101
|
Kissiwaa SA, Bagley EE. Central sensitization of the spino-parabrachial-amygdala pathway that outlasts a brief nociceptive stimulus. J Physiol 2018; 596:4457-4473. [PMID: 30004124 PMCID: PMC6138295 DOI: 10.1113/jp273976] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/29/2018] [Indexed: 12/22/2022] Open
Abstract
KEY POINTS Chronic pain is disabling because sufferers form negative associations between pain and activities, such as work, leading to the sufferer limiting these activities. Pain information arriving in the amygdala is responsible for forming these associations and contributes to us feeling bad when we are in pain. Ongoing injuries enhance the delivery of pain information to the amygdala. If we want to understand why chronic pain can continue without ongoing injury, it is important to know whether this facilitation continues once the injury has healed. In the present study, we show that a 2 min noxious heat stimulus, without ongoing injury, is able to enhance delivery of pain information to the amygdala for 3 days. If the noxious heat stimulus is repeated, this enhancement persists even longer. These changes may prime this information pathway so that subsequent injuries may feel even worse and the associative learning that results in pain-related avoidance may be promoted. ABSTRACT Pain is an important defence against dangers in our environment; however, some clinical conditions produce pain that outlasts this useful role and persists even after the injury has healed. The experience of pain consists of somatosensory elements of intensity and location, negative emotional/aversive feelings and subsequent restrictions on lifestyle as a result of a learned association between certain activities and pain. The amygdala contributes negative emotional value to nociceptive sensory information and forms the association between an aversive response and the environment in which it occurs. It is able to form this association because it receives nociceptive information via the spino-parabrachio-amygdaloid pathway and polymodal sensory information via cortical and thalamic inputs. Synaptic plasticity occurs at the parabrachial-amygdala synapse and other brain regions in chronic pain conditions with ongoing injury; however, very little is known about how plasticity occurs in conditions with no ongoing injury. Using immunohistochemistry, electrophysiology and behavioural assays, we show that a brief nociceptive stimulus with no ongoing injury is able to produce long-lasting synaptic plasticity at the rat parabrachial-amygdala synapse. We show that this plasticity is caused by an increase in postsynaptic AMPA receptors with a transient change in the AMPA receptor subunit, similar to long-term potentiation. Furthermore, this synaptic potentiation primes the synapse so that a subsequent noxious stimulus causes prolonged potentiation of the nociceptive information flow into the amygdala. As a result, a second injury could have an increased negative emotional value and promote associative learning that results in pain-related avoidance.
Collapse
Affiliation(s)
- Sarah A Kissiwaa
- Discipline of Pharmacology and Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| | - Elena E Bagley
- Discipline of Pharmacology and Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| |
Collapse
|
102
|
Wallrafen R, Dresbach T. The Presynaptic Protein Mover Is Differentially Expressed Across Brain Areas and Synapse Types. Front Neuroanat 2018; 12:58. [PMID: 30057527 PMCID: PMC6053503 DOI: 10.3389/fnana.2018.00058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/21/2018] [Indexed: 01/12/2023] Open
Abstract
The assembly and function of presynaptic nerve terminals relies on evolutionarily conserved proteins. A small number of presynaptic proteins occurs only in vertebrates. These proteins may add specialized functions to certain synapses, thus increasing synaptic heterogeneity. Here, we show that the vertebrate-specific synaptic vesicle (SV) protein mover is differentially distributed in the forebrain and cerebellum of the adult mouse. Using a quantitative immunofluorescence approach, we compare the expression of mover to the expression of the general SV marker synaptophysin in 16 brain areas. We find that mover is particularly abundant in the septal nuclei (SNu), ventral pallidum (VPa), amygdala and hippocampus. Within the hippocampus, mover is predominantly associated with excitatory synapses. Its levels are low in layers that receive afferent input from the entorhinal cortex, and high in layers harboring intra-hippocampal circuits. In contrast, mover levels are high in all nuclei of the amygdala, and mover is associated with inhibitory synapses in the medioposterior amygdala. Our data reveal a striking heterogeneity in the abundance of mover on three levels, i.e., between brain areas, within individual brain areas and between synapse types. This distribution suggests a role for mover in providing specialization to subsets of synapses, thereby contributing to the functional diversity of brain areas.
Collapse
Affiliation(s)
| | - Thomas Dresbach
- Synaptogenesis Group, Institute of Anatomy and Embryology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
103
|
McGarry LM, Carter AG. Prefrontal Cortex Drives Distinct Projection Neurons in the Basolateral Amygdala. Cell Rep 2018; 21:1426-1433. [PMID: 29117549 DOI: 10.1016/j.celrep.2017.10.046] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/01/2017] [Accepted: 10/11/2017] [Indexed: 01/16/2023] Open
Abstract
The prefrontal cortex (PFC) regulates emotional behavior via top-down control of the basolateral amygdala (BLA). However, the influence of PFC inputs on the different projection pathways within the BLA remains largely unexplored. Here, we combine whole-cell recordings and optogenetics to study these cell-type specific connections in mouse BLA. We characterize PFC inputs onto three distinct populations of BLA neurons that project to the PFC, ventral hippocampus, or nucleus accumbens. We find that PFC-evoked synaptic responses are strongest at amygdala-cortical and amygdala-hippocampal neurons and much weaker at amygdala-striatal neurons. We assess the mechanisms for this targeting and conclude that it reflects fewer connections onto amygdala-striatal neurons. Given the similar intrinsic properties of these cells, this connectivity allows the PFC to preferentially activate amygdala-cortical and amygdala-hippocampal neurons. Together, our findings reveal how PFC inputs to the BLA selectively drive feedback projections to the PFC and feedforward projections to the hippocampus.
Collapse
Affiliation(s)
- Laura M McGarry
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Adam G Carter
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA.
| |
Collapse
|
104
|
Wei J, Zhong P, Qin L, Tan T, Yan Z. Chemicogenetic Restoration of the Prefrontal Cortex to Amygdala Pathway Ameliorates Stress-Induced Deficits. Cereb Cortex 2018; 28:1980-1990. [PMID: 28498919 PMCID: PMC6018994 DOI: 10.1093/cercor/bhx104] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/27/2017] [Indexed: 01/11/2023] Open
Abstract
Corticosteroid stress hormones exert a profound impact on cognitive and emotional processes. Understanding the neuronal circuits that are altered by chronic stress is important for counteracting the detrimental effects of stress in a brain region- and cell type-specific manner. Using the chemogenetic tool, Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), which enables the remote, noninvasive and long-lasting modulation of cellular activity and signal transduction in discrete neuronal populations in vivo, we sought to identify the specific pathways that play an essential role in stress responses. We found that prolonged severe stress induced the diminished glutamatergic projection from pyramidal neurons in prefrontal cortex (PFC) to GABAergic interneurons in basolateral amygdala (BLA), leading to the loss of feedforward inhibition and ensuing hyperexcitability of BLA principal neurons, which caused a variety of behavioral abnormalities. Activating PFC pyramidal neurons with hM3D(Gq) DREADD restored the functional connection between PFC and BLA in stressed animals, resulting in the rescue of recognition memory, normalization of locomotor activity and reduction of aggressive behaviors. Inhibiting BLA principal neurons directly with hM4D(Gi) DREADD also blocked BLA hyperactivity and aggressive behaviors in stressed animals. These results have offered an effective avenue to counteract the stress-induced disruption of circuitry homeostasis.
Collapse
Affiliation(s)
- Jing Wei
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Luye Qin
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Tao Tan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| |
Collapse
|
105
|
Krabbe S, Gründemann J, Lüthi A. Amygdala Inhibitory Circuits Regulate Associative Fear Conditioning. Biol Psychiatry 2018; 83:800-809. [PMID: 29174478 DOI: 10.1016/j.biopsych.2017.10.006] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 11/16/2022]
Abstract
Associative memory formation is essential for an animal's survival by ensuring adaptive behavioral responses in an ever-changing environment. This is particularly important under conditions of immediate threats such as in fear learning. One of the key brain regions involved in associative fear learning is the amygdala. The basolateral amygdala is the main entry site for sensory information to the amygdala complex, and local plasticity in excitatory basolateral amygdala principal neurons is considered to be crucial for learning of conditioned fear responses. However, activity and plasticity of excitatory circuits are tightly controlled by local inhibitory interneurons in a spatially and temporally defined manner. In this review, we provide an updated view on how distinct interneuron subtypes in the basolateral amygdala contribute to the acquisition and extinction of conditioned fear memories.
Collapse
Affiliation(s)
- Sabine Krabbe
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Jan Gründemann
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Andreas Lüthi
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
106
|
Jiang H, Kim HF. Anatomical Inputs From the Sensory and Value Structures to the Tail of the Rat Striatum. Front Neuroanat 2018; 12:30. [PMID: 29773980 PMCID: PMC5943565 DOI: 10.3389/fnana.2018.00030] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/05/2018] [Indexed: 11/17/2022] Open
Abstract
The caudal region of the rodent striatum, called the tail of the striatum (TS), is a relatively small area but might have a distinct function from other striatal subregions. Recent primate studies showed that this part of the striatum has a unique function in encoding long-term value memory of visual objects for habitual behavior. This function might be due to its specific connectivity. We identified inputs to the rat TS and compared those with inputs to the dorsomedial striatum (DMS) in the same animals. The TS directly received anatomical inputs from both sensory structures and value-coding regions, but the DMS did not. First, inputs from the sensory cortex and sensory thalamus to the TS were found; visual, auditory, somatosensory and gustatory cortex and thalamus projected to the TS but not to the DMS. Second, two value systems innervated the TS; dopamine and serotonin neurons in the lateral part of the substantia nigra pars compacta (SNc) and dorsal raphe nucleus projected to the TS, respectively. The DMS received inputs from the separate group of dopamine neurons in the medial part of the SNc. In addition, learning-related regions of the limbic system innervated the TS; the temporal areas and the basolateral amygdala selectively innervated the TS, but not the DMS. Our data showed that both sensory and value-processing structures innervated the TS, suggesting its plausible role in value-guided sensory-motor association for habitual behavior.
