2 receptor expression in the nucleus accumbens of adult rats. Neurosci Lett 2015; 594:133-6. [DOI: 10.1016/j.neulet.2015.03.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 01/11/2023]
|
32
|
Lakehayli S, Said N, Battas O, Hakkou F, Tazi A. Prenatal stress alters sensitivity to benzodiazepines in adult rats. Neurosci Lett 2015; 591:187-191. [DOI: 10.1016/j.neulet.2015.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
|
33
|
Biezonski DK, Trifilieff P, Meszaros J, Javitch JA, Kellendonk C. Evidence for limited D1 and D2 receptor coexpression and colocalization within the dorsal striatum of the neonatal mouse. J Comp Neurol 2015; 523:1175-89. [PMID: 25556545 DOI: 10.1002/cne.23730] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 01/09/2023]
Abstract
The striatum is the major input nucleus of the basal ganglia involved in reward processing, goal-directed behaviors, habit learning, and motor control. The striatum projects to the basal ganglia output nuclei via the "direct" and "indirect" pathways, which can be distinguished by their projection fields and their opposing effects on behavior. In adult animals, the functional opposition is modulated by the differential actions of D1 and D2 dopamine receptors (D1R, D2R), the expression of which is largely separated between these pathways. To determine whether a similar degree of separation exists earlier in development, we used dual-label immunohistochemistry to map dorsal-striatal D1R and D2R expression at the promoter level in postnatal day 0 (PD0) Drd1a-tdTomato/Drd2-GFP BAC transgenic mice, and at the receptor level by costaining for native D1R and D2R in wildtype (WT) PD0 animals. To assess for potential molecular interactions between D1R and D2R we also employed a recently developed proximity-ligation assay (PLA). Limited coexpression and colocalization of the D1R and D2R proteins was found in clusters of neurons endemic to the "patch" compartment as identified by costaining with tyrosine hydroxylase, but not outside these clusters. Moreover, in contrast to our recent findings where we failed to detect a D1R-D2R PLA signal in the adult striatum, in PD0 striatum we did identify a clear PLA signal for this pair of receptors. This colocalization at close proximity points to a possible role for D1R/D2R-mediated crosstalk in early striatal ontogeny.
Collapse
Affiliation(s)
- Dominik K Biezonski
- Department of Psychiatry, Division of Molecular Therapeutics, Columbia University, New York State Psychiatric Institute, New York, New York, 10032, USA
| | | | | | | | | |
Collapse
|
34
|
Reynaert ML, Marrocco J, Gatta E, Mairesse J, Van Camp G, Fagioli F, Maccari S, Nicoletti F, Morley-Fletcher S. A Self-Medication Hypothesis for Increased Vulnerability to Drug Abuse in Prenatally Restraint Stressed Rats. PERINATAL PROGRAMMING OF NEURODEVELOPMENT 2015; 10:101-20. [DOI: 10.1007/978-1-4939-1372-5_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
35
|
Prenatal stress and adult drug-seeking behavior: interactions with genes and relation to nondrug-related behavior. ADVANCES IN NEUROBIOLOGY 2015; 10:75-100. [PMID: 25287537 DOI: 10.1007/978-1-4939-1372-5_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Addiction inflicts large personal, social, and economic burdens, yet its etiology is poorly defined and effective treatments are lacking. As with other neuropsychiatric disorders, addiction is characterized by a core set of symptoms and behaviors that are believed to be influenced by complex gene-environment interactions. Our group focuses on the interaction between early stress and genetic background in determining addiction vulnerability. Prior work by our group and others has indicated that a history of prenatal stress (PNS) in rodents elevates adult drug seeking in a number of behavioral paradigms. The focus of the present chapter is to summarize work in the area of PNS and addiction models as well as our recent studies of PNS on drug seeking in different strains of mice as a strategy to dissect gene-environment interactions underlying cocaine addiction vulnerability. These studies indicate that ability of PNS to elevate adult cocaine seeking is strain dependent. Further, PNS also alters other nondrug behaviors in a fashion that is dependent on different strains and independent from the strain dependence of drug seeking. Thus, it appears that the ability of PNS to alter behavior related to different psychiatric conditions is orthogonal, with similar nonspecific susceptibility to prenatal stress across genetic backgrounds but with the genetic background determining the specific nature of the PNS effects. Finally, the advent of recombinant inbred mouse strains is allowing us to determine the genetic bases of these gene-environment interactions. Understanding these effects will have broad implications to determining the nature of vulnerability to addiction and perhaps other disorders.
