1
|
Rodriguez KA, Mattox N, Desme C, Hall LV, Wu Y, Pruden SM. Harnessing technology to measure individual differences in spatial thinking in early childhood from a relational developmental systems perspective. ADVANCES IN CHILD DEVELOPMENT AND BEHAVIOR 2024; 67:236-272. [PMID: 39260905 DOI: 10.1016/bs.acdb.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
According to the Relational Developmental Systems perspective, the development of individual differences in spatial thinking (e.g., mental rotation, spatial reorientation, and spatial language) are attributed to various psychological (e.g., children's cognitive strategies), biological (e.g., structure and function of hippocampus), and cultural systems (e.g., caregiver spatial language input). Yet, measuring the development of individual differences in spatial thinking in young children, as well as the psychological, biological, and cultural systems that influence the development of these abilities, presents unique challenges. The current paper outlines ways to harness available technology including eye-tracking, eye-blink conditioning, MRI, Zoom, and LENA technology, to study the development of individual differences in young children's spatial thinking. The technologies discussed offer ways to examine children's spatial thinking development from different levels of analyses (i.e., psychological, biological, cultural), thereby allowing us to advance the study of developmental theory. We conclude with a discussion of the use of artificial intelligence.
Collapse
Affiliation(s)
- Karinna A Rodriguez
- Florida International University, Department of Psychology, Miami, FL, United States.
| | - Nick Mattox
- Florida International University, Department of Psychology, Miami, FL, United States
| | - Carlos Desme
- Florida International University, Department of Psychology, Miami, FL, United States
| | - LaTreese V Hall
- Florida International University, Department of Psychology, Miami, FL, United States
| | - Yinbo Wu
- Florida International University, Department of Psychology, Miami, FL, United States
| | - Shannon M Pruden
- Florida International University, Department of Psychology, Miami, FL, United States
| |
Collapse
|
2
|
Tapp ZM, Cornelius S, Oberster A, Kumar JE, Atluri R, Witcher KG, Oliver B, Bray C, Velasquez J, Zhao F, Peng J, Sheridan J, Askwith C, Godbout JP, Kokiko-Cochran ON. Sleep fragmentation engages stress-responsive circuitry, enhances inflammation and compromises hippocampal function following traumatic brain injury. Exp Neurol 2022; 353:114058. [PMID: 35358498 PMCID: PMC9068267 DOI: 10.1016/j.expneurol.2022.114058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/04/2022] [Accepted: 03/24/2022] [Indexed: 02/08/2023]
Abstract
Traumatic brain injury (TBI) impairs the ability to restore homeostasis in response to stress, indicating hypothalamic-pituitary-adrenal (HPA)-axis dysfunction. Many stressors result in sleep disturbances, thus mechanical sleep fragmentation (SF) provides a physiologically relevant approach to study the effects of stress after injury. We hypothesize SF stress engages the dysregulated HPA-axis after TBI to exacerbate post-injury neuroinflammation and compromise recovery. To test this, male and female mice were given moderate lateral fluid percussion TBI or sham-injury and left undisturbed or exposed to daily, transient SF for 7- or 30-days post-injury (DPI). Post-TBI SF increases cortical expression of interferon- and stress-associated genes characterized by inhibition of the upstream regulator NR3C1 that encodes glucocorticoid receptor (GR). Moreover, post-TBI SF increases neuronal activity in the hippocampus, a key intersection of the stress-immune axes. By 30 DPI, TBI SF enhances cortical microgliosis and increases expression of pro-inflammatory glial signaling genes characterized by persistent inhibition of the NR3C1 upstream regulator. Within the hippocampus, post-TBI SF exaggerates microgliosis and decreases CA1 neuronal activity. Downstream of the hippocampus, post-injury SF suppresses neuronal activity in the hypothalamic paraventricular nucleus indicating decreased HPA-axis reactivity. Direct application of GR agonist, dexamethasone, to the CA1 at 30 DPI increases GR activity in TBI animals, but not sham animals, indicating differential GR-mediated hippocampal action. Electrophysiological assessment revealed TBI and SF induces deficits in Schaffer collateral long-term potentiation associated with impaired acquisition of trace fear conditioning, reflecting dorsal hippocampal-dependent cognitive deficits. Together these data demonstrate that post-injury SF engages the dysfunctional post-injury HPA-axis, enhances inflammation, and compromises hippocampal function. Therefore, external stressors that disrupt sleep have an integral role in mediating outcome after brain injury.
Collapse
Affiliation(s)
- Zoe M Tapp
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Sydney Cornelius
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Alexa Oberster
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA
| | - Julia E Kumar
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Ravitej Atluri
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Kristina G Witcher
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Braedan Oliver
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Chelsea Bray
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - John Velasquez
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Fangli Zhao
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Juan Peng
- Center for Biostatistics, The Ohio State University, 320-55 Lincoln Tower, 1800 Cannon Drive, Columbus, OH 43210, USA.
| | - John Sheridan
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA; Division of Biosciences, College of Dentistry, The Ohio State University, 305 W. 12(th) Ave, Columbus, OH 43210, USA.
| | - Candice Askwith
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA.
| | - Jonathan P Godbout
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Olga N Kokiko-Cochran
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, USA.
| |
Collapse
|
3
|
Staples MC, Herman MA, Lockner JW, Avchalumov Y, Kharidia KM, Janda KD, Roberto M, Mandyam CD. Isoxazole-9 reduces enhanced fear responses and retrieval in ethanol-dependent male rats. J Neurosci Res 2021; 99:3047-3065. [PMID: 34496069 PMCID: PMC10112848 DOI: 10.1002/jnr.24932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022]
Abstract
Plasticity in the dentate gyrus (DG) is strongly influenced by ethanol, and ethanol experience alters long-term memory consolidation dependent on the DG. However, it is unclear if DG plasticity plays a role in dysregulation of long-term memory consolidation during abstinence from chronic ethanol experience. Outbred male Wistar rats experienced 7 weeks of chronic intermittent ethanol vapor exposure (CIE). Seventy-two hours after CIE cessation, CIE and age-matched ethanol-naïve Air controls experienced auditory trace fear conditioning (TFC). Rats were tested for cue-mediated retrieval in the fear context either twenty-four hours (24 hr), ten days (10 days), or twenty-one days (21 days) later. CIE rats showed enhanced freezing behavior during TFC acquisition compared to Air rats. Air rats showed significant fear retrieval, and this behavior did not differ at the three time points. In CIE rats, fear retrieval increased over time during abstinence, indicating an incubation in fear responses. Enhanced retrieval at 21 days was associated with reduced structural and functional plasticity of ventral granule cell neurons (GCNs) and reduced expression of synaptic proteins important for neuronal plasticity. Systemic treatment with the drug Isoxazole-9 (Isx-9; small molecule that stimulates DG plasticity) during the last week and a half of CIE blocked altered acquisition and retrieval of fear memories in CIE rats during abstinence. Concurrently, Isx-9 modulated the structural and functional plasticity of ventral GCNs and the expression of synaptic proteins in the ventral DG. These findings identify that abstinence-induced disruption of fear memory consolidation occurs via altered plasticity within the ventral DG, and that Isx-9 prevented these effects.
Collapse
Affiliation(s)
| | - Melissa A. Herman
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jonathan W. Lockner
- Departments of Chemistry and Immunology, Scripps Research, La Jolla, CA, USA
| | | | | | - Kim D. Janda
- Departments of Chemistry and Immunology, Scripps Research, La Jolla, CA, USA
| | - Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, Scripps Research, La Jolla, CA, USA
| | - Chitra D. Mandyam
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
4
|
Rule L, Yang J, Watkin H, Hall J, Brydges NM. Environmental enrichment rescues survival and function of adult-born neurons following early life stress. Mol Psychiatry 2021; 26:1898-1908. [PMID: 32286496 DOI: 10.1038/s41380-020-0718-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/12/2020] [Accepted: 03/24/2020] [Indexed: 01/06/2023]
Abstract
Adverse experiences early in life are associated with the development of psychiatric illnesses. The hippocampus is likely to play pivotal role in generating these effects: it undergoes significant development during childhood and is extremely reactive to stress. In rodent models, stress in the pre-pubertal period impairs adult hippocampal neurogenesis (AHN) and behaviours which rely on this process. In normal adult animals, environmental enrichment (EE) is a potent promoter of AHN and hippocampal function. Whether exposure to EE during adolescence can restore normal hippocampal function and AHN following pre-pubertal stress (PPS) is unknown. We investigated EE as a treatment for reduced AHN and hippocampal function following PPS in a rodent model. Stress was administered between post-natal days (PND) 25-27, EE from PND 35 to early adulthood, when behavioural testing and assessment of AHN took place. PPS enhanced fear reactions to a conditioned stimulus (CS) following a trace fear protocol and reduced the survival of 4-week-old adult-born neurons throughout the adult hippocampus. Furthermore, we show that fewer adult-born neurons were active during recall of the CS stimulus following PPS. All effects were reversed by EE. Our results demonstrate lasting effects of PPS on the hippocampus and highlight the utility of EE during adolescence for restoring normal hippocampal function. EE during adolescence is a promising method of enhancing impaired hippocampal function resulting from early life stress, and due to multiple benefits (low cost, few side effects, widespread availability) should be more thoroughly explored as a treatment option in human sufferers of childhood adversity.
Collapse
Affiliation(s)
- Lowenna Rule
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Jessica Yang
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Holly Watkin
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.,MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Nichola Marie Brydges
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
5
|
Ehlers VL, Smies CW, Moyer JR. Apoaequorin differentially modulates fear memory in adult and aged rats. Brain Behav 2020; 10:e01832. [PMID: 32945630 PMCID: PMC7667302 DOI: 10.1002/brb3.1832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Cognitive deficits during aging are pervasive across species and learning paradigms. One of the major mechanisms thought to play a role in age-related memory decline is dysregulated calcium (Ca2+ ) homeostasis. Aging is associated with impaired function of several calcium-regulatory mechanisms, including calcium-binding proteins that normally support intracellular Ca2+ regulation. This age-related calcium-binding protein dysfunction and changes in expression lead to disrupted maintenance of intracellular Ca2+ , thus contributing to memory decline. Other work has found that age-related cognitive deficits can be mitigated by either blocking Ca2+ entry into the cytosol or preventing its release from intracellular Ca2+ stores. However, the effect of calcium-binding protein administration on cognitive function during aging is not well-understood. Our laboratory has previously shown that the calcium-binding protein apoaequorin (AQ) is neuroprotective during oxygen-glucose deprivation, a model of in vitro ischemia characterized by calcium-induced excitotoxicity. The current experiments assessed the effect of direct dorsal hippocampal AQ infusion on trace and context fear memory in adult and aged rats. METHODS Adult (3-6 months) and aged (22-26 months) male F344 rats were randomly assigned to different experimental infusion groups before undergoing trace fear conditioning and testing. In experiment 1, rats received bilateral dorsal hippocampal infusions of either vehicle or AQ (4% w/v) 24 hr before trace fear conditioning. In experiment 2, rats received bilateral dorsal hippocampal infusions of either vehicle or 4% AQ 1 hr before trace fear conditioning and 1 hr before testing. RESULTS Aged rats displayed impaired trace and context fear memory. While a single AQ infusion 24 hr before trace fear conditioning was insufficient to rescue age-related trace fear memory deficits, AQ infusion 1 hr before both conditioning and testing abolished age-related context fear memory deficits. CONCLUSIONS These results suggest that intrahippocampal infusion of AQ may reverse aging-related deficits in hippocampus-dependent context fear memory.