Collapse
Affiliation(s)
- Haiyan Jiang
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, South Korea.,Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, South Korea
| | - Hyoung F Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, South Korea.,Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, South Korea
| |
Collapse
|
107
|
Hiser J, Koenigs M. The Multifaceted Role of the Ventromedial Prefrontal Cortex in Emotion, Decision Making, Social Cognition, and Psychopathology. Biol Psychiatry 2018; 83:638-647. [PMID: 29275839 PMCID: PMC5862740 DOI: 10.1016/j.biopsych.2017.10.030] [Citation(s) in RCA: 583] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 10/06/2017] [Accepted: 10/30/2017] [Indexed: 12/21/2022]
Abstract
The ventromedial prefrontal cortex (vmPFC) has been implicated in a variety of social, cognitive, and affective functions that are commonly disrupted in mental illness. In this review, we summarize data from a diverse array of human and animal studies demonstrating that the vmPFC is a key node of cortical and subcortical networks that subserve at least three broad domains of psychological function linked to psychopathology. One track of research indicates that the vmPFC is critical for the representation of reward- and value-based decision making, through interactions with the ventral striatum and amygdala. A second track of research demonstrates that the vmPFC is critical for the generation and regulation of negative emotion, through its interactions with the amygdala, bed nucleus of the stria terminalis, periaqueductal gray, hippocampus, and dorsal anterior cingulate cortex. A third track of research shows the importance of the vmPFC in multiple aspects of social cognition, such as facial emotion recognition, theory-of-mind ability, and processing self-relevant information, through its interactions with the posterior cingulate cortex, precuneus, dorsomedial PFC, and amygdala. We then present meta-analytic data revealing distinct subregions within the vmPFC that correspond to each of these three functions, as well as the associations between these subregions and specific psychiatric disorders (depression, posttraumatic stress disorder, addiction, social anxiety disorder, bipolar disorder, schizophrenia, and attention-deficit/hyperactivity disorder). We conclude by describing several translational possibilities for clinical studies of vmPFC-based circuits, including neuropsychological assessment of transdiagnostic functions, anatomical targets for intervention, predictors of treatment response, markers of treatment efficacy, and subtyping within disorders.
Collapse
Affiliation(s)
- Jaryd Hiser
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI,
USA,Department of Psychology, University of Wisconsin-Madison, Madison, WI,
USA
| | - Michael Koenigs
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
108
|
Parnaudeau S, Bolkan SS, Kellendonk C. The Mediodorsal Thalamus: An Essential Partner of the Prefrontal Cortex for Cognition. Biol Psychiatry 2018; 83:648-656. [PMID: 29275841 PMCID: PMC5862748 DOI: 10.1016/j.biopsych.2017.11.008] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 01/17/2023]
Abstract
Deficits in cognition are a core feature of many psychiatric conditions, including schizophrenia, where the severity of such deficits is a strong predictor of long-term outcome. Impairment in cognitive domains such as working memory and behavioral flexibility has typically been associated with prefrontal cortex (PFC) dysfunction. However, there is increasing evidence that the PFC cannot be dissociated from its main thalamic counterpart, the mediodorsal thalamus (MD). Since the causal relationships between MD-PFC abnormalities and cognitive impairment, as well as the neuronal mechanisms underlying them, are difficult to address in humans, animal models have been employed for mechanistic insight. In this review, we discuss anatomical, behavioral, and electrophysiological findings from animal studies that provide a new understanding on how MD-PFC circuits support higher-order cognitive function. We argue that the MD may be required for amplifying and sustaining cortical representations under different behavioral conditions. These findings advance a new framework for the broader involvement of distributed thalamo-frontal circuits in cognition and point to the MD as a potential therapeutic target for improving cognitive deficits in schizophrenia and other disorders.
Collapse
Affiliation(s)
- Sébastien Parnaudeau
- Sorbonne Universités, Université Pierre et Marie Curie Paris 06, Institut de Biologie Paris Seine UM119, Neuroscience Paris Seine, Centre National de la Recherche Scientifique UMR8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Scott S Bolkan
- Graduate Program in Neurobiology and Behavior, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Christoph Kellendonk
- Departments of Pharmacology and Psychiatry, Columbia University, College of Physicians and Surgeons, New York, New York; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York.
| |
Collapse
|
109
|
Jacob Y, Gilam G, Lin T, Raz G, Hendler T. Anger Modulates Influence Hierarchies Within and Between Emotional Reactivity and Regulation Networks. Front Behav Neurosci 2018; 12:60. [PMID: 29681803 PMCID: PMC5897670 DOI: 10.3389/fnbeh.2018.00060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/15/2018] [Indexed: 12/31/2022] Open
Abstract
Emotion regulation is hypothesized to be mediated by the interactions between emotional reactivity and regulation networks during the dynamic unfolding of the emotional episode. Yet, it remains unclear how to delineate the effective relationships between these networks. In this study, we examined the aforementioned networks’ information flow hierarchy during viewing of an anger provoking movie excerpt. Anger regulation is particularly essential for averting individuals from aggression and violence, thus improving prosocial behavior. Using subjective ratings of anger intensity we differentiated between low and high anger periods of the film. We then applied the Dependency Network Analysis (DEPNA), a newly developed graph theory method to quantify networks’ node importance during the two anger periods. The DEPNA analysis revealed that the impact of the ventromedial prefrontal cortex (vmPFC) was higher in the high anger condition, particularly within the regulation network and on the connections between the reactivity and regulation networks. We further showed that higher levels of vmPFC impact on the regulation network were associated with lower subjective anger intensity during the high-anger cinematic period, and lower trait anger levels. Supporting and replicating previous findings, these results emphasize the previously acknowledged central role of vmPFC in modulating negative affect. We further show that the impact of the vmPFC relies on its correlational influence on the connectivity between reactivity and regulation networks. More importantly, the hierarchy network analysis revealed a link between connectivity patterns of the vmPFC and individual differences in anger reactivity and trait, suggesting its potential therapeutic role.
Collapse
Affiliation(s)
- Yael Jacob
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Center for Brain Functions, Wohl Institute for Advanced Imaging, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gadi Gilam
- Tel Aviv Center for Brain Functions, Wohl Institute for Advanced Imaging, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel.,Systems Neuroscience and Pain Laboratory, Department of Anesthesia, Perioperative and Pain Medicine, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Tamar Lin
- Tel Aviv Center for Brain Functions, Wohl Institute for Advanced Imaging, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gal Raz
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Center for Brain Functions, Wohl Institute for Advanced Imaging, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Film and Television Department, Tel Aviv University, Tel Aviv, Israel
| | - Talma Hendler
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Center for Brain Functions, Wohl Institute for Advanced Imaging, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
110
|
Dario MFR, Sara T, Estela CO, Margarita PM, Guillermo ET, Fernando RDF, Javier SL, Carmen P. Stress, Depression, Resilience and Ageing: A Role for the LPA-LPA1 Pathway. Curr Neuropharmacol 2018; 16:271-283. [PMID: 28699486 PMCID: PMC5843979 DOI: 10.2174/1570159x15666170710200352] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/26/2017] [Accepted: 06/30/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Chronic stress affects health and the quality of life, with its effects being particularly relevant in ageing due to the psychobiological characteristics of this population. However, while some people develop psychiatric disorders, especially depression, others seem very capable of dealing with adversity. There is no doubt that along with the identification of neurobiological mechanisms involved in developing depression, discovering which factors are involved in positive adaptation under circumstances of extreme difficulty will be crucial for promoting resilience. METHODS Here, we review recent work in our laboratory, using an animal model lacking the LPA1 receptor, together with pharmacological studies and clinical evidence for the possible participation of the LPA1 receptor in mood and resilience to stress. RESULTS Substantial evidence has shown that the LPA1 receptor is involved in emotional regulation and in coping responses to chronic stress, which, if dysfunctional, may induce vulnerability to stress and predisposition to the development of depression. Given that there is commonality of mechanisms between those involved in negative consequences of stress and in ageing, this is not surprising, considering that the LPA1 receptor may be involved in coping with adversity during ageing. CONCLUSION Alterations in this receptor may be a susceptibility factor for the presence of depression and cognitive deficits in the elderly population. However, because this is only a promising hypothesis based on previous data, future studies should focus on the involvement of the LPA-LPA1 pathway in coping with stress and resilience in ageing.
Collapse
Affiliation(s)
- Moreno-Fernández Román Dario
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Tabbai Sara
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Castilla-Ortega Estela
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga; Málaga 29010, Spain
| | - Pérez-Martín Margarita
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de
Málaga; Málaga 29071, Spain
| | - Estivill-Torrús Guillermo
- Unidad de Gestión Clínica de Neurociencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitarios de Málaga, Málaga, Spain
| | - Rodríguez de Fonseca Fernando
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga; Málaga 29010, Spain
| | - Santin Luis Javier
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| | - Pedraza Carmen
- Departamento de Psicobiología y Metodología de las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga; Málaga 29071, Spain
| |
Collapse
|
111
|
Rozeske RR, Jercog D, Karalis N, Chaudun F, Khoder S, Girard D, Winke N, Herry C. Prefrontal-Periaqueductal Gray-Projecting Neurons Mediate Context Fear Discrimination. Neuron 2018; 97:898-910.e6. [DOI: 10.1016/j.neuron.2017.12.044] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/07/2017] [Accepted: 12/27/2017] [Indexed: 01/22/2023]
|
112
|
Chang YH, Liu SW, Chang CH. Pharmacological activation of the lateral orbitofrontal cortex on regulation of learned fear and extinction. Neurobiol Learn Mem 2018; 148:30-37. [DOI: 10.1016/j.nlm.2017.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 12/18/2017] [Accepted: 12/29/2017] [Indexed: 11/26/2022]
|
113
|
Rubio-Casillas A, Fernández-Guasti A. The dose makes the poison: from glutamate-mediated neurogenesis to neuronal atrophy and depression. Rev Neurosci 2018; 27:599-622. [PMID: 27096778 DOI: 10.1515/revneuro-2015-0066] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/04/2016] [Indexed: 12/21/2022]
Abstract
Experimental evidence has demonstrated that glutamate is an essential factor for neurogenesis, whereas another line of research postulates that excessive glutamatergic neurotransmission is associated with the pathogenesis of depression. The present review shows that such paradox can be explained within the framework of hormesis, defined as biphasic dose responses. Low glutamate levels activate adaptive stress responses that include proteins that protect neurons against more severe stress. Conversely, abnormally high levels of glutamate, resulting from increased release and/or decreased removal, cause neuronal atrophy and depression. The dysregulation of the glutamatergic transmission in depression could be underlined by several factors including a decreased inhibition (γ-aminobutyric acid or serotonin) or an increased excitation (primarily within the glutamatergic system). Experimental evidence shows that the activation of N-methyl-D-aspartate receptor (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPAR) can exert two opposite effects on neurogenesis and neuron survival depending on the synaptic or extrasynaptic concentration. Chronic stress, which usually underlies experimental and clinical depression, enhances glutamate release. This overactivates NMDA receptors (NMDAR) and consequently impairs AMPAR activity. Various studies show that treatment with antidepressants decreases plasma glutamate levels in depressed individuals and regulates glutamate receptors by reducing NMDAR function by decreasing the expression of its subunits and by potentiating AMPAR-mediated transmission. Additionally, it has been shown that chronic treatment with antidepressants having divergent mechanisms of action (including tricyclics, selective serotonin reuptake inhibitors, and ketamine) markedly reduced depolarization-evoked glutamate release in the hippocampus. These data, taken together, suggest that the glutamatergic system could be a final common pathway for antidepressant treatments.
