1
|
Martz J, Shelton MA, Langen TJ, Srinivasan S, Seney ML, Kentner AC. Peripubertal antagonism of corticotropin-releasing factor receptor 1 results in sustained changes in behavioral plasticity and the transcriptomic profile of the amygdala. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.14.607957. [PMID: 39185241 PMCID: PMC11343213 DOI: 10.1101/2024.08.14.607957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Peripuberty is a significant period of neurodevelopment with long-lasting effects on the brain and behavior. Blocking type 1 corticotropin-releasing factor receptors (CRFR1) in neonatal and peripubertal rats attenuates detrimental effects of early-life stress on neural plasticity, behavior, and stress hormone action, long after exposure to the drug has ended. CRFR1 antagonism can also impact neural and behavioral development in the absence of stressful stimuli, suggesting sustained alterations under baseline conditions. To investigate this further, we administered the CRFR1 antagonist (CRFR1a) R121919 to young adolescent male and female rats across 4 days. Following each treatment, rats were tested for locomotion, social behavior, mechanical allodynia, or prepulse inhibition (PPI). Acute CRFR1 blockade immediately reduced PPI in peripubertal males, but not females. In adulthood, each assay was repeated without CRFR1a exposure to test for long-term effects of the adolescent treatment. Males continued to experience deficits in PPI while females displayed altered locomotion, PPI, and social behavior. In the adult amygdala, peripubertal CRFR1a induced alterations in pathways related to neural plasticity and stress in males. In females, pathways related to central nervous system myelination, cell junction organization, and glutamatergic regulation of synaptic transmission were affected. Understanding how acute exposure to neuropharmacological agents can have sustained impacts on brain and behavior, in the absence of further exposures, has important clinical implications for developing adolescents.
Collapse
|
2
|
Corley C, Craig A, Sadek S, Marusich JA, Chehimi SN, White AM, Holdiness LJ, Reiner BC, Gipson CD. Enhancing translation: A need to leverage complex preclinical models of addictive drugs to accelerate substance use treatment options. Pharmacol Biochem Behav 2024; 243:173836. [PMID: 39067531 PMCID: PMC11344688 DOI: 10.1016/j.pbb.2024.173836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Preclinical models of addictive drugs have been developed for decades to model aspects of the clinical experience in substance use disorders (SUDs). These include passive exposure as well as volitional intake models across addictive drugs and have been utilized to also measure withdrawal symptomatology and potential neurobehavioral mechanisms underlying relapse to drug seeking or taking. There are a number of Food and Drug Administration (FDA)-approved medications for SUDs, however, many demonstrate low clinical efficacy as well as potential sex differences, and we also note gaps in the continuum of care for certain aspects of clinical experiences in individuals who use drugs. In this review, we provide a comprehensive update on both frequently utilized and novel behavioral models of addiction with a focus on translational value to the clinical experience and highlight the need for preclinical research to follow epidemiological trends in drug use patterns to stay abreast of clinical treatment needs. We then note areas in which models could be improved to enhance the medications development pipeline through efforts to enhance translation of preclinical models. Next, we describe neuroscience efforts that can be leveraged to identify novel biological mechanisms to enhance medications development efforts for SUDs, focusing specifically on advances in brain transcriptomics approaches that can provide comprehensive screening and identification of novel targets. Together, the confluence of this review demonstrates the need for careful selection of behavioral models and methodological parameters that better approximate the clinical experience combined with cutting edge neuroscience techniques to advance the medications development pipeline for SUDs.
Collapse
Affiliation(s)
- Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Samar N Chehimi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lexi J Holdiness
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
3
|
Derman RC, Lattal KM. Sex-dependent effects of acute stress in adolescence or adulthood on appetitive motivation. Psychopharmacology (Berl) 2024; 241:1645-1662. [PMID: 38753027 DOI: 10.1007/s00213-024-06587-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/02/2024] [Indexed: 07/25/2024]
Abstract
RATIONALE Intensely stressful experiences can lead to long-lasting changes in appetitive and aversive behaviors. In humans, post-traumatic stress disorder increases the risk of comorbid appetitive disorders including addiction and obesity. We have previously shown that an acute stressful experience in adult male rats suppresses motivation for natural reward. OBJECTIVES We examine the impact of sex and age on the effects of intense stress on action-based (instrumental) and stimulus-based (Pavlovian) motivation for natural reward (food). METHODS Rats received 15 unsignaled footshocks (stress) in a single session followed by appetitive training and testing in a distinct context. In Experiment 1, stress occurred in either adolescence (PN28) or adulthood (PN70) with appetitive training and testing beginning on PN71 for all rats. In Experiment 2, stress and appetitive training/testing occurred in adolescence. RESULTS Acute stress in adolescent females suppressed instrumental motivation assessed with progressive ratio testing when testing occurred in late adolescence or in adulthood, whereas in males stress in adolescence did not suppress instrumental motivation. Acute stress in adulthood did not alter instrumental motivation. In contrast, Pavlovian motivation assessed with single-outcome Pavlovian-to-instrumental transfer (SO-PIT) was consistently enhanced in females following adolescent or adult stress. In males, however, stress in adolescence had no effect, whereas stress in adulthood attenuated SO-PIT. CONCLUSIONS Acute stress in adolescence or adulthood altered instrumental motivation and stimulus-triggered Pavlovian motivation in a sex and developmentally specific manner. These findings suggest that the persistent effects of acute stress on Pavlovian and instrumental motivational processes differ in females and males, and that males may be less vulnerable to the deleterious effects of intense stress during adolescence on appetitive motivation.
Collapse
Affiliation(s)
- Rifka C Derman
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
4
|
Durand-de Cuttoli R, Martínez-Rivera FJ, Li L, Minier-Toribio A, Dong Z, Cai DJ, Russo SJ, Nestler EJ, Sweis BM. A Double Hit of Social and Economic Stress in Mice Precipitates Changes in Decision-Making Strategies. Biol Psychiatry 2024; 96:67-78. [PMID: 38141911 PMCID: PMC11168892 DOI: 10.1016/j.biopsych.2023.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND Economic stress can serve as a second hit for people who have already accumulated a history of adverse life experiences. How one recovers from a setback is a core feature of resilience but is seldom captured in animal studies. METHODS We challenged mice in a novel 2-hit stress model by first exposing them to chronic social defeat stress and then testing adaptations to increasing reward scarcity on a neuroeconomic task. Mice were tested across months on the Restaurant Row task, during which they foraged daily for their primary source of food while on a limited time budget in a closed-economy system. An abrupt transition into a reward-scarce environment elicits an economic challenge, precipitating a drop in food intake and body weight to which mice must respond to survive. RESULTS We found that mice with a history of social stress mounted a robust behavioral response to this economic challenge that was achieved through a complex redistribution of time allocation among competing opportunities. Interestingly, we found that mice with a history of social defeat displayed changes in the development of decision-making policies during the recovery process that are important not only for ensuring food security necessary for survival but also prioritizing subjective value and that these changes emerged only for certain types of choices. CONCLUSIONS These findings indicate that an individual's capacity to recover from economic challenges depends on that person's prior history of stress and can affect multiple decision-making aspects of subjective well-being, thus highlighting a motivational balance that may be altered in stress-related disorders such as depression.
Collapse
Affiliation(s)
- Romain Durand-de Cuttoli
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Long Li
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Angélica Minier-Toribio
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zhe Dong
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Denise J Cai
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brian M Sweis
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
5
|
Martz J, Shelton MA, Geist L, Seney ML, Kentner AC. Sex differences in offspring risk and resilience following 11β-hydroxylase antagonism in a rodent model of maternal immune activation. Neuropsychopharmacology 2024; 49:1078-1090. [PMID: 38007547 PMCID: PMC11109257 DOI: 10.1038/s41386-023-01771-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/21/2023] [Accepted: 11/08/2023] [Indexed: 11/27/2023]
Abstract
Maternal immune activation (MIA) puts offspring at greater risk for neurodevelopmental disorders associated with impaired social behavior. While it is known that immune signaling through maternal, placental, and fetal compartments contributes to these phenotypical changes, it is unknown to what extent the stress response to illness is involved and how it can be harnessed for potential interventions. To this end, on gestational day 15, pregnant rat dams were administered the bacterial mimetic lipopolysaccharide (LPS; to induce MIA) alongside metyrapone, a clinically available 11β-hydroxylase (11βHSD) inhibitor used to treat hypercortisolism in pregnant, lactating, and neonatal populations. Maternal, placental, and fetal brain levels of corticosterone and placental 11βHSD enzymes type 1 and 2 were measured 3-hrs post treatment. Offspring social behaviors were evaluated across critical phases of development. MIA was associated with increased maternal, placental, and fetal brain corticosterone concentrations that were diminished with metyrapone exposure. Metyrapone protected against reductions in placental 11βHSD2 in males only, suggesting that less corticosterone was inactivated in female placentas. Behaviorally, metyrapone-exposure attenuated MIA-induced social disruptions in juvenile, adolescent, and adult males, while females were unaffected or performed worse. Metyrapone-exposure reversed MIA-induced transcriptional changes in monoamine-, glutamate-, and GABA-related genes in adult male ventral hippocampus, but not in females. Taken together, these findings illustrate that MIA-induced HPA responses act alongside the immune system to produce behavioral deficits. As a clinically available drug, the sex-specific benefits and constraints of metyrapone should be investigated further as a potential means of reducing neurodevelopmental risks due to gestational MIA.
Collapse
Affiliation(s)
- Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Micah A Shelton
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Laurel Geist
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA.
| |
Collapse
|
6
|
Valentino RJ, Nair SG, Volkow ND. Neuroscience in addiction research. J Neural Transm (Vienna) 2024; 131:453-459. [PMID: 37947883 DOI: 10.1007/s00702-023-02713-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
The prevention and treatment of addiction (moderate to severe substance use disorder-SUD) have remained challenging because of the dynamic and complex interactions between multiple biological and social determinants that shape SUD. The pharmacological landscape is ever changing and the use of multiple drugs is increasingly common, requiring an unraveling of pharmacological interactions to understand the effects. There are different stages in the trajectory from drug use to addiction that are characterized by distinct cognitive and emotional features. These are directed by different neurobiological processes that require identification and characterization including those that underlie the high co-morbidity with other disorders. Finally, there is substantial individual variability in the susceptibility to develop SUD because there are multiple determinants, including genetics, sex, developmental trajectories and times of drug exposures, and psychosocial and environmental factors including commercial determinants that influence drug availability. Elucidating how these factors interact to determine risk is essential for identifying the biobehavioral basis of addiction and developing prevention and treatment strategies. Basic research is tasked with addressing each of these challenges. The recent proliferation of technological advances that allow for genetic manipulation, visualization of molecular reactions and cellular activity in vivo, multiscale whole brain mapping across the life span, and the mining of massive data sets including multimodality human brain imaging are accelerating our ability to understand how the brain functions and how drugs influence it. Here, we highlight how the application of these tools to the study of addiction promises to illuminate its neurobiological basis and guide strategies for prevention and treatment.
