51
|
Melugin PR, Nolan SO, Siciliano CA. Bidirectional causality between addiction and cognitive deficits. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:371-407. [PMID: 33648674 PMCID: PMC8566632 DOI: 10.1016/bs.irn.2020.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cognitive deficits are highly comorbid with substance use disorders. Deficits span multiple cognitive domains, are associated with disease severity across substance classes, and persist long after cessation of substance use. Furthermore, recovery of cognitive function during protracted abstinence is highly predictive of treatment adherence, relapse, and overall substance use disorder prognosis, suggesting that addiction may be best characterized as a disease of executive dysfunction. While the association between cognitive deficits and substance use disorders is clear, determining causalities is made difficult by the complex interplay between these variables. Cognitive dysfunction present prior to first drug use can act as a risk factor for substance use initiation, likelihood of pathology, and disease trajectory. At the same time, substance use can directly cause cognitive impairments even in individuals without preexisting deficits. Thus, parsing preexisting risk factors from substance-induced adaptations, and how they may interact, poses significant challenges. Here, focusing on psychostimulants and alcohol, we review evidence from clinical literature implicating cognitive deficits as a risk factor for addiction, a consequence of substance use, and the role the prefrontal cortex plays in these phenomena. We then review corresponding preclinical literature, highlighting the high degree of congruency between animal and human studies, and emphasize the unique opportunity that animal models provide to test causality between cognitive phenotypes and substance use, and to investigate the underlying neurobiology at a cellular and molecular level. Together, we provide an accessible resource for assessing the validity and utility of forward- and reverse-translation between these clinical and preclinical literatures.
Collapse
Affiliation(s)
- Patrick R Melugin
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, United States
| | - Suzanne O Nolan
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, United States
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, United States.
| |
Collapse
|
52
|
Sorby-Adams AJ, Schneider WT, Goncalves RP, Knolle F, Morton AJ. Measuring executive function in sheep (Ovis aries) using visual stimuli in a semi-automated operant system. J Neurosci Methods 2020; 351:109009. [PMID: 33340554 DOI: 10.1016/j.jneumeth.2020.109009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/27/2020] [Accepted: 11/19/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cognitive impairment is a distinguishing feature of many neurodegenerative diseases. The intra-dimensional (ID) extra-dimensional (ED) attentional set shift task is part of a clinical battery of tests used to evaluate executive function in Huntington's and Alzheimer's disease patients. The IDED task, however, has not translated well to pre-clinical rodent models of neurological disease. NEW METHOD The ability to perform executive tasks coupled with a long lifespan makes sheep (Ovis aries) an ideal species for modelling cognitive decline in progressive neurodegenerative conditions. We describe the methodology for testing the performance of sheep in the IDED task using a semi-automated system in which visual stimuli are presented as coloured letters on computer screens. RESULTS During each stage of IDED testing, all sheep (n = 12) learned successfully to discriminate between different colours and letters. Sheep were quick to learn the rules of acquisition at each stage. They required significantly more trials to reach criterion (p < 0.05) and made more errors (p < 0.05) following stimulus reversal, with the exception of the ED shift (p > 0.05). COMPARISON WITH EXISTING METHOD(S) Previous research shows that sheep can perform IDED set shifting in a walk-through maze using solid objects with two changeable dimensions (colour and shape) as the stimuli. Presenting the stimuli on computer screens provides better validity, greater task flexibility and higher throughput than the walk-through maze. CONCLUSION All sheep completed each stage of the task, with a range of abilities expected in an outbred population. The IDED task described is ideally suited as a quantifiable and clinically translatable measure of executive function in sheep.
Collapse
Affiliation(s)
- A J Sorby-Adams
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - W T Schneider
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - R P Goncalves
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - F Knolle
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom; Department of Neurology, Klinikum recht der Isar, Technical University Munich, Munich, Germany
| | - A J Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, United Kingdom.
| |
Collapse
|
53
|
Hatzipantelis C, Langiu M, Vandekolk TH, Pierce TL, Nithianantharajah J, Stewart GD, Langmead CJ. Translation-Focused Approaches to GPCR Drug Discovery for Cognitive Impairments Associated with Schizophrenia. ACS Pharmacol Transl Sci 2020; 3:1042-1062. [PMID: 33344888 PMCID: PMC7737210 DOI: 10.1021/acsptsci.0c00117] [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: 08/20/2020] [Indexed: 01/07/2023]
Abstract
There are no effective therapeutics for cognitive impairments associated with schizophrenia (CIAS), which includes deficits in executive functions (working memory and cognitive flexibility) and episodic memory. Compounds that have entered clinical trials are inadequate in terms of efficacy and/or tolerability, highlighting a clear translational bottleneck and a need for a cohesive preclinical drug development strategy. In this review we propose hippocampal-prefrontal-cortical (HPC-PFC) circuitry underlying CIAS-relevant cognitive processes across mammalian species as a target source to guide the translation-focused discovery and development of novel, procognitive agents. We highlight several G protein-coupled receptors (GPCRs) enriched within HPC-PFC circuitry as therapeutic targets of interest, including noncanonical approaches (biased agonism and allosteric modulation) to conventional clinical targets, such as dopamine and muscarinic acetylcholine receptors, along with prospective novel targets, including the orphan receptors GPR52 and GPR139. We also describe the translational limitations of popular preclinical cognition tests and suggest touchscreen-based assays that probe cognitive functions reliant on HPC-PFC circuitry and reflect tests used in the clinic, as tests of greater translational relevance. Combining pharmacological and behavioral testing strategies based in HPC-PFC circuit function creates a cohesive, translation-focused approach to preclinical drug development that may improve the translational bottleneck currently hindering the development of treatments for CIAS.
Collapse
Affiliation(s)
- Cassandra
J. Hatzipantelis
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Monica Langiu
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Teresa H. Vandekolk
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Tracie L. Pierce
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Jess Nithianantharajah
- Florey
Institute of Neuroscience
and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Gregory D. Stewart
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Christopher J. Langmead
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
54
|
West RK, Rodgers SP, Leasure JL. Neural Perturbations Associated With Recurrent Binge Alcohol in Male and Female Rats. Alcohol Clin Exp Res 2020; 45:365-374. [PMID: 33295022 DOI: 10.1111/acer.14529] [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: 07/03/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Binge drinking, characterized by brief periods of high intoxication interspersed with periods of abstinence, appears to be particularly damaging to the brain. Binge drinking is increasing among American women, yet few preclinical studies have assessed sex differences in the neurobehavioral effects of binge alcohol. METHODS Adult Long-Evans rats were administered 4 g/kg ethanol (EtOH; or an isocaloric control dose) via intragastric gavage once-weekly. Brains were collected after 3 or 8 binge doses, and immunohistochemistry for mature neurons (NeuN), microglia (Iba1), neurogenesis (DCX), and reactive astrogliosis (vimentin) performed. Stereology was used to quantify target cell populations in the hippocampus and medial prefrontal cortex (mPFC). In a separate cohort of animals, cognition (spatial navigation and reversal learning), affect (tickling-evoked ultrasonic vocalizations), and task-induced c-fos activation were assessed after 3 or 8 binge doses. RESULTS Blood EtOH concentration did not differ significantly between females (175 ± 3.6 mg/dl) and males (180 ± 3.7 mg/dl) and did not change significantly over time, indicating that tolerance did not develop. After 3 or 8 binge doses, the number of granule neurons in the hippocampal dentate gyrus of both sexes was significantly reduced in comparison with controls, although there was no binge effect on newly generated neurons. Moreover, 8 (but not 3) binge doses significantly increased the total number of microglia and the number of partially activated microglia in the hippocampus and mPFC in both sexes. There was no detectable reactive astrogliosis (vimentin) in either region at any timepoint. There was no effect of binge alcohol on behavior outcomes in either sex, but binged rats showed increased cellular activation in the mPFC following reversal learning. CONCLUSIONS Our data indicate that recurrent binge alcohol results in similar neural damage and neuroimmune activation in alcohol-vulnerable corticolimbic brain regions in males and females.
Collapse
Affiliation(s)
- Rebecca K West
- Department of Psychology, University of Houston, Houston, Texas
| | | | - J Leigh Leasure
- Department of Psychology, University of Houston, Houston, Texas.,Department of Biology & Biochemistry, University of Houston, Houston, Texas
| |
Collapse
|
55
|
Waddell J, Hill E, Tang S, Jiang L, Xu S, Mooney SM. Choline Plus Working Memory Training Improves Prenatal Alcohol-Induced Deficits in Cognitive Flexibility and Functional Connectivity in Adulthood in Rats. Nutrients 2020; 12:E3513. [PMID: 33202683 PMCID: PMC7696837 DOI: 10.3390/nu12113513] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/27/2020] [Accepted: 11/12/2020] [Indexed: 12/28/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is the leading known cause of intellectual disability, and may manifest as deficits in cognitive function, including working memory. Working memory capacity and accuracy increases during adolescence when neurons in the prefrontal cortex undergo refinement. Rats exposed to low doses of ethanol prenatally show deficits in working memory during adolescence, and in cognitive flexibility in young adulthood. The cholinergic system plays a crucial role in learning and memory processes. Here we report that the combination of choline and training on a working memory task during adolescence significantly improved cognitive flexibility (performance on an attentional set shifting task) in young adulthood: 92% of all females and 81% of control males formed an attentional set, but only 36% of ethanol-exposed males did. Resting state functional magnetic resonance imaging showed that functional connectivity among brain regions was different between the sexes, and was altered by prenatal ethanol exposure and by choline + training. Connectivity, particularly between prefrontal cortex and striatum, was also different in males that formed a set compared with those that did not. Together, these findings indicate that prenatal exposure to low doses of ethanol has persistent effects on brain functional connectivity and behavior, that these effects are sex-dependent, and that an adolescent intervention could mitigate some of the effects of prenatal ethanol exposure.
Collapse
Affiliation(s)
- Jaylyn Waddell
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (E.H.); (S.M.M.)
| | - Elizabeth Hill
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (E.H.); (S.M.M.)
| | - Shiyu Tang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.T.); (L.J.); (S.X.)
| | - Li Jiang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.T.); (L.J.); (S.X.)
| | - Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.T.); (L.J.); (S.X.)
| | - Sandra M. Mooney
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (E.H.); (S.M.M.)
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| |
Collapse
|
56
|
Moretti J, Poh EZ, Bolland SJ, Harvey AR, Albrecht MA, Rodger J. Concurrent LI-rTMS induces changes in c-Fos expression but not behavior during a progressive ratio task with adult ephrin-A2A5 -/- mice. Behav Brain Res 2020; 400:113011. [PMID: 33181182 DOI: 10.1016/j.bbr.2020.113011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/04/2020] [Accepted: 11/05/2020] [Indexed: 12/28/2022]
Abstract
Changes within the dopaminergic system induced by repetitive transcranial magnetic stimulation (rTMS) may contribute to its therapeutic effects; however, dopamine-related behavioral effects of rTMS have not been widely investigated. We recently showed that ephrin-A2A5-/- mice completed significantly fewer trials in a visual task than wildtype mice, and that concurrent low-intensity (LI-) rTMS during the task could partially rescue the abnormal behavior [Poh et al. 2018, eNeuro, vol. 5]. Here, we investigated whether the behavioral differences in ephrin-A2A5-/- mice are due to abnormal motivation, primarily a dopamine-modulated behavior, and whether LI-rTMS would increase motivation. Ephrin-A2A5-/- and wildtype mice underwent 14 daily sessions of progressive ratio (PR) tasks and received either sham or LI-rTMS during the first 10 min. Ephrin-A2A5-/- mice responded more than wildtype comparisons, and LI-rTMS did not influence task performance for either strain. Therefore concurrent stimulation does not influence motivation in a PR task. However, ephrin-A2A5-/- mice did have abnormal performance in the PR tasks after a change in the PR schedule which suggests perseverative behavior. We stained for c-Fos in the prelimbic area (PrL), ventral tegmental area and nucleus accumbens (NAc) core and shell to examine neuronal activity from the final PR session. Sham ephrin-A2A5-/- mice had lower c-Fos expression in the PrL and NAc vs. wildtype mice. Ephrin-A2A5-/- mice that received LI-rTMS showed c-Fos expression closer to wildtype levels in the NAc. Combined with high PR performance, ephrin-A2A5-/- mice show an abnormal shift to habitual responding and LI-rTMS may attenuate this shift.