Collapse
|
114
|
Memory Retention Involves the Ventrolateral Orbitofrontal Cortex: Comparison with the Basolateral Amygdala. Neuropsychopharmacology 2018; 43:373-383. [PMID: 28664926 PMCID: PMC5729558 DOI: 10.1038/npp.2017.139] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 11/08/2022]
Abstract
The orbitofrontal cortex (OFC) is thought to link stimuli and actions with anticipated outcomes in order to sustain flexible behavior in an ever-changing environment. How it retains these associations to guide future behavior is less well-defined. Here we focused on one subregion of this heterogeneous structure, the ventrolateral OFC (VLO). CaMKII-driven inhibitory Gi-coupled designer receptors exclusively activated by designer drugs (DREADDs) were infused and subsequently activated by their ligand Clozapine-N-oxide (CNO) in conjunction with fear extinction training (a form of aversive conditioning) and response-outcome conditioning (a form of appetitive conditioning). Gi-DREADD-mediated inactivation of the VLO during extinction conditioning interfered with fear extinction memory, resulting in sustained freezing when mice were later tested drug-free. Similarly, Gi-DREADD-mediated inactivation in conjunction with response-outcome conditioning caused a later decay in goal-directed responding-that is, mice were unable to select actions based on the likelihood that they would be rewarded in a sustainable manner. By contrast, inhibitory Gi-DREADDs in the basolateral amygdala (BLA) impaired the acquisition of both conditioned fear extinction and response-outcome conditioning, as expected based on prior studies using other inactivation techniques. Meanwhile, DREADD-mediated inhibition of the dorsolateral striatum enhanced response-outcome conditioning, also in line with prior reports. Together, our findings suggest that learning-related neuroplasticity in the VLO may be necessary for memory retention in both appetitive and aversive domains.
Collapse
|
115
|
Saitoh A, Suzuki S, Soda A, Ohashi M, Yamada M, Oka JI, Nagase H, Yamada M. The delta opioid receptor agonist KNT-127 in the prelimbic medial prefrontal cortex attenuates veratrine-induced anxiety-like behaviors in mice. Behav Brain Res 2018; 336:77-84. [DOI: 10.1016/j.bbr.2017.08.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/21/2017] [Accepted: 08/28/2017] [Indexed: 10/19/2022]
|
116
|
Abstract
The activity of neural circuits that underpin particular behaviours are one of the most interesting questions in neurobiology today. This understanding will not only lead to a detailed understanding of learning and memory formation, but also provides a platform for the development of novel therapeutic approaches to a range of neurological disorders that afflict humans. Among the different behavioural paradigms, Pavlovian fear conditioning and its extinction are two of the most extensively used to study acquisition, consolidation and retrieval of fear-related memories. The amygdala, medial prefrontal cortex (mPFC) and hippocampus are three regions with extensive bidirectional connections, and play key roles in fear processing. In this chapter, we summarise our current understanding of the structure and physiological role of these three regions in fear learning and extinction.
Collapse
Affiliation(s)
- Roger Marek
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Pankaj Sah
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
117
|
Heslin K, Coutellier L. Npas4 deficiency and prenatal stress interact to affect social recognition in mice. GENES BRAIN AND BEHAVIOR 2018; 17:e12448. [PMID: 29227584 DOI: 10.1111/gbb.12448] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/30/2017] [Accepted: 12/07/2017] [Indexed: 01/21/2023]
Abstract
Neurodevelopmental disorders such as autism spectrum disorders and schizophrenia have an expansive array of reported genetic and environmental contributing factors. However, none of these factors alone can account for a substantial proportion of cases of either disorder. Instead, many gene-by-environment interactions are responsible for neurodevelopmental disturbances that lead to these disorders. The current experiment used heterozygous knock-out mice to examine a potential interaction between 2 factors commonly linked to neurodevelopmental disorders and cognitive deficit: imbalanced excitatory/inhibitory signaling in the cortex and prenatal stress (PNS) exposure. Both of these factors have been linked to disrupt GABAergic signaling in the prefrontal cortex (PFC), a common feature of neurodevelopmental disorders. The neuronal PAS domain protein 4 (Npas4) gene is instrumental in regulation of the excitatory/inhibitory balance in the cortex and hippocampus in response to activation. Npas4 heterozygous and wild-type male and female mice were exposed to either PNS or standard gestation, then evaluated during adulthood in social and anxiety behavioral measures. The combination of PNS and Npas4 deficiency in male mice impaired social recognition. This behavioral deficit was associated with decreased parvalbumin and cFos protein expression in the infralimbic region of the PFC following social stimulation in Npas4 heterozygous males. In contrast, females displayed fewer behavioral effects and molecular changes in PFC in response to PNS and decreased Npas4.
Collapse
Affiliation(s)
- K Heslin
- Department of Psychology, The Ohio State University, Columbus, Ohio
| | - L Coutellier
- Department of Psychology, The Ohio State University, Columbus, Ohio.,Department of Neuroscience, The Ohio State University, Columbus, Ohio
| |
Collapse
|
118
|
Saha R, Shrivastava K, Jing L, Schayek R, Maroun M, Kriebel M, Volkmer H, Richter-Levin G. Perturbation of GABAergic Synapses at the Axon Initial Segment of Basolateral Amygdala Induces Trans-regional Metaplasticity at the Medial Prefrontal Cortex. Cereb Cortex 2017; 28:395-410. [DOI: 10.1093/cercor/bhx300] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Indexed: 12/14/2022] Open
Affiliation(s)
- Rinki Saha
- Sagol Department of Neurobiology, University of Haifa, Haifa 31905, Israel
| | | | - Liang Jing
- The Institute for the Study of Affective Neuroscience, University of Haif, Haifa 31905, Israel
| | - Rachel Schayek
- Sagol Department of Neurobiology, University of Haifa, Haifa 31905, Israel
| | - Mouna Maroun
- Sagol Department of Neurobiology, University of Haifa, Haifa 31905, Israel
| | - Martin Kriebel
- NMI Natural and Medical Sciences Institute, University of Tübingen, 72770 Reutlingen, Germany
| | - Hansjürgen Volkmer
- NMI Natural and Medical Sciences Institute, University of Tübingen, 72770 Reutlingen, Germany
| | - Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Haifa 31905, Israel
- Department of Psychology, University of Haifa, Haifa 31905, Israel
- The Institute for the Study of Affective Neuroscience, University of Haif, Haifa 31905, Israel
| |
Collapse
|
119
|
N-palmitoylethanolamide in the anterior cingulate cortex attenuates inflammatory pain behaviour indirectly via a CB1 receptor-mediated mechanism. Pain 2017; 157:2687-2696. [PMID: 27649266 DOI: 10.1097/j.pain.0000000000000687] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The neural substrates and mechanisms mediating the antinociceptive effects of the endogenous bioactive lipid, N-palmitoylethanolamide (PEA), require further investigation. We investigated the effects of exogenous PEA administration into the anterior cingulate cortex (ACC), an important brain region linked with cognitive and affective modulation of pain, on formalin-evoked nociceptive behaviour in rats. Potential involvement of peroxisome proliferator-activated receptor isoforms (PPAR) α and γ or endocannabinoid-mediated entourage effects at cannabinoid1 (CB1) receptors or transient receptor potential subfamily V member 1 (TRPV1) in mediating the effects of PEA was also investigated. Intra-ACC administration of PEA significantly attenuated the first and early second phases of formalin-evoked nociceptive behaviour. This effect was attenuated by the CB1 receptor antagonist AM251, but not by the PPARα antagonist GW6471, the PPARγ antagonist GW9662, or the TRPV1 antagonist 5'-iodo resiniferatoxin. All antagonists, administered alone, significantly reduced formalin-evoked nociceptive behaviour, suggesting facilitatory/permissive roles for these receptors in the ACC in inflammatory pain. Post-mortem tissue analysis revealed a strong trend for increased levels of the endocannabinoid anandamide in the ACC of rats that received intra-ACC PEA. Expression of c-Fos, a marker of neuronal activity, was significantly reduced in the basolateral nucleus of the amygdala, but not in the central nucleus of the amygdala, the rostral ventromedial medulla or the dorsal horn of the spinal cord. In conclusion, these data indicate that PEA in the ACC can reduce inflammatory pain-related behaviour, possibly via AEA-induced activation of CB1 receptors and associated modulation of neuronal activity in the basolateral amygdala.
Collapse
|
120
|
Amygdala-mediated mechanisms regulate visceral hypersensitivity in adult females following early life stress: importance of the glucocorticoid receptor and corticotropin-releasing factor. Pain 2017; 158:296-305. [PMID: 27849648 DOI: 10.1097/j.pain.0000000000000759] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alterations in amygdala activity are apparent in women who report a history of early life stress (ELS) and those diagnosed with chronic pain disorders. Chronic stress in adulthood induces visceral hypersensitivity by alterations in glucocorticoid receptor (GR) and corticotropin-releasing factor (CRF) expression within the central amygdala (CeA). Here, we hypothesized that unpredictable ELS, previously shown to induce visceral hypersensitivity in adult female rats, alters GR and CRF expression in the CeA. After neonatal ELS, visceral sensitivity and GR and CRF gene expression were quantified in adult female rats. After unpredictable ELS, adult female rats exhibited visceral hypersensitivity and increased expression of GR and CRF in the CeA. After predictable ELS, adult female rats demonstrated normosensitive behavioral pain responses and upregulation of GR but not CRF in the CeA. After the ELS paradigms, visceral sensitivity and gene expression within the CeA were unaffected in adult male rats. The role of GR and CRF in modulating visceral sensitivity in adult female rats after ELS was investigated using oligodeoxynucleotide sequences targeted to the CeA for knockdown of GR or CRF. Knockdown of GR increased visceral sensitivity in all rats but revealed an exaggerated visceral hypersensitivity in females with a history of predictable or unpredictable ELS compared with that of controls. Knockdown of CRF expression or antagonism of CRF1R in the CeA attenuated visceral hypersensitivity after unpredictable ELS. This study highlights a shift in GR and CRF regulation within the CeA after ELS that underlies the development of visceral hypersensitivity in adulthood.
Collapse
|
121
|
Plasticity in the Interoceptive System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:59-74. [DOI: 10.1007/978-3-319-62817-2_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
122
|
Schienle A, Übel S, Wabnegger A. When opposites lead to the same: a direct comparison of explicit and implicit disgust regulation via fMRI. Soc Cogn Affect Neurosci 2017; 12:445-451. [PMID: 27665000 PMCID: PMC5390737 DOI: 10.1093/scan/nsw144] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/21/2016] [Indexed: 11/24/2022] Open
Abstract
Cognitive reappraisal and placebo administration constitute two different approaches for modulating one’s own emotional state. Whereas reappraisal is an explicit (effortful) type of self-regulation, placebo treatment initiates implicit processes of affective control. The brain mechanisms underlying these processes have not been directly compared with each other up until now; doing this enables the identification of distinct and shared neuronal features. We conducted a functional magnetic resonance imaging study with 45 women, who were presented with disgusting and neutral images in a block design, at three experimental sessions, over 3 consecutive days. They were asked to passively view the images in one session, engage in reappraisal in another, and in another session they received a placebo pill: a disgust-reducing ‘anti-nausea drug’. Relative to passive viewing, both reappraisal and placebo treatment effectively reduced the experienced disgust intensity. In the placebo condition, this reduction was associated with decreased activation of the insula and the dorsolateral prefrontal cortex (DLPFC). In contrast, reappraisal induced increased activation in both regions. Furthermore, both regulation strategies were associated with opposite patterns of connectivity in a network encompassing the amygdala, the insula and the DLPFC. Only placebo administration led to a reduced coupling in this network.