Collapse
Affiliation(s)
- Rita J Valentino
- National Institute On Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
| | - Sunila G Nair
- National Institute On Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Nora D Volkow
- National Institute On Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Reguilón MD, Manzanedo C, Miñarro J, Rodríguez-Arias M. Stress inoculation during adolescence attenuates social stress-induced increase in ethanol intake in adult male mice. Neuropharmacology 2024; 246:109838. [PMID: 38199295 DOI: 10.1016/j.neuropharm.2024.109838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/05/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Social stress exposure heightens the risk of substance abuse disorder development, especially when endured during adolescence, influencing long-term mental health. This study investigates early-life stress's potential to confer resilience against later-life stressors. To investigate this hypothesis, we examined the impact of a single social defeat (SD) incident during adolescent mice's lives on subsequent voluntary ethanol consumption following repeated adult social stress exposure. Half of the adolescent mice experienced SD at postnatal day 28. Three weeks later (postnatal day 49), defeated groups encountered four confrontations with aggressive residents every 72 h, while control groups were exposed to non-resident exploration. A day after the last SD, defeated mice were classified as resilient or susceptible based on their response to a social interaction test (SIT), a model for depressive behavior. To assess ethanol consumption during young adulthood, researchers used the 'drinking in the dark' and oral ethanol self-administration paradigms. Stress inoculation (IS) slightly increased resilient animals in the SIT. In mice without IS exposure during adolescence, susceptible defeated mice displayed higher ethanol consumption and motivation than control and resilient mice. IS in adolescence effectively counteracted this effect, as IS-SD groups, whether resilient or susceptible, showed no increase in ethanol intake. These groups also exhibited similar motivation to control, measured by the progressive ratio. Notably, elevated IL-6 levels seen in SD-S mice were absent in IS-exposed mice. Additionally, IS-exposed groups had lower prefrontal cortex IL-6 and CX3CL1 levels. These findings support the hypothesis that IS, induced by moderate-intensity stress during adolescence, can enhance resilience to more severe stressors in adulthood.
Collapse
Affiliation(s)
- Marina D Reguilón
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - Carmen Manzanedo
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - José Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain.
| |
Collapse
|
8
|
Deckers C, Karbalaei R, Miles NA, Harder EV, Witt E, Harris EP, Reissner K, Wimmer ME, Bangasser DA. Early resource scarcity causes cortical astrocyte enlargement and sex-specific changes in the orbitofrontal cortex transcriptome in adult rats. Neurobiol Stress 2024; 29:100607. [PMID: 38304302 PMCID: PMC10831308 DOI: 10.1016/j.ynstr.2024.100607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 02/03/2024] Open
Abstract
Astrocyte morphology affects function, including the regulation of glutamatergic signaling. This morphology changes dynamically in response to the environment. However, how early life manipulations alter adult cortical astrocyte morphology is underexplored. Our lab uses brief postnatal resource scarcity, the limited bedding and nesting (LBN) manipulation, in rats. We previously found that LBN augments maternal behaviors and promotes later resilience to adult addiction-related behaviors, reducing impulsivity, risky decision-making, and morphine self-administration. These behaviors rely on glutamatergic transmission in the medial orbitofrontal (mOFC) and medial prefrontal (mPFC) cortex. Here we tested whether LBN changed astrocyte morphology in the mOFC and mPFC of adult rats using a novel viral approach that, unlike traditional markers, fully labels astrocytes. Prior exposure to LBN causes an increase in the surface area and volume of astrocytes in the mOFC and mPFC of adult males and females relative to control-raised rats. We next used bulk RNA sequencing of OFC tissue to assess transcriptional changes that could increase astrocyte size in LBN rats. LBN caused mainly sex-specific changes in differentially expressed genes. Pathway analysis revealed that OFC glutamatergic signaling is altered by LBN in males and females, but the gene changes in that pathway differed across sex. This may represent a convergent sex difference where glutamatergic signaling, which affects astrocyte morphology, is altered by LBN via sex-specific mechanisms. Collectively, these studies highlight that astrocytes may be an important cell type that mediates the effect of early resource scarcity on adult brain function.
Collapse
Affiliation(s)
- Claire Deckers
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, USA
| | - Reza Karbalaei
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, USA
| | - Nylah A. Miles
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, USA
| | - Eden V. Harder
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily Witt
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Erin P. Harris
- Neuroscience Institute, Georgia State University, Atlanta, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, USA
| | - Kathryn Reissner
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mathieu E. Wimmer
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, USA
| | - Debra A. Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, USA
- Neuroscience Institute, Georgia State University, Atlanta, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, USA
| |
Collapse
|
9
|
Parel ST, Bennett SN, Cheng CJ, Timmermans OC, Fiori LM, Turecki G, Peña CJ. Transcriptional signatures of early-life stress and antidepressant treatment efficacy. Proc Natl Acad Sci U S A 2023; 120:e2305776120. [PMID: 38011563 DOI: 10.1073/pnas.2305776120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/10/2023] [Indexed: 11/29/2023] Open
Abstract
Individuals with a history of early-life stress (ELS) tend to have an altered course of depression and lower treatment response rates. Research suggests that ELS alters brain development, but the molecular changes in the brain following ELS that may mediate altered antidepressant response have not been systematically studied. Sex and gender also impact the risk of depression and treatment response. Here, we leveraged existing RNA sequencing datasets from 1) blood samples from depressed female- and male-identifying patients treated with escitalopram or desvenlafaxine and assessed for treatment response or failure; 2) the nucleus accumbens (NAc) of female and male mice exposed to ELS and/or adult stress; and 3) the NAc of mice after adult stress, antidepressant treatment with imipramine or ketamine, and assessed for treatment response or failure. We find that transcriptomic signatures of adult stress after a history of ELS correspond with transcriptomic signatures of treatment nonresponse, across species and multiple classes of antidepressants. Transcriptomic correspondence with treatment outcome was stronger among females and weaker among males. We next pharmacologically tested these predictions in our mouse model of early-life and adult social defeat stress and treatment with either chronic escitalopram or acute ketamine. Among female mice, the strongest predictor of behavior was an interaction between ELS and ketamine treatment. Among males, however, early experience and treatment were poor predictors of behavior, mirroring our bioinformatic predictions. These studies provide neurobiological evidence for molecular adaptations in the brain related to sex and ELS that contribute to antidepressant treatment response.
Collapse
Affiliation(s)
- Sero Toriano Parel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | - Shannon N Bennett
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | - Cindy J Cheng
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | | | - Laura M Fiori
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
| | - Gustavo Turecki
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
| | | |
Collapse
|
10
|
Lonstein JS, Vitale EM, Olekanma D, McLocklin A, Pence N, Bredewold R, Veenema AH, Johnson AW, Burt SA. Anxiety, aggression, reward sensitivity, and forebrain dopamine receptor expression in a laboratory rat model of early-life disadvantage. Dev Psychobiol 2023; 65:e22421. [PMID: 37860907 DOI: 10.1002/dev.22421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/24/2023] [Accepted: 08/17/2023] [Indexed: 10/21/2023]
Abstract
Despite early-life disadvantage (ELD) in humans being a highly heterogenous construct, it consistently predicts negative neurobehavioral outcomes. The numerous environmental contributors and neural mechanisms underlying ELD remain unclear, though. We used a laboratory rat model to evaluate the effects of limited resources and/or heavy metal exposure on mothers and their adult male and female offspring. Dams and litters were chronically exposed to restricted (1-cm deep) or ample (4-cm deep) home cage bedding postpartum, with or without lead acetate (0.1%) in their drinking water from insemination through 1-week postweaning. Restricted-bedding mothers showed more pup-directed behaviors and behavioral fragmentation, while lead-exposed mothers showed more nestbuilding. Restricted bedding-raised male offspring showed higher anxiety and aggression. Either restricted bedding or lead exposure impaired goal-directed performance in a reinforcer devaluation task in females, whereas restricted bedding alone disrupted it in males. Lead exposure, but not limited bedding, also reduced sucrose reward sensitivity in a progressive ratio task in females. D1 and D2 receptor mRNA in the medial prefrontal cortex and nucleus accumbens (NAc) were each affected by the early-life treatments and differently between the sexes. Most notably, adult males (but not females) exposed to both early-life treatments had greatly increased D1 receptor mRNA in the NAc core. These results illuminate neural mechanisms through which ELD threatens neurobehavioral development and highlight forebrain dopamine as a factor.
Collapse
Affiliation(s)
- Joseph S Lonstein
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Erika M Vitale
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Doris Olekanma
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Andrew McLocklin
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Nathan Pence
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Remco Bredewold
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Alexa H Veenema
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Alexander W Johnson
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - S Alexandra Burt
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
11
|
Rocks D, Jaric I, Bellia F, Cham H, Greally JM, Suzuki M, Kundakovic M. Early-life stress and ovarian hormones alter transcriptional regulation in the nucleus accumbens resulting in sex-specific responses to cocaine. Cell Rep 2023; 42:113187. [PMID: 37777968 PMCID: PMC10753961 DOI: 10.1016/j.celrep.2023.113187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/29/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023] Open
Abstract
Early-life stress and ovarian hormones contribute to increased female vulnerability to cocaine addiction. Here, we reveal molecular substrates in the reward area, the nucleus accumbens, through which these female-specific factors affect immediate and conditioning responses to cocaine. We find shared involvement of X chromosome inactivation-related and estrogen signaling-related gene regulation in enhanced conditioning responses following early-life stress and during the low-estrogenic state in females. Low-estrogenic females respond to acute cocaine by opening neuronal chromatin enriched for the sites of ΔFosB, a transcription factor implicated in chronic cocaine response and addiction. Conversely, high-estrogenic females respond to cocaine by preferential chromatin closing, providing a mechanism for limiting cocaine-driven chromatin and synaptic plasticity. We find that physiological estrogen withdrawal, early-life stress, and absence of one X chromosome all nullify the protective effect of a high-estrogenic state on cocaine conditioning in females. Our findings offer a molecular framework to enable understanding of sex-specific neuronal mechanisms underlying cocaine use disorder.