Collapse
Affiliation(s)
- Vanessa L Ehlers
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Chad W Smies
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - James R Moyer
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.,Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| |
Collapse
|
6
|
Yousuf H, Ehlers VL, Sehgal M, Song C, Moyer JR. Modulation of intrinsic excitability as a function of learning within the fear conditioning circuit. Neurobiol Learn Mem 2019; 167:107132. [PMID: 31821881 DOI: 10.1016/j.nlm.2019.107132] [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: 10/01/2019] [Accepted: 11/27/2019] [Indexed: 11/28/2022]
Abstract
Experience-dependent neuronal plasticity is a fundamental substrate of learning and memory. Intrinsic excitability is a form of neuronal plasticity that can be altered by learning and indicates the pattern of neuronal responding to external stimuli (e.g. a learning or synaptic event). Associative fear conditioning is one form of learning that alters intrinsic excitability, reflecting an experience-dependent change in neuronal function. After fear conditioning, intrinsic excitability changes are evident in brain regions that are a critical part of the fear circuit, including the amygdala, hippocampus, retrosplenial cortex, and prefrontal cortex. Some of these changes are transient and/or reversed by extinction as well as learning-specific (i.e. they are not observed in neurons from control animals). This review will explore how intrinsic neuronal excitability changes within brain structures that are critical for fear learning, and it will also discuss evidence promoting intrinsic excitability as a vital mechanism of associative fear memories. This work has raised interesting questions regarding the role of fear learning in changes of intrinsic excitability within specific subpopulations of neurons, including those that express immediate early genes and thus demonstrate experience-dependent activity, as well as in neurons classified as having a specific firing type (e.g. burst-spiking vs. regular-spiking). These findings have interesting implications for how intrinsic excitability can serve as a neural substrate of learning and memory, and suggest that intrinsic plasticity within specific subpopulations of neurons may promote consolidation of the memory trace in a flexible and efficient manner.
Collapse
Affiliation(s)
- Hanna Yousuf
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - Vanessa L Ehlers
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - Megha Sehgal
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - Chenghui Song
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - James R Moyer
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA; Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA.
| |
Collapse
|
7
|
Brydges NM, Moon A, Rule L, Watkin H, Thomas KL, Hall J. Sex specific effects of pre-pubertal stress on hippocampal neurogenesis and behaviour. Transl Psychiatry 2018; 8:271. [PMID: 30531788 PMCID: PMC6288078 DOI: 10.1038/s41398-018-0322-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022] Open
Abstract
Experience of traumatic events in childhood is linked to an elevated risk of developing psychiatric disorders in adulthood. The neurobiological mechanisms underlying this phenomenon are not fully understood. The limbic system, particularly the hippocampus, is significantly impacted by childhood trauma. In particular, it has been hypothesised that childhood stress may impact adult hippocampal neurogenesis (AHN) and related behaviours, conferring increased risk for later mental illness. Stress in utero can lead to impaired hippocampal synaptic plasticity, and stress in the first 2-3 weeks of life reduces AHN in animal models. Less is known about the effects of stress in the post-weaning, pre-pubertal phase, a developmental time-point more akin to human childhood. Therefore, we investigated persistent effects of pre-pubertal stress (PPS) on functional and molecular aspects of the hippocampus. AHN was altered following PPS in male rats only. Specifically males showed reduced production of new neurons following PPS, but increased survival in the ventral dentate gyrus. In adult males, but not females, pattern separation and trace fear conditioning, behaviours that rely heavily on AHN, were also impaired after PPS. PPS also increased the expression of parvalbumin-positive GABAergic interneurons in the ventral dentate gyrus and increased glutamic acid decarboxylase 67 expression in the ventral hilus, in males only. Our results demonstrate the lasting effects of PPS on the hippocampus in a sex- and time-dependent manner, provide a potential mechanistic link between PPS and later behavioural impairments, and highlight sex differences in vulnerability to neuropsychiatric conditions after early-life stress.
Collapse
Affiliation(s)
- Nichola Marie Brydges
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| | - Anna Moon
- 0000 0001 0807 5670grid.5600.3Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK
| | - Lowenna Rule
- 0000 0001 0807 5670grid.5600.3Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK
| | - Holly Watkin
- 0000 0001 0807 5670grid.5600.3Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK
| | - Kerrie L. Thomas
- 0000 0001 0807 5670grid.5600.3Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK ,0000 0001 0807 5670grid.5600.3School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX UK
| | - Jeremy Hall
- 0000 0001 0807 5670grid.5600.3Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK ,0000 0001 0807 5670grid.5600.3MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ UK
| |
Collapse
|
8
|
Pilkiw M, Takehara-Nishiuchi K. Neural representations of time-linked memory. Neurobiol Learn Mem 2018; 153:57-70. [PMID: 29614377 DOI: 10.1016/j.nlm.2018.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 10/17/2022]
Abstract
Many cognitive processes, such as episodic memory and decision making, rely on the ability to form associations between two events that occur separately in time. The formation of such temporal associations depends on neural representations of three types of information: what has been presented (trace holding), what will follow (temporal expectation), and when the following event will occur (explicit timing). The present review seeks to link these representations with firing patterns of single neurons recorded while rodents and non-human primates associate stimuli, outcomes, and motor responses over time intervals. Across these studies, two distinct firing patterns were observed in the hippocampus, neocortex, and striatum: some neurons change firing rates during or shortly after the stimulus presentation and sustain the firing rate stably or sidlingly during the subsequent intervals (tonic firings). Other neurons transiently change firing rates during a specific moment within the time intervals (phasic firings), and as a group, they form a sequential firing pattern that covers the entire interval. Clever task designs used in some of these studies collectively provide evidence that both tonic and phasic firing responses represent trace holding, temporal expectation, and explicit timing. Subsequently, we applied machine-learning based classification approaches to the two firing patterns within the same dataset collected from rat medial prefrontal cortex during trace eyeblink conditioning. This quantitative analysis revealed that phasic-firing patterns showed greater selectivity for stimulus identity and temporal position than tonic-firing patterns. Our summary illuminates distributed neural representations of temporal association in the forebrain and generates several ideas for future investigations.
Collapse
Affiliation(s)
- Maryna Pilkiw
- Department of Cell and Systems Biology, University of Toronto, Toronto M5S 3G3, Canada
| | - Kaori Takehara-Nishiuchi
- Department of Cell and Systems Biology, University of Toronto, Toronto M5S 3G3, Canada; Department of Psychology, University of Toronto, Toronto M5S 3G3, Canada; Neuroscience Program, University of Toronto, Toronto M5S 3G3, Canada.
| |
Collapse
|
9
|
Reboreda A, Theissen FM, Valero-Aracama MJ, Arboit A, Corbu MA, Yoshida M. Do TRPC channels support working memory? Comparing modulations of TRPC channels and working memory through G-protein coupled receptors and neuromodulators. Behav Brain Res 2018; 354:64-83. [PMID: 29501506 DOI: 10.1016/j.bbr.2018.02.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
Working memory is a crucial ability we use in daily life. However, the cellular mechanisms supporting working memory still remain largely unclear. A key component of working memory is persistent neural firing which is believed to serve short-term (hundreds of milliseconds up to tens of seconds) maintenance of necessary information. In this review, we will focus on the role of transient receptor potential canonical (TRPC) channels as a mechanism underlying persistent firing. Many years of in vitro work have been suggesting a crucial role of TRPC channels in working memory and temporal association tasks. If TRPC channels are indeed a central mechanism for working memory, manipulations which impair or facilitate working memory should have a similar effect on TRPC channel modulation. However, modulations of working memory and TRPC channels were never systematically compared, and it remains unanswered whether TRPC channels indeed contribute to working memory in vivo or not. In this article, we review the effects of G-protein coupled receptors (GPCR) and neuromodulators, including acetylcholine, noradrenalin, serotonin and dopamine, on working memory and TRPC channels. Based on comparisons, we argue that GPCR and downstream signaling pathways that activate TRPC, generally support working memory, while those that suppress TRPC channels impair it. However, depending on the channel types, areas, and systems tested, this is not the case in all studies. Further work to clarify involvement of specific TRPC channels in working memory tasks and how they are affected by neuromodulators is still necessary in the future.
Collapse
Affiliation(s)
- Antonio Reboreda
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany.
| | - Frederik M Theissen
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Maria J Valero-Aracama
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 17, 91054 Erlangen, Germany
| | - Alberto Arboit
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Mihaela A Corbu
- Ruhr University Bochum (RUB), Universitätsstraße 150, 44801, Bochum, Germany
| | - Motoharu Yoshida
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany; Center for Behavioral Brain Sciences, 39106, Magdeburg, Germany.
| |
Collapse
|
10
|
Carvalho MC, Veloni AC, Genaro K, Brandão ML. Behavioral sensitization induced by dorsal periaqueductal gray electrical stimulation is counteracted by NK1 receptor antagonism in the ventral hippocampus and central nucleus of the amygdala. Neurobiol Learn Mem 2018. [PMID: 29519453 DOI: 10.1016/j.nlm.2018.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A single threatening experience may change the behavior of an animal in a long-lasting way and elicit generalized behavioral responses to a novel threatening situation that is unrelated to the original aversive experience. Electrical stimulation (ES) of the dorsal periaqueductal gray (dPAG) produces a range of defensive reactions, characterized by freezing, escape, and post-stimulation freezing (PSF). The latter reflects the processing of ascending aversive information to prosencephalic structures, including the central nucleus of the amygdala (CeA), which allows the animal to evaluate the consequences of the aversive situation. This process is modulated by substance P (SP) and its preferred receptor, neurokinin 1 (NK1). The ventral hippocampus (VH) has been associated with the processing of aversive information and expression of emotional reactions with negative valence, but the participation of the VH in the expression of these defensive responses has not been investigated. The VH is rich in NK1 receptor expression and has a high density of SP-containing fibers. The present study examined the role of NK1 receptors in the VH in the expression of defensive responses and behavioral sensitization that were induced by dPAG-ES. Rats were implanted with an electrode in the dPAG for ES, and a cannula was implanted in the VH or CeA for injections of vehicle (phosphate-buffered saline) or the NK1 receptor antagonist spantide (100 pmol/0.2 μL. Spantide reduced the duration of PSF that was evoked by dPAG-ES, without changing the aversive freezing or escape thresholds. One and 7 days later, exploratory behavior was evaluated in independent groups of rats in the elevated plus maze (EPM). dPAG-ES in rats that received vehicle caused higher aversion to the open arms of the EPM compared with rats that did not receive dPAG stimulation at both time intervals. Injections of spantide in the VH or CeA prevented the proaversive effects of dPAG-ES in the EPM only 1 day later. These findings suggest that NK1 receptors are activated in both the VH and CeA during the processing of aversive information that derives from dPAG-ES. As previously shown for the CeA, SP/NK1 receptors in the VH are recruited during PSF that is evoked by dPAG-ES, suggesting that a 24-h time window is susceptible to interventions with NK1 antagonists that block the passage of aversive information from the dPAG to higher brain areas.