Collapse
|
36
|
Pallarés ME, Antonelli MC. Hormonal modulation of catecholaminergic neurotransmission in a prenatal stress model. ADVANCES IN NEUROBIOLOGY 2015; 10:45-59. [PMID: 25287535 DOI: 10.1007/978-1-4939-1372-5_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Our laboratory has a long-standing interest in the effects of prenatal stress (PS) on various neurotransmitter pathways and the morphology of the developing brain as well as in behavioural aspects of the offspring. Employing a commonly used PS paradigm in which the dams were subjected to restraint stress during the last week of gestation, we observed that several of these pathways were altered in the offspring brain. In this chapter, we will summarize and discuss the results obtained with the main catecholaminergic pathways, namely dopamine (DA) and norepinephrine (NE). In our hands, PS produces an increase in dopamine D2-type receptors in limbic areas, a decreased DA release after amphetamine stimulation in prefrontal cortex (PFC) and an increase in NE release in the same area of the adult offspring brain. In addition, DA uptake is altered at prepubertal stages that persist through adulthood. However, the expression of the step-limiting enzyme of the DA synthesis, tyrosine hydroxylase (TH), is only impaired at early stages of development after PS in the neuronal bodies. At the nuclear regulation level, dopaminergic transcription factors Nurr1 and Ptx3 showed a high vulnerability to PS showing changes along the lifespan. It was striking to observe that many impairments observed in most of these pathways differed depending on whether they were tested before or after puberty indicating a particular sensitivity of the systems to variations in gonadal hormones peaks. In fact, we observed that PS induced long-term effects on the male offspring reproductive system and spermatogenesis development, particularly by inducing a long-term imbalance of circulating sexual hormone levels. Our findings suggest that PS exerts long-term effects on various neurotransmitter pathways altering the normal connectivity between brain areas. Since the developing forebrain was shown to be influenced by androgen exposure, and PS was shown to disrupt prenatal testosterone surges, our results suggest that prenatal insults might be affecting the organizational role of androgens during brain development and differentially modulating their activational role during pubertal brain maturation.
Collapse
Affiliation(s)
- María Eugenia Pallarés
- Instituto de Biología Celular y Neurociencias, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
37
|
Gillies G, Virdee K, McArthur S, Dalley J. Sex-dependent diversity in ventral tegmental dopaminergic neurons and developmental programing: A molecular, cellular and behavioral analysis. Neuroscience 2014; 282:69-85. [PMID: 24943715 PMCID: PMC4245713 DOI: 10.1016/j.neuroscience.2014.05.033] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/12/2014] [Accepted: 05/18/2014] [Indexed: 02/02/2023]
Abstract
The knowledge that diverse populations of dopaminergic neurons within the ventral tegmental area (VTA) can be distinguished in terms of their molecular, electrophysiological and functional properties, as well as their differential projections to cortical and subcortical regions has significance for key brain functions, such as the regulation of motivation, working memory and sensorimotor control. Almost without exception, this understanding has evolved from landmark studies performed in the male sex. However, converging evidence from both clinical and pre-clinical studies illustrates that the structure and functioning of the VTA dopaminergic systems are intrinsically different in males and females. This may be driven by sex differences in the hormonal environment during adulthood ('activational' effects) and development (perinatal and/or pubertal 'organizational' effects), as well as genetic factors, especially the SRY gene on the Y chromosome in males, which is expressed in a sub-population of adult midbrain dopaminergic neurons. Stress and stress hormones, especially glucocorticoids, are important factors which interact with the VTA dopaminergic systems in order to achieve behavioral adaptation and enable the individual to cope with environmental change. Here, also, there is male/female diversity not only during adulthood, but also in early life when neurobiological programing by stress or glucocorticoid exposure differentially impacts dopaminergic developmental trajectories in male and female brains. This may have enduring consequences for individual resilience or susceptibility to pathophysiological change induced by stressors in later life, with potential translational significance for sex bias commonly found in disorders involving dysfunction of the mesocorticolimbic dopaminergic systems. These findings highlight the urgent need for a better understanding of the sexual dimorphism in the VTA if we are to improve strategies for the prevention and treatment of debilitating conditions which differentially affect men and women in their prevalence and nature, including schizophrenia, attention/deficit hyperactivity disorder, autism spectrum disorders, anxiety, depression and addiction.