Collapse
Affiliation(s)
- Jessica Moretti
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia; School of Human Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Eugenia Z Poh
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia; School of Human Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Samuel J Bolland
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | | | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia; Perron Institute for Neurological and Translational Science, Perth, WA, Australia.
| |
Collapse
|
57
|
Durairaja A, Fendt M. Orexin deficiency modulates cognitive flexibility in a sex-dependent manner. GENES BRAIN AND BEHAVIOR 2020; 20:e12707. [PMID: 33070452 DOI: 10.1111/gbb.12707] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/01/2020] [Accepted: 10/14/2020] [Indexed: 12/18/2022]
Abstract
Cognitive flexibility is an important executive function and refers to the ability to adapt behaviors in response to changes in the environment. Of note, many brain disorders are associated with impairments in cognitive flexibility. Several classical neurotransmitter systems including dopamine, acetylcholine and noradrenaline are shown to be important for cognitive flexibility, however, there is not much known about the role of neuropeptides. The neuropeptide orexin, which is brain-widely released by neurons in the lateral hypothalamus, is a major player in maintaining sleep/wake cycle, feeding behavior, arousal, and motivational behavior. Recent studies showed a role of orexin in attention, cognition and stress-induced attenuation of cognitive flexibility by disrupting orexin signaling locally or systemically. However, it is not known so far whether brain-wide reduction or loss of orexin affects cognitive flexibility. We investigated this question by testing male and female orexin-deficient mice in the attentional set shifting task (ASST), an established paradigm of cognitive flexibility. We found that orexin deficiency impaired the intra-dimensional shift phase of the ASST selectively in female homozygous orexin-deficient mice and improved the first reversal learning phase selectively in male homozygous orexin-deficient mice. We also found that these orexin-mediated sex-based modulations of cognitive flexibility were not correlated with trait anxiety, narcoleptic episodes, and reward consumption. Our findings highlight a sexually dimorphic role of orexin in regulating cognitive flexibility and the need for further investigations of sex-specific functions of the orexin circuitry.
Collapse
Affiliation(s)
- Archana Durairaja
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany.,Center of Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
58
|
Wang PY, Boboila C, Chin M, Higashi-Howard A, Shamash P, Wu Z, Stein NP, Abbott LF, Axel R. Transient and Persistent Representations of Odor Value in Prefrontal Cortex. Neuron 2020; 108:209-224.e6. [PMID: 32827456 DOI: 10.1016/j.neuron.2020.07.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 03/10/2020] [Accepted: 07/24/2020] [Indexed: 11/19/2022]
Abstract
The representation of odor in olfactory cortex (piriform) is distributive and unstructured and can only be afforded behavioral significance upon learning. We performed 2-photon imaging to examine the representation of odors in piriform and in two downstream areas, the orbitofrontal cortex (OFC) and the medial prefrontal cortex (mPFC), as mice learned olfactory associations. In piriform, we observed that odor responses were largely unchanged during learning. In OFC, 30% of the neurons acquired robust responses to conditioned stimuli (CS+) after learning, and these responses were gated by internal state and task context. Moreover, direct projections from piriform to OFC can be entrained to elicit learned olfactory behavior. CS+ responses in OFC diminished with continued training, whereas persistent representations of both CS+ and CS- odors emerged in mPFC. Optogenetic silencing indicates that these two brain structures function sequentially to consolidate the learning of appetitive associations.
Collapse
Affiliation(s)
- Peter Y Wang
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Cristian Boboila
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Matthew Chin
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Alexandra Higashi-Howard
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA; Howard Hughes Medical Institute, Columbia University, New York, NY 10027, USA
| | - Philip Shamash
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Zheng Wu
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Nicole P Stein
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - L F Abbott
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Richard Axel
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA; Howard Hughes Medical Institute, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
59
|
Cho KKA, Davidson TJ, Bouvier G, Marshall JD, Schnitzer MJ, Sohal VS. Cross-hemispheric gamma synchrony between prefrontal parvalbumin interneurons supports behavioral adaptation during rule shift learning. Nat Neurosci 2020; 23:892-902. [PMID: 32451483 PMCID: PMC7347248 DOI: 10.1038/s41593-020-0647-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
Organisms must learn new strategies to adapt to changing environments. Activity in different neurons often exhibits synchronization that can dynamically enhance their communication and might create flexible brain states that facilitate changes in behavior. We studied the role of gamma-frequency (~40 Hz) synchrony between prefrontal parvalbumin (PV) interneurons in mice learning multiple new cue-reward associations. Voltage indicators revealed cell-type-specific increases of cross-hemispheric gamma synchrony between PV interneurons when mice received feedback that previously learned associations were no longer valid. Disrupting this synchronization by delivering out-of-phase optogenetic stimulation caused mice to perseverate on outdated associations, an effect not reproduced by in-phase stimulation or out-of-phase stimulation at other frequencies. Gamma synchrony was specifically required when new associations used familiar cues that were previously irrelevant to behavioral outcomes, not when associations involved new cues or for reversing previously learned associations. Thus, gamma synchrony is indispensable for reappraising the behavioral salience of external cues.
Collapse
Affiliation(s)
- Kathleen K A Cho
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas J Davidson
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Guy Bouvier
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Jesse D Marshall
- Department of Organismic & Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Mark J Schnitzer
- Departments of Biology and Applied Physics, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Vikaas S Sohal
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
60
|
Conley TE, Beaudin SA, Lasley SM, Fornal CA, Hartman J, Uribe W, Khan T, Strupp BJ, Smith DR. Early postnatal manganese exposure causes arousal dysregulation and lasting hypofunctioning of the prefrontal cortex catecholaminergic systems. J Neurochem 2020; 153:631-649. [PMID: 31811785 PMCID: PMC7261255 DOI: 10.1111/jnc.14934] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022]
Abstract
Studies have reported associations between environmental manganese (Mn) exposure and impaired cognition, attention, impulse control, and fine motor function in children. Our recent rodent studies established that elevated Mn exposure causes these impairments. Here, rats were exposed orally to 0, 25, or 50 mg Mn kg-1 day-1 during early postnatal life (PND 1-21) or lifelong to determine whether early life Mn exposure causes heightened behavioral reactivity in the open field, lasting changes in the catecholaminergic systems in the medial prefrontal cortex (mPFC), altered dendritic spine density, and whether lifelong exposure exacerbates these effects. We also assessed astrocyte reactivity (glial fibrillary acidic protein, GFAP), and astrocyte complement C3 and S100A10 protein levels as markers of A1 proinflammatory or A2 anti-inflammatory reactive astrocytes. Postnatal Mn exposure caused heightened behavioral reactivity during the first 5-10 min intervals of daily open field test sessions, consistent with impairments in arousal regulation. Mn exposure reduced the evoked release of norepinephrine (NE) and caused decreased protein levels of tyrosine hydroxylase (TH), dopamine (DA) and NE transporters, and DA D1 receptors, along with increased DA D2 receptors. Mn also caused a lasting increase in reactive astrocytes (GFAP) exhibiting increased A1 and A2 phenotypes, with a greater induction of the A1 proinflammatory phenotype. These results demonstrate that early life Mn exposure causes broad lasting hypofunctioning of the mPFC catecholaminergic systems, consistent with the impaired arousal regulation, attention, impulse control, and fine motor function reported in these animals, suggesting that mPFC catecholaminergic dysfunction may underlie similar impairments reported in Mn-exposed children.
Collapse
Affiliation(s)
- Travis E. Conley
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Stephane A. Beaudin
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Stephen M. Lasley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, 61605, USA
| | - Casimir A. Fornal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, 61605, USA
| | - Jasenia Hartman
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Walter Uribe
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Tooba Khan
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Barbara J. Strupp
- Division of Nutritional Sciences and Department of Psychology, Cornell University, Ithaca, NY, 14853, USA
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| |
Collapse
|
61
|
Morè L, Lauterborn JC, Papaleo F, Brambilla R. Enhancing cognition through pharmacological and environmental interventions: Examples from preclinical models of neurodevelopmental disorders. Neurosci Biobehav Rev 2020; 110:28-45. [PMID: 30981451 DOI: 10.1016/j.neubiorev.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022]
Abstract
In this review we discuss the role of environmental and pharmacological treatments to enhance cognition with special regards to neurodevelopmental related disorders and aging. How the environment influences brain structure and function, and the interactions between rearing conditions and gene expression, are fundamental questions that are still poorly understood. We propose a model that can explain some of the discrepancies in findings for effects of environmental enrichment on outcome measures. Evidence of a direct causal correlation of nootropics and treatments that enhanced cognition also will be presented, and possible molecular mechanisms that include neurotrophin signaling and downstream pathways underlying these processes are discussed. Finally we review recent findings achieved with a wide set of behavioral and cognitive tasks that have translational validity to humans, and should be useful for future work on devising appropriate therapies. As will be discussed, the collective findings suggest that a combinational therapeutic approach of environmental enrichment and nootropics could be particularly successful for improving learning and memory in both developmental disorders and normal aging.
Collapse
Affiliation(s)
- Lorenzo Morè
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2XT, Preston, UK.
| | - Julie C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92617, USA.
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163, Genova, Italy.
| | - Riccardo Brambilla
- Neuroscience and Mental Health Research Institute (NMHRI), Division of Neuroscience, School of Biosciences, Cardiff University, CF24 4HQ, Cardiff, UK.
| |
Collapse
|
62
|
Britten RA, Duncan VD, Fesshaye A, Rudobeck E, Nelson GA, Vlkolinsky R. Altered Cognitive Flexibility and Synaptic Plasticity in the Rat Prefrontal Cortex after Exposure to Low (≤15 cGy) Doses of 28Si Radiation. Radiat Res 2020; 193:223-235. [DOI: 10.1667/rr15458.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
| | | | | | - Emil Rudobeck
- Department of Basic Sciences, Loma Linda University, Loma Linda, California, 92354
| | - Gregory A. Nelson
- Department of Basic Sciences, Loma Linda University, Loma Linda, California, 92354
| | - Roman Vlkolinsky
- Department of Basic Sciences, Loma Linda University, Loma Linda, California, 92354
| |
Collapse
|
63
|
Thomas AW, Delevich K, Chang I, Wilbrecht L. Variation in early life maternal care predicts later long range frontal cortex synapse development in mice. Dev Cogn Neurosci 2020; 41:100737. [PMID: 31786477 PMCID: PMC6994474 DOI: 10.1016/j.dcn.2019.100737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 11/03/2019] [Accepted: 11/18/2019] [Indexed: 12/31/2022] Open
Abstract
Empirical and theoretical work suggests that early postnatal experience may inform later developing synaptic connectivity to adapt the brain to its environment. We hypothesized that early maternal experience may program the development of synaptic density on long range frontal cortex projections. To test this idea, we used maternal separation (MS) to generate environmental variability and examined how MS affected 1) maternal care and 2) synapse density on virally-labeled long range axons of offspring reared in MS or control conditions. We found that MS and variation in maternal care predicted bouton density on dorsal frontal cortex axons that terminated in the basolateral amygdala (BLA) and dorsomedial striatum (DMS) with more, fragmented care associated with higher density. The effects of maternal care on these distinct axonal projections of the frontal cortex were manifest at different ages. Maternal care measures were correlated with frontal cortex → BLA bouton density at mid-adolescence postnatal (P) day 35 and frontal cortex → DMS bouton density in adulthood (P85). Meanwhile, we found no evidence that MS or maternal care affected bouton density on ascending orbitofrontal cortex (OFC) or BLA axons that terminated in the dorsal frontal cortices. Our data show that variation in early experience can alter development in a circuit-specific and age-dependent manner that may be relevant to understanding the effects of early life adversity.