Collapse
Affiliation(s)
- M C Carvalho
- Departamento de Psicologia, Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, SP, Brazil; Instituto de Neurociências e Comportamento, INeC, Ribeirão Preto, SP, Brazil.
| | - A C Veloni
- Departamento de Psicologia, Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, SP, Brazil; Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, SP, Brazil
| | - K Genaro
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, SP, Brazil; Instituto de Neurociências e Comportamento, INeC, Ribeirão Preto, SP, Brazil
| | - M L Brandão
- Instituto de Neurociências e Comportamento, INeC, Ribeirão Preto, SP, Brazil
| |
Collapse
|
11
|
Marks WN, Kalynchuk LE. Repeated corticosterone enhances the acquisition and recall of trace fear conditioning. Physiol Behav 2017; 182:40-45. [DOI: 10.1016/j.physbeh.2017.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/18/2017] [Accepted: 09/22/2017] [Indexed: 12/27/2022]
|
12
|
Witkin JM, Li J, Gilmour G, Mitchell SN, Carter G, Gleason SD, Seidel WF, Eastwood BJ, McCarthy A, Porter WJ, Reel J, Gardinier KM, Kato AS, Wafford KA. Electroencephalographic, cognitive, and neurochemical effects of LY3130481 (CERC-611), a selective antagonist of TARP-γ8-associated AMPA receptors. Neuropharmacology 2017; 126:257-270. [PMID: 28757050 DOI: 10.1016/j.neuropharm.2017.07.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/27/2017] [Accepted: 07/26/2017] [Indexed: 11/18/2022]
Abstract
6-[(1S)-1-[1-[5-(2-hydroxyethoxy)-2-pyridyl]pyrazol-3-yl]ethyl]-3H-1,3-benzothiazol-2-one (LY3130481 or CERC-611) is a selective antagonist of AMPA receptors containing transmembrane AMPA receptor regulatory protein (TARP) γ-8. This molecule has been characterized as a potent and efficacious anticonvulsant in an array of acute and chronic epilepsy models in rodents. The present set of experiments was designed to assess the effects of LY3130481 on the electroencephelogram (EEG), cognitive function, and neurochemical outflow. LY3130481 disrupted food-maintained responding in rats and spontaneous alternation in a Y-maze in mice. In rat fear conditioning, LY3130481 caused a deficit in trace (hippocampal-dependent), but not in delay fear conditioning. Although these effects on cognitive performances were observed, the known cognitive-impairing anticonvulsant, topiramate, did not always produce deficits under these assay conditions. LY3130481 produced modest increases in wake times in rats. In addition, LY3130481 was able to attenuate some impairing effects of standard antiepileptic drugs. The motor-impairing effects of the lacosamide were attenuated by LY3130481 as was the decrease in non-rapid-eye movement sleep induced by carbamazepine. Evaluation of the effect of LY3130481 on neurotransmitter and metabolite efflux in the rat medial prefrontal cortex, using in vivo microdialysis, revealed significant increases in the pro-cognitive and wake-promoting neurotransmitters, histamine and acetylcholine, as well as in serotonin, telemethylhistamine, 5-HIAA, HVA and MHPG. LY3130481 thus presents a novel behavioral profile that will have to be evaluated in patients to fully appreciate its implications for therapeutics. LY3130481 is currently under clinical development as CERC-611 as an antiepileptic.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA.
| | - Jennifer Li
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Gary Gilmour
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Stephen N Mitchell
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Guy Carter
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Scott D Gleason
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Wesley F Seidel
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Brian J Eastwood
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Andrew McCarthy
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK
| | - Warren J Porter
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jon Reel
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Kevin M Gardinier
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Akihiko S Kato
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Keith A Wafford
- Lilly Research Laboratories, Eli Lilly and Company, Windlesham, Surrey, UK.
| |
Collapse
|
13
|
Enhanced theta synchronization correlates with the successful retrieval of trace fear memory. Biochem Biophys Res Commun 2016; 480:608-614. [DOI: 10.1016/j.bbrc.2016.10.101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/25/2016] [Indexed: 11/19/2022]
|
14
|
Connor DA, Gould TJ. The role of working memory and declarative memory in trace conditioning. Neurobiol Learn Mem 2016; 134 Pt B:193-209. [PMID: 27422017 PMCID: PMC5755400 DOI: 10.1016/j.nlm.2016.07.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 07/07/2016] [Accepted: 07/11/2016] [Indexed: 01/18/2023]
Abstract
Translational assays of cognition that are similarly implemented in both lower and higher-order species, such as rodents and primates, provide a means to reconcile preclinical modeling of psychiatric neuropathology and clinical research. To this end, Pavlovian conditioning has provided a useful tool for investigating cognitive processes in both lab animal models and humans. This review focuses on trace conditioning, a form of Pavlovian conditioning typified by the insertion of a temporal gap (i.e., trace interval) between presentations of a conditioned stimulus (CS) and an unconditioned stimulus (US). This review aims to discuss pre-clinical and clinical work investigating the mnemonic processes recruited for trace conditioning. Much work suggests that trace conditioning involves unique neurocognitive mechanisms to facilitate formation of trace memories in contrast to standard Pavlovian conditioning. For example, the hippocampus and prefrontal cortex (PFC) appear to play critical roles in trace conditioning. Moreover, cognitive mechanistic accounts in human studies suggest that working memory and declarative memory processes are engaged to facilitate formation of trace memories. The aim of this review is to integrate cognitive and neurobiological accounts of trace conditioning from preclinical and clinical studies to examine involvement of working and declarative memory.
Collapse
Affiliation(s)
- David A Connor
- Department of Psychology, Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Thomas J Gould
- Department of Psychology, Neuroscience Program, Temple University, Philadelphia, PA 19122, United States.
| |
Collapse
|
15
|
Koh MT, Shao Y, Sherwood A, Smith DR. Impaired hippocampal-dependent memory and reduced parvalbumin-positive interneurons in a ketamine mouse model of schizophrenia. Schizophr Res 2016; 171:187-94. [PMID: 26811256 PMCID: PMC4762714 DOI: 10.1016/j.schres.2016.01.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/09/2016] [Accepted: 01/11/2016] [Indexed: 01/23/2023]
Abstract
The hippocampus of patients with schizophrenia displays aberrant excess neuronal activity which affects cognitive function. Animal models of the illness have recapitulated the overactivity in the hippocampus, with a corresponding regionally localized reduction of inhibitory interneurons, consistent with that observed in patients. To better understand whether cognitive function is similarly affected in these models of hippocampal overactivity, we tested a ketamine mouse model of schizophrenia for cognitive performance in hippocampal- and medial prefrontal cortex (mPFC)-dependent tasks. We found that adult mice exposed to ketamine during adolescence were impaired on a trace fear conditioning protocol that relies on the integrity of the hippocampus. Conversely, the performance of the mice was normal on a delayed response task that is sensitive to mPFC damage. We confirmed that ketamine-exposed mice had reduced parvalbumin-positive interneurons in the hippocampus, specifically in the CA1, but not in the mPFC in keeping with the behavioral findings. These results strengthened the utility of the ketamine model for preclinical investigations of hippocampal overactivity in schizophrenia.
Collapse
Affiliation(s)
- Ming Teng Koh
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA.
| | | | | | | |
Collapse
|
16
|
Deal AL, Erickson KJ, Shiers SI, Burman MA. Limbic system development underlies the emergence of classical fear conditioning during the third and fourth weeks of life in the rat. Behav Neurosci 2016; 130:212-30. [PMID: 26820587 DOI: 10.1037/bne0000130] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Classical fear conditioning creates an association between an aversive stimulus and a neutral stimulus. Although the requisite neural circuitry is well understood in mature organisms, the development of these circuits is less well studied. The current experiments examine the ontogeny of fear conditioning and relate it to neuronal activation assessed through immediate early gene (IEG) expression in the amygdala, hippocampus, perirhinal cortex, and hypothalamus of periweanling rats. Rat pups were fear conditioned, or not, during the third or fourth weeks of life. Neuronal activation was assessed by quantifying expression of FBJ osteosarcoma oncogene (FOS) using immunohistochemistry (IHC) in Experiment 1. Fos and early growth response gene-1 (EGR1) expression was assessed using qRT-PCR in Experiment 2. Behavioral data confirm that both auditory and contextual fear continue to emerge between PD 17 and 24. The IEG expression data are highly consistent with these behavioral results. IHC results demonstrate significantly more FOS protein expression in the basal amygdala of fear-conditioned PD 23 subjects compared to control subjects, but no significant difference at PD 17. qRT-PCR results suggest specific activation of the amygdala only in older subjects during auditory fear expression. A similar effect of age and conditioning status was also observed in the perirhinal cortex during both contextual and auditory fear expression. Overall, the development of fear conditioning occurring between the third and fourth weeks of life appears to be at least partly attributable to changes in activation of the amygdala and perirhinal cortex during fear conditioning or expression. (PsycINFO Database Record
Collapse
|
17
|
Reichelt AC, Maniam J, Westbrook RF, Morris MJ. Dietary-induced obesity disrupts trace fear conditioning and decreases hippocampal reelin expression. Brain Behav Immun 2015; 43:68-75. [PMID: 25043993 DOI: 10.1016/j.bbi.2014.07.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 06/25/2014] [Accepted: 07/08/2014] [Indexed: 12/23/2022] Open
Abstract
Both obesity and over-consumption of palatable high fat/high sugar "cafeteria" diets in rats has been shown to induce cognitive deficits in executive function, attention and spatial memory. Adult male Sprague-Dawley rats were fed a diet that supplemented standard lab chow with a range of palatable foods eaten by people for 8 weeks, or regular lab chow. Memory was assessed using a trace fear conditioning procedure, whereby a conditioned stimulus (CS) is presented for 10s and then 30s after its termination a foot shock (US) is delivered. We assessed freezing to the CS (flashing light) in a neutral context, and freezing in the context associated with footshock. A dissociation was observed between levels of freezing in the context and to the CS associated with footshock. Cafeteria diet fed rats froze less than control chow fed rats in the context associated with footshock (P<0.01), indicating that encoding of a hippocampus-dependent context representation was impaired in these rats. Conversely, cafeteria diet fed rats froze more (P<0.05) to the CS than chow fed rats, suggesting that when hippocampal function was compromised the cue was the best predictor of footshock, as contextual information was not encoded. Dorsal hippocampal mRNA expression of inflammatory and neuroplasticity markers was analysed at the end of the experiment, 10 weeks of diet. Of these, mRNA expression of reelin, which is known to be important in long term potentiation and neuronal plasticity, was significantly reduced in cafeteria diet fed rats (P=0.003). This implicates reductions in hippocampal plasticity in the contextual fear memory deficits seen in the cafeteria diet fed rats.
