1
|
Bihorac J, Salem Y, Lückemann L, Schedlowski M, Doenlen R, Engler H, Mark MD, Dombrowski K, Spoida K, Hadamitzky M. Investigations on the Ability of the Insular Cortex to Process Peripheral Immunosuppression. J Neuroimmune Pharmacol 2024; 19:40. [PMID: 39078442 PMCID: PMC11289148 DOI: 10.1007/s11481-024-10143-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/14/2024] [Indexed: 07/31/2024]
Abstract
The brain and immune system communicate through complex bidirectional pathways, but the specificity by which the brain perceives or even remembers alterations in immune homeostasis is still poorly understood. Recent data revealed that immune-related information under peripheral inflammatory conditions, termed as "immunengram", were represented in specific neuronal ensembles in the insular cortex (IC). Chemogenetic reactivation of these neuronal ensembles was sufficient to retrieve the inflammatory stages, indicating that the brain can store and retrieve specific immune responses. Against this background, the current approach was designed to investigate the ability of the IC to process states of immunosuppression pharmacologically induced by the mechanistic target of rapamycin (mTOR) inhibitor rapamycin. We here show that the IC perceives the initial state of immunosuppression, reflected by increased deep-brain electroencephalography (EEG) activity during acute immunosuppressive drug treatment. Following an experienced period of immunosuppression, though, diminished splenic cytokine production as formerly induced by rapamycin could not be reinstated by nonspecific chemogenetic activation or inhibition of the IC. These findings suggest that the information of a past, or experienced status of pharmacologically induced immunosuppression is not represented in the IC. Together, the present work extends the view of immune-to-brain communication during the states of peripheral immunosuppression and foster the prominent role of the IC for interoception.
Collapse
Affiliation(s)
- Julia Bihorac
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, 45147, Germany
| | - Yasmin Salem
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, 45147, Germany
| | - Laura Lückemann
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, 45147, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, 45147, Germany
- Department of Clinical Neuroscience, Osher Center for Integrative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Raphael Doenlen
- Center of Phenogenomics, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, 45147, Germany
| | - Melanie D Mark
- Behavioral Neuroscience, Faculty for Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Kirsten Dombrowski
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, 45147, Germany
| | - Katharina Spoida
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Martin Hadamitzky
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, 45147, Germany.
| |
Collapse
|
2
|
Keiser AA, Dong TN, Kramár EA, Butler CW, Chen S, Matheos DP, Rounds JS, Rodriguez A, Beardwood JH, Augustynski AS, Al-Shammari A, Alaghband Y, Alizo Vera V, Berchtold NC, Shanur S, Baldi P, Cotman CW, Wood MA. Specific exercise patterns generate an epigenetic molecular memory window that drives long-term memory formation and identifies ACVR1C as a bidirectional regulator of memory in mice. Nat Commun 2024; 15:3836. [PMID: 38714691 PMCID: PMC11076285 DOI: 10.1038/s41467-024-47996-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/15/2024] [Indexed: 05/10/2024] Open
Abstract
Exercise has beneficial effects on cognition throughout the lifespan. Here, we demonstrate that specific exercise patterns transform insufficient, subthreshold training into long-term memory in mice. Our findings reveal a potential molecular memory window such that subthreshold training within this window enables long-term memory formation. We performed RNA-seq on dorsal hippocampus and identify genes whose expression correlate with conditions in which exercise enables long-term memory formation. Among these genes we found Acvr1c, a member of the TGF ß family. We find that exercise, in any amount, alleviates epigenetic repression at the Acvr1c promoter during consolidation. Additionally, we find that ACVR1C can bidirectionally regulate synaptic plasticity and long-term memory in mice. Furthermore, Acvr1c expression is impaired in the aging human and mouse brain, as well as in the 5xFAD mouse model, and over-expression of Acvr1c enables learning and facilitates plasticity in mice. These data suggest that promoting ACVR1C may protect against cognitive impairment.
Collapse
Affiliation(s)
- Ashley A Keiser
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Tri N Dong
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Enikö A Kramár
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Christopher W Butler
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
- Department of Neurology, University of California Irvine, Irvine, CA, 92697, USA
| | - Siwei Chen
- Institute for Genomics and Bioinformatics, School of Information and Computer Science, University of California, Irvine, Irvine, CA, 92697, USA
| | - Dina P Matheos
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Jacob S Rounds
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Alyssa Rodriguez
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Joy H Beardwood
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Agatha S Augustynski
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Ameer Al-Shammari
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Yasaman Alaghband
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Vanessa Alizo Vera
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Nicole C Berchtold
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
- Department of Neurology, University of California Irvine, Irvine, CA, 92697, USA
| | - Sharmin Shanur
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, School of Information and Computer Science, University of California, Irvine, Irvine, CA, 92697, USA
| | - Carl W Cotman
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA
- Department of Neurology, University of California Irvine, Irvine, CA, 92697, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA.
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, CA, 92697, USA.
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
3
|
Liu D, Hu SW, Wang D, Zhang Q, Zhang X, Ding HL, Cao JL. An Ascending Excitatory Circuit from the Dorsal Raphe for Sensory Modulation of Pain. J Neurosci 2024; 44:e0869232023. [PMID: 38124016 PMCID: PMC10860493 DOI: 10.1523/jneurosci.0869-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
The dorsal raphe nucleus (DRN) is an important nucleus in pain regulation. However, the underlying neural pathway and the function of specific cell types remain unclear. Here, we report a previously unrecognized ascending facilitation pathway, the DRN to the mesoaccumbal dopamine (DA) circuit, for regulating pain. Chronic pain increased the activity of DRN glutamatergic, but not serotonergic, neurons projecting to the ventral tegmental area (VTA) (DRNGlu-VTA) in male mice. The optogenetic activation of DRNGlu-VTA circuit induced a pain-like response in naive male mice, and its inhibition produced an analgesic effect in male mice with neuropathic pain. Furthermore, we discovered that DRN ascending pathway regulated pain through strengthened excitatory transmission onto the VTA DA neurons projecting to the ventral part of nucleus accumbens medial shell (vNAcMed), thereby activated the mesoaccumbal DA neurons. Correspondingly, optogenetic manipulation of this three-node pathway bilaterally regulated pain behaviors. These findings identified a DRN ascending excitatory pathway that is crucial for pain sensory processing, which can potentially be exploited toward targeting pain disorders.
Collapse
Affiliation(s)
- Di Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Su-Wan Hu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Di Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Qi Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiao Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
4
|
Munari L, Patel V, Johnson N, Mariottini C, Prabha S, Blitzer RD, Iyengar R. Memory discrimination is promoted by the expression of the transcription repressor WT1 in the dentate gyrus. Front Behav Neurosci 2023; 17:1130840. [PMID: 37830039 PMCID: PMC10564998 DOI: 10.3389/fnbeh.2023.1130840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 08/14/2023] [Indexed: 10/14/2023] Open
Abstract
The hippocampus is critical for the precise formation of contextual memories. Overlapping inputs coming from the entorhinal cortex are processed by the trisynaptic pathway to form distinct memories. Disruption in any step of the circuit flow can lead to a lack of memory precision, and to memory interference. We have identified the transcriptional repressor Wilm's Tumor 1 (WT1) as an important regulator of synaptic plasticity involved in memory discrimination in the hippocampus. In male mice, using viral and transgenic approaches, we showed that WT1 deletion in granule cells of the dentate gyrus (DG) disrupts memory discrimination. With electrophysiological methods, we then identified changes in granule cells' excitability and DG synaptic transmission indicating that WT1 knockdown in DG granule cells disrupts the inhibitory feedforward input from mossy fibers to CA3 by decreasing mIPSCs and shifting the normal excitatory/inhibitory (E/I) balance in the DG → CA3 circuit in favor of excitation. Finally, using a chemogenetic approach, we established a causal link between granule cell hyperexcitability and memory discrimination impairments. Our results suggest that WT1 enables a circuit-level computation that drives pattern discrimination behavior.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ravi Iyengar
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
5
|
Chen YT, Arano R, Guo J, Saleem U, Li Y, Xu W. Inhibitory hippocampus-medial septum projection controls locomotion and exploratory behavior. Front Synaptic Neurosci 2023; 15:1042858. [PMID: 37091878 PMCID: PMC10116069 DOI: 10.3389/fnsyn.2023.1042858] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Although the hippocampus is generally considered a cognitive center for spatial representation, learning, and memory, increasing evidence supports its roles in regulating locomotion. However, the neuronal mechanisms of the hippocampal regulation of locomotion and exploratory behavior remain unclear. In this study, we found that the inhibitory hippocampal synaptic projection to the medial septum (MS) bi-directionally controls the locomotor speed of mice. The activation of the MS-projecting interneurons in the hippocampus or the activation of the hippocampus-originated inhibitory synaptic terminals in the MS decreased locomotion and exploratory behavior. On the other hand, the inhibition of the hippocampus-originated inhibitory synaptic terminals in the MS increased locomotion. Unlike the septal projecting interneurons, the activation of the hippocampal interneurons projecting to the retrosplenial cortex did not change animal locomotion. Therefore, this study reveals a specific long-range inhibitory synaptic output from the hippocampus to the medial septum in the regulation of animal locomotion.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Xu
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
6
|
Goyal D, Kumar H. In Vivo and 3D Imaging Technique(s) for Spatiotemporal Mapping of Pathological Events in Experimental Model(s) of Spinal Cord Injury. ACS Chem Neurosci 2023; 14:809-819. [PMID: 36787542 DOI: 10.1021/acschemneuro.2c00643] [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: 02/16/2023] Open
Abstract
Endothelial damage, astrogliosis, microgliosis, and neuronal degeneration are the most common events after spinal cord injury (SCI). Studies highlighted that studying the spatiotemporal profile of these events might provide a deeper understanding of the pathophysiology of SCI. For imaging of these events, available conventional techniques such as 2-dimensional histology and immunohistochemistry (IHC) are well established and frequently used to visualize and detect the altered expression of the protein of interest involved in these events. However, the technique requires the physical sectioning of the tissue, and results are also open to misinterpretation. Currently, researchers are focusing more attention toward the advanced tools for imaging the spinal cord's various physiological and pathological parameters. The tools include two-photon imaging, light sheet fluorescence microscopy, in vivo imaging system with fluorescent probes, and in vivo chemical and fluorescent protein-expressing viral-tracers. These techniques outperform the limitations associated with conventional techniques in various aspects, such as optical sectioning of tissue, 3D reconstructed imaging, and imaging of particular planes of interest. In addition to this, these techniques are minimally invasive and less time-consuming. In this review, we will discuss the various advanced imaging methodologies that will evolve in the future to explore the fundamental mechanisms after SCI.
Collapse
Affiliation(s)
- Divya Goyal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat India, 382355
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat India, 382355
| |
Collapse
|
7
|
Cinalli DA, Cohen SJ, Calubag M, Oz G, Zhou L, Stackman RW. DREADD-inactivation of dorsal CA1 pyramidal neurons in mice impairs retrieval of object and spatial memories. Hippocampus 2023; 33:6-17. [PMID: 36468186 DOI: 10.1002/hipo.23484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/02/2022] [Accepted: 11/19/2022] [Indexed: 12/12/2022]
Abstract
The hippocampus, a medial temporal lobe brain region, is critical for the consolidation of information from short-term memory into long-term episodic memory and for spatial memory that enables navigation. Hippocampal damage in humans has been linked to amnesia and memory loss, characteristic of Alzheimer's disease and other dementias. Numerous studies indicate that the rodent hippocampus contributes significantly to long-term memory for spatial and nonspatial information. For example, muscimol-induced depression of CA1 neuronal activity in the dorsal hippocampus impairs the encoding, consolidation, and retrieval of nonspatial object memory in mice. Here, a chemogenetic designer receptor exclusively activated by designer drugs (DREADDs) approach was used to test the selective involvement of CA1 pyramidal neurons in memory retrieval for objects and for spatial location in a cohort of male C57BL/6J mice. Activation of the inhibitory (hM4Di) DREADDs receptor expressed in CA1 neurons significantly impaired the retrieval of object memory in the spontaneous object recognition task and of spatial memory in the Morris water maze. Silencing of CA1 neuronal activity in hM4Di-expressing mice was confirmed by comparing Fos expression in vehicle- and clozapine-N-oxide-treated mice after exploration of a novel environment. Histological analyses revealed that expression of the hM4Di receptor was limited to CA1 neurons of the dorsal hippocampus. These results suggest that a common subset of CA1 neurons (i.e., those expressing hM4Di receptors) in mouse hippocampus contributed to the retrieval of long-term memory for nonspatial and spatial information. Our findings support the view that the contribution of the rodent hippocampus is like that of the primate hippocampus, specifically essential for global memory. Our results further validate mice as a suitable model system to study the neurobiological mechanisms of human episodic memory, but also in developing treatments and understanding the underlying causes of diseases affecting long-term memory, such as Alzheimer's disease.
