201
|
Zeng Y, Hu D, Yang W, Hayashinaka E, Wada Y, Watanabe Y, Zeng Q, Cui Y. A voxel-based analysis of neurobiological mechanisms in placebo analgesia in rats. Neuroimage 2018; 178:602-612. [DOI: 10.1016/j.neuroimage.2018.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/22/2018] [Accepted: 06/04/2018] [Indexed: 12/19/2022] Open
|
202
|
Dik A, Saffari R, Zhang M, Zhang W. Contradictory effects of erythropoietin on inhibitory synaptic transmission in left and right prelimbic cortex of mice. Neurobiol Stress 2018; 9:113-123. [PMID: 30450377 PMCID: PMC6234276 DOI: 10.1016/j.ynstr.2018.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/08/2018] [Accepted: 08/24/2018] [Indexed: 12/28/2022] Open
Abstract
Erythropoietin (EPO) has been shown to improve cognitive function in mammals as well as in patients of psychiatric diseases by directly acting on the brain. In addition, EPO attenuates the synaptic transmission and enhances short- and long-term synaptic plasticity in hippocampus of mice, although there are still many discrepancies between different studies. It has been suggested that the divergences of different studies take root in different in-vivo application schemata or in long-term trophic effects of EPO. In the current study, we investigated the direct effects of EPO in slices of prelimbic cortex (PrL) by acute ex-vivo application of EPO, so that the erythropoietic or other trophic effects could be entirely excluded. Our results showed that the EPO effects were contradictory between the left and the right PrL. It enhanced the inhibitory transmission in the left and depressed the inhibitory transmission in the right PrL. Strikingly, this lateralized effect of EPO could be consistently found in individual bi-lateral PrL of all tested mice. Thus, our data suggest that EPO differentially modulates the inhibitory synaptic transmission of neuronal networks in the left and the right PrL. We hypothesize that such lateralized effects of EPO contribute to the development of the lateralization of stress reaction in PFC and underlie the altered bilateral GAGAergic synaptic transmission and oscillation patterns under stress that impact the central emotional and cognitive control in physiology as well as in pathophysiology. EPO showed fast effects on inhibitory transmission in the prefrontal cortex of mice. EPO enhanced the inhibitory transmission in the left and depressed it in the right prelimbic cortex of mice. The expression of EPOR in GAD+-neurons is different between the left and right PFC.
Collapse
Affiliation(s)
- Andre Dik
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Münster, Germany.,Department of Neurology, University of Muenster, Germany
| | - Roja Saffari
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Münster, Germany
| | - Mingyue Zhang
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Münster, Germany
| | - Weiqi Zhang
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Münster, Germany
| |
Collapse
|
203
|
Chaves-Coira I, Martín-Cortecero J, Nuñez A, Rodrigo-Angulo ML. Basal Forebrain Nuclei Display Distinct Projecting Pathways and Functional Circuits to Sensory Primary and Prefrontal Cortices in the Rat. Front Neuroanat 2018; 12:69. [PMID: 30158859 PMCID: PMC6104178 DOI: 10.3389/fnana.2018.00069] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/27/2018] [Indexed: 12/25/2022] Open
Abstract
Recent evidence supports that specific projections between different basal forebrain (BF) nuclei and their cortical targets are necessary to modulate cognitive functions in the cortex. We tested the hypothesis of the existence of specific neuronal populations in the BF linking with specific sensory, motor, and prefrontal cortices in rats. Neuronal tracing techniques were performed using retrograde tracers injected in the primary somatosensory (S1), auditory (A1), and visual (V1) cortical areas, in the medial prefrontal cortex (mPFC) as well as in BF nuclei. Results indicate that the vertical and horizontal diagonal band of Broca (VDB/HDB) nuclei target specific sensory cortical areas and maintains reciprocal projections with the prelimbic/infralimbic (PL/IL) area of the mPFC. The basal magnocellular nucleus (B nucleus) has more widespread targets in the sensory-motor cortex and does not project to the PL/IL cortex. Optogenetic stimulation was used to establish if BF neurons modulate whisker responses recorded in S1 and PL/IL cortices. We drove the expression of high levels of channelrhodopsin-2, tagged with a fluorescent protein (ChR2-eYFP) by injection of a virus in HDB or B nuclei. Blue-light pulses were delivered to the BF through a thin optic fiber to stimulate these neurons. Blue-light stimulation directed toward the HDB facilitated whisker responses in S1 cortex through activation of muscarinic receptors. The same optogenetic stimulation of HDB induced an inhibition of whisker responses in mPFC by activation of nicotinic receptors. Blue-light stimulation directed toward the B nucleus had lower effects than HDB stimulation. Our findings pointed the presence of specific neuronal networks between the BF and the cortex that may play different roles in the control of cortical activity.
Collapse
Affiliation(s)
- Irene Chaves-Coira
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Martín-Cortecero
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Angel Nuñez
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Margarita L Rodrigo-Angulo
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
204
|
Shikanai H, Oshima N, Kawashima H, Kimura SI, Hiraide S, Togashi H, Iizuka K, Ohkura K, Izumi T. N-methyl-d-aspartate receptor dysfunction in the prefrontal cortex of stroke-prone spontaneously hypertensive rat/Ezo as a rat model of attention deficit/hyperactivity disorder. Neuropsychopharmacol Rep 2018; 38:61-66. [PMID: 30106260 PMCID: PMC7292284 DOI: 10.1002/npr2.12007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/10/2018] [Accepted: 01/11/2018] [Indexed: 11/07/2022] Open
Abstract
AIM We previously reported that stroke-prone spontaneously hypertensive rat/Ezo (SHRSP/Ezo) has high validity as an attention deficit/hyperactivity disorder (AD/HD) animal model, based on its behavioral phenotypes, such as inattention, hyperactivity, and impulsivity. Fronto-cortical dysfunction is implicated in the pathogenesis of AD/HD. In this study, we investigated prefrontal cortex (PFC) function in SHRSP/Ezo rats by electrophysiological methods and radioreceptor assay. METHODS We recorded excitatory postsynaptic potential in layer V pyramidal neurons in the PFC by intracellular recording method to assess synaptic plasticity in the form of long-term potentiation (LTP). We also performed N-methyl-d-aspartate acid (NMDA) receptor binding assay in the PFC and hippocampus using radiolabeled NMDA receptor antagonist [3 H]MK-801. RESULTS Theta-burst stimulation induced LTP in the PFC of genetic control, WKY/Ezo, whereas failed to induce LTP in that of SHRSP/Ezo. The Kd value of [3 H]MK-801 binding for NMDA receptors in the PFC of SHRSP/Ezo was higher than in the WKY/Ezo. Neither the Bmax nor Kd of [3 H]MK-801 binding in the SHRSP/Ezo hippocampus was significantly different to WKY/Ezo. CONCLUSION These results suggest that the AD/HD animal model SHRSP/Ezo has NMDA receptor dysfunction in the PFC.
Collapse
Affiliation(s)
- Hiroki Shikanai
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| | - Nobuhiro Oshima
- Department of Biophysical Sciences, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| | - Hidekazu Kawashima
- Department of Biophysical Sciences, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| | - Shin-Ichi Kimura
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| | - Sachiko Hiraide
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| | - Hiroko Togashi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| | - Kenji Iizuka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| | - Kazue Ohkura
- Department of Biophysical Sciences, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| | - Takeshi Izumi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Japan
| |
Collapse
|
205
|
Dissecting executive control circuits with neuron types. Neurosci Res 2018; 141:13-22. [PMID: 30110598 DOI: 10.1016/j.neures.2018.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/30/2018] [Accepted: 07/23/2018] [Indexed: 12/17/2022]
Abstract
Executive control supports our ability to behave flexibly in accordance with a given situation. In order to fully understand how cortical circuits achieve this task, we need to determine the intrinsic physiological and connection profiles of neuron types and analyze their functional roles during behavior. This article introduces current knowledge regarding neuron type classification in the cortex and reviews our understanding of how each neuron type is incorporated in the functional cortical circuit to implement executive control. Recent work using neuron-type specific imaging/recording has begun to reveal significant functional organizations of pyramidal neurons and their subtypes depending on the layers and long-range projection targets. GABAergic interneurons also make crucial contributions to executive control in a subtype-specific manner. Vasoactive intestinal peptide (VIP)-positive interneurons are preferentially recruited by top-down inputs from higher-order cortical regions and amplify the signals in pyramidal neurons by inhibiting other interneuron subtypes. Particularly in the prefrontal cortex, one of the hierarchically highest cortices, executive control signals are regulated by the VIP neuron-mediated disinhibition and robustly maintained through recurrent connections at a long time scale. The differences and commonalities in the functional organization between sensory areas and the prefrontal cortex are discussed.
Collapse
|
206
|
Bedwell SA, Tinsley CJ. Mapping of fine-scale rat prefrontal cortex connections: Evidence for detailed ordering of inputs and outputs connecting the temporal cortex and sensory-motor regions. Eur J Neurosci 2018; 48:1944-1963. [DOI: 10.1111/ejn.14068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 06/11/2018] [Accepted: 07/04/2018] [Indexed: 11/29/2022]
Affiliation(s)
| | - Chris J. Tinsley
- School of Science and Technology; Nottingham Trent University; Nottingham UK
| |
Collapse
|
207
|
Rubin LH, Sacktor N, Creighton J, Du Y, Endres CJ, Pomper MG, Coughlin JM. Microglial activation is inversely associated with cognition in individuals living with HIV on effective antiretroviral therapy. AIDS 2018; 32:1661-1667. [PMID: 29746297 DOI: 10.1097/qad.0000000000001858] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Despite viral suppression, HIV-associated cognitive impairment persists and may be partially due to persistent immune signalling by cells of the myeloid-lineage. Here, we aimed to understand the contribution of activated microglia located in vulnerable brain regions (e.g. frontal, subcortical) of HIV-infected, virally suppressed (HIV+VS) individuals in relation to cognitive and motor function. DESIGN Twenty-one HIV+VS individuals underwent PET with [11C]DPA-713 to image the translocator protein 18 kDa (TSPO), a marker of microglial activation, and completed a comprehensive neuropsychological test battery. METHODS Multivariable linear regressions were used to examine the contribution of [11C]DPA-713 binding to cognitive performance. RESULTS Higher [11C]DPA-713 binding was associated with lower cognition among HIV+VS individuals. [11C]DPA-713 binding in middle frontal gyrus/frontal cortex, hippocampus/temporal cortex and occipital cortex was inversely associated with performance on a number of cognitive domains, including verbal memory, processing speed/attention/concentration, executive function, working memory and motor function. [C]DPA-713 binding in parietal cortex, cerebellum and thalamus was associated with only specific cognitive domains including visual construction and verbal memory. Binding was not associated with global cognitive performance. CONCLUSION The findings add to the growing body of evidence that immune-mediated brain injury may contribute to domain specific, HIV-associated, cognitive vulnerabilities despite viral suppression.