Collapse
Affiliation(s)
- Amy C Reichelt
- School of Medical Sciences, The University of New South Wales, Sydney, Australia; School of Psychology, The University of New South Wales, Sydney, Australia
| | - Jayanthi Maniam
- School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | | | - Margaret J Morris
- School of Medical Sciences, The University of New South Wales, Sydney, Australia.
| |
Collapse
|
18
|
Burman MA, Erickson KJ, Deal AL, Jacobson RE. Contextual and auditory fear conditioning continue to emerge during the periweaning period in rats. PLoS One 2014; 9:e100807. [PMID: 24977415 PMCID: PMC4076234 DOI: 10.1371/journal.pone.0100807] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/29/2014] [Indexed: 01/19/2023] Open
Abstract
Anxiety disorders often emerge during childhood. Rodent models using classical fear conditioning have shown that different types of fear depend upon different neural structures and may emerge at different stages of development. For example, some work has suggested that contextual fear conditioning generally emerges later in development (postnatal day 23–24) than explicitly cued fear conditioning (postnatal day 15–17) in rats. This has been attributed to an inability of younger subjects to form a representation of the context due to an immature hippocampus. However, evidence that contextual fear can be observed in postnatal day 17 subjects and that cued fear conditioning continues to emerge past this age raises questions about the nature of this deficit. The current studies examine this question using both the context pre-exposure facilitation effect for immediate single-shock contextual fear conditioning and traditional cued fear conditioning using Sprague-Dawley rats. The data suggest that both cued and contextual fear conditioning are continuing to develop between PD 17 and 24, consistent with development occurring the in essential fear conditioning circuit.
Collapse
Affiliation(s)
- Michael A. Burman
- Department of Psychology, Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States of America
- * E-mail:
| | - Kristen J. Erickson
- Department of Psychology, Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States of America
| | - Alex L. Deal
- Department of Psychology, Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States of America
| | - Rose E. Jacobson
- Department of Biology, University of New England, Biddeford, Maine, United States of America
| |
Collapse
|
19
|
Greenberg A, Ward-Flanagan R, Dickson CT, Treit D. ANI inactivation: Unconditioned anxiolytic effects of anisomycin in the ventral hippocampus. Hippocampus 2014; 24:1308-16. [DOI: 10.1002/hipo.22312] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2014] [Indexed: 12/22/2022]
Affiliation(s)
| | | | - Clayton T. Dickson
- Centre for Neuroscience; University of Alberta; Edmonton Alberta
- Department of Psychology; University of Alberta; Edmonton Alberta
- Department of Physiology; University of Alberta; Edmonton Alberta
| | - Dallas Treit
- Centre for Neuroscience; University of Alberta; Edmonton Alberta
- Department of Psychology; University of Alberta; Edmonton Alberta
| |
Collapse
|
20
|
Cornelisse S, van Ast VA, Joëls M, Kindt M. Delayed effects of cortisol enhance fear memory of trace conditioning. Psychoneuroendocrinology 2014; 40:257-68. [PMID: 24485497 DOI: 10.1016/j.psyneuen.2013.11.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 10/31/2013] [Accepted: 11/18/2013] [Indexed: 12/18/2022]
Abstract
Corticosteroids induce rapid non-genomic effects followed by slower genomic effects that are thought to modulate cognitive function in opposite and complementary ways. It is presently unknown how these time-dependent effects of cortisol affect fear memory of delay and trace conditioning. This distinction is of special interest because the neural substrates underlying these types of conditioning may be differently affected by time-dependent cortisol effects. Delay conditioning is predominantly amygdala-dependent, while trace conditioning additionally requires the hippocampus. Here, we manipulated the timing of cortisol action during acquisition of delay and trace fear conditioning, by randomly assigning 63 men to one of three possible groups: (1) receiving 10mg hydrocortisone 240 min (slow cort) or (2) 60 min (rapid cort) before delay and trace acquisition, or (3) placebo at both times, in a double-blind design. The next day, we tested memory for trace and delay conditioning. Fear potentiated startle responses, skin conductance responses and unconditioned stimulus expectancy scores were measured throughout the experiment. The fear potentiated startle data show that cortisol intake 240 min before actual fear acquisition (slow cort) uniquely strengthened subsequent trace conditioned memory. No effects of cortisol delivery 60 min prior to fear acquisition were found on any measure of fear memory. Our findings emphasize that slow, presumably genomic, but not more rapid effects of corticosteroids enhance hippocampal-dependent fear memories. On a broader level, our findings underline that basic experimental research and clinically relevant pharmacological treatments employing corticosteroids should acknowledge the timing of corticosteroid administration relative to the learning phase, or therapeutic intervention.
Collapse
Affiliation(s)
- Sandra Cornelisse
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, UMC Utrecht, Utrecht, The Netherlands; Priority Program Brain and Cognition, University of Amsterdam, The Netherlands.
| | - Vanessa A van Ast
- Department of Clinical Psychology, University of Amsterdam, Amsterdam, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Kapittelweg 29, 6525, EN The Netherlands.
| | - Marian Joëls
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, UMC Utrecht, Utrecht, The Netherlands; Priority Program Brain and Cognition, University of Amsterdam, The Netherlands
| | - Merel Kindt
- Department of Clinical Psychology, University of Amsterdam, Amsterdam, The Netherlands; Priority Program Brain and Cognition, University of Amsterdam, The Netherlands
| |
Collapse
|
21
|
Raybuck JD, Lattal KM. Bridging the interval: theory and neurobiology of trace conditioning. Behav Processes 2014; 101:103-11. [PMID: 24036411 PMCID: PMC3943893 DOI: 10.1016/j.beproc.2013.08.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/25/2013] [Accepted: 08/09/2013] [Indexed: 12/26/2022]
Abstract
An early finding in the behavioral analysis of learning was that conditioned responding weakens as the conditioned stimulus (CS) and unconditioned stimulus (US) are separated in time. This "trace" conditioning effect has been the focus of years of research in associative learning. Theoretical accounts of trace conditioning have focused on mechanisms that allow associative learning to occur across long intervals between the CS and US. These accounts have emphasized degraded contingency effects, timing mechanisms, and inhibitory learning. More recently, study of the neurobiology of trace conditioning has shown that even a short interval between the CS and US alters the circuitry recruited for learning. Here, we review some of the theoretical and neurobiological mechanisms underlying trace conditioning with an emphasis on recent studies of trace fear conditioning. Findings across many studies have implications not just for how we think about time and conditioning, but also for how we conceptualize fear conditioning in general, suggesting that circuitry beyond the usual suspects needs to be incorporated into current thinking about fear, learning, and anxiety.
Collapse
Affiliation(s)
- Jonathan D Raybuck
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States.
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States.
| |
Collapse
|
22
|
Agster KL, Burwell RD. Hippocampal and subicular efferents and afferents of the perirhinal, postrhinal, and entorhinal cortices of the rat. Behav Brain Res 2013; 254:50-64. [PMID: 23872326 DOI: 10.1016/j.bbr.2013.07.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 07/01/2013] [Accepted: 07/05/2013] [Indexed: 01/06/2023]
Abstract
Available evidence suggests there is functional differentiation among hippocampal and parahippocampal subregions and along the dorsoventral (septotemporal) axis of the hippocampus. The aim of this study was to characterize and compare the efferent and afferent connections of perirhinal areas 35 and 36, postrhinal cortex, and the lateral and medial entorhinal areas (LEA and MEA) with dorsal and ventral components of the hippocampal formation (dentate gyrus, hippocampus cornu ammonis fields, and subiculum) as well as the presubiculum, and the parasubiculum. The entorhinal connections were also characterized with respect to the LEA and MEA dentate gyrus-projecting bands. In general, the entorhinal connections with the hippocampal formation are much stronger than the perirhinal and postrhinal connections. The entorhinal cortex projects strongly to all components of the hippocampal formation, whereas the perirhinal and postrhinal cortices project weakly and only to CA1 and the subiculum. In addition, the postrhinal cortex preferentially targets the dorsal CA1 and subiculum, whereas the perirhinal cortex targets ventral subiculum. Similarly, the perirhinal cortex receives more input from ventral hippocampal formation structures and the postrhinal cortex receives more input from dorsal hippocampal structures. The LEA and the MEA medial band are more strongly interconnected with ventral hippocampal structures, whereas the MEA lateral band is more interconnected with dorsal hippocampal structures. With regard to the presubiculum and parasubiculum, the postrhinal cortex and the MEA lateral band receive stronger input from the dorsal presubiculum and caudal parasubiculum. In contrast, the LEA and MEA medial bands receive stronger input from the ventral presubiculum and rostral parasubiculum.
Collapse
Affiliation(s)
- Kara L Agster
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | | |
Collapse
|
23
|
Pisansky MT, Wickham RJ, Su J, Fretham S, Yuan LL, Sun M, Gewirtz JC, Georgieff MK. Iron deficiency with or without anemia impairs prepulse inhibition of the startle reflex. Hippocampus 2013; 23:952-62. [PMID: 23733517 DOI: 10.1002/hipo.22151] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2013] [Indexed: 12/29/2022]
Abstract
Iron deficiency (ID) during early life causes long-lasting detrimental cognitive sequelae, many of which are linked to alterations in hippocampus function, dopamine synthesis, and the modulation of dopaminergic circuitry by the hippocampus. These same features have been implicated in the origins of schizophrenia, a neuropsychiatric disorder with significant cognitive impairments. Deficits in sensorimotor gating represent a reliable endophenotype of schizophrenia that can be measured by prepulse inhibition (PPI) of the acoustic startle reflex. Using two rodent model systems, we investigated the influence of early-life ID on PPI in adulthood. To isolate the role of hippocampal iron in PPI, our mouse model utilized a timed (embryonic day 18.5), hippocampus-specific knockout of Slc11a2, a gene coding an important regulator of cellular iron uptake, the divalent metal transport type 1 protein (DMT-1). Our second model used a classic rat dietary-based global ID during gestation, a condition that closely mimics human gestational ID anemia (IDA). Both models exhibited impaired PPI in adulthood. Furthermore, our DMT-1 knockout model displayed reduced long-term potentiation (LTP) and elevated paired-pulse facilitation (PPF), electrophysiological results consistent with previous findings in the IDA rat model. These results, in combination with previous findings demonstrating impaired hippocampus functioning and altered dopaminergic and glutamatergic neurotransmission, suggest that iron availability within the hippocampus is critical for the neurodevelopmental processes underlying sensorimotor gating. Ultimately, evidence of reduced PPI in both of our models may offer insights into the roles of fetal ID and the hippocampus in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Marc T Pisansky
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Cox D, Czerniawski J, Ree F, Otto T. Time course of dorsal and ventral hippocampal involvement in the expression of trace fear conditioning. Neurobiol Learn Mem 2013; 106:316-23. [PMID: 23747568 DOI: 10.1016/j.nlm.2013.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 05/21/2013] [Accepted: 05/24/2013] [Indexed: 11/25/2022]
Abstract
While a number of early studies demonstrated that hippocampal damage attenuates the expression of recent, but not remotely trained tasks, an emerging body of evidence has shown that damage to, or inactivation of, the hippocampus often impairs recall across a wide range of training-testing intervals. Collectively, these data suggest that the time course of hippocampal involvement in the storage or recall of previously-acquired memories may differ according to hippocampal subregion and the particular learning task under consideration. The present study examined the contributions of dorsal (DH) and ventral (VH) hippocampus to the expression of previously-acquired trace fear conditioning, a form of Pavlovian conditioning in which the offset of an initially neutral cue or cues and the onset of an aversive stimulus is separated by a temporal (trace) interval. Specifically, either saline or the GABA-A agonist muscimol was infused into DH or VH prior to testing either 1, 7, 28, or 42 days after trace fear conditioning. The results revealed a marked dissociation: pre-testing inactivation of DH failed to impair performance at any time-point, while pre-testing inactivation of VH impaired performance at all time-points. Importantly, pre-testing inactivation of VH had no effect on the performance of previously-acquired delay conditioning, suggesting that the deficits observed in trace conditioning cannot be attributed to a deficit in performance of the freezing response. Collectively, these data suggest that VH, but not DH, remains a neuroanatomical locus critical to the recall or expression of trace fear conditioning over an extended period of time.