Collapse
Affiliation(s)
- A Wren Thomas
- Helen Wills Neuroscience Graduate Program, University of California, Berkeley, CA, 94720, USA
| | - Kristen Delevich
- Department of Psychology, University of California, Berkeley, CA, 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720 USA
| | - Irene Chang
- Department of Psychology, University of California, Berkeley, CA, 94720, USA
| | - Linda Wilbrecht
- Department of Psychology, University of California, Berkeley, CA, 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720 USA.
| |
Collapse
|
64
|
Cognition- and circuit-based dysfunction in a mouse model of 22q11.2 microdeletion syndrome: effects of stress. Transl Psychiatry 2020; 10:41. [PMID: 32066701 PMCID: PMC7026063 DOI: 10.1038/s41398-020-0687-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/19/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022] Open
Abstract
Genetic microdeletion at the 22q11 locus is associated with very high risk for schizophrenia. The 22q11.2 microdeletion (Df(h22q11)/+) mouse model shows cognitive deficits observed in this disorder, some of which can be linked to dysfunction of the prefrontal cortex (PFC). We used behavioral (n = 10 per genotype), electrophysiological (n = 7 per genotype per group), and neuroanatomical (n = 5 per genotype) techniques to investigate schizophrenia-related pathology of Df(h22q11)/+ mice, which showed a significant decrease in the total number of parvalbumin positive interneurons in the medial PFC. The Df(h22q11)/+ mice when tested on PFC-dependent behavioral tasks, including gambling tasks, perform significantly worse than control animals while exhibiting normal behavior on hippocampus-dependent tasks. They also show a significant decrease in hippocampus-medial Prefrontal cortex (H-PFC) synaptic plasticity (long-term potentiation, LTP). Acute platform stress almost abolished H-PFC LTP in both wild-type and Df(h22q11)/+ mice. H-PFC LTP was restored to prestress levels by clozapine (3 mg/kg i.p.) in stressed Df(h22q11)/+ mice, but the restoration of stress-induced LTP, while significant, was similar between wild-type and Df(h22q11)/+ mice. A medial PFC dysfunction may underlie the negative and cognitive symptoms in human 22q11 deletion carriers, and these results are relevant to the current debate on the utility of clozapine in such subjects.
Collapse
|
65
|
Nimitvilai S, Lopez MF, Woodward JJ. Sex-dependent differences in ethanol inhibition of mouse lateral orbitofrontal cortex neurons. Addict Biol 2020; 25:e12698. [PMID: 30468275 DOI: 10.1111/adb.12698] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/21/2018] [Accepted: 10/23/2018] [Indexed: 01/27/2023]
Abstract
Biological differences between males and females likely influence responses to alcohol and the propensity to engage in excessive drinking. In both humans and rodents, females escalate alcohol use and develop addiction-like behaviors faster than males, while males exhibit more severe withdrawal symptoms during abstinence. The mechanisms underlying these differences are not yet known but may reflect fundamental differences in the ethanol sensitivity of neurons in reward and control areas of the brain. To address this question, we recorded current-evoked spiking of lateral orbitofrontal cortex (lOFC) neurons in male and female C57BL/6J mice following acute and chronic exposure to ethanol. Ethanol (11-66 mM) reduced firing of lOFC neurons but produced less inhibition in neurons from female mice. As previously reported for male mice, the glycine receptor blocker strychnine blocked ethanol inhibition of spiking of lOFC neurons from female mice and prevented the ethanol-induced increase in tonic current. Following chronic intermittent ethanol (CIE) exposure, current-evoked spiking of lOFC neurons was significantly enhanced with a greater effect observed in males. After CIE treatment, acute ethanol had no effect on spiking in neurons from male mice, while it produced a slight but significant decrease in firing in females. Finally, like male mice, the inhibitory effect of the glycine transport inhibitor sarcosine was blunted in CIE-exposed female mice. Together, these results suggest that while lOFC neurons in male and female mice are similarly affected by ethanol, there are significant differences in sensitivity that may contribute to differences in alcohol actions between males and females.
Collapse
Affiliation(s)
- Sudarat Nimitvilai
- Department of NeuroscienceMedical University of South Carolina Charleston South Carolina
| | - Marcelo F. Lopez
- Department of Psychiatry and Behavioral SciencesMedical University of South Carolina Charleston South Carolina
| | - John J. Woodward
- Department of NeuroscienceMedical University of South Carolina Charleston South Carolina
- Department of Psychiatry and Behavioral SciencesMedical University of South Carolina Charleston South Carolina
| |
Collapse
|
66
|
Why we need nonhuman primates to study the role of ventromedial prefrontal cortex in the regulation of threat- and reward-elicited responses. Proc Natl Acad Sci U S A 2019; 116:26297-26304. [PMID: 31871181 DOI: 10.1073/pnas.1902288116] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The ventromedial prefrontal cortex (vmPFC) is consistently implicated in the cognitive and emotional symptoms of many psychiatric disorders, but the causal mechanisms of its involvement remain unknown. In part, this is because of the poor characterization of the disorders and their symptoms, and the focus of experimental studies in animals on subcortical (rather than cortical) dysregulation. Moreover, even in those experimental studies that have focused on the vmPFC, the preferred animal model for such research has been the rodent, in which there are marked differences in the organization of this region to that seen in humans, and thus the extent of functional homology is unclear. There is also a paucity of well-defined behavioral paradigms suitable for translating disorder-relevant findings across species. With these considerations in mind, we discuss the value of nonhuman primates (NHPs) in bridging the translational gap between human and rodent studies. We focus on recent investigations into the involvement in reward and threat processing of 2 major regions of the vmPFC, areas 25 and 32 in NHPs and their anatomical homologs, the infralimbic and prelimbic cortex, in rodents. We highlight potential similarities, but also differences between species, and consider them in light of the extent to which anatomical homology reflects functional homology, the expansion of the PFC in human and NHPs, and most importantly how they can guide future studies to improve the translatability of findings from preclinical animal studies into the clinic.
Collapse
|
67
|
Goodwill HL, Manzano-Nieves G, LaChance P, Teramoto S, Lin S, Lopez C, Stevenson RJ, Theyel BB, Moore CI, Connors BW, Bath KG. Early Life Stress Drives Sex-Selective Impairment in Reversal Learning by Affecting Parvalbumin Interneurons in Orbitofrontal Cortex of Mice. Cell Rep 2019; 25:2299-2307.e4. [PMID: 30485800 PMCID: PMC6310486 DOI: 10.1016/j.celrep.2018.11.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/18/2018] [Accepted: 10/31/2018] [Indexed: 01/25/2023] Open
Abstract
Poverty, displacement, and parental stress represent potent sources of early life stress (ELS). Stress disproportionately affects females, who are at increased risk for stress-related pathologies associated with cognitive impairment. Mechanisms underlying stress-associated cognitive impairment and enhanced risk of females remain unknown. Here, ELS is associated with impaired rule-reversal (RR) learning in females, but not males. Impaired performance was associated with decreased expression and density of interneurons expressing parvalbumin (PV+) in orbitofrontal cortex (OFC), but not other inter-neuron subtypes. Optogenetic silencing of PV+ inter-neuron activity in OFC of control mice phenocopied RR learning deficits observed in ELS females. Localization of reversal learning deficits to PV+ interneurons in OFC was confirmed by optogenetic studies in which neurons in medial prefrontal cortex (mPFC) were silenced and associated with select deficits in rule-shift learning. Sex-, cell-, and region-specific effects show altered PV+ interneuron development can be a driver of sex differences in cognitive dysfunction. Goodwill et al. investigate the effect of early life stress (ELS) on cognitive development in a mouse model. Using a combination of genetic, histological, optogenetic, and behavioral techniques, they find that ELS leads to female-selective impairments in the ability to engage in rule reversal, but not other forms of attentional learning. Impairments are associated with diminished parvalbumin (PV) expression and a decreased density of PV+ interneurons in the orbitofrontal cortex (OFC).
Collapse
Affiliation(s)
- Haley L Goodwill
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | | | - Patrick LaChance
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Sana Teramoto
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Shirley Lin
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Chelsea Lopez
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Rachel J Stevenson
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, USA
| | - Brian B Theyel
- Department of Psychiatry and Human Behavior, Brown University, Providence, RI 02912, USA
| | | | - Barry W Connors
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Kevin G Bath
- Department of Cognitive, Linguistic and Psychological Sciences, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
68
|
Mancini N, Hranova S, Weber J, Weiglein A, Schleyer M, Weber D, Thum AS, Gerber B. Reversal learning in Drosophila larvae. Learn Mem 2019; 26:424-435. [PMID: 31615854 PMCID: PMC6796787 DOI: 10.1101/lm.049510.119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/09/2019] [Indexed: 01/08/2023]
Abstract
Adjusting behavior to changed environmental contingencies is critical for survival, and reversal learning provides an experimental handle on such cognitive flexibility. Here, we investigate reversal learning in larval Drosophila Using odor-taste associations, we establish olfactory reversal learning in the appetitive and the aversive domain, using either fructose as a reward or high-concentration sodium chloride as a punishment, respectively. Reversal learning is demonstrated both in differential and in absolute conditioning, in either valence domain. In differential conditioning, the animals are first trained such that an odor A is paired, for example, with the reward whereas odor B is not (A+/B); this is followed by a second training phase with reversed contingencies (A/B+). In absolute conditioning, odor B is omitted, such that the animals are first trained with paired presentations of A and reward, followed by unpaired training in the second training phase. Our results reveal "true" reversal learning in that the opposite associative effects of both the first and the second training phase are detectable after reversed-contingency training. In what is a surprisingly quick, one-trial contingency adjustment in the Drosophila larva, the present study establishes a simple and genetically easy accessible study case of cognitive flexibility.
Collapse
Affiliation(s)
- Nino Mancini
- Department of Genetics, Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
| | - Sia Hranova
- Institute for Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Julia Weber
- Department of Genetics, Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
| | - Aliće Weiglein
- Department of Genetics, Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
| | - Michael Schleyer
- Department of Genetics, Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
| | - Denise Weber
- Institute for Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Andreas S Thum
- Institute for Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Bertram Gerber
- Department of Genetics, Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
- Institute for Biology, Otto von Guericke University, 39106 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| |
Collapse
|
69
|
Marquardt K, Cavanagh JF, Brigman JL. Alcohol exposure in utero disrupts cortico-striatal coordination required for behavioral flexibility. Neuropharmacology 2019; 162:107832. [PMID: 31678398 DOI: 10.1016/j.neuropharm.2019.107832] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/09/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022]
Abstract
Deficits in behavioral flexibility are a hallmark of multiple psychiatric, neurological, and substance use disorders. These deficits are often marked by decreased function of the prefrontal cortex (PFC); however, the genesis of such executive deficits remains understudied. Here we report how the most preventable cause of developmental disability, in utero exposure to alcohol, alters cortico-striatal circuit activity leading to impairments in behavioral flexibility in adulthood. We utilized a translational touch-screen task coupled with in vivo electrophysiology in adult mice to examine single unit and coordinated activity of the lateral orbital frontal cortex (OFC) and dorsolateral striatum (DS) during flexible behavior. Prenatal alcohol exposure (PAE) decreased OFC, and increased DS, single unit activity during reversal learning and altered the number of choice responsive neurons in both regions. PAE also decreased coordinated activity within the OFC and DS as measured by oscillatory field activity and altered spike-field coupling. Furthermore, PAE led to sustained connectivity between regions past what was seen in control animals. These findings suggest that PAE causes altered coordination within and between the OFC and DS, promoting maladaptive perseveration. Our model suggests that in optimally functioning mice OFC disengages the DS and updates the newly changed reward contingency, whereas in PAE animals, aberrant and persistent OFC to DS signaling drives behavioral inflexibility during early reversal sessions. Together, these findings demonstrate how developmental exposure alters circuit-level activity leading to behavioral deficits and suggest a critical role for coordination of neural timing during behaviors requiring executive function.