Collapse
Affiliation(s)
- G.E. Gillies
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK,Corresponding author. Address: Division of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK. Tel: +44-(0)-20-7594-7050.
| | - K. Virdee
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK
| | - S. McArthur
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London EC1 6BQ, UK
| | - J.W. Dalley
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychiatry, University of Cambridge, Addenbrooke’s Hospital, Hill’s Road, Cambridge CB2 2QQ, UK
| |
Collapse
|
38
|
Babenko O, Kovalchuk I, Metz GAS. Stress-induced perinatal and transgenerational epigenetic programming of brain development and mental health. Neurosci Biobehav Rev 2014; 48:70-91. [PMID: 25464029 DOI: 10.1016/j.neubiorev.2014.11.013] [Citation(s) in RCA: 331] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/19/2014] [Accepted: 11/17/2014] [Indexed: 12/20/2022]
Abstract
Research efforts during the past decades have provided intriguing evidence suggesting that stressful experiences during pregnancy exert long-term consequences on the future mental wellbeing of both the mother and her baby. Recent human epidemiological and animal studies indicate that stressful experiences in utero or during early life may increase the risk of neurological and psychiatric disorders, arguably via altered epigenetic regulation. Epigenetic mechanisms, such as miRNA expression, DNA methylation, and histone modifications are prone to changes in response to stressful experiences and hostile environmental factors. Altered epigenetic regulation may potentially influence fetal endocrine programming and brain development across several generations. Only recently, however, more attention has been paid to possible transgenerational effects of stress. In this review we discuss the evidence of transgenerational epigenetic inheritance of stress exposure in human studies and animal models. We highlight the complex interplay between prenatal stress exposure, associated changes in miRNA expression and DNA methylation in placenta and brain and possible links to greater risks of schizophrenia, attention deficit hyperactivity disorder, autism, anxiety- or depression-related disorders later in life. Based on existing evidence, we propose that prenatal stress, through the generation of epigenetic alterations, becomes one of the most powerful influences on mental health in later life. The consideration of ancestral and prenatal stress effects on lifetime health trajectories is critical for improving strategies that support healthy development and successful aging.
Collapse
Affiliation(s)
- Olena Babenko
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada T1K 3M4; Department of Biological Sciences, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada T1K 3M4
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada T1K 3M4
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada T1K 3M4
| |
Collapse
|
39
|
Boersma GJ, Moghadam AA, Cordner ZA, Tamashiro KL. Prenatal stress and stress coping style interact to predict metabolic risk in male rats. Endocrinology 2014; 155:1302-12. [PMID: 24467745 PMCID: PMC3959611 DOI: 10.1210/en.2013-1874] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Both prenatal stress (PNS) exposure and a passive stress-coping style have been identified as risk factors for insulin resistance in rats. In the current study, we test the hypothesis that PNS and stress-coping style may interact in predicting susceptibility for metabolic disease. To test this hypothesis, adult male control and PNS offspring were behaviorally characterized using a defensive burying test to have either a passive or proactive stress-coping style. In adulthood, all rats were fed either a standard chow or a high-fat diet for 3 weeks. After 3 weeks of diet exposure, glucose and insulin levels were assessed during an oral glucose tolerance test. Under high-fat diet conditions, PNS rats display elevated glucose and insulin responses to the oral glucose tolerance test, indicative of glucose intolerance. Interestingly, these effects of PNS were far more pronounced in rats characterized by a passive stress-coping style. Additionally, the passively coping PNS rats also gained more weight on the high-fat diet than all other rats tested. This observation suggests that a stressful prenatal environment in combination with a passive stress-coping strategy may prime an individual to be sensitive to diet-induced obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Gretha J Boersma
- Department of Psychiatry and Behavioral Sciences (G.J.B., A.A.M., Z.A.C., K.L.T.), and The Mood Disorders Center (G.J.B., Z.A.C., K.L.T.), Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205