Collapse
Affiliation(s)
- Devin Rocks
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Ivana Jaric
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Fabio Bellia
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Heining Cham
- Department of Psychology, Fordham University, Bronx, NY, USA
| | - John M Greally
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Masako Suzuki
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Marija Kundakovic
- Department of Biological Sciences, Fordham University, Bronx, NY, USA.
| |
Collapse
|
12
|
Giovanniello J, Bravo-Rivera C, Rosenkranz A, Matthew Lattal K. Stress, associative learning, and decision-making. Neurobiol Learn Mem 2023; 204:107812. [PMID: 37598745 DOI: 10.1016/j.nlm.2023.107812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/02/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Exposure to acute and chronic stress has significant effects on the basic mechanisms of associative learning and memory. Stress can both impair and enhance associative learning depending on type, intensity, and persistence of the stressor, the subject's sex, the context that the stress and behavior is experienced in, and the type of associative learning taking place. In some cases, stress can cause or exacerbate the maladaptive behavior that underlies numerous psychiatric conditions including anxiety disorders, obsessive-compulsive disorder, post-traumatic stress disorder, substance use disorder, and others. Therefore, it is critical to understand how the varied effects of stress, which may normally facilitate adaptive behavior, can also become maladaptive and even harmful. In this review, we highlight several findings of associative learning and decision-making processes that are affected by stress in both human and non-human subjects and how they are related to one another. An emerging theme from this work is that stress biases behavior towards less flexible strategies that may reflect a cautious insensitivity to changing contingencies. We consider how this inflexibility has been observed in different associative learning procedures and suggest that a goal for the field should be to clarify how factors such as sex and previous experience influence this inflexibility.
Collapse
Affiliation(s)
| | - Christian Bravo-Rivera
- Departments of Psychiatry and Anatomy & Neurobiology, University of Puerto Rico School of Medicine, San Juan, PR 00935, United States.
| | - Amiel Rosenkranz
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Chicago Medical School, Rosalind Franklin University of Medicine and Science, United States.
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States.
| |
Collapse
|
13
|
Singleton S, Sneddon C, Bakina A, Lambert JJ, Hales TG. Early-life adversity increases morphine tolerance and persistent inflammatory hypersensitivity through upregulation of δ opioid receptors in mice. Pain 2023; 164:2253-2264. [PMID: 37171192 PMCID: PMC10502877 DOI: 10.1097/j.pain.0000000000002925] [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: 01/05/2023] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 05/13/2023]
Abstract
ABSTRACT Exposure to severely stressful events during childhood is associated with poor health outcomes in later life, including chronic pain and substance use disorder. However, the mediators and mechanisms are unclear. We investigated the impact of a well-characterized mouse model of early-life adversity, fragmented maternal care (FC) between postnatal day 2 and 9, on nociception, inflammatory hypersensitivity, and responses to morphine. Male and female mice exposed to FC exhibited prolonged basal thermal withdrawal latencies and decreased mechanical sensitivity. In addition, morphine had reduced potency in mice exposed to FC and their development of tolerance to morphine was accelerated. Quantitative PCR analysis in several brain regions and the spinal cords of juvenile and adult mice revealed an impact of FC on the expression of genes encoding opioid peptide precursors and their receptors. These changes included enhanced abundance of δ opioid receptor transcript in the spinal cord. Acute inflammatory hypersensitivity (induced by hind paw administration of complete Freund's adjuvant) was unaffected by exposure to FC. However, after an initial recovery of mechanical hypersensitivity, there was a reappearance in mice exposed to FC by day 15, which was not seen in control mice. Changes in nociception, morphine responses, and hypersensitivity associated with FC were apparent in males and females but were absent from mice lacking δ receptors or β-arrestin2. These findings suggest that exposure to early-life adversity in mice enhances δ receptor expression leading to decreased basal sensitivity to noxious stimuli coupled with accelerated morphine tolerance and enhanced vulnerability to persistent inflammatory hypersensitivity.
Collapse
Affiliation(s)
- Sam Singleton
- The Institute of Academic Anaesthesia, Division of Cellular and Systems Medicine, School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - Claire Sneddon
- The Institute of Academic Anaesthesia, Division of Cellular and Systems Medicine, School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - Alice Bakina
- The Institute of Academic Anaesthesia, Division of Cellular and Systems Medicine, School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - Jeremy J. Lambert
- The Institute of Academic Anaesthesia, Division of Cellular and Systems Medicine, School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - Tim G. Hales
- The Institute of Academic Anaesthesia, Division of Cellular and Systems Medicine, School of Medicine, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
14
|
Maulik M, Looschen K, Smith C, Johnson K, Carman AF, Nagisetty C, Corriveau K, Salisbury C, Deschepper K, Michels M, Henderson-Redmond AN, Morgan DJ, Mitra S. Postpartum scarcity-adversity inflicts sex-specific cerebellar adaptations and reward behaviors in adolescence. Pharmacol Biochem Behav 2023; 231:173620. [PMID: 37625522 PMCID: PMC10565883 DOI: 10.1016/j.pbb.2023.173620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Early life adversity in the form of poor postnatal care is a major developmental stressor impacting behavior later in life. Previous studies have shown the impact of early life stress on neurobehavioral abnormalities. Specifically, research has demonstrated how limited bedding and nesting (LBN) materials can cause behavioral deficits in adulthood. There is, however, a limited understanding of how LBN influences sex-specific neurobehavioral adaptation in adolescence, a developmental stage susceptible to psychiatric diseases including substance use disorder. LBN and stress-naive c57BL/6 adolescent male and female mouse offspring were tested for a battery of behaviors including open field, novel object recognition, elevated plus maze, social preference, and morphine-induced conditioned place preference. There was a significant sex-specific deficit in social preference in male mice exposed to LBN compared to stress-naïve counterparts and both LBN males and females had a higher preference towards the drug-paired chamber in the morphine-induced conditioned place preference test. These behavioral deficits were concomitant with sex-specific increases in the transcription factor, Klf9 in the deep cerebellar nuclei (DCN) of males. Further, mRNA levels of the circadian gene Bmal1, which is known to be transcriptionally regulated by Klf9, were decreased in the DCN. Since Bmal1 has recently been implicated in extracellular matrix modulation, we examined perineuronal nets (PNN) and observed depleted PNN in the DCN of males but not female LBN mice. Overall, we provide a novel understanding of how postpartum adversity impinges on the cerebellar extracellular matrix homeostasis, likely, through disruption of the circadian axis by Klf9 that might underlie sex-specific behavioral adaptations in adolescence.
Collapse
Affiliation(s)
- Malabika Maulik
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Kassandra Looschen
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Colton Smith
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Khyla Johnson
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Alaina F Carman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Cherishma Nagisetty
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Katilyn Corriveau
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Colin Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Kayla Deschepper
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Madison Michels
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Angela N Henderson-Redmond
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Daniel J Morgan
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Swarup Mitra
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States; Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA.
| |
Collapse
|
15
|
Danoff JS, Ramos EN, Hinton TD, Perkeybile AM, Graves AJ, Quinn GC, Lightbody-Cimer AR, Gordevičius J, Milčiūtė M, Brooke RT, Carter CS, Bales KL, Erisir A, Connelly JJ. Father's care uniquely influences male neurodevelopment. Proc Natl Acad Sci U S A 2023; 120:e2308798120. [PMID: 37487074 PMCID: PMC10400995 DOI: 10.1073/pnas.2308798120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/21/2023] [Indexed: 07/26/2023] Open
Abstract
Mammalian infants depend on parental care for survival, with numerous consequences for their behavioral development. We investigated the epigenetic and neurodevelopmental mechanisms mediating the impact of early biparental care on development of alloparenting behavior, or caring for offspring that are not one's own. We find that receiving high parental care early in life leads to slower epigenetic aging of both sexes and widespread male-specific differential expression of genes related to synaptic transmission and autism in the nucleus accumbens. Examination of parental care composition indicates that high-care fathers promote a male-specific increase in excitatory synapses and increases in pup retrieval behavior as juveniles. Interestingly, females raised by high-care fathers have the opposite behavioral response and display fewer pup retrievals. These results support the concept that neurodevelopmental trajectories are programmed by different features of early-life parental care and reveal that male neurodevelopmental processes are uniquely sensitive to care by fathers.
Collapse
Affiliation(s)
- Joshua S. Danoff
- Department of Psychology, University of Virginia, Charlottesville, VA22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA22904
| | - Erin N. Ramos
- Department of Psychology, University of Virginia, Charlottesville, VA22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA22904
| | - Taylor D. Hinton
- Department of Psychology, University of Virginia, Charlottesville, VA22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA22904
| | - Allison M. Perkeybile
- Department of Psychology, University of Virginia, Charlottesville, VA22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA22904
| | - Andrew J. Graves
- Department of Psychology, University of Virginia, Charlottesville, VA22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA22904
| | - Graham C. Quinn
- Department of Psychology, University of Virginia, Charlottesville, VA22904
| | | | | | - Milda Milčiūtė
- Epigenetic Clock Development Foundation, Torrance, CA90502
| | | | - C. Sue Carter
- Department of Psychology, University of Virginia, Charlottesville, VA22904
| | - Karen L. Bales
- Department of Psychology, University of California, Davis, CA95616
| | - Alev Erisir
- Department of Psychology, University of Virginia, Charlottesville, VA22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA22904
| | - Jessica J. Connelly
- Department of Psychology, University of Virginia, Charlottesville, VA22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA22904
| |
Collapse
|
16
|
Deckers C, Karbalaei R, Miles NA, Harder EV, Witt E, Harris EP, Reissner K, Wimmer ME, Bangasser DA. Early resource scarcity causes cortical astrocyte enlargement and sex-specific changes in the orbitofrontal cortex transcriptome in adult rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.01.547315. [PMID: 37425737 PMCID: PMC10327175 DOI: 10.1101/2023.07.01.547315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Astrocyte morphology affects function, including the regulation of glutamatergic signaling. This morphology changes dynamically in response to the environment. However, how early life manipulations alter adult cortical astrocyte morphology is underexplored. Our lab uses brief postnatal resource scarcity, the limited bedding and nesting (LBN) manipulation, in rats. We previously found that LBN promotes later resilience to adult addiction-related behaviors, reducing impulsivity, risky decision-making, and morphine self-administration. These behaviors rely on glutamatergic transmission in the medial orbitofrontal (mOFC) and medial prefrontal (mPFC) cortex. Here we tested whether LBN changed astrocyte morphology in the mOFC and mPFC of adult rats using a novel viral approach that, unlike traditional markers, fully labels astrocytes. Prior exposure to LBN causes an increase in the surface area and volume of astrocytes in the mOFC and mPFC of adult males and females relative to control-raised rats. We next used bulk RNA sequencing of OFC tissue to assess transcriptional changes that could increase astrocyte size in LBN rats. LBN caused mainly sex-specific changes in differentially expressed genes. However, Park7, which encodes for the protein DJ-1 that alters astrocyte morphology, was increased by LBN across sex. Pathway analysis revealed that OFC glutamatergic signaling is altered by LBN in males and females, but the gene changes in that pathway differed across sex. This may represent a convergent sex difference where glutamatergic signaling, which affects astrocyte morphology, is altered by LBN via sex-specific mechanisms. Collectively, these studies highlight that astrocytes may be an important cell type that mediates the effect of early resource scarcity on adult brain function.