Collapse
Affiliation(s)
- David A Cinalli
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, USA
| | - Sarah J Cohen
- Jupiter Life Science Initiative, John D. MacArthur Campus, Florida Atlantic University, Jupiter, Florida, USA
| | - Mariah Calubag
- Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida, USA
| | - Goksu Oz
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, USA.,Florida Atlantic University and Max Planck Florida Institute Joint Integrative Biology - Neuroscience Ph.D. Program, Florida Atlantic University, Jupiter, Florida, USA.,International Max Planck Research School for Synapses and Circuits, Florida Atlantic University and Max Planck Florida Institute for Neuroscience, Jupiter, Florida, USA
| | - Lylybell Zhou
- Alexander W. Dreyfoos High School of the Arts, West Palm Beach, Florida, USA
| | - Robert W Stackman
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, USA.,Jupiter Life Science Initiative, John D. MacArthur Campus, Florida Atlantic University, Jupiter, Florida, USA.,Florida Atlantic University and Max Planck Florida Institute Joint Integrative Biology - Neuroscience Ph.D. Program, Florida Atlantic University, Jupiter, Florida, USA.,International Max Planck Research School for Synapses and Circuits, Florida Atlantic University and Max Planck Florida Institute for Neuroscience, Jupiter, Florida, USA
| |
Collapse
|
8
|
Nebeling FC, Poll S, Justus LC, Steffen J, Keppler K, Mittag M, Fuhrmann M. Microglial motility is modulated by neuronal activity and correlates with dendritic spine plasticity in the hippocampus of awake mice. eLife 2023; 12:83176. [PMID: 36749020 PMCID: PMC9946443 DOI: 10.7554/elife.83176] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Microglia, the resident immune cells of the brain, play a complex role in health and disease. They actively survey the brain parenchyma by physically interacting with other cells and structurally shaping the brain. Yet, the mechanisms underlying microglial motility and significance for synapse stability, especially in the hippocampus during adulthood, remain widely unresolved. Here, we investigated the effect of neuronal activity on microglial motility and the implications for the formation and survival of dendritic spines on hippocampal CA1 neurons in vivo. We used repetitive two-photon in vivo imaging in the hippocampus of awake and anesthetized mice to simultaneously study the motility of microglia and their interaction with dendritic spines. We found that CA3 to CA1 input is sufficient to modulate microglial process motility. Simultaneously, more dendritic spines emerged in mice after awake compared to anesthetized imaging. Interestingly, the rate of microglial contacts with individual dendritic spines and dendrites was associated with the stability, removal, and emergence of dendritic spines. These results suggest that microglia might sense neuronal activity via neurotransmitter release and actively participate in synaptic rewiring of the hippocampal neural network during adulthood. Further, this study has profound relevance for hippocampal learning and memory processes.
Collapse
Affiliation(s)
| | - Stefanie Poll
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| | - Lena Christine Justus
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| | - Julia Steffen
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| | - Kevin Keppler
- Light Microscopy Facility, German Center for Neurodegenerative DiseasesBonnGermany
| | - Manuel Mittag
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| | - Martin Fuhrmann
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| |
Collapse
|
9
|
Su H, Nakauchi S, Sumikawa K. Nicotine-mediated activation of α2 nAChR-expressing OLM cells in developing mouse brains disrupts OLM cell-mediated control of LTP in adolescence. Neurobiol Learn Mem 2022; 194:107674. [PMID: 36029955 PMCID: PMC9835838 DOI: 10.1016/j.nlm.2022.107674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/08/2022] [Accepted: 08/19/2022] [Indexed: 01/14/2023]
Abstract
Early postnatal nicotine exposure, a rodent model of smoking during pregnancy, affects hippocampal synaptic plasticity and memory. Here, we investigated the role of α2 nAChR-expressing OLM (α2-OLM) cells in LTP in unexposed and postnatal nicotine-exposed mice. We found that reduced α2 nAChR-dependent activation of OLM cells in α2 heterozygous knockout mice prevented LTP, whereas enhanced α2 nAChR-dependent activation of OLM cells in heterozygous knockin mice expressing hypersensitive α2 nAChRs facilitated LTP. Both optogenetic and chemogenetic activation of α2-OLM cells facilitated LTP as nicotine did. However, in postnatal nicotine-exposed mice, expressing chemogenetic hM3Dq receptors in α2-OLM cells, LTP was facilitated and both nicotinic and chemogenetic activation of α2-OLM cells prevented rather than facilitated LTP. These results demonstrate a critical role of α2-OLM cell activation in LTP as well as altered α2-OLM cell function in postnatal nicotine-exposed mice. To determine whether nicotine-mediated α2 nAChR activation in developing brains causes facilitated LTP and altered nicotinic modulation of LTP in adolescence, we used homozygous knockin mice expressing hypersensitive α2 nAChRs as a way to selectively activate α2-OLM cells. In the knockin mice, postnatal exposure to a low dose of nicotine, which had no effect on LTP in wild-type mice, is sufficient to cause facilitated LTP and altered nicotinic modulation of LTP as found in wild-type mice exposed to a higher dose of nicotine. Thus, the nicotine-mediated activation of α2 nAChRs on OLM cells in developing brains disrupts the α2-OLM cell-mediated control of LTP in adolescence that might be linked to impaired memory.
Collapse
Affiliation(s)
- Hailing Su
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697-4550, USA
| | - Sakura Nakauchi
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697-4550, USA
| | - Katumi Sumikawa
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697-4550, USA.
| |
Collapse
|
10
|
Chao OY, Nikolaus S, Yang YM, Huston JP. Neuronal circuitry for recognition memory of object and place in rodent models. Neurosci Biobehav Rev 2022; 141:104855. [PMID: 36089106 PMCID: PMC10542956 DOI: 10.1016/j.neubiorev.2022.104855] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022]
Abstract
Rats and mice are used for studying neuronal circuits underlying recognition memory due to their ability to spontaneously remember the occurrence of an object, its place and an association of the object and place in a particular environment. A joint employment of lesions, pharmacological interventions, optogenetics and chemogenetics is constantly expanding our knowledge of the neural basis for recognition memory of object, place, and their association. In this review, we summarize current studies on recognition memory in rodents with a focus on the novel object preference, novel location preference and object-in-place paradigms. The evidence suggests that the medial prefrontal cortex- and hippocampus-connected circuits contribute to recognition memory for object and place. Under certain conditions, the striatum, medial septum, amygdala, locus coeruleus and cerebellum are also involved. We propose that the neuronal circuitry for recognition memory of object and place is hierarchically connected and constructed by different cortical (perirhinal, entorhinal and retrosplenial cortices), thalamic (nucleus reuniens, mediodorsal and anterior thalamic nuclei) and primeval (hypothalamus and interpeduncular nucleus) modules interacting with the medial prefrontal cortex and hippocampus.
Collapse
Affiliation(s)
- Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Susanne Nikolaus
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University, 40225 Düsseldorf, Germany.
| |
Collapse
|
11
|
Dong TN, Kramár EA, Beardwood JH, Al-Shammari A, Wood MA, Keiser AA. Temporal endurance of exercise-induced benefits on hippocampus-dependent memory and synaptic plasticity in female mice. Neurobiol Learn Mem 2022; 194:107658. [PMID: 35811066 PMCID: PMC9901197 DOI: 10.1016/j.nlm.2022.107658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/20/2022] [Accepted: 07/04/2022] [Indexed: 02/08/2023]
Abstract
Exercise facilitates hippocampal neurogenesis and neuroplasticity that in turn, promotes cognitive function. Our previous studies have demonstrated that in male mice, voluntary exercise enables hippocampus-dependent learning in conditions that are normally subthreshold for long-term memory formation in sedentary animals. Such cognitive enhancement can be maintained long after exercise has ceased and can be re-engaged by a subsequent subthreshold exercise session, suggesting exercise-induced benefits are temporally dynamic. In females, the extent to which the benefits of exercise can be maintained and the mechanisms underlying this maintenance have yet to be defined. Here, we examined the exercise parameters required to initiate and maintain the benefits of exercise in female C57BL/6J mice. Using a subthreshold version of the hippocampus-dependent task called object-location memory (OLM) task, we show that 14d of voluntary exercise enables learning under subthreshold acquisition conditions in female mice. Following the initial exercise, a 7d sedentary delay results in diminished performance, which can be re-facilitated when animals receive 2d of reactivating exercise following the sedentary delay. Assessment of estrous cycle reveals enhanced wheel running activity during the estrus phase relative to the diestrus phase, whereas estrous phase on training or test had no effect on OLM performance. Utilizing the same exercise parameters, we demonstrate that 14d of exercise enhances long-term potentiation (LTP) in the CA1 region of the hippocampus, an effect that persists throughout the sedentary delay and following the reactivating exercise session. Previous studies have proposed exercise-induced BDNF upregulation as the mechanism underlying exercise-mediated benefits on synaptic plasticity and cognition. However, our assessment of hippocampal Bdnf mRNA expression following memory retrieval reveals no difference between exercise conditions and control, suggesting that persistent Bdnf upregulation may not be required for maintenance of exercise-induced benefits. Together, our data indicate that 14d of voluntary exercise can initiate long-lasting benefits on neuroplasticity and cognitive function in female mice, establishing the first evidence on the temporal endurance of exercise-induced benefits in females.
Collapse
Affiliation(s)
- T N Dong
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - E A Kramár
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - J H Beardwood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - A Al-Shammari
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - M A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - A A Keiser
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States.
| |
Collapse
|
12
|
Eriksson H, Rössler OG, Thiel G. Tyrosine hydroxylase gene promoter activity is upregulated in female catecholaminergic neuroblastoma cells following activation of a Gαq-coupled designer receptor. Neurochem Int 2022; 160:105407. [PMID: 35995267 DOI: 10.1016/j.neuint.2022.105407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 10/31/2022]
Abstract
Tyrosine hydroxylase is the rate-limiting enzyme of catecholamine biosynthesis that catalyzes the conversion of L-tyrosine to L-3,4-dihydroxyphenylalanine. The tyrosine hydroxylase gene is regulated by extracellular signaling molecules such as epidermal growth factor, nerve growth factor and steroids. Here, we investigated whether the activity of the tyrosine hydroxylase gene promoter is upregulated by activation of G protein-coupled receptors, the largest group of plasma membrane receptors. We used catecholaminergic neuroblastoma cells as a cellular model and chromatin-integrated tyrosine hydroxylase promoter-luciferase reporter genes. The results show that stimulation of Rαq, a Gαq-coupled designer receptor, triggered transcription of a reporter gene driven by the tyrosine hydroxylase promoter. Transcription was attenuated by overexpression of regulator of G-protein signaling-2, which activates the GTPase activity of the G protein α-subunit, and by a truncated, dominant-negative mutant of phospholipase Cβ3. Extracellular signal-regulated protein kinase was identified as the signal transducer. At the transcriptional level, tyrosine hydroxylase promoter activity was found to be controlled by the transcription factor CREB. Expression experiments with the adenoviral regulator protein E1A, an inhibitor of CBP/p300 histone acetyltransferases, showed that transcription of the reporter gene controlled by the tyrosine hydroxylase is under epigenetic control. We identified the protein phosphatases MAP kinase phosphatase-1 and calcineurin as part of a shutdown device of the signaling cascade linking Rαq designer receptor activation to tyrosine hydroxylase gene transcription. We conclude that tyrosine hydroxylase promoter activity is controlled by Gαq-coupled receptors.