Collapse
|
123
|
Siddiqui SA, Singh S, Ranjan V, Ugale R, Saha S, Prakash A. Enhanced Histone Acetylation in the Infralimbic Prefrontal Cortex is Associated with Fear Extinction. Cell Mol Neurobiol 2017; 37:1287-1301. [PMID: 28097489 PMCID: PMC11482208 DOI: 10.1007/s10571-017-0464-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/09/2017] [Indexed: 12/20/2022]
Abstract
The molecular processes that establish fear memory are complex and involve a combination of genetic and epigenetic influences. Dysregulation of these processes can manifest in humans as a range of fear-related anxiety disorders like post-traumatic stress disorders (PTSD). In the present study, immunohistochemistry for acetyl H3, H4, c-fos, CBP (CREB-binding protein) in the infralimbic prefrontal cortex (IL-PFC) and prelimbic prefrontal cortex (PL-PFC) of mPFC (medial prefrontal cortex) and basal amygdala (BA), lateral amygdala (LA), centrolateral amygdala (CeL), centromedial amygdala (CeM) of the amygdala was performed to link region-specific histone acetylation to fear and extinction learning. It was found that the PL-PFC and IL-PFC along with the sub-regions of the amygdala responded differentially to the fear learning and extinction. Following fear learning, c-fos and CBP expression and acetylation of H3 and H4 increased in the BA, LA, CeM, and CeL and the PL-PFC but not in the IL-PFC as compared to the naive control. Similarly, following extinction learning, c-fos and CBP expression increased in BA, LA, CeL, and IL-PFC but not in PL-PFC and CeM as compared to the naive control and conditioned group. However, the acetylation of H3 increased in both IL and PL as opposed to H4 which increased only in the IL-PFC following extinction learning. Overall, region-specific activation in amygdala and PFC following fear and extinction learning as evident by the c-fos activation paralleled the H3/H4 acetylation in these regions. These results suggest that the differential histone acetylation in the PFC and amygdala subnuclei following fear learning and extinction may be associated with the region-specific changes in the neuronal activation pattern resulting in more fear/less fear.
Collapse
Affiliation(s)
| | - Sanjay Singh
- Department of Biotechnology, BabasahebBhimrao Ambedkar University, Lucknow, India
| | - Vandana Ranjan
- Department of Biotechnology, BabasahebBhimrao Ambedkar University, Lucknow, India
| | - Rajesh Ugale
- Department of Pharmaceutical Sciences, RTM Nagpur University, Nagpur, India
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, BabasahebBhimrao Ambedkar University, Lucknow, India
| | - Anand Prakash
- Department of Biotechnology, BabasahebBhimrao Ambedkar University, Lucknow, India.
| |
Collapse
|
124
|
Wu ZH, Zhang QJ, Du CX, Xi Y, Li WJ, Guo FY, Yu SQ, Yang YX, Liu J. Prelimbic α1-adrenoceptors are involved in the regulation of depressive-like behaviors in the hemiparkinsonian rats. Brain Res Bull 2017; 134:99-108. [DOI: 10.1016/j.brainresbull.2017.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 07/04/2017] [Accepted: 07/10/2017] [Indexed: 01/21/2023]
|
125
|
Chang CH, Ho TW. Inhibitory modulation of medial prefrontal cortical activation on lateral orbitofrontal cortex-amygdala information flow. J Physiol 2017; 595:6065-6076. [PMID: 28678402 DOI: 10.1113/jp274568] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/29/2017] [Indexed: 01/20/2023] Open
Abstract
KEY POINTS The basolateral complex of the amygdala (BLA) receives input from the lateral orbitofrontal cortex (lOFC) for cue-outcome contingencies and the medial prefrontal cortex (mPFC) for emotion control. Here we examined how the mPFC modulates lOFC-BLA information flow. We found that the majority of BLA neurons responsive to lOFC stimulation were also responsive to mPFC stimulation. Activation of the mPFC exerted an inhibitory modulation of the lOFC-BLA pathway, which was reversed with intra-amygdala blockade of GABAergic receptors. mPFC tetanus potentiated the lOFC-BLA pathway, but did not alter its inhibitory modulatory gating. These results show that the mPFC potently inhibits lOFC drive of the BLA in a GABA-dependent manner, which is informative in understanding the normal and potential pathophysiological state of emotion and contingency associations in regulating behaviour. ABSTRACT Several neocortical projections converge onto the basolateral complex of the amygdala (BLA), including the lateral orbitofrontal cortex (lOFC) and the medial prefrontal cortex (mPFC). Lateral orbitofrontal input to the BLA is important for cue-outcome contingencies, while medial prefrontal input is essential for emotion control. In this study, we examined how the mPFC, specifically the infralimbic division of the mPFC, modulates lOFC-BLA information flow, using combined in vivo extracellular single-unit recordings and pharmacological manipulations in anaesthetized rats. We found that the majority (over 95%) of BLA neurons that responded to lOFC stimulation also responded to mPFC stimulation. Compared to basal condition, pharmacological (N-methyl-d-aspartate) or electrical activation of the mPFC exerted an inhibitory modulation of the lOFC-BLA pathway, which was reversed with intra-amygdala blockade of GABAergic receptors with combined GABAA and GABAB antagonists (bicuculline and saclofen). Moreover, mPFC tetanus potentiated the lOFC-BLA pathway, but mPFC tetanus or low-frequency stimulation did not alter its inhibitory modulatory gating on the lOFC-BLA pathway. These results show that the mPFC potently inhibits lOFC drive of BLA neurons in a GABA-dependent manner. Our result is informative in understanding the normal and potential pathophysiological state of emotion and contingency associations regulating behaviour.
Collapse
Affiliation(s)
- Chun-Hui Chang
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan, 30013
| | - Ta-Wen Ho
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, 30013
| |
Collapse
|
126
|
Differential Expression of Munc13-2 Produces Unique Synaptic Phenotypes in the Basolateral Amygdala of C57BL/6J and DBA/2J Mice. J Neurosci 2017; 36:10964-10977. [PMID: 27798178 DOI: 10.1523/jneurosci.1785-16.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/28/2016] [Indexed: 11/21/2022] Open
Abstract
C57BL/6J (B6) and DBA/2J (D2) mice are well known to differentially express a number of behavioral phenotypes, including anxiety-like behavior, fear conditioning, and drug self-administration. However, the cellular mechanisms contributing to these differences remain unclear. Given the basolateral amygdala (BLA) contributes to these behaviors, we characterized strain-dependent differences in presynaptic and postsynaptic function in BLA neurons by integrating electrophysiological, biochemical, and genetic approaches to identify specific molecular mechanisms. We found that D2 glutamatergic synapses expressed enhanced release probability and lower sensitivity to both the inhibitory effects of low extracellular calcium and facilitation by phorbol esters. Furthermore, repetitive stimulation of BLA afferents at low (2 Hz) or high (40 Hz) frequencies revealed that B6 terminals, relative to D2 terminals, were more sensitive to synaptic fatigue principally because of reduced vesicle recycling rates. Additionally, B6 synapses exhibited more robust augmentation of spontaneous release after repetitive stimulation relative to the D2 strain. In silico analysis of the inheritance of synaptic physiology from an array of BXD recombinant inbred strains (Jansen et al., 2011) identified a segment on chromosome 4 containing the gene encoding Munc13-2, which has calcium-/phorbol ester-binding domains and controls presynaptic function. We subsequently found that B6 mice express substantially higher levels of Munc13-2 compared with the D2 strain whereas expression of several release-related proteins, including Munc13-1, was equivalent. We then knocked down the expression of Munc13-2 in B6 mice using a short hairpin RNA and found this recapitulated the presynaptic phenotype of D2 BLA synapses. SIGNIFICANCE STATEMENT DBA/2J and C57BL/6J mice have been used to understand the genetic mechanisms controlling behaviors related to a number of psychiatric illnesses. However, the fundamental neurobiological mechanisms producing these behavioral characteristics remain unresolved. Here we identify a critical family of presynaptic proteins differentially expressed by these strains that control strain-dependent synaptic physiology. This family of proteins regulates excitation/secretion coupling, vesicle recycling, and short-term plasticity throughout the CNS. Thus, differential inheritance of proteins like Munc13-2 has broad implications for genetic control over a wide variety of pathological behaviors. Importantly, these proteins also contain a large number of modulatory sites, making them attractive potential targets for the development of novel neuropharmaceutical treatments.
Collapse
|
127
|
Sabihi S, Dong SM, Maurer SD, Post C, Leuner B. Oxytocin in the medial prefrontal cortex attenuates anxiety: Anatomical and receptor specificity and mechanism of action. Neuropharmacology 2017; 125:1-12. [PMID: 28655609 DOI: 10.1016/j.neuropharm.2017.06.024] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 01/12/2023]
Abstract
Numerous studies in animals and humans have established that oxytocin (OT) reduces anxiety. In rats, the prelimbic (PL) subregion of the medial prefrontal cortex (mPFC) is among the brain areas implicated in the anxiolytic actions of OT. However, questions remain about the anatomical and receptor specificity of OT and its mechanism of action. Here we assessed whether the regulation of anxiety by mPFC OT is restricted to the PL subregion and evaluated whether oxytocin receptor (OTR) activation is required for OT to have an anxiolytic effect. We also examined whether OT interacts with GABA in the mPFC to reduce anxiety and investigated the extent to which OT in the mPFC affects activation of mPFC GABA neurons as well as neuronal activation in the amygdala, a primary target of the mPFC which is part of the neural network regulating anxiety. We found that OT reduced anxiety-like behavior when delivered to the PL, but not infralimbic or anterior cingulate subregions of the mPFC. The anxiolytic effect of OT in the PL mPFC was blocked by pretreatment with an OTR, but not a vasopressin receptor, antagonist as well as with a GABAA receptor antagonist. Lastly, administration of OT to the PL mPFC was accompanied by increased activation of GABA neurons in the PL mPFC and altered neuronal activation of the amygdala following anxiety testing. These results demonstrate that OT in the PL mPFC attenuates anxiety-related behavior and may do so by engaging GABAergic neurons which ultimately modulate downstream brain regions implicated in anxiety.