Collapse
|
208
|
Panayi MC, Killcross S. Functional heterogeneity within the rodent lateral orbitofrontal cortex dissociates outcome devaluation and reversal learning deficits. eLife 2018; 7:e37357. [PMID: 30044220 PMCID: PMC6101941 DOI: 10.7554/elife.37357] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 07/24/2018] [Indexed: 01/12/2023] Open
Abstract
The orbitofrontal cortex (OFC) is critical for updating reward-directed behaviours flexibly when outcomes are devalued or when task contingencies are reversed. Failure to update behaviour in outcome devaluation and reversal learning procedures are considered canonical deficits following OFC lesions in non-human primates and rodents. We examined the generality of these findings in rodents using lesions of the rodent lateral OFC (LO) in instrumental action-outcome and Pavlovian cue-outcome devaluation procedures. LO lesions disrupted outcome devaluation in Pavlovian but not instrumental procedures. Furthermore, although both anterior and posterior LO lesions disrupted Pavlovian outcome devaluation, only posterior LO lesions were found to disrupt reversal learning. Posterior but not anterior LO lesions were also found to disrupt the attribution of motivational value to Pavlovian cues in sign-tracking. These novel dissociable task- and subregion-specific effects suggest a way to reconcile contradictory findings between rodent and non-human primate OFC research.
Collapse
Affiliation(s)
- Marios C Panayi
- School of PsychologyThe University of New South WalesKensingtonAustralia
- Department of Experimental PsychologyUniversity of OxfordOxfordUnited Kingdom
| | - Simon Killcross
- School of PsychologyThe University of New South WalesKensingtonAustralia
| |
Collapse
|
209
|
Contributions of medial prefrontal cortex to decision making involving risk of punishment. Neuropharmacology 2018; 139:205-216. [PMID: 30009836 DOI: 10.1016/j.neuropharm.2018.07.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022]
Abstract
The prefrontal cortex (PFC) plays an important role in several forms of cost-benefit decision making. Its contributions to decision making under risk of explicit punishment, however, are not well understood. A rat model was used to investigate the role of the medial PFC (mPFC) and its monoaminergic innervation in a Risky Decision-making Task (RDT), in which rats chose between a small, "safe" food reward and a large, "risky" food reward accompanied by varying probabilities of mild footshock punishment. Inactivation of mPFC increased choice of the large, risky reward when the punishment probability increased across the session ("ascending RDT"), but decreased choice of the large, risky reward when the punishment probability decreased across the session ("descending RDT"). In contrast, enhancement of monoamine availability via intra-mPFC amphetamine reduced choice of the large, risky reward only in the descending RDT. Systemic administration of amphetamine reduced choice of the large, risky reward in both the ascending and descending RDT; however, this reduction was not attenuated by concurrent mPFC inactivation, indicating that mPFC is not a critical locus of amphetamine's effects on risk taking. These findings suggest that mPFC plays an important role in adapting choice behavior in response to shifting risk contingencies, but not necessarily in risk-taking behavior per se.
Collapse
|
210
|
Jhang J, Lee H, Kang MS, Lee HS, Park H, Han JH. Anterior cingulate cortex and its input to the basolateral amygdala control innate fear response. Nat Commun 2018; 9:2744. [PMID: 30013065 PMCID: PMC6048069 DOI: 10.1038/s41467-018-05090-y] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 06/12/2018] [Indexed: 11/23/2022] Open
Abstract
Prefrontal brain areas are implicated in the control of fear behavior. However, how prefrontal circuits control fear response to innate threat is poorly understood. Here, we show that the anterior cingulate cortex (ACC) and its input to the basolateral nucleus of amygdala (BLA) contribute to innate fear response to a predator odor in mice. Optogenetic inactivation of the ACC enhances freezing response to fox urine without affecting conditioned freezing. Conversely, ACC stimulation robustly inhibits both innate and conditioned freezing. Circuit tracing and slice patch recordings demonstrate a monosynaptic glutamatergic connectivity of ACC-BLA but no or very sparse ACC input to the central amygdala. Finally, our optogenetic manipulations of the ACC-BLA projection suggest its inhibitory control of innate freezing response to predator odors. Together, our results reveal the role of the ACC and its projection to BLA in innate fear response to olfactory threat stimulus. Brain circuits that control innate fear response are essential for an animal’s survival. Here, the authors report how the anterior cingulate cortex and its projection to amygdala control the innate fear response in mice.
Collapse
Affiliation(s)
- Jinho Jhang
- Department of Biological Sciences, KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Hyoeun Lee
- Department of Structure & Function of Neural Network, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, Korea
| | - Min Soo Kang
- Department of Biological Sciences, KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Han-Sol Lee
- Department of Biological Sciences, KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Hyungju Park
- Department of Structure & Function of Neural Network, Korea Brain Research Institute (KBRI), 61 Cheomdan-ro, Dong-gu, Daegu, 41068, Korea.
| | - Jin-Hee Han
- Department of Biological Sciences, KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea.
| |
Collapse
|
211
|
Mouse MRI shows brain areas relatively larger in males emerge before those larger in females. Nat Commun 2018; 9:2615. [PMID: 29976930 PMCID: PMC6033927 DOI: 10.1038/s41467-018-04921-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 06/04/2018] [Indexed: 01/08/2023] Open
Abstract
Sex differences exist in behaviors, disease and neuropsychiatric disorders. Sexual dimorphisms however, have yet to be studied across the whole brain and across a comprehensive time course of postnatal development. Here, we use manganese-enhanced MRI (MEMRI) to longitudinally image male and female C57BL/6J mice across 9 time points, beginning at postnatal day 3. We recapitulate findings on canonically dimorphic areas, demonstrating MEMRI’s ability to study neuroanatomical sex differences. We discover, upon whole-brain volume correction, that neuroanatomical regions larger in males develop earlier than those larger in females. Groups of areas with shared sexually dimorphic developmental trajectories reflect behavioral and functional networks, and expression of genes involved with sex processes. Also, post-pubertal neuroanatomy is highly individualized, and individualization occurs earlier in males. Our results demonstrate the ability of MEMRI to reveal comprehensive developmental differences between male and female brains, which will improve our understanding of sex-specific predispositions to various neuropsychiatric disorders. Sex differences occur in various aspects of neurodevelopment. Here the authors use manganese-enhanced MRI at nine different postnatal stages to detail the development of structural sex differences in the mouse brain.
Collapse
|
212
|
Kawaguchi Y. Pyramidal Cell Subtypes and Their Synaptic Connections in Layer 5 of Rat Frontal Cortex. Cereb Cortex 2018; 27:5755-5771. [PMID: 29028949 DOI: 10.1093/cercor/bhx252] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/06/2017] [Indexed: 12/31/2022] Open
Abstract
The frontal cortical areas make a coordinated response that generates appropriate behavior commands, using individual local circuits with corticostriatal and corticocortical connections in longer time scales than sensory areas. In secondary motor cortex (M2), situated between the prefrontal and primary motor areas, major subtypes of layer 5 corticostriatal cells are crossed-corticostriatal (CCS) cells innervating both sides of striatum, and corticopontine (CPn) cells projecting to the ipsilateral striatum and pontine nuclei. CCS cells innervate CPn cells unidirectionally: the former are therefore hierarchically higher than the latter among L5 corticostriatal cells. CCS cells project directly to both frontal and nonfrontal areas. On the other hand, CPn cells innervate the thalamus and layer 1a of frontal areas, where thalamic fibers relaying basal ganglia outputs are distributed. Thus, CCS cells can make activities of frontal areas in concert with those of nonfrontal area using corticocortical loops, whereas CPn cells are more involved in closed corticostriatal loops than CCS cells. Since reciprocal connections between CPn cells with facilitatory synapses may be related to persistent activity, CPn cells play a key role of longer time constant processes in corticostriatal as well as in corticocortical loops between the frontal areas.
Collapse
Affiliation(s)
- Yasuo Kawaguchi
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, Okazaki 444-8787, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Japan
| |
Collapse
|
213
|
Mathis V, Barbelivien A, Majchrzak M, Mathis C, Cassel JC, Lecourtier L. The Lateral Habenula as a Relay of Cortical Information to Process Working Memory. Cereb Cortex 2018; 27:5485-5495. [PMID: 28334072 DOI: 10.1093/cercor/bhw316] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/19/2016] [Indexed: 11/14/2022] Open
Abstract
Working memory is a cognitive ability allowing the temporary storage of information to solve problems or adjust behavior. While working memory is known to mainly depend on the medial prefrontal cortex (mPFC), very few is known about how cortical information are relayed subcortically. By its connectivity, the lateral habenula (lHb) might act as a subcortical relay for cortical information. Indeed, the lHb receives inputs from several mPFC subregions, and recent findings suggest a role for the lHb in online processing of spatial information, a fundamental aspect of working memory. In rats, in a delayed non-matching to position paradigm, using focal microinjections of the GABAA agonist muscimol we showed that inactivation of the lHb (16 ng in 0.2 µL per side), as well as disconnection between the prelimbic region of the mPFC (mPFC/PrL, 32 ng in 0.4 µL in one hemisphere) and the lHb (16 ng in 0.2 µL in the lHb in the contralateral hemisphere) impaired working memory. The deficits were unlikely to result from motivational or motor deficits as muscimol did not affect reward collection or cue responding latencies, and did not increase the number of omissions. These results show for the first time the implication of the lHb in mPFC-dependent memory processes, likely as a relay of mPFC/PrL information. They also open new perspectives in the understanding of the top-down processing of high-level cognitive functions.