Collapse
Affiliation(s)
- Kristin Marquardt
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - James F Cavanagh
- Department of Psychology, University of New Mexico, Albuquerque, NM, USA
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA; New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
70
|
Exacerbation of the credit assignment problem in rats with lesions of the medial prefrontal cortex is revealed by Bayesian analysis of behavior in the pre-solution period of learning. Behav Brain Res 2019; 372:112037. [DOI: 10.1016/j.bbr.2019.112037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 01/07/2023]
|
71
|
Spatial learning and flexibility in 129S2/SvHsd and C57BL/6J mouse strains using different variants of the Barnes maze. Behav Pharmacol 2019; 29:688-700. [PMID: 30212384 DOI: 10.1097/fbp.0000000000000433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Behavioural flexibility is the ability to switch between tasks and strategies following a change in rules, and involves intact functioning of the medial prefrontal cortex. Impairments of behavioural flexibility have frequently been reported in patients with schizophrenia and rodents with disruption/dysfunction of the prefrontal cortex. The discovery of a mutation in the disrupted in schizophrenia 1 (DISC1) gene in the 129 mouse strain suggests that these mice may be exploited as a 'naturally occurring' model of schizophrenia. The aim of this present study was to assess cognition and behavioural flexibility of 129S2/SvHsd mice in comparison with C57BL/6J mice in the Barnes maze, using three different maze variations that consisted of either 8, 16 or 32 holes. Whereas C57BL/6J mice were able to perform both acquisition and reversal learning in all three mazes, 129S2/SvHsd mice displayed impairments dependent on the complexity of the test. Intact acquisition and reversal occurred in the 8-hole maze; intact acquisition, but impaired reversal, was evident in the 16-hole maze and impaired acquisition was evident in the most difficult 32-hole test. Furthermore, analysis of search strategies confirmed strain differences in the adoption of spatial searches across both acquisition and reversal trials. 129S2/SvHsd mice displayed fewer spatial-type trials than C57BL/6J mice and instead employed more random or serial/chaining search behaviours. The deficits observed in both cognition and behavioural flexibility support the notion of the 129 mouse strain as a potential model of schizophrenia.
Collapse
|
72
|
Sey NYA, Gómez-A A, Madayag AC, Boettiger CA, Robinson DL. Adolescent intermittent ethanol impairs behavioral flexibility in a rat foraging task in adulthood. Behav Brain Res 2019; 373:112085. [PMID: 31319133 DOI: 10.1016/j.bbr.2019.112085] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/19/2022]
Abstract
Alcohol exposure is linked to behavioral flexibility deficits in humans, but it is unclear when the critical exposure occurred or if alcohol exposure alone is sufficient to produce behavior deficits. Increasing evidence shows that binge levels of alcohol during adolescence are particularly harmful to the brain, producing physiological and behavioral effects that can persist into adulthood. The present study determined whether adolescent intermittent ethanol (AIE) in rats impaired action selection in a discriminative stimulus task using a foraging response. Rats were exposed to ethanol during adolescence (5 g/kg/day, IG, 2-days-on/2-days-off, postnatal day 25-54). In adulthood, they learned to dig for food reward buried in one of two media, cued with one of two odors. AIE and control rats both learned to discriminate between olfactory cues, but AIE rats were impaired when reversing that learned association (first intra-dimensional reversal). However, AIE rats were faster to reinstate the original odor discrimination rule (second reversal), suggesting perseverative behavior. Next, the reward location was cued by digging media rather than odor. Both groups learned this extra-dimensional shift; however, control rats were slower to reach criterion. These findings are consistent with studies of people with substance abuse disorder, who learn new stimulus-response associations similarly to, or better than, control subjects, but perseverate when attempting to replace a well-learned association. These data suggest that adolescent binge-alcohol exposure contributes to behavioral flexibility deficits observed in adulthood.
Collapse
Affiliation(s)
- Nancy Y A Sey
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA; Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, USA
| | - Alexander Gómez-A
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA
| | - Aric C Madayag
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA
| | - Charlotte A Boettiger
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA; Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, USA; Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Donita L Robinson
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA; Neuroscience Curriculum, University of North Carolina, Chapel Hill, NC, USA; Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
73
|
Neuwirth LS, Masood S, Anderson DW, Schneider JS. The attention set-shifting test is sensitive for revealing sex-based impairments in executive functions following developmental lead exposure in rats. Behav Brain Res 2019; 366:126-134. [PMID: 30878351 PMCID: PMC6732195 DOI: 10.1016/j.bbr.2019.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/22/2019] [Accepted: 03/11/2019] [Indexed: 12/19/2022]
Abstract
The literature on lead (Pb) exposure has focused in large part on hippocampal-based learning and memory deficits, although frontoexecutive dysfunctions are known to exist in Pb-exposed humans. This study examined the effects of perinatal (PERI) and early postnatal (EPN) developmental low-level Pb-exposures in rats on frontoexecutive functions, using the Attention Set-Shift Test (ASST). Control males and females performed the ASST similarly. Male EPN rats had difficulty with simple discrimination (SD) of odors and failed to complete the compound discrimination (CD) stage of the ASST. All other Pb-exposed rats completed the training and testing. Male PERI rats performed worse on the SD, intradimensional (ID), and intradimensional-reversal (ID-Rev) ASST stages when compared to male Control rats. Female EPN rats performed similar to Controls on the ID-Rev rats, whereas PERI rats performed better the trials-to-criterion on the ID-Rev than EPN and Control rats. Pb-exposed female rats had significant difficulty performing the ED/ED-Rev stages, with the number of trials-to-criterion double that required by Pb-exposed and Control male rats and Control female rats. Together, the ASST results showed that developmental Pb-exposure induces frontoexecutive dysfunction that persists into adulthood, with different sex-based vulnerabilities dependent upon the time-period of neurotoxicant exposure.
Collapse
Affiliation(s)
- Lorenz S Neuwirth
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY 11568, United States; SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury NY 11568, United States; Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States.
| | - Sidrah Masood
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY 11568, United States; SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury NY 11568, United States
| | - David W Anderson
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| | - Jay S Schneider
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| |
Collapse
|
74
|
Manning EE, Dombrovski AY, Torregrossa MM, Ahmari SE. Impaired instrumental reversal learning is associated with increased medial prefrontal cortex activity in Sapap3 knockout mouse model of compulsive behavior. Neuropsychopharmacology 2019; 44:1494-1504. [PMID: 30587851 PMCID: PMC6785097 DOI: 10.1038/s41386-018-0307-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/12/2018] [Accepted: 12/04/2018] [Indexed: 01/29/2023]
Abstract
Convergent functional neuroimaging findings implicate hyperactivity across the prefrontal cortex (PFC) and striatum in the neuropathology of obsessive compulsive disorder (OCD). The impact of cortico-striatal circuit hyperactivity on executive functions subserved by these circuits is unclear, because impaired recruitment of PFC has also been observed in OCD patients during paradigms assessing cognitive flexibility. To investigate the relationship between cortico-striatal circuit disturbances and cognitive functioning relevant to OCD, Sapap3 knockout mice (KOs) and littermate controls were tested in an instrumental reversal-learning paradigm to assess cognitive flexibility. Cortical and striatal activation associated with reversal learning was assessed via quantitative analysis of expression of the immediate early gene cFos and generalized linear mixed-effects models. Sapap3-KOs displayed heterogeneous reversal-learning performance, with almost half (n = 13/28) failing to acquire the reversed contingency, while the other 15/28 had similar acquisition as controls. Notably, reversal impairments were not correlated with compulsive grooming severity. cFos analysis revealed that reversal performance declined as medial PFC (mPFC) activity increased in Sapap3-KOs. No such relationship was observed in controls. Our studies are among the first to describe cognitive impairments in a transgenic OCD-relevant model, and demonstrate pronounced heterogeneity among Sapap3-KOs. These findings suggest that increased neural activity in mPFC is associated with impaired reversal learning in Sapap3-KOs, providing a likely neural basis for this observed heterogeneity. The Sapap3-KO model is thus a useful tool for future mechanistic studies to determine how mPFC hyperactivity contributes to OCD-relevant cognitive dysfunction.
Collapse
Affiliation(s)
- Elizabeth E Manning
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Alexandre Y Dombrovski
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Susanne E Ahmari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
75
|
Wickens MM, Deutschmann AU, McGrath AG, Parikh V, Briand LA. Glutamate receptor interacting protein acts within the prefrontal cortex to blunt cocaine seeking. Neuropharmacology 2019; 157:107672. [PMID: 31233823 DOI: 10.1016/j.neuropharm.2019.107672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/03/2019] [Accepted: 06/12/2019] [Indexed: 12/17/2022]
Abstract
Glutamate receptor interacting protein (GRIP) is a neuronal scaffolding protein that anchors GluA2-containing AMPA receptors to the cell membrane. GRIP plays a critical role in activity-dependent synaptic plasticity, including that which occurs after drug exposure. Given that cocaine administration alters glutamate receptor trafficking within the prefrontal cortex (PFC), a better understanding of the role of receptor trafficking proteins could lead to a more complete understanding of addictive phenotypes. AMPA receptor trafficking in general, and GRIP specifically, is known to play a role in cocaine seeking and conditioned reward in the nucleus accumbens, but its role in the PFC has not been characterized. The current study demonstrates that conditional deletion of GRIP1 in the medial prefrontal cortex increases the motivation for cocaine and potentiates cue-induced reinstatement of cocaine seeking in male and female mice. As no effects of PFC GRIP1 deletion were seen in reinstatement of food seeking, strategy set-shifting, or reversal learning the effects on cocaine seeking are not related to generalized alterations in cognitive function. While disrupting GRIP1 might be expected to lead to decreased AMPA transmission, our electrophysiological data indicate an increase in sEPSC amplitude in the prefrontal cortex and a corresponding decrease in paired pulse facilitation in the nucleus accumbens. Taken together this suggests a strengthening of the PFC to NAc input following prefrontal GRIP1 deletion that may mediate the enhanced drug seeking behavior.
Collapse
Affiliation(s)
| | | | | | - Vinay Parikh
- Department of Psychology, Temple University, USA; Neuroscience Program, Temple University, USA
| | - Lisa A Briand
- Department of Psychology, Temple University, USA; Neuroscience Program, Temple University, USA.
| |
Collapse
|
76
|
Biró S, Lasztóczi B, Klausberger T. A Visual Two-Choice Rule-Switch Task for Head-Fixed Mice. Front Behav Neurosci 2019; 13:119. [PMID: 31244622 PMCID: PMC6562896 DOI: 10.3389/fnbeh.2019.00119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/17/2019] [Indexed: 12/02/2022] Open
Abstract
Cognitive flexibility is the innate ability of the brain to change mental processes and to modify behavioral responses according to an ever-changing environment. As our brain has a limited capacity to process the information of our surroundings in any given moment, it uses sets as a strategy to aid neural processing systems. With assessing the capability of shifting between task sets, it is possible to test cognitive flexibility and executive functions. The most widely used neuropsychological task for the evaluation of these functions in humans is the Wisconsin Card Sorting Test (WCST), which requires the subject to alter response strategies and use previously irrelevant information to solve a problem. The test has proven clinical relevance, as poor performance has been reported in multiple neuropsychiatric conditions. Although, similar tasks have been used in pre-clinical rodent research, many are limited because of their manual-based testing procedures and their hardware attenuates neuronal recordings. We developed a two-choice rule-switch task whereby head-fixed C57BL/6 mice had to choose correctly one of the two virtual objects presented to retrieve a small water reward. The animals learnt to discriminate the visual cues and they successfully switched their strategies according to the related rules. We show that reaching successful performance after the rule changes required more trials in this task and that animals took more time to execute decisions when the two rules were in conflict. We used optogenetics to inhibit temporarily the medial prefrontal cortex (mPFC) during reward delivery and consumption, which significantly increased the number of trials needed to perform the second rule successfully (i.e., succeed in switching between rules), compared to control experiments. Furthermore, by assessing two types of error animals made after the rule switch, we show that interfering with the positive feedback integration, but leaving the negative feedback processing intact, does not influence the initial disengagement from the first rule, but impedes the maintenance of the newly acquired response set. These findings support the role of prefrontal networks in mice for cognitive flexibility, which is impaired during numerous neuropsychiatric diseases, such as schizophrenia and depression.