Collapse
Affiliation(s)
- Claire Deckers
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia
| | - Reza Karbalaei
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia
| | - Nylah A Miles
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia
| | - Eden V Harder
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Emily Witt
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Erin P Harris
- Neuroscience Institute, Georgia State University, Atlanta
- Center for Behavioral Neuroscience, Georgia State University, Atlanta
| | - Kathryn Reissner
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia
- Neuroscience Institute, Georgia State University, Atlanta
- Center for Behavioral Neuroscience, Georgia State University, Atlanta
| |
Collapse
|
17
|
Calpe-López C, Martínez-Caballero MÁ, García-Pardo MP, Aguilar MA. Resilience to the short- and long-term behavioral effects of intermittent repeated social defeat in adolescent male mice. Pharmacol Biochem Behav 2023:173574. [PMID: 37315696 DOI: 10.1016/j.pbb.2023.173574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/16/2023] [Accepted: 06/03/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Exposure to intermittent repeated social defeat (IRSD) increases the sensitivity of mice to the rewarding effects of cocaine in the conditioned place preference (CPP) paradigm. Some animals are resilient to this effect of IRSD, though research exploring this inconsistency in adolescent mice is scarce. Thus, our aim was to characterize the behavioral profile of mice exposed to IRSD during early adolescence and to explore a potential association with resilience to the short- and long-term effects of IRSD. METHODS Thirty-six male C57BL/6 mice were exposed to IRSD during early adolescence (PND 27, 30, 33 and 36), while another 10 male mice did not undergo stress (controls). Defeated mice and controls then carried out the following battery of behavioral tests; the Elevated Plus Maze, Hole-Board and Social Interaction Test on PND 37, and the Tail Suspension and Splash tests on PND 38. Three weeks later, all the mice were submitted to the CPP paradigm with a low dose of cocaine (1.5 mg/kg). RESULTS IRSD during early adolescence induced depressive-like behavior in the Social Interaction and Splash tests and increased the rewarding effects of cocaine. Mice with low levels of submissive behavior during episodes of defeat were resilient to the short- and long-term effects of IRSD. In addition, resilience to the short-term effects of IRSD on social interaction and grooming behavior predicted resilience to the long-term effects of IRSD on cocaine reward. CONCLUSION Our findings help to characterize the nature of resilience to the effects of social stress during adolescence.
Collapse
Affiliation(s)
- Claudia Calpe-López
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behavior Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain
| | - Maria Ángeles Martínez-Caballero
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behavior Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain
| | - Maria Pilar García-Pardo
- Department of Psychology and Sociology, Faculty of Social Sciences, University of Zaragoza, Teruel, Spain
| | - Maria Asunción Aguilar
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behavior Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain.
| |
Collapse
|
18
|
de Carvalho G, Khoja S, Haile MT, Chen LY. Early life adversity impaired dorsal striatal synaptic transmission and behavioral adaptability to appropriate action selection in a sex-dependent manner. Front Synaptic Neurosci 2023; 15:1128640. [PMID: 37091877 PMCID: PMC10116150 DOI: 10.3389/fnsyn.2023.1128640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/10/2023] [Indexed: 04/25/2023] Open
Abstract
Early life adversity (ELA) is a major health burden in the United States, with 62% of adults reporting at least one adverse childhood experience. These experiences during critical stages of brain development can perturb the development of neural circuits that mediate sensory cue processing and behavioral regulation. Recent studies have reported that ELA impaired the maturation of dendritic spines on neurons in the dorsolateral striatum (DLS) but not in the dorsomedial striatum (DMS). The DMS and DLS are part of two distinct corticostriatal circuits that have been extensively implicated in behavioral flexibility by regulating and integrating action selection with the reward value of those actions. To date, no studies have investigated the multifaceted effects of ELA on aspects of behavioral flexibility that require alternating between different action selection strategies or higher-order cognitive processes, and the underlying synaptic transmission in corticostriatal circuitries. To address this, we employed whole-cell patch-clamp electrophysiology to assess the effects of ELA on synaptic transmission in the DMS and DLS. We also investigated the effects of ELA on the ability to update action control in response to outcome devaluation in an instrumental learning paradigm and reversal of action-outcome contingency in a water T-maze paradigm. At the circuit level, ELA decreased corticostriatal glutamate transmission in male but not in female mice. Interestingly, in DMS, glutamate transmission is decreased in male ELA mice, but increased in female ELA mice. ELA impaired the ability to update action control in response to reward devaluation in a context that promotes goal-directedness in male mice and induced deficits in reversal learning. Overall, our findings demonstrate the sex- and region-dependent effects of ELA on behavioral flexibility and underlying corticostriatal glutamate transmission. By establishing a link between ELA and circuit mechanisms underlying behavioral flexibility, our findings will begin to identify novel molecular mechanisms that can represent strategies for treating behavioral inflexibility in individuals who experienced early life traumatic incidents.
Collapse
Affiliation(s)
- Gregory de Carvalho
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Sheraz Khoja
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Mulatwa T Haile
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Lulu Y Chen
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
- UCI-Conte Center, UCI-NIMH, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
19
|
Bardo MT, Chandler CM, Denehy ED, Carper BA, Prendergast MA, Nolen TL. Effect of the glucocorticoid receptor antagonist PT150 on acquisition and escalation of fentanyl self-administration following early-life stress. Exp Clin Psychopharmacol 2023; 31:362-369. [PMID: 35587421 PMCID: PMC10084834 DOI: 10.1037/pha0000577] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
There is comorbidity between posttraumatic stress disorder (PTSD) and opioid use disorder (OUD), perhaps because PTSD-like stressful experiences early in life alter the hypothalamic-pituitary-adrenal stress axis to increase the risk for OUD. The present study determined if the glucocorticoid receptor antagonist PT150 reduces the escalation of fentanyl intake in rats exposed to a "two-hit" model of early-life stress (isolation rearing and acute stress). Male and female rats were raised during adolescence in either isolated or social housing and then were given either a single acute stress (restraint and cold-water swim) or control treatment in young adulthood. Rats were then treated daily with PT150 (50 mg/kg, oral) or placebo and were tested for acquisition of fentanyl self-administration in 1-hr sessions, followed by escalation across 6-hr sessions. Regardless of PT150 treatment or sex, acquisition of fentanyl self-administration in 1-hr sessions was greater in isolate-housed rats compared to social-housed rats; the acute stress manipulation did not have an effect on self-administration even though it transiently increased plasma corticosterone levels. During the 6-hr sessions, escalation of fentanyl was observed across all treatment groups; however, there was a significant PT150 Treatment × Sex interaction. While males self-administered more than females overall, PT150 decreased intake in males and increased intake in females, thus negating the sex difference. Although PT150 may serve as an effective treatment for reducing the risk of OUD following early-life stress in males, further work is needed to determine the mechanism underlying the differential effects of PT150 in males and females. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
| | | | - Emily D. Denehy
- Department of Psychology, University of Kentucky, 40536, USA
| | | | | | - Tracy L. Nolen
- Research Triangle Institute, Research Triangle Park, NC, USA
| |
Collapse
|
20
|
Simmons SC, Grecco GG, Atwood BK, Nugent FS. Effects of prenatal opioid exposure on synaptic adaptations and behaviors across development. Neuropharmacology 2023; 222:109312. [PMID: 36334764 PMCID: PMC10314127 DOI: 10.1016/j.neuropharm.2022.109312] [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: 09/09/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
In this review, we focus on prenatal opioid exposure (POE) given the significant concern for the mental health outcomes of children with parents affected by opioid use disorder (OUD) in the view of the current opioid crisis. We highlight some of the less explored interactions between developmental age and sex on synaptic plasticity and associated behavioral outcomes in preclinical POE research. We begin with an overview of the rich literature on hippocampal related behaviors and plasticity across POE exposure paradigms. We then discuss recent work on reward circuit dysregulation following POE. Additional risk factors such as early life stress (ELS) could further influence synaptic and behavioral outcomes of POE. Therefore, we include an overview on the use of preclinical ELS models where ELS exposure during key critical developmental periods confers considerable vulnerability to addiction and stress psychopathology. Here, we hope to highlight the similarity between POE and ELS on development and maintenance of opioid-induced plasticity and altered opioid-related behaviors where similar enduring plasticity in reward circuits may occur. We conclude the review with some of the limitations that should be considered in future investigations. This article is part of the Special Issue on 'Opioid-induced addiction'.
Collapse
Affiliation(s)
- Sarah C Simmons
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Greg G Grecco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Brady K Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fereshteh S Nugent
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
21
|
Sex differences in addiction-relevant behavioral outcomes in rodents following early life stress. ADDICTION NEUROSCIENCE 2023; 6. [PMID: 37101684 PMCID: PMC10124992 DOI: 10.1016/j.addicn.2023.100067] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In humans, exposure to early life stress (ELS) is an established risk factor for the development of substance use disorders (SUDs) during later life. Similarly, rodents exposed to ELS involving disrupted mother-infant interactions, such as maternal separation (MS) or adverse caregiving due to scarcity-adversity induced by limited bedding and nesting (LBN) conditions, also exhibit long-term alterations in alcohol and drug consumption. In both humans and rodents, there is a range of addiction-related behaviors that are associated with drug use and even predictive of subsequent SUDs. In rodents, these include increased anxiety-like behavior, impulsivity, and novelty-seeking, altered alcohol and drug intake patterns, as well as disrupted reward-related processes involving consummatory and social behaviors. Importantly, the expression of these behaviors often varies throughout the lifespan. Moreover, preclinical studies suggest that sex differences play a role in how exposure to ELS impacts reward and addiction-related phenotypes as well as underlying brain reward circuitry. Here, addiction-relevant behavioral outcomes and mesolimbic dopamine (DA) dysfunction resulting from ELS in the form of MS and LBN are discussed with a focus on age- and sex-dependent effects. Overall, these findings suggest that ELS may increase susceptibility for later life drug use and SUDs by interfering with the normal maturation of reward-related brain and behavioral function.