Collapse
Affiliation(s)
- Victor Mathis
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, F-67000 Strasbourg, France.,LNCA, UMR 7364, CNRS, F-67000 Strasbourg, France
| | - Alexandra Barbelivien
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, F-67000 Strasbourg, France.,LNCA, UMR 7364, CNRS, F-67000 Strasbourg, France
| | - Monique Majchrzak
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, F-67000 Strasbourg, France.,LNCA, UMR 7364, CNRS, F-67000 Strasbourg, France
| | - Chantal Mathis
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, F-67000 Strasbourg, France.,LNCA, UMR 7364, CNRS, F-67000 Strasbourg, France
| | - Jean-Christophe Cassel
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, F-67000 Strasbourg, France.,LNCA, UMR 7364, CNRS, F-67000 Strasbourg, France
| | - Lucas Lecourtier
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, F-67000 Strasbourg, France.,LNCA, UMR 7364, CNRS, F-67000 Strasbourg, France
| |
Collapse
|
214
|
Negrón-Oyarzo I, Espinosa N, Aguilar-Rivera M, Fuenzalida M, Aboitiz F, Fuentealba P. Coordinated prefrontal-hippocampal activity and navigation strategy-related prefrontal firing during spatial memory formation. Proc Natl Acad Sci U S A 2018; 115:7123-7128. [PMID: 29915053 PMCID: PMC6142212 DOI: 10.1073/pnas.1720117115] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Learning the location of relevant places in the environment is crucial for survival. Such capacity is supported by a distributed network comprising the prefrontal cortex and hippocampus, yet it is not fully understood how these structures cooperate during spatial reference memory formation. Hence, we examined neural activity in the prefrontal-hippocampal circuit in mice during acquisition of spatial reference memory. We found that interregional oscillatory coupling increased with learning, specifically in the slow-gamma frequency (20 to 40 Hz) band during spatial navigation. In addition, mice used both spatial and nonspatial strategies to navigate and solve the task, yet prefrontal neuronal spiking and oscillatory phase coupling were selectively enhanced in the spatial navigation strategy. Lastly, a representation of the behavioral goal emerged in prefrontal spiking patterns exclusively in the spatial navigation strategy. These results suggest that reference memory formation is supported by enhanced cortical connectivity and evolving prefrontal spiking representations of behavioral goals.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Instituto de Fisiología, and Centro de Neurobiología y Fisiopatología Integrativa, Facultad de Ciencias, Universidad de Valparaíso, 2340000 Valparaíso, Chile
- Centro Integrativo de Neurociencias y Departamento de Psiquiatría, Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile
| | - Nelson Espinosa
- Centro Integrativo de Neurociencias y Departamento de Psiquiatría, Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile
| | - Marcelo Aguilar-Rivera
- Centro Integrativo de Neurociencias y Departamento de Psiquiatría, Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile
| | - Marco Fuenzalida
- Instituto de Fisiología, and Centro de Neurobiología y Fisiopatología Integrativa, Facultad de Ciencias, Universidad de Valparaíso, 2340000 Valparaíso, Chile
| | - Francisco Aboitiz
- Centro Integrativo de Neurociencias y Departamento de Psiquiatría, Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile
| | - Pablo Fuentealba
- Centro Integrativo de Neurociencias y Departamento de Psiquiatría, Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile;
- Centro de Investigación en Nanotecnología y Materiales Avanzados, Pontificia Universidad Católica de Chile, 7820436 Santiago, Chile
| |
Collapse
|
215
|
Sekeres MJ, Winocur G, Moscovitch M. The hippocampus and related neocortical structures in memory transformation. Neurosci Lett 2018; 680:39-53. [DOI: 10.1016/j.neulet.2018.05.006] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/23/2022]
|
216
|
Wang X, Allen WE, Wright MA, Sylwestrak EL, Samusik N, Vesuna S, Evans K, Liu C, Ramakrishnan C, Liu J, Nolan GP, Bava FA, Deisseroth K. Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science 2018; 361:science.aat5691. [PMID: 29930089 PMCID: PMC6339868 DOI: 10.1126/science.aat5691] [Citation(s) in RCA: 776] [Impact Index Per Article: 129.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/01/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022]
Abstract
Retrieving high-content gene-expression information while retaining three-dimensional (3D) positional anatomy at cellular resolution has been difficult, limiting integrative understanding of structure and function in complex biological tissues. We developed and applied a technology for 3D intact-tissue RNA sequencing, termed STARmap (spatially-resolved transcript amplicon readout mapping), which integrates hydrogel-tissue chemistry, targeted signal amplification, and in situ sequencing. The capabilities of STARmap were tested by mapping 160 to 1020 genes simultaneously in sections of mouse brain at single-cell resolution with high efficiency, accuracy, and reproducibility. Moving to thick tissue blocks, we observed a molecularly defined gradient distribution of excitatory-neuron subtypes across cubic millimeter-scale volumes (>30,000 cells) and a short-range 3D self-clustering in many inhibitory-neuron subtypes that could be identified and described with 3D STARmap.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - William E Allen
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.,Neuroscience Program, Stanford University, CA 94305, USA
| | - Matthew A Wright
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.,Department of Psychiatry and Behavioral Sciences, Stanford University, CA 94305, USA
| | - Emily L Sylwestrak
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Nikolay Samusik
- Baxter Laboratory, Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Sam Vesuna
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kathryn Evans
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Cindy Liu
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Jia Liu
- Department of Chemical Engineering, Stanford University, CA 94305, USA
| | - Garry P Nolan
- Baxter Laboratory, Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA.
| | - Felice-Alessio Bava
- Baxter Laboratory, Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA.
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA. .,Department of Psychiatry and Behavioral Sciences, Stanford University, CA 94305, USA.,Howard Hughes Medical Institute, Stanford University, CA 94305, USA
| |
Collapse
|
217
|
Rouault M, Koechlin E. Prefrontal function and cognitive control: from action to language. Curr Opin Behav Sci 2018. [DOI: 10.1016/j.cobeha.2018.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
218
|
Abstract
OBJECTIVE Stroke patients often suffer from delayed disturbances of mood and cognition. In rodents, the prefrontal cortex (PFC) is involved in both higher order cognition and emotion. Our objective was to determine if bilateral focal ischaemic lesions restricted to the medial prefrontal cortex (mPFC) could be used to model post-stroke anxiety and/or cognitive deficits. METHODS Groups of adult male Sprague-Dawley rats (n=9) received bilateral injections of either endothelin-1 (ET-1) (400 pmol) or vehicle (artificial cerebrospinal fluid) into the mPFC and were tested at various times using both a test of temporal order memory and in an elevated plus maze. Lesions were verified histologically. RESULTS ET-1 lesioned rats had reduced mobility on post-surgery day 8 that had resolved by day 29 at which time they spent significantly more time in the closed arm of the plus maze CONCLUSION: We conclude that ischaemic lesions localised to the mPFC can be used to model post-stroke anxiety in rats.
Collapse
|
219
|
Bueno-Junior LS, Leite JP. Input Convergence, Synaptic Plasticity and Functional Coupling Across Hippocampal-Prefrontal-Thalamic Circuits. Front Neural Circuits 2018; 12:40. [PMID: 29875637 PMCID: PMC5975431 DOI: 10.3389/fncir.2018.00040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/26/2018] [Indexed: 01/19/2023] Open
Abstract
Executive functions and working memory are long known to involve the prefrontal cortex (PFC), and two PFC-projecting areas: midline/paramidline thalamus (MLT) and cornus ammonis 1 (CA1)/subiculum of the hippocampal formation (HF). An increasing number of rodent electrophysiology studies are examining these substrates together, thus providing circuit-level perspectives on input convergence, synaptic plasticity and functional coupling, as well as insights into cognition mechanisms and brain disorders. Our review article puts this literature into a method-oriented narrative. As revisited throughout the text, limbic thalamic and hippocampal afferents to the PFC gate one another’s inputs, which in turn are modulated by PFC interneurons and ascending monoaminergic projections. In addition, long-term synaptic plasticity, paired-pulse facilitation (PPF), and event-related potentials (ERP) dynamically vary across PFC-related circuits during learning paradigms and drug effects. Finally, thalamic-prefrontal loops, which have been shown to amplify both cognitive processes and limbic seizures, are also being implicated as relays in the prefrontal-hippocampal feedback, contributing to spatial navigation and decision making. Based on these issues, we conclude the review with a critical synthesis and some research directions.
Collapse
Affiliation(s)
- Lezio S Bueno-Junior
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Joao P Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
220
|
Ye Y, Mastwal S, Cao VY, Ren M, Liu Q, Zhang W, Elkahloun AG, Wang KH. Dopamine is Required for Activity-Dependent Amplification of Arc mRNA in Developing Postnatal Frontal Cortex. Cereb Cortex 2018; 27:3600-3608. [PMID: 27365296 DOI: 10.1093/cercor/bhw181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The activity-regulated gene Arc/Arg3.1 encodes a postsynaptic protein crucially involved in glutamatergic synaptic plasticity. Genetic mutations in Arc pathway and altered Arc expression in human frontal cortex have been associated with schizophrenia. Although Arc expression has been reported to vary with age, what mechanisms regulate Arc mRNA levels in frontal cortex during postnatal development remains unclear. Using quantitative mRNA analysis of mouse frontal cortical tissues, we mapped the developmental profiles of Arc expression and found that its mRNA levels are sharply amplified near the end of the second postnatal week, when mouse pups open their eyes for the first time after birth. Surprisingly, electrical stimulation of the frontal cortex before eye-opening is not sufficient to drive the amplification of Arc mRNA. Instead, this amplification needs both electrical stimulation and dopamine D1-type receptor (D1R) activation. Furthermore, visual stimuli-driven amplification of Arc mRNA is also dependent on D1R activation and dopamine neurons located in the ventral midbrain. These results indicate that dopamine is required to drive activity-dependent amplification of Arc mRNA in the developing postnatal frontal cortex and suggest that joint electrical and dopaminergic activation is essential to establish the normal expression pattern of a schizophrenia-associated gene during frontal cortical development.