Collapse
Affiliation(s)
- Szabolcs Biró
- Center for Brain Research, Division of Cognitive Neurobiology, Medical University of Vienna, Vienna, Austria
| | - Bálint Lasztóczi
- Center for Brain Research, Division of Cognitive Neurobiology, Medical University of Vienna, Vienna, Austria
| | - Thomas Klausberger
- Center for Brain Research, Division of Cognitive Neurobiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
77
|
Sumitomo A, Yukitake H, Hirai K, Horike K, Ueta K, Chung Y, Warabi E, Yanagawa T, Kitaoka S, Furuyashiki T, Narumiya S, Hirano T, Niwa M, Sibille E, Hikida T, Sakurai T, Ishizuka K, Sawa A, Tomoda T. Ulk2 controls cortical excitatory-inhibitory balance via autophagic regulation of p62 and GABAA receptor trafficking in pyramidal neurons. Hum Mol Genet 2019; 27:3165-3176. [PMID: 29893844 DOI: 10.1093/hmg/ddy219] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 06/04/2018] [Indexed: 01/21/2023] Open
Abstract
Autophagy plays an essential role in intracellular degradation and maintenance of cellular homeostasis in all cells, including neurons. Although a recent study reported a copy number variation of Ulk2, a gene essential for initiating autophagy, associated with a case of schizophrenia (SZ), it remains to be studied whether Ulk2 dysfunction could underlie the pathophysiology of the disease. Here we show that Ulk2 heterozygous (Ulk2+/-) mice have upregulated expression of sequestosome-1/p62, an autophagy-associated stress response protein, predominantly in pyramidal neurons of the prefrontal cortex (PFC), and exhibit behavioral deficits associated with the PFC functions, including attenuated sensorimotor gating and impaired cognition. Ulk2+/- neurons showed imbalanced excitatory-inhibitory neurotransmission, due in part to selective down-modulation of gamma-aminobutyric acid (GABA)A receptor surface expression in pyramidal neurons. Genetically reducing p62 gene dosage or suppressing p62 protein levels with an autophagy-inducing agent restored the GABAA receptor surface expression and rescued the behavioral deficits in Ulk2+/- mice. Moreover, expressing a short peptide that specifically interferes with the interaction of p62 and GABAA receptor-associated protein, a protein that regulates endocytic trafficking of GABAA receptors, also restored the GABAA receptor surface expression and rescued the behavioral deficits in Ulk2+/- mice. Thus, the current study reveals a novel mechanism linking deregulated autophagy to functional disturbances of the nervous system relevant to SZ, through regulation of GABAA receptor surface presentation in pyramidal neurons.
Collapse
Affiliation(s)
- Akiko Sumitomo
- Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Yukitake
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kazuko Hirai
- Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kouta Horike
- Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Keisho Ueta
- Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Youjin Chung
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eiji Warabi
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Toru Yanagawa
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Shiho Kitaoka
- CREST Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Pharmacology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Tomoyuki Furuyashiki
- CREST Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Pharmacology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Shuh Narumiya
- CREST Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoo Hirano
- Department of Biophysics, Kyoto University Graduate School of Science, Kyoto, Japan
| | - Minae Niwa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Takatoshi Hikida
- Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Sakurai
- Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koko Ishizuka
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Toshifumi Tomoda
- Department of Research and Drug Discovery, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
78
|
Paredes D, Morilak DA. A Rodent Model of Exposure Therapy: The Use of Fear Extinction as a Therapeutic Intervention for PTSD. Front Behav Neurosci 2019; 13:46. [PMID: 30914932 PMCID: PMC6421316 DOI: 10.3389/fnbeh.2019.00046] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/21/2019] [Indexed: 12/28/2022] Open
Abstract
The symptoms of post-traumatic stress disorder (PTSD) include cognitive impairment related to medial prefrontal cortical dysfunction. Indeed, a deficit of cognitive flexibility, i.e., an inability to modify previously learned thoughts and behaviors based on changes in the environment, may underlie many of the other symptoms of PTSD, such as changes in mood, hyper-arousal, intrusive thoughts, exaggerated and over-generalized fear, and avoidance behavior. Cognitive-behavioral therapies target the cognitive dysfunction observed in PTSD patients, training them to recalibrate stress-related perceptions, interpretations and responses. Preclinically, the extinction of conditioned fear bears resemblance to one form of cognitive therapy, exposure therapy, whereby an individual learns, through repeated exposure to a fear-provoking stimulus in a safe environment, that the stimulus no longer signals imminent threat, and their fear response is suppressed. In this review article, we highlight recent findings from our lab using fear extinction as a preclinical model of exposure therapy in rodents exposed to chronic unpredictable stress (CUS). We specifically focus on the therapeutic effects of extinction on stress-compromised set-shifting as a measure of cognitive flexibility, and active vs. passive coping behavior as a measure of avoidance. Finally, we discuss mechanisms involving activity and plasticity in the medial prefrontal cortex (mPFC) necessary for the therapeutic effects of extinction on cognitive flexibility and active coping.
Collapse
Affiliation(s)
- Denisse Paredes
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, San Antonio, TX, United States
| | - David A Morilak
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, San Antonio, TX, United States.,South Texas Veterans Health Care System (STVHCS), San Antonio, TX, United States
| |
Collapse
|
79
|
Bonnavion P, Fernández EP, Varin C, de Kerchove d’Exaerde A. It takes two to tango: Dorsal direct and indirect pathways orchestration of motor learning and behavioral flexibility. Neurochem Int 2019; 124:200-214. [DOI: 10.1016/j.neuint.2019.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/12/2018] [Accepted: 01/08/2019] [Indexed: 12/27/2022]
|
80
|
Cho WH, Park JC, Jeon WK, Cho J, Han JS. Superior Place Learning of C57BL/6 vs. DBA/2 Mice Following Prior Cued Learning in the Water Maze Depends on Prefrontal Cortical Subregions. Front Behav Neurosci 2019; 13:11. [PMID: 30760989 PMCID: PMC6361835 DOI: 10.3389/fnbeh.2019.00011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 01/15/2019] [Indexed: 01/02/2023] Open
Abstract
The participation of the prefrontal cortex (PFC), hippocampus, and dorsal striatum in switching the learning task from cued to place learning were examined in C57BL/6 and DBA/2 mice, by assessing changed levels of phosphorylated CREB (pCREB). Mice of both strains first received cued training in a water maze for 4 days (4 trials per day), and were then assigned to one of four groups, one with no place training, and three with different durations of place training (2, 4, or 8 days). Both strains showed equal performance in cued training. After the switch to place training, C57BL/6 mice with 2 or 4 days of training performed significantly better than DBA/2 mice, but their superiority disappeared during the second half of an 8 days-place training period. The pCREB levels of these mice were measured 30 min after place training and compared with those of mice that received only cued training. Changes in pCREB levels of C57BL/6 mice were greater in the hippocampal CA3, hippocampal dentate gyrus, orbitofrontal and medial PFC than those of DBA/2 mice, when mice of both received the switched place training for 2 days. We further investigated the roles of orbitofrontal and medial PFC among these brain regions showing strain differences, by destroying each region using selective neurotoxins. C57BL/6 mice with orbitofrontal lesions were slower to acquire the place learning and continued to use the cued search acquired during the cued training phase. These findings indicate that mouse orbitofrontal cortex (OFC) pCREB is associated with behavioral flexibility such as the ability to switch a learning task.
Collapse
Affiliation(s)
- Woo-Hyun Cho
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| | - Jung-Cheol Park
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| | - Won Kyung Jeon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon, South Korea.,Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Jeiwon Cho
- Department of Medical Science, College of Medicine, Catholic Kwandong University International St. Mary's Hospital, Incheon, South Korea.,Institute for Bio-Medical Convergence, Incheon St. Mary's Hospital, The Catholic University of Korea, Incheon, South Korea
| | - Jung-Soo Han
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| |
Collapse
|
81
|
Abstract
Executive functions (EFs) include high-order cognitive abilities such as working memory, inhibitory control, cognitive flexibility, planning, reasoning, and problem solving. EFs enable humans to achieve goals, adapt to novel everyday life situations, and manage social interactions. Traditionally EFs have been associated with frontal lobe functioning. More recent evidence shows that posterior and subcortical regions also play a crucial role in EF processing, especially in the integration of sensory information and emotion. This chapter reviews the variety of EFs and their neural underpinning, based on lesion mapping and neuroimaging studies, as well as the evidence for rehabilitation interventions, neuropsychological assessment based on standard and ecologically valid tests, development, and genetic predisposition for recovery of executive functions after brain injury. Taken together, the EFs are critical for unique human abilities and more careful analyses of their subcomponents may help the development of targeted translational interventions to improve them.
Collapse
|
82
|
Piantadosi PT, Lieberman AG, Pickens CL, Bergstrom HC, Holmes A. A novel multichoice touchscreen paradigm for assessing cognitive flexibility in mice. ACTA ACUST UNITED AC 2018; 26:24-30. [PMID: 30559117 PMCID: PMC6298539 DOI: 10.1101/lm.048264.118] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 11/09/2018] [Indexed: 01/16/2023]
Abstract
Cognitive flexibility refers to various processes which enable behaviors to be modified on the basis of a change in the contingencies between stimuli or responses and their associated outcomes. Reversal learning is a form of cognitive flexibility which measures the ability to adjust responding based on a switch in the stimulus–outcome contingencies of, typically two, perceptually distinct stimuli. Reversal tasks have provided valuable insight into the neural basis of cognitive flexibility, implicating brain regions including the lateral orbitofrontal cortex (lOFC) and dorsomedial prefrontal cortex (dmPFC). However, with two-stimulus reversal, it is difficult to determine whether response errors are due excessive perseveration, deficient learning, or other problems with updating. To address this limitation, we developed a mouse three-choice touchscreen-based visual reversal task, in which the contingencies of two stimuli were switched on reversal but a third, simultaneously presented, stimulus was never reinforced. We found that, in male C57BL/6J mice, responding at the previously rewarded stimulus predominated over the newly and never-reinforced stimuli during early reversal. Next, we showed that acute pharmacological inhibition of lOFC, but not dmPFC, impaired early reversal performance, relative to noninactivated controls. Interestingly, however, lOFC inactivation deficits were characterized by increased choice of the never-reinforced stimulus and a decrease in (perseverative-like) responding at the previously rewarded stimulus. These effects are inconsistent with the historical notion of lOFC mediating response inhibition and closer to recent views of the lOFC's role in response/outcome tracking. Overall, these findings provide initial support the utility of this novel paradigm for studying cognitive flexibility and its underlying neural substrates.