Collapse
Affiliation(s)
- David Cox
- Program in Behavioral and Systems Neuroscience, Department of Psychology, Rutgers University, 152 Frelinghuysen Rd., Piscataway, NJ 08854, USA.
| | | | | | | |
Collapse
|
25
|
Fournier NM, Botterill JJ, Marks WN, Guskjolen AJ, Kalynchuk LE. Impaired recruitment of seizure-generated neurons into functional memory networks of the adult dentate gyrus following long-term amygdala kindling. Exp Neurol 2013; 244:96-104. [DOI: 10.1016/j.expneurol.2012.11.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 11/17/2012] [Accepted: 11/21/2012] [Indexed: 02/04/2023]
|
26
|
Moustafa AA, Wufong E, Servatius RJ, Pang KCH, Gluck MA, Myers CE. Why trace and delay conditioning are sometimes (but not always) hippocampal dependent: a computational model. Brain Res 2012. [PMID: 23178699 DOI: 10.1016/j.brainres.2012.11.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A recurrent-network model provides a unified account of the hippocampal region in mediating the representation of temporal information in classical eyeblink conditioning. Much empirical research is consistent with a general conclusion that delay conditioning (in which the conditioned stimulus CS and unconditioned stimulus US overlap and co-terminate) is independent of the hippocampal system, while trace conditioning (in which the CS terminates before US onset) depends on the hippocampus. However, recent studies show that, under some circumstances, delay conditioning can be hippocampal-dependent and trace conditioning can be spared following hippocampal lesion. Here, we present an extension of our prior trial-level models of hippocampal function and stimulus representation that can explain these findings within a unified framework. Specifically, the current model includes adaptive recurrent collateral connections that aid in the representation of intra-trial temporal information. With this model, as in our prior models, we argue that the hippocampus is not specialized for conditioned response timing, but rather is a general-purpose system that learns to predict the next state of all stimuli given the current state of variables encoded by activity in recurrent collaterals. As such, the model correctly predicts that hippocampal involvement in classical conditioning should be critical not only when there is an intervening trace interval, but also when there is a long delay between CS onset and US onset. Our model simulates empirical data from many variants of classical conditioning, including delay and trace paradigms in which the length of the CS, the inter-stimulus interval, or the trace interval is varied. Finally, we discuss model limitations, future directions, and several novel empirical predictions of this temporal processing model of hippocampal function and learning.
Collapse
Affiliation(s)
- Ahmed A Moustafa
- Department of Veterans Affairs, New Jersey Health Care System, East Orange, NJ, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Kryukov VI. Towards a unified model of pavlovian conditioning: short review of trace conditioning models. Cogn Neurodyn 2012; 6:377-98. [PMID: 24082960 PMCID: PMC3438324 DOI: 10.1007/s11571-012-9195-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Revised: 12/12/2011] [Accepted: 02/03/2012] [Indexed: 12/18/2022] Open
Abstract
There are three basic paradigms of classical conditioning: delay, trace and context conditioning where presentation of a conditioned stimulus (CS) or a context typically predicts an unconditioned stimulus (US). In delay conditioning CS and US normally coterminate, whereas in trace conditioning an interval of time exists between CS termination and US onset. The modeling of trace conditioning is a rather difficult computational problem and is a challenge to the behavior and connectionist approaches mainly due to a time gap between CS and US. To account for trace conditioning, Pavlov (Conditioned reflexes: an investigation of the physiological activity of the cerebral cortex, Oxford University Press, London, 1927) postulated the existence of a stimulus "trace" in the nervous system. Meanwhile, there exist many other options for solving this association problem. There are several excellent reviews of computational models of classical conditioning but none has thus far been devoted to trace conditioning. Eight representative models of trace conditioning aimed at building a prospective model are being reviewed below in a brief form. As a result, one of them, comprising the most important features of its predecessors, can be suggested as a real candidate for a unified model of trace conditioning.
Collapse
Affiliation(s)
- V. I. Kryukov
- St. Daniel Monastery, Danilovsky Val 22, 115191 Moscow, Russia
| |
Collapse
|
28
|
Dorsal hippocampal lesions disrupt Pavlovian delay conditioning and conditioned-response timing. Behav Brain Res 2012; 230:259-67. [PMID: 22366272 DOI: 10.1016/j.bbr.2012.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 01/16/2012] [Accepted: 02/08/2012] [Indexed: 11/20/2022]
Abstract
The involvement of the rat dorsal hippocampus (dhpc) in Pavlovian conditioning and timing of conditioned responding was examined in an appetitive preparation in which presentation of a relatively long, 40-s auditory conditioned stimulus (CS) was followed immediately by food delivery. Dorsal hippocampal lesions impaired Pavlovian conditioning in this task. They also produced a deficit in interval timing, replicating previous findings with short CSs. The conditioning and timing deficits observed are consistent with the findings from single-unit recording studies in other species, and suggest that the involvement of the dhpc in Pavlovian processes could be more general than is assumed by many of the current theories of hippocampal function.
Collapse
|
29
|
|
30
|
Pagani JH, Lee HJ, Young WS. Postweaning, forebrain-specific perturbation of the oxytocin system impairs fear conditioning. GENES BRAIN AND BEHAVIOR 2011; 10:710-9. [PMID: 21668734 DOI: 10.1111/j.1601-183x.2011.00709.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Oxytocin (Oxt) and vasopressin (Avp) are important for a wide variety of behaviors and the use of transgenic mice lacking the peptides or their receptors, particularly when their loss is spatially and temporally manipulated, offers an opportunity to closely examine their role in a particular behavior. We used a cued fear conditioning paradigm to examine associative learning in three lines of transgenic mice: mice that constitutively lack vasopressin 1a (Avpr1a(-/-)) or Oxt receptors (Oxtr(-/-)) and mice that have Oxt receptor loss restricted to the forebrain that begins postweaning (Oxtr(FB/FB)). Oxtr(-/-) and Avpr1a(-/-) mice have normal conditioned freezing. Oxtr(FB/FB) mice have a reduction in freezing behavior during acquisition, as well as during context and cue retention. In addition to reduction of Oxtr in the central nucleus of the amygdala, in vitro receptor autoradiography showed that the Oxtr(FB/FB) mice have significantly reduced levels of Avpr1a only in that structure. Our results show that postweaning alteration of the distribution of Oxtr receptors is critically important for fear behavior, an effect mirrored in the neural structures that mediate it. While constitutive knockouts of Oxtr and Avpr1a are useful for identifying the neural underpinnings of some behaviors, compensatory mechanisms within some circuits may obscure other behavioral roles.
Collapse
Affiliation(s)
- J H Pagani
- Section on Neural Gene Expression, NIMH, NIH, DHHS, Bethesda, MD 20892-4483, USA
| | | | | |
Collapse
|
31
|
Murawski NJ, Stanton ME. Effects of dose and period of neonatal alcohol exposure on the context preexposure facilitation effect. Alcohol Clin Exp Res 2011; 35:1160-70. [PMID: 21352243 DOI: 10.1111/j.1530-0277.2011.01449.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alcohol exposure in the rat on postnatal days (PD) 4 to 9 is known to partially damage the hippocampus and to impair hippocampus-dependent behavioral tasks. We previously reported that PD4 to 9 alcohol exposure eliminated the context preexposure facilitation effect (CPFE) in juvenile rats, a hippocampus-dependent variant of contextual fear conditioning. In the CPFE, context exposure and immediate shock occur on successive occasions and this produces conditioned freezing relative to a control group that is not preexposed to the training context. Here, we extend our earlier findings by examining the effects of neonatal alcohol administered at multiple doses or over different neonatal exposure periods. METHOD Rat pups (male and female) were exposed to a single binge dose of alcohol at one of 3 doses (2.75, 4.00, or 5.25 g/kg/d) over PD4 to 9 (Experiment 1) or to 5.25 g over PD4 to 6 or PD7 to 9 (Experiment 2). Sham-intubated (SI) and undisturbed (UD) rats served as controls. On PD31, rats were preexposed to either the training context (Pre) or an alternate context (No-Pre). On PD32, rats received an immediate unsignaled footshock (1.5 mA, 2 seconds) in the training context. Finally, on PD33, all rats were returned to the training context and tested for contextual freezing over a 5-minute period. RESULTS Undisturbed- and SI-Pre rats showed the CPFE, i.e., context preexposure facilitated contextual conditioning, relative to their No-Pre counterparts. The immediate shock deficit was present in all No-Pre groups, regardless of previous alcohol exposure. In Experiment 1, blood alcohol level was negatively correlated with contextual freezing. Group 2.75 g-Pre did not differ from controls. Group 4.00 g-Pre froze significantly less than Groups UD- and SI-Pre but more than Group 5.25-Pre, which showed the immediate shock deficit. In Experiment 2, alcohol exposure limited to PD7 to 9, but not PD4 to 6, disrupted the CPFE. CONCLUSIONS This is the first demonstration of dose-related impairment on a hippocampus-dependent task produced by neonatal alcohol exposure in the rat. Exposure period effects support previous studies of alcohol and spatial learning. The CPFE is a more sensitive behavioral task that can be used to elucidate developmental alcohol-induced deficits over a range of alcohol doses that are more relevant to human exposure levels.