Collapse
Affiliation(s)
- Yizhou Ye
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Surjeet Mastwal
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vania Yu Cao
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ming Ren
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qing Liu
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenyu Zhang
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abdel G Elkahloun
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kuan Hong Wang
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
221
|
Ferguson BR, Gao WJ. PV Interneurons: Critical Regulators of E/I Balance for Prefrontal Cortex-Dependent Behavior and Psychiatric Disorders. Front Neural Circuits 2018; 12:37. [PMID: 29867371 PMCID: PMC5964203 DOI: 10.3389/fncir.2018.00037] [Citation(s) in RCA: 360] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/17/2018] [Indexed: 01/20/2023] Open
Abstract
Elucidating the prefrontal cortical microcircuit has been challenging, given its role in multiple complex behaviors, including working memory, cognitive flexibility, attention, social interaction and emotional regulation. Additionally, previous methodological limitations made it difficult to parse out the contribution of certain neuronal subpopulations in refining cortical representations. However, growing evidence supports a fundamental role of fast-spiking parvalbumin (PV) GABAergic interneurons in regulating pyramidal neuron activity to drive appropriate behavioral responses. Further, their function is heavily diminished in the prefrontal cortex (PFC) in numerous psychiatric diseases, including schizophrenia and autism. Previous research has demonstrated the importance of the optimal balance of excitation and inhibition (E/I) in cortical circuits in maintaining the efficiency of cortical information processing. Although we are still unraveling the mechanisms of information representation in the PFC, the E/I balance seems to be crucial, as pharmacological, chemogenetic and optogenetic approaches for disrupting E/I balance induce impairments in a range of PFC-dependent behaviors. In this review, we will explore two key hypotheses. First, PV interneurons are powerful regulators of E/I balance in the PFC, and help optimize the representation and processing of supramodal information in PFC. Second, diminishing the function of PV interneurons is sufficient to generate an elaborate symptom sequelae corresponding to those observed in a range of psychiatric diseases. Then, using this framework, we will speculate on whether this circuitry could represent a platform for the development of therapeutic interventions in disorders of PFC function.
Collapse
Affiliation(s)
- Brielle R Ferguson
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States.,Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, United States
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
222
|
Abstract
A longstanding controversy in neuroscience pertains to differences in human prefrontal cortex (PFC) compared with other primate species; specifically, is human PFC disproportionately large? Distinctively human behavioral capacities related to higher cognition and affect presumably arose from evolutionary modifications since humans and great apes diverged from a common ancestor about 6–8 Mya. Accurate determination of regional differences in the amount of cortical gray and subcortical white matter content in humans, great apes, and Old World monkeys can further our understanding of the link between structure and function of the human brain. Using tissue volume analyses, we show a disproportionately large amount of gray and white matter corresponding to PFC in humans compared with nonhuman primates. Humans have the largest cerebral cortex among primates. The question of whether association cortex, particularly prefrontal cortex (PFC), is disproportionately larger in humans compared with nonhuman primates is controversial: Some studies report that human PFC is relatively larger, whereas others report a more uniform PFC scaling. We address this controversy using MRI-derived cortical surfaces of many individual humans, chimpanzees, and macaques. We present two parcellation-based PFC delineations based on cytoarchitecture and function and show that a previously used morphological surrogate (cortex anterior to the genu of the corpus callosum) substantially underestimates PFC extent, especially in humans. We find that the proportion of cortical gray matter occupied by PFC in humans is up to 1.9-fold greater than in macaques and 1.2-fold greater than in chimpanzees. The disparity is even more prominent for the proportion of subcortical white matter underlying the PFC, which is 2.4-fold greater in humans than in macaques and 1.7-fold greater than in chimpanzees.
Collapse
|
223
|
Varodayan FP, Sidhu H, Kreifeldt M, Roberto M, Contet C. Morphological and functional evidence of increased excitatory signaling in the prelimbic cortex during ethanol withdrawal. Neuropharmacology 2018; 133:470-480. [PMID: 29471053 PMCID: PMC5865397 DOI: 10.1016/j.neuropharm.2018.02.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/26/2018] [Accepted: 02/16/2018] [Indexed: 02/06/2023]
Abstract
Excessive alcohol consumption in humans induces deficits in decision making and emotional processing, which indicates a dysfunction of the prefrontal cortex (PFC). The present study aimed to determine the impact of chronic intermittent ethanol (CIE) inhalation on mouse medial PFC pyramidal neurons. Data were collected 6-8 days into withdrawal from 7 weeks of CIE exposure, a time point when mice exhibit behavioral symptoms of withdrawal. We found that spine maturity in prelimbic (PL) layer 2/3 neurons was increased, while dendritic spines in PL layer 5 neurons or infralimbic (IL) neurons were not affected. Corroborating these morphological observations, CIE enhanced glutamatergic transmission in PL layer 2/3 pyramidal neurons, but not IL layer 2/3 neurons. Contrary to our predictions, these cellular alterations were associated with improved, rather than impaired, performance in reversal learning and strategy switching tasks in the Barnes maze at an earlier stage of chronic ethanol exposure (5-7 days withdrawal from 3 to 4 weeks of CIE), which could result from the anxiety-like behavior associated with ethanol withdrawal. Altogether, this study adds to a growing body of literature indicating that glutamatergic activity in the PFC is upregulated following chronic ethanol exposure, and identifies PL layer 2/3 pyramidal neurons as a sensitive target of synaptic remodeling. It also indicates that the Barnes maze is not suitable to detect deficits in cognitive flexibility in CIE-withdrawn mice.
Collapse
Affiliation(s)
| | - Harpreet Sidhu
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Max Kreifeldt
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Marisa Roberto
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Candice Contet
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA.
| |
Collapse
|
224
|
Mashhoori A, Hashemnia S, McNaughton BL, Euston DR, Gruber AJ. Rat anterior cingulate cortex recalls features of remote reward locations after disfavoured reinforcements. eLife 2018; 7:29793. [PMID: 29664400 PMCID: PMC5931797 DOI: 10.7554/elife.29793] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 04/04/2018] [Indexed: 12/30/2022] Open
Abstract
The anterior cingulate cortex (ACC) encodes information supporting mnemonic and cognitive processes. We show here that a rat’s position can be decoded with high spatiotemporal resolution from ACC activity. ACC neurons encoded the current state of the animal and task, except for brief excursions that sometimes occurred at target feeders. During excursions, the decoded position became more similar to a remote target feeder than the rat’s physical position. Excursions recruited activation of neurons encoding choice and reward, and the likelihood of excursions at a feeder was inversely correlated with feeder preference. These data suggest that the excursion phenomenon was related to evaluating real or fictive choice outcomes, particularly after disfavoured reinforcements. We propose that the multiplexing of position with choice-related information forms a mental model isomorphic with the task space, which can be mentally navigated via excursions to recall multimodal information about the utility of remote locations.
Collapse
Affiliation(s)
- Ali Mashhoori
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Alberta, Canada
| | - Saeedeh Hashemnia
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Alberta, Canada
| | - Bruce L McNaughton
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Alberta, Canada
| | - David R Euston
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Alberta, Canada
| | - Aaron J Gruber
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Alberta, Canada
| |
Collapse
|
225
|
Collins DP, Anastasiades PG, Marlin JJ, Carter AG. Reciprocal Circuits Linking the Prefrontal Cortex with Dorsal and Ventral Thalamic Nuclei. Neuron 2018; 98:366-379.e4. [PMID: 29628187 DOI: 10.1016/j.neuron.2018.03.024] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 02/01/2018] [Accepted: 03/14/2018] [Indexed: 12/12/2022]
Abstract
Reciprocal interactions between the prefrontal cortex (PFC) and thalamus play a critical role in cognition, but the underlying circuits remain poorly understood. Here we use optogenetics to dissect the specificity and dynamics of cortico-thalamo-cortical networks in the mouse brain. We find that cortico-thalamic (CT) neurons in prelimbic PFC project to both mediodorsal (MD) and ventromedial (VM) thalamus, where layer 5 and 6 inputs activate thalamo-cortical (TC) neurons with distinct temporal profiles. We show that TC neurons in MD and VM in turn make distinct connections in PFC, with MD preferentially and strongly activating layer 2/3 cortico-cortical (CC) neurons. Finally, we assess local connections from superficial CC to deep CT neurons, which link thalamo-cortical and cortico-thalamic networks within the PFC. Together our findings indicate that PFC strongly drives neurons in the thalamus, whereas MD and VM indirectly influence reciprocally connected neurons in the PFC, providing a mechanistic understanding of these circuits.
Collapse
Affiliation(s)
- David P Collins
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Paul G Anastasiades
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Joseph J Marlin
- 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
|
226
|
Pfarr S, Schaaf L, Reinert JK, Paul E, Herrmannsdörfer F, Roßmanith M, Kuner T, Hansson AC, Spanagel R, Körber C, Sommer WH. Choice for Drug or Natural Reward Engages Largely Overlapping Neuronal Ensembles in the Infralimbic Prefrontal Cortex. J Neurosci 2018; 38:3507-3519. [PMID: 29483279 PMCID: PMC6596043 DOI: 10.1523/jneurosci.0026-18.2018] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/04/2018] [Accepted: 02/09/2018] [Indexed: 12/19/2022] Open
Abstract
Cue-reward associations form distinct memories that can drive appetitive behaviors and are involved in craving for both drugs and natural rewards. Distinct sets of neurons, so-called neuronal ensembles, in the infralimbic area (IL) of the medial prefrontal cortex (mPFC) play a key role in alcohol seeking. Whether this ensemble is specific for alcohol or controls reward seeking in general remains unclear. Here, we compared IL ensembles formed upon recall of drug (alcohol) or natural reward (saccharin) memories in male Wistar rats. Using an experimental framework that allows identification of two distinct reward-associated ensembles within the same animal, we found that cue-induced seeking of either alcohol or saccharin activated ensembles of similar size and organization, whereby these ensembles consist of largely overlapping neuronal populations. Thus, the IL seems to act as a general integration hub for reward seeking behavior, but also contains subsets of neurons that encode for the different rewards.SIGNIFICANCE STATEMENT Cue-reward associations form distinct memories that can act as drivers of appetitive behaviors and are involved in craving for natural rewards as well as for drugs. Distinct sets of neurons, so-called neuronal ensembles, in the infralimbic area of the mPFC play a key role in cue-triggered reward seeking. However, it is unclear whether these ensembles act as broadly tuned controllers of approach behavior or represent the learned associations between specific cues and rewards. Using an experimental framework that allows identification of two distinct reward-associated ensembles within the same animal we find largely overlapping neuronal populations. Repeated activation by two distinct events could reflect the linking of the two memory traces within the same neuron.