Collapse
|
22
|
Williams AV, Peña CJ, Ramos-Maciel S, Laman-Maharg A, Ordoñez-Sanchez E, Britton M, Durbin-Johnson B, Settles M, Hao R, Yokoyama S, Xu C, Luo PX, Dwyer T, Bhela S, Black AM, Labonté B, Serafini RA, Ruiz A, Neve RL, Zachariou V, Nestler EJ, Trainor BC. Comparative Transcriptional Analyses in the Nucleus Accumbens Identifies RGS2 as a Key Mediator of Depression-Related Behavior. Biol Psychiatry 2022; 92:942-951. [PMID: 36075764 PMCID: PMC9794384 DOI: 10.1016/j.biopsych.2022.06.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Major depressive disorder is one of the most commonly diagnosed mental illnesses worldwide, with a higher prevalence in women than in men. Although currently available pharmacological therapeutics help many individuals, they are not effective for most. Animal models have been important for the discovery of molecular alterations in stress and depression, but difficulties in adapting animal models of depression for females has impeded progress in developing novel therapeutic treatments that may be more efficacious for women. METHODS Using the California mouse social defeat model, we took a multidisciplinary approach to identify stress-sensitive molecular targets that have translational relevance for women. We determined the impact of stress on transcriptional profiles in male and female California mouse nucleus accumbens (NAc) and compared these results with data from postmortem samples of the NAc from men and women diagnosed with major depressive disorder. RESULTS Our cross-species computational analyses identified Rgs2 (regulator of G protein signaling 2) as a transcript downregulated by social defeat stress in female California mice and in women with major depressive disorder. RGS2 plays a key role in signal regulation of neuropeptide and neurotransmitter receptors. Viral vector-mediated overexpression of Rgs2 in the NAc restored social approach and sucrose preference in stressed female California mice. CONCLUSIONS These studies show that Rgs2 acting in the NAc has functional properties that translate to changes in anxiety- and depression-related behavior. Future studies should investigate whether targeting Rgs2 represents a novel target for treatment-resistant depression in women.
Collapse
Affiliation(s)
- Alexia V Williams
- Department of Psychology, University of California, Davis, Davis, California
| | - Catherine J Peña
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Princeton Neuroscience Institute, Princeton, New Jersey
| | | | | | - Evelyn Ordoñez-Sanchez
- Department of Psychology, University of California, Davis, Davis, California; Department of Psychology, Temple University, Philadelphia, Pennsylvania
| | - Monica Britton
- Bioinformatics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, California
| | | | - Matt Settles
- Bioinformatics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, California
| | - Rebecca Hao
- Department of Psychology, University of California, Davis, Davis, California
| | - Sae Yokoyama
- Department of Psychology, University of California, Davis, Davis, California
| | - Christine Xu
- Department of Psychology, University of California, Davis, Davis, California
| | - Pei X Luo
- Department of Psychology, University of California, Davis, Davis, California
| | - Tjien Dwyer
- Department of Psychology, University of California, Davis, Davis, California
| | - Shanu Bhela
- Department of Psychology, University of California, Davis, Davis, California
| | - Alexis M Black
- Department of Psychology, University of California, Davis, Davis, California
| | - Benoit Labonté
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry and Neuroscience, Laval University, Québec, Quebec, Canada
| | - Randal Alex Serafini
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anne Ruiz
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, Boston, Massachusetts
| | - Venetia Zachariou
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brian C Trainor
- Department of Psychology, University of California, Davis, Davis, California.
| |
Collapse
|
23
|
Williams AV, Flowers J, Coates KS, Ingram A, Hehn AT, Dupuis M, Wimmer ME, Venniro M, Bangasser DA. Early resource scarcity alters motivation for natural rewards in a sex- and reinforcer-dependent manner. Psychopharmacology (Berl) 2022; 239:3929-3937. [PMID: 36301314 PMCID: PMC9817039 DOI: 10.1007/s00213-022-06264-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/13/2022] [Indexed: 01/11/2023]
Abstract
RATIONALE Early life adversity impacts reward-related behaviors, including reward seeking for drugs of abuse. However, the effects of early stress on natural rewards, such as food and social rewards, which have strong implications for symptoms of psychiatric conditions such as major depressive disorder (MDD), are understudied. To fill this gap, we used the limited bedding and nesting (LBN) procedure to assess the impact of early resource scarcity on motivational drive for both food and social rewards in rats. METHODS Male and female Long Evans rats were reared in either an LBN environment, with limited nesting materials and no enrichment, from their postnatal day 2-9 or control environment with ample nesting materials and enrichment. As adults, they were tested for reward-seeking behavior on progressive ratio operant tasks: food reward (sucrose) or social reward (access to a same-sex/age conspecific). RESULTS We observed sex differences in the impact of LBN on motivation for natural rewards. In males, LBN increased motivation for both a sucrose and social reward. In females, LBN reduced motivation for sucrose but had no effect on social reward. CONCLUSIONS These results suggest that the effects of LBN on motivation for natural rewards are both sex- and reinforcer-dependent, with males and females showing differential motivation for food and social rewards following early scarcity. Our previous data revealed an LBN-driven reduction in motivation for morphine in males and no effect in females, highlighting the reinforcer-dependent impact of early resource scarcity on motivated behavior more widely.
Collapse
Affiliation(s)
- Alexia V Williams
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - James Flowers
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Kennedy S Coates
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Atiba Ingram
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Alexandra T Hehn
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Molly Dupuis
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Marco Venniro
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA.
- Neuroscience Institute, Georgia State University, GA, 30303-5030, Atlanta, USA.
| |
Collapse
|
24
|
Got milk? Maternal immune activation during the mid-lactational period affects nutritional milk quality and adolescent offspring sensory processing in male and female rats. Mol Psychiatry 2022; 27:4829-4842. [PMID: 36056174 PMCID: PMC9771965 DOI: 10.1038/s41380-022-01744-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 01/14/2023]
Abstract
Previous studies have underscored the importance of breastfeeding and parental care on offspring development and behavior. However, their contribution as dynamic variables in animal models of early life stress are often overlooked. In the present study, we investigated how lipopolysaccharide (LPS)-induced maternal immune activation (MIA) on postnatal day (P)10 affects maternal care, milk, and offspring development. MIA was associated with elevated milk corticosterone concentrations on P10, which recovered by P11. In contrast, both milk triglyceride and percent creamatocrit values demonstrated a prolonged decrease following inflammatory challenge. Adolescent MIA offspring were heavier, which is often suggestive of poor early life nutrition. While MIA did not decrease maternal care quality, there was a significant compensatory increase in maternal licking and grooming the day following inflammatory challenge. However, this did not protect against disrupted neonatal huddling or later-life alterations in sensorimotor gating, conditioned fear, mechanical allodynia, or reductions in hippocampal parvalbumin expression in MIA offspring. MIA-associated changes in brain and behavior were likely driven by differences in milk nutritional values and not by direct exposure to LPS or inflammatory molecules as neither LPS binding protein nor interleukin-6 milk levels differed between groups. These findings reflected comparable microbiome and transcriptomic patterns at the genome-wide level. Animal models of early life stress can impact both parents and their offspring. One mechanism that can mediate the effects of such stressors is changes to maternal lactation quality which our data show can confer multifaceted and compounding effects on offspring physiology and behavior.
Collapse
|
25
|
Hamdan JN, Sierra-Fonseca JA, Flores RJ, Saucedo S, Miranda-Arango M, O’Dell LE, Gosselink KL. Early-life adversity increases anxiety-like behavior and modifies synaptic protein expression in a region-specific manner. Front Behav Neurosci 2022; 16:1008556. [PMID: 36338879 PMCID: PMC9626971 DOI: 10.3389/fnbeh.2022.1008556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/29/2022] [Indexed: 02/11/2024] Open
Abstract
Early-life adversity (ELA) can induce persistent neurological changes and increase the risk for developing affective or substance use disorders. Disruptions to the reward circuitry of the brain and pathways serving motivation and emotion have been implicated in the link between ELA and altered adult behavior. The molecular mechanisms that mediate the long-term effects of ELA, however, are not fully understood. We examined whether ELA in the form of neonatal maternal separation (MatSep) modifies behavior and synaptic protein expression in adults. We hypothesized that MatSep would affect dopaminergic and glutamatergic signaling and enhance sensitivity to methamphetamine (Meth) reward or increase anxiety. Male Wistar rats were subjected to MatSep for 180 min/d on postnatal days (PND) 2-14 and allowed to grow to adulthood (PND 60) with no further manipulation. The hippocampus (Hipp), medial prefrontal cortex (mPFC), nucleus accumbens (NAc), and caudate putamen (CPu) were isolated from one subgroup of animals and subjected to Western blot and protein quantitation for tyrosine hydroxylase (TH), α-synuclein (ALPHA), NMDA receptor (NMDAR), dopamine receptor-1 (D1) and -2 (D2), dopamine transporter (DAT), and postsynaptic density 95 (PSD95). Separate group of animals were tested for anxiety-like behavior and conditioned place preference (CPP) to Meth at 0.0, 0.1, and 1.0 mg/kg doses. MatSep rats displayed an increase in basal levels of anxiety-like behavior compared to control animals. MatSep rats also demonstrated CPP to Meth, but their responses did not differ significantly from controls at any drug dose. Increased NMDAR, D2, and ALPHA expression was observed in the NAc and CPu following MatSep; D2 and ALPHA levels were also elevated in the mPFC, along with DAT. MatSep rats had reduced D1 expression in the mPFC and Hipp, with the Hipp also showing a reduction in D2. Only the CPu showed elevated TH and decreased DAT expression levels. No significant changes were found in PSD95 expression in MatSep rats. In conclusion, ELA is associated with long-lasting and region-specific changes in synaptic protein expression that diminish dopamine neurotransmission and increase anxiety-like behavior in adults. These findings illustrate potential mechanisms through which ELA may increase vulnerability to stress-related illness.