Collapse
Affiliation(s)
- Helen Eriksson
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421, Homburg, Germany
| | - Oliver G Rössler
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421, Homburg, Germany
| | - Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421, Homburg, Germany.
| |
Collapse
|
13
|
Liu Y, Deng SL, Li LX, Zhou ZX, Lv Q, Wang ZY, Wang F, Chen JG. A circuit from dorsal hippocampal CA3 to parvafox nucleus mediates chronic social defeat stress-induced deficits in preference for social novelty. SCIENCE ADVANCES 2022; 8:eabe8828. [PMID: 35196094 PMCID: PMC8865774 DOI: 10.1126/sciadv.abe8828] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
The preference for social novelty is crucial to the social life of humans and rodents. However, the neural mechanisms underlying social novelty preference are poorly understood. Here, we found that chronic social defeat stress (CSDS) reduced the preference for social novelty in mice by impairing the response of CaMKIIα+ neurons in the CA3 region of dorsal hippocampus (dCA3) during approach to an unfamiliar mouse. The deficits of social novelty preference in CSDS-treated mice were reversed by activating the output from dCA3 to the GABAergic neurons in the lateral septum (LS). The activation of GABAergic projection from LS recruited a circuit that inhibited the Foxb1+ neurons in the parvafox nucleus (PFN), which drove social avoidance by projecting to the lateral periaqueductal gray (lPAG). These results suggest that a previously unidentified circuit of dCA3CaMKIIα+→LSGABA+→PFNFoxb1+→lPAG mediates the deficits of social novelty preference induced by CSDS.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Si-Long Deng
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang-Xia Li
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zi-Xiang Zhou
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiu Lv
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhong-Yuan Wang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fang Wang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Jian-Guo Chen
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| |
Collapse
|
14
|
Kawai M, Imaizumi K, Ishikawa M, Shibata S, Shinozaki M, Shibata T, Hashimoto S, Kitagawa T, Ago K, Kajikawa K, Shibata R, Kamata Y, Ushiba J, Koga K, Furue H, Matsumoto M, Nakamura M, Nagoshi N, Okano H. Long-term selective stimulation of transplanted neural stem/progenitor cells for spinal cord injury improves locomotor function. Cell Rep 2021; 37:110019. [PMID: 34818559 DOI: 10.1016/j.celrep.2021.110019] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
In cell transplantation therapy for spinal cord injury (SCI), grafted human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs) mainly differentiate into neurons, forming synapses in a process similar to neurodevelopment. In the developing nervous system, the activity of immature neurons has an important role in constructing and maintaining new synapses. Thus, we investigate how enhancing the activity of transplanted hiPSC-NS/PCs affects both the transplanted cells themselves and the host tissue. We find that chemogenetic stimulation of hiPSC-derived neural cells enhances cell activity and neuron-to-neuron interactions in vitro. In a rodent model of SCI, consecutive and selective chemogenetic stimulation of transplanted hiPSC-NS/PCs also enhances the expression of synapse-related genes and proteins in surrounding host tissues and prevents atrophy of the injured spinal cord, thereby improving locomotor function. These findings provide a strategy for enhancing activity within the graft to improve the efficacy of cell transplantation therapy for SCI.
Collapse
Affiliation(s)
- Momotaro Kawai
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Kent Imaizumi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Shinsuke Shibata
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Munehisa Shinozaki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Takahiro Shibata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Shogo Hashimoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Takahiro Kitagawa
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Kentaro Ago
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Keita Kajikawa
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Reo Shibata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Yasuhiro Kamata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Junichi Ushiba
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku, Yokohama 223-8522, Japan
| | - Keisuke Koga
- Department of Neurophysiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Hidemasa Furue
- Department of Neurophysiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| |
Collapse
|
15
|
Bubb EJ, Aggleton JP, O’Mara SM, Nelson AJD. Chemogenetics Reveal an Anterior Cingulate-Thalamic Pathway for Attending to Task-Relevant Information. Cereb Cortex 2021; 31:2169-2186. [PMID: 33251536 PMCID: PMC7945017 DOI: 10.1093/cercor/bhaa353] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/28/2022] Open
Abstract
In a changing environment, organisms need to decide when to select items that resemble previously rewarded stimuli and when it is best to switch to other stimulus types. Here, we used chemogenetic techniques to provide causal evidence that activity in the rodent anterior cingulate cortex and its efferents to the anterior thalamic nuclei modulate the ability to attend to reliable predictors of important outcomes. Rats completed an attentional set-shifting paradigm that first measures the ability to master serial discriminations involving a constant stimulus dimension that reliably predicts reinforcement (intradimensional-shift), followed by the ability to shift attention to a previously irrelevant class of stimuli when reinforcement contingencies change (extradimensional-shift). Chemogenetic disruption of the anterior cingulate cortex (Experiment 1) as well as selective disruption of anterior cingulate efferents to the anterior thalamic nuclei (Experiment 2) impaired intradimensional learning but facilitated 2 sets of extradimensional-shifts. This pattern of results signals the loss of a corticothalamic system for cognitive control that preferentially processes stimuli resembling those previously associated with reward. Previous studies highlight a separate medial prefrontal system that promotes the converse pattern, that is, switching to hitherto inconsistent predictors of reward when contingencies change. Competition between these 2 systems regulates cognitive flexibility and choice.
Collapse
Affiliation(s)
- Emma J Bubb
- School of Psychology, Cardiff University, Wales CF10 3AT, UK
| | - John P Aggleton
- School of Psychology, Cardiff University, Wales CF10 3AT, UK
| | - Shane M O’Mara
- Institute of Neuroscience, Trinity College Dublin D02 PN40, Ireland
| | | |
Collapse
|
16
|
Keiser AA, Kramár EA, Dong T, Shanur S, Pirodan M, Ru N, Acharya MM, Baulch JE, Limoli CL, Wood MA. Systemic HDAC3 inhibition ameliorates impairments in synaptic plasticity caused by simulated galactic cosmic radiation exposure in male mice. Neurobiol Learn Mem 2021; 178:107367. [PMID: 33359392 PMCID: PMC8456980 DOI: 10.1016/j.nlm.2020.107367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 12/18/2022]
Abstract
Deep space travel presents a number of measurable risks including exposure to a spectrum of radiations of varying qualities, termed galactic cosmic radiation (GCR) that are capable of penetrating the spacecraft, traversing through the body and impacting brain function. Using rodents, studies have reported that exposure to simulated GCR leads to cognitive impairments associated with changes in hippocampus function that can persist as long as one-year post exposure with no sign of recovery. Whether memory can be updated to incorporate new information in mice exposed to GCR is unknown. Further, mechanisms underlying long lasting impairments in cognitive function as a result of GCR exposure have yet to be defined. Here, we examined whether whole body exposure to simulated GCR using 6 ions and doses of 5 or 30 cGy interfered with the ability to update an existing memory or impact hippocampal synaptic plasticity, a cellular mechanism believed to underlie memory processes, by examining long term potentiation (LTP) in acute hippocampal slices from middle aged male mice 3.5-5 months after radiation exposure. Using a modified version of the hippocampus-dependent object location memory task developed by our lab termed "Objects in Updated Locations" (OUL) task we find that GCR exposure impaired hippocampus-dependent memory updating and hippocampal LTP 3.5-5 months after exposure. Further, we find that impairments in LTP are reversed through one-time systemic subcutaneous injection of the histone deacetylase 3 inhibitor RGFP 966 (10 mg/kg), suggesting that long lasting impairments in cognitive function may be mediated at least in part, through epigenetic mechanisms.
Collapse
Affiliation(s)
- A A Keiser
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - E A Kramár
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - T Dong
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - S Shanur
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - M Pirodan
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - N Ru
- Department of Radiation Oncology, University of California, Irvine 92697-2695, United States
| | - M M Acharya
- Department of Radiation Oncology, University of California, Irvine 92697-2695, United States
| | - J E Baulch
- Department of Radiation Oncology, University of California, Irvine 92697-2695, United States
| | - C L Limoli
- Department of Radiation Oncology, University of California, Irvine 92697-2695, United States.
| | - M A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States.
| |
Collapse
|
17
|
Kahn JB, Port RG, Anderson SA, Coulter DA. Modular, Circuit-Based Interventions Rescue Hippocampal-Dependent Social and Spatial Memory in a 22q11.2 Deletion Syndrome Mouse Model. Biol Psychiatry 2020; 88:710-718. [PMID: 32682567 PMCID: PMC7554065 DOI: 10.1016/j.biopsych.2020.04.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/09/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND 22q11.2 deletion syndrome (22qDS) manifests with myriad symptoms, including multiple neuropsychiatric disorders. Complications associated with the polygenic haploinsufficiency make 22qDS symptoms particularly difficult to manage with traditional therapeutic approaches. However, the varying mechanistic consequences often culminate to generate inappropriate regulation of neuronal circuit activity. We explored whether managing this aberrant activity in adults could be a therapeutically beneficial strategy. METHODS To assess and dissect hippocampal circuit function, we performed functional imaging in acute slices and targeted eloquent circuits (specific subcircuits tied to specific behavioral tasks) to provide relevant behavioral outputs. For example, the ventral and dorsal CA1 regions critically support social and spatial discrimination, respectively. We focally introduced chemogenetic constructs in 34 control and 24 22qDS model mice via adeno-associated viral vectors, driven by excitatory neuron-specific promoter elements, to manipulate circuit recruitment in an on-demand fashion. RESULTS 22qDS model mice exhibited CA1 excitatory ensemble hyperexcitability and concomitant behavioral deficits in both social and spatial memory. Remarkably, acute chemogenetic inhibition of pyramidal cells successfully corrected memory deficits and did so in a regionally specific manner: ventrally targeted constructs rescued only social behavior, while those expressed dorsally selectively affected spatial memory. Additionally, manipulating activity in control mice could recapitulate the memory deficits in a regionally specific manner. CONCLUSIONS These data suggest that retuning activity dysregulation can rescue function in disease-altered circuits, even in the face of a polygenetic haploinsufficiency with a strong developmental component. Targeting circuit excitability in a focal, modular manner may prove to be an effective therapeutic for treatment-resistant symptoms of mental illness.
Collapse
Affiliation(s)
- Julia B. Kahn
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Russell G. Port
- Departments of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Stewart A. Anderson
- Departments of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Douglas A. Coulter
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Departments of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| |
Collapse
|
18
|
Krueger JN, Wilmot JH, Teratani-Ota Y, Puhger KR, Nemes SE, Crestani AP, Lafreniere MM, Wiltgen BJ. Amnesia for context fear is caused by widespread disruption of hippocampal activity. Neurobiol Learn Mem 2020; 175:107295. [PMID: 32822864 PMCID: PMC8562570 DOI: 10.1016/j.nlm.2020.107295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/26/2022]
Abstract
The hippocampus plays an essential role in the formation and retrieval of episodic memories in humans and contextual memories in animals. However, amnesia is not always observed when this structure is compromised. To determine why this is the case, we compared the effects of several different circuit manipulations on memory retrieval and hippocampal activity. Mice were first trained on context fear conditioning and then optogenetic and chemogenetic tools were used to alter activity during memory retrieval. We found that retrieval was only impaired when manipulations caused widespread changes (increases or decreases) in hippocampal activity. Widespread increases occurred when pyramidal cells were excited and widespread decreases were found when GABAergic neurons were stimulated. Direct hyperpolarization of excitatory neurons only moderately reduced activity and did not produce amnesia. Surprisingly, widespread decreases in hippocampal activity did not prevent retrieval if they occurred gradually prior to testing. This suggests that intact brain regions can express contextual memories if they are given adequate time to compensate for the loss of the hippocampus.