Collapse
Affiliation(s)
- Sara Sabihi
- The Ohio State University, Department of Psychology, Columbus, OH 43210, United States
| | - Shirley M Dong
- The Ohio State University, Department of Psychology, Columbus, OH 43210, United States
| | - Skyler D Maurer
- The Ohio State University, Department of Psychology, Columbus, OH 43210, United States
| | - Caitlin Post
- The Ohio State University, Department of Psychology, Columbus, OH 43210, United States
| | - Benedetta Leuner
- The Ohio State University, Department of Psychology, Columbus, OH 43210, United States; The Ohio State University, Department of Neuroscience, Columbus, OH 43210, United States; The Ohio State University, Behavioral Neuroendocrinology Group, Columbus, OH 43210, United States.
| |
Collapse
|
128
|
Cádiz-Moretti B, Abellán-Álvaro M, Pardo-Bellver C, Martínez-García F, Lanuza E. Afferent and efferent projections of the anterior cortical amygdaloid nucleus in the mouse. J Comp Neurol 2017; 525:2929-2954. [DOI: 10.1002/cne.24248] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 01/26/2023]
Affiliation(s)
- Bernardita Cádiz-Moretti
- Unitat Mixta de Neuroanatomia Funcional UV-UJI - Dept. de Biologia Cel·lular i Biologia Funcional, Facultat de Ciències Biològiques, Universitat de València; Burjassot 46100 València Spain
| | - María Abellán-Álvaro
- Unitat Mixta de Neuroanatomia Funcional UV-UJI - Dept. de Biologia Cel·lular i Biologia Funcional, Facultat de Ciències Biològiques, Universitat de València; Burjassot 46100 València Spain
| | - Cecília Pardo-Bellver
- Unitat Mixta de Neuroanatomia Funcional UV-UJI - Dept. de Biologia Cel·lular i Biologia Funcional, Facultat de Ciències Biològiques, Universitat de València; Burjassot 46100 València Spain
| | - Fernando Martínez-García
- Unitat Mixta de Neuroanatomia Funcional UV-UJI - Unitat Predepartamental de Medicina, Fac. Ciències de la Salut, Universitat Jaume I; Castelló de la Plana Spain
| | - Enrique Lanuza
- Unitat Mixta de Neuroanatomia Funcional UV-UJI - Dept. de Biologia Cel·lular i Biologia Funcional, Facultat de Ciències Biològiques, Universitat de València; Burjassot 46100 València Spain
| |
Collapse
|
129
|
Garcia AF, Nakata KG, Ferguson SM. Viral strategies for targeting cortical circuits that control cocaine-taking and cocaine-seeking in rodents. Pharmacol Biochem Behav 2017; 174:33-41. [PMID: 28552825 DOI: 10.1016/j.pbb.2017.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/08/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022]
Abstract
Addiction to cocaine is a chronic disease characterized by persistent drug-taking and drug-seeking behaviors, and a high likelihood of relapse. The prefrontal cortex (PFC) has long been implicated in the development of cocaine addiction, and relapse. However, the PFC is a heterogeneous structure, and understanding the role of PFC subdivisions, cell types and afferent/efferent connections is critical for gaining a comprehensive picture of the contribution of the PFC in addiction-related behaviors. Here we provide an update on the role of the PFC in cocaine addiction from recent work that used viral-mediated optogenetic and chemogenetic tools to study the role of the PFC in drug-taking and drug-seeking behavior in rodents. Following overviews of rodent PFC neuroanatomy and of viral-mediated optogenetic and chemogenetic techniques, we review studies of manipulations within the PFC, followed by a review of work that utilized targeted manipulations to PFC inputs and outputs.
Collapse
Affiliation(s)
- Aaron F Garcia
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States; Neuroscience Graduate Program, University of Washington, Seattle, WA, United States
| | - Kanichi G Nakata
- Neuroscience Graduate Program, University of Washington, Seattle, WA, United States
| | - Susan M Ferguson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States; Neuroscience Graduate Program, University of Washington, Seattle, WA, United States; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States.
| |
Collapse
|
130
|
Chen W, Wang Y, Wang X, Li H. Neural circuits involved in the renewal of extinguished fear. IUBMB Life 2017; 69:470-478. [PMID: 28464461 DOI: 10.1002/iub.1636] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/14/2017] [Indexed: 12/22/2022]
Abstract
The last 10 years have witnessed a substantial progress in understanding the neural mechanisms for the renewal of the extinguished fear memory. Based on the theory of fear extinction, exposure therapy has been developed as a typical cognitive behavioral therapy for posttraumatic stress disorder. Although the fear memory can be extinguished by repeated presentation of conditioned stimulus without unconditioned stimulus, the fear memory is not erased and tends to relapse outside of extinction context, which is referred to as renewal. Therefore, the renewal is regarded as a great obstruction interfering with the effect of exposure therapy. In recent years, there has been a great deal of studies in understanding the neurobiological underpinnings of fear renewal. These offer a foundation upon which novel therapeutic interventions for the renewal may be built. This review focuses on behavioral, anatomical and electrophysiological studies that interpret roles of the hippocampus, prelimbic cortex and amygdala as well as the connections between them for the renewal of the extinguished fear. Additionally, this review suggests the possible pathways for the renewal: (1) the prelimbic cortex may integrate contextual information from hippocampal inputs and project to the basolateral amygdala to mediate the renewal of extinguished fear memory; the ventral hippocampus may innervate the activities of the basolateral amygdala or the central amygdala directly for the renewal. © 2017 IUBMB Life, 69(7):470-478, 2017.
Collapse
Affiliation(s)
- Weihai Chen
- Faculty of Psychology, Southwest University, Chongqing, China.,Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, China
| | - Yan Wang
- Faculty of Psychology, Southwest University, Chongqing, China.,Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, China
| | - Xiaqing Wang
- Faculty of Psychology, Southwest University, Chongqing, China.,Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, China
| | - Hong Li
- Faculty of Psychology, Southwest University, Chongqing, China.,Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, China
| |
Collapse
|
131
|
Vergara MD, Keller VN, Fuentealba JA, Gysling K. Activation of type 4 dopaminergic receptors in the prelimbic area of medial prefrontal cortex is necessary for the expression of innate fear behavior. Behav Brain Res 2017; 324:130-137. [PMID: 28212942 DOI: 10.1016/j.bbr.2017.01.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 11/25/2022]
Abstract
The prelimbic area (PL) of the medial Prefrontal cortex (mPFC) is involved in the acquisition and expression of conditioned and innate fear. Both types of fear share several neuronal pathways. It has been documented that dopamine (DA) plays an important role in the regulation of aversive memories in the mPFC. The exposure to an aversive stimulus, such as the smell of a predator odor or the exposure to footshock stress is accompanied by an increase in mPFC DA release. Evidence suggests that the type 4 dopaminergic receptor (D4R) is the molecular target through which DA modulates fear expression. In fact, the mPFC is the brain region with the highest expression of D4R; however, the role of D4R in the expression of innate fear has not been fully elucidated. Therefore, the principal objective of this work was to evaluate the participation of mPFC D4R in the expression of innate fear. Rats were exposed to the elevated plus-maze (EPM) and to the cat odor paradigm after the intra PL injection of L-745,870, selective D4R antagonist, to measure the expression of fear-related behaviors. Intra PL injection of L-745,870 increased the time spent in the EPM open arms and decreased freezing behavior in the cat odor paradigm. Our results also showed that D4R is expressed in GABAergic and pyramidal neurons in the PL region of PFC. Thus, D4R antagonism in the PL decreases the expression of innate fear-behavior indicating that the activation of D4R in the PL is necessary for the expression of innate fear-behavior.
Collapse
Affiliation(s)
- Macarena D Vergara
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Victor N Keller
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José A Fuentealba
- Department of Pharmacy, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Katia Gysling
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
132
|
Shapiro LP, Parsons RG, Koleske AJ, Gourley SL. Differential expression of cytoskeletal regulatory factors in the adolescent prefrontal cortex: Implications for cortical development. J Neurosci Res 2017; 95:1123-1143. [PMID: 27735056 PMCID: PMC5352542 DOI: 10.1002/jnr.23960] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/04/2016] [Accepted: 09/12/2016] [Indexed: 12/27/2022]
Abstract
The prevalence of depression, anxiety, schizophrenia, and drug and alcohol use disorders peaks during adolescence. Further, up to 50% of "adult" mental health disorders emerge in adolescence. During adolescence, the prefrontal cortex (PFC) undergoes dramatic structural reorganization, in which dendritic spines and synapses are refined, pruned, and stabilized. Understanding the molecular mechanisms that underlie these processes should help to identify factors that influence the development of psychiatric illness. Here we briefly discuss the anatomical connections of the medial and orbital prefrontal cortex (mPFC and OFC, respectively). We then present original findings suggesting that dendritic spines on deep-layer excitatory neurons in the mouse mPFC and OFC prune at different adolescent ages, with later pruning in the OFC. In parallel, we used Western blotting to define levels of several cytoskeletal regulatory proteins during early, mid-, and late adolescence, focusing on tropomyosin-related kinase receptor B (TrkB) and β1-integrin-containing receptors and select signaling partners. We identified regional differences in the levels of several proteins in early and midadolescence that then converged in early adulthood. We also observed age-related differences in TrkB levels, both full-length and truncated isoforms, Rho-kinase 2, and synaptophysin in both PFC subregions. Finally, we identified changes in protein levels in the dorsal and ventral hippocampus that were distinct from those in the PFC. We conclude with a general review of the manner in which TrkB- and β1-integrin-mediated signaling influences neuronal structure in the postnatal brain. Elucidating the role of cytoskeletal regulatory factors throughout adolescence may identify critical mechanisms of PFC development. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lauren P Shapiro
- Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia
- Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Ryan G Parsons
- Department of Psychology and Neuroscience Institute, Graduate Program in Integrative Neuroscience, Program in Neuroscience, Stony Brook University, Stony Brook, New York
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Department of Neurobiology, Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia
| |
Collapse
|
133
|
Reichard RA, Subramanian S, Desta MT, Sura T, Becker ML, Ghobadi CW, Parsley KP, Zahm DS. Abundant collateralization of temporal lobe projections to the accumbens, bed nucleus of stria terminalis, central amygdala and lateral septum. Brain Struct Funct 2017; 222:1971-1988. [PMID: 27704219 PMCID: PMC5378696 DOI: 10.1007/s00429-016-1321-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
Abstract
Behavioral flexibility is subserved in part by outputs from the cerebral cortex to telencephalic subcortical structures. In our earlier evaluation of the organization of the cortical-subcortical output system (Reynolds and Zahm, J Neurosci 25:11757-11767, 2005), retrograde double-labeling was evaluated in the prefrontal cortex following tracer injections into pairs of the following subcortical telencephalic structures: caudate-putamen, core and shell of the accumbens (Acb), bed nucleus of stria terminalis (BST) and central nucleus of the amygdala (CeA). The present study was done to assess patterns of retrograde labeling in the temporal lobe after similar paired tracer injections into most of the same telencephalic structures plus the lateral septum (LS). In contrast to the modest double-labeling observed in the prefrontal cortex in the previous study, up to 60-80 % of neurons in the basal and accessory basal amygdaloid nuclei and amygdalopiriform transition area exhibited double-labeling in the present study. The most abundant double-labeling was generated by paired injections into structures affiliated with the extended amygdala, including the CeA, BST and Acb shell. Injections pairing the Acb core with the BST or CeA produced significantly fewer double-labeled neurons. The ventral subiculum exhibited modest amounts of double-labeling associated with paired injections into the Acb, BST, CeA and LS. The results raise the issue of how an extraordinarily collateralized output from the temporal lobe may contribute to behavioral flexibility.