Collapse
Affiliation(s)
- Jameel N. Hamdan
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
- Antharis Therapeutics, San Diego, CA, United States
| | - Jorge A. Sierra-Fonseca
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
- Department of Science, Chatham University, Pittsburgh, PA, United States
| | - Rodolfo J. Flores
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
- National Institutes of Health, National Institute of General Medical Sciences, Bethesda, MD, United States
| | - Sigifredo Saucedo
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
| | - Manuel Miranda-Arango
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
| | - Laura E. O’Dell
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
| | - Kristin L. Gosselink
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
- Department of Physiology and Pathology, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| |
Collapse
|
26
|
Perinatal Morphine Exposure Leads to Sex-Dependent Executive Function Deficits and Microglial Changes in Mice. eNeuro 2022; 9:ENEURO.0238-22.2022. [PMID: 36216505 PMCID: PMC9581576 DOI: 10.1523/eneuro.0238-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/19/2022] [Accepted: 08/29/2022] [Indexed: 01/13/2023] Open
Abstract
Children exposed prenatally to opioids are at an increased risk for behavioral problems and executive function deficits. The prefrontal cortex (PFC) and amygdala (AMG) regulate executive function and social behavior and are sensitive to opioids prenatally. Opioids can bind to toll-like receptor 4 (TLR4) to activate microglia, which may be developmentally important for synaptic pruning. Therefore, we tested the effects of perinatal morphine exposure on executive function and social behavior in male and female mouse offspring, along with microglial-related and synaptic-related outcomes. Dams were injected once daily subcutaneously with saline (n = 8) or morphine (MO; 10 mg/kg; n = 12) throughout pregestation, gestation, and lactation until offspring were weaned on postnatal day 21 (P21). Male MO offspring had impairments in attention and accuracy in the five-choice serial reaction time task, while female MO offspring were less affected. Targeted gene expression analysis at P21 in the PFC identified alterations in microglial-related and TLR4-related genes, while immunohistochemical analysis in adult brains indicated decreased microglial Iba1 and phagocytic CD68 proteins in the PFC and AMG in males, but females had an increase. Further, both male and female MO offspring had increased social preference. Overall, these data demonstrate male vulnerability to executive function deficits in response to perinatal opioid exposure and evidence for disruptions in neuron-microglial signaling.
Collapse
|
27
|
DeRosa H, Caradonna SG, Tran H, Marrocco J, Kentner AC. Milking It for All It's Worth: The Effects of Environmental Enrichment on Maternal Nurturance, Lactation Quality, and Offspring Social Behavior. eNeuro 2022; 9:ENEURO.0148-22.2022. [PMID: 35995560 PMCID: PMC9417599 DOI: 10.1523/eneuro.0148-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/10/2022] [Accepted: 07/15/2022] [Indexed: 12/17/2022] Open
Abstract
Breastfeeding confers robust benefits to offspring development in terms of growth, immunity, and neurophysiology. Similarly, improving environmental complexity, i.e., environmental enrichment (EE), contributes developmental advantages to both humans and laboratory animal models. However, the impact of environmental context on maternal care and milk quality has not been thoroughly evaluated, nor are the biological underpinnings of EE on offspring development understood. Here, Sprague Dawley rats were housed and bred in either EE or standard-housed (SD) conditions. EE dams gave birth to a larger number of pups, and litters were standardized and cross-fostered across groups on postnatal day (P)1. Maternal milk samples were then collected on P1 (transitional milk phase) and P10 (mature milk phase) for analysis. While EE dams spent less time nursing, postnatal enrichment exposure was associated with heavier offspring bodyweights. Milk from EE mothers had increased triglyceride levels, a greater microbiome diversity, and a significantly higher abundance of bacterial families related to bodyweight and energy metabolism. These differences reflected comparable transcriptomic changes at the genome-wide level. In addition to changes in lactational quality, we observed elevated levels of cannabinoid receptor 1 in the hypothalamus of EE dams, and sex-dependent and time-dependent effects of EE on offspring social behavior. Together, these results underscore the multidimensional impact of the combined neonatal and maternal environments on offspring development and maternal health. Moreover, they highlight potential deficiencies in the use of "gold standard" laboratory housing in the attempt to design translationally relevant animal models in biomedical research.
Collapse
Affiliation(s)
- Holly DeRosa
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115
| | | | - Hieu Tran
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115
| | - Jordan Marrocco
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065
- Department of Biology, Touro University, New York, NY 10023
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115
| |
Collapse
|
28
|
Baracz SJ, Robinson KJ, Wright AL, Turner AJ, McGregor IS, Cornish JL, Everett NA. Oxytocin as an adolescent treatment for methamphetamine addiction after early life stress in male and female rats. Neuropsychopharmacology 2022; 47:1561-1573. [PMID: 35581382 PMCID: PMC9206013 DOI: 10.1038/s41386-022-01336-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/02/2022] [Accepted: 04/27/2022] [Indexed: 11/08/2022]
Abstract
Early life stress (ELS) is associated with perturbed neural development and augmented vulnerability to mental health disorders, including addiction. How ELS changes the brain to increase addiction risk is poorly understood, and there are no therapies which target this ELS-induced vulnerability. ELS disrupts the oxytocin system, which can modulate addiction susceptibility, suggesting that targeting the oxytocin system may be therapeutic in this ELS-addiction comorbidity. Therefore, we determined whether adolescent oxytocin treatment after ELS could: (1) reduce vulnerability to anxiety, social deficits, and methamphetamine-taking and reinstatement; and (2) restore hypothalamic oxytocin and corticotropin-releasing factor expressing neurons and peripheral oxytocin and corticosterone levels. Long Evans pups underwent maternal separation (MS) for either 15 min or 360 min on postnatal days (PND) 1-21. During adolescence (PNDs 28-42), rats received a daily injection of either oxytocin or saline. In Experiment 1, adult rats were assessed using the elevated plus-maze, social interaction procedure, and methamphetamine self-administration procedure, including extinction, and cue-, methamphetamine- and yohimbine-induced reinstatement. In Experiment 2, plasma for enzyme immunoassays and brain tissue for immunofluorescence were collected from adult rats after acute stress exposure. Adolescent oxytocin treatment ameliorated ELS-induced anxiety and reduced methamphetamine- and yohimbine-induced reinstatement in both sexes, and suppressed methamphetamine intake and facilitated extinction in males only. Additionally, adolescent oxytocin treatment after ELS restored oxytocin-immunoreactive cells and stress-induced oxytocin levels in males, and attenuated stress-induced corticosterone levels in both sexes. Adolescent oxytocin treatment reverses some of the ELS effects on later-life psychopathology and vulnerability to addiction.
Collapse
Affiliation(s)
- Sarah J Baracz
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia.
- School of Psychology, University of Sydney, Camperdown, NSW, 2006, Australia.
- Centre for Emotional Health, Macquarie University, North Ryde, NSW, 2109, Australia.
| | - Katherine J Robinson
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Amanda L Wright
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Anita J Turner
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Iain S McGregor
- School of Psychology, University of Sydney, Camperdown, NSW, 2006, Australia
- Lambert Initiative of Cannabinoid Therapeutics, Brain and Mind Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Jennifer L Cornish
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
- Centre for Emotional Health, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Nicholas A Everett
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
- School of Psychology, University of Sydney, Camperdown, NSW, 2006, Australia
| |
Collapse
|
29
|
Levis SC, Birnie MT, Bolton JL, Perrone CR, Montesinos JS, Baram TZ, Mahler SV. Enduring disruption of reward and stress circuit activities by early-life adversity in male rats. Transl Psychiatry 2022; 12:251. [PMID: 35705547 PMCID: PMC9200783 DOI: 10.1038/s41398-022-01988-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
In humans, early-life adversity (ELA) such as trauma, poverty, and chaotic environment is linked to increased risk of later-life emotional disorders including depression and substance abuse. These disorders involve underlying disruption of reward circuits and likely vary by sex. Accordingly, we previously found that ELA leads to anhedonia for natural rewards and cocaine in male rodents, whereas in females ELA instead increases vulnerability to addiction-like use of opioid drugs and palatable food. While these findings suggest that ELA-induced disruption of reward circuitry may differ between the sexes, the specific circuit nodes that are influenced by ELA in either sex remain poorly understood. Here, in adult male Sprague-Dawley rats, we ask how ELA impacts opioid addiction-relevant behaviors that we previously tested after ELA in females. We probe potential circuit mechanisms in males by assessing opioid-associated neuronal activation in stress and reward circuit nodes including nucleus accumbens (NAc), amygdala, medial prefrontal cortex (mPFC), and paraventricular thalamus. We find that ELA diminishes opioid-seeking behaviors in males, and alters heroin-induced activation of NAc, PFC, and amygdala, suggesting a potential circuit-based mechanism. These studies demonstrate that ELA leads to behavioral and neurobiological disruptions consistent with anhedonia in male rodents, unlike the increased opioid seeking we previously saw in females. Our findings, taken together with our prior work, suggest that men and women could face qualitatively different mental health consequences of ELA, which may be essential for individually tailoring future intervention strategies.
Collapse
Affiliation(s)
- Sophia C Levis
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA, USA.
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, USA.
| | - Matthew T Birnie
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Jessica L Bolton
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Christina R Perrone
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, USA
| | - Johanna S Montesinos
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Stephen V Mahler
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
30
|
Sanchez EO, Bangasser DA. The effects of early life stress on impulsivity. Neurosci Biobehav Rev 2022; 137:104638. [PMID: 35341796 DOI: 10.1016/j.neubiorev.2022.104638] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/19/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023]
Abstract
Elevated impulsivity is a symptom shared by various psychiatric disorders such as substance use disorder, bipolar disorder, and attention-deficit/hyperactivity disorder. However, impulsivity is not a unitary construct and impulsive behaviors fall into two subcategories: impulsive action and impulsive choice. Impulsive choice refers to the tendency to prefer immediate, small rewards over delayed, large rewards, whereas impulsive action involves difficulty inhibiting rash, premature, or mistimed behaviors. These behaviors are mediated by the mesocorticolimbic dopamine (DA) system, which consists of projections from the ventral tegmental area to the nucleus accumbens and prefrontal cortex. Early life stress (ELS) alters both impulsive choice and impulsive action in rodents. ELS also changes DA receptor expression, transmission, and activity within the mesocorticolimbic system. This review integrates the dopamine, impulsivity, and ELS literature to provide evidence that ELS alters impulsivity via inducing changes in the mesocorticolimbic DA system. Understanding how ELS affects brain circuits associated with impulsivity can help advance treatments aimed towards reducing impulsivity symptoms in a variety of psychiatric disorders.
Collapse
Affiliation(s)
- Evelyn Ordoñes Sanchez
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA.