Collapse
Affiliation(s)
- Jamie N Krueger
- Center for Neuroscience, University of California Davis, 1544 Newton Ct., Davis, CA 95618, United States.
| | - Jacob H Wilmot
- Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| | - Yusuke Teratani-Ota
- Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| | - Kyle R Puhger
- Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| | - Sonya E Nemes
- Center for Neuroscience, University of California Davis, 1544 Newton Ct., Davis, CA 95618, United States.
| | - Ana P Crestani
- Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| | - Marrisa M Lafreniere
- Center for Neuroscience, University of California Davis, 1544 Newton Ct., Davis, CA 95618, United States.
| | - Brian J Wiltgen
- Center for Neuroscience, University of California Davis, 1544 Newton Ct., Davis, CA 95618, United States; Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| |
Collapse
|
19
|
Echagarruga CT, Gheres KW, Norwood JN, Drew PJ. nNOS-expressing interneurons control basal and behaviorally evoked arterial dilation in somatosensory cortex of mice. eLife 2020; 9:e60533. [PMID: 33016877 PMCID: PMC7556878 DOI: 10.7554/elife.60533] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022] Open
Abstract
Cortical neural activity is coupled to local arterial diameter and blood flow. However, which neurons control the dynamics of cerebral arteries is not well understood. We dissected the cellular mechanisms controlling the basal diameter and evoked dilation in cortical arteries in awake, head-fixed mice. Locomotion drove robust arterial dilation, increases in gamma band power in the local field potential (LFP), and increases calcium signals in pyramidal and neuronal nitric oxide synthase (nNOS)-expressing neurons. Chemogenetic or pharmocological modulation of overall neural activity up or down caused corresponding increases or decreases in basal arterial diameter. Modulation of pyramidal neuron activity alone had little effect on basal or evoked arterial dilation, despite pronounced changes in the LFP. Modulation of the activity of nNOS-expressing neurons drove changes in the basal and evoked arterial diameter without corresponding changes in population neural activity.
Collapse
Affiliation(s)
| | - Kyle W Gheres
- Molecular, Cellular, and Integrative Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
| | - Jordan N Norwood
- Cell and Developmental Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
| | - Patrick J Drew
- Bioengineering Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Molecular, Cellular, and Integrative Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Cell and Developmental Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Departments of Engineering Science and Mechanics, Biomedical Engineering, and Neurosurgery, Pennsylvania State UniversityUniversity ParkUnited States
| |
Collapse
|
20
|
Schwabe MR, Taxier LR, Frick KM. It takes a neural village: Circuit-based approaches for estrogenic regulation of episodic memory. Front Neuroendocrinol 2020; 59:100860. [PMID: 32781195 PMCID: PMC7669700 DOI: 10.1016/j.yfrne.2020.100860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
Cognitive behaviors, such as episodic memory formation, are complex processes involving coordinated activity in multiple brain regions. However, much of the research on hormonal regulation of cognition focuses on manipulation of one region at a time or provides a single snapshot of how a systemic treatment affects multiple brain regions without investigating how these regions might interact to mediate hormone effects. Here, we use estrogenic regulation of episodic memory as an example of how circuit-based approaches may be incorporated into future studies of hormones and cognition. We first review basic episodic memory circuitry, rapid mechanisms by which 17β-estradiol can alter circuit activity, and current knowledge about 17β-estradiol's effects on episodic memory. Next, we outline approaches that researchers can employ to consider circuit effects in their estrogen research and provide examples of how these methods have been used to examine hormonal regulation of memory and other behaviors.
Collapse
Affiliation(s)
- Miranda R Schwabe
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
21
|
Marciante AB, Farmer GE, Cunningham JT. G q DREADD activation of CaMKIIa MnPO neurons stimulates nitric oxide activity. J Neurophysiol 2020; 124:591-609. [PMID: 32697679 PMCID: PMC7500373 DOI: 10.1152/jn.00239.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 11/22/2022] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) modify cellular activity following administration of the exogenous ligand clozapine-N-oxide (CNO). However, some reports indicate CNO may have off-target effects. The current studies investigate the use of Gq DREADDs in CaMKIIa-expressing neurons in the median preoptic nucleus (MnPO). Male Sprague-Dawley rats (250 g) anesthetized with isoflurane were stereotaxically microinjected in the MnPO with the Gq DREADD (AAV5-CaMKIIa-HM3D-mCherry) or control virus (AAV5-CaMKIIa-mCherry). Following a 2-wk recovery, rats were used for either immunohistochemical Fos analysis or in vitro patch-clamp electrophysiology. In Gq DREADD-injected rats, CNO induced significant increases in Fos staining in the MnPO and in regions that receive direct or indirect projections from the MnPO. In electrophysiological studies, CNO depolarized and augmented firing frequency in both Gq DREADD-positive neurons (Gq DREADD) as well as unlabeled MnPO neurons in slices from Gq DREADD-injected rats (Gq DREADDx). Gq DREADDx neurons also displayed increases in spontaneous postsynaptic current (sPSC) frequency in response to CNO. Additionally, CaMKIIa-positive MnPO neurons, which also express nitric oxide synthase (NOS), were treated with Nω-nitro-l-arginine (l-NNA; competitive inhibitor of NOS) and hemoglobin (NO scavenger) to assess the role of NO in Gq DREADDx neuron recruitment. Both l-NNA and hemoglobin blocked CNO-induced effects in Gq DREADDx neurons without affecting Gq DREADD neurons. These findings indicate that Gq DREADD-mediated activation of CaMKIIa/NOS expressing neurons in the MnPO can influence the activity of neighboring neurons. Future studies utilizing the use of Gq DREADDs will need to consider the potential recruitment of additional cell populations.NEW & NOTEWORTHY Rats were injected in the median preoptic nucleus (MnPO) with either an adeno-associated virus (AAV) and excitatory (Gq) designer receptor exclusively activated by designer drugs (DREADD) construct or a control AAV. In the Gq DREADD-injected rats only, clozapine-N-oxide (CNO) increased Fos staining in the MnPO and its targets and increased neuron action potential frequency. In electrophysiology experiments with slices with DREADD cells, unlabeled cells were activated and this was likely due to nitric oxide release by the DREADD cells.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Fort Worth, Texas
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Fort Worth, Texas
| | | |
Collapse
|
22
|
Poll S, Mittag M, Musacchio F, Justus LC, Giovannetti EA, Steffen J, Wagner J, Zohren L, Schoch S, Schmidt B, Jackson WS, Ehninger D, Fuhrmann M. Memory trace interference impairs recall in a mouse model of Alzheimer’s disease. Nat Neurosci 2020; 23:952-958. [DOI: 10.1038/s41593-020-0652-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
|
23
|
Hamilton KA, Santhakumar V. Current ex Vivo and in Vitro Approaches to Uncovering Mechanisms of Neurological Dysfunction after Traumatic Brain Injury. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 14:18-24. [PMID: 32548365 PMCID: PMC7297186 DOI: 10.1016/j.cobme.2020.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Traumatic brain injury often leads to progressive alterations at the molecular to circuit levels resulting in epilepsy and memory impairments. Ex vivo and in vitro models have provided a powerful platform for investigating the multimodal alteration after trauma. Recent ex vivo analyses using voltage sensitive dye imaging, optogenetics, and glutamate uncaging have revealed circuit abnormalities following in vivo brain injury. In vitro injury models have enabled examination of early and progressive changes in activity while development of three-dimensional organoids derived from human induced pluripotent stem cells have opened novel avenues for injury research. Here, we highlight recent advances in ex vivo and in vitro systems, focusing on their potential for advancing mechanistic understandings, possible limitations, and implications for therapeutics.
Collapse
Affiliation(s)
- Kelly Andrew Hamilton
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
| | - Vijayalakshmi Santhakumar
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
24
|
Anterior insula stimulation suppresses appetitive behavior while inducing forebrain activation in alcohol-preferring rats. Transl Psychiatry 2020; 10:150. [PMID: 32424183 PMCID: PMC7235223 DOI: 10.1038/s41398-020-0833-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 02/08/2023] Open
Abstract
The anterior insular cortex plays a key role in the representation of interoceptive effects of drug and natural rewards and their integration with attention, executive function, and emotions, making it a potential target region for intervention to control appetitive behaviors. Here, we investigated the effects of chemogenetic stimulation or inhibition of the anterior insula on alcohol and sucrose consumption. Excitatory or inhibitory designer receptors (DREADDs) were expressed in the anterior insula of alcohol-preferring rats by means of adenovirus-mediated gene transfer. Rats had access to either alcohol or sucrose solution during intermittent sessions. To characterize the brain network recruited by chemogenetic insula stimulation we measured brain-wide activation patterns using pharmacological magnetic resonance imaging (phMRI) and c-Fos immunohistochemistry. Anterior insula stimulation by the excitatory Gq-DREADDs significantly attenuated both alcohol and sucrose consumption, whereas the inhibitory Gi-DREADDs had no effects. In contrast, anterior insula stimulation failed to alter locomotor activity or deprivation-induced water drinking. phMRI and c-Fos immunohistochemistry revealed downstream activation of the posterior insula and medial prefrontal cortex, as well as of the mediodorsal thalamus and amygdala. Our results show the critical role of the anterior insula in regulating reward-directed behavior and delineate an insula-centered functional network associated with the effects of insula stimulation. From a translational perspective, our data demonstrate the therapeutic potential of circuit-based interventions and suggest that potentiation of insula excitability with neuromodulatory methods, such as repetitive transcranial magnetic stimulation (rTMS), could be useful in the treatment of alcohol use disorders.
Collapse
|
25
|
Kelly MP, Heckman PRA, Havekes R. Genetic manipulation of cyclic nucleotide signaling during hippocampal neuroplasticity and memory formation. Prog Neurobiol 2020; 190:101799. [PMID: 32360536 DOI: 10.1016/j.pneurobio.2020.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/14/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Decades of research have underscored the importance of cyclic nucleotide signaling in memory formation and synaptic plasticity. In recent years, several new genetic techniques have expanded the neuroscience toolbox, allowing researchers to measure and modulate cyclic nucleotide gradients with high spatiotemporal resolution. Here, we will provide an overview of studies using genetic approaches to interrogate the role cyclic nucleotide signaling plays in hippocampus-dependent memory processes and synaptic plasticity. Particular attention is given to genetic techniques that measure real-time changes in cyclic nucleotide levels as well as newly-developed genetic strategies to transiently manipulate cyclic nucleotide signaling in a subcellular compartment-specific manner with high temporal resolution.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, VA Bldg1, 3(rd) Fl, D-12, Columbia, 29209, SC, USA.
| | - Pim R A Heckman
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
26
|
Fredericks JM, Dash KE, Jaskot EM, Bennett TW, Lerchner W, Dold G, Ide D, Cummins AC, Der Minassian VH, Turchi JN, Richmond BJ, Eldridge MAG. Methods for mechanical delivery of viral vectors into rhesus monkey brain. J Neurosci Methods 2020; 339:108730. [PMID: 32302596 DOI: 10.1016/j.jneumeth.2020.108730] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Modern molecular tools make it possible to manipulate neural activity in a reversible and cell-type specific manner. For rhesus monkey research, molecular tools are generally introduced via viral vectors. New instruments designed specifically for use in monkey research are needed to enhance the efficiency and reliability of vector delivery. NEW METHOD A suite of multi-channel injection devices was developed to permit efficient and uniform vector delivery to cortical regions of the monkey brain. Manganese was co-infused with virus to allow rapid post-surgical confirmation of targeting accuracy using MRI. A needle guide was designed to increase the accuracy of sub-cortical targeting using stereotaxic co-ordinates. RESULTS The multi-channel injection devices produced dense, uniform coverage of dorsal surface cortex, ventral surface cortex, and intra-sulcal cortex, respectively. Co-infusion of manganese with the viral vector allowed for immediate verification of injection accuracy. The needle guide improved accuracy of targeting sub-cortical structures by preventing needle deflection. COMPARISON WITH EXISTING METHOD(S) The current methods, hand-held injections or single slow mechanical injection, for surface cortex transduction do not, in our hands, produce the density and uniformity of coverage provided by the injector arrays and associated infusion protocol. CONCLUSIONS The efficiency and reliability of vector delivery has been considerably improved by the development of new methods and instruments. This development should facilitate the translation of chemo- and optogenetic studies performed in smaller animals to larger animals such as rhesus monkeys.