| | | | | | | |
Collapse
|
40
|
Sex-dependent and non-monotonic enhancement and unmasking of methylmercury neurotoxicity by prenatal stress. Neurotoxicology 2014; 41:123-40. [PMID: 24502960 DOI: 10.1016/j.neuro.2014.01.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/20/2014] [Accepted: 01/28/2014] [Indexed: 11/20/2022]
Abstract
Methylmercury (MeHg) and prenatal stress (PS) are risk factors for neurotoxicity that may co-occur in human populations. Because they also share biological substrates and can produce common behavioral deficits, this study examined their joint effects on behavioral and neurochemical effects in male and female rats. Dams had access to 0, 0.5 or 2.5ppm MeHg chloride drinking water from two to three weeks prior to breeding through weaning. Half of the dams in each of these treatment groups also underwent PS on gestational days 16-17. This yielded 6 groups/gender: 0-NS, 0-PS, 0.5-NS, 0.5-PS, 2.5-NS, and 2.5-PS. Behavioral testing began in young adulthood and included fixed interval (FI) schedule-controlled behavior, novel object recognition (NOR) and locomotor activity, behaviors previously demonstrated to be sensitive to MeHg and/or mediated by brain mesocorticolimbic dopamine glutamate systems targeted by both MeHg and PS. Behavioral deficits were more pronounced in females and included impaired NOR recognition memory only under conditions of combined MeHg and PS, while non-monotonic reductions in FI response rates occurred, with greatest effects at the 0.5ppm concentration; the less reduced 2.5ppm FI response rates were further reduced under conditions of PS (2.5-PS). Correspondingly, many neurochemical changes produced by MeHg were only seen under conditions of PS, particularly in striatum in males and in hippocampus and nucleus accumbens in females, regions of significance to the mediation of FI and NOR performance. Collectively these findings demonstrate sex-dependent and non-monotonic effects of developmental MeHg exposure that can be unmasked or enhanced by PS, particularly for behavioral outcomes in females, but for both sexes in neurochemical changes, that were observed at MeHg exposure concentrations that did not influence either reproductive outcomes or maternal behavior. Thus, assessment of risks associated with MeHg may be underestimated in the absence of other extant risk factors with which it may share common substrates and effects.
Collapse
|
41
|
Stroud LR, Papandonatos G, Shenassa E, Rodriguez D, Niaura R, LeWinn K, Lipsitt LP, Buka SL. Prenatal glucocorticoids and maternal smoking during pregnancy independently program adult nicotine dependence in daughters: a 40-year prospective study. Biol Psychiatry 2014; 75:47-55. [PMID: 24034414 PMCID: PMC3858529 DOI: 10.1016/j.biopsych.2013.07.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 06/25/2013] [Accepted: 07/12/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Maternal smoking during pregnancy (MSDP) is an independent risk factor for offspring nicotine dependence (ND), but mechanisms remain unknown. We investigated prenatal glucocorticoid (cortisol) and androgen (testosterone) associations with offspring ND over 40 years and the possibility that prenatal glucocorticoids and androgens would mediate links between MSDP and offspring ND. METHODS Participants were 1086 mother-adult offspring pairs (59% female) from the New England Family Study, a 40-year longitudinal follow-up of the Collaborative Perinatal Project. MSDP was assessed prospectively at each prenatal visit. Maternal cortisol, testosterone, and cotinine (nicotine metabolite) were assayed from third trimester maternal sera. Offspring lifetime ND was assessed via structured interview. RESULTS Significant bivariate associations emerged for: 1) MSDP/cotinine and lifetime ND; and 2) maternal cortisol and lifetime ND, for daughters only. In multivariate models, maternal cortisol and MSDP/cotinine remained significantly and independently associated with increased odds of lifetime ND of daughters. However, cortisol did not mediate the MSDP-lifetime ND relation. No associations emerged between maternal testosterone and offspring ND. CONCLUSIONS Results provide the first evidence in support of prenatal glucocorticoid programming of adult ND over 40 years in daughters only. Our study highlights two independent prenatal pathways leading to increased risk for ND in daughters: elevated prenatal glucocorticoids and MSDP/nicotine exposure. Daughter-specific effects of glucocorticoid and MSDP programming over 40 years highlight the breadth and persistence of sexually dimorphic programming effects in humans. Results do not support androgen programming of offspring ND.