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA.
| |
Collapse
|
31
|
Levis SC, Baram TZ, Mahler SV. Neurodevelopmental origins of substance use disorders: Evidence from animal models of early-life adversity and addiction. Eur J Neurosci 2022; 55:2170-2195. [PMID: 33825217 PMCID: PMC8494863 DOI: 10.1111/ejn.15223] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 01/06/2023]
Abstract
Addiction is a chronic relapsing disorder with devastating personal, societal, and economic consequences. In humans, early-life adversity (ELA) such as trauma, neglect, and resource scarcity are linked with increased risk of later-life addiction, but the brain mechanisms underlying this link are still poorly understood. Here, we focus on data from rodent models of ELA and addiction, in which causal effects of ELA on later-life responses to drugs and the neurodevelopmental mechanisms by which ELA increases vulnerability to addiction can be determined. We first summarize evidence for a link between ELA and addiction in humans, then describe how ELA is commonly modeled in rodents. Since addiction is a heterogeneous disease with many individually varying behavioral aspects that may be impacted by ELA, we next discuss common rodent assays of addiction-like behaviors. We then summarize the specific addiction-relevant behavioral phenotypes caused by ELA in male and female rodents and discuss some of the underlying changes in brain reward and stress circuits that are likely responsible. By better understanding the behavioral and neural mechanisms by which ELA promotes addiction vulnerability, we hope to facilitate development of new approaches for preventing or treating addiction in those with a history of ELA.
Collapse
Affiliation(s)
- Sophia C. Levis
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA
| | - Tallie Z. Baram
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA
- Department of Pediatrics, University of California Irvine, Irvine, CA
| | - Stephen V. Mahler
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA
| |
Collapse
|
32
|
Eck SR, Palmer JL, Bavley CC, Karbalaei R, Ordoñes Sanchez E, Flowers J, Holley A, Wimmer ME, Bangasser DA. Effects of early life adversity on male reproductive behavior and the medial preoptic area transcriptome. Neuropsychopharmacology 2022; 47:1231-1239. [PMID: 35102257 PMCID: PMC9019015 DOI: 10.1038/s41386-022-01282-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/18/2021] [Accepted: 01/14/2022] [Indexed: 02/02/2023]
Abstract
Early life adversity can alter reproductive development in humans, changing the timing of pubertal onset and sexual activity. One common form of early adversity is limited access to resources. This adversity can be modeled in rats using the limited bedding/nesting model (LBN), in which dams and pups are placed in a low resource environment from pups' postnatal days 2-9. Our laboratory previously found that adult male rats raised in LBN conditions have elevated levels of plasma estradiol compared to control males. In females, LBN had no effect on plasma hormone levels, pubertal timing, or estrous cycle duration. Estradiol mediates male reproductive behaviors. Thus, here we compared reproductive behaviors in adult males exposed to LBN vs. control housing. LBN males acquired the suite of reproductive behaviors (mounts, intromissions, and ejaculations) more quickly than their control counterparts over 3 weeks of testing. However, there was no effect of LBN in males on puberty onset or masculinization of certain brain regions, suggesting LBN effects on estradiol and reproductive behaviors manifest after puberty. In male and female rats, we next used RNA sequencing to characterize LBN-induced transcriptional changes in the medial preoptic area (mPOA), which underlies male reproductive behaviors. LBN produced sex-specific alterations in gene expression, with many transcripts showing changes in opposite directions. Numerous transcripts altered by LBN in males are regulated by estradiol, linking hormonal changes to molecular changes in the mPOA. Pathway analysis revealed that LBN induced changes in neurosignaling and immune signaling in males and females, respectively. Collectively, these studies reveal novel neurobiological mechanisms by which early life adversity can alter reproductive strategies.
Collapse
Affiliation(s)
- Samantha R. Eck
- grid.264727.20000 0001 2248 3398Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Jamie L. Palmer
- grid.264727.20000 0001 2248 3398Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Charlotte C. Bavley
- grid.264727.20000 0001 2248 3398Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Reza Karbalaei
- grid.264727.20000 0001 2248 3398Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Evelyn Ordoñes Sanchez
- grid.264727.20000 0001 2248 3398Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - James Flowers
- grid.264727.20000 0001 2248 3398Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Amanda Holley
- grid.411024.20000 0001 2175 4264Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Mathieu E. Wimmer
- grid.264727.20000 0001 2248 3398Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Debra A. Bangasser
- grid.264727.20000 0001 2248 3398Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| |
Collapse
|
33
|
Singh A, Xie Y, Davis A, Wang ZJ. Early social isolation stress increases addiction vulnerability to heroin and alters c-Fos expression in the mesocorticolimbic system. Psychopharmacology (Berl) 2022; 239:1081-1095. [PMID: 34997861 DOI: 10.1007/s00213-021-06024-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/04/2021] [Indexed: 01/02/2023]
Abstract
RATIONALE Adverse psychosocial factors during early childhood or adolescence compromise neural structure and brain function, inducing susceptibility for many psychiatric disorders such as substance use disorder. Nevertheless, the mechanisms underlying early life stress-induced addiction vulnerability is still unclear, especially for opioids. OBJECTIVES To address this, we used a mouse heroin self-administration model to examine how chronic early social isolation (ESI) stress (5 weeks, beginning at weaning) affects the behavioral and neural responses to heroin during adulthood. RESULTS We found that ESI stress did not alter the acquisition for sucrose or heroin self-administration, nor change the motivation for sucrose on a progressive ratio schedule. However, ESI stress induced an upward shift of heroin dose-response curve in female mice and increased motivation and seeking for heroin in both sexes. Furthermore, we examined the neuronal activity (measured by c-Fos expression) within the key brain regions of the mesocorticolimbic system, including the prelimbic cortex (PrL), infralimbic cortex (IL), nucleus accumbens (NAc) core and shell, caudate putamen, and ventral tegmental area (VTA). We found that ESI stress dampened c-Fos expression in the PrL, IL, and VTA after 14-day forced abstinence, while augmented the neuronal responses to heroin-predictive context and cue in the IL and NAc core. Moreover, ESI stress disrupted the association between c-Fos expression and attempted infusions during heroin-seeking test in the PrL. CONCLUSIONS These data indicate that ESI stress leads to increased seeking and motivation for heroin, and this may be associated with distinct changes in neuronal activities in different subregions of the mesocorticolimbic system.
Collapse
Affiliation(s)
- Archana Singh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS, 66045, USA
| | - Yang Xie
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS, 66045, USA
| | - Ashton Davis
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS, 66045, USA
| | - Zi-Jun Wang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS, 66045, USA.
| |
Collapse
|
34
|
Baker PM, Mathis V, Lecourtier L, Simmons SC, Nugent FS, Hill S, Mizumori SJY. Lateral Habenula Beyond Avoidance: Roles in Stress, Memory, and Decision-Making With Implications for Psychiatric Disorders. Front Syst Neurosci 2022; 16:826475. [PMID: 35308564 PMCID: PMC8930415 DOI: 10.3389/fnsys.2022.826475] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/10/2022] [Indexed: 01/02/2023] Open
Abstract
In this Perspective review, we highlight some of the less explored aspects of lateral habenula (LHb) function in contextual memory, sleep, and behavioral flexibility. We provide evidence that LHb is well-situated to integrate different internal state and multimodal sensory information from memory-, stress-, motivational-, and reward-related circuits essential for both survival and decision making. We further discuss the impact of early life stress (ELS) on LHb function as an example of stress-induced hyperactivity and dysregulation of neuromodulatory systems within the LHb that promote anhedonia and motivational deficits following ELS. We acknowledge that recent technological advancements in manipulation and recording of neural circuits in simplified and well-controlled behavioral paradigms have been invaluable in our understanding of the critical role of LHb in motivation and emotional regulation as well as the involvement of LHb dysfunction in stress-induced psychopathology. However, we also argue that the use of ethologically-relevant behaviors with consideration of complex aspects of decision-making is warranted for future studies of LHb contributions in a wide range of psychiatric illnesses. We conclude this Perspective with some of the outstanding issues for the field to consider where a multi-systems approach is needed to investigate the complex nature of LHb circuitry interactions with environmental stimuli that predisposes psychiatric disorders.
Collapse
Affiliation(s)
- Phillip M. Baker
- Department of Psychology, Seattle Pacific University, Seattle, WA, United States
- *Correspondence: Phillip M. Baker,
| | - Victor Mathis
- CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, Center National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France
| | - Lucas Lecourtier
- CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Université de Strasbourg, Strasbourg, France
- Lucas Lecourtier,
| | - Sarah C. Simmons
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Fereshteh S. Nugent
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Fereshteh S. Nugent,
| | - Sierra Hill
- Department of Psychology, Seattle Pacific University, Seattle, WA, United States
| | - Sheri J. Y. Mizumori
- Department of Psychology, University of Washington, Seattle, WA, United States
- Sheri J. Y. Mizumori,
| |
Collapse
|
35
|
Cotella EM, Nawreen N, Moloney RD, Martelle SE, Oshima KM, Lemen P, NiBlack JN, Julakanti RR, Fitzgerald M, Baccei ML, Herman JP. Adolescent Stress Confers Resilience to Traumatic Stress Later in Life: Role of the Prefrontal Cortex. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 3:274-282. [PMID: 37124346 PMCID: PMC10140393 DOI: 10.1016/j.bpsgos.2022.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/25/2022] [Accepted: 02/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background Adolescent brains are sensitive to stressors. However, under certain circumstances, developmental stress can promote an adaptive phenotype, allowing individuals to cope better with adverse situations in adulthood, thereby contributing to resilience. Methods Sprague Dawley rats (50 males, 48 females) were subjected to adolescent chronic variable stress (adol CVS) for 2 weeks at postnatal day 45. At postnatal day 85, a group was subjected to single prolonged stress (SPS). After a week, animals were evaluated in an auditory-cued fear conditioning paradigm, and neuronal recruitment during reinstatement was assessed by Fos expression. Patch clamp electrophysiology (17-35 cells/group) was performed in male rats to examine physiological changes associated with resilience. Results Adol CVS blocked fear potentiation evoked by SPS. We observed that SPS impaired extinction (males) and enhanced reinstatement (both sexes) of the conditioned freezing response. Prior adol CVS prevented both effects. SPS effects were associated with a reduction of infralimbic (IL) cortex neuronal recruitment after reinstatement in males and increased engagement of the central amygdala in females, both also prevented by adol CVS, suggesting different neurocircuits involved in generating resilience between sexes. We explored the mechanism behind reduced IL recruitment in males by studying the intrinsic excitability of IL pyramidal neurons. SPS reduced excitability of IL neurons, and prior adol CVS prevented this effect. Conclusions Our data indicate that adolescent stress can impart resilience to the effects of traumatic stress on neuroplasticity and behavior. Our data provide a mechanistic link behind developmental stress-induced behavioral resilience and prefrontal (IL) cortical excitability in males.