Collapse
Affiliation(s)
| | | | - Janine K Reinert
- Institute of Anatomy and Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, 69120 Heidelberg, Germany
| | | | - Frank Herrmannsdörfer
- Institute of Anatomy and Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, 69120 Heidelberg, Germany
| | | | - Thomas Kuner
- Institute of Anatomy and Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, 69120 Heidelberg, Germany
| | | | | | - Christoph Körber
- Institute of Anatomy and Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, 69120 Heidelberg, Germany
| | - Wolfgang H Sommer
- Institute of Psychopharmacology,
- Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany, and
| |
Collapse
|
227
|
Wass C, Sauce B, Pizzo A, Matzel LD. Dopamine D1 receptor density in the mPFC responds to cognitive demands and receptor turnover contributes to general cognitive ability in mice. Sci Rep 2018; 8:4533. [PMID: 29540721 PMCID: PMC5852043 DOI: 10.1038/s41598-018-22668-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/27/2018] [Indexed: 01/11/2023] Open
Abstract
In both humans and mice, performance on tests of intelligence or general cognitive ability (GCA) is related to dopamine D1 receptor-mediated activity in the prelimbic cortex, and levels of DRD1 mRNA predict the GCA of mice. Here we assessed the turnover rate of D1 receptors as well as the expression level of the D1 chaperone protein (DRiP78) in the medial PPC (mPFC) of mice to determine whether rate of receptor turnover was associated with variations in the GCA of genetically heterogeneous mice. Following assessment of GCA (aggregate performance on four diverse learning tests) mice were administered an irreversible dopamine receptor antagonist (EEDQ), after which the density of new D1 receptors were quantified. GCA was positively correlated with both the rate of D1 receptor recovery and levels of DRiP78. Additionally, the density of D1 receptors was observed to increase within 60 min (or less) in response to intense demands on working memory, suggesting that a pool of immature receptors was available to accommodate high cognitive loads. These results provide evidence that innate general cognitive abilities are related to D1 receptor turnover rates in the prefrontal cortex, and that an intracellular pool of immature D1 receptors are available to accommodate cognitive demands.
Collapse
Affiliation(s)
- Christopher Wass
- Department of Psychology, Program in Behavioral and Systems Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Bruno Sauce
- Department of Psychology, Program in Behavioral and Systems Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Alessandro Pizzo
- Department of Psychology, Program in Behavioral and Systems Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Louis D Matzel
- Department of Psychology, Program in Behavioral and Systems Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
228
|
Priya V, Srikumar BN, Shankaranarayana Rao BS. Contrasting effects of pre-training on acquisition of operant and radial arm maze tasks in rats. J Integr Neurosci 2018:JIN077. [PMID: 29562551 DOI: 10.3233/jin-180077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Performing multiple tasks either simultaneously, in rapid alternation or in succession, is routine in daily life. Further, testing rodents in a battery of tests is common both in drug discovery and behavioral phenotyping research. However, learning of new tasks can be influenced by prior experience(s). There has been some research on 'switching cost' involved in the transition from one behavior to another. However, there has been no specific assessment of the effect of learning an operant paradigm on performance in a spatial memory task and vice versa. Accordingly, we evaluated task switching between two forms of learning paradigms, operant conditioning and radial arm maze (RAM) tasks. In experiment 1, rats were trained for operant conditioning with food reward followed by a partially baited RAM task. In experiment 2, rats were trained first on a RAM task followed by operant learning. Pre-training on the operant task, impaired the acquisition of the RAM. On the contrary, pre-training on the RAM enhanced operant performance. Our study reveals significant effects of the test order on task-switching in rats. This knowledge can be useful when framing test sequences in test batteries for drug discovery research and screening genetically modified mice.
Collapse
Affiliation(s)
- V Priya
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bengaluru - 560 029, India
| | - B N Srikumar
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bengaluru - 560 029, India
| | - B S Shankaranarayana Rao
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bengaluru - 560 029, India
| |
Collapse
|
229
|
Konstantoudaki X, Chalkiadaki K, Vasileiou E, Kalemaki K, Karagogeos D, Sidiropoulou K. Prefrontal cortical-specific differences in behavior and synaptic plasticity between adolescent and adult mice. J Neurophysiol 2018; 119:822-833. [DOI: 10.1152/jn.00189.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adolescence is a highly vulnerable period for the emergence of major neuropsychological disorders and is characterized by decreased cognitive control and increased risk-taking behavior and novelty-seeking. The prefrontal cortex (PFC) is involved in the cognitive control of impulsive and risky behavior. Although the PFC is known to reach maturation later than other cortical areas, little information is available regarding the functional changes from adolescence to adulthood in PFC, particularly compared with other primary cortical areas. This study aims to understand the development of PFC-mediated, compared with non-PFC-mediated, cognitive functions. Toward this aim, we performed cognitive behavioral tasks in adolescent and adult mice and subsequently investigated synaptic plasticity in two different cortical areas. Our results showed that adolescent mice exhibit impaired performance in PFC-dependent cognitive tasks compared with adult mice, whereas their performance in non-PFC-dependent tasks is similar to that of adults. Furthermore, adolescent mice exhibited decreased long-term potentiation (LTP) within upper-layer synapses of the PFC but not the barrel cortex. Blocking GABAA receptor function significantly augments LTP in both the adolescent and adult PFC. No change in intrinsic excitability of PFC pyramidal neurons was observed between adolescent and adult mice. Finally, increased expression of the NR2A subunit of the N-methyl-d-aspartate receptors is found only in the adult PFC, a change that could underlie the emergence of LTP. In conclusion, our results demonstrate physiological and behavioral changes during adolescence that are specific to the PFC and could underlie the reduced cognitive control in adolescents. NEW & NOTEWORTHY This study reports that adolescent mice exhibit impaired performance in cognitive functions dependent on the prefrontal cortex but not in cognitive functions dependent on other cortical regions. The current results propose reduced synaptic plasticity in the upper layers of the prefrontal cortex as a cellular correlate of this weakened cognitive function. This decreased synaptic plasticity is due to reduced N-methyl-d-aspartate receptor expression but not due to dampened intrinsic excitability or enhanced GABAergic signaling during adolescence.
Collapse
Affiliation(s)
| | | | | | - Katerina Kalemaki
- Division of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology – Hellas, Heraklion, Greece
| | - Domna Karagogeos
- Division of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology – Hellas, Heraklion, Greece
| | | |
Collapse
|
230
|
Grünewald L, Becker N, Camphausen A, O'Leary A, Lesch KP, Freudenberg F, Reif A. Expression of the ADHD candidate gene Diras2 in the brain. J Neural Transm (Vienna) 2018; 125:913-923. [PMID: 29488099 DOI: 10.1007/s00702-018-1867-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 02/21/2018] [Indexed: 12/21/2022]
Abstract
The distinct subgroup of the Ras family member 2 (DIRAS2) gene has been found to be associated with attention-deficit/hyperactivity disorder (ADHD) in one of our previous studies. This gene is coding for a small Ras GTPase with unknown function. DIRAS2 is highly expressed in the brain. However, the exact neural expression pattern of this gene was unknown so far. Therefore, we investigated the expressional profile of DIRAS2 in the human and murine brain. In the present study, qPCR analyses in the human and in the developing mouse brain, immunocytological double staining on murine hippocampal primary cells and RNA in situ hybridization (ISH) on brain sections of C57BL/6J wild-type mice, have been used to reveal the expression pattern of DIRAS2 in the brain. We could show that DIRAS2 expression in the human brain is the highest in the hippocampus and the cerebral cortex, which is in line with the ISH results in the mouse brain. During mouse brain development, Diras2 levels strongly increase from prenatal to late postnatal stages. Co-expression studies indicate Diras2 expression in glutamatergic and catecholaminergic neurons. Our findings support the idea of DIRAS2 as a candidate gene for ADHD as the timeline of its expression as well as the brain regions and cell types that show Diras2 expression correspond to those assumed to underlie the pathomechanisms of the disease.
Collapse
Affiliation(s)
- Lena Grünewald
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany.
| | - Nils Becker
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Annika Camphausen
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| | - Klaus-Peter Lesch
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany
| |
Collapse
|
231
|
Okazaki H, Hayashi-Takagi A, Nagaoka A, Negishi M, Ucar H, Yagishita S, Ishii K, Toyoizumi T, Fox K, Kasai H. Calcineurin knockout mice show a selective loss of small spines. Neurosci Lett 2018; 671:99-102. [PMID: 29427598 DOI: 10.1016/j.neulet.2018.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/26/2018] [Accepted: 02/03/2018] [Indexed: 01/26/2023]
Abstract
Calcineurin is required for long-term depression and activity-dependent spine shrinkage, and calcineurin mutations have been identified in patients with schizophrenia. Moreover, mice with conditional knockout of calcineurin B (CNB-KO) exhibit behavioral abnormalities suggestive of schizophrenia. Changes in the dendritic spines of these mice, however, have not been investigated. We therefore examined the dendritic spines of CNB-KO mice, and observed a significant reduction in small spines and an increase in large spines in the prefrontal and visual cortices. The effect of CNB-KO on the spine sizes was relatively moderate, possibly due to the presence of spontaneous fluctuations (dynamics) in the dendritic spines themselves. Thus, CNB-KO mice showed a spine phenotype similar to those recently reported in patients with schizophrenia.