Collapse
Affiliation(s)
- Laura R. Stroud
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University
| | | | - Edmond Shenassa
- Program in Maternal-Child Health, School of Public Health, University of Maryland
| | - Daniel Rodriguez
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University
| | - Raymond Niaura
- Schroeder Institute for Tobacco Research and Policy Studies, American Legacy Foundation
| | - Kaja LeWinn
- Department of Psychiatry, University of California, San Francisco
| | | | - Stephen L. Buka
- Department of Epidemiology, School of Public Health, Brown University
| |
Collapse
|
42
|
Initial d2 dopamine receptor sensitivity predicts cocaine sensitivity and reward in rats. PLoS One 2013; 8:e78258. [PMID: 24223783 PMCID: PMC3817276 DOI: 10.1371/journal.pone.0078258] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 09/10/2013] [Indexed: 11/24/2022] Open
Abstract
The activation of dopamine receptors within the mesolimbic dopamine system is known to be involved in the initiation and maintenance of cocaine use. Expression of the D2 dopamine receptor subtype has been implicated as both a predisposing factor and consequence of chronic cocaine use. It is unclear whether there is a predictive relationship between D2 dopamine receptor function and cocaine sensitivity that would enable cocaine abuse. Therefore, we exploited individual differences in behavioral responses to D2 dopamine receptor stimulation to test its relationship with cocaine-mediated behaviors. Outbred, male Sprague-Dawley rats were initially characterized by their locomotor responsiveness to the D2 dopamine receptor agonist, quinpirole, in a within-session ascending dose-response regimen (0, 0.1, 0.3 & 1.0 mg/kg, sc). Rats were classified as high or low quinpirole responders (HD2 and LD2, respectively) by a median split of their quinpirole-induced locomotor activity. Rats were subsequently tested for differences in the psychostimulant effects of cocaine by measuring changes in cocaine-induced locomotor activity (5 and 15 mg/kg, ip). Rats were also tested for differences in the development of conditioned place preference to a low dose of cocaine (7.5 mg/kg, ip) that does not reliably produce a cocaine conditioned place preference. Finally, rats were tested for acquisition of cocaine self-administration and maintenance responding on fixed ratio 1 and 5 schedules of reinforcement, respectively. Results demonstrate that HD2 rats have enhanced sensitivity to the locomotor stimulating properties of cocaine, display greater cocaine conditioned place preference, and self-administer more cocaine compared to LD2 animals. These findings suggest that individual differences in D2 dopamine receptor sensitivity may be predictive of cocaine sensitivity and reward.