Collapse
Affiliation(s)
- Patrick T Piantadosi
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health, Bethesda, Maryland 20852, USA
| | - Abby G Lieberman
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health, Bethesda, Maryland 20852, USA
| | - Charles L Pickens
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health, Bethesda, Maryland 20852, USA
| | - Hadley C Bergstrom
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health, Bethesda, Maryland 20852, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health, Bethesda, Maryland 20852, USA
| |
Collapse
|
83
|
Jewell JS, Duncan VD, Fesshaye A, Tondin A, Macadat E, Britten RA. Exposure to ≤15 cGy of 600 MeV/n 56Fe Particles Impairs Rule Acquisition but not Long-Term Memory in the Attentional Set-Shifting Assay. Radiat Res 2018; 190:565-575. [DOI: 10.1667/rr15085.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Jessica S. Jewell
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Vania D. Duncan
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Arriyam Fesshaye
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Abigail Tondin
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Evangeline Macadat
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Richard A. Britten
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| |
Collapse
|
84
|
Soma S, Suematsu N, Yoshida J, Ríos A, Shimegi S. Discretion for behavioral selection affects development of habit formation after extended training in rats. Behav Processes 2018; 157:291-300. [PMID: 30366108 DOI: 10.1016/j.beproc.2018.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/27/2018] [Accepted: 10/22/2018] [Indexed: 01/01/2023]
Abstract
As training progresses, animals show a transition from goal-dependent behavior to goal-independent behavior (habitual responses). Habit formation is influenced by several factors, including the amount of training and action-outcome contingency. However, it remains unknown whether and how discretion for behavioral selection influences habit formation. To this end, we trained male rats in two types of two-alternative forced-choice task: visual association and nonvisual association tasks. In the first type of task, rats learned the association between reward and a visual cue, the position of which was randomly changed per trial so that rats had to make a judgmental decision about which choice delivered the reward in each trial (discreet judgment group); in the second type of task, the rats learned that a reward was delivered after either choice following task initiation (uncontrolled judgment group). To test the sensitivity to contingency manipulation, the extinction tests were conducted in short- and long-term trained groups, with the result that the overtrained rats in the uncontrolled judgment group, but not the other three groups, showed less sensitivity. To further investigate the reward sensitivity in the long-term trained groups from another perspective, we continuously and periodically altered the reward size for each trial. The rats of the discreet judgment group changed intertrial intervals depending on reward size, while this tendency was weaker in the uncontrolled judgment group. These results suggest that discreet judgment maintained goal-directed rat behavior, whereas uncontrolled judgment led to the development of habit-like behavior.
Collapse
Affiliation(s)
- Shogo Soma
- Graduate School of Medicine, Osaka University, Toyonaka, Osaka, 560-0043, Japan; Brain Science Institute, Tamagawa University, Machida, Tokyo, 194-8610, Japan.
| | - Naofumi Suematsu
- Graduate School of Medicine, Osaka University, Toyonaka, Osaka, 560-0043, Japan
| | - Junichi Yoshida
- Brain Science Institute, Tamagawa University, Machida, Tokyo, 194-8610, Japan
| | - Alain Ríos
- Brain Science Institute, Tamagawa University, Machida, Tokyo, 194-8610, Japan; Academia de Fisiología, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Colonia Casco de Santo Tomás, 11340, D.F., Mexico
| | - Satoshi Shimegi
- Graduate School of Medicine, Osaka University, Toyonaka, Osaka, 560-0043, Japan; Center for Education in Liberal Arts and Sciences, Osaka University, Toyonaka, Osaka, 560-0043, Japan
| |
Collapse
|
85
|
Miguel PM, Deniz BF, Deckmann I, Confortim HD, Diaz R, Laureano DP, Silveira PP, Pereira LO. Prefrontal cortex dysfunction in hypoxic-ischaemic encephalopathy contributes to executive function impairments in rats: Potential contribution for attention-deficit/hyperactivity disorder. World J Biol Psychiatry 2018; 19:547-560. [PMID: 28105895 DOI: 10.1080/15622975.2016.1273551] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The attention-deficit/hyperactivity disorder (ADHD) compromises the quality of life of individuals including adaptation to the social environment. ADHD aetiology includes perinatal conditions such as hypoxic-ischaemic events; preclinical studies have demonstrated attentional deficits and impulsive-hyperactive outcomes after neonatal hypoxic and/or ischaemic intervention, but data are missing to understand this relationship. Thus, the aim of this study was to evaluate executive function (EF) and impulsivity, and tissue integrity and dopaminergic function in the prefrontal cortex (PFC) of rats submitted to hypoxia-ischaemia (HI). METHODS At postnatal day (PND) 7, male Wistar rats were divided into control (n = 10) and HI groups (n = 11) and the HI procedure was conducted. At PND60, the animals were tested in the attentional set-shifting (ASS) task to EF and in the tolerance to delay of reward for assessment of impulsivity. After, morphological analysis and the dopaminergic system were evaluated in the PFC. RESULTS Animals subjected to HI had impairments in EF evidenced by a behavioural inflexibility that was correlated to PFC atrophy. Moreover, HI animals presented reduced D2 receptors in the ipsilateral side of ischaemia in the PFC. CONCLUSIONS Animals submitted to HI presented impaired EF associated with tissue atrophy and dopaminergic disturbance in the PFC.
Collapse
Affiliation(s)
- Patrícia Maidana Miguel
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Bruna Ferrary Deniz
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Iohanna Deckmann
- b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Heloísa Deola Confortim
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Ramiro Diaz
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Daniela Pereira Laureano
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Patrícia Pelufo Silveira
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,c Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,d Ludmer Centre for Neuroinformatics and Mental Health , Douglas Mental Health University Institute, McGill University , Montreal , QC , Canada
| | - Lenir Orlandi Pereira
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| |
Collapse
|
86
|
Infralimbic cortex is required for learning alternatives to prelimbic promoted associations through reciprocal connectivity. Nat Commun 2018; 9:2727. [PMID: 30006525 PMCID: PMC6045592 DOI: 10.1038/s41467-018-05318-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 06/25/2018] [Indexed: 11/30/2022] Open
Abstract
Prefrontal cortical areas mediate flexible adaptive control of behavior, but the specific contributions of individual areas and the circuit mechanisms through which they interact to modulate learning have remained poorly understood. Using viral tracing and pharmacogenetic techniques, we show that prelimbic (PreL) and infralimbic cortex (IL) exhibit reciprocal PreL↔IL layer 5/6 connectivity. In set-shifting tasks and in fear/extinction learning, activity in PreL is required during new learning to apply previously learned associations, whereas activity in IL is required to learn associations alternative to previous ones. IL→PreL connectivity is specifically required during IL-dependent learning, whereas reciprocal PreL↔IL connectivity is required during a time window of 12–14 h after association learning, to set up the role of IL in subsequent learning. Our results define specific and opposing roles of PreL and IL to together flexibly support new learning, and provide circuit evidence that IL-mediated learning of alternative associations depends on direct reciprocal PreL↔IL connectivity. Prelimbic (PL) and infralimbic (IL) cortical areas are known to have complementary roles in learning and decision making. Here the authors report reciprocal connectivity between the two areas and elucidate their functional impact on different aspects of learning.
Collapse
|
87
|
Girotti M, Adler SM, Bulin SE, Fucich EA, Paredes D, Morilak DA. Prefrontal cortex executive processes affected by stress in health and disease. Prog Neuropsychopharmacol Biol Psychiatry 2018; 85:161-179. [PMID: 28690203 PMCID: PMC5756532 DOI: 10.1016/j.pnpbp.2017.07.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/01/2017] [Accepted: 07/05/2017] [Indexed: 11/23/2022]
Abstract
Prefrontal cortical executive functions comprise a number of cognitive capabilities necessary for goal directed behavior and adaptation to a changing environment. Executive dysfunction that leads to maladaptive behavior and is a symptom of psychiatric pathology can be instigated or exacerbated by stress. In this review we survey research addressing the impact of stress on executive function, with specific focus on working memory, attention, response inhibition, and cognitive flexibility. We then consider the neurochemical pathways underlying these cognitive capabilities and, where known, how stress alters them. Finally, we review work exploring potential pharmacological and non-pharmacological approaches that can ameliorate deficits in executive function. Both preclinical and clinical literature indicates that chronic stress negatively affects executive function. Although some of the circuitry and neurochemical processes underlying executive function have been characterized, a great deal is still unknown regarding how stress affects these processes. Additional work focusing on this question is needed in order to make progress on developing interventions that ameliorate executive dysfunction.
Collapse
Affiliation(s)
- Milena Girotti
- Department of Pharmacology, Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA.
| | - Samantha M Adler
- Department of Pharmacology, Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - Sarah E Bulin
- Department of Pharmacology, Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - Elizabeth A Fucich
- Department of Pharmacology, Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - Denisse Paredes
- Department of Pharmacology, Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA
| | - David A Morilak
- Department of Pharmacology, Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA
| |
Collapse
|
88
|
Gray NE, Zweig JA, Caruso M, Martin MD, Zhu JY, Quinn JF, Soumyanath A. Centella asiatica increases hippocampal synaptic density and improves memory and executive function in aged mice. Brain Behav 2018; 8:e01024. [PMID: 29920983 PMCID: PMC6043711 DOI: 10.1002/brb3.1024] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Centella asiatica is a plant used for centuries to enhance memory. We have previously shown that a water extract of Centella asiatica (CAW) attenuates age-related spatial memory deficits in mice and improves neuronal health. Yet the effect of CAW on other cognitive domains remains unexplored as does its mechanism of improving age-related cognitive impairment. This study investigates the effects of CAW on a variety of cognitive tasks as well as on synaptic density and mitochondrial and antioxidant pathways. METHODS Twenty-month-old CB6F1 mice were treated with CAW (2 mg/ml) in their drinking water for 2 weeks prior to behavioral testing. Learning, memory, and executive function were assessed using the novel object recognition task (NORT), object location memory task (OLM), and odor discrimination reversal learning (ODRL) test. Tissue was collected for Golgi analysis of spine density as well as assessment of mitochondrial, antioxidant, and synaptic proteins. RESULTS CAW improved performance in all behavioral tests suggesting effects on hippocampal and cortical dependent memory as well as on prefrontal cortex mediated executive function. There was also an increase in synaptic density in the treated animals, which was accompanied by increased expression of the antioxidant response gene NRF2 as well as the mitochondrial marker porin. CONCLUSIONS These data show that CAW can increase synaptic density as well as antioxidant and mitochondrial proteins and improve multiple facets of age-related cognitive impairment. Because mitochondrial dysfunction and oxidative stress also accompany cognitive impairment in many pathological conditions this suggests a broad therapeutic utility of CAW.
Collapse
Affiliation(s)
- Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Jonathan A Zweig
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Marjoen D Martin
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Jennifer Y Zhu
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, Oregon
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
89
|
Tantra M, Guo L, Kim J, Zainolabidin N, Eulenburg V, Augustine GJ, Chen AI. Conditional deletion of Cadherin 13 perturbs Golgi cells and disrupts social and cognitive behaviors. GENES, BRAIN, AND BEHAVIOR 2018; 17:e12466. [PMID: 29446202 PMCID: PMC6635760 DOI: 10.1111/gbb.12466] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 12/21/2022]
Abstract
Inhibitory interneurons mediate the gating of synaptic transmission and modulate the activities of neural circuits. Disruption of the function of inhibitory networks in the forebrain is linked to impairment of social and cognitive behaviors, but the involvement of inhibitory interneurons in the cerebellum has not been assessed. We found that Cadherin 13 (Cdh13), a gene implicated in autism spectrum disorder and attention-deficit hyperactivity disorder, is specifically expressed in Golgi cells within the cerebellar cortex. To assess the function of Cdh13 and utilize the manipulation of Cdh13 expression in Golgi cells as an entry point to examine cerebellar-mediated function, we generated mice carrying Cdh13-floxed alleles and conditionally deleted Cdh13 with GlyT2::Cre mice. Loss of Cdh13 results in a decrease in the expression/localization of GAD67 and reduces spontaneous inhibitory postsynaptic current (IPSC) in cerebellar Golgi cells without disrupting spontaneous excitatory postsynaptic current (EPSC). At the behavioral level, loss of Cdh13 in the cerebellum, piriform cortex and endopiriform claustrum have no impact on gross motor coordination or general locomotor behaviors, but leads to deficits in cognitive and social abilities. Mice lacking Cdh13 exhibit reduced cognitive flexibility and loss of preference for contact region concomitant with increased reciprocal social interactions. Together, our findings show that Cdh13 is critical for inhibitory function of Golgi cells, and that GlyT2::Cre-mediated deletion of Cdh13 in non-executive centers of the brain, such as the cerebellum, may contribute to cognitive and social behavioral deficits linked to neurological disorders.