Collapse
Affiliation(s)
- Nathen J Murawski
- Department of Psychology, University of Delaware, Newark, DE 19716, USA.
| | | |
Collapse
|
32
|
Albrechet-Souza L, Borelli KG, Almada RC, Brandão ML. Midazolam reduces the selective activation of the rhinal cortex by contextual fear stimuli. Behav Brain Res 2010; 216:631-8. [PMID: 20851717 DOI: 10.1016/j.bbr.2010.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/01/2010] [Accepted: 09/06/2010] [Indexed: 12/01/2022]
Abstract
Independent brain circuits appear to underlie different forms of conditioned fear, depending on the type of conditioning used, such as a context or explicit cue paired with footshocks. Several clinical reports have associated damage to the medial temporal lobe (MTL) with retrograde amnesia. Although a number of studies have elucidated the neural circuits underlying conditioned fear, the involvement of MTL components in the aversive conditioning paradigm is still unclear. To address this issue, we assessed freezing responses and Fos protein expression in subregions of the rhinal cortex and ventral hippocampus of rats following exposure to a context, light or tone previously paired with footshock (Experiment 1). A comparable degree of freezing was observed in the three types of conditioned fear, but with distinct patterns of Fos distribution. The groups exposed to cued fear conditioning did not show changes in Fos expression, whereas the group subjected to contextual fear conditioning showed selective activation of the ectorhinal (Ect), perirhinal (Per), and entorhinal (Ent) cortices, with no changes in the ventral hippocampus. We then examined the effects of the benzodiazepine midazolam injected bilaterally into these three rhinal subregions in the expression of contextual fear conditioning (Experiment 2). Midazolam administration into the Ect, Per, and Ent reduced freezing responses. These findings suggest that contextual and explicit stimuli endowed with aversive properties through conditioning recruit distinct brain areas, and the rhinal cortex appears to be critical for storing context-, but not explicit cue-footshock, associations.
Collapse
|
33
|
The role of nicotinic acetylcholine receptors in the medial prefrontal cortex and hippocampus in trace fear conditioning. Neurobiol Learn Mem 2010; 94:353-63. [PMID: 20727979 DOI: 10.1016/j.nlm.2010.08.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 08/01/2010] [Accepted: 08/15/2010] [Indexed: 12/28/2022]
Abstract
Acute nicotine enhances multiple types of learning including trace fear conditioning but the underlying neural substrates of these effects are not well understood. Trace fear conditioning critically involves the medial prefrontal cortex and hippocampus, which both express nicotinic acetylcholine receptors (nAChRs). Therefore, nicotine could act in either or both areas to enhance trace fear conditioning. To identify the underlying neural areas and nAChR subtypes, we examined the effects of infusion of nicotine, or nicotinic antagonists dihydro-beta-erythroidine (DHβE: high-affinity nAChRs) or methyllycaconitine (MLA: low-affinity nAChRs) into the dorsal hippocampus, ventral hippocampus, and medial prefrontal cortex (mPFC) on trace and contextual fear conditioning. We found that the effects of nicotine on trace and contextual fear conditioning vary by brain region and nAChR subtype. The dorsal hippocampus was involved in the effects of nicotine on both trace and contextual fear conditioning but each task was sensitive to different doses of nicotine. Additionally, dorsal hippocampal infusion of the antagonist DHβE produced deficits in trace but not contextual fear conditioning. Nicotine infusion into the ventral hippocampus produced deficits in both trace and contextual fear conditioning. In the mPFC, nicotine enhanced trace but not contextual fear conditioning. Interestingly, infusion of the antagonists MLA or DHβE in the mPFC also enhanced trace fear conditioning. These findings suggest that nicotine acts on different substrates to enhance trace versus contextual fear conditioning, and that nicotine-induced desensitization of nAChRs in the mPFC may contribute to the effects of nicotine on trace fear conditioning.
Collapse
|
34
|
Burman MA, Hamilton KL, Gewirtz JC. Role of corticosterone in trace and delay conditioned fear-potentiated startle in rats. Behav Neurosci 2010; 124:294-9. [PMID: 20364889 DOI: 10.1037/a0018911] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Emotional events often lead to particularly strong memory formation. Corticosterone, the final product of hypothalamic-pituitary-adrenal (HPA)-axis activation, has been suggested to play a critical role in this effect. Although a great deal of work has implicated the amygdala as a necessary structure for the effects of corticosterone, other studies have suggested a critical role for the hippocampus in determining the involvement of corticosterone. The current experiments examined this question by disrupting corticosterone synthesis with administration of metyrapone (25 or 100 mg/kg) prior to training in either dorsal hippocampus-independent delay fear conditioning or dorsal hippocampus-dependent trace fear conditioning. Metyrapone administration 2 hrs prior to training significantly attenuated corticosterone secretion during training, but these effects were transient as corticosterone levels were similar to control subjects following the test session. As hypothesized, only trace fear conditioning was impaired. This suggests that only fear conditioning tasks that are dependent on the dorsal hippocampus require HPA-axis activation in order to be learned.
Collapse
Affiliation(s)
- Michael A Burman
- Department of Psychology, University of New England, Biddeford, ME 04005, USA.
| | | | | |
Collapse
|
35
|
Boccardi M, Ganzola R, Rossi R, Sabattoli F, Laakso MP, Repo-Tiihonen E, Vaurio O, Könönen M, Aronen HJ, Thompson PM, Frisoni GB, Tiihonen J. Abnormal hippocampal shape in offenders with psychopathy. Hum Brain Mapp 2010; 31:438-47. [PMID: 19718651 DOI: 10.1002/hbm.20877] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Posterior hippocampal volumes correlate negatively with the severity of psychopathy, but local morphological features are unknown. The aim of this study was to investigate hippocampal morphology in habitually violent offenders having psychopathy. Manual tracings of hippocampi from magnetic resonance images of 26 offenders (age: 32.5 +/- 8.4), with different degrees of psychopathy (12 high, 14 medium psychopathy based on the Psychopathy Checklist Revised), and 25 healthy controls (age: 34.6 +/- 10.8) were used for statistical modelling of local changes with a surface-based radial distance mapping method. Both offenders and controls had similar hippocampal volume and asymmetry ratios. Local analysis showed that the high psychopathy group had a significant depression along the longitudinal hippocampal axis, on both the dorsal and ventral aspects, when compared with the healthy controls and the medium psychopathy group. The opposite comparison revealed abnormal enlargement of the lateral borders in both the right and left hippocampi of both high and medium psychopathy groups versus controls, throughout CA1, CA2-3 and the subicular regions. These enlargement and reduction effects survived statistical correction for multiple comparisons in the main contrast (26 offenders vs. 25 controls) and in most subgroup comparisons. A statistical check excluded a possible confounding effect from amphetamine and polysubstance abuse. These results indicate that habitually violent offenders exhibit a specific abnormal hippocampal morphology, in the absence of total gray matter volume changes, that may relate to different autonomic modulation and abnormal fear-conditioning.
Collapse
Affiliation(s)
- Marina Boccardi
- LENITEM Laboratory of Epidemiology, Neuroimaging, and Telemedicine, IRCCS San Giovanni di Dio-Fatebenefratelli, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ohmura Y, Izumi T, Yamaguchi T, Tsutsui-Kimura I, Yoshida T, Yoshioka M. The serotonergic projection from the median raphe nucleus to the ventral hippocampus is involved in the retrieval of fear memory through the corticotropin-releasing factor type 2 receptor. Neuropsychopharmacology 2010; 35:1271-8. [PMID: 20072117 PMCID: PMC3055345 DOI: 10.1038/npp.2009.229] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several different studies have separately established that serotonin, corticotropin-releasing factor (CRF) receptors, and the hippocampus are involved in fear memory retrieval. The main aim of this study is to connect these separate studies. To assess the levels of anxiety/fear, we used the contextual fear-conditioning test and the elevated plus maze test as memory-dependent and memory-independent tasks, respectively. We injected CRF receptor antagonists or vehicle into the median raphe nucleus (MRN) 10 min before behavioral tests. As a result, 1000 ng of astressin 2B (CRF(2) receptor antagonist), but not 250 ng of antalarmin (CRF(1) receptor antagonist), significantly suppressed the expression rate of freezing behavior in the contextual fear-conditioning test. However, in the elevated plus maze test, there was no difference between astressin 2B-injected rats and saline-injected rats in the time spent in open arms. Neither the amount of exploratory behavior nor the moving distance in the EPM of astressin 2B-injected rats differed from that of vehicle-injected rats. Moreover, when we assessed the extracellular serotonin release in the ventral hippocampus in freely moving rats through in vivo microdialysis, it was shown that the blockade of the CRF(2) receptor in the MRN suppressed serotonin release in the ventral hippocampus during fear memory retrieval. These results indicated that endogenous CRF and/or related ligands that were released in the MRN could activate the CRF(2) receptor and stimulate serotonin release in the ventral hippocampus, thereby inducing fear memory retrieval.
Collapse
Affiliation(s)
- Yu Ohmura
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan,Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Takeshi Izumi
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Taku Yamaguchi
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Iku Tsutsui-Kimura
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takayuki Yoshida
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Mitsuhiro Yoshioka
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan,Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, N15 W7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan. Tel: +81 11 706 5059; Fax: +81 11 706 7872; E-mail:
| |
Collapse
|
37
|
Foster JA, Burman MA. Evidence for hippocampus-dependent contextual learning at postnatal day 17 in the rat. Learn Mem 2010; 17:259-66. [PMID: 20427514 DOI: 10.1101/lm.1755810] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Long-term memory for fear of an environment (contextual fear conditioning) emerges later in development (postnatal day; PD 23) than long-term memory for fear of discrete stimuli (PD 17). As contextual, but not explicit cue, fear conditioning relies on the hippocampus; this has been interpreted as evidence that the hippocampus is not fully developed until PD 23. Alternatively, the hippocampus may be functional prior to PD 23, but unable to cooperate with the amygdala for fearful learning. The current experiments investigate this by separating the phases of conditioning across developmental stages. Rats were allowed to learn about the context on one day and to form the fearful association on another. Rats exposed to the context on PD 17 exhibited significant fear only when trained and tested a week later (PD 23, 24), but not on consecutive days (PD 18, 19), demonstrating that rats can learn about a context as early as PD 17. Further experiments clarify that it is associative mechanisms that are developing between PD 18 and 23. Finally, the hippocampus was lesioned prior to training to ensure the task is being solved in a hippocampus-dependent manner. These data provide compelling evidence that the hippocampus is functional for contextual learning as early as PD 17, however, its connection to the amygdala or other relevant brain structures may not yet be fully developed.