Collapse
Affiliation(s)
- J Megan Fredericks
- Laboratory of Neuropsychology, NIMH/NIH/DHHS, Bethesda, MD, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10014, USA
| | - Kiana E Dash
- Laboratory of Neuropsychology, NIMH/NIH/DHHS, Bethesda, MD, USA
| | - Emilia M Jaskot
- Laboratory of Neuropsychology, NIMH/NIH/DHHS, Bethesda, MD, USA
| | | | - Walter Lerchner
- Laboratory of Neuropsychology, NIMH/NIH/DHHS, Bethesda, MD, USA
| | - George Dold
- Section on Instrumentation, NIH/DHHS, Bethesda, MD, USA
| | - David Ide
- Section on Instrumentation, NIH/DHHS, Bethesda, MD, USA
| | | | | | - Janita N Turchi
- Laboratory of Neuropsychology, NIMH/NIH/DHHS, Bethesda, MD, USA
| | | | | |
Collapse
|
27
|
The medial prefrontal cortex - hippocampus circuit that integrates information of object, place and time to construct episodic memory in rodents: Behavioral, anatomical and neurochemical properties. Neurosci Biobehav Rev 2020; 113:373-407. [PMID: 32298711 DOI: 10.1016/j.neubiorev.2020.04.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/25/2020] [Accepted: 04/06/2020] [Indexed: 12/31/2022]
Abstract
Rats and mice have been demonstrated to show episodic-like memory, a prototype of episodic memory, as defined by an integrated memory of the experience of an object or event, in a particular place and time. Such memory can be assessed via the use of spontaneous object exploration paradigms, variably designed to measure memory for object, place, temporal order and object-location inter-relationships. We review the methodological properties of these tests, the neurobiology about time and discuss the evidence for the involvement of the medial prefrontal cortex (mPFC), entorhinal cortex (EC) and hippocampus, with respect to their anatomy, neurotransmitter systems and functional circuits. The systematic analysis suggests that a specific circuit between the mPFC, lateral EC and hippocampus encodes the information for event, place and time of occurrence into the complex episodic-like memory, as a top-down regulation from the mPFC onto the hippocampus. This circuit can be distinguished from the neuronal component memory systems for processing the individual information of object, time and place.
Collapse
|
28
|
Grella SL, Fortin AH, McKissick O, Leblanc H, Ramirez S. Odor modulates the temporal dynamics of fear memory consolidation. ACTA ACUST UNITED AC 2020; 27:150-163. [PMID: 32179657 PMCID: PMC7079569 DOI: 10.1101/lm.050690.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/31/2019] [Indexed: 01/14/2023]
Abstract
Systems consolidation (SC) theory proposes that recent, contextually rich memories are stored in the hippocampus (HPC). As these memories become remote, they are believed to rely more heavily on cortical structures within the prefrontal cortex (PFC), where they lose much of their contextual detail and become schematized. Odor is a particularly evocative cue for intense remote memory recall and despite these memories being remote, they are highly contextual. In instances such as posttraumatic stress disorder (PTSD), intense remote memory recall can occur years after trauma, which seemingly contradicts SC. We hypothesized that odor may shift the organization of salient or fearful memories such that when paired with an odor at the time of encoding, they are delayed in the de-contextualization process that occurs across time, and retrieval may still rely on the HPC, where memories are imbued with contextually rich information, even at remote time points. We investigated this by tagging odor- and non-odor-associated fear memories in male c57BL/6 mice and assessed recall and c-Fos expression in the dorsal CA1 (dCA1) and prelimbic cortex (PL) 1 or 21 d later. In support of SC, our data showed that recent memories were more dCA1-dependent whereas remote memories were more PL-dependent. However, we also found that odor influenced this temporal dynamic, biasing the memory system from the PL to the dCA1 when odor cues were present. Behaviorally, inhibiting the dCA1 with activity-dependent DREADDs had no effect on recall at 1 d and unexpectedly caused an increase in freezing at 21 d. Together, these findings demonstrate that odor can shift the organization of fear memories at the systems level.
Collapse
Affiliation(s)
- Stephanie L Grella
- Psychological and Brain Sciences, Boston University, Boston, Massachusetts 02215, USA
| | - Amanda H Fortin
- Psychological and Brain Sciences, Boston University, Boston, Massachusetts 02215, USA
| | - Olivia McKissick
- Psychological and Brain Sciences, Boston University, Boston, Massachusetts 02215, USA
| | - Heloise Leblanc
- Psychological and Brain Sciences, Boston University, Boston, Massachusetts 02215, USA
| | - Steve Ramirez
- Psychological and Brain Sciences, Boston University, Boston, Massachusetts 02215, USA
| |
Collapse
|
29
|
Wang YC, Galeffi F, Wang W, Li X, Lu L, Sheng H, Hoffmann U, Turner DA, Yang W. Chemogenetics-mediated acute inhibition of excitatory neuronal activity improves stroke outcome. Exp Neurol 2020; 326:113206. [PMID: 31962128 DOI: 10.1016/j.expneurol.2020.113206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/07/2020] [Accepted: 01/17/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND PURPOSE Ischemic stroke significantly perturbs neuronal homeostasis leading to a cascade of pathologic events causing brain damage. In this study, we assessed acute stroke outcome after chemogenetic inhibition of forebrain excitatory neuronal activity. METHODS We generated hM4Di-TG transgenic mice expressing the inhibitory hM4Di, a Designer Receptors Exclusively Activated by Designer Drugs (DREADD)-based chemogenetic receptor, in forebrain excitatory neurons. Clozapine-N-oxide (CNO) was used to activate hM4Di DREADD. Ischemic stroke was induced by transient occlusion of the middle cerebral artery. Neurologic function and infarct volumes were evaluated. Excitatory neuronal suppression in the hM4Di-TG mouse forebrain was assessed electrophysiologically in vitro and in vivo, based on evoked synaptic responses, and in vivo based on occurrence of potassium-induced cortical spreading depolarizations. RESULTS Detailed characterization of hM4Di-TG mice confirmed that evoked synaptic responses in both in vitro hippocampal slices and in vivo motor cortex were significantly reduced after CNO-mediated activation of the inhibitory hM4Di DREADD. Further, CNO treatment had no obvious effects on physiology and motor function in either control or hM4Di-TG mice. Importantly, hM4Di-TG mice treated with CNO at either 10 min before ischemia or 30 min after reperfusion exhibited significantly improved neurologic function and smaller infarct volumes compared to CNO-treated control mice. Mechanistically, we showed that potassium-induced cortical spreading depression episodes were inhibited, including frequency and duration of DC shift, in CNO-treated hM4Di-TG mice. CONCLUSIONS Our data demonstrate that acute inhibition of a subset of excitatory neurons after ischemic stroke can prevent brain injury and improve functional outcome. This study, together with the previous work in optogenetic neuronal modulation during the chronic phase of stroke, supports the notion that targeting neuronal activity is a promising strategy in stroke therapy.
Collapse
Affiliation(s)
- Ya-Chao Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | | | - Wei Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Department of Anesthesiology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Xuan Li
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Liping Lu
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Huaxin Sheng
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Ulrike Hoffmann
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Dennis A Turner
- Research and Surgery Services, Durham VAMC, Durham, NC, USA; Departments of Neurosurgery, Neurobiology and Biomedical Engineering, Duke University Medical Center, Durham, NC, USA
| | - Wei Yang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
30
|
Chemogenetic Activation of Excitatory Neurons Alters Hippocampal Neurotransmission in a Dose-Dependent Manner. eNeuro 2019; 6:ENEURO.0124-19.2019. [PMID: 31645362 PMCID: PMC6860986 DOI: 10.1523/eneuro.0124-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 11/21/2022] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADD)-based chemogenetic tools are extensively used to manipulate neuronal activity in a cell type-specific manner. Whole-cell patch-clamp recordings indicate membrane depolarization, coupled with increased neuronal firing rate, following administration of the DREADD ligand, clozapine-N-oxide (CNO) to activate the Gq-coupled DREADD, hM3Dq. Although hM3Dq has been used to enhance neuronal firing in order to manipulate diverse behaviors, often within 30 min to 1 h after CNO administration, the physiological effects on excitatory neurotransmission remain poorly understood. We investigated the influence of CNO-mediated hM3Dq DREADD activation on distinct aspects of hippocampal excitatory neurotransmission at the Schaffer collateral-CA1 synapse in hippocampal slices derived from mice expressing hM3Dq in Ca2+/calmodulin-dependent protein kinase α (CamKIIα)-positive excitatory neurons. Our results indicate a clear dose-dependent effect on field EPSP (fEPSP) slope, with no change noted at the lower dose of CNO (1 µM) and a significant, long-term decline in fEPSP slope observed at higher doses (5-20 µM). Further, we noted a robust θ burst stimulus (TBS) induced long-term potentiation (LTP) in the presence of the lower CNO (1 µM) dose, which was significantly attenuated at the higher CNO (20 µM) dose. Whole-cell patch-clamp recording revealed both complex dose-dependent regulation of excitability, and spontaneous and evoked activity of CA1 pyramidal neurons in response to hM3Dq activation across CNO concentrations. Our data indicate that CNO-mediated activation of the hM3Dq DREADD results in dose-dependent regulation of excitatory hippocampal neurotransmission and highlight the importance of careful interpretation of behavioral experiments involving chemogenetic manipulation.
Collapse
|
31
|
Sun Y, Jin S, Lin X, Chen L, Qiao X, Jiang L, Zhou P, Johnston KG, Golshani P, Nie Q, Holmes TC, Nitz DA, Xu X. CA1-projecting subiculum neurons facilitate object-place learning. Nat Neurosci 2019; 22:1857-1870. [PMID: 31548723 PMCID: PMC6819262 DOI: 10.1038/s41593-019-0496-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 08/09/2019] [Indexed: 11/09/2022]
Abstract
Recent anatomical evidence suggests a functionally significant back-projection pathway from the subiculum to the CA1. Here we show that the afferent circuitry of CA1-projecting subicular neurons is biased by inputs from CA1 inhibitory neurons and the visual cortex, but lacks input from the entorhinal cortex. Efferents of the CA1-projecting subiculum neurons also target the perirhinal cortex, an area strongly implicated in object-place learning. We identify a critical role for CA1-projecting subicular neurons in object-location learning and memory, and show that this projection modulates place-specific activity of CA1 neurons and their responses to displaced objects. Together, these experiments reveal a novel pathway by which cortical inputs, particularly those from the visual cortex, reach the hippocampal output region CA1. Our findings also implicate this circuitry in the formation of complex spatial representations and learning of object-place associations.
Collapse
Affiliation(s)
- Yanjun Sun
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Suoqin Jin
- Department of Mathematics and Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Xiaoxiao Lin
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Lujia Chen
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Xin Qiao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Li Jiang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Pengcheng Zhou
- Department of Statistics and Center for Theoretical Neuroscience, Columbia University, New York, NY, USA
| | - Kevin G Johnston
- Department of Mathematics and Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Peyman Golshani
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- West Los Angeles VA Medical Center, Los Angeles, CA, USA
| | - Qing Nie
- Department of Mathematics and Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Douglas A Nitz
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA.
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA.