Collapse
Affiliation(s)
- Rhett A Reichard
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Suriya Subramanian
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Mikiyas T Desta
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Tej Sura
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Mary L Becker
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Comeron W Ghobadi
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Kenneth P Parsley
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Daniel S Zahm
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA.
| |
Collapse
|
134
|
Chang CH. Lateral Orbitofrontal Cortical Modulation on the Medial Prefrontal Cortex-Amygdala Pathway: Differential Regulation of Intra-Amygdala GABAA and GABAB Receptors. Int J Neuropsychopharmacol 2017; 20:602-610. [PMID: 28444246 PMCID: PMC5492808 DOI: 10.1093/ijnp/pyx027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The basolateral complex of the amygdala receives inputs from neocortical areas, including the medial prefrontal cortex and lateral orbitofrontal cortex. Earlier studies have shown that lateral orbitofrontal cortex activation exerts an inhibitory gating on medial prefrontal cortex-amygdala information flow. Here we examined the individual role of GABAA and GABAB receptors in this process. METHODS In vivo extracellular single-unit recordings were done in anesthetized rats. We searched amygdala neurons that fire in response to medial prefrontal cortex activation, tested lateral orbitofrontal cortex gating at different delays (lateral orbitofrontal cortex-medial prefrontal cortex delays: 25, 50, 100, 250, 500, and 1000 milliseconds), and examined differential contribution of GABAA and GABAB receptors with iontophoresis. RESULTS Relative to baseline, lateral orbitofrontal cortex stimulation exerted an inhibitory modulatory gating on the medial prefrontal cortex-amygdala pathway and was effective up to a long delay of 500 ms (long-delay latencies at 100, 250, and 500 milliseconds). Moreover, blockade of intra-amygdala GABAA receptors with bicuculline abolished the lateral orbitofrontal cortex inhibitory gating at both short- (25 milliseconds) and long-delay (100 milliseconds) intervals, while blockade of GABAB receptors with saclofen reversed the inhibitory gating at long delay (100 milliseconds) only. Among the majority of the neurons examined (8 of 9), inactivation of either GABAA or GABAB receptors during baseline did not change evoked probability per se, suggesting that local feed-forward inhibitory mechanism is pathway specific. CONCLUSIONS Our results suggest that the effect of lateral orbitofrontal cortex inhibitory modulatory gating was effective up to 500 milliseconds and that intra-amygdala GABAA and GABAB receptors differentially modulate the short- and long-delay lateral orbitofrontal cortex inhibitory gating on the medial prefrontal cortex-amygdala pathway.
Collapse
Affiliation(s)
- Chun-hui Chang
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
135
|
Endogenous opioids regulate moment-to-moment neuronal communication and excitability. Nat Commun 2017; 8:14611. [PMID: 28327612 PMCID: PMC5364458 DOI: 10.1038/ncomms14611] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 01/14/2017] [Indexed: 01/12/2023] Open
Abstract
Fear and emotional learning are modulated by endogenous opioids but the cellular basis for this is unknown. The intercalated cells (ITCs) gate amygdala output and thus regulate the fear response. Here we find endogenous opioids are released by synaptic stimulation to act via two distinct mechanisms within the main ITC cluster. Endogenously released opioids inhibit glutamate release through the δ-opioid receptor (DOR), an effect potentiated by a DOR-positive allosteric modulator. Postsynaptically, the opioids activate a potassium conductance through the μ-opioid receptor (MOR), suggesting for the first time that endogenously released opioids directly regulate neuronal excitability. Ultrastructural localization of endogenous ligands support these functional findings. This study demonstrates a new role for endogenously released opioids as neuromodulators engaged by synaptic activity to regulate moment-to-moment neuronal communication and excitability. These distinct actions through MOR and DOR may underlie the opposing effect of these receptor systems on anxiety and fear. The endogenous opioid system regulates fear and anxiety, but the underlying cellular mechanism is unclear. Winters et al. shows that in the intercalated cells (ITC) of the amygdala, endogenous opioids suppress glutamatergic inputs via the δ-opioid receptor presynaptically, and reduce the excitability of ITCs via the μ-opioid receptor postsynaptically.
Collapse
|
136
|
Kerestes R, Chase HW, Phillips ML, Ladouceur CD, Eickhoff SB. Multimodal evaluation of the amygdala's functional connectivity. Neuroimage 2017; 148:219-229. [PMID: 28089676 PMCID: PMC5416470 DOI: 10.1016/j.neuroimage.2016.12.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 12/26/2022] Open
Abstract
The amygdala is one of the most extensively studied human brain regions and undisputedly plays a central role in many psychiatric disorders. However, an outstanding question is whether connectivity of amygdala subregions, specifically the centromedial (CM), laterobasal (LB) and superficial (SF) nuclei, are modulated by brain state (i.e., task vs. rest). Here, using a multimodal approach, we directly compared meta-analytic connectivity modeling (MACM) and specific co-activation likelihood estimation (SCALE)-derived estimates of CM, LB and SF task-based co-activation to the functional connectivity of these nuclei as assessed by resting state fmri (rs-fmri). Finally, using a preexisting resting state functional connectivity-derived cortical parcellation, we examined both MACM and rs-fmri amygdala subregion connectivity with 17 large-scale networks, to explicitly address how the amygdala interacts with other large-scale neural networks. Analyses revealed strong differentiation of CM, LB and SF connectivity patterns with other brain regions, both in task-dependent and task-independent contexts. All three regions, however, showed convergent connectivity with the right ventrolateral prefrontal cortex (VLPFC) that was not driven by high base rate levels of activation. Similar patterns of connectivity across rs-fmri and MACM were observed for each subregion, suggesting a similar network architecture of amygdala connectivity with the rest of the brain across tasks and resting state for each subregion, that may be modified in the context of specific task demands. These findings support animal models that posit a parallel model of amygdala functioning, but importantly, also modify this position to suggest integrative processing in the amygdala.
Collapse
Affiliation(s)
- Rebecca Kerestes
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Henry W Chase
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mary L Phillips
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cecile D Ladouceur
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Germany; Institute of Clinical Neuroscience and Medical Psychology, Heinrich-Heine University Düsseldorf, Germany; Institute of Systems Neuroscience, School of Medicine, Heinrich-Heine University Düsseldorf, Germany
| |
Collapse
|
137
|
Zimmermann KS, Yamin JA, Rainnie DG, Ressler KJ, Gourley SL. Connections of the Mouse Orbitofrontal Cortex and Regulation of Goal-Directed Action Selection by Brain-Derived Neurotrophic Factor. Biol Psychiatry 2017; 81:366-377. [PMID: 26786312 PMCID: PMC4871791 DOI: 10.1016/j.biopsych.2015.10.026] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 10/27/2015] [Accepted: 10/27/2015] [Indexed: 01/30/2023]
Abstract
BACKGROUND Distinguishing between actions that are more likely or less likely to be rewarded is a critical aspect of goal-directed decision making. However, neuroanatomic and molecular mechanisms are not fully understood. METHODS We used anterograde tracing, viral-mediated gene silencing, functional disconnection strategies, pharmacologic rescue, and designer receptors exclusively activated by designer drugs (DREADDs) to determine the anatomic and functional connectivity between the orbitofrontal cortex (OFC) and the amygdala in mice. In particular, we knocked down brain-derived neurotrophic factor (Bdnf) bilaterally in the OFC or generated an OFC-amygdala "disconnection" by pairing unilateral OFC Bdnf knockdown with lesions of the contralateral amygdala. We characterized decision-making strategies using a task in which mice selected actions based on the likelihood that they would be reinforced. Additionally, we assessed the effects of DREADD-mediated OFC inhibition on the consolidation of action-outcome conditioning. RESULTS As in other species, the OFC projects to the basolateral amygdala and dorsal striatum in mice. Bilateral Bdnf knockdown within the ventrolateral OFC and unilateral Bdnf knockdown accompanied by lesions of the contralateral amygdala impede goal-directed response selection, implicating BDNF-expressing OFC projection neurons in selecting actions based on their consequences. The tyrosine receptor kinase B agonist 7,8-dihydroxyflavone rescues action selection and increases dendritic spine density on excitatory neurons in the OFC. Rho-kinase inhibition also rescues goal-directed response strategies, linking neural remodeling with outcome-based decision making. Finally, DREADD-mediated OFC inhibition weakens new action-outcome memory. CONCLUSIONS Activity-dependent and BDNF-dependent neuroplasticity within the OFC coordinate outcome-based decision making through interactions with the amygdala. These interactions break reward-seeking habits, a putative factor in multiple psychopathologies.
Collapse
Affiliation(s)
- Kelsey S. Zimmermann
- Department of Pediatrics, Emory University, Atlanta, GA USA,Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA,Yerkes National Primate Research Center, Emory University, Atlanta, GA USA,Graduate Program in Neuroscience, Emory University, Atlanta, GA USA
| | - John A. Yamin
- Department of Pediatrics, Emory University, Atlanta, GA USA,Yerkes National Primate Research Center, Emory University, Atlanta, GA USA
| | - Donald G. Rainnie
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA,Yerkes National Primate Research Center, Emory University, Atlanta, GA USA,Graduate Program in Neuroscience, Emory University, Atlanta, GA USA
| | - Kerry J. Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA,Yerkes National Primate Research Center, Emory University, Atlanta, GA USA,Graduate Program in Neuroscience, Emory University, Atlanta, GA USA,Howard Hughes Medical Institute, Bethesda, MD, USA
| | - Shannon L. Gourley
- Department of Pediatrics, Emory University, Atlanta, GA USA,Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA,Yerkes National Primate Research Center, Emory University, Atlanta, GA USA,Graduate Program in Neuroscience, Emory University, Atlanta, GA USA
| |
Collapse
|
138
|
Optogenetic Examination of Prefrontal-Amygdala Synaptic Development. J Neurosci 2017; 37:2976-2985. [PMID: 28193691 DOI: 10.1523/jneurosci.3097-16.2017] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/18/2017] [Accepted: 02/05/2017] [Indexed: 11/21/2022] Open
Abstract
A brain network comprising the medial prefrontal cortex (mPFC) and amygdala plays important roles in developmentally regulated cognitive and emotional processes. However, very little is known about the maturation of mPFC-amygdala circuitry. We conducted anatomical tracing of mPFC projections and optogenetic interrogation of their synaptic connections with neurons in the basolateral amygdala (BLA) at neonatal to adult developmental stages in mice. Results indicate that mPFC-BLA projections exhibit delayed emergence relative to other mPFC pathways and establish synaptic transmission with BLA excitatory and inhibitory neurons in late infancy, events that coincide with a massive increase in overall synaptic drive. During subsequent adolescence, mPFC-BLA circuits are further modified by excitatory synaptic strengthening as well as a transient surge in feedforward inhibition. The latter was correlated with increased spontaneous inhibitory currents in excitatory neurons, suggesting that mPFC-BLA circuit maturation culminates in a period of exuberant GABAergic transmission. These findings establish a time course for the onset and refinement of mPFC-BLA transmission and point to potential sensitive periods in the development of this critical network.SIGNIFICANCE STATEMENT Human mPFC-amygdala functional connectivity is developmentally regulated and figures prominently in numerous psychiatric disorders with a high incidence of adolescent onset. However, it remains unclear when synaptic connections between these structures emerge or how their properties change with age. Our work establishes developmental windows and cellular substrates for synapse maturation in this pathway involving both excitatory and inhibitory circuits. The engagement of these substrates by early life experience may support the ontogeny of fundamental behaviors but could also lead to inappropriate circuit refinement and psychopathology in adverse situations.