Collapse
|
43
|
Converse AK, Moore CF, Moirano JM, Ahlers EO, Larson JA, Engle JW, Barnhart TE, Murali D, Christian BT, DeJesus OT, Holden JE, Nickles RJ, Schneider ML. Prenatal stress induces increased striatal dopamine transporter binding in adult nonhuman primates. Biol Psychiatry 2013; 74:502-10. [PMID: 23726316 PMCID: PMC3775901 DOI: 10.1016/j.biopsych.2013.04.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 04/22/2013] [Accepted: 04/25/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND To determine the effects in adult offspring of maternal exposure to stress and alcohol during pregnancy, we imaged striatal and midbrain dopamine transporter (DAT) binding by positron emission tomography in rhesus monkeys (Macaca mulatta). We also evaluated the relationship between DAT binding and behavioral responses previously found to relate to dopamine D2 receptor density (responsivity to tactile stimuli, performance on a learning task, and behavior during a learning task). METHODS Subjects were adult offspring derived from a 2 × 2 experiment in which pregnant monkeys were randomly assigned to control, daily mild stress exposure (acoustic startle), voluntary consumption of moderate-level alcohol, or both daily stress and alcohol. Adult offspring (n = 38) were imaged by positron emission tomography with the DAT ligand [(18)F]2β-carbomethoxy-3β-(4-chlorophenyl)-8-(2-fluoroethyl)-nortropane ([(18)F]FECNT). RESULTS Results showed that prenatal stress yielded an overall increase of 15% in [(18)F]FECNT binding in the striatum (p = .016), 17% greater binding in the putamen (p = .012), and 13% greater binding in the head of the caudate (p = .028) relative to animals not exposed to prenatal stress. Striatal [(18)F]FECNT binding correlated negatively with habituation to repeated tactile stimulation and positively with tactile responsivity. There were no significant effects of prenatal alcohol exposure on [(18)F]FECNT binding. CONCLUSIONS Maternal exposure to mild daily stress during pregnancy yielded increases in striatal DAT availability that were apparent in adult offspring and were associated with behavioral characteristics reflecting tactile hyperresponsivity, a condition associated with problem behaviors in children.
Collapse
Affiliation(s)
- Alexander K. Converse
- Waisman Center, University of Wisconsin-Madison,Corresponding author: , tel. 001/608/265.6604, T123 Waisman Center, University of Wisconsin-Madison, Madison, WI 53704
| | | | | | | | - Julie A. Larson
- Harlow Center for Biological Psychology, University of Wisconsin-Madison,Department of Kinesiology, University of Wisconsin-Madison
| | | | | | | | | | | | - James E. Holden
- Department of Medical Physics, University of Wisconsin-Madison
| | | | - Mary L. Schneider
- Harlow Center for Biological Psychology, University of Wisconsin-Madison,Department of Psychology, University of Wisconsin-Madison,Department of Kinesiology, University of Wisconsin-Madison
| |
Collapse
|
44
|
Pallarés ME, Baier CJ, Adrover E, Monteleone MC, Brocco MA, Antonelli MC. Age-Dependent Effects of Prenatal Stress on the Corticolimbic Dopaminergic System Development in the Rat Male Offspring. Neurochem Res 2013; 38:2323-35. [DOI: 10.1007/s11064-013-1143-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 08/23/2013] [Accepted: 08/26/2013] [Indexed: 12/15/2022]
|
45
|
Marrocco J, Mairesse J, Bucci D, Lionetto L, Battaglia G, Consolazione M, Ravasi L, Simmaco M, Morley-Fletcher S, Maccari S, Nicoletti F. Early life stress causes refractoriness to haloperidol-induced catalepsy. Mol Pharmacol 2013; 84:244-51. [PMID: 23716620 DOI: 10.1124/mol.113.085530] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The use of classic antipsychotic drugs is limited by the occurrence of extrapyramidal motor symptoms, which are caused by dopamine (DA) receptor blockade in the neostriatum. We examined the impact of early-life stress on haloperidol-induced catalepsy using the rat model of prenatal restraint stress (PRS). Adult "PRS rats," i.e., the offspring of mothers exposed to restraint stress during pregnancy, were resistant to catalepsy induced by haloperidol (0.5-5 mg/kg i.p.) or raclopride (2 mg/kg s.c.). Resistance to catalepsy in PRS rats did not depend on reductions in blood or striatal levels, as compared with unstressed control rats. PRS rats also showed a greater behavioral response to the DA receptor agonist, apomorphine, suggesting that PRS causes enduring neuroplastic changes in the basal ganglia motor circuit. To examine the activity of this circuit, we performed a stereological counting of c-Fos(+) neurons in the external and internal globus pallidus, subthalamic nucleus, and ventral motor thalamic nuclei. Remarkably, the number of c-Fos(+) neurons in ventral motor thalamic nuclei was higher in PRS rats than in unstressed controls, both under basal conditions and in response to single or repeated injections with haloperidol. Ventral motor thalamic nuclei contain exclusively excitatory projection neurons that convey the basal ganglia motor programming to the cerebral cortex. Hence, an increased activity of ventral motor thalamic nuclei nicely explains the refractoriness of PRS rats to haloperidol-induced catalepsy. Our data raise the interesting possibility that early-life stress is protective against extrapyramidal motor effects of antipsychotic drugs in the adult life.