- Department of Computer Science, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
32
|
Hamlett ED, Ledreux A, Gilmore A, Vazey EM, Aston-Jones G, Boger HA, Paredes D, Granholm ACE. Inhibitory designer receptors aggravate memory loss in a mouse model of down syndrome. Neurobiol Dis 2019; 134:104616. [PMID: 31678403 DOI: 10.1016/j.nbd.2019.104616] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/06/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022] Open
Abstract
The pontine nucleus locus coeruleus (LC) is the primary source of noradrenergic (NE) projections to the brain and is important for working memory, attention, and cognitive flexibility. Individuals with Down syndrome (DS) develop Alzheimer's disease (AD) with high penetrance and often exhibit working memory deficits coupled with degeneration of LC-NE neurons early in the progression of AD pathology. Designer receptors exclusively activated by designer drugs (DREADDs) are chemogenetic tools that allow targeted manipulation of discrete neuronal populations in the brain without the confounds of off-target effects. We utilized male Ts65Dn mice (a mouse model for DS), and male normosomic (NS) controls to examine the effects of inhibitory DREADDs delivered via an AAV vector under translational control of the synthetic PRSx8, dopamine β hydroxylase (DβH) promoter. This chemogenetic tool allowed LC inhibition upon administration of the inert DREADD ligand, clozapine-N-oxide (CNO). DREADD-mediated LC inhibition impaired performance in a novel object recognition task and reversal learning in a spatial task. DREADD-mediated LC inhibition gave rise to an elevation of α-adrenoreceptors both in NS and in Ts65Dn mice. Further, microglial markers showed that the inhibitory DREADD stimulation led to increased microglial activation in the hippocampus in Ts65Dn but not in NS mice. These findings strongly suggest that LC signaling is important for intact memory and learning in Ts65Dn mice and disruption of these neurons leads to increased inflammation and dysregulation of adrenergic receptors.
Collapse
Affiliation(s)
- Eric D Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Aurélie Ledreux
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA
| | - Anah Gilmore
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA
| | - Elena M Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Gary Aston-Jones
- Rutgers Brain Health Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Heather A Boger
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Daniel Paredes
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA
| | | |
Collapse
|
33
|
Panthi S, Leitch B. The impact of silencing feed-forward parvalbumin-expressing inhibitory interneurons in the cortico-thalamocortical network on seizure generation and behaviour. Neurobiol Dis 2019; 132:104610. [PMID: 31494287 DOI: 10.1016/j.nbd.2019.104610] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/10/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022] Open
Abstract
Feed-forward inhibition (FFI) is an essential mechanism within the brain, to regulate neuronal firing and prevent runaway excitation. In the cortico-thalamocortical (CTC) network, fast spiking parvalbumin-expressing (PV+) inhibitory interneurons regulate the firing of pyramidal cells in the cortex and relay neurons in the thalamus. PV+ interneuron dysfunction has been implicated in several neurological disorders, including epilepsy. Previously, we demonstrated that loss of excitatory AMPA-receptors, specifically at synapses on PV+ interneurons in CTC feedforward microcircuits, occurs in the stargazer mouse model of absence epilepsy. These mice present with absence seizures characterized by spike and wave discharges (SWDs) on electroencephalogram (EEG) and concomitant behavioural arrest, similar to childhood absence epilepsy. The aim of the current study was to investigate the impact of loss of FFI within the CTC on absence seizure generation and behaviour using new Designer Receptor Exclusively Activated by Designer Drug (DREADD) technology. We crossed PV-Cre mice with Cre-dependent hM4Di DREADD strains of mice, which allowed Cre-recombinase-mediated restricted expression of inhibitory Gi-DREADDs in PV+ interneurons. We then tested the impact of global and focal (within the CTC network) silencing of PV+ interneurons. CNO mediated silencing of all PV+ interneurons by intraperitoneal injection caused the impairment of motor control, decreased locomotion and increased anxiety in a dose-dependent manner. Such silencing generated pathological oscillations similar to absence-like seizures. Focal silencing of PV+ interneurons within cortical or thalamic feedforward microcircuits, induced SWD-like oscillations and associated behavioural arrest. Epileptiform activity on EEG appeared significantly sooner after focal injection compared to peripheral injection of CNO. However, the mean duration of each oscillatory burst and spike frequency was similar, irrespective of mode of CNO delivery. No significant changes were observed in vehicle-treated or non-DREADD wild-type control animals. These data suggest that dysfunctional feed-forward inhibition in CTC microcircuits may be an important target for future therapy strategies for some patients with absence seizures. Additionally, silencing of PV+ interneurons in other brain regions may contribute to anxiety related neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Sandesh Panthi
- Department of Anatomy, School of Biomedical Sciences, and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Beulah Leitch
- Department of Anatomy, School of Biomedical Sciences, and Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
34
|
Kahn JB, Port RG, Yue C, Takano H, Coulter DA. Circuit-based interventions in the dentate gyrus rescue epilepsy-associated cognitive dysfunction. Brain 2019; 142:2705-2721. [PMID: 31363737 PMCID: PMC6736326 DOI: 10.1093/brain/awz209] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 12/15/2022] Open
Abstract
Temporal lobe epilepsy is associated with significant structural pathology in the hippocampus. In the dentate gyrus, the summative effect of these pathologies is massive hyperexcitability in the granule cells, generating both increased seizure susceptibility and cognitive deficits. To date, therapeutic approaches have failed to improve the cognitive symptoms in fully developed, chronic epilepsy. As the dentate's principal signalling population, the granule cells' aggregate excitability has the potential to provide a mechanistically-independent downstream target. We examined whether normalizing epilepsy-associated granule cell hyperexcitability-without correcting the underlying structural circuit disruptions-would constitute an effective therapeutic approach for cognitive dysfunction. In the systemic pilocarpine mouse model of temporal lobe epilepsy, the epileptic dentate gyrus excessively recruits granule cells in behavioural contexts, not just during seizure events, and these mice fail to perform on a dentate-mediated spatial discrimination task. Acutely reducing dorsal granule cell hyperactivity in chronically epileptic mice via either of two distinct inhibitory chemogenetic receptors rescued behavioural performance such that they responded comparably to wild type mice. Furthermore, recreating granule cell hyperexcitability in control mice via excitatory chemogenetic receptors, without altering normal circuit anatomy, recapitulated spatial memory deficits observed in epileptic mice. However, making the granule cells overly quiescent in both epileptic and control mice again disrupted behavioural performance. These bidirectional manipulations reveal that there is a permissive excitability window for granule cells that is necessary to support successful behavioural performance. Chemogenetic effects were specific to the targeted dorsal hippocampus, as hippocampal-independent and ventral hippocampal-dependent behaviours remained unaffected. Fos expression demonstrated that chemogenetics can modulate granule cell recruitment via behaviourally relevant inputs. Rather than driving cell activity deterministically or spontaneously, chemogenetic intervention merely modulates the behaviourally permissive activity window in which the circuit operates. We conclude that restoring appropriate principal cell tuning via circuit-based therapies, irrespective of the mechanisms generating the disease-related hyperactivity, is a promising translational approach.
Collapse
Affiliation(s)
- Julia B Kahn
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Russell G Port
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Cuiyong Yue
- The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Hajime Takano
- The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas A Coulter
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Vetere G, Borreca A, Pignataro A, Conforto G, Giustizieri M, Marinelli S, Ammassari-Teule M. Coincident Pre- and Post-Synaptic Cortical Remodelling Disengages Episodic Memory from Its Original Context. Mol Neurobiol 2019; 56:8513-8523. [PMID: 31267371 DOI: 10.1007/s12035-019-01652-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/15/2019] [Indexed: 11/28/2022]
Abstract
The view that the neocortex is remotely recruited for long-term episodic memory recall is challenged by data showing that an intense transcriptional and synaptic activity is detected in this region immediately after training. By measuring markers of synaptic activity at recent and remote time points from contextual fear conditioning (CFC), we could show that pre-synaptic changes are selectively detected 1 day post-training when the memory is anchored to the training context. Differently, pre- and post-synaptic changes are detected 14 days post-training when the memory generalizes to other contexts. Confirming that coincident pre- and post-synaptic remodelling mediates the disengagement of memory from its original context, DREADDs-mediated enhancement of cortical neuron activity during CFC training anticipates expression of a schematic memory and observation of bilateral synaptic remodelling. Together, our data show that the plastic properties of cortical synapses vary over time and specialise in relation to the quality of memory.
Collapse
Affiliation(s)
- Gisella Vetere
- Department of Experimental Neuroscience, Laboratory of Psychobiology, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy.,Laboratoire Plasticité du Cerveau, ESPCI-Ecole Supérieure de Physique et Chimie Industrielle, Paris, France
| | - Antonella Borreca
- Department of Experimental Neuroscience, Laboratory of Psychobiology, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy.,Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Rome, Italy.,Consiglio Nazionale delle Ricerche, Istituto di Neuroscienze, Milan, Italy
| | - Annabella Pignataro
- Department of Experimental Neuroscience, Laboratory of Psychobiology, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy.,Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Rome, Italy
| | - Giulia Conforto
- Department of Experimental Neuroscience, Laboratory of Psychobiology, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy
| | | | | | - Martine Ammassari-Teule
- Department of Experimental Neuroscience, Laboratory of Psychobiology, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy. .,Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Rome, Italy.
| |
Collapse
|
36
|
Chen X, Liu Z, Ma C, Ma L, Liu X. Parvalbumin Interneurons Determine Emotional Valence Through Modulating Accumbal Output Pathways. Front Behav Neurosci 2019; 13:110. [PMID: 31139063 PMCID: PMC6527764 DOI: 10.3389/fnbeh.2019.00110] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/02/2019] [Indexed: 11/13/2022] Open
Abstract
Parvalbumin (PV) expressing GABAergic interneurons provide large source of GABA to spiny projection neurons (SPNs) in the striatum. However, the roles of PV+ interneurons in the regulation of SPNs in the ventral striatum and emotional states are largely unknown. Here, we investigated whether stimulation of ventral striatal (accumbal) PV+ interneurons would drive emotional valence in mice. We found that during conditioned place preference (CPP) training, activation of accumbal PV+ interneurons evoked place preference while suppressing them resulted in conditioned place aversion (CPA). Activation of PV+ interneurons during place conditioning increased Fos expression in SPNs in the direct pathway (dSPNs) and impaired lithium chloride-induced CPA. Activation of dSPNs and SPNs in the indirect pathway (iSPNs) induced CPP and CPA, respectively; conversely, suppression of dSPNs or iSPNs induced CPA or CPP. In addition, activation or suppression of calretinin-expressing (CR) GABAergic interneurons did not induce place preference or aversion. These data suggest that PV+ interneurons can bidirectionally determine the emotional valence through their regulation of accumbal SPN activities and raise the possibility that manipulation of PV+ interneuron activity may have the potential to alter emotional valence and treat related mental disorders.
Collapse
Affiliation(s)
- Xi Chen
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhiyuan Liu
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Chaonan Ma
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Lan Ma
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xing Liu
- The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences and the Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
37
|
López AJ, Jia Y, White AO, Kwapis JL, Espinoza M, Hwang P, Campbell R, Alaghband Y, Chitnis O, Matheos DP, Lynch G, Wood MA. Medial habenula cholinergic signaling regulates cocaine-associated relapse-like behavior. Addict Biol 2019; 24:403-413. [PMID: 29430793 PMCID: PMC6087687 DOI: 10.1111/adb.12605] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/08/2017] [Accepted: 01/08/2018] [Indexed: 01/19/2023]
Abstract
Propensity to relapse, even following long periods of abstinence, is a key feature in substance use disorders. Relapse and relapse‐like behaviors are known to be induced, in part, by re‐exposure to drug‐associated cues. Yet, while many critical nodes in the neural circuitry contributing to relapse have been identified and studied, a full description of the networks driving reinstatement of drug‐seeking behaviors is lacking. One area that may provide further insight to the mechanisms of relapse is the habenula complex, an epithalamic region composed of lateral and medial (MHb) substructures, each with unique cell and target populations. Although well conserved across vertebrate species, the functions of the MHb are not well understood. Recent research has demonstrated that the MHb regulates nicotine aversion and withdrawal. However, it remains undetermined whether MHb function is limited to nicotine and aversive stimuli or if MHb circuit regulates responses to other drugs of abuse. Advances in circuit‐level manipulations now allow for cell‐type and temporally specific manipulations during behavior, specifically in spatially restrictive brain regions, such as the MHb. In this study, we focus on the response of the MHb to reinstatement of cocaine‐associated behavior, demonstrating that cocaine‐primed reinstatement of conditioned place preference engages habenula circuitry. Using chemogenetics, we demonstrate that MHb activity is sufficient to induce reinstatement behavior. Together, these data identify the MHb as a key hub in the circuitry underlying reinstatement and may serve as a target for regulating relapse‐like behaviors.