Collapse
Affiliation(s)
- Jennifer A Foster
- Program in Neuroscience, Bates College, Lewiston, Maine 04240-6028, USA
| | | |
Collapse
|
38
|
Murawski NJ, Stanton ME. Variants of contextual fear conditioning are differentially impaired in the juvenile rat by binge ethanol exposure on postnatal days 4-9. Behav Brain Res 2010; 212:133-42. [PMID: 20385174 DOI: 10.1016/j.bbr.2010.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 04/01/2010] [Accepted: 04/03/2010] [Indexed: 11/17/2022]
Abstract
Neonatal ethanol exposure in the rat is known to partially damage the hippocampus, but such exposure causes only modest or inconsistent deficits on hippocampus-dependent behavioral tasks. This may reflect variable sensitivity of these tasks or residual function following partial hippocampal injury. The context preexposure facilitation effect (CPFE) is a variant of context conditioning in which context exposure and immediate shock occur on successive occasions. During testing, preexposed rats freeze more than non-preexposed controls. The CPFE is more sensitive to anterograde hippocampal injury than standard contextual fear conditioning (e.g., Rudy JW, O'Reilly RC. Conjunctive representations, the hippocampus, and contextual fear conditioning. Cogn Affect Behav Neurosci 2001;1:66-82). We report that rats exposed to a high binge dose of ethanol (5.25g/kg/day) over postnatal days [PD] 4-9 failed to demonstrate the CPFE when preexposed to the conditioning context on PD31, relative to sham-intubated and undisturbed controls (Experiment 1). Neonatal alcohol disrupted conditioned freezing to a much lesser extent relative to controls when context preexposure was followed by a standard context conditioning trial rather than immediate shock (Experiment 2). Fear conditioning to a discrete auditory cue (tone) was unaffected by neonatal alcohol exposure ruling out possible performance effects (Experiment 3). These findings suggest that the CPFE is an especially sensitive task for detecting hippocampal injury produced by neonatal alcohol. Mixed results with other tasks may reflect residual hippocampal function and/or the use of alternate neurobehavioral systems or "strategies" following alcohol-induced brain damage.
Collapse
|
39
|
Wamsley EJ, Antrobus JS. The expression of trace conditioning during non-REM sleep and its relation to subjective experience. Neurobiol Learn Mem 2009; 92:283-91. [DOI: 10.1016/j.nlm.2009.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 02/03/2009] [Accepted: 02/21/2009] [Indexed: 11/29/2022]
|
40
|
Hunt PS, Jacobson SE, Torok EJ. Deficits in trace fear conditioning in a rat model of fetal alcohol exposure: dose-response and timing effects. Alcohol 2009; 43:465-74. [PMID: 19801276 PMCID: PMC2758299 DOI: 10.1016/j.alcohol.2009.08.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 08/15/2009] [Accepted: 08/17/2009] [Indexed: 11/28/2022]
Abstract
In humans, prenatal alcohol exposure can result in significant impairments in several types of learning and memory, including declarative and spatial memory. Animal models have been useful for confirming that many of the observed effects are the result of alcohol exposure, and not secondary to poor maternal nutrition or adverse home environments. Wagner and Hunt (2006) reported that rats exposed to ethanol during the neonatal period (postnatal days [PDs] 4-9) exhibited impaired trace fear conditioning when trained as adolescents, but were unaffected in delay fear conditioning. The present series of three experiments represent a more detailed analysis of ethanol-induced deficits in trace conditioning. In Experiment 1, the dose of ethanol given to neonates was varied (3.0, 4.0, or 5.0g/kg/day). There was a dose-dependent reduction in trace conditioning, with the poorest performance observed in animals treated with the highest dose. In Experiment 2, it was found that the impairment in trace conditioning resulting from neonatal ethanol exposure was dependent on the duration of the trace interval used for training; less learning was evident in ethanol-exposed animals trained with longer trace interval durations. These results confirm other reports of delay-dependent memory deficits. Finally, Experiment 3 determined that ethanol exposure limited to the first half of the neonatal period (PDs 4-6) was more detrimental to later trace conditioning than exposure during the second half (PDs 7-9). These results support the hypothesis that trace-conditioning impairments resulting from early ethanol exposure are due to the drug's teratogenic effects on the developing hippocampus, as the findings parallel those observed in animals with discrete hippocampal lesions. Comparisons between delay and trace fear-conditioning performance in animals exposed to ethanol during the brain growth spurt provide a model system to study both selective learning impairments and possible treatment approaches for humans with fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Pamela S Hunt
- Department of Psychology, College of William and Mary, Williamsburg, VA 23187-8795, USA.
| | | | | |
Collapse
|
41
|
Raybuck JD, Gould TJ. Nicotine withdrawal-induced deficits in trace fear conditioning in C57BL/6 mice--a role for high-affinity beta2 subunit-containing nicotinic acetylcholine receptors. Eur J Neurosci 2009; 29:377-87. [PMID: 19200240 DOI: 10.1111/j.1460-9568.2008.06580.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nicotine alters cognitive processes that include working memory and long-term memory. Trace fear conditioning may involve working memory during acquisition while also allowing the assessment of long-term memory. The present study used trace fear conditioning in C57BL/6 mice to investigate the effects of acute nicotine, chronic nicotine and withdrawal of chronic nicotine on processes active during acquisition and recall 24 h later and to examine the nicotinic acetylcholine receptor subtypes (nAChRs) involved in withdrawal deficits in trace fear conditioning. During training, acute nicotine (0.09 mg/kg) enhanced, but chronic nicotine (6.3 mg/kg/day, 13 days) and withdrawal of chronic nicotine (6.3 mg/kg/day, 12 days) had no significant effect on, acquisition of trace conditioning. At recall, acute treatment enhanced conditioning while chronic nicotine had no effect and withdrawal of chronic nicotine resulted in deficits. Antagonist-precipitated withdrawal was used to characterize the nAChRs involved in the withdrawal deficits. The low-affinity nAChR antagonist MLA (1.5, 3 or 9 mg/kg) had no effect on trace fear conditioning, but the high-affinity nAChR antagonist DHbetaE (3 mg/kg) precipitated deficits in trace fear conditioning if administered at training or training and testing, but not if administered at testing alone. The beta2 nAChR subunit is involved in the withdrawal effects as withdrawal of chronic nicotine produced deficits in trace fear conditioning in wildtype but not in beta2-knockout mice. Thus, nicotine alters processes involved in both acquisition and long-term memory of trace fear conditioning, and high-affinity beta2 subunit-containing nAChRs are critically involved in the effects of nicotine withdrawal on trace fear conditioning.
Collapse
Affiliation(s)
- J D Raybuck
- Center for Substance Abuse Research & Department of Psychology, 1701n 13th, Weiss Hall, 657, Temple University, Philadelphia, PA 19122, USA
| | | |
Collapse
|
42
|
Czerniawski J, Yoon T, Otto T. Dissociating space and trace in dorsal and ventral hippocampus. Hippocampus 2009; 19:20-32. [PMID: 18651617 DOI: 10.1002/hipo.20469] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Emerging evidence suggests that the hippocampus can be anatomically and functionally dissociated along its septotemporal axis into dorsal and ventral subregions. With respect to function, we have recently demonstrated that pre-training excitotoxic lesions of ventral, but not dorsal, hippocampus impair the acquisition of trace fear conditioning, whereas post-training lesions of either dorsal or ventral hippocampus impair the subsequent expression of trace fear conditioning (Yoon and Otto (2007) Neurobiol Learn Mem 87:464-475). In addition to trace fear conditioning, dorsal and ventral hippocampus appear to be differentially involved in a number of spatial memory tasks. The present study examined the effects of temporary inactivation of dorsal or ventral hippocampus on the acquisition and expression of trace fear conditioning and on performance of a spatial delayed reinforced alternation task. The findings demonstrate a double dissociation of dorsal and ventral hippocampal function: inactivation of ventral, but not dorsal, hippocampus attenuated the acquisition and expression of trace fear conditioning, whereas inactivation of dorsal, but not ventral, hippocampus dramatically impaired performance in the delayed reinforced alternation task. These data further support the notion that dorsal and ventral hippocampus contribute differentially to performance in a variety of paradigms.
Collapse
|
43
|
Esclassan F, Coutureau E, Di Scala G, Marchand AR. Differential contribution of dorsal and ventral hippocampus to trace and delay fear conditioning. Hippocampus 2009; 19:33-44. [PMID: 18683846 DOI: 10.1002/hipo.20473] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Trace conditioning relies on the maintained representation of a stimulus across a trace interval, and may involve a persistent trace of the conditioned stimulus (CS) and/or a contribution of contextual conditioning. The role of hippocampal structures in these two types of conditioning was studied by means of pretraining lesions and reversible inactivation of the hippocampus in rats. Similar levels of conditioning to a tone CS and to the context were obtained with a trace interval of 30 s. Neurotoxic lesions of the whole hippocampus or reversible muscimol inactivation of the ventral hippocampus impaired both contextual and tone freezing in both trace- and delay-conditioned rats. Dorsal hippocampal injections impaired contextual freezing and trace conditioning, but not delay conditioning. No dissociation between trace and contextual conditioning was observed under any of these conditions. Altogether, these data indicate that the ventral and dorsal parts of the hippocampus compute different aspects of trace conditioning, with the ventral hippocampus being involved in fear and anxiety processes, and the dorsal hippocampus in the temporal and contextual aspects of event representation.
Collapse
Affiliation(s)
- Frederic Esclassan
- Université de Bordeaux, Centre de Neurosciences Intégratives et Cognitives, C.N.R.S. UMR 5228, Talence, France
| | | | | | | |
Collapse
|
44
|
Female rats learn trace memories better than male rats and consequently retain a greater proportion of new neurons in their hippocampi. Proc Natl Acad Sci U S A 2009; 106:2927-32. [PMID: 19188598 DOI: 10.1073/pnas.0809650106] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Learning increases the survival of new cells that are generated in the hippocampal formation before the training experience, especially if the animal learns to associate stimuli across time [Gould E, Beylin A, Tanapat P, Reeves A, Shors TJ (1999) Nat Neurosci 2:260-265]. All relevant studies have been conducted on male rats, despite evidence for sex differences in this type of learning. In the present study, we asked whether sex differences in learning influence the survival of neurons generated in the adult hippocampus. Male and female adult rats were injected with one dose of bromodeoxyuridine (BrdU; 200 mg/kg), to label one population of dividing cells. One week later, half of the animals were trained with a temporal learning task of trace eyeblink conditioning, while the other half were not trained. Animals were killed 1 day after training (12 days after the BrdU injection). Hippocampal tissue was stained for BrdU and a marker of immature neurons, doublecortin. Both sexes learned to emit the conditioned eyeblink response during the trace interval. As a consequence, more new neurons remained in their hippocampi than in sex-matched controls. In individual animals, the number of surviving cells correlated positively with asymptotic performance; those that expressed more learned responses retained more new neurons. However, animals that learned very well retained even more new cells if they required many trials to do so. Because females emitted more learned responses than males did, they retained nearly twice as many new cells per unit volume of tissue. This effect was most evident in the ventral region of the hippocampal formation. Thus, sex differences in learning alter the anatomical structure of the hippocampus. As a result, male and female brains continue to differentiate in adulthood.