Collapse
Affiliation(s)
- Hitoshi Okazaki
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Akiko Hayashi-Takagi
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Laboratory of Medical Neuroscience, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi-city, Gunma, Japan; PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Akira Nagaoka
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Makiko Negishi
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hasan Ucar
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sho Yagishita
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kazuhiko Ishii
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Taro Toyoizumi
- RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Kevin Fox
- School of Bioscience, Cardiff University, Cardiff, UK
| | - Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
232
|
Ouhaz Z, Fleming H, Mitchell AS. Cognitive Functions and Neurodevelopmental Disorders Involving the Prefrontal Cortex and Mediodorsal Thalamus. Front Neurosci 2018; 12:33. [PMID: 29467603 PMCID: PMC5808198 DOI: 10.3389/fnins.2018.00033] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/15/2018] [Indexed: 11/13/2022] Open
Abstract
The mediodorsal nucleus of the thalamus (MD) has been implicated in executive functions (such as planning, cognitive control, working memory, and decision-making) because of its significant interconnectivity with the prefrontal cortex (PFC). Yet, whilst the roles of the PFC have been extensively studied, how the MD contributes to these cognitive functions remains relatively unclear. Recently, causal evidence in monkeys has demonstrated that in everyday tasks involving rapid updating (e.g., while learning something new, making decisions, or planning the next move), the MD and frontal cortex are working in close partnership. Furthermore, researchers studying the MD in rodents have been able to probe the underlying mechanisms of this relationship to give greater insights into how the frontal cortex and MD might interact during the performance of these essential tasks. This review summarizes the circuitry and known neuromodulators of the MD, and considers the most recent behavioral, cognitive, and neurophysiological studies conducted in monkeys and rodents; in total, this evidence demonstrates that MD makes a critical contribution to cognitive functions. We propose that communication occurs between the MD and the frontal cortex in an ongoing, fluid manner during rapid cognitive operations, via the means of efference copies of messages passed through transthalamic routes; the conductance of these messages may be modulated by other brain structures interconnected to the MD. This is similar to the way in which other thalamic structures have been suggested to carry out forward modeling associated with rapid motor responding and visual processing. Given this, and the marked thalamic pathophysiology now identified in many neuropsychiatric disorders, we suggest that changes in the different subdivisions of the MD and their interconnections with the cortex could plausibly give rise to a number of the otherwise disparate symptoms (including changes to olfaction and cognitive functioning) that are associated with many different neuropsychiatric disorders. In particular, we will focus here on the cognitive symptoms of schizophrenia and suggest testable hypotheses about how changes to MD-frontal cortex interactions may affect cognitive processes in this disorder.
Collapse
Affiliation(s)
- Zakaria Ouhaz
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Hugo Fleming
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Anna S Mitchell
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
233
|
Igonina TN, Ragaeva DS, Tikhonova MA, Petrova OM, Herbeck YE, Rozhkova IN, Amstislavskaya TG, Amstislavsky SY. Neurodevelopment and behavior in neonatal OXYS rats with genetically determined accelerated senescence. Brain Res 2018; 1681:75-84. [DOI: 10.1016/j.brainres.2017.12.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/14/2017] [Accepted: 12/18/2017] [Indexed: 12/29/2022]
|
234
|
Notter T, Coughlin JM, Gschwind T, Weber-Stadlbauer U, Wang Y, Kassiou M, Vernon AC, Benke D, Pomper MG, Sawa A, Meyer U. Translational evaluation of translocator protein as a marker of neuroinflammation in schizophrenia. Mol Psychiatry 2018; 23:323-334. [PMID: 28093569 DOI: 10.1038/mp.2016.248] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/14/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023]
Abstract
Positron emission tomography (PET) imaging with radiotracers that target translocator protein 18 kDa (TSPO) has become a popular approach to assess putative neuroinflammatory processes and associated microglia activation in psychotic illnesses. It remains unclear, however, whether TSPO imaging can accurately capture low-grade inflammatory processes such as those present in schizophrenia and related disorders. Therefore, we evaluated the validity of TSPO as a disease-relevant marker of inflammation using a translational approach, which combined neurodevelopmental and neurodegenerative mouse models with PET imaging in patients with recent-onset schizophrenia and matched controls. Using an infection-mediated neurodevelopmental mouse model, we show that schizophrenia-relevant behavioral abnormalities and increased inflammatory cytokine expression are associated with reduced prefrontal TSPO levels. On the other hand, TSPO was markedly upregulated in a mouse model of acute neurodegeneration and reactive gliosis, which was induced by intrahippocampal injection of kainic acid. In both models, the changes in TSPO levels were not restricted to microglia but emerged in various cell types, including microglia, astrocytes and vascular endothelial cells. Human PET imaging using the second-generation TSPO radiotracer [11C]DPA-713 revealed a strong trend towards reduced TSPO binding in the middle frontal gyrus of patients with recent-onset schizophrenia, who were previously shown to display increased levels of inflammatory cytokines in peripheral and central tissues. Together, our findings challenge the common assumption that central low-grade inflammation in schizophrenia is mirrored by increased TSPO expression or ligand binding. Our study further underscores the need to interpret altered TSPO binding in schizophrenia with caution, especially when measures of TSPO are not complemented with other markers of inflammation. Unless more selective microglial markers are available for PET imaging, quantification of cytokines and other inflammatory biomarkers, along with their molecular signaling pathways, may be more accurate in attempts to characterize inflammatory profiles in schizophrenia and other mental disorders that lack robust reactive gliosis.
Collapse
Affiliation(s)
- T Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - J M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - T Gschwind
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - U Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Y Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - M Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW, Australia
- Discipline of Medical Radiation Sciences, The University of Sydney, Sydney, NSW, Australia
| | - A C Vernon
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - D Benke
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - M G Pomper
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - U Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
235
|
Fluid network dynamics in the prefrontal cortex during multiple strategy switching. Nat Commun 2018; 9:309. [PMID: 29358717 PMCID: PMC5778086 DOI: 10.1038/s41467-017-02764-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/26/2017] [Indexed: 01/08/2023] Open
Abstract
Coordinated shifts of neuronal activity in the prefrontal cortex are associated with strategy adaptations in behavioural tasks, when animals switch from following one rule to another. However, network dynamics related to multiple-rule changes are scarcely known. We show how firing rates of individual neurons in the prelimbic and cingulate cortex correlate with the performance of rats trained to change their navigation multiple times according to allocentric and egocentric strategies. The concerted population activity exhibits a stable firing during the performance of one rule but shifted to another neuronal firing state when a new rule is learnt. Interestingly, when the same rule is presented a second time within the same session, neuronal firing does not revert back to the original neuronal firing state, but a new activity-state is formed. Our data indicate that neuronal firing of prefrontal cortical neurons represents changes in strategy and task-performance rather than specific strategies or rules. Population activity of neurons in the prefrontal cortex has been shown to represent cognitive strategy and rules. Here the authors report that when the same rule is repeated on multiple occasions in the task, it is accompanied each time by a new prefrontal firing rate state.
Collapse
|
236
|
Dopamine Development in the Mouse Orbital Prefrontal Cortex Is Protracted and Sensitive to Amphetamine in Adolescence. eNeuro 2018; 5:eN-NWR-0372-17. [PMID: 29333488 PMCID: PMC5762649 DOI: 10.1523/eneuro.0372-17.2017] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 02/04/2023] Open
Abstract
The prefrontal cortex (PFC) is divided into subregions, including the medial and orbital prefrontal cortices. Dopamine connectivity in the medial PFC (mPFC) continues to be established throughout adolescence as the result of the continuous growth of axons that innervated the nucleus accumbens (NAcc) prior to adolescence. During this period, dopamine axons remain vulnerable to environmental influences, such as drugs used recreationally by humans. The developmental trajectory of the orbital prefrontal dopamine innervation remains almost completely unstudied. Nonetheless, the orbital PFC (oPFC) is critical for some of the most complex functions of the PFC and is disrupted by drugs of abuse, both in adolescent humans and rodents. Here, we use quantitative neuroanatomy, axon-initiated viral-vector recombination, and pharmacology in mice to determine the spatiotemporal development of the dopamine innervation to the oPFC and its vulnerability to amphetamine in adolescence. We find that dopamine innervation to the oPFC also continues to increase during adolescence and that this increase is due to the growth of new dopamine axons to this region. Furthermore, amphetamine in adolescence dramatically reduces the number of presynaptic sites on oPFC dopamine axons. In contrast, dopamine innervation to the piriform cortex is not protracted across adolescence and is not impacted by amphetamine exposure during adolescence, indicating that dopamine development during adolescence is a uniquely prefrontal phenomenon. This renders these fibers, and the PFC in general, particularly vulnerable to environmental risk factors during adolescence, such as recreational drug use.
Collapse
|
237
|
Virtanen MA, Lacoh CM, Fiumelli H, Kosel M, Tyagarajan S, de Roo M, Vutskits L. Development of inhibitory synaptic inputs on layer 2/3 pyramidal neurons in the rat medial prefrontal cortex. Brain Struct Funct 2018; 223:1999-2012. [PMID: 29322238 DOI: 10.1007/s00429-017-1602-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 12/28/2017] [Indexed: 02/03/2023]
Abstract
Inhibitory control of pyramidal neurons plays a major role in governing the excitability in the brain. While spatial mapping of inhibitory inputs onto pyramidal neurons would provide important structural data on neuronal signaling, studying their distribution at the single cell level is difficult due to the lack of easily identifiable anatomical proxies. Here, we describe an approach where in utero electroporation of a plasmid encoding for fluorescently tagged gephyrin into the precursors of pyramidal cells along with ionotophoretic injection of Lucifer Yellow can reliably and specifically detect GABAergic synapses on the dendritic arbour of single pyramidal neurons. Using this technique and focusing on the basal dendritic arbour of layer 2/3 pyramidal cells of the medial prefrontal cortex, we demonstrate an intense development of GABAergic inputs onto these cells between postnatal days 10 and 20. While the spatial distribution of gephyrin clusters was not affected by the distance from the cell body at postnatal day 10, we found that distal dendritic segments appeared to have a higher gephyrin density at later developmental stages. We also show a transient increase around postnatal day 20 in the percentage of spines that are carrying a gephyrin cluster, indicative of innervation by a GABAergic terminal. Since the precise spatial arrangement of synaptic inputs is an important determinant of neuronal responses, we believe that the method described in this work may allow a better understanding of how inhibition settles together with excitation, and serve as basics for further modelling studies focusing on the geometry of dendritic inhibition during development.