Collapse
Affiliation(s)
- Evelin M. Cotella
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
- Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Nawshaba Nawreen
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio
| | - Rachel D. Moloney
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Susan E. Martelle
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Kristen M. Oshima
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Paige Lemen
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Jordan N. NiBlack
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Reetu R. Julakanti
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Maureen Fitzgerald
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Mark L. Baccei
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio
- Department of Anesthesiology, Pain Research Center, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - James P. Herman
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, Ohio
- Veterans Affairs Medical Center, Cincinnati, Ohio
- Address correspondence to James P. Herman, Ph.D.
| |
Collapse
|
36
|
Olsen CM, Corrigan JD. Does Traumatic Brain Injury Cause Risky Substance Use or Substance Use Disorder? Biol Psychiatry 2022; 91:421-437. [PMID: 34561027 PMCID: PMC8776913 DOI: 10.1016/j.biopsych.2021.07.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/07/2021] [Accepted: 07/12/2021] [Indexed: 01/22/2023]
Abstract
There is a high co-occurrence of risky substance use among adults with traumatic brain injury (TBI), although it is unknown if the neurologic sequelae of TBI can promote this behavior. We propose that to conclude that TBI can cause risky substance use, it must be determined that TBI precedes risky substance use, that confounders with the potential to increase the likelihood of both TBI and risky substance use must be ruled out, and that there must be a plausible mechanism of action. In this review, we address these factors by providing an overview of key clinical and preclinical studies and list plausible mechanisms by which TBI could increase risky substance use. Human and animal studies have identified an association between TBI and risky substance use, although the strength of this association varies. Factors that may limit detection of this relationship include differential variability due to substance, sex, age of injury, and confounders that may influence the likelihood of both TBI and risky substance use. We propose possible mechanisms by which TBI could increase substance use that include damage-associated neuroplasticity, chronic changes in neuroimmune signaling, and TBI-associated alterations in brain networks.
Collapse
Affiliation(s)
- Christopher M Olsen
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - John D Corrigan
- Department of Physical Medicine & Rehabilitation, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
37
|
Toussaint AB, Foster W, Jones JM, Kaufmann S, Wachira M, Hughes R, Bongiovanni AR, Famularo ST, Dunham BP, Schwark R, Karbalaei R, Dressler C, Bavley CC, Fried NT, Wimmer ME, Abdus-Saboor I. Chronic paternal morphine exposure increases sensitivity to morphine-derived pain relief in male progeny. SCIENCE ADVANCES 2022; 8:eabk2425. [PMID: 35171664 PMCID: PMC8849295 DOI: 10.1126/sciadv.abk2425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Parental history of opioid exposure is seldom considered when prescribing opioids for pain relief. To explore whether parental opioid exposure may affect sensitivity to morphine in offspring, we developed a "rat pain scale" with high-speed imaging, machine learning, and mathematical modeling in a multigenerational model of paternal morphine self-administration. We find that the most commonly used tool to measure mechanical sensitivity in rodents, the von Frey hair, is not painful in rats during baseline conditions. We also find that male progeny of morphine-treated sires had no baseline changes in mechanical pain sensitivity but were more sensitive to the pain-relieving effects of morphine. Using RNA sequencing across pain-relevant brain regions, we identify gene expression changes within the regulator of G protein signaling family of proteins that may underlie this multigenerational phenotype. Together, this rat pain scale revealed that paternal opioid exposure increases sensitivity to morphine's pain-relieving effects in male offspring.
Collapse
Affiliation(s)
- Andre B. Toussaint
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - William Foster
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Jessica M. Jones
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Samuel Kaufmann
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Meghan Wachira
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Robert Hughes
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Angela R. Bongiovanni
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Sydney T. Famularo
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Benjamin P. Dunham
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Ryan Schwark
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Reza Karbalaei
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Carmen Dressler
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Charlotte C. Bavley
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Nathan T. Fried
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Mathieu E. Wimmer
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
- Corresponding author.
| |
Collapse
|
38
|
Sangha S. Elevated dopamine in the amygdala disrupts infant's approach to mother: Implications for development of neurotypical social behaviors and networks. Neuron 2021; 109:3900-3902. [PMID: 34914916 DOI: 10.1016/j.neuron.2021.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In this issue of Neuron, Opendak et al. (2021) use a suite of techniques that are typically challenging in infant rat pups to examine the role of dopaminergic input to the basolateral amygdala in social behavior deficits in response to early-life adversity.
Collapse
Affiliation(s)
- Susan Sangha
- Department of Psychiatry, Indiana University School of Medicine, IU Health Neuroscience Center, 355 W. 16(th) Street, Suite 4800, Indianapolis, IN 46202-7176, USA.
| |
Collapse
|
39
|
Developmental Shifts in Amygdala Activity during a High Social Drive State. J Neurosci 2021; 41:9308-9325. [PMID: 34611026 DOI: 10.1523/jneurosci.1414-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/27/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022] Open
Abstract
Amygdala abnormalities characterize several psychiatric disorders with prominent social deficits and often emerge during adolescence. The basolateral amygdala (BLA) bidirectionally modulates social behavior and has increased sensitivity during adolescence. We tested how an environmentally-driven social state is regulated by the BLA in adults and adolescent male rats. We found that a high social drive state caused by brief social isolation increases age-specific social behaviors and increased BLA neuronal activity. Chemogenetic inactivation of BLA decreased the effect of high social drive on social engagement. High social drive preferentially enhanced BLA activity during social engagement; however, the effect of social opportunity on BLA activity was greater during adolescence. While this identifies a substrate underlying age differences in social drive, we then determined that high social drive increased BLA NMDA GluN2B expression and sensitivity to antagonism increased with age. Further, the effect of a high social drive state on BLA activity during social engagement was diminished by GluN2B blockade in an age-dependent manner. These results demonstrate the necessity of the BLA for environmentally driven social behavior, its sensitivity to social opportunity, and uncover a maturing role for BLA and its GluN2B receptors in social engagement.SIGNIFICANCE STATEMENT Social engagement during adolescence is a key component of healthy development. Social drive provides the impetus for social engagement and abnormalities underlie social symptoms of depression and anxiety. While adolescence is characterized by transitions in social drive and social environment sensitivity, little is known about the neural basis for these changes. We found that amygdala activity is uniquely sensitive to social environment during adolescence compared with adulthood, and is required for expression of heightened social drive. In addition, the neural substrates shift toward NMDA dependence in adulthood. These results are the first to demonstrate a unique neural signature of higher social drive and begin to uncover the underlying factors that heighten social engagement during adolescence.
Collapse
|
40
|
Hanson JL, Williams AV, Bangasser DA, Peña CJ. Impact of Early Life Stress on Reward Circuit Function and Regulation. Front Psychiatry 2021; 12:744690. [PMID: 34744836 PMCID: PMC8563782 DOI: 10.3389/fpsyt.2021.744690] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022] Open
Abstract
Early life stress - including experience of child maltreatment, neglect, separation from or loss of a parent, and other forms of adversity - increases lifetime risk of mood, anxiety, and substance use disorders. A major component of this risk may be early life stress-induced alterations in motivation and reward processing, mediated by changes in the nucleus accumbens (NAc) and ventral tegmental area (VTA). Here, we review evidence of the impact of early life stress on reward circuit structure and function from human and animal models, with a focus on the NAc. We then connect these results to emerging theoretical models about the indirect and direct impacts of early life stress on reward circuit development. Through this review and synthesis, we aim to highlight open research questions and suggest avenues of future study in service of basic science, as well as applied insights. Understanding how early life stress alters reward circuit development, function, and motivated behaviors is a critical first step toward developing the ability to predict, prevent, and treat stress-related psychopathology spanning mood, anxiety, and substance use disorders.
Collapse
Affiliation(s)
- Jamie L. Hanson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Alexia V. Williams
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Debra A. Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Catherine J. Peña
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| |
Collapse
|
41
|
Shupe EA, Clinton SM. Neonatal resource scarcity alters maternal care and impacts offspring core temperature and growth in rats. Dev Psychobiol 2021; 63:e22144. [PMID: 34053070 DOI: 10.1002/dev.22144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022]
Abstract
Stressful experiences during childhood, including poverty and inconsistent parental care, can enhance vulnerability for worsened physical and mental health outcomes in adulthood. Using Sprague Dawley rats, the present study explored the impact of limited resource availability on maternal behavior and physiological and emotional behavior outcomes in the offspring. Early life adversity was induced by incorporating aspects of the limited bedding and nesting and scarcity models, wherein limited resource availability has previously been shown to provoke unpredictable or adverse maternal care respectively. In our hands, neonatal limited bedding (NLB) stress during postnatal days (P)2-9 altered maternal care, augmenting pup-directed behaviors and reducing self-directed behaviors, and modestly increased the frequency of transitions between discrete behaviors across consecutive timed observations. NLB-exposed pups had lower core body temperatures immediately following the stressful manipulation and exhibited decreased body weight gain across development. However, NLB exposure did not impact adult offspring's social or emotional behavior outcomes in the three-chamber social interaction, novelty-suppressed feeding, splash, or forced swim tests. These findings add to the literature demonstrating that early life adversity impacts maternal care in rodents and can disrupt certain metabolic and thermoregulatory outcomes in the offspring.
Collapse
Affiliation(s)
- Elizabeth A Shupe
- School of Neuroscience, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, Virginia, USA
| | - Sarah M Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, Virginia, USA
| |
Collapse
|
42
|
Kooiker CL, Birnie MT, Baram TZ. The Paraventricular Thalamus: A Potential Sensor and Integrator of Emotionally Salient Early-Life Experiences. Front Behav Neurosci 2021; 15:673162. [PMID: 34079442 PMCID: PMC8166219 DOI: 10.3389/fnbeh.2021.673162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/20/2021] [Indexed: 02/03/2023] Open
Abstract
Early-life experiences influence a broad spectrum of behaviors throughout the lifespan that contribute to resilience or vulnerability to mental health disorders. Yet, how emotionally salient experiences early in life are encoded, stored, and processed and the mechanisms by which they influence future behaviors remain poorly understood. The paraventricular nucleus of the thalamus (PVT) is a key structure in modulating positive and negative experiences and behaviors in adults. However, little is known of the PVT's role in encoding and integrating emotionally salient experiences that occur during neonatal, infancy, and childhood periods. In this review, we (1) describe the functions and connections of the PVT and its regulation of behavior, (2) introduce novel technical approaches to elucidating the role of the PVT in mediating enduring changes in adult behaviors resulting from early-life experiences, and (3) conclude that PVT neurons of neonatal rodents are engaged by both positive and negative emotionally salient experiences, and their activation may enduringly govern future behavior-modulating PVT activity during emotionally salient contexts.
Collapse
Affiliation(s)
- Cassandra L. Kooiker
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, United States
| | - Matthew T. Birnie
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z. Baram
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|