Collapse
Affiliation(s)
- Alberto J. López
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Yousheng Jia
- Department of Anatomy and Neurobiology, School of Medicine; University of California; Irvine CA USA
| | - André O. White
- Department of Biological Sciences, Neuroscience and Behavior; Mount Holyoke College; South Hadley MA USA
| | - Janine L. Kwapis
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Monica Espinoza
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Philip Hwang
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Rianne Campbell
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Yasaman Alaghband
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Om Chitnis
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Dina P. Matheos
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| | - Gary Lynch
- Department of Anatomy and Neurobiology, School of Medicine; University of California; Irvine CA USA
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, Ayala School of Biological Sciences; University of California; Irvine CA USA
- UC Irvine Center for Addiction Neuroscience, Ayala School of Biological Sciences; University of California; Irvine CA USA
- Center for the Neurobiology of Learning and Memory, Ayala School of Biological Sciences; University of California; Irvine CA USA
| |
Collapse
|
38
|
Kang S, Li J, Zuo W, Chen P, Gregor D, Fu R, Han X, Bekker A, Ye JH. Downregulation of M-channels in lateral habenula mediates hyperalgesia during alcohol withdrawal in rats. Sci Rep 2019; 9:2714. [PMID: 30804373 PMCID: PMC6389965 DOI: 10.1038/s41598-018-38393-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 12/21/2018] [Indexed: 01/21/2023] Open
Abstract
Hyperalgesia often occurs in alcoholics, especially during abstinence, yet the underlying mechanisms remain elusive. The lateral habenula (LHb) has been implicated in the pathophysiology of pain and alcohol use disorders. Suppression of m-type potassium channels (M-channels) has been found to contribute to the hyperactivity of LHb neurons of rats withdrawn from chronic alcohol administration. Here, we provided evidence that LHb M-channels may contribute to hyperalgesia. Compared to alcohol naïve counterparts, in male Long-Evans rats at 24-hours withdrawal from alcohol administration under the intermittent access paradigm for eight weeks, hyperalgesia was evident (as measured by paw withdrawal latencies in the Hargreaves Test), which was accompanied with higher basal activities of LHb neurons in brain slices, and lower M-channel protein expression. Inhibition of LHb neurons by chemogenetics, or pharmacological activation of M-channels, as well as overexpression of M-channels' subunit KCNQ3, relieved hyperalgesia and decreased relapse-like alcohol consumption. In contrast, chemogenetic activation of LHb neurons induced hyperalgesia in alcohol-naive rats. These data reveal a central role for the LHb in hyperalgesia during alcohol withdrawal, which may be due in part to the suppression of M-channels and, thus, highlights M-channels in the LHb as a potential therapeutic target for hyperalgesia in alcoholics.
Collapse
Affiliation(s)
- Seungwoo Kang
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jing Li
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Wanhong Zuo
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Pei Chen
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Danielle Gregor
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Rao Fu
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Xiao Han
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Alex Bekker
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA. .,Pharmacology, Physiology, and Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.
| |
Collapse
|
39
|
Chemogenetic activation of ventral tegmental area GABA neurons, but not mesoaccumbal GABA terminals, disrupts responding to reward-predictive cues. Neuropsychopharmacology 2019; 44:372-380. [PMID: 29875446 PMCID: PMC6300533 DOI: 10.1038/s41386-018-0097-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 05/03/2018] [Accepted: 05/05/2018] [Indexed: 12/19/2022]
Abstract
Cues predicting rewards can gain motivational properties and initiate reward-seeking behaviors. Dopamine projections from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) are critical in regulating cue-motivated responding. Although, approximately one third of mesoaccumbal projection neurons are GABAergic, it is unclear how this population influences motivational processes and cue processing. This is largely due to our inability to pharmacologically probe circuit level contributions of VTA-GABA, which arises from diverse sources, including multiple GABA afferents, interneurons, and projection neurons. Here we used a combinatorial viral vector approach to restrict activating Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) to GABA neurons in the VTA of wild-type rats trained to respond during a distinct audiovisual cue for sucrose. We measured different aspects of motivation for the cue or primary reinforcer, while chemogenetically activating either the VTA-GABA neurons or their projections to the NAc. Activation of VTA-GABA neurons decreased cue-induced responding and accuracy, while increasing latencies to respond to the cue and obtain the reward. Perseverative and spontaneous responses decreased, yet the rats persisted in entering the reward cup when the cue and reward were absent. However, activation of the VTA-GABA terminals in the accumbens had no effect on any of these behaviors. Together, we demonstrate that VTA-GABA neuron activity preferentially attenuates the ability of cues to trigger reward-seeking, while some aspects of the motivation for the reward itself are preserved. Additionally, the dense VTA-GABA projections to the NAc do not influence the motivational salience of the cue.
Collapse
|
40
|
Zhang T, Chen P, Li W, Sha S, Wang Y, Yuan Z, Shen B, Chen L. Cognitive deficits in mice lacking Nsun5, a cytosine-5 RNA methyltransferase, with impairment of oligodendrocyte precursor cells. Glia 2018; 67:688-702. [PMID: 30485550 DOI: 10.1002/glia.23565] [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: 05/19/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 12/15/2022]
Abstract
Williams-Beuren syndrome (WBS) is a microdeletion disorder with cognitive phenotype. NSUN5 gene, which encodes a cytosine-5 RNA methyltransferase, is located in WBS deletion locus. To investigate the influence of NSUN5 deletion on cognitive behaviors, we produced single-gene Nsun5 knockout (Nsun5-KO) mice. Here, we report that adult Nsun5-KO mice showed spatial cognitive deficits. Size of the brain and hippocampal structures and the number of CA1 or CA3 pyramidal cells in Nsun5-KO mice did not differ from WT mice. Basal properties of Schaffer collateral-CA1 synaptic transmission in Nsun5-KO mice were unchanged, but NMDA receptor (NMDAr)-dependent long-term potentiation (LTP) was not induced. The NMDA-evoked current in CA1 pyramidal cells was reduced in Nsun5-KO mice without the changes in expression and phosphorylation of NMDAr subunits NR2A and NR2B. Although the protein level of AMPA receptor subunit GluR2 was attenuated in Nsun5-KO mice, the AMPA-evoked current was not altered. Hippocampal immuno-staining showed the selective expression of Nsun5 in NG2 or PDGFRα labeled oligodendrocyte precursor cells (OPCs), but not in pyramidal cells or astrocytes. Analysis of RT-PCR determined the Nsun5 expression in purified populations of OPCs rather than neurons or astrocytes. The Nsun5 deficiency led to decreases in the number and neurite outgrowth of OPCs in the hippocampal CA1 and DG, with the decline in NG2 expression and OPCs proliferation. These findings indicate that the Nsun5 deletion suppresses NMDAr activity in neuronal cells probably through the disrupted development and function of OPCs, leading to deficits in NMDAr-dependent LTP and spatial cognitive abilities.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Peipei Chen
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Wei Li
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Sha Sha
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Zihao Yuan
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Bin Shen
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Physiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
41
|
Miyamoto K, Ishikura KI, Kume K, Ohsawa M. Astrocyte-neuron lactate shuttle sensitizes nociceptive transmission in the spinal cord. Glia 2018; 67:27-36. [DOI: 10.1002/glia.23474] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 05/17/2018] [Accepted: 05/25/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Keisuke Miyamoto
- Department of Neuropharmacology; Graduate School of Pharmaceutical Sciences, Nagoya City University; Nagoya Japan
| | - Kei-ichiro Ishikura
- Department of Neuropharmacology; Graduate School of Pharmaceutical Sciences, Nagoya City University; Nagoya Japan
| | - Kazuhiko Kume
- Department of Neuropharmacology; Graduate School of Pharmaceutical Sciences, Nagoya City University; Nagoya Japan
| | - Masahiro Ohsawa
- Department of Neuropharmacology; Graduate School of Pharmaceutical Sciences, Nagoya City University; Nagoya Japan
| |
Collapse
|
42
|
Chemogenetic inactivation of the dorsal hippocampus and medial prefrontal cortex, individually and concurrently, impairs object recognition and spatial memory consolidation in female mice. Neurobiol Learn Mem 2018; 156:103-116. [PMID: 30408525 PMCID: PMC7310386 DOI: 10.1016/j.nlm.2018.11.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/25/2018] [Accepted: 11/03/2018] [Indexed: 01/23/2023]
Abstract
The dorsal hippocampus (DH) and medial prefrontal cortex (mPFC) are brain regions essential for processing and storing episodic memory. In rodents, the DH has a well-established role in supporting the consolidation of episodic-like memory in tasks such as object recognition and object placement. However, the role of the mPFC in the consolidation of episodic-like memory tasks remains controversial. Therefore, the present study examined involvement of the DH and mPFC, alone and in combination, in object and spatial recognition memory consolidation in ovariectomized female mice. To this end, we utilized two types of inhibitory Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) to inactivate the DH alone, the mPFC alone, or both brain regions concurrently immediately after object training to assess the role of each region in the consolidation of object recognition and spatial memories. Our results using single and multiplexed DREADDS suggest that excitatory activity in the DH and mPFC, alone or in combination, is required for the successful consolidation of object recognition and spatial memories. Together, these studies provide critical insight into how the DH and mPFC work in concert to facilitate memory consolidation in female mice.
Collapse
|
43
|
Sarno E, Robison AJ. Emerging role of viral vectors for circuit-specific gene interrogation and manipulation in rodent brain. Pharmacol Biochem Behav 2018; 174:2-8. [PMID: 29709585 PMCID: PMC6369584 DOI: 10.1016/j.pbb.2018.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/10/2018] [Accepted: 04/24/2018] [Indexed: 01/11/2023]
Abstract
Over the past half century, novel tools have allowed the characterization of myriad molecular underpinnings of neural phenomena including synaptic function, neurogenesis and neurodegeneration, membrane excitability, and neurogenetics/epigenetics. More recently, transgenic mice have made possible cell type-specific explorations of these phenomena and have provided critical models of many neurological and psychiatric diseases. However, it has become clear that many critical areas of study require tools allowing the study and manipulation of individual neural circuits within the brain, and viral vectors have come to the forefront in driving these circuit-specific studies. Here, we present a surface-level review of the general classes of viral vectors used for study of the brain, along with their suitability for circuit-specific studies. We then cover in detail a new long-lasting, retrograde expressing form of herpes simplex virus termed LT-HSV that has become highly useful in circuit-based studies. We detail some of its current uses and propose a variety of future uses for this critical new tool, including circuit-based transgene overexpression, gene editing, and gene expression profiling.