Collapse
|
45
|
Maren S. Pavlovian fear conditioning as a behavioral assay for hippocampus and amygdala function: cautions and caveats. Eur J Neurosci 2009; 28:1661-6. [PMID: 18973583 DOI: 10.1111/j.1460-9568.2008.06485.x] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pavlovian fear conditioning has become an important model for investigating the neural substrates of learning and memory in rats, mice and humans. The hippocampus and amygdala are widely believed to be essential for fear conditioning to contexts and discrete cues, respectively. Indeed, this parsing of function within the fear circuit has been used to leverage fear conditioning as a behavioral assay of hippocampal and amygdala function, particularly in transgenic mouse models. Recent work, however, blurs the anatomical segregation of cue and context conditioning and challenges the necessity for the hippocampus and amygdala in fear learning. Moreover, nonassociative factors may influence the performance of fear responses under a variety of conditions. Caution must therefore be exercised when using fear conditioning as a behavioral assay for hippocampal- and amygdala-dependent learning.
Collapse
Affiliation(s)
- Stephen Maren
- Department of Psychology and Neuroscience Program, University of Michigan, Ann Arbor, MI 48109-1043, USA.
| |
Collapse
|
46
|
Effects of gestational iron deficiency on fear conditioning in juvenile and adult rats. Brain Res 2008; 1237:195-203. [PMID: 18789313 DOI: 10.1016/j.brainres.2008.08.079] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 08/20/2008] [Accepted: 08/21/2008] [Indexed: 11/22/2022]
Abstract
The hippocampus is especially sensitive to the effects of gestational and neonatal iron deficiency, even after iron repletion. This study compared the effects of iron deficiency, maintained from gestational day 2 to postnatal day (P)7, on "delay" and "trace" fear conditioning. Only the latter paradigm is critically dependent on the dorsal hippocampus. In different groups of rats, fear conditioning commenced either prior to puberty (P28 or P35) or after puberty (P56). Fear conditioning was measured using fear-potentiated startle. Both delay and trace fear conditioning were diminished by iron deficiency at P28 and P35. Hippocampal expression of the plasticity-related protein PKC-gamma was increased through trace fear conditioning, but reduced at P35 in the iron-deficient group. Trace fear conditioning was enhanced by prior iron deficiency in the P56 group. This unanticipated finding in iron-repleted adults is consistent with the effects of developmental iron deficiency on inhibitory avoidance learning, but contrasts with the persistent deleterious long-term effects of a more severe iron-deficiency protocol, suggesting that degree and duration of iron deficiency affects the possibility of recovery from its deleterious effects.
Collapse
|
47
|
Engin E, Treit D. The effects of intra-cerebral drug infusions on animals' unconditioned fear reactions: a systematic review. Prog Neuropsychopharmacol Biol Psychiatry 2008; 32:1399-419. [PMID: 18495312 DOI: 10.1016/j.pnpbp.2008.03.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 03/25/2008] [Accepted: 03/26/2008] [Indexed: 10/22/2022]
Abstract
Intra-cerebral (i.c.) microinfusion of selective receptor agonists and antagonists into behaving animals can provide both neuroanatomical and neurochemical insights into the neural mechanisms of anxiety. However, there have been no systematic reviews of the results of this experimental approach that include both a range of unconditioned anxiety reactions and a sufficiently broad theoretical context. Here we focus on amino acid, monoamine, cholinergic and peptidergic receptor ligands microinfused into neural structures previously implicated in anxiety, and subsequent behavioral effects in animal models of unconditioned anxiety or fear. GABAA receptor agonists and glutamate receptor antagonists produced the most robust anxiolytic-like behavioral effects, in the majority of neural substrates and animal models. In contrast, ligands of the other receptor systems had more selective, site-specific anti-anxiety effects. For example, 5-HT1A receptor agonists produced anxiolytic-like effects in the raphe nuclei, but inconsistent effects in the amygdala, septum, and hippocampus. Conversely, 5-HT3 receptor antagonists produced anxiolytic-like effects in the amygdala but not in the raphe nuclei. Nicotinic receptor agonists produced anxiolytic-like effects in the raphe and anxiogenic effects in the septum and hippocampus. Unexpectedly, physostigmine, a general cholinergic agonist, produced anxiolytic-like effects in the hippocampus. Neuropeptide receptors, although they are popular targets for the development of selective anxiolytic agents, had the least reliable effects across different animal models and brain structures, perhaps due in part to the fact that selective receptor ligands are relatively scarce. While some inconsistencies in the microinfusion data can easily be attributed to pharmacological variables such as dose or ligand selectivity, in other instances pharmacological explanations are more difficult to invoke: e.g., even the same dose of a known anxiolytic compound (midazolam) with a known mechanism of action (the benzodiazepine-GABAA receptor complex), can selectively affect different fear reactions depending upon the different subregions of the nucleus into which it is infused (CeA versus BLA). These particular functional dissociations are important and may depend on the ability of a GABAA receptor agonist to interact with distinct isoforms and combinations of GABAA receptor subunits (e.g., alpha1-6, beta1-3, Upsilon1-2, delta), many of which are unevenly distributed throughout the brain. Although this molecular hypothesis awaits thorough evaluation, the microinfusion data overall give some support for a model of "anxiety" that is functionally segregated along different levels of a neural hierarchy, analogous in some ways to the organization of sensorimotor systems.
Collapse
Affiliation(s)
- Elif Engin
- Department of Psychology, University of Alberta, P449 Biological Sciences Building, Edmonton, Canada AB T6G 2E9
| | | |
Collapse
|
48
|
Raman L, Hamilton KL, Gewirtz JC, Rao R. Effects of chronic hypoxia in developing rats on dendritic morphology of the CA1 subarea of the hippocampus and on fear-potentiated startle. Brain Res 2007; 1190:167-74. [PMID: 18083146 DOI: 10.1016/j.brainres.2007.11.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 11/12/2007] [Accepted: 11/15/2007] [Indexed: 10/22/2022]
Abstract
Chronic hypoxia (CH) present in infants with cyanotic congenital heart disease may be responsible for subsequent cognitive deficits seen in these children. In a rat model of CH [10% O(2) between postnatal day (P) 3 and 28], we have demonstrated significant alterations in energy metabolism and excitatory neurotransmission in the developing hippocampus. These alterations may adversely affect dendritic morphology, which is a highly energy-dependent and excitatory neurotransmitter-mediated event, and hippocampus-mediated behaviors. We measured the apical segment length of dendrites in pyramidal neurons of the CA1 region of the hippocampus using microtubule-associated protein-2 (MAP-2) histochemistry on P28 while the animals were hypoxic (n=8 in CH and n=6 in control), and on P56 after the animals had been normoxic for 4 weeks (n=8/group). We also compared dorsal hippocampus-dependent trace fear conditioning and dorsal hippocampus-independent delay fear conditioning on P56. Developmental trajectory of the apical segment length was similar in CH and controls, decreasing between P28 and P56. However, when compared with the controls, the apical segment length was longer in the CH group on both P28 [55.11+/-2.30 microm (CH) vs. 40.52+/-1.20 microm (control), p<0.001] and P56 [44.01+/-1.56 microm (CH) vs. 31.75+/-1.31 microm (control), p<0.001], suggesting the persistence of an immature dendritic architecture. Both trace and delay fear conditioning were decreased in the CH group, suggesting functional abnormality beyond the dorsal hippocampus. These structural and functional alterations may contribute to the cognitive deficits seen in infants at risk for CH.
Collapse
Affiliation(s)
- Lakshmi Raman
- Department of Pediatrics, Hennepin County Medical Center, University of Minnesota, Minneapolis, MN 55415, USA.
| | | | | | | |
Collapse
|
49
|
Petrides T, Georgopoulos P, Kostopoulos G, Papatheodoropoulos C. The GABAA receptor-mediated recurrent inhibition in ventral compared with dorsal CA1 hippocampal region is weaker, decays faster and lasts less. Exp Brain Res 2007; 177:370-83. [PMID: 16988819 DOI: 10.1007/s00221-006-0681-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Accepted: 08/15/2006] [Indexed: 11/26/2022]
Abstract
Hippocampal functions appear to be segregated along the dorso-ventral axis of the structure. Differences at the cellular and local neuronal network level may be involved in this functional segregation. In this study the characteristics of CA1 recurrent inhibition (RI) were measured and compared between dorsal (DH, n = 95) and ventral (VH, n = 60) hippocampal slices, using recordings of suprathreshold field potentials. RI strength was estimated as the percentile decrease of the population spike (PS) amplitude evoked with an orthodromic stimulus (at the Schaffer collaterals) when preceded by an antidromic stimulus (at the alveus). Varying the interpulse interval (IPI) between the two stimuli, we estimated RI duration. Alvear stimulation produced significant PS suppression in both VH and DH at every IPI tested, from 10 to 270 ms. Moreover, gradually more oblique DH (but not VH) slices displayed increasing RI, which at IPIs < or = 125 ms was reversibly abolished by the GABAA receptor antagonist picrotoxin (10 microM). The GABAA-mediated RI, measured under the blockade of GABAB receptors, was weaker, decayed faster and lasted less in VH compared to DH slices, regardless of the slice orientation. Specifically, in VH compared to DH, the PS suppression at 20 ms was 34.4 +/- 4.5% versus 69.9 +/- 6.5% (P < 0.001), the time constant of RI decay was 29 +/- 2.4 versus 87.5 +/- 13.6 ms (P < 0.01) and the duration was 50 versus 125 ms (P < 0.001). Thus, GABAA-mediated RI may control the CA1 excitatory output less effectively in VH compared to DH. The observed dorso-ventral differences in RI contribute to the longitudinal diversification of the structure and may underlie to some extent the region-specificity of hippocampal functions.
Collapse
Affiliation(s)
- Theodoros Petrides
- Department of Physiology, Medical School, University of Patras, 26 504 Patras, Greece
| | | | | | | |
Collapse
|
50
|
Abstract
Although current models of hippocampal function stress its well-known role in cognitive functions, historically it has also been viewed as a neural mediator of emotion. Here, we review recent evidence from intrahippocampal infusion studies in animals that support a distinctive role of the hippocampus in anxiety, independent of its roles in learning and memory. Specifically, gamma-aminobutyric acid type A receptor agonists, both direct and indirect, reliably inhibit a number of animals' untrained anxiety reactions when microinfused into the hippocampus, whereas gamma-aminobutyric acid type A receptor antagonists do not. Intrahippocampal infusions of glutamatergic, serotonergic and cholinergic compounds also produce statistically reliable antianxiety effects, but the results vary as a function of specific anxiety reactions, and to some extent specific intrahippocampal targets. One hypothesis that may accommodate some of this variability is that anxiety is functionally segregated within the hippocampus, with ventral subregions more involved in anxiety-related processes, and dorsal subregions more involved with cognitive processes. Another possibility is that different hippocampal functions (e.g. memory and anxiety) are mediated by different neurotransmitter systems and/or different receptor subtypes within the hippocampus. Although there is some evidence that supports the latter hypothesis, the evidence for the former is not conclusive. Overall, however, the data clearly suggest that the hippocampus is importantly and directly involved in the mediation of untrained anxiety reactions in animals.
Collapse
Affiliation(s)
- Elif Engin
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|