Collapse
|
139
|
Morphological, structural, and functional alterations of the prefrontal cortex and the basolateral amygdala after early lesion of the rat mediodorsal thalamus. Brain Struct Funct 2017; 222:2527-2545. [DOI: 10.1007/s00429-016-1354-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 12/16/2016] [Indexed: 01/06/2023]
|
140
|
Al Aïn S, Perry RE, Nuñez B, Kayser K, Hochman C, Brehman E, LaComb M, Wilson DA, Sullivan RM. Neurobehavioral assessment of maternal odor in developing rat pups: implications for social buffering. Soc Neurosci 2017; 12:32-49. [PMID: 26934130 PMCID: PMC5033694 DOI: 10.1080/17470919.2016.1159605] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Social support can attenuate the behavioral and stress hormone response to threat, a phenomenon called social buffering. The mother's social buffering of the infant is one of the more robust examples; yet we understand little about the neurobiology. Using a rodent model, we explore the neurobiology of social buffering by assessing neural processing of the maternal odor, a major cue controlling social buffering in rat pups. We used pups before (postnatal day (PN) 7) and after (PN14, PN23) the functional emergence of social buffering. Pups were injected with 14C 2-deoxyglucose (2-DG) and presented with the maternal odor, a control preferred odor incapable of social buffering (acetophenone), or no odor. Brains were removed, processed for autoradiography and brain areas identified as important in adult social buffering were assessed, including the amygdala basolateral complex (Basolateral Amygdala [BLA]), medial prefrontal cortex (mPFC), and anterior cingulate cortex (ACC). Results suggest dramatic changes in the processing of maternal odor. PN7 pups show mPFC and ACC activation, although PN14 pups showed no activation of the mPFC, ACC, or BLA. All brain areas assessed were recruited by PN23. Additional analysis suggests substantial changes in functional connectivity across development. Together, these results imply complex nonlinear transitions in the neurobiology of social buffering in early life that may provide insight into the changing role of the mother in supporting social buffering.
Collapse
Affiliation(s)
- Syrina Al Aïn
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Rosemarie E. Perry
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
- Neuroscience and Physiology, NYU Sackler Institute, New York University School of Medicine, New York, NY, USA
| | - Bestina Nuñez
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
| | - Kassandra Kayser
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
| | - Chase Hochman
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Elizabeth Brehman
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Miranda LaComb
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Donald A. Wilson
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York, USA
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
141
|
Buck KJ, Chen G, Kozell LB. Limbic circuitry activation in ethanol withdrawal is regulated by a chromosome 1 locus. Alcohol 2017; 58:153-160. [PMID: 27989609 DOI: 10.1016/j.alcohol.2016.09.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/03/2016] [Accepted: 09/03/2016] [Indexed: 11/19/2022]
Abstract
Physiological dependence and associated withdrawal episodes are thought to constitute a motivational force sustaining alcohol use/abuse and contributing to relapse in alcoholics. Although no animal model exactly duplicates alcoholism, models for specific factors, including the withdrawal syndrome, are useful for identifying potential genetic and neural determinants of liability in humans. We previously identified highly significant quantitative trait loci (QTLs) with large effects on predisposition to withdrawal after chronic and acute alcohol exposure in mice and mapped these loci to the same region of chromosome 1 (Alcdp1 and Alcw1, respectively). The present studies utilize a novel Alcdp1/Alcw1 congenic model (in which an interval spanning Alcdp1 and Alcw1 from the C57BL/6J donor strain [build GRCm38 150.3-174.6 Mb] has been introgressed onto a uniform inbred DBA/2J genetic background) known to demonstrate significantly less severe chronic and acute withdrawal compared to appropriate background strain animals. Here, using c-Fos induction as a high-resolution marker of neuronal activation, we report that male Alcdp1/Alcw1 congenic animals demonstrate significantly less alcohol withdrawal-associated neural activation compared to appropriate background strain animals in the prelimbic and cingulate cortices of the prefrontal cortex as well as discrete regions of the extended amygdala (i.e., basolateral) and extended basal ganglia (i.e., dorsolateral striatum, and caudal substantia nigra pars reticulata). These studies are the first to begin to elucidate circuitry by which this confirmed addiction-relevant QTL could influence behavior. This circuitry overlaps limbic circuitry involved in stress, providing additional mechanistic information. Alcdp1/Alcw1 maps to a region syntenic with human chromosome 1q, where multiple studies find significant associations with risk for alcoholism.
Collapse
Affiliation(s)
- Kari J Buck
- Department of Behavioral Neuroscience, Portland Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239, USA.
| | - Gang Chen
- Department of Behavioral Neuroscience, Portland Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Laura B Kozell
- Department of Behavioral Neuroscience, Portland Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
142
|
Hummer TA, Phan KL, Kern DW, McClintock MK. A human chemosignal modulates frontolimbic activity and connectivity in response to emotional stimuli. Psychoneuroendocrinology 2017; 75:15-25. [PMID: 27768980 DOI: 10.1016/j.psyneuen.2016.09.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 11/29/2022]
Abstract
Evidence suggests the putative human pheromone Δ4,16-androstadien-3-one (androstadienone), a natural component of human sweat, increases attention to emotional information when passively inhaled, even in minute amounts. However, the neural mechanisms underlying androstadienone's impact on the perception of emotional stimuli have not been clarified. To characterize how the compound modifies neural circuitry while attending to emotional information, 22 subjects (11 women) underwent two fMRI scanning sessions, one with an androstadienone solution and one with a carrier control solution alone on their upper lip. During each session, participants viewed blocks of emotionally positive, negative, or neutral images. The BOLD response to emotional images (relative to neutral images) was greater during exposure to androstadienone in right orbitofrontal and lateral prefrontal cortex, particularly during positive image blocks. Androstadienone did not impact the response to social images, compared to nonsocial images, and results were not related to participant sex or olfactory sensitivity. To examine how androstadienone influences effective connectivity of this network, a dynamic causal model was employed with primary visual cortex (V1), amygdala, prefrontal cortex, and orbitofrontal cortex on each side. These models indicated that emotional images increased the drive from V1 to the amygdala during the control session. With androstadienone present, this drive to amygdala was decreased specifically for positive images, which drove downstream increases in orbitofrontal and prefrontal activity. This evidence suggests that androstadienone may act as a chemical signal to increase attention to positively valenced information via modifications to amygdala connectivity.
Collapse
Affiliation(s)
- Tom A Hummer
- Department of Psychiatry, Indiana University School of Medicine, United States.
| | - K Luan Phan
- Department of Psychiatry, University of Illinois at Chicago and Mental Health Service Line, Jesse Brown VA, Chicago, IL, United States
| | - David W Kern
- Institute for Mind and Biology and Department of Comparative Human Development, The University of Chicago, United States
| | - Martha K McClintock
- Institute for Mind and Biology and Department of Comparative Human Development, The University of Chicago, United States; Department of Psychology, The University of Chicago, United States
| |
Collapse
|
143
|
Activation and blockade of prelimbic 5-HT6 receptors produce different effects on depressive-like behaviors in unilateral 6-hydroxydopamine-induced Parkinson's rats. Neuropharmacology 2016; 110:25-36. [DOI: 10.1016/j.neuropharm.2016.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/10/2016] [Accepted: 07/12/2016] [Indexed: 11/15/2022]
|
144
|
Cao B, Wang J, Shahed M, Jelfs B, Chan RHM, Li Y. Vagus Nerve Stimulation Alters Phase Synchrony of the Anterior Cingulate Cortex and Facilitates Decision Making in Rats. Sci Rep 2016; 6:35135. [PMID: 27731403 PMCID: PMC5059720 DOI: 10.1038/srep35135] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 09/26/2016] [Indexed: 11/09/2022] Open
Abstract
Vagus nerve stimulation (VNS) can enhance memory and cognitive functions in both rats and humans. Studies have shown that VNS influenced decision-making in epileptic patients. However, the sites of action involved in the cognitive-enhancement are poorly understood. By employing a conscious rat model equipped with vagus nerve cuff electrode, we assess the role of chronic VNS on decision-making in rat gambling task (RGT). Simultaneous multichannel-recordings offer an ideal setup to test the hypothesis that VNS may induce alterations of in both spike-field-coherence and synchronization of theta oscillations across brain areas in the anterior cingulate cortex (ACC) and basolateral amygdala (BLA). Daily VNS, administered immediately following training sessions of RGT, caused an increase in 'good decision-maker' rats. Neural spikes in the ACC became synchronized with the ongoing theta oscillations of local field potential (LFP) in BLA following VNS. Moreover, cross-correlation analysis revealed synchronization between the ACC and BLA. Our results provide specific evidence that VNS facilitates decision-making and unveils several important roles for VNS in regulating LFP and spike phases, as well as enhancing spike-phase coherence between key brain areas involved in cognitive performance. These data may serve to provide fundamental notions regarding neurophysiological biomarkers for therapeutic VNS in cognitive impairment.
Collapse
Affiliation(s)
- Bing Cao
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
| | - Jun Wang
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
| | - Mahadi Shahed
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
| | - Beth Jelfs
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
- Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Rosa H. M. Chan
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
- Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Ying Li
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
- Shenzhen Key Lab of Neuropsychiatric Modulation, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Hong Kong
- School of Veterinary Medicine, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
145
|
Paine TA, Swedlow N, Swetschinski L. Decreasing GABA function within the medial prefrontal cortex or basolateral amygdala decreases sociability. Behav Brain Res 2016; 317:542-552. [PMID: 27732892 DOI: 10.1016/j.bbr.2016.10.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/28/2016] [Accepted: 10/07/2016] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Decreased sociability is a symptom of psychiatric conditions including autism-spectrum disorder and schizophrenia. Both of these conditions are associated with decreases in GABA function, particularly in the medial prefrontal cortex (PFC) and the basolateral amygdala (BLA); structures that are components of the social brain. Here, we determined if decreasing GABA transmission within either the PFC or the BLA decreases social behavior. METHODS Rats were implanted with cannulae aimed at either the medial PFC or the BLA and then were tested on up to 4 behavioral tests following bilateral infusions of 0.5μl bicuculline methiodide (BMI, a GABAA receptor antagonist) at doses of 0, 25, or 50ng/μl. Rats were tested in the social interaction test, the social preference test, the sucrose preference test and for locomotor activity (BLA infusions only). RESULTS Intra-BLA or PFC BMI infusions decreased the amount of time and the number of social interactions in the social interaction test. Further, in the social preference test, rats infused with 50ng BMI no longer exhibited a preference to explore a social over a non-social stimulus. The change in sociability was not due to a change in reward processing or locomotor behavior. DISCUSSION Decreasing GABA transmission in either the medial PFC or BLA decreased sociability. Thus, changes in GABA signaling observed in conditions such as autism or schizophrenia may mediate the social withdrawal characteristic of these conditions. Moreover, they suggest that social withdrawal may be treated by drugs that potentiate GABA transmission.