Collapse
Affiliation(s)
- Erika Sarno
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
44
|
Alaghband Y, Kramár E, Kwapis JL, Kim ES, Hemstedt TJ, López AJ, White AO, Al-Kachak A, Aimiuwu OV, Bodinayake KK, Oparaugo NC, Han J, Lattal KM, Wood MA. CREST in the Nucleus Accumbens Core Regulates Cocaine Conditioned Place Preference, Cocaine-Seeking Behavior, and Synaptic Plasticity. J Neurosci 2018; 38:9514-9526. [PMID: 30228227 PMCID: PMC6209848 DOI: 10.1523/jneurosci.2911-17.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 08/29/2018] [Accepted: 09/06/2018] [Indexed: 11/21/2022] Open
Abstract
Epigenetic mechanisms result in persistent changes at the cellular level that can lead to long-lasting behavioral adaptations. Nucleosome remodeling is a major epigenetic mechanism that has not been well explored with regards to drug-seeking behaviors. Nucleosome remodeling is performed by multi-subunit complexes that interact with DNA or chromatin structure and possess an ATP-dependent enzyme to disrupt nucleosome-DNA contacts and ultimately regulate gene expression. Calcium responsive transactivator (CREST) is a transcriptional activator that interacts with enzymes involved in both histone acetylation and nucleosome remodeling. Here, we examined the effects of knocking down CREST in the nucleus accumbens (NAc) core on drug-seeking behavior and synaptic plasticity in male mice as well as drug-seeking in male rats. Knocking down CREST in the NAc core results in impaired cocaine-induced conditioned place preference (CPP) as well as theta-induced long-term potentiation in the NAc core. Further, similar to the CPP findings, using a self-administration procedure, we found that CREST knockdown in the NAc core of male rats had no effect on instrumental responding for cocaine itself on a first-order schedule, but did significantly attenuate responding on a second-order chain schedule, in which responding has a weaker association with cocaine. Together, these results suggest that CREST in the NAc core is required for cocaine-induced CPP, synaptic plasticity, as well as cocaine-seeking behavior.SIGNIFICANCE STATEMENT This study demonstrates a key role for the role of Calcium responsive transactivator (CREST), a transcriptional activator, in the nucleus accumbens (NAc) core with regard to cocaine-induced conditioned place preference (CPP), self-administration (SA), and synaptic plasticity. CREST is a unique transcriptional regulator that can recruit enzymes from two different major epigenetic mechanisms: histone acetylation and nucleosome remodeling. In this study we also found that the level of potentiation in the NAc core correlated with whether or not animals formed a CPP. Together the results indicate that CREST is a key downstream regulator of cocaine action in the NAc.
Collapse
Affiliation(s)
- Yasaman Alaghband
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Irvine Center for Addiction Neuroscience
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Enikö Kramár
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Janine L Kwapis
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, California 92697
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Earnest S Kim
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| | - Thekla J Hemstedt
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Alberto J López
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Irvine Center for Addiction Neuroscience
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - André O White
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Irvine Center for Addiction Neuroscience
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Amni Al-Kachak
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Osasumwen V Aimiuwu
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Kasuni K Bodinayake
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Nicole C Oparaugo
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - Joseph Han
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory,
- Irvine Center for Addiction Neuroscience
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, California 92697
- Center for the Neurobiology of Learning and Memory, Irvine, California, 92697, and
| |
Collapse
|
45
|
Wiegert JS, Pulin M, Gee CE, Oertner TG. The fate of hippocampal synapses depends on the sequence of plasticity-inducing events. eLife 2018; 7:39151. [PMID: 30311904 PMCID: PMC6205809 DOI: 10.7554/elife.39151] [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: 06/12/2018] [Accepted: 10/11/2018] [Indexed: 01/31/2023] Open
Abstract
Synapses change their strength in response to specific activity patterns. This functional plasticity is assumed to be the brain’s primary mechanism for information storage. We used optogenetic stimulation of rat hippocampal slice cultures to induce long-term potentiation (LTP), long-term depression (LTD), or both forms of plasticity in sequence. Two-photon imaging of spine calcium signals allowed us to identify stimulated synapses and to follow their fate for the next 7 days. We found that plasticity-inducing protocols affected the synapse’s chance for survival: LTP increased synaptic stability, LTD destabilized synapses, and the effect of the last stimulation protocol was dominant over earlier stimulations. Interestingly, most potentiated synapses were resistant to depression-inducing protocols delivered 24 hr later. Our findings suggest that activity-dependent changes in the transmission strength of individual synapses are transient, but have long-lasting consequences for synaptic lifetime.
Collapse
Affiliation(s)
- J Simon Wiegert
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mauro Pulin
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Elizabeth Gee
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Epigenetic regulation of the circadian gene Per1 contributes to age-related changes in hippocampal memory. Nat Commun 2018; 9:3323. [PMID: 30127461 PMCID: PMC6102273 DOI: 10.1038/s41467-018-05868-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 07/27/2018] [Indexed: 01/13/2023] Open
Abstract
Aging is accompanied by impairments in both circadian rhythmicity and long-term memory. Although it is clear that memory performance is affected by circadian cycling, it is unknown whether age-related disruption of the circadian clock causes impaired hippocampal memory. Here, we show that the repressive histone deacetylase HDAC3 restricts long-term memory, synaptic plasticity, and experience-induced expression of the circadian gene Per1 in the aging hippocampus without affecting rhythmic circadian activity patterns. We also demonstrate that hippocampal Per1 is critical for long-term memory formation. Together, our data challenge the traditional idea that alterations in the core circadian clock drive circadian-related changes in memory formation and instead argue for a more autonomous role for circadian clock gene function in hippocampal cells to gate the likelihood of long-term memory formation. Circadian rhythms are known to modulate memory, but it’s not known whether clock genes in the hippocampus are required for memory consolidation. Here, the authors show that epigenetic regulation of clock gene Period1 in the hippocampus regulates memory and contributes to age-related memory decline, independent of circadian rhythms.
Collapse
|
47
|
Treating a novel plasticity defect rescues episodic memory in Fragile X model mice. Mol Psychiatry 2018; 23:1798-1806. [PMID: 29133950 PMCID: PMC5951717 DOI: 10.1038/mp.2017.221] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/02/2017] [Accepted: 07/28/2017] [Indexed: 11/18/2022]
Abstract
Episodic memory, a fundamental component of human cognition, is significantly impaired in autism. We believe we report the first evidence for this problem in the Fmr1-knockout (KO) mouse model of Fragile X syndrome and describe potentially treatable underlying causes. The hippocampus is critical for the formation and use of episodes, with semantic (cue identity) information relayed to the structure via the lateral perforant path (LPP). The unusual form of synaptic plasticity expressed by the LPP (lppLTP) was profoundly impaired in Fmr1-KOs relative to wild-type mice. Two factors contributed to this defect: (i) reduced GluN1 subunit levels in synaptic NMDA receptors and related currents, and (ii) impaired retrograde synaptic signaling by the endocannabinoid 2-arachidonoylglycerol (2-AG). Studies using a novel serial cue paradigm showed that episodic encoding is dependent on both the LPP and the endocannabinoid receptor CB1, and is strikingly impaired in Fmr1-KOs. Enhancing 2-AG signaling rescued both lppLTP and learning in the mutants. Thus, two consequences of the Fragile-X mutation converge on plasticity at one site in hippocampus to prevent encoding of a basic element of cognitive memory. Collectively, the results suggest a clinically plausible approach to treatment.
Collapse
|
48
|
Wei J, Zhong P, Qin L, Tan T, Yan Z. Chemicogenetic Restoration of the Prefrontal Cortex to Amygdala Pathway Ameliorates Stress-Induced Deficits. Cereb Cortex 2018; 28:1980-1990. [PMID: 28498919 PMCID: PMC6018994 DOI: 10.1093/cercor/bhx104] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/27/2017] [Indexed: 01/11/2023] Open
Abstract
Corticosteroid stress hormones exert a profound impact on cognitive and emotional processes. Understanding the neuronal circuits that are altered by chronic stress is important for counteracting the detrimental effects of stress in a brain region- and cell type-specific manner. Using the chemogenetic tool, Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), which enables the remote, noninvasive and long-lasting modulation of cellular activity and signal transduction in discrete neuronal populations in vivo, we sought to identify the specific pathways that play an essential role in stress responses. We found that prolonged severe stress induced the diminished glutamatergic projection from pyramidal neurons in prefrontal cortex (PFC) to GABAergic interneurons in basolateral amygdala (BLA), leading to the loss of feedforward inhibition and ensuing hyperexcitability of BLA principal neurons, which caused a variety of behavioral abnormalities. Activating PFC pyramidal neurons with hM3D(Gq) DREADD restored the functional connection between PFC and BLA in stressed animals, resulting in the rescue of recognition memory, normalization of locomotor activity and reduction of aggressive behaviors. Inhibiting BLA principal neurons directly with hM4D(Gi) DREADD also blocked BLA hyperactivity and aggressive behaviors in stressed animals. These results have offered an effective avenue to counteract the stress-induced disruption of circuitry homeostasis.
Collapse
Affiliation(s)
- Jing Wei
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Luye Qin
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Tao Tan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| |
Collapse
|
49
|
Astrocytic Activation Generates De Novo Neuronal Potentiation and Memory Enhancement. Cell 2018; 174:59-71.e14. [PMID: 29804835 DOI: 10.1016/j.cell.2018.05.002] [Citation(s) in RCA: 349] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/31/2018] [Accepted: 04/27/2018] [Indexed: 12/20/2022]
Abstract
Astrocytes respond to neuronal activity and were shown to be necessary for plasticity and memory. To test whether astrocytic activity is also sufficient to generate synaptic potentiation and enhance memory, we expressed the Gq-coupled receptor hM3Dq in CA1 astrocytes, allowing their activation by a designer drug. We discovered that astrocytic activation is not only necessary for synaptic plasticity, but also sufficient to induce NMDA-dependent de novo long-term potentiation in the hippocampus that persisted after astrocytic activation ceased. In vivo, astrocytic activation enhanced memory allocation; i.e., it increased neuronal activity in a task-specific way only when coupled with learning, but not in home-caged mice. Furthermore, astrocytic activation using either a chemogenetic or an optogenetic tool during acquisition resulted in memory recall enhancement on the following day. Conversely, directly increasing neuronal activity resulted in dramatic memory impairment. Our findings that astrocytes induce plasticity and enhance memory may have important clinical implications for cognitive augmentation treatments.
Collapse
|
50
|
Cembrowski MS, Phillips MG, DiLisio SF, Shields BC, Winnubst J, Chandrashekar J, Bas E, Spruston N. Dissociable Structural and Functional Hippocampal Outputs via Distinct Subiculum Cell Classes. Cell 2018; 173:1280-1292.e18. [PMID: 29681453 DOI: 10.1016/j.cell.2018.03.031] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/22/2018] [Accepted: 03/14/2018] [Indexed: 12/30/2022]
Abstract
The mammalian hippocampus, comprised of serially connected subfields, participates in diverse behavioral and cognitive functions. It has been postulated that parallel circuitry embedded within hippocampal subfields may underlie such functional diversity. We sought to identify, delineate, and manipulate this putatively parallel architecture in the dorsal subiculum, the primary output subfield of the dorsal hippocampus. Population and single-cell RNA-seq revealed that the subiculum can be divided into two spatially adjacent subregions associated with prominent differences in pyramidal cell gene expression. Pyramidal cells occupying these two regions differed in their long-range inputs, local wiring, projection targets, and electrophysiological properties. Leveraging gene-expression differences across these regions, we use genetically restricted neuronal silencing to show that these regions differentially contribute to spatial working memory. This work provides a coherent molecular-, cellular-, circuit-, and behavioral-level demonstration that the hippocampus embeds structurally and functionally dissociable streams within its serial architecture.
Collapse
Affiliation(s)
- Mark S Cembrowski
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr., Ashburn, VA 20147, USA.
| | - Matthew G Phillips
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr., Ashburn, VA 20147, USA
| | - Salvatore F DiLisio
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr., Ashburn, VA 20147, USA
| | - Brenda C Shields
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr., Ashburn, VA 20147, USA
| | - Johan Winnubst
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr., Ashburn, VA 20147, USA
| | - Jayaram Chandrashekar
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr., Ashburn, VA 20147, USA
| | - Erhan Bas
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr., Ashburn, VA 20147, USA
| | - Nelson Spruston
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr., Ashburn, VA 20147, USA.
| |
Collapse
|