Collapse
|
46
|
Macrì S, Ceci C, Canese R, Laviola G. Prenatal stress and peripubertal stimulation of the endocannabinoid system differentially regulate emotional responses and brain metabolism in mice. PLoS One 2012; 7:e41821. [PMID: 22848620 PMCID: PMC3405010 DOI: 10.1371/journal.pone.0041821] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/26/2012] [Indexed: 12/22/2022] Open
Abstract
The central endocannabinoid system (ECS) and the hypothalamic-pituitary-adrenal-axis mediate individual responses to emotionally salient stimuli. Their altered developmental adjustment may relate to the emergence of emotional disturbances. Although environmental influences regulate the individual phenotype throughout the entire lifespan, their effects may result particularly persistent during plastic developmental stages (e.g. prenatal life and adolescence). Here, we investigated whether prenatal stress – in the form of gestational exposure to corticosterone supplemented in the maternal drinking water (100 mg/l) during the last week of pregnancy – combined with a pharmacological stimulation of the ECS during adolescence (daily fatty acid amide hydrolase URB597 i.p. administration - 0.4 mg/kg - between postnatal days 29–38), influenced adult mouse emotional behaviour and brain metabolism measured through in vivo quantitative magnetic resonance spectroscopy. Compared to control mice, URB597-treated subjects showed, in the short-term, reduced locomotion and, in the long term, reduced motivation to execute operant responses to obtain palatable rewards paralleled by reduced levels of inositol and taurine in the prefrontal cortex. Adult mice exposed to prenatal corticosterone showed increased behavioural anxiety and reduced locomotion in the elevated zero maze, and altered brain metabolism (increased glutamate and reduced taurine in the hippocampus; reduced inositol and N-Acetyl-Aspartate in the hypothalamus). Present data further corroborate the view that prenatal stress and pharmacological ECS stimulation during adolescence persistently regulate emotional responses in adulthood. Yet, whilst we hypothesized these factors to be interactive in nature, we observed that the consequences of prenatal corticosterone administration were independent from those of ECS drug-induced stimulation during adolescence.
Collapse
Affiliation(s)
- Simone Macrì
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Roma, Italy.
| | | | | | | |
Collapse
|
47
|
Cory-Slechta DA, Virgolini MB, Liu S, Weston D. Enhanced stimulus sequence-dependent repeated learning in male offspring after prenatal stress alone or in conjunction with lead exposure. Neurotoxicology 2012; 33:1188-202. [PMID: 22796262 DOI: 10.1016/j.neuro.2012.06.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/29/2012] [Accepted: 06/29/2012] [Indexed: 10/28/2022]
Abstract
Both lead (Pb) exposure and prenatal stress (PS) can produce cognitive deficits, and in a prior study we demonstrated enhanced cognitive deficits in repeated learning of female rats exposed to both of these developmental insults (Cory-Slechta et al., 2010). However, PS can also lead to improved cognitive outcomes that are both gender- and context-dependent. Thus, the current study examined whether Pb ± PS likewise produced repeated learning deficits in males, either after maternal or lifetime Pb exposure. Repeated learning was evaluated using a multiple schedule of repeated learning and performance that required learning 3-response sequences in male offspring that had been subjected to either maternal Pb (0 or 150 ppm) or lifetime Pb exposure (0 or 50 ppm) beginning two months prior to dam breeding, to prenatal immobilization restraint stress (gestational days 16-17), or to both Pb and PS. Blood Pb, corticosterone, hippocampal glucocorticoid receptor density and brain monoamines were also measured. In contrast to outcomes in females, sequence-specific enhancements of repeated learning accuracy were produced by PS, particularly when combined with Pb, results that appeared to be more robust in combination with lifetime than maternal Pb exposure. A common behavioral mechanism of these improvements appears to be an increased reinforcement density associated with increased response rates and shorter session times seen with PS ± Pb that could shorten time to reinforcement. Trends toward lower levels of nucleus accumbens dopamine activity seen after both maternal Pb and lifetime Pb combined with PS suggest a possible role for this region/neurotransmitter in enhanced accuracy, whereas PS ± Pb-induced corticosterone changes did not exhibit an obvious systematic relationship to accuracy enhancements. While PS ± Pb-based increases in accuracy appear to be an improved outcome, the benefits of increased response rate are by no means universal, but highly context-dependent and can lead to adverse behavioral effects in other conditions.