Collapse
Affiliation(s)
- Mari A Virtanen
- Department of Basic Neuroscience, University of Geneva Medical School, Geneva, Switzerland.,Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Claudia Marvine Lacoh
- Department of Basic Neuroscience, University of Geneva Medical School, Geneva, Switzerland
| | - Hubert Fiumelli
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Markus Kosel
- Department of Psychiatry, University of Geneva Medical School, Geneva, Switzerland
| | - Shiva Tyagarajan
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Mathias de Roo
- Department of Basic Neuroscience, University of Geneva Medical School, Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, University Hospitals of Geneva, 4, rue Gabrielle-Perret-Gentil, 1211, Geneva 4, Switzerland
| | - Laszlo Vutskits
- Department of Basic Neuroscience, University of Geneva Medical School, Geneva, Switzerland. .,Department of Anesthesiology, Pharmacology and Intensive Care, University Hospitals of Geneva, 4, rue Gabrielle-Perret-Gentil, 1211, Geneva 4, Switzerland.
| |
Collapse
|
238
|
Sparks DW, Tian MK, Sargin D, Venkatesan S, Intson K, Lambe EK. Opposing Cholinergic and Serotonergic Modulation of Layer 6 in Prefrontal Cortex. Front Neural Circuits 2018; 11:107. [PMID: 29354034 PMCID: PMC5758509 DOI: 10.3389/fncir.2017.00107] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/13/2017] [Indexed: 01/28/2023] Open
Abstract
Prefrontal cortex is a hub for attention processing and receives abundant innervation from cholinergic and serotonergic afferents. A growing body of evidence suggests that acetylcholine (ACh) and serotonin (5-HT) have opposing influences on tasks requiring attention, but the underlying neurophysiology of their opposition is unclear. One candidate target population is medial prefrontal layer 6 pyramidal neurons, which provide feedback modulation of the thalamus, as well as feed-forward excitation of cortical interneurons. Here, we assess the response of these neurons to ACh and 5-HT using whole cell recordings in acute brain slices from mouse cortex. With application of exogenous agonists, we show that individual layer 6 pyramidal neurons are bidirectionally-modulated, with ACh and 5-HT exerting opposite effects on excitability across a number of concentrations. Next, we tested the responses of layer 6 pyramidal neurons to optogenetic release of endogenous ACh or 5-HT. These experiments were performed in brain slices from transgenic mice expressing channelrhodopsin in either ChAT-expressing cholinergic neurons or Pet1-expressing serotonergic neurons. Light-evoked endogenous neuromodulation recapitulated the effects of exogenous neurotransmitters, showing opposing modulation of layer 6 pyramidal neurons by ACh and 5-HT. Lastly, the addition of 5-HT to either endogenous or exogenous ACh significantly suppressed the excitation of pyramidal neurons in prefrontal layer 6. Taken together, this work suggests that the major corticothalamic layer of prefrontal cortex is a substrate for opposing modulatory influences on neuronal activity that could have implications for regulation of attention.
Collapse
Affiliation(s)
- Daniel W Sparks
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Michael K Tian
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Derya Sargin
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Katheron Intson
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Evelyn K Lambe
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
239
|
Watanabe K, Funahashi S. Toward an understanding of the neural mechanisms underlying dual-task performance: Contribution of comparative approaches using animal models. Neurosci Biobehav Rev 2018; 84:12-28. [DOI: 10.1016/j.neubiorev.2017.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 08/09/2017] [Accepted: 08/11/2017] [Indexed: 10/19/2022]
|
240
|
Manning EE, Ahmari SE. How can preclinical mouse models be used to gain insight into prefrontal cortex dysfunction in obsessive-compulsive disorder? Brain Neurosci Adv 2018; 2:2398212818783896. [PMID: 32166143 PMCID: PMC7058260 DOI: 10.1177/2398212818783896] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/18/2018] [Indexed: 01/09/2023] Open
Abstract
Obsessive-compulsive disorder is a debilitating psychiatric disorder that is characterised by perseverative thoughts and behaviours. Cognitive and affective disturbances play a central role in this illness, and it is therefore not surprising that clinical neuroimaging studies have demonstrated widespread alterations in prefrontal cortex functioning in patients. Preclinical mouse experimental systems provide the opportunity to gain mechanistic insight into the neurobiological changes underlying prefrontal cortex dysfunction through new technologies that allow measurement and manipulation of activity in discrete neural populations in awake, behaving mice. However, recent preclinical research has focused on striatal dysfunction, and has therefore provided relatively little insight regarding the role of the prefrontal cortex in obsessive-compulsive disorder-relevant behaviours. Here, we will discuss a number of translational prefrontal cortex-dependent paradigms, including obsessive-compulsive disorder-relevant tasks that produce compulsive responding, and how they can be leveraged in this context. Drawing on recent examples that have led to mechanistic insight about specific genes, cell types and circuits that mediate prefrontal cortex contributions to distinct aspects of cognition, we will provide a framework for applying similar strategies to identify neural mechanisms underlying obsessive-compulsive disorder-relevant behavioural domains. We propose that research using clinically relevant paradigms will accelerate translation of findings from preclinical mouse models, thus supporting the development of novel therapeutics targeted to specific pathophysiological mechanisms.
Collapse
Affiliation(s)
| | - Susanne E. Ahmari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
241
|
Nurmasitoh T, Sari DCR, Partadiredja G. The effects of black garlic on the working memory and pyramidal cell number of medial prefrontal cortex of rats exposed to monosodium glutamate. Drug Chem Toxicol 2017; 41:324-329. [DOI: 10.1080/01480545.2017.1414833] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Titis Nurmasitoh
- Department of Physiology, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Physiology, Faculty of Medicine, Universitas Islam Indonesia, Yogyakarta, Indonesia
| | - Dwi Cahyani Ratna Sari
- Department of Anatomy, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ginus Partadiredja
- Department of Physiology, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
242
|
NLGN1 and NLGN2 in the prefrontal cortex: their role in memory consolidation and strengthening. Curr Opin Neurobiol 2017; 48:122-130. [PMID: 29278843 DOI: 10.1016/j.conb.2017.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/27/2017] [Accepted: 12/10/2017] [Indexed: 12/21/2022]
Abstract
The prefrontal cortex (PFC) is critical for memory formation, but the underlying molecular mechanisms are poorly understood. Clinical and animal model studies have shown that changes in PFC excitation and inhibition are important for cognitive functions as well as related disorders. Here, we discuss recent findings revealing the roles of the excitatory and inhibitory synaptic proteins neuroligin 1 (NLGN1) and NLGN2 in the PFC in memory formation and modulation of memory strength. We propose that shifts in NLGN1 and NLGN2 expression in specific excitatory and inhibitory neuronal subpopulations in response to experience regulate the dynamic processes of memory consolidation and strengthening. Because excitatory/inhibitory imbalances accompany neuropsychiatric disorders in which strength and flexibility of representations play important roles, understanding these mechanisms may suggest novel therapies.
Collapse
|
243
|
Baldo BA. Prefrontal Cortical Opioids and Dysregulated Motivation: A Network Hypothesis. Trends Neurosci 2017; 39:366-377. [PMID: 27233653 DOI: 10.1016/j.tins.2016.03.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 02/06/2023]
Abstract
Loss of inhibitory control over appetitively motivated behavior occurs in multiple psychiatric disorders, including drug abuse, behavioral addictions, and eating disorders with binge features. In this opinion article, novel actions of μ-opioid peptides in the prefrontal cortex (PFC) that could contribute to inhibitory control deficits will be discussed. Evidence has accrued to suggest that excessive intra-PFC μ-opioid receptor (μ-OR) signaling alters the PFC response to excitatory drive, resulting in supernormal and incoherent recruitment of multiple PFC output pathways. Affected pathways include functionally opposed PFC→hypothalamus 'appetitive driver' and PFC→striatum 'appetitive limiter' projections. This network perturbation engenders disorganized, impulsive appetitive responses. Evidence supporting this hypothesis from human imaging and animal studies will be discussed, and combinatorial drug treatments targeting μ-ORs and specific PFC subcortical targets will be explored.
Collapse
Affiliation(s)
- Brian A Baldo
- Department of Psychiatry, University of Wisconsin-Madison School of Medicine and Public Health, 6001 Research Park Blvd, Madison, WI 53719, USA.
| |
Collapse
|
244
|
Abdurasulova IN, Ekimova IV, Matsulevich AV, Gazizova AR, Klimenko VM, Pastukhov YF. Impairment of non-associative learning in a rat experimental model of preclinical stage of Parkinson's disease. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2017; 476:188-190. [PMID: 29101619 DOI: 10.1134/s0012496617050039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Indexed: 11/23/2022]
Abstract
An experimental model of the preclinical stage of Parkinson's disease was induced by double intranasal administration of the proteasome inhibitor lactacystin. The results demonstrated signs of cognitive impairments expressed as impaired non-associative learning. This was related to degeneration of one-third of dopaminergic neurons in the ventral tegmental area of the midbrain and their axons in the dorsolateral prefrontal cortex. Impairment of non-associative learning may be an early non-motor marker of Parkinson's disease indicating the start of neurodegenerative processes in the dopaminergic mesocortical system of the brain.
Collapse
Affiliation(s)
- I N Abdurasulova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia. .,Institute of Experimental Medicine, St. Petersburg, Russia.
| | - I V Ekimova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - A V Matsulevich
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.,Institute of Experimental Medicine, St. Petersburg, Russia
| | - A R Gazizova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - V M Klimenko
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.,Institute of Experimental Medicine, St. Petersburg, Russia
| | - Yu F Pastukhov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
245
|
|
246
|
Making Dopamine Connections in Adolescence. Trends Neurosci 2017; 40:709-719. [PMID: 29032842 DOI: 10.1016/j.tins.2017.09.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/07/2017] [Accepted: 09/14/2017] [Indexed: 12/13/2022]
Abstract
A dramatic maturational process ongoing in adolescence is prefrontal cortex development, including its dopamine innervation. Dopamine axons grow from the striatum to the prefrontal cortex, the only known case of long-distance axon growth during adolescence. This is coordinated by the Netrin-1 guidance cue receptor DCC (deleted in colorectal cancer), which in turn controls the intrinsic development of the prefrontal cortex itself. Stimulant drugs in adolescence alter DCC in dopamine neurons and, in turn prefrontal cortex maturation, impacting cognitive abilities. Variations in DCC expression are linked to psychiatric conditions of prefrontal cortex dysfunction, and microRNA regulation of DCC may be key to determining adolescent vulnerability or resilience. Since early interventions are proving to effectively ameliorate disease outcome, the Netrin-1 system is a promising therapeutic target.