Collapse
Affiliation(s)
- Tracie A Paine
- Department of Neuroscience, Oberlin College, Oberlin, OH, 44074, United States.
| | - Nathan Swedlow
- Department of Neuroscience, Oberlin College, Oberlin, OH, 44074, United States
| | - Lucien Swetschinski
- Department of Neuroscience, Oberlin College, Oberlin, OH, 44074, United States
| |
Collapse
|
146
|
Cervera-Ferri A, Teruel-Martí V, Barceló-Molina M, Martínez-Ricós J, Luque-García A, Martínez-Bellver S, Adell A. Characterization of oscillatory changes in hippocampus and amygdala after deep brain stimulation of the infralimbic prefrontal cortex. Physiol Rep 2016; 4:4/14/e12854. [PMID: 27449812 PMCID: PMC4962070 DOI: 10.14814/phy2.12854] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 06/16/2016] [Indexed: 01/14/2023] Open
Abstract
Deep brain stimulation (DBS) is a new investigational therapy that has generated positive results in refractory depression. Although the neurochemical and behavioral effects of DBS have been examined, less attention has been paid to the influence of DBS on the network dynamics between different brain areas, which could contribute to its therapeutic effects. Herein, we set out to identify the effects of 1 h DBS in the infralimbic cortex (IL) on the oscillatory network dynamics between hippocampus and basolateral amygdala (BLA), two regions implicated in depression and its treatment. Urethane-anesthetized rats with bilaterally implanted electrodes in the IL were exposed to 1 h constant stimulation of 130 Hz of frequency, 60 μA of constant current intensity and biphasic pulse width of 80 μsec. After a period of baseline recording, local field potentials (LFP) were recorded with formvar-insulated stainless steel electrodes. DBS of the IL increased the power of slow wave (SW, <1.5 Hz) and theta (3-12 Hz) frequencies in the hippocampus and BLA Furthermore, IL DBS caused a precise coupling in different frequency bands between both brain structures. The increases in SW band synchronization in hippocampus and BLA after DBS suggest that these changes may be important for the improvement of depressive behavior. In addition, the augmentation in theta synchrony might contribute to improvement in emotional and cognitive processes.
Collapse
Affiliation(s)
- Ana Cervera-Ferri
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embriology, Faculty of Medicine and Odontology University of Valencia, Valencia, 46010, Spain
| | - Vicent Teruel-Martí
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embriology, Faculty of Medicine and Odontology University of Valencia, Valencia, 46010, Spain
| | - Moises Barceló-Molina
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embriology, Faculty of Medicine and Odontology University of Valencia, Valencia, 46010, Spain Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain
| | - Joana Martínez-Ricós
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embriology, Faculty of Medicine and Odontology University of Valencia, Valencia, 46010, Spain
| | - Aina Luque-García
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embriology, Faculty of Medicine and Odontology University of Valencia, Valencia, 46010, Spain Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain
| | - Sergio Martínez-Bellver
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embriology, Faculty of Medicine and Odontology University of Valencia, Valencia, 46010, Spain Department of Cell Biology and Parasitology, Faculty of Medicine and Odontology University of Valencia, Valencia, 46010, Spain
| | - Albert Adell
- Institute of Biomedicine and Biotechnology of Cantabria, IBBTEC (CSIC University of Cantabria), Santander, 39011, Spain
| |
Collapse
|
147
|
The Medial Orbitofrontal Cortex Regulates Sensitivity to Outcome Value. J Neurosci 2016; 36:4600-13. [PMID: 27098701 DOI: 10.1523/jneurosci.4253-15.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/08/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED An essential component of goal-directed decision-making is the ability to maintain flexible responding based on the value of a given reward, or "reinforcer." The medial orbitofrontal cortex (mOFC), a subregion of the ventromedial prefrontal cortex, is uniquely positioned to regulate this process. We trained mice to nose poke for food reinforcers and then stimulated this region using CaMKII-driven Gs-coupled designer receptors exclusively activated by designer drugs (DREADDs). In other mice, we silenced the neuroplasticity-associated neurotrophin brain-derived neurotrophic factor (BDNF). Activation of Gs-DREADDs increased behavioral sensitivity to reinforcer devaluation, whereas Bdnf knockdown blocked sensitivity. These changes were accompanied by modifications in breakpoint ratios in a progressive ratio task, and they were recapitulated in Bdnf(+/-)mice. Replacement of BDNF selectively in the mOFC in Bdnf(+/-)mice rescued behavioral deficiencies, as well as phosphorylation of extracellular-signal regulated kinase 1/2 (ERK1/2). Thus, BDNF expression in the mOFC is both necessary and sufficient for the expression of typical effort allocation relative to an anticipated reinforcer. Additional experiments indicated that expression of the immediate-early gene c-fos was aberrantly elevated in the Bdnf(+/-)dorsal striatum, and BDNF replacement in the mOFC normalized expression. Also, systemic administration of an MAP kinase kinase inhibitor increased breakpoint ratios, whereas the addition of discrete cues bridging the response-outcome contingency rescued breakpoints in Bdnf(+/-)mice. We argue that BDNF-ERK1/2 in the mOFC is a key regulator of "online" goal-directed action selection. SIGNIFICANCE STATEMENT Goal-directed response selection often involves predicting the consequences of one's actions and the value of potential payoffs. Lesions or chemogenetic inactivation of the medial orbitofrontal cortex (mOFC) in rats induces failures in retrieving outcome identity memories (Bradfield et al., 2015), suggesting that the healthy mOFC serves to access outcome value information when it is not immediately observable and thereby guide goal-directed decision-making. Our findings suggest that the mOFC also bidirectionally regulates effort allocation for a given reward and that expression of the neurotrophin BDNF in the mOFC is both necessary and sufficient for mice to sustain stable representations of reinforcer value.
Collapse
|
148
|
Careaga MBL, Girardi CEN, Suchecki D. Understanding posttraumatic stress disorder through fear conditioning, extinction and reconsolidation. Neurosci Biobehav Rev 2016; 71:48-57. [PMID: 27590828 DOI: 10.1016/j.neubiorev.2016.08.023] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 07/20/2016] [Accepted: 08/16/2016] [Indexed: 11/25/2022]
Abstract
Careaga MBL, Girardi CEN, Suchecki D. Understanding posttraumatic stress disorder through fear conditioning, extinction and reconsolidation. NEUROSCI BIOBEHAV REV -Posttraumatic stress disorder (PTSD) is a psychopathology characterized by exacerbation of fear response. A dysregulated fear response may be explained by dysfunctional learning and memory, a hypothesis that was proposed decades ago. A key component of PTSD is fear conditioning and the study of this phenomenon in laboratory has expanded the understanding of the underlying neurobiological changes in PTSD. Furthermore, traumatic memories are strongly present even years after the trauma and maintenance of this memory is usually related to behavioral and physiological maladaptive responses. Persistence of traumatic memory may be explained by a dysregulation of two memory processes: extinction and reconsolidation. The former may explain the over-expression of fear responses as an imbalance between traumatic and extinction memory. The latter, in turn, explains the maintenance of fear responses as a result of enhancing trauma-related memories. Thus, this review will discuss the importance of fear conditioning for the establishment of PTSD and how failure in extinction or abnormal reconsolidation may contribute to the maintenance of fear response overtime.
Collapse
Affiliation(s)
| | - Carlos Eduardo Neves Girardi
- Departamento de Psicobiologia - Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deborah Suchecki
- Departamento de Psicobiologia - Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.
| |
Collapse
|
149
|
Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry 2016; 3:760-773. [PMID: 27475769 PMCID: PMC6135092 DOI: 10.1016/s2215-0366(16)00104-8] [Citation(s) in RCA: 2058] [Impact Index Per Article: 228.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/09/2016] [Accepted: 03/11/2016] [Indexed: 12/17/2022]
Abstract
Drug addiction represents a dramatic dysregulation of motivational circuits that is caused by a combination of exaggerated incentive salience and habit formation, reward deficits and stress surfeits, and compromised executive function in three stages. The rewarding effects of drugs of abuse, development of incentive salience, and development of drug-seeking habits in the binge/intoxication stage involve changes in dopamine and opioid peptides in the basal ganglia. The increases in negative emotional states and dysphoric and stress-like responses in the withdrawal/negative affect stage involve decreases in the function of the dopamine component of the reward system and recruitment of brain stress neurotransmitters, such as corticotropin-releasing factor and dynorphin, in the neurocircuitry of the extended amygdala. The craving and deficits in executive function in the so-called preoccupation/anticipation stage involve the dysregulation of key afferent projections from the prefrontal cortex and insula, including glutamate, to the basal ganglia and extended amygdala. Molecular genetic studies have identified transduction and transcription factors that act in neurocircuitry associated with the development and maintenance of addiction that might mediate initial vulnerability, maintenance, and relapse associated with addiction.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA.
| | - Nora D Volkow
- National Institute on Drug Abuse, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
150
|
Do Monte FH, Quirk GJ, Li B, Penzo MA. Retrieving fear memories, as time goes by…. Mol Psychiatry 2016; 21:1027-36. [PMID: 27217148 PMCID: PMC4956525 DOI: 10.1038/mp.2016.78] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/22/2016] [Accepted: 04/05/2016] [Indexed: 12/12/2022]
Abstract
Research in fear conditioning has provided a comprehensive picture of the neuronal circuit underlying the formation of fear memories. In contrast, our understanding of the retrieval of fear memories is much more limited. This disparity may stem from the fact that fear memories are not rigid, but reorganize over time. To bring some clarity and raise awareness about the time-dependent dynamics of retrieval circuits, we review current evidence on the neuronal circuitry participating in fear memory retrieval at both early and late time points following auditory fear conditioning. We focus on the temporal recruitment of the paraventricular nucleus of the thalamus (PVT) for the retrieval and maintenance of fear memories. Finally, we speculate as to why retrieval circuits change with time, and consider the functional strategy of recruiting structures not previously considered as part of the retrieval circuit.
Collapse
Affiliation(s)
- Fabricio H. Do Monte
- Departments of Psychiatry and Anatomy & Neurobiology, University of Puerto Rico School of Medicine, PO Box 365067, San Juan 00936, Puerto Rico
| | - Gregory J. Quirk
- Departments of Psychiatry and Anatomy & Neurobiology, University of Puerto Rico School of Medicine, PO Box 365067, San Juan 00936, Puerto Rico
| | - Bo Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Mario A. Penzo
- National Institute of Mental Health, 35 Convent Drive, Bldg. 35A Room 2E621, Bethesda, MD 20850
| |
Collapse
|