Collapse
Affiliation(s)
- M. Tantra
- School of Biological SciencesNanyang Technological University (NTU)Singapore
- School of Life SciencesUniversity of WarwickCoventryUK
| | - L. Guo
- School of Biological SciencesNanyang Technological University (NTU)Singapore
- School of Life SciencesUniversity of WarwickCoventryUK
| | - J. Kim
- Lee Kong Chian School of MedicineNanyang Technological University (NTU)Singapore
| | - N. Zainolabidin
- School of Biological SciencesNanyang Technological University (NTU)Singapore
- School of Life SciencesUniversity of WarwickCoventryUK
| | - V. Eulenburg
- Institute of BiochemistryFriedrich‐Alexander University Erlangen‐NurembergErlangenGermany
| | - G. J. Augustine
- Lee Kong Chian School of MedicineNanyang Technological University (NTU)Singapore
- Institute of Molecular and Cell BiologySingapore
| | - A. I. Chen
- School of Biological SciencesNanyang Technological University (NTU)Singapore
- School of Life SciencesUniversity of WarwickCoventryUK
- Institute of Molecular and Cell BiologySingapore
| |
Collapse
|
90
|
Tyler CRS, Smoake JJW, Solomon ER, Villicana E, Caldwell KK, Allan AM. Sex-Dependent Effects of the Histone Deacetylase Inhibitor, Sodium Valproate, on Reversal Learning After Developmental Arsenic Exposure. Front Genet 2018; 9:200. [PMID: 29963072 PMCID: PMC6013562 DOI: 10.3389/fgene.2018.00200] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/22/2018] [Indexed: 01/18/2023] Open
Abstract
Several studies have demonstrated that exposure to arsenic in drinking water adversely affects brain development and cognitive function in adulthood. While the mechanism by which arsenic induces adverse neurological outcomes remains elusive, studies suggest a link between reduced levels of histone acetylation and impaired performance on a variety of behavioral tasks following arsenic exposure. Using our developmental arsenic exposure (DAE) paradigm, we have previously reported reduced histone acetylation and associated histone acetyltransferase enzyme expression in the frontal cortex of C57BL/6J adult male mice, with no changes observed in the female frontal cortex. In the present study, we sought to determine if DAE produced sex-dependent deficits in frontal cortical executive function using the Y-maze acquisition and reversal learning tasks, which are specific for assessing cognitive flexibility. Further, we tested whether the administration of valproic acid, a class I-IIa histone deacetylase inhibitor, was able to mitigate behavioral and biochemical changes resulting from DAE. As anticipated, DAE inhibited acquisition and reversal learning performance in adult male, but not female, mice. Valproate treatment for 2 weeks restored reversal performance in the male arsenic-exposed offspring, while not affecting female performance. Protein levels of HDACs 1, 2, and 5 were elevated following behavioral assessment but only in DAE male mice; restoration of appropriate HDAC levels occurred after valproate treatment and was concurrent with improved behavioral performance, particularly during reversal learning. Female frontal cortical levels of HDAC enzymes were not impacted by DAE or valproate treatment. Finally, mRNA expression levels of brain-derived neurotrophic factor, Bdnf, which has been implicated in the control of frontal cortical flexibility and is regulated by HDAC5, were elevated in DAE male mice and restored to normal levels following HDACi treatment. Levels of mRNA encoding glutamate receptor ionotropic NMDA type subunits, which have been linked to cognitive flexibility, were not related to the reversal learning deficit in the DAE mice and were not altered by HDACi treatments. These findings demonstrate that DAE alters frontal cortical HDAC levels and Bdnf expression in males, but not females, and that these molecular changes are associated with sex-dependent differences in cognitive flexibility in a reversal-learning task.
Collapse
Affiliation(s)
| | - Jane J W Smoake
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Elizabeth R Solomon
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Estrella Villicana
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Kevin K Caldwell
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Andrea M Allan
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
91
|
Scheggia D, Mastrogiacomo R, Mereu M, Sannino S, Straub RE, Armando M, Managò F, Guadagna S, Piras F, Zhang F, Kleinman JE, Hyde TM, Kaalund SS, Pontillo M, Orso G, Caltagirone C, Borrelli E, De Luca MA, Vicari S, Weinberger DR, Spalletta G, Papaleo F. Variations in Dysbindin-1 are associated with cognitive response to antipsychotic drug treatment. Nat Commun 2018; 9:2265. [PMID: 29891954 PMCID: PMC5995960 DOI: 10.1038/s41467-018-04711-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/15/2018] [Indexed: 01/25/2023] Open
Abstract
Antipsychotics are the most widely used medications for the treatment of schizophrenia spectrum disorders. While such drugs generally ameliorate positive symptoms, clinical responses are highly variable in terms of negative symptoms and cognitive impairments. However, predictors of individual responses have been elusive. Here, we report a pharmacogenetic interaction related to a core cognitive dysfunction in patients with schizophrenia. We show that genetic variations reducing dysbindin-1 expression can identify individuals whose executive functions respond better to antipsychotic drugs, both in humans and in mice. Multilevel ex vivo and in vivo analyses in postmortem human brains and genetically modified mice demonstrate that such interaction between antipsychotics and dysbindin-1 is mediated by an imbalance between the short and long isoforms of dopamine D2 receptors, leading to enhanced presynaptic D2 function within the prefrontal cortex. These findings reveal one of the pharmacodynamic mechanisms underlying individual cognitive response to treatment in patients with schizophrenia, suggesting a potential approach for improving the use of antipsychotic drugs.
Collapse
Affiliation(s)
- Diego Scheggia
- Department of Neuroscience and Brain Technologies, Genetics of Cognition laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital Center Lausanne, Prilly-Lausanne, CH-1008, Switzerland
| | - Rosa Mastrogiacomo
- Department of Neuroscience and Brain Technologies, Genetics of Cognition laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Maddalena Mereu
- Department of Neuroscience and Brain Technologies, Genetics of Cognition laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
- Dipartimento di Scienze del Farmaco, Universita' degli Studi di Padova, Largo Meneghetti 2, 35131, Padova, Italy
| | - Sara Sannino
- Department of Neuroscience and Brain Technologies, Genetics of Cognition laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Richard E Straub
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, 21205, USA
| | - Marco Armando
- Department of Neuroscience, Bambino Gesù Children's Hospital, Piazza Sant'Onofrio 4, 00100, Rome, Italy
| | - Francesca Managò
- Department of Neuroscience and Brain Technologies, Genetics of Cognition laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Simone Guadagna
- Department of Neuroscience and Brain Technologies, Genetics of Cognition laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Fabrizio Piras
- IRCCS Santa Lucia Foundation, Neuropsychiatry Laboratory, 00179, Rome, Italy
| | - Fengyu Zhang
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, 21205, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, 21205, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, 21205, USA
| | - Sanne S Kaalund
- Research Laboratory for Stereology and Neuroscience, Bispebjerg University Hospital, 2400, Copenhagen, NV, Denmark
| | - Maria Pontillo
- Department of Neuroscience, Bambino Gesù Children's Hospital, Piazza Sant'Onofrio 4, 00100, Rome, Italy
| | - Genny Orso
- IRCCS E. Medea Scientific Institute, 23842, Bosisio Parini, Italy
| | - Carlo Caltagirone
- IRCCS Santa Lucia Foundation, Neuropsychiatry Laboratory, 00179, Rome, Italy
| | | | - Maria A De Luca
- Department of Biomedical Sciences, Università di Cagliari, 09124, Cagliari, Italy
| | - Stefano Vicari
- Department of Neuroscience, Bambino Gesù Children's Hospital, Piazza Sant'Onofrio 4, 00100, Rome, Italy
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, 21205, USA
- Departments of Psychiatry, Neurology, Neuroscience and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Gianfranco Spalletta
- IRCCS Santa Lucia Foundation, Neuropsychiatry Laboratory, 00179, Rome, Italy
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Francesco Papaleo
- Department of Neuroscience and Brain Technologies, Genetics of Cognition laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy.
| |
Collapse
|
92
|
Vandenberg A, Lin WC, Tai LH, Ron D, Wilbrecht L. Mice engineered to mimic a common Val66Met polymorphism in the BDNF gene show greater sensitivity to reversal in environmental contingencies. Dev Cogn Neurosci 2018; 34:34-41. [PMID: 29909248 PMCID: PMC6596311 DOI: 10.1016/j.dcn.2018.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/30/2018] [Accepted: 05/28/2018] [Indexed: 12/22/2022] Open
Abstract
A new line of mice,Val68Met, mimic human BDNF Val66Met polymorphism. New knock-in BDNF Met mice reverse more efficiently than Val in two separate tasks. Supports theory that BDNF Met allele confers greater sensitivity to the environment. Reversal performance can be dissociated from go/no-go and extinction performance. Phenotypes differ between newer and older BDNF Val66Met mouse models.
A common human polymorphism in the gene that encodes brain derived neurotrophic factor (BDNF), Val66Met, is considered a marker of vulnerability for mental health issues and has been associated with cognitive impairment. An alternate framework has been proposed in which “risk alleles” are reinterpreted as “plasticity alleles” that confer vulnerability in adverse environments and positive effects in neutral or positive environments (Belsky et al., 2009). These frameworks produce divergent predictions for tests of learning and cognitive flexibility. Here, we examined multiple aspects of learning and cognitive flexibility in a relatively new BDNF Val66Met mouse model (BDNF Val68Met, Warnault et al., 2016), including multiple choice discrimination and reversal, go/no-go learning and reversal, and appetitive extinction learning. We found that mice homozygous for the Met allele show more efficient reversal learning in two different paradigms, but learn at rates comparable to Val homozygotes on the multiple choice discrimination task, a go/no-go task, and in appetitive extinction. Our results dissociate reversal performance from go/no-go learning and appetitive extinction and support the plasticity allele framework that suggests BDNF Met carriers are potentially more sensitive to changes in the environment.
Collapse
Affiliation(s)
- Angela Vandenberg
- Neuroscience Graduate Program, University of California, San Francisco, CA, 94158, USA
| | - Wan Chen Lin
- Department of Psychology, University of California, Berkeley, CA, 94720, USA
| | - Lung-Hao Tai
- Department of Psychology, University of California, Berkeley, CA, 94720, USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Linda Wilbrecht
- Department of Psychology, University of California, Berkeley, CA, 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720 USA.
| |
Collapse
|
93
|
Ferguson BR, Gao WJ. PV Interneurons: Critical Regulators of E/I Balance for Prefrontal Cortex-Dependent Behavior and Psychiatric Disorders. Front Neural Circuits 2018; 12:37. [PMID: 29867371 PMCID: PMC5964203 DOI: 10.3389/fncir.2018.00037] [Citation(s) in RCA: 360] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/17/2018] [Indexed: 01/20/2023] Open
Abstract
Elucidating the prefrontal cortical microcircuit has been challenging, given its role in multiple complex behaviors, including working memory, cognitive flexibility, attention, social interaction and emotional regulation. Additionally, previous methodological limitations made it difficult to parse out the contribution of certain neuronal subpopulations in refining cortical representations. However, growing evidence supports a fundamental role of fast-spiking parvalbumin (PV) GABAergic interneurons in regulating pyramidal neuron activity to drive appropriate behavioral responses. Further, their function is heavily diminished in the prefrontal cortex (PFC) in numerous psychiatric diseases, including schizophrenia and autism. Previous research has demonstrated the importance of the optimal balance of excitation and inhibition (E/I) in cortical circuits in maintaining the efficiency of cortical information processing. Although we are still unraveling the mechanisms of information representation in the PFC, the E/I balance seems to be crucial, as pharmacological, chemogenetic and optogenetic approaches for disrupting E/I balance induce impairments in a range of PFC-dependent behaviors. In this review, we will explore two key hypotheses. First, PV interneurons are powerful regulators of E/I balance in the PFC, and help optimize the representation and processing of supramodal information in PFC. Second, diminishing the function of PV interneurons is sufficient to generate an elaborate symptom sequelae corresponding to those observed in a range of psychiatric diseases. Then, using this framework, we will speculate on whether this circuitry could represent a platform for the development of therapeutic interventions in disorders of PFC function.