Collapse
|
247
|
Cruz-Rizzolo RJ, Limieri LL, de Paiva IR, Ribeiro JOB, Pimenta TF, Pinato L, Ervolino E, Casatti CA, Guissoni Campos LM, Liberti EA. Protein malnutrition during gestation and early life decreases neuronal size in the medial prefrontal cortex of post-pubertal rats. IBRO Rep 2017; 3:65-71. [PMID: 30135943 PMCID: PMC6084879 DOI: 10.1016/j.ibror.2017.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 10/28/2022] Open
Abstract
Retrospective studies in human populations indicate that protein deprivation during pregnancy and early life (early protein malnutrition, EPM) is associated with cognitive impairments, learning disabilities and may represent a risk factor for the late onset of some psychiatric disorders, fundamentally schizophrenia, a condition where the prefrontal cortex plays an important role. The purpose of this study was to analyze whether EPM affects structural aspects of the rat medial prefrontal cortex (mPFC), such as cortical volume, neuronal density and neuronal soma size, which seem altered in patients with schizophrenia. For this, a rat model of EPM (5% casein from conception to postnatal day 60) was adopted and the rat mPFC volume, total number of neurons and average neuronal volume were evaluated on postnatal day 60 (post-pubertal animals) by histo- and immunohistochemical techniques using unbiased stereological analysis. EPM did not alter the number of NeuN+ neurons in the rat mPFC. However, a very significant decrease in mPFC volume and average neuronal size was observed in malnourished rats. Although the present study does not establish causal relationships between malnutrition and schizophrenia, our results may indicate a similar structural phenomenon in these two situations.
Collapse
Affiliation(s)
| | - Laís Leal Limieri
- Department of Basic Sciences, São Paulo State University, Araçatuba, SP, Brazil
| | | | | | | | - Luciana Pinato
- Department of Speech-Language and Hearing Therapy, São Paulo State University, Marilia, SP, Brazil
| | - Edilson Ervolino
- Department of Basic Sciences, São Paulo State University, Araçatuba, SP, Brazil
| | | | | | - Edson Aparecido Liberti
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| |
Collapse
|
248
|
Watt MJ, Weber MA, Davies SR, Forster GL. Impact of juvenile chronic stress on adult cortico-accumbal function: Implications for cognition and addiction. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79. [PMID: 28642080 PMCID: PMC5610933 DOI: 10.1016/j.pnpbp.2017.06.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Repeated exposure to stress during childhood is associated with increased risk for neuropsychiatric illness, substance use disorders and other behavioral problems in adulthood. However, it is not clear how chronic childhood stress can lead to emergence of such a wide range of symptoms and disorders in later life. One possible explanation lies in stress-induced disruption to the development of specific brain regions associated with executive function and reward processing, deficits in which are common to the disorders promoted by childhood stress. Evidence of aberrations in prefrontal cortex and nucleus accumbens function following repeated exposure of juvenile (pre- and adolescent) organisms to a variety of different stressors would account not only for the similarity in symptoms across the wide range of childhood stress-associated mental illnesses, but also their persistence into adulthood in the absence of further stress. Therefore, the goal of this review is to evaluate the current knowledge regarding disruption to executive function and reward processing in adult animals or humans exposed to chronic stress over the juvenile period, and the underlying neurobiology, with particular emphasis on the prefrontal cortex and nucleus accumbens. First, the role of these brain regions in mediating executive function and reward processing is highlighted. Second, the neurobehavioral development of these systems is discussed to illustrate how juvenile stress may exert long-lasting effects on prefrontal cortex-accumbal activity and related behavioral functions. Finally, a critical review of current animal and human findings is presented, which strongly supports the supposition that exposure to chronic stress (particularly social aggression and isolation in animal studies) in the juvenile period produces impairments in executive function in adulthood, especially in working memory and inhibitory control. Chronic juvenile stress also results in aberrations to reward processing and seeking, with increased sensitivity to drugs of abuse particularly noted in animal models, which is in line with greater incidence of substance use disorders seen in clinical studies. These consequences are potentially mediated by monoamine and glutamatergic dysfunction in the prefrontal cortex and nucleus accumbens, providing translatable therapeutic targets. However, the predominant use of male subjects and social-based stressors in preclinical studies points to a clear need for determining how both sex differences and stressor heterogeneity may differentially contribute to stress-induced changes to substrates mediating executive function and reward processing, before the impact of chronic juvenile stress in promoting adult psychopathology can be fully understood.
Collapse
|
249
|
Santana N, Artigas F. Laminar and Cellular Distribution of Monoamine Receptors in Rat Medial Prefrontal Cortex. Front Neuroanat 2017; 11:87. [PMID: 29033796 PMCID: PMC5625028 DOI: 10.3389/fnana.2017.00087] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/15/2017] [Indexed: 01/03/2023] Open
Abstract
The prefrontal cortex (PFC) is deeply involved in higher brain functions, many of which are altered in psychiatric conditions. The PFC exerts a top-down control of most cortical and subcortical areas through descending pathways and is densely innervated by axons emerging from the brainstem monoamine cell groups, namely, the dorsal and median raphe nuclei (DR and MnR, respectively), the ventral tegmental area and the locus coeruleus (LC). In turn, the activity of these cell groups is tightly controlled by afferent pathways arising from layer V PFC pyramidal neurons. The reciprocal connectivity between PFC and monoamine cell groups is of interest to study the pathophysiology and treatment of severe psychiatric disorders, such as major depression and schizophrenia, inasmuch as antidepressant and antipsychotic drugs target monoamine receptors/transporters expressed in these areas. Here we review previous reports examining the presence of monoamine receptors in pyramidal and GABAergic neurons of the PFC using double in situ hybridization. Additionally, we present new data on the quantitative layer distribution (layers I, II-III, V, and VI) of monoamine receptor-expressing cells in the cingulate (Cg), prelimbic (PrL) and infralimbic (IL) subfields of the medial PFC (mPFC). The receptors examined include serotonin 5-HT1A, 5-HT2A, 5-HT2C, and 5-HT3, dopamine D1 and D2 receptors, and α1A-, α1B-, and α1D-adrenoceptors. With the exception of 5-HT3 receptors, selectively expressed by layers I-III GABA interneurons, the rest of monoamine receptors are widely expressed by pyramidal and GABAergic neurons in intermediate and deep layers of mPFC (5-HT2C receptors are also expressed in layer I). This complex distribution suggests that monoamines may modulate the communications between PFC and cortical/subcortical areas through the activation of receptors expressed by neurons in intermediate (e.g., 5-HT1A, 5-HT2A, α1D-adrenoceptors, dopamine D1 receptors) and deep layers (e.g., 5-HT1A, 5-HT2A, α1A-adrenoceptors, dopamine D2 receptors), respectively. Overall, these data provide a detailed framework to better understand the role of monoamines in the processing of cognitive and emotional signals by the PFC. Likewise, they may be helpful to characterize brain circuits relevant for the therapeutic action of antidepressant and antipsychotic drugs and to improve their therapeutic action, overcoming the limitations of current drugs.
Collapse
Affiliation(s)
- Noemí Santana
- Systems Neuropharmacology, Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain
| | - Francesc Artigas
- Systems Neuropharmacology, Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
250
|
Gawlak M, Szulczyk B, Berłowski A, Grzelka K, Stachurska A, Pełka J, Czarzasta K, Małecki M, Kurowski P, Nurowska E, Szulczyk P. Age-dependent expression of Nav1.9 channels in medial prefrontal cortex pyramidal neurons in rats. Dev Neurobiol 2017; 77:1371-1384. [PMID: 28913981 DOI: 10.1002/dneu.22537] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 12/19/2022]
Abstract
Developmental changes that occur in the prefrontal cortex during adolescence alter behavior. These behavioral alterations likely stem from changes in prefrontal cortex neuronal activity, which may depend on the properties and expression of ion channels. Nav1.9 sodium channels conduct a Na+ current that is TTX resistant with a low threshold and noninactivating over time. The purpose of this study was to assess the presence of Nav1.9 channels in medial prefrontal cortex (mPFC) layer II and V pyramidal neurons in young (20-day old), late adolescent (60-day old), and adult (6- to 7-month old) rats. First, we demonstrated that layer II and V mPFC pyramidal neurons in slices obtained from young rats exhibited a TTX-resistant, low-threshold, noninactivating, and voltage-dependent Na+ current. The mRNA expression of the SCN11a gene (which encodes the Nav1.9 channel) in mPFC tissue was significantly higher in young rats than in late adolescent and adult rats. Nav1.9 protein was immunofluorescently labeled in mPFC cells in slices and analyzed via confocal microscopy. Nav1.9 immunolabeling was present in layer II and V mPFC pyramidal neurons and was more prominent in the neurons of young rats than in the neurons of late adolescent and adult rats. We conclude that Nav1.9 channels are expressed in layer II and V mPFC pyramidal neurons and that Nav1.9 protein expression in the mPFC pyramidal neurons of late adolescent and adult rats is lower than that in the neurons of young rats. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1371-1384, 2017.
Collapse
Affiliation(s)
- Maciej Gawlak
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| | - Bartłomiej Szulczyk
- Department of Drug Technology and Pharmaceutical Biotechnology, The Medical University of Warsaw, Warsaw, Poland
| | - Adam Berłowski
- Department of Physiology and Pathophysiology, The Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Grzelka
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| | - Anna Stachurska
- Department of Molecular Biology, The Medical University of Warsaw, Warsaw, Poland
| | - Justyna Pełka
- Department of Physiology and Pathophysiology, The Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Czarzasta
- Laboratory of Experimental and Clinical Physiology, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Małecki
- Department of Molecular Biology, The Medical University of Warsaw, Warsaw, Poland
| | - Przemysław Kurowski
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| | - Ewa Nurowska
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| | - Paweł Szulczyk
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|