Collapse
Affiliation(s)
- D A Cory-Slechta
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | | | | | | |
Collapse
|
48
|
Baier CJ, Katunar MR, Adrover E, Pallarés ME, Antonelli MC. Gestational restraint stress and the developing dopaminergic system: an overview. Neurotox Res 2012; 22:16-32. [PMID: 22215534 DOI: 10.1007/s12640-011-9305-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 12/15/2011] [Accepted: 12/20/2011] [Indexed: 12/22/2022]
Abstract
Prenatal stress exerts a strong impact on fetal brain development in rats impairing adaptation to stressful conditions, subsequent vulnerability to anxiety, altered sexual function, and enhanced propensity to self-administer drugs. Most of these alterations have been attributed to changes in the neurotransmitter dopamine (DA). In humans; dysfunction of dopaminergic system is associated with development of several neurological disorders, such as Parkinson disease, schizophrenia, attention-deficit hyperactivity disorder, and depression. Evidences provided by animal research, as well as retrospective studies in humans, pointed out that exposure to adverse events in early life can alter adult behaviors and neurochemical indicators of midbrain DA activity, suggesting that the development of the DA system is sensitive to disruption by exposure to early stressors. The purpose of this article is to provide a general overview of published studies and our own study related to the effect of prenatal insults on the development of DA metabolism and biology, focusing mainly in articles involving prenatal-restraint stress protocols in rats. We will also attempt to make a correlation between theses alterations and DA-related pathological processes in humans.
Collapse
Affiliation(s)
- Carlos J Baier
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
49
|
Abstract
Basic research in animals represents a fruitful approach to study the neurobiological basis of brain and behavioral disturbances relevant to neuropsychiatric disease and to establish and evaluate novel pharmacological therapies for their treatment. In the context of schizophrenia, there are models employing specific experimental manipulations developed according to specific pathophysiological or etiological hypotheses. The use of selective lesions in adult animals and the acute administration of psychotomimetic agents are indispensable tools in the elucidation of the contribution of specific brain regions or neurotransmitters to the genesis of a specific symptom or collection of symptoms and enjoy some degrees of predictive validity. However, they may be inaccurate, if not inadequate, in capturing the etiological mechanisms or ontology of the disease needed for a complete understanding of the disease and may be limited in the discovery of novel compounds for the treatment of negative and cognitive symptoms of schizophrenia. Under the prevailing consensus of schizophrenia as a disease of neurodevelopmental origin, we have seen the establishment of neurodevelopmental animal models which aim to identify the etiological processes whereby the brain, following specific triggering events, develops into a "schizophrenia-like brain" over time. Many neurodevelopmental models such as the neonatal ventral hippocampus (vHPC) lesion, methylazoxymethanol (MAM), and prenatal immune activation models can mimic a broad spectrum of behavioral, cognitive, and pharmacological abnormalities directly implicated in schizophrenic disease. These models allow pharmacological screens against multiple and coexisting schizophrenia-related dysfunctions while incorporating the disease-relevant concept of abnormal brain development. The multiplicity of existing models is testimonial to the multifactorial nature of schizophrenia, and there are ample opportunities for their integration. Indeed, one ultimate goal must be to incorporate the successes of distinct models into one unitary account of the complex disorder of schizophrenia and to use such unitary approaches in the further development and evaluation of novel antipsychotic treatment strategies.
Collapse
|
50
|
Salihagić-Kadić A, Medić M, Kurjak A, Andonotopo W, Azumendi G, Hafner T, Milenković D. Four-dimensional sonography in the assessment of fetal functional neurodevelopment and behavioral patterns. ACTA ACUST UNITED AC 2011. [DOI: 10.3109/14722240500190640] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|