Collapse
Affiliation(s)
- Brielle R Ferguson
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States.,Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, United States
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
94
|
Effects of monoamines on the intrinsic excitability of lateral orbitofrontal cortex neurons in alcohol-dependent and non-dependent female mice. Neuropharmacology 2018; 137:1-12. [PMID: 29689260 DOI: 10.1016/j.neuropharm.2018.04.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/10/2018] [Accepted: 04/19/2018] [Indexed: 11/20/2022]
Abstract
Changes in brain reward and control systems of frontal cortical areas including the orbitofrontal cortex (OFC) are associated with alcohol use disorders (AUD). The OFC is extensively innervated by monoamines, and drugs that target monoamine receptors have been used to treat a number of neuropsychiatric diseases, including AUDs. Recent findings from this laboratory demonstrate that D2, α2-adrenergic and 5HT1A receptors all decrease the intrinsic excitability of lateral OFC (lOFC) neurons in naïve male mice and that this effect is lost in mice exposed to repeated cycles of chronic intermittent ethanol (CIE) vapor. As biological sex differences may influence an individual's response to alcohol and contribute to the propensity to engage in addictive behaviors, we examined whether monoamines have similar effects on lOFC neurons in control and CIE exposed female mice. Dopamine, norepinephrine and serotonin all decreased spiking of lOFC neurons in naïve females via activation of Giα-coupled D2, α2-adrenergic and 5HT1A receptors, respectively. Firing was also inhibited by the direct GIRK channel activator ML297, while blocking these channels with barium eliminated the inhibitory actions of monoamines. Following CIE treatment, evoked spiking of lOFC neurons from female mice was significantly enhanced and monoamines and ML297 no longer inhibited firing. Unlike in male mice, the enhanced firing of neurons from CIE exposed female mice was not associated with changes in the after-hyperpolarization and the small-conductance potassium channel blocker apamin had no effect on current-evoked tail currents from either control or CIE exposed female mice. These results suggest that while CIE exposure alters monoamine regulation of OFC neuron firing similarly in males and female mice, there are sex-dependent differences in processes that regulate the intrinsic excitability of these neurons.
Collapse
|
95
|
Kőszeghy Á, Lasztóczi B, Forro T, Klausberger T. Spike-Timing of Orbitofrontal Neurons Is Synchronized With Breathing. Front Cell Neurosci 2018; 12:105. [PMID: 29731709 PMCID: PMC5920025 DOI: 10.3389/fncel.2018.00105] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/03/2018] [Indexed: 11/28/2022] Open
Abstract
The orbitofrontal cortex (OFC) has been implicated in a multiplicity of complex brain functions, including representations of expected outcome properties, post-decision confidence, momentary food-reward values, complex flavors and odors. As breathing rhythm has an influence on odor processing at primary olfactory areas, we tested the hypothesis that it may also influence neuronal activity in the OFC, a prefrontal area involved also in higher order processing of odors. We recorded spike timing of orbitofrontal neurons as well as local field potentials (LFPs) in awake, head-fixed mice, together with the breathing rhythm. We observed that a large majority of orbitofrontal neurons showed robust phase-coupling to breathing during immobility and running. The phase coupling of action potentials to breathing was significantly stronger in orbitofrontal neurons compared to cells in the medial prefrontal cortex. The characteristic synchronization of orbitofrontal neurons with breathing might provide a temporal framework for multi-variable processing of olfactory, gustatory and reward-value relationships.
Collapse
Affiliation(s)
- Áron Kőszeghy
- Division of Cognitive Neurobiology, Center for Brain Research, Medizinische Universität Wien, Vienna, Austria
| | - Bálint Lasztóczi
- Division of Cognitive Neurobiology, Center for Brain Research, Medizinische Universität Wien, Vienna, Austria
| | - Thomas Forro
- Division of Cognitive Neurobiology, Center for Brain Research, Medizinische Universität Wien, Vienna, Austria
| | - Thomas Klausberger
- Division of Cognitive Neurobiology, Center for Brain Research, Medizinische Universität Wien, Vienna, Austria
| |
Collapse
|
96
|
Marshall CA, King KM, Kortagere S. Limitations of the rat medial forebrain lesion model to study prefrontal cortex mediated cognitive tasks in Parkinson's disease. Brain Res 2018; 1702:105-113. [PMID: 29608880 DOI: 10.1016/j.brainres.2018.03.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 01/08/2023]
Abstract
Parkinson's Disease (PD) is a progressive movement disorder characterized by the loss of dopaminergic neurons in the midbrain. Besides motor impairment, PD patients exhibit non-motor symptoms that negatively impact their quality of life and often manifest prior to motor deficits. One such symptom is mild cognitive impairment (PD-MCI), which is comprised of deficits in executive function such as working memory, attention, cognitive flexibility, and spatial memory. The 6-hydroxydopamine (6-OHDA) induced unilateral medial forebrain bundle (MFB) lesion animal model successfully recapitulates PD motor impairment but is also used to assess non-motor deficits. The present study utilizes a unilateral 6-OHDA induced MFB lesion rodent model to investigate prefrontal cortex (PFC)-mediated cognitive processes that are impaired in PD patients. In a test of attentional set shifting, PD rodents demonstrated deficits in simple discrimination, but not in rule reversal or extradimensional shifts. PD rodents also exhibited deficits in a temporal order memory task but had no deficits in novel/spatial object recognition or object-in-place tasks. These results reveal limitations of the 6-OHDA induced unilateral MFB lesion model to completely recapitulate PD-MCI symptoms suggesting a need for better lesion models to study PD-MCI.
Collapse
Affiliation(s)
- Courtney A Marshall
- Department of Neurobiology and Anatomy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19129, United States
| | - Kirsten M King
- Department of Neurobiology and Anatomy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19129, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19129, United States.
| |
Collapse
|
97
|
Britten RA, Jewell JS, Duncan VD, Hadley MM, Macadat E, Musto AE, Tessa CL. Impaired Attentional Set-Shifting Performance after Exposure to 5 cGy of 600 MeV/n28Si Particles. Radiat Res 2018; 189:273-282. [DOI: 10.1667/rr14627.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Richard A. Britten
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507
- Leroy T Canoles Jr. Cancer Center, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Jessica S. Jewell
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Vania D. Duncan
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Melissa M. Hadley
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Evangeline Macadat
- Department of a Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Alberto E. Musto
- Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Chiara La Tessa
- NSRL Brookhaven National Laboratories, Upton, New York 11973
- University of Trento, Povo Trento 38122, Italy
| |
Collapse
|
98
|
Tait DS, Bowman EM, Neuwirth LS, Brown VJ. Assessment of intradimensional/extradimensional attentional set-shifting in rats. Neurosci Biobehav Rev 2018; 89:72-84. [PMID: 29474818 DOI: 10.1016/j.neubiorev.2018.02.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/06/2018] [Accepted: 02/19/2018] [Indexed: 01/07/2023]
Abstract
The rat intradimensional/extradimensional (ID/ED) task, first described by Birrell and Brown 18 years ago, has become the predominant means by which attentional set-shifting is investigated in rodents: the use of rats in the task has been described in over 135 publications by researchers from nearly 90 universities and pharmaceutical companies. There is variation in the protocols used by different groups, including differences in apparatus, stimuli (both stimulus dimensions and exemplars within), and also the methodology. Nevertheless, most of these variations seem to be of little consequence: there is remarkable similarity in the profile of published data, with consistency of learning rates and in the size and reliability of the set-shifting and reversal 'costs'. However, we suspect that there may be inconsistent data that is unpublished or perhaps 'failed experiments' that may have been caused by unintended deviations from effective protocols. The purpose of this review is to describe our approach and the rationale behind certain aspects of the protocol, including common pitfalls that are encountered when establishing an effective local protocol.
Collapse
Affiliation(s)
- David S Tait
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, South Street, St Andrews, Fife, KY16 9JP, UK.
| | - Eric M Bowman
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, South Street, St Andrews, Fife, KY16 9JP, UK
| | - Lorenz S Neuwirth
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, 11568, USA; SUNY Neuroscience Research Institute, Old Westbury, NY, 11568, USA
| | - Verity J Brown
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, South Street, St Andrews, Fife, KY16 9JP, UK
| |
Collapse
|
99
|
Page CE, Alexander J, Shepard R, Coutellier L. Npas4 deficiency interacts with adolescent stress to disrupt prefrontal GABAergic maturation and adult cognitive flexibility. GENES BRAIN AND BEHAVIOR 2018; 17:e12459. [DOI: 10.1111/gbb.12459] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/08/2017] [Accepted: 01/11/2018] [Indexed: 01/03/2023]
Affiliation(s)
- C. E. Page
- Department of Neuroscience; The Ohio State University; Columbus Ohio
| | - J. Alexander
- Department of Neuroscience; The Ohio State University; Columbus Ohio
| | - R. Shepard
- Department of Psychology; The Ohio State University; Columbus Ohio
| | - L. Coutellier
- Department of Neuroscience; The Ohio State University; Columbus Ohio
- Department of Psychology; The Ohio State University; Columbus Ohio
| |
Collapse
|
100
|
Roles of Prefrontal Cortex and Mediodorsal Thalamus in Task Engagement and Behavioral Flexibility. J Neurosci 2018; 38:2569-2578. [PMID: 29437889 DOI: 10.1523/jneurosci.1728-17.2018] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 01/19/2023] Open
Abstract
Behavioral tasks involving auditory cues activate inhibitory neurons within auditory cortex, leading to a reduction in the amplitude of auditory evoked response potentials (ERPs). One hypothesis is that this process, termed "task engagement," may enable context-dependent behaviors. Here we set out to determine (1) whether the medial prefrontal cortex (mPFC) plays a role in task engagement and (2) how task engagement relates to the context-dependent processing of auditory cues in male and female mice performing a decision-making task that can be guided by either auditory or visual cues. We found that, in addition to auditory ERP suppression, task engagement is associated with increased mPFC activity and an increase in theta band (4-7 Hz) synchronization between the mPFC and auditory cortex. Optogenetically inhibiting the mPFC eliminates the task engagement-induced auditory ERP suppression, while also preventing mice from switching between auditory and visual cue-based rules. However, mPFC inhibition, which eliminates task engagement-induced auditory ERP suppression, did not prevent mice from making decisions based on auditory cues. Furthermore, a more specific manipulation, selective disruption of mPFC outputs to the mediodorsal (MD) thalamus, is sufficient to prevent switching between auditory and visual rules but does not affect auditory ERPs. Based on these findings, we conclude that (1) the mPFC contributes to both task engagement and behavioral flexibility; (2) mPFC-MD projections are important for behavioral flexibility but not task engagement; and (3) task engagement, evidenced by the suppression of cortical responses to sensory input, is not required for sensory cue-guided decision making.SIGNIFICANCE STATEMENT When rodents perform choice-selection tasks based on sensory cues, neural responses to these cues are modulated compared with task-free conditions. Here we demonstrate that this phenomenon depends on the prefrontal cortex and thus represents a form of "top-down" regulation. However, we also show that this phenomenon is not critical for task performance, as rodents can make decisions based on specific sensory cues even when the task-dependent modulation of responses to those cues is abolished. Furthermore, disrupting one specific set of prefrontal outputs impairs rule switching but not the task-dependent modulation of sensory responses. These results show that the prefrontal cortex comprises multiple circuits that mediate dissociable functions related to behavioral flexibility and sensory processing.
Collapse
|